1
|
Sharma R, Ghosh R, Kumar S, Komal K, Kumar M. Decoding pancreatic cancer: key risk factors across genetics and lifestyle. Expert Rev Mol Diagn 2025; 25:95-99. [PMID: 39918844 DOI: 10.1080/14737159.2025.2464563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025]
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| |
Collapse
|
2
|
Dieli R, Lioy R, Crispo F, Cascelli N, Martinelli M, Lerose R, Telesca D, Milella MR, Colella M, Loperte S, Mazzoccoli C. The Oncoprotein Mucin 1 in Pancreatic Cancer Onset and Progression: Potential Clinical Implications. Biomolecules 2025; 15:275. [PMID: 40001578 PMCID: PMC11853026 DOI: 10.3390/biom15020275] [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: 12/11/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy characterized by poor prognosis, therapeutic resistance, and frequent recurrence. Current therapeutic options for PDAC include surgery, radiotherapy, immunological and targeted approaches. However, all these therapies provide only a slight improvement in patient survival. Consequently, the discovery of novel specific targets is becoming a priority to develop more effective treatments for PDAC. Mucin 1 (MUC1), a transmembrane glycoprotein, is aberrantly glycosylated and frequently overexpressed in pancreatic cancer. Recent studies highlighted the role of this oncoprotein in pancreatic carcinogenesis and its involvement in the acquisition of typical aggressive features of PDAC, like local invasion, metastases, and drug resistance. This review explores the mechanisms by which MUC1 contributes to cancer onset and progression, with a focus on its potential role as a biomarker and novel therapeutic target for pancreatic adenocarcinoma treatment.
Collapse
Affiliation(s)
- Rosalia Dieli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Rosa Lioy
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Nicoletta Cascelli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Mara Martinelli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Rosa Lerose
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Donatella Telesca
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Maria Rita Milella
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Marco Colella
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Simona Loperte
- Institute of Methodologies for Environmental Analysis, National Research Council, 85050 Tito Scalo, Italy;
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| |
Collapse
|
3
|
Choudhury S, Ghosh S, Chakraborty P, Pal S, Ghosh K, Saha S, Midha J, Sankar V, Mohata A, Chattopadhyay BK, Ghosh S, Das S, Basu B, Sikdar N. Epidermal Growth Factor Receptor (EGFR) and SMAD4 negatively correlated in the progression of gallbladder cancer in Eastern Indian patients. BMC Gastroenterol 2024; 24:446. [PMID: 39623311 PMCID: PMC11613908 DOI: 10.1186/s12876-024-03485-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND AND INTRODUCTION Two and half percent of the Indian population suffer from gallbladder cancer (GBC). The primary factors that lead GBC are associated with mutation of several protooncogenes such as EGFR, ERBB2, Myc, and CCND1 along with dysregulation of several tumor suppressor genes such as SMAD4 and CDKN2A. Bacterial infection caused by S.typhi and H.pylori are also hypothesized to be potential factors driving GBC. AIMS This study aims to investigate the molecular mechanisms driving the progression of gallbladder adenocarcinoma in Eastern Indian patients. We specifically focussed on analyzing the mutational status of the KRAS gene, examining the amplification of the ERBB2/Her2-neu gene, and evaluating the expression patterns of six dysregulated genes (CCND1, MYC, EGFR, ERBB2/Her2-neu, CDKN2A, SMAD4). Additionally, we assessed the expression status of TGF-beta, the association between bacterial infections (S. Typhi and H. pylori) and GBC, and the impact of single nucleotide polymorphisms in ERBB2/Her2-neu and CCND1 genes within this population. METHODS Sixty-seven samples from GBC-diagnosed patients, 26 other unrelated GBC samples for validation cohort, and 68 gallstone tissue samples were collected for this study. Genomic DNA from normal as well as tumor tissues were isolated, exon 2 and exon 3 of KRAS gene were amplified along, DNA sequenced and analyzed. KRAS codon 12 mutation was detected by allele specific PCR (ASPCR) method. Amplification of UreC A (coding for urease subunit α), VacA (coding for Vacuolating cytotoxin A) and CagA genes (coding for cytotoxin-associated gene A) in H.pylori were amplified using PCR. Similarly, FlicC (coding for flagellin gene C) in S.typhi was amplified using PCR. The ERBB2/Her2-neu SNP I655V, and CCND1 SNP A870G were analyzed using PCR followed by RFLP. Expression studies of CCND1, Myc, CDKN2A, ERBB2/Her2-neu, EGFR, and SMAD4 genes were measured in GBC tumor tissues by sybr green quantitative RT PCR. RESULTS The oncogenes (EGFR and ERBB2/Her2-neu) were statistically significantly overexpressed and the tumor suppressor gene (SMAD4) downregulated in our GBC tumor patient samples. The EGFR and SMAD4 genes were negatively correlated (r = -0.01) in GBC patients and the data is statistically significant and validated through IHC technique. A significant downregulation of TGF-beta had also been observed. Lower frequency (i.e. 11.5%) of KRAS mutation in GBC tumor was observed. CONCLUSIONS EGFR and SMAD4 expression were found to be negatively correlated in GBC tissue samples. ERBB2 overexpression/amplification was observed in 30% of the GBC samples. We also found a low percentage of GBC samples to show KRAS codon 12 mutation in Indian GBC patient population, as had been previously documented in pancreatic cancers.
Collapse
Affiliation(s)
- Sounetra Choudhury
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Sandip Ghosh
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Research Institute, Kolkata, India
| | - Prosenjeet Chakraborty
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Sayari Pal
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Koustuv Ghosh
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India
| | - Subhankar Saha
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | - Jitesh Midha
- Department of General Surgery, SSKM Hospital and IPGME&R, Kolkata, India
| | - Vinu Sankar
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | - Abhisek Mohata
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | | | - Shibajyoti Ghosh
- Department of General Surgery, Medical College and Hospital, Kolkata, India
| | - Soumen Das
- Department of General Surgery, SSKM Hospital and IPGME&R, Kolkata, India
| | - Biswarup Basu
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Research Institute, Kolkata, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata, 700108, India.
- Estuarine and Coastal Studies Foundation, Howrah, India.
| |
Collapse
|
4
|
Zohud O, Lone IM, Midlej K, Nashef A, Iraqi FA. Smad4 Heterozygous Knockout Effect on Pancreatic and Body Weight in F1 Population Using Collaborative Cross Lines. BIOLOGY 2024; 13:918. [PMID: 39596873 PMCID: PMC11592182 DOI: 10.3390/biology13110918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
Smad4, a critical tumor suppressor gene, plays a significant role in pancreatic biology and tumorigenesis. Genetic background and sex are known to influence phenotypic outcomes, but their impact on pancreatic weight in Smad4-deficient mice remains unclear. This study investigates the impact of Smad4 deficiency on pancreatic weight in first-generation (F1) mice from diverse collaborative cross (CC) lines, focusing on the influence of genetic background and sex. F1 mice were generated by crossbreeding female CC mice with C57BL/6J-Smad4tm1Mak males. Genotyping confirmed the presence of Smad4 knockout alleles. Mice were housed under standard conditions, euthanized at 80 weeks, and their pancreatic weights were measured, adjusted for body weight, and analyzed for effects of Smad4 deficiency, sex, and genetic background. The overall population of F1 mice showed a slight but non-significant increase in adjusted pancreatic weights in heterozygous knockout mice compared to wild-type mice. Sex-specific analysis revealed no significant difference in males but a significant increase in adjusted pancreatic weights in heterozygous knockout females. Genetic background analysis showed that lines CC018 and CC025 substantially increased adjusted pancreatic weights in heterozygous knockout mice. In contrast, other lines showed no significant difference or varied non-significant changes. The interplay between genetic background and sex further influenced these outcomes. Smad4 deficiency affects pancreatic weight in a manner significantly modulated by genetic background and sex. This study highlights the necessity of considering these factors in genetic research and therapeutic development, demonstrating the value of the collaborative cross mouse population in dissecting complex genetic interactions.
Collapse
Affiliation(s)
- Osayd Zohud
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.Z.); (I.M.L.); (K.M.)
| | - Iqbal M. Lone
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.Z.); (I.M.L.); (K.M.)
| | - Kareem Midlej
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.Z.); (I.M.L.); (K.M.)
| | - Aysar Nashef
- Department of Oral and Maxillofacial Surgery, Baruch Padeh Medical Center, Poriya 1528001, Israel;
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan 5290002, Israel
- Department of Oral and Maxillofacial Surgery, Meir Medical Center, Kfar Saba Affiliated to the Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel Aviv 6997801, Israel; (O.Z.); (I.M.L.); (K.M.)
| |
Collapse
|
5
|
Lee M, Ham H, Lee J, Lee ES, Chung CH, Kong DH, Park JR, Lee DK. TGF-β-Induced PAUF Plays a Pivotal Role in the Migration and Invasion of Human Pancreatic Ductal Adenocarcinoma Cell Line Panc-1. Int J Mol Sci 2024; 25:11420. [PMID: 39518973 PMCID: PMC11546992 DOI: 10.3390/ijms252111420] [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: 10/12/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic adenocarcinoma upregulated factor (PAUF) was initially identified as a secreted protein that is substantially expressed in pancreatic ductal adenocarcinoma (PDAC). PAUF also affects invasiveness, motility, and the proliferation of cells in several types of cancer. Recently, PAUF was reported to play a pivotal role in the TLR4-mediated migration and invasion of PDAC cells. However, the mechanism inducing PAUF expression and its functional role in TGF-β-stimulated PDAC cells have not yet been studied. Thus, we first assessed whether TGF-β regulates PAUF expression in several PDAC cell lines and found a significant increase in PAUF expression in Smad signaling-positive Panc-1 cells treated with TGF-β. We also found that the PAUF promoter region contains a Smad-binding element. TGF-β-treated Panc-1 cells showed an increase in PAUF promoter activity, but this effect was not observed in TGF-β-stimulated Smad4-null BxPC-3 cells. Restoring Smad4 expression increased the PAUF promoter activity and expression in Smad4-overexpressing BxPC-3 cells treated with TGF-β. We further found that PAUF aggravated the TGF-β-induced epithelial-mesenchymal transition (EMT) in Panc-1 and BxPC-3 cells via the activation of MEK-ERK signaling. These results indicate that TGF-β/Smad signaling-mediated upregulation of PAUF plays a crucial role in EMT progression by activating the TGF-β-mediated MEK-ERK signaling pathway.
