1
|
Wen YR, Lin XW, Zhou YW, Xu L, Zhang JL, Chen CY, He J. N-glycan biosignatures as a potential diagnostic biomarker for early-stage pancreatic cancer. World J Gastrointest Oncol 2024; 16:659-669. [PMID: 38577461 PMCID: PMC10989390 DOI: 10.4251/wjgo.v16.i3.659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a 5-year survival rate of less than 10%, owing to its late-stage diagnosis. Early detection of pancreatic cancer (PC) can significantly increase survival rates. AIM To identify the serum biomarker signatures associated with early-stage PDAC by serum N-glycan analysis. METHODS An extensive patient cohort was used to determine a biomarker signature, including patients with PDAC that was well-defined at an early stage (stages I and II). The biomarker signature was derived from a case-control study using a case-cohort design consisting of 29 patients with stage I, 22 with stage II, 4 with stage III, 16 with stage IV PDAC, and 88 controls. We used multiparametric analysis to identify early-stage PDAC N-glycan signatures and developed an N-glycan signature-based diagnosis model called the "Glyco-model". RESULTS The biomarker signature was created to discriminate samples derived from patients with PC from those of controls, with a receiver operating characteristic area under the curve of 0.86. In addition, the biomarker signature combined with cancer antigen 19-9 could discriminate patients with PDAC from controls, with a receiver operating characteristic area under the curve of 0.919. Glyco-model demonstrated favorable diagnostic performance in all stages of PC. The diagnostic sensitivity for stage I PDAC was 89.66%. CONCLUSION In a prospective validation study, this serum biomarker signature may offer a viable method for detecting early-stage PDAC.
Collapse
Affiliation(s)
- Yan-Rong Wen
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Xia-Wen Lin
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yu-Wen Zhou
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Lei Xu
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Jun-Li Zhang
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Cui-Ying Chen
- Department of Research and Development, Sysdiagno (Nanjing) Biotech Co., Ltd, Nanjing 210008, Jiangsu Province, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
2
|
Budka J, Debowski D, Mai S, Narajczyk M, Hac S, Rolka K, Vrettos EI, Tzakos AG, Inkielewicz-Stepniak I. Design, Synthesis, and Antitumor Evaluation of an Opioid Growth Factor Bioconjugate Targeting Pancreatic Ductal Adenocarcinoma. Pharmaceutics 2024; 16:283. [PMID: 38399336 PMCID: PMC10892429 DOI: 10.3390/pharmaceutics16020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents a formidable challenge with high lethality and limited effective drug treatments. Its heightened metastatic potential further complicates the prognosis. Owing to the significant toxicity of current chemotherapeutics, compounds like [Met5]-enkephalin, known as opioid growth factor (OGF), have emerged in oncology clinical trials. OGF, an endogenous peptide interacting with the OGF receptor (OGFr), plays a crucial role in inhibiting cell proliferation across various cancer types. This in vitro study explores the potential anticancer efficacy of a newly synthesized OGF bioconjugate in synergy with the classic chemotherapeutic agent, gemcitabine (OGF-Gem). The study delves into assessing the impact of the OGF-Gem conjugate on cell proliferation inhibition, cell cycle regulation, the induction of cellular senescence, and apoptosis. Furthermore, the antimetastatic potential of the OGF-Gem conjugate was demonstrated through evaluations using blood platelets and AsPC-1 cells with a light aggregometer. In summary, this article demonstrates the cytotoxic impact of the innovative OGF-Gem conjugate on pancreatic cancer cells in both 2D and 3D models. We highlight the potential of both the OGF-Gem conjugate and OGF alone in effectively inhibiting the ex vivo pancreatic tumor cell-induced platelet aggregation (TCIPA) process, a phenomenon not observed with Gem alone. Furthermore, the confirmed hemocompatibility of OGF-Gem with platelets reinforces its promising potential. We anticipate that this conjugation strategy will open avenues for the development of potent anticancer agents.
Collapse
Affiliation(s)
- Justyna Budka
- Department of Pharmaceutical Pathophysiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Dawid Debowski
- Department of Molecular Biochemistry, University of Gdansk, 80-309 Gdansk, Poland
| | - Shaoshan Mai
- Department of Pharmaceutical Pathophysiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Magdalena Narajczyk
- Bioimaging Laboratory, Faculty of Biology, University of Gdansk, 80-309 Gdansk, Poland
| | - Stanislaw Hac
- Department of General Endocrine and Transplant Surgery, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Krzysztof Rolka
- Department of Molecular Biochemistry, University of Gdansk, 80-309 Gdansk, Poland
| | | | - Andreas G. Tzakos
- Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece
- University Research Center of Ioannina, Institute of Materials Science and Computing, 45110 Ioannina, Greece
| | | |
Collapse
|
3
|
Zhao R, Hu Z, Zhang X, Huang S, Yu G, Wu Z, Yu W, Lu J, Ruan B. The oncogenic mechanisms of the Janus kinase-signal transducer and activator of transcription pathway in digestive tract tumors. Cell Commun Signal 2024; 22:68. [PMID: 38273295 PMCID: PMC10809652 DOI: 10.1186/s12964-023-01421-9] [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: 09/06/2023] [Accepted: 12/03/2023] [Indexed: 01/27/2024] Open
Abstract
Digestive tract tumors are heterogeneous and involve the dysregulation of multiple signaling pathways. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway plays a notable role in the oncogenesis of digestive tract tumors. Typically activated by pro-inflammatory cytokines, it regulates important biological processes, such as cell growth, differentiation, apoptosis, immune responses, and inflammation. The aberrant activation of this pathway manifests in different forms, including mutations in JAKs, overexpression of cytokine receptors, and sustained STAT activation, and contributes to promoting the malignant characteristics of cancer cells, including uncontrolled proliferation, resistance to apoptosis, enhanced invasion and metastasis, angiogenesis, acquisition of stem-like properties, and drug resistance. Numerous studies have shown that aberrant activation of the JAK-STAT pathway is closely related to the development and progression of digestive tract tumors, contributing to tumor survival, angiogenesis, changes in the tumor microenvironment, and even immune escape processes. In addition, this signaling pathway also affects the sensitivity of digestive tract tumors to chemotherapy and targeted therapy. Therefore, it is crucial to comprehensively understand the oncogenic mechanisms underlying the JAK-STAT pathway in order to develop effective therapeutic strategies against digestive tract tumors. Currently, several JAK-STAT inhibitors are undergoing clinical and preclinical trials as potential treatments for various human diseases. However, further investigation is required to determine the role of this pathway, as well as the effectiveness and safety of its inhibitors, especially in the context of digestive tract tumors. In this review, we provide an overview of the structure, classic activation, and negative regulation of the JAK-STAT pathway. Furthermore, we discuss the pathogenic mechanisms of JAK-STAT signaling in different digestive tract tumors, with the aim of identifying potential novel therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Ruihong Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhangmin Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Xiaoli Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Shujuan Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Guodong Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Zhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| | - Bing Ruan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, National Medical Center for Infectious Diseases, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
4
|
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
|
5
|
Xuan SH, Hua ML, Xiang Z, He XL, Huang L, Jiang C, Dong P, Wu J. Roles of cancer stem cells in gastrointestinal cancers. World J Stem Cells 2023; 15:209-220. [PMID: 37181004 PMCID: PMC10173810 DOI: 10.4252/wjsc.v15.i4.209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/25/2023] [Accepted: 03/27/2023] [Indexed: 04/26/2023] Open
Abstract
Cancer stem cells (CSCs) are the main cause of tumor growth, invasion, metastasis and recurrence. Recently, CSCs have been extensively studied to identify CSC-specific surface markers as well as signaling pathways that play key roles in CSCs self-renewal. The involvement of CSCs in the pathogenesis of gastrointestinal (GI) cancers also highlights these cells as a priority target for therapy. The diagnosis, prognosis and treatment of GI cancer have always been a focus of attention. Therefore, the potential application of CSCs in GI cancers is receiving increasing attention. This review summarizes the role of CSCs in GI cancers, focusing on esophageal cancer, gastric cancer, liver cancer, colorectal cancer, and pancreatic cancer. In addition, we propose CSCs as potential targets and therapeutic strategies for the effective treatment of GI cancers, which may provide better guidance for clinical treatment of GI cancers.
