1
|
Abuelafia AM, Santofimia-Castaño P, Estaras M, Grasso D, Chuluyan E, Lomberk G, Urrutia R, Dusetti N, Fraunhoffer N, Iovanna J. KRAS inhibition reverses chemotherapy resistance promoted by therapy-induced senescence-like in pancreatic ductal adenocarcinoma. Transl Oncol 2025; 57:102421. [PMID: 40382842 DOI: 10.1016/j.tranon.2025.102421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that chemotherapy can accumulate senescent-like cells within tumor tissues, a phenomenon linked to therapy resistance. The aim of this study is to analyze the senescence-like state of after-treatment persistent cells associated with KRAS mutational status to offer a therapeutic strategy to target these cells in pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN Three commercial cell lines and five patient-derived primary cell cultures with different KRAS statuses were studied following gemcitabine treatment. Senescence-like status was assessed using SA-β-gal, together with cell cycle regulators such as p21. Additionally, KRAS mutations were modulated using MRTX1133 and AMG-510, and the signaling pathways ERK and AKT were analyzed and modulated in vitro. Finally, p21 expression, associated with the senescence-like state, on patient outcomes and treatment response was analyzed in publicly available bulk RNA-seq and single-nucleus datasets. RESULTS We observed an overexpression of p21 alongside an increase in SA-β-gal signal in response to gemcitabine treatment, indicating the induction of a senescence-like state. Specific inhibition of KRAS G12D or G12C mutations reduced SA-β-gal signal and sensitized PDAC cells to gemcitabine. Moreover, ERK inhibition but not AKT inhibition decreased SA-β-gal signal. Additionally, we characterized p21 expression levels in relation to patient outcomes and found that they are modulated by treatment. CONCLUSIONS This dual-targeted therapeutic strategy holds promises for overcoming the challenges posed by KRAS-driven cancers, particularly in addressing the formidable obstacle of pancreatic cancer.
Collapse
Affiliation(s)
- Analia Meilerman Abuelafia
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Matias Estaras
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France
| | - Daniel Grasso
- Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina; Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eduardo Chuluyan
- Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Buenos Aires University, Center for Pharmacological and Botanical Studies, Faculty of Medicine, National Council for Scientific and Technical Research, Buenos Aires C1121ABG, Argentina; Buenos Aires University, Faculty of Medicine, Department of Microbiology, Parasitology and Immunology, Buenos Aires C1121ABG, Argentina
| | - Gwen Lomberk
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Raul Urrutia
- Genomic Science and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina
| | - Nicolas Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Buenos Aires University, Center for Pharmacological and Botanical Studies, Faculty of Medicine, National Council for Scientific and Technical Research, Buenos Aires C1121ABG, Argentina.
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, 163 Avenue de Luminy, 13288 Marseille, Equipe labélisée Ligue Nationale contre le cancer, France; Programa franco-argentino de estudio del Cáncer de Páncreas, Argentina; Hospital de Alta Complejidad El Cruce, Florencio Varela, Buenos Aires, Argentina; University Arturo Jauretche, Florencio Varela, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Kluz N, Kraj L, Chmiel P, Przybyłkowski AM, Wyrwicz L, Stec R, Szymański Ł. Correlation Between Antihypertensive Drugs and Survival Among Patients with Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:3945. [PMID: 39682132 DOI: 10.3390/cancers16233945] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
There is a growing prevalence of pancreatic cancer, accompanied by accelerated disease progression and diminished survival rates. Radical resection with clear margins remains the sole viable option for achieving a long-term cure in patients. In cases of advanced, unresectable, and metastatic disease, chemotherapy based on leucovorin, 5-fluorouracil, irinotecan, oxaliplatin, gemcitabine, or nab-paclitaxel represents the cornerstone of the treatment. Considering the limited treatment options available following initial therapy, the strategy of repurposing commonly prescribed drugs such as antihypertensives into anti-cancer therapies in palliative treatment represents a promising avenue for enhancing survival in patients with pancreatic ductal adenocarcinoma. The repurposing of existing drugs is typically a more cost-effective and expedient strategy than the development of new ones. The potential for antihypertensive drugs to be employed as adjunctive therapies could facilitate a more comprehensive treatment approach by targeting multiple pathways involved in cancer progression and acquired resistance to treatment. Antihypertensive medications, particularly those belonging to the pharmacological classes of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and calcium channel blockers, are commonly prescribed and have well-established safety profiles, particularly among patients with pancreatic cancer who are affected by multiple comorbidities. Therefore, we emphasize the preclinical and clinical evidence supporting the use of antihypertensive agents in the treatment of pancreatic cancer, emphasizing their beneficial chemosensitizing effects.