Collapse
Affiliation(s)
- Miso Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| | - Hyejun Ham
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| | - Jiyeong Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| | - Eun Soo Lee
- Department of Internal Medicine, Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju 26426, Gangwon-do, Republic of Korea; (E.S.L.); (C.H.C.)
| | - Choon Hee Chung
- Department of Internal Medicine, Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju 26426, Gangwon-do, Republic of Korea; (E.S.L.); (C.H.C.)
| | - Deok-Hoon Kong
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| | - Jeong-Ran Park
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| | - Dong-Keon Lee
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon 24341, Gangwon-do, Republic of Korea; (M.L.); (H.H.); (J.L.); (D.-H.K.)
| |
Collapse
|
6
|
Wang B, Jiang Y, Zhu J, Wu H, Wu J, Li L, Huang J, Xiao Z, He Y. Fully-automated production of [ 68Ga]Ga-Trivehexin for clinical application and its biodistribution in healthy volunteers. Front Oncol 2024; 14:1445415. [PMID: 39156699 PMCID: PMC11327152 DOI: 10.3389/fonc.2024.1445415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Background The αvβ6-integrin targeting trimeric ligand [68Ga]Ga-Trivehexin has emerged as a promising candidate for clinical application due to its clinical imaging potentials in various malignant cancers. Our objective was to develop a simplified and reproducible module-based automated synthesis protocol to expand its availability in clinical application. Methods The pH value and the precursor load of radiolabeling were explored using an iQS-TS fully-automated module. Radiochemical purity was evaluated by radio-HPLC and radio-TLC. The ethanol content, radionuclide purity and identity, bacterial endotoxins, sterility, and stability of the final product [68Ga]Ga-Trivehexin were all tested. Biodistribution of [68Ga]Ga-Trivehexin in healthy volunteers was also conducted. Results The synthesis was explored and established using fully-automated module with outstanding radiochemical purity (>99%). Considering molar activity and economic costs, a pH of 3.6 and precursor dose of 30 μg were determined to be optimal. All relevant quality control parameters were tested and met the requirement of European Pharmacopoeia. In vitro stability test and imaging in healthy volunteer indicated the practical significance in clinical routines. Conclusions A fully-automated synthesis protocol for [68Ga]Ga-Trivehexin using the iQS-TS synthesis module was achieved and conformed to the clinical quality standards. Clinical trial registration ClinicalTrials.gov, NCT05835570. Registered 28 April 2023, https://www.clinicaltrials.gov/study/NCT05835570.
Collapse
Affiliation(s)
- Binchen Wang
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yaqun Jiang
- Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaxu Zhu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huiqin Wu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianyuan Wu
- Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ling Li
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianying Huang
- Clinical Trial Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhiwei Xiao
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yong He
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Caggiano EG, Taniguchi CM. UCP2 and pancreatic cancer: conscious uncoupling for therapeutic effect. Cancer Metastasis Rev 2024; 43:777-794. [PMID: 38194152 PMCID: PMC11156755 DOI: 10.1007/s10555-023-10157-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024]
Abstract
Pancreatic cancer has an exaggerated dependence on mitochondrial metabolism, but methods to specifically target the mitochondria without off target effects in normal tissues that rely on these organelles is a significant challenge. The mitochondrial uncoupling protein 2 (UCP2) has potential as a cancer-specific drug target, and thus, we will review the known biology of UCP2 and discuss its potential role in the pathobiology and future therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Emily G Caggiano
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Stefanoudakis D, Frountzas M, Schizas D, Michalopoulos NV, Drakaki A, Toutouzas KG. Significance of TP53, CDKN2A, SMAD4 and KRAS in Pancreatic Cancer. Curr Issues Mol Biol 2024; 46:2827-2844. [PMID: 38666907 PMCID: PMC11049225 DOI: 10.3390/cimb46040177] [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: 02/28/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
The present review demonstrates the major tumor suppressor genes, including TP53, CDKN2A and SMAD4, associated with pancreatic cancer. Each gene's role, prevalence and impact on tumor development and progression are analyzed, focusing on the intricate molecular landscape of pancreatic cancer. In addition, this review underscores the prognostic significance of specific mutations, such as loss of TP53, and explores some potential targeted therapies tailored to these molecular signatures. The findings highlight the importance of genomic analyses for risk assessment, early detection and the design of personalized treatment approaches in pancreatic cancer. Overall, this review provides a comprehensive analysis of the molecular intricacies of pancreatic tumors, paving the way for more effective and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Stefanoudakis
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Maximos Frountzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos V. Michalopoulos
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| | - Alexandra Drakaki
- Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Konstantinos G. Toutouzas
- First Propaedeutic Department of Surgery, Hippocration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (N.V.M.)
| |
Collapse
|
9
|
Fan G, Yan Q, Chen Y, Han M, Wu Z, Ruan S, Zhuang H, Gou Q, Hou B. FLNB overexpression promotes tumor progression and associates with immune suppression, evasion and stemness in pancreatic cancer. Am J Cancer Res 2024; 14:709-726. [PMID: 38455418 PMCID: PMC10915313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/11/2024] [Indexed: 03/09/2024] Open
Abstract
Pancreatic cancer (PC) is an immunosuppressive cancer. Immune-based therapies that enhance or recruit antitumor immune cells into the tumor microenvironment (TME) remain promising strategies for PC treatment. Consequently, a deeper understanding of the molecular mechanisms involved in PC immune suppression is critical for developing immune-based therapies to improve survival rates. In this study, weighted gene co-expression network analysis (WGCNA) was used to identify Filamin B (FLNB) correlated with the infiltration of CD8+ T cells and tumor-associated macrophages (TAMs). The clinical significance and potential biological function of FLNB were evaluated using bioinformatic analysis. The oncogenic role of FLNB in PC was determined using in vitro and in vivo studies. We further analyzed possible associations between FLNB expression and tumor immunity using CIBERSORT, single sample gene set enrichment analysis, and ESTIMATE algorithms. We found FLNB was overexpressed in PC tissues and was correlated with poorer overall survival, tumor recurrence, larger tumor size, and higher histologic grade. Moreover, FLNB overexpression was associated with the mutation status and expression of driver genes, especially for KRAS and SMAD4. Functional enrichment analysis identified the role of FLNB in the regulation of cell cycle, focal adhesion, vascular formation, and immune regulation. Knockdown of FLNB expression inhibited cancer cell proliferation and migration in-vitro and suppressed tumor growth in-vivo. Furthermore, FLNB overexpression caused high infiltration of Treg cells, Th2 cells, and TAMs, but reduced infiltration of CD8+ T cells and Th1/Th2. Collectively, our findings suggest FLNB promotes PC progression and may be a novel biomarker for PC.
Collapse
Affiliation(s)
- Guoyong Fan
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
| | - Qian Yan
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
- School of Medicine South China University of TechnologyGuangzhou, Guangdong, China
| | - Yubin Chen
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
- School of Medicine South China University of TechnologyGuangzhou, Guangdong, China
| | - Mingqian Han
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
| | - Zelong Wu
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
| | - Shiye Ruan
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
| | - Hongkai Zhuang
- Sun Yat-sen University Cancer CenterGuangzhou, Guangdong, China
| | - Qing Gou
- Department of Interventional Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
| | - Baohua Hou
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhou, Guangdong, China
- School of Medicine South China University of TechnologyGuangzhou, Guangdong, China
- Heyuan People’s HospitalHeyuan, Guangdong, China
| |
Collapse
|
10
|
Hartupee C, Nagalo BM, Chabu CY, Tesfay MZ, Coleman-Barnett J, West JT, Moaven O. Pancreatic cancer tumor microenvironment is a major therapeutic barrier and target. Front Immunol 2024; 15:1287459. [PMID: 38361931 PMCID: PMC10867137 DOI: 10.3389/fimmu.2024.1287459] [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: 09/01/2023] [Accepted: 01/04/2024] [Indexed: 02/17/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is projected to become the 2nd leading cause of cancer-related deaths in the United States. Limitations in early detection and treatment barriers contribute to the lack of substantial success in the treatment of this challenging-to-treat malignancy. Desmoplasia is the hallmark of PDAC microenvironment that creates a physical and immunologic barrier. Stromal support cells and immunomodulatory cells face aberrant signaling by pancreatic cancer cells that shifts the complex balance of proper repair mechanisms into a state of dysregulation. The product of this dysregulation is the desmoplastic environment that encases the malignant cells leading to a dense, hypoxic environment that promotes further tumorigenesis, provides innate systemic resistance, and suppresses anti-tumor immune invasion. This desmoplastic environment combined with the immunoregulatory events that allow it to persist serve as the primary focus of this review. The physical barrier and immune counterbalance in the tumor microenvironment (TME) make PDAC an immunologically cold tumor. To convert PDAC into an immunologically hot tumor, tumor microenvironment could be considered alongside the tumor cells. We discuss the complex network of microenvironment molecular and cellular composition and explore how they can be targeted to overcome immuno-therapeutic challenges.
Collapse
Affiliation(s)
- Conner Hartupee
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
- The Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
| | - Chiswili Y. Chabu
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, United States
- Siteman Cancer Center, Washington University, St. Louis, MO, United States
| | - Mulu Z. Tesfay
- Department of Pathology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
| | - Joycelynn Coleman-Barnett
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
| | - Omeed Moaven
- Division of Surgical Oncology, Department of Surgery, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Department of Interdisciplinary Oncology, Louisiana Cancer Research Center, Louisiana State University (LSU) Health, New Orleans, LA, United States
- Louisiana State University - Louisiana Children's Medical Center (LSU - LCMC) Cancer Center, New Orleans, LA, United States
| |
Collapse
|
11
|
Guo X, Wang P, Li Y, Chang Y, Wang X. Microbiomes in pancreatic cancer can be an accomplice or a weapon. Crit Rev Oncol Hematol 2024; 194:104262. [PMID: 38199428 DOI: 10.1016/j.critrevonc.2024.104262] [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: 08/31/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024] Open
Abstract
Recently, several investigations have linked the microbiome to pancreatic cancer progression. It is critical to reveal the role of different microbiomes in the occurrence, development, and treatment of pancreatic cancer. The current review summarizes the various microbiota types in pancreatic cancer while updating and supplementing the mechanisms of the representative gut, pancreatic, and oral microbiota, and their metabolites during its pathogenesis and therapeutic intervention. Several novel strategies have been introduced based on the tumor-associated microbiome to optimize the early diagnosis and prognosis of pancreatic cancer. The pros and cons involving different microbiomes in treating pancreatic cancer are discussed. The microbiome-related clinical trials for pancreatic cancer theranostics are outlined. This convergence of cutting-edge knowledge will provide feasible ideas for developing innovative therapies against pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoyu Guo
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Pan Wang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yuan Li
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yawei Chang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xiaobing Wang
- All authors are from the National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
12
|
Stanilov N, Velikova T, Stanilova S. Navigating the Cytokine Seas: Targeting Cytokine Signaling Pathways in Cancer Therapy. Int J Mol Sci 2024; 25:1009. [PMID: 38256080 PMCID: PMC10815616 DOI: 10.3390/ijms25021009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains one of the leading causes of morbidity and mortality worldwide, necessitating continuous efforts to develop effective therapeutic strategies. Over the years, advancements in our understanding of the complex interplay between the immune system and cancer cells have led to the development of immunotherapies that revolutionize cancer treatment. Cytokines, as key regulators of the immune response, are involved in both the initiation and progression of cancer by affecting inflammation and manipulating multiple intracellular signaling pathways that regulate cell growth, proliferation, and migration. Cytokines, as key regulators of inflammation, have emerged as promising candidates for cancer therapy. This review article aims to provide an overview of the significance of cytokines in cancer development and therapy by highlighting the importance of targeting cytokine signaling pathways as a potential therapeutic approach.