Collapse
Affiliation(s)
- Shi-Hai Xuan
- Department of Laboratory Medicine, The People’s Hospital of Dongtai City, Dongtai 224299, Jiangsu Province, China
| | - Meng-Lu Hua
- School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Ze Xiang
- School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Xiang-Lin He
- School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Lan Huang
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| | - Chun Jiang
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| | - Peng Dong
- Hangzhou Institute of Cardiovascular Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Jian Wu
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| |
Collapse
|
6
|
Effects of chloroquine and hydroxychloroquine on the sensitivity of pancreatic cancer cells to targeted therapies. Adv Biol Regul 2023; 87:100917. [PMID: 36243652 DOI: 10.1016/j.jbior.2022.100917] [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: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 11/20/2022]
Abstract
Approaches to improve pancreatic cancer therapy are essential as this disease has a very bleak outcome. Approximately 80% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). PDAC is a cancer which is difficult to effectively treat as it is often detected late in the disease process. Almost all PDACs (over 90%) have activating mutations in the GTPase gene KRAS. These mutations result in constitutive KRas activation and the mobilization of downstream pathways such as the Raf/MEK/ERK pathway. Small molecule inhibitors of key components of the KRas/Raf/MEK/ERK pathways as well as monoclonal antibodies (MoAbs) specific for upstream growth factor receptors such insulin like growth factor-1 receptor (IGF1-R) and epidermal growth factor receptors (EGFRs) have been developed and have been evaluated in clinical trials. An additional key regulatory gene frequently mutated (∼75%) in PDAC is the TP53 tumor suppressor gene which controls the transcription of multiple genes involved in cell cycle progression, apoptosis, metabolism, cancer progression and other growth regulatory processes. Small molecule mutant TP53 reactivators have been developed which alter the structure of mutant TP53 protein and restore some of its antiproliferative activities. Some mutant TP53 reactivators have been examined in clinical trials with patients with mutant TP53 genes. Inhibitors to the TP53 negative regulator Mouse Double Minute 2 (MDM2) have been developed and analyzed in clinical trials. Chloroquine and hydroxychloroquine are established anti-malarial and anti-inflammatory drugs that also prevent the induction of autophagy which can have effects on cancer survival. Chloroquine and hydroxychloroquine have also been examined in various clinical trials. Recent studies are suggesting effective treatment of PDAC patients may require chemotherapy as well as targeting multiple pathways and biochemical processes.
Collapse
|
7
|
Yu D, Zhao Z, Wang L, Qiao S, Yang Z, Wen Q, Zhu G. SOX21-AS1 activated by STAT6 promotes pancreatic cancer progression via up-regulation of SOX21. J Transl Med 2022; 20:511. [PMID: 36335356 PMCID: PMC9636668 DOI: 10.1186/s12967-022-03521-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/08/2022] [Indexed: 11/06/2022] Open
Abstract
Background Pancreatic cancer (PC) is a highly malignant tumor which threatens human’s health. Long non-coding RNAs (lncRNAs) are implicated in many cancers, including PC, but their mechanisms in PC have not yet been entirely clarified. We focused on revealing the potential function of lncRNA SOX21-AS1 in PC. Methods Functional assays assessed SOX21-AS1 function on PC progression. Bioinformatics analysis, along with mechanism assays were taken to unmask the regulatory mechanism SOX21-AS1 may exert in PC cells. Results SOX21-AS1 possessed a high expression level in PC cells. SOX21-AS1 absence suppressed PC cell proliferation, migration, stemness and epithelial-mesenchymal transition (EMT) while elevated cell apoptosis. SOX21-AS1 positively regulated its nearby gene SRY-box transcription factor 21 (SOX21) at post-transcriptional level. Through mechanism assays, we uncovered that SOX21-AS1 sponged SOX21-AS1 to elevate SOX21 mRNA and recruited ubiquitin-specific peptidase 10 (USP10) to deubiquitinate and stabilize SOX21 protein. Moreover, signal transducer and activator of transcription 6 (STAT6) could transcriptionally activate SOX21-AS1 and SOX21 expression. Conclusions SOX21-AS1 aggravated the malignant development of PC, which might provide the utility value for PC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03521-5.
Collapse
|
8
|
Tarannum M, Vivero-Escoto JL. Nanoparticle-based therapeutic strategies targeting major clinical challenges in pancreatic cancer treatment. Adv Drug Deliv Rev 2022; 187:114357. [PMID: 35605679 DOI: 10.1016/j.addr.2022.114357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Collapse
|
9
|
Sally Á, McGowan R, Finn K, Moran BM. Current and Future Therapies for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14102417. [PMID: 35626020 PMCID: PMC9139531 DOI: 10.3390/cancers14102417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Pancreatic cancer is the fourth leading cause of cancer-related mortality worldwide. The poor survival associated with this disease is due to delayed diagnosis, a lack of reliable biomarkers, and tumour resistance to treatment. Currently, surgery is the only curative treatment option, but few patients are eligible for this procedure. Developing resistance to current chemotherapies such as gemcitabine has led to a reduction in effective therapy options for patients and an urgent requirement for the development of novel therapeutic avenues. Potential success has been noted in therapeutic approaches such as synthetic lethality and immunotherapy. An array of clinical trials are currently recruiting, primarily in the area of monoclonal antibodies in combination with other therapies such as chemotherapy and immune checkpoint inhibitors. This review article aims to highlight the potential these therapies have to improve patient prognosis and survival. Abstract Pancreatic cancer is one of the leading causes of cancer-related death worldwide. This is due to delayed diagnosis and resistance to traditional chemotherapy. Delayed diagnosis is often due to the broad range of non-specific symptoms that are associated with the disease. Resistance to current chemotherapies, such as gemcitabine, develops due to genetic mutations that are either intrinsic or acquired. This has resulted in poor patient prognosis and, therefore, justifies the requirement for new targeted therapies. A synthetic lethality approach, that targets specific loss-of-function mutations in cancer cells, has shown great potential in pancreatic ductal adenocarcinoma (PDAC). Immunotherapies have also yielded promising results in the development of new treatment options, with several currently undergoing clinical trials. The utilisation of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell transfer, and vaccines have shown success in several neoplasms such as breast cancer and B-cell malignancies and, therefore, could hold the same potential in PDAC treatment. These therapeutic strategies could have the potential to be at the forefront of pancreatic cancer therapy in the future. This review focuses on currently approved therapies for PDAC, the challenges associated with them, and future directions of therapy including synthetically lethal approaches, immunotherapy, and current clinical trials.