Collapse
Affiliation(s)
- Noyko Stanilov
- Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Spaska Stanilova
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
13
|
Jiang Z, Zheng X, Li M, Liu M. Improving the prognosis of pancreatic cancer: insights from epidemiology, genomic alterations, and therapeutic challenges. Front Med 2023; 17:1135-1169. [PMID: 38151666 DOI: 10.1007/s11684-023-1050-6] [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: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/29/2023]
Abstract
Pancreatic cancer, notorious for its late diagnosis and aggressive progression, poses a substantial challenge owing to scarce treatment alternatives. This review endeavors to furnish a holistic insight into pancreatic cancer, encompassing its epidemiology, genomic characterization, risk factors, diagnosis, therapeutic strategies, and treatment resistance mechanisms. We delve into identifying risk factors, including genetic predisposition and environmental exposures, and explore recent research advancements in precursor lesions and molecular subtypes of pancreatic cancer. Additionally, we highlight the development and application of multi-omics approaches in pancreatic cancer research and discuss the latest combinations of pancreatic cancer biomarkers and their efficacy. We also dissect the primary mechanisms underlying treatment resistance in this malignancy, illustrating the latest therapeutic options and advancements in the field. Conclusively, we accentuate the urgent demand for more extensive research to enhance the prognosis for pancreatic cancer patients.
Collapse
Affiliation(s)
- Zhichen Jiang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of General Surgery, Division of Gastroenterology and Pancreas, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Xiaohao Zheng
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
14
|
Kumar L, Kumar S, Sandeep K, Patel SKS. Therapeutic Approaches in Pancreatic Cancer: Recent Updates. Biomedicines 2023; 11:1611. [PMID: 37371705 DOI: 10.3390/biomedicines11061611] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer is a significant challenge for effective treatment due to its complex mechanism, different progressing stages, and lack of adequate procedures for screening and identification. Pancreatic cancer is typically identified in its advanced progression phase with a low survival of ~5 years. Among cancers, pancreatic cancer is also considered a high mortality-causing casualty over other accidental or disease-based mortality, and it is ranked seventh among all mortality-associated cancers globally. Henceforth, developing diagnostic procedures for its early detection, understanding pancreatic cancer-linked mechanisms, and various therapeutic strategies are crucial. This review describes the recent development in pancreatic cancer progression, mechanisms, and therapeutic approaches, including molecular techniques and biomedicines for effectively treating cancer.
Collapse
Affiliation(s)
- Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan 173229, India
| | - Sanjay Kumar
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida 201310, India
| | - Kumar Sandeep
- Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India
| | | |
Collapse
|
15
|
Myo Min KK, Ffrench CB, Jessup CF, Shepherdson M, Barreto SG, Bonder CS. Overcoming the Fibrotic Fortress in Pancreatic Ductal Adenocarcinoma: Challenges and Opportunities. Cancers (Basel) 2023; 15:2354. [PMID: 37190281 PMCID: PMC10137060 DOI: 10.3390/cancers15082354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
An overabundance of desmoplasia in the tumour microenvironment (TME) is one of the defining features that influences pancreatic ductal adenocarcinoma (PDAC) development, progression, metastasis, and treatment resistance. Desmoplasia is characterised by the recruitment and activation of fibroblasts, heightened extracellular matrix deposition (ECM) and reduced blood supply, as well as increased inflammation through an influx of inflammatory cells and cytokines, creating an intrinsically immunosuppressive TME with low immunogenic potential. Herein, we review the development of PDAC, the drivers that initiate and/or sustain the progression of the disease and the complex and interwoven nature of the cellular and acellular components that come together to make PDAC one of the most aggressive and difficult to treat cancers. We review the challenges in delivering drugs into the fortress of PDAC tumours in concentrations that are therapeutic due to the presence of a highly fibrotic and immunosuppressive TME. Taken together, we present further support for continued/renewed efforts focusing on aspects of the extremely dense and complex TME of PDAC to improve the efficacy of therapy for better patient outcomes.
Collapse
Affiliation(s)
- Kay K. Myo Min
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; (K.K.M.M.); (C.B.F.)
| | - Charlie B. Ffrench
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; (K.K.M.M.); (C.B.F.)
| | - Claire F. Jessup
- College of Medicine & Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Mia Shepherdson
- College of Medicine & Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Hepatopancreatobiliary & Liver Transplant Unit, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Savio George Barreto
- College of Medicine & Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Hepatopancreatobiliary & Liver Transplant Unit, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Claudine S. Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; (K.K.M.M.); (C.B.F.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
16
|
Balasundaram A, Kumar S U, D TK, Anil Dedge A, R G, K SS, R S, C GPD. The targeted next-generation sequence revealed SMAD4, AKT1, and TP53 mutations from circulating cell-free DNA of breast cancer and its effect on protein structure - A computational approach. J Biomol Struct Dyn 2023; 41:15584-15597. [PMID: 37011004 DOI: 10.1080/07391102.2023.2191122] [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: 07/22/2022] [Accepted: 03/06/2023] [Indexed: 04/04/2023]
Abstract
Breast cancer biomarkers that detect marginally advanced stages are still challenging. The detection of specific abnormalities, targeted therapy selection, prognosis, and monitoring of treatment effectiveness over time are all made possible by circulating free DNA (cfDNA) analysis. The proposed study will detect specific genetic abnormalities from the plasma cfDNA of a female breast cancer patient by sequencing a cancer-related gene panel (MGM455 - Oncotrack Ultima), including 56 theranostic genes (SNVs and small INDELs). Initially, we determined the pathogenicity of the observed mutations using PredictSNP, iStable, Align-GVGD, and ConSurf servers. As a next step, molecular dynamics (MD) was implemented to determine the functional significance of SMAD4 mutation (V465M). Lastly, the mutant gene relationships were examined using the Cytoscape plug-in GeneMANIA. Using ClueGO, we determined the gene's functional enrichment and integrative analysis. The structural characteristics of SMAD4 V465M protein by MD simulation analysis further demonstrated that the mutation was deleterious. The simulation showed that the native structure was more significantly altered by the SMAD4 (V465M) mutation. Our findings suggest that SMAD4 V465M mutation might be significantly associated with breast cancer, and other patient-found mutations (AKT1-E17K and TP53-R175H) are synergistically involved in the process of SMAD4 translocate to nuclease, which affects the target gene translation. Therefore, this combination of gene mutations could alter the TGF-β signaling pathway in BC. We further proposed that the SMAD4 protein loss may contribute to an aggressive phenotype by inhibiting the TGF-β signaling pathway. Thus, breast cancer's SMAD4 (V465M) mutation might increase their invasive and metastatic capabilities.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ambritha Balasundaram
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Udhaya Kumar S
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Thirumal Kumar D
- Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai, Tamil Nadu, India
| | - Aditi Anil Dedge
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Gnanasambandan R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Satish Srinivas K
- Department of Radiation Oncology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - Siva R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
17
|
Kung H, Yu J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. MedComm (Beijing) 2023; 4:e216. [PMID: 36814688 PMCID: PMC9939368 DOI: 10.1002/mco2.216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with a high rate of recurrence and a dismal 5-year survival rate. Contributing to the poor prognosis of PDAC is the lack of early detection, a complex network of signaling pathways and molecular mechanisms, a dense and desmoplastic stroma, and an immunosuppressive tumor microenvironment. A recent shift toward a neoadjuvant approach to treating PDAC has been sparked by the numerous benefits neoadjuvant therapy (NAT) has to offer compared with upfront surgery. However, certain aspects of NAT against PDAC, including the optimal regimen, the use of radiotherapy, and the selection of patients that would benefit from NAT, have yet to be fully elucidated. This review describes the major signaling pathways and molecular mechanisms involved in PDAC initiation and progression in addition to the immunosuppressive tumor microenvironment of PDAC. We then review current guidelines, ongoing research, and future research directions on the use of NAT based on randomized clinical trials and other studies. Finally, the current use of and research regarding targeted therapy for PDAC are examined. This review bridges the molecular understanding of PDAC with its clinical significance, development of novel therapies, and shifting directions in treatment paradigm.
Collapse
Affiliation(s)
- Heng‐Chung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yu
- Departments of Medicine and OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
18
|
He N, Lang X, Wang C, Lv C, Li M, Sun R, Zhang J. Expression of MSTN/Smad signaling pathway genes and its association with meat quality in Tibetan sheep ( Ovis aries). Food Sci Nutr 2023; 11:1836-1845. [PMID: 37051366 PMCID: PMC10084970 DOI: 10.1002/fsn3.3216] [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: 09/14/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 01/20/2023] Open
Abstract
Tibetan sheep is a unique breed living in Qinghai-Tibet Plateau. Since MSTN/Smad signaling pathway plays a critical role in the regulation of muscle development, we aimed to study the mutton quality, mRNA expression of main transduction genes in the MSTN/Smad signaling pathway, and the effects of those genes on the mutton quality of Tibetan sheep in this study. Six-month-old Qinghai-Tibetan sheep were selected, slaughtered, and their Longissimus lumborum, semitendinosus muscle, arm triceps, and quadriceps femoris muscle were collected. The mutton quality was evaluated, and gene expression and their association with the mutton quality were analyzed using RT-qPCR. The results showed that the indexes of mutton quality were not significantly different between ewes and rams (p > .05) except for Warner-Bratzler shear force (WBSF) (p < .05). A total of 21 different fatty acids were detected in the muscles of Tibetan sheep, including nine types of SFA, four types of MUFA, and eight types of PUFA. The main transduction genes of the MSTN/Smad signaling pathway were found to be widely expressed in muscle tissues, but no significant differences were observed (p > .05). The correlation analysis of the main genes and mutton quality showed that MSTN was significantly correlated with redness and cooking time; Smad2, Smad3, Smad4, and TGFβRI had significant positive correlations with marbling in arm triceps; Smad3 and TGFβRII had strong negative correlations with pH24 h in Longissimus lumborum; Smad2 was negatively correlated with drip loss in Longissimus lumborum. In short, the expression level of MSTN in muscles was positively correlated with Smad2, Smad3, and Smad4 genes and negatively correlated with TGFβRII genes. Thus, the results of this study provide a theoretical basis for the regulation mechanism of the MSTN/Smad pathway on mutton quality.
Collapse
Affiliation(s)
- Na He
- College of Agriculture and Animal Husbandry/Key Laboratory of Livestock and Poultry Genetics and Breeding on the Qinghai‐Tibet Plateau, Ministry of Agriculture and Rural Affairs/Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai ProvinceQinghai UniversityXiningQinghaiChina
| | - Xia Lang
- Gansu Key Laboratory of Cattle and Sheep Germplasm and Straw FodderGansu Academy of Agricultural SciencesLanzhouChina
| | - Cailian Wang
- Gansu Key Laboratory of Cattle and Sheep Germplasm and Straw FodderGansu Academy of Agricultural SciencesLanzhouChina
| | - Cailing Lv
- College of Agriculture and Animal Husbandry/Key Laboratory of Livestock and Poultry Genetics and Breeding on the Qinghai‐Tibet Plateau, Ministry of Agriculture and Rural Affairs/Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai ProvinceQinghai UniversityXiningQinghaiChina
| | - Mingming Li
- College of Agriculture and Animal Husbandry/Key Laboratory of Livestock and Poultry Genetics and Breeding on the Qinghai‐Tibet Plateau, Ministry of Agriculture and Rural Affairs/Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai ProvinceQinghai UniversityXiningQinghaiChina
| | - Ruizhe Sun
- College of Agriculture and Animal Husbandry/Key Laboratory of Livestock and Poultry Genetics and Breeding on the Qinghai‐Tibet Plateau, Ministry of Agriculture and Rural Affairs/Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai ProvinceQinghai UniversityXiningQinghaiChina
| | - Junxia Zhang
- College of Agriculture and Animal Husbandry/Key Laboratory of Livestock and Poultry Genetics and Breeding on the Qinghai‐Tibet Plateau, Ministry of Agriculture and Rural Affairs/Plateau Livestock Genetic Resources Protection and Innovative Utilization Key Laboratory of Qinghai ProvinceQinghai UniversityXiningQinghaiChina
| |
Collapse
|
19
|
Sant D, Rojekar S, Gera S, Pallapati A, Gimenez-Roig J, Kuo TC, Padilla A, Korkmaz F, Cullen L, Chatterjee J, Shelly E, Meseck M, Miyashita S, Macdonald A, Sultana F, Barak O, Ryu V, Kim SM, Robinson C, Rosen CJ, Caminis J, Lizneva D, Haider S, Yuen T, Zaidi M. Optimizing a therapeutic humanized follicle-stimulating hormone-blocking antibody formulation by protein thermal shift assay. Ann N Y Acad Sci 2023; 1521:67-78. [PMID: 36628526 PMCID: PMC11658029 DOI: 10.1111/nyas.14952] [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] [Indexed: 01/12/2023]
Abstract
Biopharmaceutical products are formulated using several Food and Drug Administration (FDA) approved excipients within the inactive ingredient limit to maintain their storage stability and shelf life. Here, we have screened and optimized different sets of excipient combinations to yield a thermally stable formulation for the humanized follicle-stimulating hormone (FSH)-blocking antibody, MS-Hu6. We used a protein thermal shift assay in which rising temperatures resulted in the maximal unfolding of the protein at the melting temperature (Tm ). To determine the buffer and pH for a stable solution, four different buffers with a pH range from 3 to 8 were screened. This resulted in maximal Tm s at pH 5.62 for Fab in phosphate buffer and at pH 6.85 for Fc in histidine buffer. Upon testing a range of salt concentrations, MS-Hu6 was found to be more stable at lower concentrations, likely due to reduced hydrophobic effects. Molecular dynamics simulations revealed a higher root-mean-square deviation with 1 mM than with 100 mM salt, indicating enhanced stability, as noted experimentally. Among the stabilizers tested, Tween 20 was found to yield the highest Tm and reversed the salt effect. Among several polyols/sugars, trehalose and sucrose were found to produce higher thermal stabilities. Finally, binding of recombinant human FSH to MS-Hu6 in a final formulation (20 mM phosphate buffer, 1 mM NaCl, 0.001% w/v Tween 20, and 260 mM trehalose) resulted in a thermal shift (increase in Tm ) for the Fab, but expectedly not in the Fc domain. Given that we used a low dose of MS-Hu6 (1 μM), the next challenge would be to determine whether 100-fold higher, industry-standard concentrations are equally stable.
Collapse
Affiliation(s)
- Damini Sant
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Satish Rojekar
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Sakshi Gera
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Anusha Pallapati
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Judit Gimenez-Roig
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Tan-Chun Kuo
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Ashley Padilla
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Jiya Chatterjee
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Eleanor Shelly
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Marcia Meseck
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Farhath Sultana
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Orly Barak
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Cemre Robinson
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Clifford J. Rosen
- Maine Medical Center Research Institute, Scarborough, Maine 040774, USA
| | - John Caminis
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Shozeb Haider
- UCL Centre for Advanced Research Computing, School of Pharmacy, London, WC1N 1AX, UK
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology and Departments of Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| |
Collapse
|
20
|
Ayasun R, Saridogan T, Gaber O, Sahin IH. Systemic Therapy for Patients With Pancreatic Cancer: Current Approaches and Opportunities for Novel Avenues Toward Precision Medicine. Clin Colorectal Cancer 2023; 22:2-11. [PMID: 36418197 PMCID: PMC11219281 DOI: 10.1016/j.clcc.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/01/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis with a 5-year overall survival of 11%. The disease is usually diagnosed at advanced stages, and systemic chemotherapy is the standard-of-care treatment for the majority of patients with PDAC. Although novel treatment options, such as targeted therapy and immunotherapy, have achieved substantial progress leading to practice-changing results, with FDA approvals for several solid tumors so far, the progress achieved for PDAC is relatively limited. Recent studies uncovered potential therapeutic targets for patients with PDAC, and potential therapeutic opportunities are currently being further examined. Herein, we review recent advances in systemic therapy regimens, including cytotoxic agents, targeted therapies, immunotherapy, and novel therapeutic options for managing patients with PDAC. We also elaborate on molecular profiling to guide treatment and existing therapeutic opportunities that may further advance the clinical care of patients with this devastating disease.
Collapse
Affiliation(s)
| | | | - Ola Gaber
- Department of Gastrointestinal Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ibrahim Halil Sahin
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Temel HY, Kaymak Ö, Kaplan S, Bahcivanci B, Gkoutos GV, Acharjee A. Role of microbiota and microbiota-derived short-chain fatty acids in PDAC. Cancer Med 2023; 12:5661-5675. [PMID: 36205023 PMCID: PMC10028056 DOI: 10.1002/cam4.5323] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive lethal diseases among other cancer types. Gut microbiome and its metabolic regulation play a crucial role in PDAC. Metabolic regulation in the gut is a complex process that involves microbiome and microbiome-derived short-chain fatty acids (SCFAs). SCFAs regulate inflammation, as well as lipid and glucose metabolism, through different pathways. This review aims to summarize recent developments in PDAC in the context of gut and oral microbiota and their associations with short-chain fatty acid (SCFA). In addition to this, we discuss possible therapeutic applications using microbiota in PDAC.
Collapse
Affiliation(s)
- Hülya Yılmaz Temel
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Öznur Kaymak
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Seren Kaplan
- Department of Bioengineering, Faculty of EngineeringEge UniversityIzmirTurkey
| | - Basak Bahcivanci
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| | - Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUK
- National Institute for Health Research Surgical Reconstruction, Queen Elizabeth Hospital BirminghamBirminghamUK
- MRC Health Data Research UK (HDR UK)BirminghamUK
| |
Collapse
|
22
|
Murugesan P, Begum H, Tangutur AD. Inhibitor of DNA binding/differentiation proteins as IDs for pancreatic cancer: Role in pancreatic cancer initiation, development and prognosis. Gene 2023; 853:147092. [PMID: 36464175 DOI: 10.1016/j.gene.2022.147092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
A family of inhibitors of cell differentiation or DNA-binding proteins, known as ID proteins (ID1-4), function as mighty transcription factors in various cellular processes, such as inhibiting differentiation, promoting cell-cycle progression, senescence, angiogenesis, tumorigenesis, and metastasis in cancer. Pancreatic cancer represents the deadliest cancer with the lowest survival rate of 10% due to the diagnosis at an advanced fatal stage and therapeutic resistance. Modestly, the only curative option for this lethal cancer is surgery but is done in less than 15-20% of patients because of the locally aggressive and early metastatic nature. Finding the earliest biomarkers and targeting the various hallmarks of pancreatic cancer can improve the treatment and survival of pancreatic cancer patients. Therefore, herein in this review, we explore in depth the potential roles of ID proteins function in hallmarks of pancreatic cancer, signaling pathways, and its oncogenic and tumor-suppressive effects. Hence, understanding the roles of dysregulated ID proteins would provide new insights into its function in pancreatic cancer tumorigenesis.
Collapse
Affiliation(s)
- Periyasamy Murugesan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Habeebunnisa Begum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
23
|
Bauer AH, Basta DW, Hornick JL, Dong F. Loss of function SMAD4 nonstop mutations in human cancer. Histopathology 2023; 82:1098-1104. [PMID: 36740808 DOI: 10.1111/his.14880] [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: 11/08/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND SMAD4 is a tumour suppressor gene that is mutated in a variety of cancers. SMAD4 nonstop mutations, which convert stop codons to sense codons that extend transcription, have been identified in genomic databases but have not been characterised in human pathology samples. The frequency of SMAD4 nonstop mutations and the consequences of nonstop mutations on SMAD4 protein expression are unknown. METHODS We retrospectively analysed our cancer sequencing database of 38,002 tumour specimens and evaluated the spectrum of SMAD4 mutations. SMAD4 protein expression was evaluated by immunohistochemistry in tumours with SMAD4 nonstop mutations. RESULTS In total, 1956 SMAD4 mutations were identified in 1822 tumours. SMAD4 mutations were most common in tumours of the gastrointestinal tract and included nonsense variants (n = 344), frameshift indels (n = 258), splice site variants (n = 104), and missense variants at codon R361 (n = 245). In a subset of cases with immunohistochemistry, SMAD4 expression was lost in 23 of 25 tumours (92%) with protein truncating variants and in 7 of 27 tumours (26%) with missense variants. Four cases harboured SMAD4 nonstop mutations. SMAD4 nonstop mutations were identified in two pancreatic adenocarcinomas, one colonic adenocarcinoma, and one non-small cell lung carcinoma. Immunohistochemistry demonstrated loss of SMAD4 protein expression in each of the four tumours with SMAD4 nonstop mutations. CONCLUSION SMAD4 nonstop mutations are associated with loss of SMAD4 protein expression in multiple tumour types. SMAD4 nonstop mutations should be clinically interpreted as pathogenic loss of function alterations when identified in cancer sequencing panels.
Collapse
Affiliation(s)
- Anna H Bauer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,University of Missouri School of Medicine, Columbia, MO, USA
| | - David W Basta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Lauria G, Curcio R, Lunetti P, Tiziani S, Coppola V, Dolce V, Fiermonte G, Ahmed A. Role of Mitochondrial Transporters on Metabolic Rewiring of Pancreatic Adenocarcinoma: A Comprehensive Review. Cancers (Basel) 2023; 15:411. [PMID: 36672360 PMCID: PMC9857038 DOI: 10.3390/cancers15020411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer is among the deadliest cancers worldwide and commonly presents as pancreatic ductal adenocarcinoma (PDAC). Metabolic reprogramming is a hallmark of PDAC. Glucose and glutamine metabolism are extensively rewired in order to fulfil both energetic and synthetic demands of this aggressive tumour and maintain favorable redox homeostasis. The mitochondrial pyruvate carrier (MPC), the glutamine carrier (SLC1A5_Var), the glutamate carrier (GC), the aspartate/glutamate carrier (AGC), and the uncoupling protein 2 (UCP2) have all been shown to influence PDAC cell growth and progression. The expression of MPC is downregulated in PDAC and its overexpression reduces cell growth rate, whereas the other four transporters are usually overexpressed and the loss of one or more of them renders PDAC cells unable to grow and proliferate by altering the levels of crucial metabolites such as aspartate. The aim of this review is to comprehensively evaluate the current experimental evidence about the function of these carriers in PDAC metabolic rewiring. Dissecting the precise role of these transporters in the context of the tumour microenvironment is necessary for targeted drug development.
Collapse
Affiliation(s)
- Graziantonio Lauria
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Paola Lunetti
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX 78723, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Fiermonte
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy
| |
Collapse
|
25
|
Lian Y, Zeng S, Wen S, Zhao X, Fang C, Zeng N. Review and Application of Integrin Alpha v Beta 6 in the Diagnosis and Treatment of Cholangiocarcinoma and Pancreatic Ductal Adenocarcinoma. Technol Cancer Res Treat 2023; 22:15330338231189399. [PMID: 37525872 PMCID: PMC10395192 DOI: 10.1177/15330338231189399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/13/2023] [Accepted: 06/28/2023] [Indexed: 08/02/2023] Open
Abstract
Integrin Alpha v Beta 6 is expressed primarily in solid epithelial tumors, such as cholangiocarcinoma, pancreatic cancer, and colorectal cancer. It has been considered a potential and promising molecular marker for the early diagnosis and treatment of cancer. Cholangiocarcinoma and pancreatic ductal adenocarcinoma share genetic, histological, and pathophysiological similarities due to the shared embryonic origin of the bile duct and pancreas. These cancers share numerous clinicopathological characteristics, including growth pattern, poor response to conventional radiotherapy and chemotherapy, and poor prognosis. This review focuses on the role of integrin Alpha v Beta 6 in cancer progression. It addition, it reviews how the marker can be used in molecular imaging and therapeutic targets. We propose further research explorations and questions that need to be addressed. We conclude that integrin Alpha v Beta 6 may serve as a potential biomarker for cancer disease progression and prognosis.