Collapse
Affiliation(s)
- Áine Sally
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Ryan McGowan
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Department of Life Sciences, School of Science, Atlantic Technological University Sligo, Ash Lane, Ballytivnan, F91 YW50 Sligo, Ireland
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Brian Michael Moran
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Correspondence:
| |
Collapse
|
10
|
McCubrey JA, Abrams SL, Steelman LS, Cocco L, Ratti S, Martelli AM, Lombardi P, Gizak A, Duda P. APR-246-The Mutant TP53 Reactivator-Increases the Effectiveness of Berberine and Modified Berberines to Inhibit the Proliferation of Pancreatic Cancer Cells. Biomolecules 2022; 12:276. [PMID: 35204775 PMCID: PMC8961609 DOI: 10.3390/biom12020276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/10/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer. In ~75% of PDAC, the tumor suppressor TP53 gene is mutated. Novel approaches to treat cancer involve compounds called mutant TP53 reactivators. They interact with mutant TP53 proteins and restore some of their growth suppressive properties, but they may also interact with other proteins, e.g., TP63 and TP73. We examined the ability of the TP53 reactivator APR-246 to interact with eleven modified berberine compounds (NAX compounds) in the presence and absence of WT-TP53 in two PDAC cell lines: the MIA-PaCa-2, which has gain of function (GOF) TP53 mutations on both alleles, and PANC-28, which lacks expression of the WT TP53 protein. Our results indicate the TP53 reactivator-induced increase in therapeutic potential of many modified berberines.
Collapse
Affiliation(s)
- James Andrew McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Stephen L. Abrams
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Linda S. Steelman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; (S.L.A.); (L.S.S.)
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, 40126 Bologna, Italy; (L.C.); (S.R.); (A.M.M.)
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, 20026 Novate Milanese, Italy;
| | - Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, University of Wrocław, 50-335 Wroclaw, Poland; (A.G.); (P.D.)
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, 50-335 Wroclaw, Poland; (A.G.); (P.D.)
| |
Collapse
|
11
|
Sesquiterpene lactones isolated from Carpesium abrotanoides L. by LC–MS combined with HSCCC inhibit liver cancer through suppression of the JAK2/STAT3 signaling pathway. Med Chem Res 2022. [DOI: 10.1007/s00044-021-02838-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Tang H, Xue Y, Li B, Xu X, Zhang F, Guo J, Li Q, Yuan T, Chen Y, Pan Y, Ping Y, Li D. Membrane-camouflaged supramolecular nanoparticles for co-delivery of chemotherapeutic and molecular-targeted drugs with siRNA against patient-derived pancreatic carcinoma. Acta Pharm Sin B 2022; 12:3410-3426. [PMID: 35967289 PMCID: PMC9366227 DOI: 10.1016/j.apsb.2022.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer remains one of the most lethal malignancies worldwide. The combination of the first-line standard agent gemcitabine (GEM) with the molecular-targeted drug erlotinib (Er) has emerged as a promising strategy for pancreatic cancer treatment. However, the clinical benefit from this combination is still far from satisfactory due to the unfavorable drug antagonism and the fibrotic tumor microenvironment. Herein, we propose a membrane-camouflaged dual stimuli-responsive delivery system for the co-delivery of GEM and Er into pancreatic cancer cells and tissues to block the antagonism, as well as reshapes profibrotic tumor microenvironment via simultaneous delivery of small interference RNA (siRNA) for synergistic pancreatic cancer treatment. This “all-in-one” delivery system exhibits sensitive GSH and pH-dependent drug release profiles and enhances the inhibitory effects on the proliferation and migration of tumor cells in vitro. Excitingly, the systemic injection of such a biomimetic drug co-delivery system not only resulted in superior inhibitory effects against orthotopic pancreatic tumor and patient-derived tumor (PDX), but also greatly extended the survival rate of tumor-bearing mice. Our findings provide a promising therapeutic strategy against pancreatic cancer through the enhanced synergistic effect of target therapy, chemotherapy and anti-fibrotic therapy, which represents an appealing way for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Honglin Tang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanan Xue
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bowen Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Corresponding authors.
| | - Xiaojie Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fu Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Jiajing Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qijun Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tingting Yuan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubin Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Corresponding authors.
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Corresponding authors.
| |
Collapse
|
13
|
Tarannum M, Hossain MA, Holmes B, Yan S, Mukherjee P, Vivero-Escoto JL. Advanced Nanoengineering Approach for Target-Specific, Spatiotemporal, and Ratiometric Delivery of Gemcitabine-Cisplatin Combination for Improved Therapeutic Outcome in Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104449. [PMID: 34758094 PMCID: PMC8758547 DOI: 10.1002/smll.202104449] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/03/2021] [Indexed: 05/13/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an intractable malignancy with a dismal survival rate. Recent combination therapies have had a major impact on the improvement of PDAC prognosis. Nevertheless, clinically used combination regimens such as FOLFIRINOX and gemcitabine (Gem)/nab-paclitaxel still face major challenges due to lack of the safe and ratiometric delivery of multiple drugs. Here, a rationally designed mesoporous silica nanoparticle (MSN)-based platform is reported for the target-specific, spatiotemporal, ratiometric, and safe co-delivery of Gem and cisplatin (cisPt). It is shown that systemic administration of the nanoparticles results in synergistic therapeutic outcome in a syngeneic and clinically relevant genetically engineered PDAC mouse model that has rarely been used for the therapeutic evaluation of nanomedicine. This synergism is associated with a strategic engineering approach, in which nanoparticles provide redox-responsive controlled delivery and in situ differential release of Gem/cisPt drugs with the goal of overcoming resistance to Pt-based drugs. The platform is also rendered with additional tumor-specificity via a novel tumor-associated mucin1 (tMUC1)-specific antibody, TAB004. Overall, the platform suppresses tumor growth and eliminates the off-target toxicities of a highly toxic chemotherapy combination.