Collapse
Affiliation(s)
- Yunyu Lian
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Sai Wen
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Chihua Fang
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| | - Ning Zeng
- Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- First Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Clinical and Engineering Technology Center of Digital Medicine, Guangzhou, China
| |
Collapse
|
26
|
Matsumura K, Hayashi H, Uemura N, Ogata Y, Zhao L, Sato H, Shiraishi Y, Kuroki H, Kitamura F, Kaida T, Higashi T, Nakagawa S, Mima K, Imai K, Yamashita YI, Baba H. Thrombospondin-1 overexpression stimulates loss of Smad4 and accelerates malignant behavior via TGF-β signal activation in pancreatic ductal adenocarcinoma. Transl Oncol 2022; 26:101533. [PMID: 36115074 PMCID: PMC9483797 DOI: 10.1016/j.tranon.2022.101533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is characterized by abundant stroma and cancer-associated fibroblasts (CAFs) provide a favorable tumor microenvironment. Smad4 is known as tumor suppressor in several types of cancers including PDAC, and loss of Smad4 triggers accelerated cell invasiveness and metastatic potential. The thrombospondin-1 (TSP-1) can act as a major activator of latent transforming growth factor-β (TGF-β) in vivo. However, the roles of TSP-1 and the mediator of Smad4 loss and TGF-β signal activation during PDAC progression have not yet been addressed. The aim is to elucidate the biological role of TSP-1 in PDAC progression. METHODS AND RESULTS High substrate stiffness stimulated TSP-1 expression in CAFs, and TSP-1 knockdown inhibited cell proliferation with suppressed profibrogenic and activated stroma-related gene expressions in CAFs. Paracrine TSP-1 treatment for PDAC cells promoted cell proliferation and epithelial mesenchymal transition (EMT) with activated TGF-β signals such as phosphorylated Akt and Smad2/3 expressions. Surprisingly, knockdown of DPC4 (Smad4 gene) induced TSP-1 overexpression with TGF-β signal activation in PDAC cells. Interestingly, TSP-1 overexpression also induced downregulation of Smad4 expression and enhanced cell proliferation in vitro and in vivo. Treatment with LSKL peptide, which antagonizes TSP-1-mediated latent TGF-β activation, attenuated cell proliferation, migration and chemoresistance with enhanced apoptosis in PDAC cells. CONCLUSIONS TSP-1 derived from CAFs stimulates loss of Smad4 expression in cancer cells and accelerates malignant behavior by TGF-β signal activation in PDAC. TSP-1 could be a novel therapeutic target, not only for CAFs in stiff stroma, but also for cancer cells in the PDAC microenvironment.
Collapse
Affiliation(s)
- Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Liu Zhao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hideyuki Kuroki
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Fumimasa Kitamura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takayoshi Kaida
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan.
| |
Collapse
|
27
|
TRIM47 promotes glioma angiogenesis by suppressing Smad4. In Vitro Cell Dev Biol Anim 2022; 58:771-779. [PMID: 36203070 DOI: 10.1007/s11626-022-00722-6] [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: 06/07/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022]
Abstract
Angiogenesis is required for tumor progression; thus, its investigation can be useful to identify strategies for potential cancer treatments. Tripartite motif 47 (TRIM47) is involved in the progression of multiple cancers. However, its role in glioma angiogenesis is largely unknown. In this study, we first showed that TRIM47 is frequently upregulated in gliomas, and increased TRIM47 levels are correlated with microvascular density. We then examined the role of TRIM47 in cellular functions related to angiogenesis in vitro and observed that TRIM47 knockdown significantly reduced human umbilical vein endothelial cell proliferation, migration, and tube formation. We also found that TRIM47 silencing reduced vessel density and tumor volume in glioma xenografts. Mechanistically, TRIM47 negatively regulated Smad4 expression in glioma cells, and SMAD4 knockdown rescued the suppressive effects of TRIM47 silencing. Taken together, our results indicate that TRIM47 promotes angiogenesis in gliomas by downregulating SMAD4. Therefore, targeting the TRIM47/SMAD4 axis may offer an innovative approach to glioma treatment.
Collapse
|
28
|
Plaugher D, Aguilar B, Murrugarra D. Uncovering potential interventions for pancreatic cancer patients via mathematical modeling. J Theor Biol 2022; 548:111197. [PMID: 35752283 DOI: 10.1016/j.jtbi.2022.111197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/20/2022] [Accepted: 06/08/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is widely known for its poor prognosis because it is often diagnosed when the cancer is in a later stage. We built a Boolean model to analyze the microenvironment of pancreatic cancer in order to better understand the interplay between pancreatic cancer, stellate cells, and their signaling cytokines. Specifically, we have used our model to study the impact of inducing four common mutations: KRAS, TP53, SMAD4, and CDKN2A. After implementing the various mutation combinations, we used our stochastic simulator to derive aggressiveness scores based on simulated attractor probabilities and long-term trajectory approximations. These aggression scores were then corroborated with clinical data. Moreover, we found sets of control targets that are effective among common mutations. These control sets contain nodes within both the pancreatic cancer cell and the pancreatic stellate cell, including PIP3, RAF, PIK3 and BAX in pancreatic cancer cell as well as ERK and PIK3 in the pancreatic stellate cell. Many of these nodes were found to be differentially expressed among pancreatic cancer patients in the TCGA database. Furthermore, literature suggests that many of these nodes can be targeted by drugs currently in circulation. The results herein help provide a proof of concept in the path towards personalized medicine through a means of mathematical systems biology. All data and code used for running simulations, statistical analysis, and plotting is available on a GitHub repository athttps://github.com/drplaugher/PCC_Mutations.
Collapse
Affiliation(s)
- Daniel Plaugher
- Department of Mathematics, University of Kentucky, Lexington, KY, USA.
| | | | - David Murrugarra
- Department of Mathematics, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
29
|
Li Y, Zhang K, Peng L, Chen L, Gao H, Chen H. Multiple Perspectives Reveal the Role of DNA Damage Repair Genes in the Molecular Classification and Prognosis of Pancreatic Adenocarcinoma. Int J Mol Sci 2022; 23:ijms231810231. [PMID: 36142142 PMCID: PMC9499455 DOI: 10.3390/ijms231810231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly heterogeneous and immunosuppressive cancer. This study investigated the diversity of DNA damage repair (DDR) and immune microenvironment in PAAD by transcriptomic and genomic analysis. Patients with PAAD were divided into two DDR-based subtypes with distinct prognosis and molecular characteristics. The differential expression genes were mostly enriched in DDR and immune-related pathways. In order to distinguish high- and low-risk groups clinically, a DDR- and immune-based 5-gene prognostic signature (termed DPRS) was established. Patients in the high-risk group had inferior prognosis, a low level of immune checkpoint gene expression and low sensitivity to DDR-associated inhibitors. Furthermore, single-cell sequencing was used to observe the performance of the DDR-based signature in a high dimension, and immunohistochemistry was used to verify the relationship between the genes we identified and the prognosis of patients with PAAD. In conclusion, the DDR heterogeneity of PAAD was demonstrated, and a novel DDR- and immune-based risk-scoring model was constructed, which indicated the feasibility of DPRS in predicting prognosis and drug response in PAAD patients.
Collapse
Affiliation(s)
- Yujie Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ke Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Linjia Peng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huifeng Gao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hao Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-18017312356
| |
Collapse
|
30
|
Luo X, Xiao D, Zhang C, Wang G. The Roles of Exosomes upon Metallic Ions Stimulation in Bone Regeneration. J Funct Biomater 2022; 13:jfb13030126. [PMID: 36135561 PMCID: PMC9506099 DOI: 10.3390/jfb13030126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Metallic ions have been widely investigated and incorporated into bone substitutes for bone regeneration owing to their superior capacity to induce angiogenesis and osteogenesis. Exosomes are key paracrine mediators that play a crucial role in cell-to-cell communication. However, the role of exosomes in metallic ion-induced bone formation and their underlying mechanisms remain unclear. Thus, this review systematically analyzes the effects of metallic ions and metallic ion-incorporated biomaterials on exosome secretion from mesenchymal stem cells (MSCs) and macrophages, as well as the effects of secreted exosomes on inflammation, angiogenesis, and osteogenesis. In addition, possible signaling pathways involved in metallic ion-mediated exosomes, followed by bone regeneration, are discussed. Despite limited investigation, metallic ions have been confirmed to regulate exosome production and function, affecting immune response, angiogenesis, and osteogenesis. Although the underlying mechanism is not yet clear, these insights enrich our understanding of the mechanisms of the metallic ion-induced microenvironment for bone regeneration, benefiting the design of metallic ion-incorporated implants.
Collapse
Affiliation(s)
- Xuwei Luo
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong 637000, China
| | - Dongqin Xiao
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong 637000, China
- Correspondence: (D.X.); (G.W.)
| | - Chengdong Zhang
- Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong 637000, China
| | - Guanglin Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (D.X.); (G.W.)
| |
Collapse
|
31
|
Zhu C, Guan X, Zhang X, Luan X, Song Z, Cheng X, Zhang W, Qin JJ. Targeting KRAS mutant cancers: from druggable therapy to drug resistance. Mol Cancer 2022; 21:159. [PMID: 35922812 PMCID: PMC9351107 DOI: 10.1186/s12943-022-01629-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) is the most frequently mutated oncogene, occurring in a variety of tumor types. Targeting KRAS mutations with drugs is challenging because KRAS is considered undruggable due to the lack of classic drug binding sites. Over the past 40 years, great efforts have been made to explore routes for indirect targeting of KRAS mutant cancers, including KRAS expression, processing, upstream regulators, or downstream effectors. With the advent of KRAS (G12C) inhibitors, KRAS mutations are now druggable. Despite such inhibitors showing remarkable clinical responses, resistance to monotherapy of KRAS inhibitors is eventually developed. Significant progress has been made in understanding the mechanisms of drug resistance to KRAS-mutant inhibitors. Here we review the most recent advances in therapeutic approaches and resistance mechanisms targeting KRAS mutations and discuss opportunities for combination therapy.