Collapse
Affiliation(s)
- Mubin Tarannum
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Nanoscale Science Program, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Md Akram Hossain
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Bryce Holmes
- Analytical Research Laboratory, North Carolina A&T State University, Greensboro, NC, 27411, USA
| | - Shan Yan
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Pinku Mukherjee
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Juan L Vivero-Escoto
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
- Center for Biomedical Engineering and Science, The University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| |
Collapse
|
14
|
Abrams SL, Akula SM, Steelman LS, Follo ML, Cocco L, Ratti S, Martelli AM, Libra M, Falzone L, Candido S, Montalto G, Cervello M, Lombardi P, McCubrey JA. Effects of the MDM2 inhibitor Nutlin-3a on sensitivity of pancreatic cancer cells to berberine and modified berberines in the presence and absence of WT-TP53. Adv Biol Regul 2021; 83:100840. [PMID: 34866036 DOI: 10.1016/j.jbior.2021.100840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
Approaches to improve pancreatic cancer therapy are essential as this disease has a very bleak outcome. Approximately 80% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). A key regulatory gene frequently mutated (∼75%) in PDAC is the TP53 tumor suppressor gene which controls the transcription of multiple genes involved in cell cycle progression, apoptosis, cancer progression and other growth regulatory processes. The mouse double minute 2 homolog (MDM2) gene product is a nuclear-localized E3 ubiquitin ligase and negatively regulates the TP53 protein which results in its proteasomal degradation. Various MDM2 inhibitors have been isolated and examined in clinical trials, especially in patients with hematological malignancies. Nutlin-3a is one of the first MDM2 inhibitors isolated. Berberine (BBR) is a natural product found in many fruits and berries and used in traditional medicine for centuries. It has many biological effects, and some are anti-proliferative in nature. BBR may activate the expression of TP53 and inhibit cell cycle progression as well as other events important in cell growth. To understand more about the potential of compounds like BBR and chemical modified BBRs (NAX compounds) to sensitize PDAC cells to MDM2 inhibitors, we introduced either WT-TP53 or the pLXSN empty vector control into two PDAC cell lines, one lacking expression of TP53 (PANC-28) and one with gain-of-function mutant TP53 on both alleles (MIA-PaCa-2). Our results indicate that nutlin-3a was able to increase the sensitivity to BBR and certain NAX compounds. The effects of nutlin-3a were usually more substantial in those cells containing an introduced WT TP53 gene. These results highlight the importance of knowledge of the type of TP53 mutation that is present in cancer patients before the administration of drugs which function by stabilization of the TP53 protein.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Shaw M Akula
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA
| | - Matilde L Follo
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Massimo Libra
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Luca Falzone
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Saverio Candido
- Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Department of Health Promotion, Maternal and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe di Vittorio 70, Novate Milanese, 20026, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
15
|
Safe S, Shrestha R, Mohankumar K, Howard M, Hedrick E, Abdelrahim M. Transcription factors specificity protein and nuclear receptor 4A1 in pancreatic cancer. World J Gastroenterol 2021; 27:6387-6398. [PMID: 34720529 PMCID: PMC8517783 DOI: 10.3748/wjg.v27.i38.6387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/30/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Specificity protein (Sp) transcription factors (TFs) Sp1, Sp3 and Sp4, and the orphan nuclear receptor 4A1 (NR4A1) are highly expressed in pancreatic tumors and Sp1 is a negative prognostic factor for pancreatic cancer patient survival. Results of knockdown and overexpression of Sp1, Sp3 and Sp4 in pancreatic and other cancer lines show that these TFs are individually pro-oncogenic factors and loss of one Sp TF is not compensated by other members. NR4A1 is also a pro-oncogenic factor and both NR4A1 and Sp TFs exhibit similar functions in pancreatic cancer cells and regulate cell growth, survival, migration and invasion. There is also evidence that Sp TFs and NR4A1 regulate some of the same genes including survivin, epidermal growth factor receptor, PAX3-FOXO1, α5- and α6-integrins, β1-, β3- and β4-integrins; this is due to NR4A1 acting as a cofactor and mediating NR4A1/Sp1/4-regulated gene expression through GC-rich gene promoter sites. Several studies show that drugs targeting Sp downregulation or NR4A1 antagonists are highly effective inhibitors of Sp/NR4A1-regulated pathways and genes in pancreatic and other cancer cells, and the triterpenoid celastrol is a novel dual-acting agent that targets both Sp TFs and NR4A1.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77845, United States
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77845, United States
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77845, United States
| | - Marcell Howard
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77845, United States
| | - Erik Hedrick
- Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Maen Abdelrahim
- Department of Medical Oncology, Houston Methodist Hospital Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
16
|
Arias-Pinilla GA, Modjtahedi H. Therapeutic Application of Monoclonal Antibodies in Pancreatic Cancer: Advances, Challenges and Future Opportunities. Cancers (Basel) 2021; 13:1781. [PMID: 33917882 PMCID: PMC8068268 DOI: 10.3390/cancers13081781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer remains as one of the most aggressive cancer types. In the absence of reliable biomarkers for its early detection and more effective therapeutic interventions, pancreatic cancer is projected to become the second leading cause of cancer death in the Western world in the next decade. Therefore, it is essential to discover novel therapeutic targets and to develop more effective and pancreatic cancer-specific therapeutic agents. To date, 45 monoclonal antibodies (mAbs) have been approved for the treatment of patients with a wide range of cancers; however, none has yet been approved for pancreatic cancer. In this comprehensive review, we discuss the FDA approved anticancer mAb-based drugs, the results of preclinical studies and clinical trials with mAbs in pancreatic cancer and the factors contributing to the poor response to antibody therapy (e.g. tumour heterogeneity, desmoplastic stroma). MAb technology is an excellent tool for studying the complex biology of pancreatic cancer, to discover novel therapeutic targets and to develop various forms of antibody-based therapeutic agents and companion diagnostic tests for the selection of patients who are more likely to benefit from such therapy. These should result in the approval and routine use of antibody-based agents for the treatment of pancreatic cancer patients in the future.
Collapse
Affiliation(s)
- Gustavo A. Arias-Pinilla
- Department of Oncology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK;
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| | - Helmout Modjtahedi
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| |
Collapse
|
17
|
Liu S, Zhao S, Dong Y, Wang T, Niu X, Zhao L, Wang G. Antitumor activity and mechanism of resistance of the novel HDAC and PI3K dual inhibitor CUDC-907 in pancreatic cancer. Cancer Chemother Pharmacol 2021; 87:415-423. [PMID: 33392641 DOI: 10.1007/s00280-020-04210-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Pancreatic cancer is a highly malignant disease with an extremely poor prognosis. The benefit of chemotherapy treatment for pancreatic cancer is very limited. Therefore, new therapeutic targets and approaches are urgently needed for this deadly disease. Multi-target therapy is a potential and feasible treatment strategy. Given the important roles that histone deacetylases (HDACs) and phosphoinositide-3-kinase (PI3K) play in pancreatic cancer, we investigated the antitumor activity and mechanism of novel HDAC and PI3K dual inhibitor CUDC-907 in pancreatic cancer. METHODS AND RESULTS MTT assay and flow cytometric analysis were used to examine the in vitro antitumor activity of CUDC-907. A BxPC-3-derived xenograft mouse model was used to determine CUDC-907 in vivo efficacy. The TUNEL assay as used to determine apoptosis in tumors in vivo post CUDC-907 treatment. Western blots were used to determine the effect of CUDC-907 on protein levels. Our results show that CUDC-907 decreased viable cells and induced cell death in a concentration-dependent manner. Furthermore, CUDC-907 showed promising in vivo antitumor activity in the BxPC-3-derived xenograft mouse model while exhibiting tolerable toxicity. Furthermore, long-term treatment with CUDC-907 induced phosphorylation of AKT, S6 (ribosomal protein S6), and ERK (extracellular regulated protein kinase), and inhibition of PI3K (phosphatidylinositol 3-kinase), mTOR (mammalian target of rapamycin), or ERK significantly enhanced CUDC-907-induced cell death in pancreatic cell lines. CONCLUSION Taken together, these findings support the clinical development of CUDC-907 for the treatment of pancreatic cancer and identify compensatory activation of mTOR and MEK/ERK as a possible mechanism of resistance to CUDC-907.