Collapse
Affiliation(s)
- Chunxiao Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China
| | - Xinuo Zhang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China.,College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhengbo Song
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Xiangdong Cheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China. .,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China. .,School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China. .,Key Laboratory of Prevention, Diagnosis, and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, 310022, China.
| |
Collapse
|
32
|
Repositioning of Old Drugs for Novel Cancer Therapies: Continuous Therapeutic Perfusion of Aspirin and Oseltamivir Phosphate with Gemcitabine Treatment Disables Tumor Progression, Chemoresistance, and Metastases. Cancers (Basel) 2022; 14:cancers14153595. [PMID: 35892853 PMCID: PMC9331689 DOI: 10.3390/cancers14153595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Repositioning old drugs in combination with clinical standard chemotherapeutics opens a promising clinical treatment approach for patients with pancreatic cancer. This report presents a therapeutic repositioning of continuous perfusion of aspirin and oseltamivir phosphate in combination with gemcitabine treatment as an effective treatment option for pancreatic cancer. The data suggest that repositioning these drugs with continuous perfusion with gemcitabine disables chemoresistance, tumor progression, EMT program, cancer stem cells, and metastases in a preclinical mouse model of human pancreatic cancer. These promising results warrant additional investigation to assess the potential of translating into the clinical setting to improve the cancer patient prognosis for an otherwise fatal disease. Abstract Metastatic pancreatic cancer has an invariably fatal outcome, with an estimated median progression-free survival of approximately six months employing our best combination chemotherapeutic regimens. Once drug resistance develops, manifested by increased primary tumor size and new and growing metastases, patients often die rapidly from their disease. Emerging evidence indicates that chemotherapy may contribute to the development of drug resistance through the upregulation of epithelial–mesenchymal transition (EMT) pathways and subsequent cancer stem cell (CSC) enrichment. Neuraminidase-1 (Neu-1) regulates the activation of several receptor tyrosine kinases implicated in EMT induction, angiogenesis, and cellular proliferation. Here, continuous therapeutic targeting of Neu-1 using parenteral perfusion of oseltamivir phosphate (OP) and aspirin (ASA) with gemcitabine (GEM) treatment significantly disrupts tumor progression, critical compensatory signaling mechanisms, EMT program, CSC, and metastases in a preclinical mouse model of human pancreatic cancer. ASA- and OP-treated xenotumors significantly inhibited the metastatic potential when transferred into animals.
Collapse
|
33
|
Polysaccharides Extracted from Angelica sinensis (Oliv.) Diels Relieve the Malignant Characteristics of Glioma Cells through Regulating the MiR-373-3p-Mediated TGF- β/Smad4 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7469774. [PMID: 35855826 PMCID: PMC9288290 DOI: 10.1155/2022/7469774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 11/18/2022]
Abstract
Objectives Angelica sinensis polysaccharide (ASP) is a traditional herbal medicine accompanied by antitumor potential. This study aims to explore the therapeutic potential of ASP on glioma, as well as the underlying mechanisms involving microRNA-373-3p (miR-373-3p) and the TGF-β/Smad4 signaling pathway. Methods U251 cells (a human glioma cell line) were treated with different concentrations of ASP. miR-373-3p was silenced in U251 cells by the transfection of the miR-373-3p inhibitor. Cell viability and apoptosis were measured by CCK-8 assay and flow cytometry, respectively. Cell migration and invasion were detected by wound healing and transwell assays, respectively. The miR-373-3p expression was measured by RT-qPCR. The protein expressions of TGF-β and Smad4 were evaluated by both western blotting and immunofluorescence. Results ASP inhibited the viability, migration, and invasion, and enhanced the apoptosis of U251 cells in a dose-dependent manner. ASP increased miR-373-3p expression and decreased TGF-β and Smad4 expressions in U251 cells. Silencing of miR-373-3p weakened the effects of ASP on inhibiting cell viability, migration, and invasion, as well as promoting cell apoptosis. In addition, deleting miR-373-3p weakened the inhibiting effects of ASP on the TGF-β/Smad4 pathway in U251 cells. Conclusions ASP suppresses the malignant progression of glioma via regulating the miR-373-3p-mediated TGF-β/Smad4 pathway.
Collapse
|
34
|
It Takes Two to Tango: Potential Prognostic Impact of Circulating TGF-Beta and PD-L1 in Pancreatic Cancer. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070960. [PMID: 35888050 PMCID: PMC9323895 DOI: 10.3390/life12070960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly devastating disease with rising incidence and poor prognosis. The lack of reliable prognostic biomarkers hampers the individual evaluation of the survival and recurrence potential. Methods: Here, we investigate the value of plasma levels of two potential key players in molecular mechanisms underlying PDAC aggressiveness and immune evasion, soluble TGF-beta (sTGF-beta) and sPD-L1, in both metastatic and radically-resected PDAC. To this aim we prospectively enrolled 38 PDAC patients and performed appropriate statistical analyses in order to evaluate their correlation, and role in the prediction of disease relapse/progression, and patients’ outcome. Results: Metastatic patients showed lower levels of circulating sTGF-beta and higher levels of sPD-L1 compared to radically-resected patients. Moreover, a decrease in sTGF-beta levels (but not sPD-L1) was significantly associated with disease relapse in radically-resected patients. We also observed lower sTGF-beta at disease progression after first-line chemotherapy in metastatic patients, though this change was not statistically significant. We found a significant correlation between the levels of sTGF-beta and sPD-L1 before first-line chemotherapy. Conclusions: These findings support the possible interaction of TGF-beta and PD-L1 pathways and suggest that sTGF-beta and sPD-L1 might synergize and be new potential blood-based biomarkers.
Collapse
|
35
|
Ko CC, Hsieh YY, Yang PM. Long Non-Coding RNA MIR31HG Promotes the Transforming Growth Factor β-Induced Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma Cells. Int J Mol Sci 2022; 23:6559. [PMID: 35743003 PMCID: PMC9223781 DOI: 10.3390/ijms23126559] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) describes a biological process in which polarized epithelial cells are converted into highly motile mesenchymal cells. It promotes cancer cell dissemination, allowing them to form distal metastases, and also involves drug resistance in metastatic cancers. Transforming growth factor β (TGFβ) is a multifunctional cytokine that plays essential roles in development and carcinogenesis. It is a major inducer of the EMT. The MIR31 host gene (MIR31HG) is a newly identified long non-coding (lnc)RNA that exhibits ambiguous roles in cancer. In this study, a cancer genomics analysis predicted that MIR31HG overexpression was positively correlated with poorer disease-free survival of pancreatic ductal adenocarcinoma (PDAC) patients, which was associated with upregulation of genes related to TGFβ signaling and the EMT. In vitro evidence demonstrated that TGFβ induced MIR31HG expression in PDAC cells, and knockdown of MIR31HG expression reversed TGFβ-induced EMT phenotypes and cancer cell migration. Therefore, MIR31HG has an oncogenic role in PDAC by promoting the EMT.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan 71004, Taiwan;
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Taipei Medical University Shuang Ho Hospital, New Taipei City 23561, Taiwan;
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Ming Yang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- TMU and Affiliated Hospitals Pancreatic Cancer Groups, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
36
|
Sim W, Lim WM, Hii LW, Leong CO, Mai CW. Targeting pancreatic cancer immune evasion by inhibiting histone deacetylases. World J Gastroenterol 2022; 28:1934-1945. [PMID: 35664961 PMCID: PMC9150054 DOI: 10.3748/wjg.v28.i18.1934] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a vital role in maintaining the delicate balance between immune recognition and tumor development. Regardless, it is not uncommon that cancerous cells can intelligently acquire abilities to bypass the antitumor immune responses, thus allowing continuous tumor growth and development. Immune evasion has emerged as a significant factor contributing to the progression and immune resistance of pancreatic cancer. Compared with other cancers, pancreatic cancer has a tumor microenvironment that can resist most treatment modalities, including emerging immunotherapy. Sadly, the use of immunotherapy has yet to bring significant clinical breakthrough among pancreatic cancer patients, suggesting that pancreatic cancer has successfully evaded immunomodulation. In this review, we summarize the impact of genetic alteration and epigenetic modification (especially histone deacetylases, HDAC) on immune evasion in pancreatic cancer. HDAC overexpression significantly suppresses tumor suppressor genes, contributing to tumor growth and progression. We review the evidence on HDAC inhibitors in tumor eradication, improving T cells activation, restoring tumor immunogenicity, and modulating programmed death 1 interaction. We provide our perspective in targeting HDAC as a strategy to reverse immune evasion in pancreatic cancer.
Collapse
Affiliation(s)
- Wynne Sim
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Wei-Meng Lim
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Ling-Wei Hii
- School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Chee-Onn Leong
- Center for Cancer and Stem Cell Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
- AGTC Genomics, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
37
|
Qi F, Tang J, Cai Z, Wang G, Wang Z. Long non-coding RNA CATIP antisense RNA 1 (lncRNA CATIP-AS1) downregulation contributes to the progression and metastasis of thyroid cancer via epithelial-mesenchymal transition (EMT) pathway. Bioengineered 2022; 13:7592-7606. [PMID: 35264071 PMCID: PMC8973971 DOI: 10.1080/21655979.2022.2047400] [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] [Indexed: 12/20/2022] Open
Abstract
Thyroid cancer (THCA) is the most common cancer of the endocrine system across the globe. To date, the mechanism of development of THCA remains scarcely known. In this study, we aim to elucidate the long non-coding RNA CATIP antisense RNA 1 (lncRNA CATIP-AS1/CATIP-AS1) role in the pathogenesis of THCA and its regulatory mechanism. The result shows that the CATIP-AS1 was significantly downregulated in THCA tissues and cells and was associated with a poor prognosis of patients diagnosed with THCA. The overexpression of CATIP-AS1 significantly inhibited THCA cell proliferation, migration, and epithelial–mesenchymal transition (EMT) but increased the THCA cell apoptosis. We found that CATIP-AS1 endogenously sponges miR-515-5p and its overexpression could inhibit miR-515-5p regulatory effect. Moreover, the overexpression of miR-515-5p repressed the Smad4 expression level, consequently reversed the inhibiting effect of overexpressed CATIP-AS1 on the proliferation, and migration of THCA cell. It also reversed the increased THCA cell apoptosis and the downregulated-CATIP-AS1-induced cell EMT inhibition. Summarily, we demonstrated that the CATIP-AS1 promotes the progression and metastasis of THCA via EMT pathway partly through regulating the miR-515-5p and Smad4 expression in THCA cell. The CATIP-AS1 could be a promising biomarker for early THCA detection and prognosis and a possible therapeutic target for its treatment.
Collapse
Affiliation(s)
- Fujian Qi
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ji'Ao Tang
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhenling Cai
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Gang Wang
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhijun Wang
- Department of General Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
38
|
Quigley NG, Steiger K, Hoberück S, Czech N, Zierke MA, Kossatz S, Pretze M, Richter F, Weichert W, Pox C, Kotzerke J, Notni J. PET/CT imaging of head-and-neck and pancreatic cancer in humans by targeting the "Cancer Integrin" αvβ6 with Ga-68-Trivehexin. Eur J Nucl Med Mol Imaging 2022; 49:1136-1147. [PMID: 34559266 PMCID: PMC8460406 DOI: 10.1007/s00259-021-05559-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/05/2021] [Indexed: 01/26/2023]
Abstract
PURPOSE To develop a new probe for the αvβ6-integrin and assess its potential for PET imaging of carcinomas. METHODS Ga-68-Trivehexin was synthesized by trimerization of the optimized αvβ6-integrin selective cyclic nonapeptide Tyr2 (sequence: c[YRGDLAYp(NMe)K]) on the TRAP chelator core, followed by automated labeling with Ga-68. The tracer was characterized by ELISA for activities towards integrin subtypes αvβ6, αvβ8, αvβ3, and α5β1, as well as by cell binding assays on H2009 (αvβ6-positive) and MDA-MB-231 (αvβ6-negative) cells. SCID-mice bearing subcutaneous xenografts of the same cell lines were used for dynamic (90 min) and static (75 min p.i.) µPET imaging, as well as for biodistribution (90 min p.i.). Structure-activity-relationships were established by comparison with the predecessor compound Ga-68-TRAP(AvB6)3. Ga-68-Trivehexin was tested for in-human PET/CT imaging of HNSCC, parotideal adenocarcinoma, and metastatic PDAC. RESULTS Ga-68-Trivehexin showed a high αvβ6-integrin affinity (IC50 = 0.047 nM), selectivity over other subtypes (IC50-based factors: αvβ8, 131; αvβ3, 57; α5β1, 468), blockable uptake in H2009 cells, and negligible uptake in MDA-MB-231 cells. Biodistribution and preclinical PET imaging confirmed a high target-specific uptake in tumor and a low non-specific uptake in other organs and tissues except the excretory organs (kidneys and urinary bladder). Preclinical PET corresponded well to in-human results, showing high and persistent uptake in metastatic PDAC and HNSCC (SUVmax = 10-13) as well as in kidneys/urine. Ga-68-Trivehexin enabled PET/CT imaging of small PDAC metastases and showed high uptake in HNSCC but not in tumor-associated inflammation. CONCLUSIONS Ga-68-Trivehexin is a valuable probe for imaging of αvβ6-integrin expression in human cancers.