Collapse
Affiliation(s)
- Shuang Liu
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China
| | - Shoujing Zhao
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China
| | - Yang Dong
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China
| | - Tingting Wang
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China
| | - Xiaojia Niu
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Guan Wang
- Key Laboratory for Molecular Enzymology and Engineering, National Engineering Laboratory for AIDS Vaccine, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, Jilin, China.
| |
Collapse
|
18
|
Stoica AF, Chang CH, Pauklin S. Molecular Therapeutics of Pancreatic Ductal Adenocarcinoma: Targeted Pathways and the Role of Cancer Stem Cells. Trends Pharmacol Sci 2020; 41:977-993. [PMID: 33092892 DOI: 10.1016/j.tips.2020.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/01/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers in humans due to late detection and highly metastatic characteristics. PDAC cells vary in their tumorigenic capabilities with the presence of a subset of PDAC cells known as pancreatic cancer stem cells (CSCs), which are more resistant to currently used therapeutics. Here, we describe the role of CSCs and tumour stroma in developing therapeutic strategies for PDAC and suggest that developmental plasticity could be considered a hallmark of cancers. We provide an overview of the molecular targets in PDAC treatments, including targeted therapies of cellular processes such as proliferation, evasion of growth suppressors, activating metastasis, and metabolic effects. Since PDAC is an inflammation-driven cancer, we also revisit therapeutic strategies targeting inflammation and immunotherapy. Lastly, we suggest that targeting epigenetic mechanisms opens therapeutic routes for heterogeneous cancer cell populations, including CSCs.
Collapse
Affiliation(s)
- Andrei-Florian Stoica
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK
| | - Chao-Hui Chang
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK.
| |
Collapse
|
19
|
Pancreatic cancer drug-sensitivity predicted by synergy of p53-Activator Wnt Inhibitor-2 (PAWI-2) and protein biomarker expression. Invest New Drugs 2020; 39:131-141. [PMID: 32915418 DOI: 10.1007/s10637-020-00998-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/04/2020] [Indexed: 10/23/2022]
Abstract
Today, pancreatic cancer (PC) is a major health problem in the United States. It remains a challenge to develop efficacious clinically useful PC therapies. New avenues, based on translational approaches and innovative validated biomarkers could be a preclinical option to evaluate PC drug candidates or drug combinations before clinical trials. Herein, we describe evaluation of combination therapies by incorporating a novel pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) with other FDA-approved cancer drugs that have been used in PC clinical trials. PAWI-2 is a potent inhibitor of drug-resistant PC cells that has been shown to selectively ameliorate human pancreatic cancer stem cells (i.e., hPCSCs, FGβ3 cells). In the present study, we showed PAWI-2 produced therapeutic synergism with certain types of anti-cancer drugs. These drugs themselves oftentimes do not ameliorate PC cells (especially PCSCs) due to high levels of drug-resistance. PAWI-2 has the ability to rescue the potency of drugs (i.e., erlotinib, trametinib) and inhibit PC cell growth. Key molecular regulators of PAWI-2 could be used to predict synergistic/antagonistic effects between PAWI-2 and other anti-cancer drugs. Anti-cancer results showed potency could be quite accurately correlated to phosphorylation of optineurin (OPTN) in PC cells. Synergism/antagonism was also associated with inhibition of PCSC marker SOX2 that was observed in FGβ3 cells. Synergism broadens the potential use of PAWI-2 as an adjunct chemotherapy in patients with PC that have developed resistance to first-line targeted therapies or chemotherapies.
Collapse
|
20
|
Liu M, Liu W, Qin Y, Xu X, Yu X, Zhuo Q, Ji S. Regulation of metabolic reprogramming by tumor suppressor genes in pancreatic cancer. Exp Hematol Oncol 2020. [DOI: 10.1186/s40164-020-00179-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023] Open
Abstract
Abstract
Background
Pancreatic cancer continues to be one of the most aggressive malignant tumors. Work in recent years in cancer molecular biology has revealed that metabolic reprogramming is an additional hallmark of cancer that is involved in the pathogenesis of cancers, and is intricately linked to gene mutations.
Main text
However, though oncogenes such as KRAS and c-Myc play important roles in the process, and have been extensively studied, no substantial improvements in the prognosis of pancreatic cancer have seen. Therefore, some scientists have tried to explain the mechanisms of abnormal cancer metabolism from the perspective of tumor suppressor genes. In this paper, we reviewed researches about how metabolic reprogramming was regulated by tumor suppressor genes in pancreatic cancer and their clinical implications.
Conclusion
Abnormal metabolism and genetic mutations are mutually causal and complementary in tumor initiation and development. A clear understanding of how metabolic reprogramming is regulated by the mutated genes would provide important insights into the pathogenesis and ultimately treatment of pancreatic cancer.
Collapse
|
21
|
Rao X, Wan L, Jie Z, Zhu X, Yin J, Cao H. Upregulated miR-27a-3p Indicates a Poor Prognosis in Pancreatic Carcinoma Patients and Promotes the Angiogenesis and Migration by Epigenetic Silencing of GATA6 and Activating VEGFA/VEGFR2 Signaling Pathway. Onco Targets Ther 2019; 12:11241-11254. [PMID: 31908490 PMCID: PMC6927607 DOI: 10.2147/ott.s220621] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/16/2019] [Indexed: 01/03/2023] Open
Abstract
Background Abnormal miR-27a-3p expression has been frequently reported in several types of human cancer and contributes to tumor progression. However, the role and potential molecular mechanism of miR-27a-3p in the progression of pancreatic carcinoma have not been clarified. Materials and methods The expression of miR-27a-3p and GATA binding protein 6 (GATA6) in pancreatic carcinoma tissues and cell lines was evaluated by quantitative real-time PCR and Western blotting analysis. The relationship between clinical pathologic features and miR-27a-3p expression was analyzed with Chi-square test. The regulatory mechanism of miR-27a-3p on GATA6 was confirmed by luciferase reporter assay and bioinformatics analysis. The effects of miR-27a-3p by targeting GATA6 on cell angiogenesis and migration were assessed by capillary tube formation and wound healing assays. Results MiR-27a-3p expression was significantly upregulated in pancreatic carcinoma tissues and cell lines. Highly expressed miR-27a-3p was closely related to more lymph node metastasis, present peritoneal metastasis, and poor prognosis in patients with pancreatic carcinoma. MiR-27a-3p promoted migration and angiogenesis of pancreatic carcinoma cells by activating vascular endothelial growth factor A (VEGFA) and vascular endothelial growth factor receptor 2 (VEGFR2) expression. A significantly negative correlation between GATA6 mRNA and miR-27a-3 expression was found in pancreatic carcinoma samples. Modulation of miR-27a-3p could alter GATA6 expression in pancreatic carcinoma cells. GATA6 was identified as a functional target gene of miR-27a-3p, and GATA6 knockdown partially reversed the effects of miR-27a-3p siliencing on the migration and angiogenesis of pancreatic carcinoma cells by regulation of VEGFA/VEGFR2 pathway. Conclusion Upregulated miR-27a-3p indicates a poor prognosis in pancreatic carcinoma patients and promotes the angiogenesis and migration by epigenetic silencing of GATA6 and activating VEGFA/VEGFR2 signaling pathway, and indicating miR-27a-3p may be a promising therapeutic target for pancreatic carcinoma treatment.