Collapse
Affiliation(s)
| | - Katja Steiger
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Sebastian Hoberück
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Norbert Czech
- Center of Nuclear Medicine and PET/CT Bremen, Bremen, Germany
| | | | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum rechts der Isar, and Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Marc Pretze
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Frauke Richter
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Christian Pox
- Clinic of Internal Medicine, Hospital St. Joseph-Stift Bremen, Bremen, Germany
| | - Jörg Kotzerke
- Klinik und Poliklinik für Nuklearmedizin, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany
| | - Johannes Notni
- Institute of Pathology, Technische Universität München, Munich, Germany.
- Experimental Radiopharmacy, Clinic for Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
39
|
Kpeglo D, Hughes MD, Dougan L, Haddrick M, Knowles MA, Evans SD, Peyman SA. Modeling the mechanical stiffness of pancreatic ductal adenocarcinoma. Matrix Biol Plus 2022; 14:100109. [PMID: 35399702 PMCID: PMC8990173 DOI: 10.1016/j.mbplus.2022.100109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 01/18/2023] Open
Abstract
The PDAC stroma stiffness underlines its malignant behavior and drug resistance. 3D in vitro cultures must model the PDAC stroma to effectively drug efficacy. PSCs are responsible for the stroma, and its activity is increased with TGF-β. Develop a 3D culture model of PDAC, which includes PSCs and TGF-β. Assess the mechanical stiffness, stain for collagen, and investigate gemcitabine efficacy.
Despite improvements in the understanding of disease biology, pancreatic ductal adenocarcinoma (PDAC) remains the most malignant cancer of the pancreas. PDAC constitutes ∼95% of all pancreatic cancers, and it is highly resistant to therapeutics. The increased tissue rigidity, which stems from the rich fibrotic stroma in the tumor microenvironment, is central to disease development, physiology, and resistance to drug perfusion. Pancreatic stellate cells (PSCs) are responsible for overproduction of extracellular matrix in the fibrotic stroma, and this is exacerbated by the overexpression of transforming growth factor-β (TGF-β). However, there are few in vitro PDAC models, which include both PSCs and TGF-β or mimic in vivo-like tumor stiffness. In this study, we present a three-dimensional in vitro PDAC model, which includes PSCs and TGF-β, and recapitulates PDAC tissue mechanical stiffness. Using oscillatory shear rheology, we show the mechanical stiffness of the model is within range of the PDAC tissue stiffness by day 21 of culture and highlight that the matrix environment is essential to adequately capture PDAC disease. PDAC is a complex, aggressive disease with poor prognosis, and biophysically relevant in vitro PDAC models, which take into account tissue mechanics, will provide improved tumor models for effective therapeutic assessment.
Collapse
Affiliation(s)
- Delanyo Kpeglo
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
| | - Matthew D.G. Hughes
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Lorna Dougan
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Block 35, Mereside Alderley Park, Alderley Edge, SK10 4TG, UK
| | - Margaret A. Knowles
- Leeds Institute of Medical Research at St James’s (LIMR), School of Medicine, University of Leeds, LS2 9 JT, UK
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, UK
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK
- Leeds Institute of Medical Research at St James’s (LIMR), School of Medicine, University of Leeds, LS2 9 JT, UK
- Corresponding author at: Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9 JT, UK.
| |
Collapse
|
40
|
Baba AB, Rah B, Bhat GR, Mushtaq I, Parveen S, Hassan R, Hameed Zargar M, Afroze D. Transforming Growth Factor-Beta (TGF-β) Signaling in Cancer-A Betrayal Within. Front Pharmacol 2022; 13:791272. [PMID: 35295334 PMCID: PMC8918694 DOI: 10.3389/fphar.2022.791272] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
A ubiquitously expressed cytokine, transforming growth factor-beta (TGF-β) plays a significant role in various ongoing cellular mechanisms. The gain or loss-of-function of TGF-β and its downstream mediators could lead to a plethora of diseases includes tumorigenesis. Specifically, at the early onset of malignancy TGF-β act as tumour suppressor and plays a key role in clearing malignant cells by reducing the cellular proliferation and differentiation thus triggers the process of apoptosis. Subsequently, TGF-β at an advanced stage of malignancy promotes tumorigenesis by augmenting cellular transformation, epithelial-mesenchymal-transition invasion, and metastasis. Besides playing the dual roles, depending upon the stage of malignancy, TGF-β also regulates cell fate through immune and stroma components. This oscillatory role of TGF-β to fight against cancer or act as a traitor to collaborate and crosstalk with other tumorigenic signaling pathways and its betrayal within the cell depends upon the cellular context. Therefore, the current review highlights and understands the dual role of TGF-β under different cellular conditions and its crosstalk with other signaling pathways in modulating cell fate.
Collapse
|
41
|
The Role of SMAD4 Inactivation in Epithelial-Mesenchymal Plasticity of Pancreatic Ductal Adenocarcinoma: The Missing Link? Cancers (Basel) 2022; 14:cancers14040973. [PMID: 35205719 PMCID: PMC8870198 DOI: 10.3390/cancers14040973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is currently one of the deadliest cancers. Despite the progress that has been made in the research of patient care and the understanding of pancreatic cancer, the survival rate remains mediocre. SMAD4, a tumor-suppressor gene, is specifically inactivated in 50–55% of pancreatic cancers. The role of SMAD4 protein loss in PDAC remains controversial, but seems to be associated with worse overall survival and metastasis. Here, we review the function of SMAD4 inactivation in the context of a specific biological process called epithelial–mesenchymal transition, as it has been increasingly associated with tumor formation, metastasis and resistance to therapy. By improving our understanding of these molecular mechanisms, we hope to find new targets for therapy and improve the care of patients with PDAC. Abstract Pancreatic ductal adenocarcinoma (PDAC) presents a five-year survival rate of 10% and its incidence increases over the years. It is, therefore, essential to improve our understanding of the molecular mechanisms that promote metastasis and chemoresistance in PDAC, which are the main causes of death in these patients. SMAD4 is inactivated in 50% of PDACs and its loss has been associated with worse overall survival and metastasis, although some controversy still exists. SMAD4 is the central signal transducer of the transforming growth factor-beta (TGF-beta) pathway, which is notably known to play a role in epithelial–mesenchymal transition (EMT). EMT is a biological process where epithelial cells lose their characteristics to acquire a spindle-cell phenotype and increased motility. EMT has been increasingly studied due to its potential implication in metastasis and therapy resistance. Recently, it has been suggested that cells undergo EMT transition through intermediary states, which is referred to as epithelial–mesenchymal plasticity (EMP). The intermediary states are characterized by enhanced aggressiveness and more efficient metastasis. Therefore, this review aims to summarize and analyze the current knowledge on SMAD4 loss in patients with PDAC and to investigate its potential role in EMP in order to better understand its function in PDAC carcinogenesis.
Collapse
|
42
|
Dai CJ, Cao YT, Huang F, Wang YG. Multiple roles of mothers against decapentaplegic homolog 4 in tumorigenesis, stem cells, drug resistance, and cancer therapy. World J Stem Cells 2022; 14:41-53. [PMID: 35126827 PMCID: PMC8788178 DOI: 10.4252/wjsc.v14.i1.41] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/13/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
The transforming growth factor (TGF)-β signaling pathway controls many cellular processes, including proliferation, differentiation, and apoptosis. Abnormalities in the TGF-β signaling pathway and its components are closely related to the occurrence of many human diseases, including cancer. Mothers against decapentaplegic homolog 4 (Smad4), also known as deleted in pancreatic cancer locus 4, is a typical tumor suppressor candidate gene locating at q21.1 of human chromosome 18 and the common mediator of the TGF-β/Smad and bone morphogenetic protein/Smad signaling pathways. It is believed that Smad4 inactivation correlates with the development of tumors and stem cell fate decisions. Smad4 also interacts with cytokines, miRNAs, and other signaling pathways, jointly regulating cell behavior. However, the regulatory function of Smad4 in tumorigenesis, stem cells, and drug resistance is currently controversial. In addition, Smad4 represents an attractive therapeutic target for cancer. Elucidating the specific role of Smad4 is important for understanding the mechanism of tumorigenesis and cancer treatment. Here, we review the identification and characterization of Smad4, the canonical TGF-β/Smad pathway, as well as the multiple roles of Smad4 in tumorigenesis, stem cells, and drug resistance. Furthermore, we provide novel insights into the prospects of Smad4-targeted cancer therapy and the challenges that it will face in the future.
Collapse
Affiliation(s)
- Chuan-Jing Dai
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Yu-Ting Cao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| | - Fang Huang
- Department of Pathology, Zhejiang Provincial People’s Hospital of Hangzhou Medical University, Hangzhou 310014, Zhejiang Province, China
| | - Yi-Gang Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang Province, China
| |
Collapse
|
43
|
Control of Expression of Key Cell Cycle Enzymes Drives Cell Line-Specific Functions of CDK7 in Human PDAC Cells. Int J Mol Sci 2022; 23:ijms23020812. [PMID: 35054996 PMCID: PMC8775745 DOI: 10.3390/ijms23020812] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 01/16/2023] Open
Abstract
Inhibition of the dual function cell cycle and transcription kinase CDK7 is known to affect the viability of cancer cells, but the mechanisms underlying cell line-specific growth control remain poorly understood. Here, we employed a previously developed, highly specific small molecule inhibitor that non-covalently blocks ATP binding to CDK7 (LDC4297) to study the mechanisms underlying cell line-specific growth using a panel of genetically heterogeneous human pancreatic tumor lines as model system. Although LDC4297 diminished both transcription rates and CDK T-loop phosphorylation in a comparable manner, some PDAC lines displayed significantly higher sensitivity than others. We focused our analyses on two well-responsive lines (Mia-Paca2 and Panc89) that, however, showed significant differences in their viability upon extended exposure to limiting LDC4297 concentrations. Biochemical and RNAseq analysis revealed striking differences in gene expression and cell cycle control. Especially the downregulation of a group of cell cycle control genes, among them CDK1/2 and CDC25A/C, correlated well to the observed viability differences in Panc89 versus Mia-Paca2 cells. A parallel downregulation of regulatory pathways supported the hypothesis of a feedforward programmatic effect of CDK7 inhibitors, eventually causing hypersensitivity of PDAC lines.