Collapse
Affiliation(s)
- Xuefeng Rao
- Department of General Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, People's Republic of China
| | - Lihui Wan
- Department of Gastroenterology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, People's Republic of China
| | - Zhigang Jie
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Xiaoliang Zhu
- Department of General Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, People's Republic of China
| | - Junxiang Yin
- Department of General Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, People's Republic of China
| | - Hong Cao
- Department of General Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, People's Republic of China
| |
Collapse
|
22
|
Parkin A, Man J, Timpson P, Pajic M. Targeting the complexity of Src signalling in the tumour microenvironment of pancreatic cancer: from mechanism to therapy. FEBS J 2019; 286:3510-3539. [PMID: 31330086 PMCID: PMC6771888 DOI: 10.1111/febs.15011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/26/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer, a disease with extremely poor prognosis, has been notoriously resistant to virtually all forms of treatment. The dynamic crosstalk that occurs between tumour cells and the surrounding stroma, frequently mediated by intricate Src/FAK signalling, is increasingly recognised as a key player in pancreatic tumourigenesis, disease progression and therapeutic resistance. These important cues are fundamental for defining the invasive potential of pancreatic tumours, and several components of the Src and downstream effector signalling have been proposed as potent anticancer therapeutic targets. Consequently, numerous agents that block this complex network are being extensively investigated as potential antiinvasive and antimetastatic therapeutic agents for this disease. In this review, we will discuss the latest evidence of Src signalling in PDAC progression, fibrotic response and resistance to therapy. We will examine future opportunities for the development and implementation of more effective combination regimens, targeting key components of the oncogenic Src signalling axis, and in the context of a precision medicine-guided approach.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Jennifer Man
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Paul Timpson
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| | - Marina Pajic
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| |
Collapse
|
23
|
Fan K, Fan Z, Cheng H, Huang Q, Yang C, Jin K, Luo G, Yu X, Liu C. Hexokinase 2 dimerization and interaction with voltage-dependent anion channel promoted resistance to cell apoptosis induced by gemcitabine in pancreatic cancer. Cancer Med 2019; 8:5903-5915. [PMID: 31426130 PMCID: PMC6792491 DOI: 10.1002/cam4.2463] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/12/2019] [Accepted: 06/29/2019] [Indexed: 12/12/2022] Open
Abstract
Gemcitabine (GEM) is the standard chemotherapy drug for pancreatic cancer. Because of widespread drug resistance, the effect is limited. Therefore, it is urgent to reveal the underlying mechanism. Glycolysis is the most remarkable character of tumor aberrant metabolism, which plays vital roles on tumor drug resistance. Hexokinase 2 (HK2), as the key enzyme regulating the first‐step reaction of glycolysis, is overexpressed in many kinds of tumors. The putative role of HK2 resisting GEM therapy was investigated in this study. We found that HK2 was overexpressed in pancreatic cancer and associated with poor prognosis. HK2 knockdown decreased pancreatic cancer cell proliferation, migration viability, and promoted cell apoptosis in vitro. HK2 high expression in pancreatic cancer showed GEM resistance. HK2 knockdown increased the sensitivity of pancreatic cancer cell to GEM, the growth of xenograft tumor with HK2 knockdown was also further decreased with the GEM treatment compared with control in vivo. GEM‐resistant pancreatic cancer showed the increase of HK2 dimer rather than HK2 mRNA or protein. Our study revealed that the ROS derived from GEM promoted HK2 dimerization combining with voltage‐dependent anion channel, which resulted in the resistance to GEM. Meanwhile, our study established a new sight for GEM resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Kun Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Zhiyao Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Qiuyi Huang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Chao Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China.,Shanghai Pancreatic Cancer Institute, Shanghai, P.R. China.,Pancreatic Cancer Institute, Fudan University, Shanghai, P.R. China
| |
Collapse
|
24
|
Firuzi O, Che PP, El Hassouni B, Buijs M, Coppola S, Löhr M, Funel N, Heuchel R, Carnevale I, Schmidt T, Mantini G, Avan A, Saso L, Peters GJ, Giovannetti E. Role of c-MET Inhibitors in Overcoming Drug Resistance in Spheroid Models of Primary Human Pancreatic Cancer and Stellate Cells. Cancers (Basel) 2019; 11:E638. [PMID: 31072019 PMCID: PMC6562408 DOI: 10.3390/cancers11050638] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/18/2019] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic stellate cells (PSCs) are a key component of tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) and contribute to drug resistance. c-MET receptor tyrosine kinase activation plays an important role in tumorigenesis in different cancers including PDAC. In this study, effects of PSC conditioned medium (PCM) on c-MET phosphorylation (by immunocytochemistry enzyme-linked immunosorbent assay (ELISA)) and drug response (by sulforhodamine B assay) were investigated in five primary PDAC cells. In novel 3D-spheroid co-cultures of cyan fluorescence protein (CFP)-firefly luciferase (Fluc)-expressing primary human PDAC cells and green fluorescence protein (GFP)-expressing immortalized PSCs, PDAC cell growth and chemosensitivity were examined by luciferase assay, while spheroids' architecture was evaluated by confocal microscopy. The highest phospho-c-MET expression was detected in PDAC5 and its subclone sorted for "stage specific embryonic antigen-4" (PDAC5 (SSEA4)). PCM of cells pre-incubated with PDAC conditioned medium, containing increased hepatocyte growth factor (HGF) levels, made PDAC cells significantly more resistant to gemcitabine, but not to c-MET inhibitors. Hetero-spheroids containing both PSCs and PDAC5 (SSEA4) cells were more resistant to gemcitabine compared to PDAC5 (SSEA4) homo-spheroids. However, c-MET inhibitors (tivantinib, PHA-665752 and crizotinib) were equally effective in both spheroid models. Experiments with primary human PSCs confirmed the main findings. In conclusion, we developed spheroid models to evaluate PSC-PDAC reciprocal interaction, unraveling c-MET inhibition as an important therapeutic option against drug resistant PDAC.
Collapse
Affiliation(s)
- Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, 71348-14336 Shiraz, Iran.
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Pei Pei Che
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Btissame El Hassouni
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Mark Buijs
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Stefano Coppola
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, 2333 CA, Leiden, The Netherlands.
| | - Matthias Löhr
- Division of Surgery, CLINTEC, Karolinska Institutet, SE-171, Stockholm, Sweden.
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, 56124 Pisa, Italy.
| | - Rainer Heuchel
- Division of Surgery, CLINTEC, Karolinska Institutet, SE-171, Stockholm, Sweden.
| | - Ilaria Carnevale
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, 56124 Pisa, Italy.
| | - Thomas Schmidt
- Physics of Life Processes, Huygens-Kamerlingh Onnes Laboratory, Leiden University, 2333 CA, Leiden, The Netherlands.
| | - Giulia Mantini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, 91778-99191 Mashhad, Iran.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, 00185, Rome, Italy.
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, 56124 Pisa, Italy.
- Fondazione Pisana per la Scienza, 56017, Pisa, Italy.
| |
Collapse
|
25
|
Candido S, Abrams SL, Steelman LS, Lertpiriyapong K, Martelli AM, Cocco L, Ratti S, Follo MY, Murata RM, Rosalen PL, Bueno-Silva B, de Alencar SM, Lombardi P, Mao W, Montalto G, Cervello M, Rakus D, Gizak A, Lin HL, Libra M, Akula SM, McCubrey JA. Effects of the MDM-2 inhibitor Nutlin-3a on PDAC cells containing and lacking WT-TP53 on sensitivity to chemotherapy, signal transduction inhibitors and nutraceuticals. Adv Biol Regul 2019; 72:22-40. [PMID: 30898612 DOI: 10.1016/j.jbior.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 06/09/2023]
Abstract
Mutations at the TP53 gene are readily detected (approximately 50-75%) in pancreatic ductal adenocarcinoma (PDAC) patients. TP53 was previously thought to be a difficult target as it is often mutated, deleted or inactivated on both chromosomes in certain cancers. In the following study, the effects of restoration of wild-type (WT) TP53 activity on the sensitivities of MIA-PaCa-2 pancreatic cancer cells to the MDM2 inhibitor nutlin-3a in combination with chemotherapy, targeted therapy, as well as, nutraceuticals were examined. Upon introduction of the WT-TP53 gene into MIA-PaCa-2 cells, which contain a TP53 gain of function (GOF) mutation, the sensitivity to the MDM2 inhibitor increased. However, effects of nutlin-3a were also observed in MIA-PaCa-2 cells lacking WT-TP53, as upon co-treatment with nutlin-3a, the sensitivity to certain inhibitors, chemotherapeutic drugs and nutraceuticals increased. Interestingly, co-treatment with nutlin-3a and certain chemotherapeutic drug such as irinotecan and oxaliplatin resulted in antagonistic effects in cells both lacking and containing WT-TP53 activity. These studies indicate the sensitizing abilities that WT-TP53 activity can have in PDAC cells which normally lack WT-TP53, as well as, the effects that the MDM2 inhibitor nutlin-3a can have in both cells containing and lacking WT-TP53 to various therapeutic agents.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Kvin Lertpiriyapong
- Weill Cornell Medicine and the Hospital for Special Surgery, New York City, New York, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Ramiro M Murata
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Bruno Bueno-Silva
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil; Dental Research Division, Guarulhos University, Guarulhos, Brazil
| | | | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese, 20026, Italy
| | - Weifeng Mao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Giuseppe Montalto
- Dipartimento di Promozione Della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Agnieska Gizak
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Heng-Liang Lin
- Catholic Fu Jen University Hospital, New Taipei City, Taiwan
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834.
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834.
| |
Collapse
|
26
|
LncRNA TP73-AS1 sponges miR-141-3p to promote the migration and invasion of pancreatic cancer cells through the up-regulation of BDH2. Biosci Rep 2019; 39:BSR20181937. [PMID: 30643007 PMCID: PMC6418400 DOI: 10.1042/bsr20181937] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/09/2018] [Accepted: 12/28/2018] [Indexed: 12/11/2022] Open
Abstract
LncRNA TP73 antisense RNA 1T (TP73-AS1) plays an important role in human malignancies. However, the levels of TP73-AS1 and its functional mechanisms in pancreatic cancer metastasis remain unknown, and the clinical significance of TP73-AS1 in human pancreatic cancer is also unclear. In the present study, the levels of TP73-AS1 and its candidate target miR-141 in pancreatic cancer and adjacent normal tissue were detected using qRT-PCR. The association between TP73-AS1 levels and the clinicopathologic characteristics of pancreatic cancer patients were analyzed. The relationship between TP73-AS1 and miR-141, and miR-141 and its candidate target 3-hydroxybutyrate dehydrogenase type 2 (BDH2) was confirmed using dual-luciferase reporter assays. TP73-AS1 and/or miR-141 were knocked down using siRNA or an inhibitor in pancreatic cancer cells and cell migration and invasion then examined. The results showed that TP73-AS1 was up-regulated in pancreatic cancer tissue and cell lines. High levels of TP73-AS1 were correlated with poor clinicopathological characteristics and shorter overall survival. MiR-141 was a direct target for TP73-AS1, while BDH2 was a direct target for miR-141. The knockdown of TP73-AS1 significantly inhibited the migration and invasion of pancreatic cancer cells, while the miR-141 inhibitor significantly restored the migration and invasion. Therefore, TP73-AS1 positively regulated BDH2 expression by sponging miR-141. These findings suggest that TP73-AS1 serves as an oncogene and promotes the metastasis of pancreatic cancer. Moreover, TP73-AS1 could serve as a predictor and a potential drug biotarget for pancreatic cancer.
Collapse
|
27
|
Sun Q, Xu W, Ji S, Qin Y, Liu W, Hu Q, Zhang Z, Liu M, Yu X, Xu X. Role of hepatocyte nuclear factor 4 alpha in cell proliferation and gemcitabine resistance in pancreatic adenocarcinoma. Cancer Cell Int 2019; 19:49. [PMID: 30867652 PMCID: PMC6398265 DOI: 10.1186/s12935-019-0767-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 02/28/2019] [Indexed: 01/03/2023] Open
Abstract
Background Hepatocyte nuclear factor 4α (HNF4α) is a tissue-specific transcription factor that regulates the expression of numerous genes in hepatocytes and pancreatic β cells. HNF4α has been reported to affect cell proliferation and chemoresistance in several cancers. However, the role of HNF4α in pancreatic adenocarcinoma (PDAC) has not been studied extensively and remains unclear. Methods By utilizing immunohistochemical (IHC) staining, we measured the expression of HNF4α in PDAC tissues. By silencing HNF4α in PDAC cell lines, we assessed the impact of HNF4α on pancreatic cancer cell proliferation and gemcitabine sensitivity. We used CCK8 and colony formation assays to examine the effect of HNF4α on cell proliferation. A flow cytometry assay was used to assess cell apoptosis. The expression of gemcitabine-related genes was detected by quantitative real‑time PCR (qRT-PCR) and Western blotting. IHC was utilized to assess the correlation between HNF4α and human equilibrative nucleoside transporter 1 (hENT1) expression in PDAC patients. Chromatin immunoprecipitation (ChIP) and dual‑luciferase reporter assays were used to confirm that hENT1 is a target gene of HNF4α. Results Increased HNF4α expression was detected in PDAC tissues; patients with higher HNF4α expression displayed worse prognosis. To elucidate the function of HNF4α, we examined its role in pancreatic cancer cell proliferation, apoptosis and gemcitabine resistance. In HNF4α-silenced Capan-1 and MiaPaCa-2 cells, we observed decreased cell proliferation and increased sensitivity to gemcitabine compared to those of controls. The mechanism of HNF4α in gemcitabine-related chemosensitivity was then explored. In response to HNF4α silencing, the expression levels of gemcitabine-related proteins, hENT1 and deoxycytidine kinase (dCK) were significantly increased. Additionally, hENT1 was negatively correlated with HNF4α in PDAC tissue samples. Moreover, we identified hENT1 as a downstream target of HNF4α. Conclusion HNF4α is a prognostic marker for overall survival, is required for pancreatic cancer cell proliferation and promotes resistance to gemcitabine by downregulating hENT1. Therefore, targeting HNF4α might reverse gemcitabine resistance and provide novel treatment strategies for PDAC. Electronic supplementary material The online version of this article (10.1186/s12935-019-0767-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiqing Sun
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Wenyan Xu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Shunrong Ji
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Yi Qin
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Wensheng Liu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Qiangsheng Hu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Zheng Zhang
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Mengqi Liu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xianjun Yu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| | - Xiaowu Xu