Collapse
|
44
|
Glutamine-Derived Aspartate Biosynthesis in Cancer Cells: Role of Mitochondrial Transporters and New Therapeutic Perspectives. Cancers (Basel) 2022; 14:cancers14010245. [PMID: 35008407 PMCID: PMC8750728 DOI: 10.3390/cancers14010245] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary In recent years, aspartate has been increasingly acknowledged as a critical player in the metabolism of cancer cells which use this metabolite for nucleotide and protein synthesis and for redox homeostasis. Most intracellular aspartate derives from the mitochondrial catabolism of glutamine. To date at least four mitochondrial transporters have been involved in this metabolic pathway. Their involvement appears to be cancer type-specific and dependent on glutamine availability. Targeting these mitochondrial transporters may represent a new attractive strategy to fight cancer. The aim of this review is to dissect the role of each of these transporters in relation to the type of cancer and the availability of nutrients in the tumoral microenvironment. Abstract Aspartate has a central role in cancer cell metabolism. Aspartate cytosolic availability is crucial for protein and nucleotide biosynthesis as well as for redox homeostasis. Since tumor cells display poor aspartate uptake from the external environment, most of the cellular pool of aspartate derives from mitochondrial catabolism of glutamine. At least four transporters are involved in this metabolic pathway: the glutamine (SLC1A5_var), the aspartate/glutamate (AGC), the aspartate/phosphate (uncoupling protein 2, UCP2), and the glutamate (GC) carriers, the last three belonging to the mitochondrial carrier family (MCF). The loss of one of these transporters causes a paucity of cytosolic aspartate and an arrest of cell proliferation in many different cancer types. The aim of this review is to clarify why different cancers have varying dependencies on metabolite transporters to support cytosolic glutamine-derived aspartate availability. Dissecting the precise metabolic routes that glutamine undergoes in specific tumor types is of upmost importance as it promises to unveil the best metabolic target for therapeutic intervention.
Collapse
|
45
|
Wardana T, Aida A, Zhafirah R, Hirawan H, Haris Budi Widodo A, Prihastuti C. Wound healing potential of forest honey for increasing TGF-β1 protein expression in palatoplasty: In-vivo and In-silico studies. SCIENTIFIC DENTAL JOURNAL 2022. [DOI: 10.4103/sdj.sdj_95_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
46
|
van Roey R, Brabletz T, Stemmler MP, Armstark I. Deregulation of Transcription Factor Networks Driving Cell Plasticity and Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:753456. [PMID: 34888306 PMCID: PMC8650502 DOI: 10.3389/fcell.2021.753456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a very aggressive disease with 5-year survival rates of less than 10%. The constantly increasing incidence and stagnant patient outcomes despite changes in treatment regimens emphasize the requirement of a better understanding of the disease mechanisms. Challenges in treating pancreatic cancer include diagnosis at already progressed disease states due to the lack of early detection methods, rapid acquisition of therapy resistance, and high metastatic competence. Pancreatic ductal adenocarcinoma, the most prevalent type of pancreatic cancer, frequently shows dominant-active mutations in KRAS and TP53 as well as inactivation of genes involved in differentiation and cell-cycle regulation (e.g. SMAD4 and CDKN2A). Besides somatic mutations, deregulated transcription factor activities strongly contribute to disease progression. Specifically, transcriptional regulatory networks essential for proper lineage specification and differentiation during pancreas development are reactivated or become deregulated in the context of cancer and exacerbate progression towards an aggressive phenotype. This review summarizes the recent literature on transcription factor networks and epigenetic gene regulation that play a crucial role during tumorigenesis.
Collapse
Affiliation(s)
- Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
47
|
Mulqueen RM, Pokholok D, O’Connell BL, Thornton CA, Zhang F, O’Roak BJ, Link J, Yardımcı GG, Sears RC, Steemers FJ, Adey AC. High-content single-cell combinatorial indexing. Nat Biotechnol 2021; 39:1574-1580. [PMID: 34226710 PMCID: PMC8678206 DOI: 10.1038/s41587-021-00962-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
Single-cell combinatorial indexing (sci) with transposase-based library construction increases the throughput of single-cell genomics assays but produces sparse coverage in terms of usable reads per cell. We develop symmetrical strand sci ('s3'), a uracil-based adapter switching approach that improves the rate of conversion of source DNA into viable sequencing library fragments following tagmentation. We apply this chemistry to assay chromatin accessibility (s3-assay for transposase-accessible chromatin, s3-ATAC) in human cortical and mouse whole-brain tissues, with mouse datasets demonstrating a six- to 13-fold improvement in usable reads per cell compared with other available methods. Application of s3 to single-cell whole-genome sequencing (s3-WGS) and to whole-genome plus chromatin conformation (s3-GCC) yields 148- and 14.8-fold improvements, respectively, in usable reads per cell compared with sci-DNA-sequencing and sci-HiC. We show that s3-WGS and s3-GCC resolve subclonal genomic alterations in patient-derived pancreatic cancer cell lines. We expect that the s3 platform will be compatible with other transposase-based techniques, including sci-MET or CUT&Tag.
Collapse
Affiliation(s)
- Ryan M. Mulqueen
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR
| | | | - Brendan L. O’Connell
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR
| | - Casey A. Thornton
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR
| | | | - Brian J. O’Roak
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR
| | - Jason Link
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR,Oregon Health & Science University, Knight Cancer Institute, Portland, OR,Oregon Health & Science University, Brendan Colson Center for Pancreatic Care, Portland, OR
| | - Galip Gürkan Yardımcı
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR,Oregon Health & Science University, Cancer Early Detection Advanced Research Center, Portland, OR
| | - Rosalie C. Sears
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR,Oregon Health & Science University, Knight Cancer Institute, Portland, OR,Oregon Health & Science University, Brendan Colson Center for Pancreatic Care, Portland, OR,Oregon Health & Science University, Cancer Early Detection Advanced Research Center, Portland, OR
| | | | - Andrew C. Adey
- Oregon Health & Science University, Department of Molecular and Medical Genetics, Portland, OR,Oregon Health & Science University, Knight Cancer Institute, Portland, OR,Oregon Health & Science University, Cancer Early Detection Advanced Research Center, Portland, OR,Oregon Health & Science University, Department of Oncological Sciences, Portland, OR,Oregon Health & Science University, Knight Cardiovascular Institute, Portland, OR,Correspondence to
| |
Collapse
|
48
|
Kossatz S, Beer AJ, Notni J. It's Time to Shift the Paradigm: Translation and Clinical Application of Non-αvβ3 Integrin Targeting Radiopharmaceuticals. Cancers (Basel) 2021; 13:cancers13235958. [PMID: 34885066 PMCID: PMC8657165 DOI: 10.3390/cancers13235958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cancer cells often present a different set of proteins on their surface than normal cells. This also applies to integrins, a class of 24 cell surface receptors which mainly are responsible for physically anchoring cells in tissues, but also fulfil a plethora of other functions. If a certain integrin is found on tumor cells but not on normal ones, radioactive molecules (named tracers) that specifically bind to this integrin will accumulate in the cancer lesion if injected into the blood stream. The emitted radiation can be detected from outside the body and allows for localization and thus, diagnosis, of cancer. Only one of the 24 integrins, the subtype αvβ3, has hitherto been thoroughly investigated in this context. We herein summarize the most recent, pertinent research on other integrins, and argue that some of these approaches might ultimately improve the clinical management of the most lethal cancers, such as pancreatic carcinoma. Abstract For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, angiogenesis. Despite a large number of translated tracers and clinical studies, a clinical value of αvβ3-integrin imaging could not be defined yet. The focus of research has, thus, been moving slowly but steadily towards other integrin subtypes which are involved in a large variety of tumorigenic pathways. Peptidic and non-peptidic radioligands for the integrins α5β1, αvβ6, αvβ8, α6β1, α6β4, α3β1, α4β1, and αMβ2 were first synthesized and characterized preclinically. Some of these compounds, targeting the subtypes αvβ6, αvβ8, and α6β1/β4, were subsequently translated into humans during the last few years. αvβ6-Integrin has arguably attracted most attention because it is expressed by some of the cancers with the worst prognosis (above all, pancreatic ductal adenocarcinoma), which substantiates a clinical need for the respective theranostic agents. The receptor furthermore represents a biomarker for malignancy and invasiveness of carcinomas, as well as for fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), and probably even for Sars-CoV-2 (COVID-19) related syndromes. Accordingly, the largest number of recent first-in-human applications has been reported for radiolabeled compounds targeting αvβ6-integrin. The results indicate a substantial clinical value, which might lead to a paradigm change and trigger the replacement of αvβ3 by αvβ6 as the most popular integrin in theranostics.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | | | - Johannes Notni
- Department of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- TRIMT GmbH, 01454 Radeberg, Germany
- Correspondence: ; Tel.: +49-89-4140-6075; Fax: +49-89-4140-6949
| |
Collapse
|
49
|
De Summa S, Danza K, Pilato B, Matera G, Fasano R, Calabrese A, Lacalamita R, Silvestris N, Tommasi S, Argentiero A, Brunetti O. A Promising Role of TGF-β Pathway in Response to Regorafenib in Metastatic Colorectal Cancer: A Case Report. Medicina (B Aires) 2021; 57:medicina57111241. [PMID: 34833459 PMCID: PMC8619854 DOI: 10.3390/medicina57111241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancer types around the world. The prognosis of patients with advanced diseases is still poor in spite of currently available therapeutic options. Regorafenib is an oral tyrosine kinase inhibitor (TKI) approved to treat refractory metastatic colorectal cancer (mCRC). We investigated Somatic mutations in several genes involved in immunological response and cancer progression in both long/short responder mCRC patients who underwent third-line therapy with regorafenib to identify predictive biomarkers of response using Ion Torrent PGM sequencing and bioinformatic tools. We found Somatic mutations in TGFBR1, TGFBR2, and TGFBR3 genes in primary tumor and metastases samples of long-responder patients. Furthermore, our bioinformatic results show that they were mainly enriched in immune response, cell junction, and cell adhesion in long responder patients, particularly in primary tumor and metastatic sites. These data suggest that the TGF-b pattern could be the leading actor of a prolonged response to this drug.
Collapse
Affiliation(s)
- Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Katia Danza
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Brunella Pilato
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Giuseppina Matera
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Rossella Fasano
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
| | - Angela Calabrese
- Radiology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Rosanna Lacalamita
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Antonella Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
- Correspondence:
| |
Collapse
|
50
|
Miquel M, Zhang S, Pilarsky C. Pre-clinical Models of Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:748631. [PMID: 34778259 PMCID: PMC8578999 DOI: 10.3389/fcell.2021.748631] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a hostile solid malignancy coupled with an extremely high mortality rate. Metastatic disease is already found in most patients at the time of diagnosis, resulting in a 5-year survival rate below 5%. Improved comprehension of the mechanisms leading to metastasis is pivotal for the development of new targeted therapies. A key field to be improved are modeling strategies applied in assessing cancer progression, since traditional platforms fail in recapitulating the complexity of PDAC. Consequently, there is a compelling demand for new preclinical models that mirror tumor progression incorporating the pressure of the immune system, tumor microenvironment, as well as molecular aspects of PDAC. We suggest the incorporation of 3D organoids derived from genetically engineered mouse models or patients as promising new tools capable to transform PDAC pre-clinical modeling and access new frontiers in personalized medicine.
Collapse
Affiliation(s)
- Maria Miquel
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Shuman Zhang
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|