- 1Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3Pancreatic Cancer Institute, Fudan University, Shanghai, 200032 China.,4Shanghai Pancreatic Cancer Institute, Shanghai, 200032 China
| |
Collapse
|
28
|
Wang ZC, Huang FZ, Xu HB, Sun JC, Wang CF. MicroRNA-137 inhibits autophagy and chemosensitizes pancreatic cancer cells by targeting ATG5. Int J Biochem Cell Biol 2019; 111:63-71. [PMID: 30710750 DOI: 10.1016/j.biocel.2019.01.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/29/2018] [Accepted: 01/29/2019] [Indexed: 12/26/2022]
Abstract
PURPOSE Autophagy play an important role in tumor chemotherapy resistance. It has been reported that miR-137 expression was reducedand involved in the regulation of sensitivity of PC cells to chemotherapy. However, little is known about the underlying molecular mechanisms. In this study, we hypothesized that miR-137 might sensitize PC cells to chemotherapy thought regulating cell autophagy. METHODS Cell survival was determined with MTT assay. Apoptotic cells were assessed with flow cytometric analysis. Fluorescence intensity of GFP-LC3 and RFP-GFP-LC3 were examined with immunofluorescence analysis to determine the autophagy and autophagic flux level. Western blotting assay was used to determine protein expression levels of LC3II/LC3I, P62, FUNDC1 and ATG5. mRNA expression level of miR-137 was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Dual-luciferase reporter assay was used to evaluate the directly binding of miR-137 with its targets. Xenograft model was setup to evaluate tumor growth. RESULTS The results showed that doxorubicin (Dox) induced autophagy but downregulated the expression level of miR-137 in pancreatic cancer (PC) cells. In turn, overexpression of miR-137 enhanced the effect of Dox on decreasing cell survival, inducing cell apoptosis and inhibiting autophagy rather than influencing autophagic flux in PC cells. Further mechanistic study identified that ATG5 was a direct target of miR-137. Moreover, overexpression of ATG5 dramatically reversed the promotion of apoptosis and inhibition of autophagy mediated by higher expression level of miR-137. We also demonstrated that miR-137 sensitized PANC-1 cells to Dox through inhibiting ATG5 and autophagy in vivo. CONCLUSIONS Our findings demonstrated for the first time that miR-137 was able to promote sensitivity of PC cells to chemotherapy via inhibition of autophagy mediated by ATG5. Therefore, miR-137 may act as a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Zhi-Chao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, PR China
| | - Fei-Zhou Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, PR China
| | - Hong-Bo Xu
- Department of Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Ji-Chun Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, PR China
| | - Chang-Fa Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, PR China.
| |
Collapse
|
29
|
Di Carlo C, Brandi J, Cecconi D. Pancreatic cancer stem cells: Perspectives on potential therapeutic approaches of pancreatic ductal adenocarcinoma. World J Stem Cells 2018; 10:172-182. [PMID: 30631392 PMCID: PMC6325076 DOI: 10.4252/wjsc.v10.i11.172] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/10/2018] [Accepted: 10/17/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the most aggressive solid tumours of the pancreas, characterised by a five-year survival rate less than 8%. Recent reports that pancreatic cancer stem cells (PCSCs) contribute to the tumorigenesis, progression, and chemoresistance of pancreatic cancer have prompted the investigation of new therapeutic approaches able to directly target PCSCs. In the present paper the non-cancer related drugs that have been proposed to target CSCs that could potentially combat pancreatic cancer are reviewed and evaluated. The role of some pathways and deregulated proteins in PCSCs as new therapeutic targets are also discussed with a focus on selected specific inhibitors. Finally, advances in the development of nanoparticles for targeting PCSCs and site-specific drug delivery are highlighted, and their limitations considered.
Collapse
Affiliation(s)
- Claudia Di Carlo
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| | - Jessica Brandi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy.
| | - Daniela Cecconi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona 37134, Italy
| |
Collapse
|
30
|
Zhan W, Shelton CA, Greer PJ, Brand RE, Whitcomb DC. Germline Variants and Risk for Pancreatic Cancer: A Systematic Review and Emerging Concepts. Pancreas 2018; 47:924-936. [PMID: 30113427 PMCID: PMC6097243 DOI: 10.1097/mpa.0000000000001136] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer requires many genetic mutations. Combinations of underlying germline variants and environmental factors may increase the risk of cancer and accelerate the oncogenic process. We systematically reviewed, annotated, and classified previously reported pancreatic cancer-associated germline variants in established risk genes. Variants were scored using multiple criteria and binned by evidence for pathogenicity, then annotated with published functional studies and associated biological systems/pathways. Twenty-two previously identified pancreatic cancer risk genes and 337 germline variants were identified from 97 informative studies that met our inclusion criteria. Fifteen of these genes contained 66 variants predicted to be pathogenic (APC, ATM, BRCA1, BRCA2, CDKN2A, CFTR, CHEK2, MLH1, MSH2, NBN, PALB2, PALLD, PRSS1, SPINK1, TP53). Pancreatic cancer risk genes were organized into key biological mechanisms that promote pancreatic oncogenesis within an oncogenic model. Development of precision medicine approaches requires updated variant information within the framework of an oncogenic progression model. Complex risk modeling may improve interpretation of early biomarkers and guide pathway-specific treatment for pancreatic cancer in the future. Precision medicine is within reach.
Collapse
Affiliation(s)
- Wei Zhan
- School of Medicine, Tsinghua University, Beijing, China
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Celeste A. Shelton
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Phil J. Greer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Randall E. Brand
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - David C. Whitcomb
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
31
|
Abrams SL, Lertpiriyapong K, Yang LV, Martelli AM, Cocco L, Ratti S, Falasca M, Murata RM, Rosalen PL, Lombardi P, Libra M, Candido S, Montalto G, Cervello M, Steelman LS, McCubrey JA. Introduction of WT-TP53 into pancreatic cancer cells alters sensitivity to chemotherapeutic drugs, targeted therapeutics and nutraceuticals. Adv Biol Regul 2018; 69:16-34. [PMID: 29980405 DOI: 10.1016/j.jbior.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 06/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5-10%. Mutations at the TP53 gene are readily detected in pancreatic tumors isolated from PDAC patients. We have investigated the effects of restoration of wild-type (WT) TP53 activity on the sensitivity of pancreatic cancer cells to: chemotherapy, targeted therapy, as well as, nutraceuticals. Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Likewise, the sensitivity to drugs used to treat other cancers such as: doxorubicin, mitoxantrone, and 4 hydroxy tamoxifen (4HT) also increased upon introduction of WT-TP53 into MIA-PaCa-2 cells. Furthermore, the sensitivity to certain inhibitors which target: PI3K/mTORC1, PDK1, SRC, GSK-3, and biochemical processes such as proteasomal degradation and the nutraceutical berberine as increased upon introduction of WT-TP53. Furthermore, in some cases, cells with WT-TP53 were more sensitive to the combination of drugs and suboptimal doses of the MDM2 inhibitor nutlin-3a. However, TP53-independent effects of nutlin-3a were observed upon treatment with either a proteasomal or a PI3K/mTOR inhibitor. These studies indicate the sensitizing effects that WT-TP53 can have in PDAC cells which normally lack WT-TP53 to various therapeutic agents and suggest approaches to improve PDAC therapy.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Ramiro M Murata
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy; Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|