Collapse
Affiliation(s)
- Natalia Kluz
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Garbatka, Poland
| | - Paulina Chmiel
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Garbatka, Poland
| | - Adam M Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Lucjan Wyrwicz
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Cancer Research Institute, 02-781 Warsaw, Poland
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Garbatka, Poland
| |
Collapse
|
3
|
Kim J, Kahttana I, Yoon H, Chang S, Yoon S. Exploring the Potential of Enhanced Prognostic Performance of NCCN-IPI in Diffuse Large B-Cell Lymphoma by Integrating Tumor Microenvironment Markers: Stromal FOXC1 and Tumor pERK1/2 Expression. Cancer Med 2024; 13:e70305. [PMID: 39404228 PMCID: PMC11475023 DOI: 10.1002/cam4.70305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND FOXC1 and ERK1-2 are proteins implicated in aggressive biological behavior of various malignancies including lymphomas. MATERIAL AND METHODS We investigate the additive prognostic value of stromal FOXC1 expression and tumor phosphorylated ERK1-2 (pERK1-2) expression to the established National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI), in 92 diffuse large B-cell lymphoma (DLBCL) cases. Multidimensional analysis using statistics and machine learning (ML) models assessed prognostic value of established clinicopathologic variables with stromal FOXC1 and tumor pERK1-2 expressions. RESULTS Both high FOXC1 stroma group and high pERK1-2 tumor group were significantly associated with shorter progression-free survival (PFS) and overall survival (OS) compared with low group (p = 0.015, 0.034 and p = 0.025, 0.025 each respectively). In multivariable analysis, high FOXC1 stromal expression was an independent prognostic factor of OS (p = 0.037). The addition of stromal FOXC1 and tumor pERK1-2 to the NCCN-IPI score significantly improved prediction of time to death compared with NCCN-IPI score alone (Harrell's C-index = 0.801 vs. 0.764; p = 0.030). ML models reconfirmed the addition of stromal FOXC1 expression and tumor pERK1-2 to NCCN-IPI score had the highest C-index (0.952) among combinations. Stromal FOXC1 and tumor pERK1-2 were determinants of DLBCL prognosis, whose addition significantly improved prognostic performance of the NCCN-IPI.
Collapse
Affiliation(s)
- Ji‐Ye Kim
- Department of Pathology, Ilsan Paik HospitalInje University College of MedicineGoyang‐siGyeonggi‐doRepublic of Korea
- Department of PathologyYonsei University College of Medicine, Severance HospitalSeoulRepublic of Korea
| | - Ibadullah Kahttana
- Division of Electronics and Information EngineeringJeonbuk National UniversityJeonju‐siRepublic of Korea
| | - Hyonok Yoon
- College of PharmacyResearch Institute of Pharmaceutical Sciences, Gyeongsang National UniversityJinju‐siRepublic of Korea
| | - Sunhee Chang
- Department of Pathology, Ilsan Paik HospitalInje University College of MedicineGoyang‐siGyeonggi‐doRepublic of Korea
| | - Sun Och Yoon
- Department of PathologyYonsei University College of Medicine, Severance HospitalSeoulRepublic of Korea
| |
Collapse
|
4
|
Kumar V, Sethi B, Staller DW, Shrestha P, Mahato RI. Gemcitabine elaidate and ONC201 combination therapy for inhibiting pancreatic cancer in a KRAS mutated syngeneic mouse model. Cell Death Discov 2024; 10:158. [PMID: 38553450 PMCID: PMC10980688 DOI: 10.1038/s41420-024-01920-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
Approximately 90% of pancreatic cancer (PC) contain KRAS mutations. Mutated KRAS activates the downstream oncogenic PI3K/AKT and MEK signaling pathways and induces drug resistance. However, targeting both pathways with different drugs can also lead to excessive toxicity. ONC201 is a dual PI3K/AKT and MEK pathway inhibitor with an excellent safety profile that targets death receptor 5 (DR5) to induce apoptosis. Gemcitabine (GEM) is a first-line chemotherapy in PC, but it is metabolically unstable and can be stabilized by a prodrug approach. In this study, phospho-Akt, phospho-mTOR, and phospho-ERK protein expressions were evaluated in patient PDAC-tissues (n = 10). We used lipid-gemcitabine (L_GEM) conjugate, which is more stable and enters the cells by passive diffusion. Further, we evaluated the efficacy of L_GEM and ONC201 in PC cells and "KrasLSL-G12D; p53LoxP; Pdx1-CreER (KPC) triple mutant xenograft tumor-bearing mice. PDAC patient tissues showed significantly higher levels of p-AKT (Ser473), p-ERK (T202/T204), and p-mTOR compared to surrounding non-cancerous tissues. ONC201 in combination with L_GEM, showed a superior inhibitory effect on the growth of MIA PaCa-2 cells. In our in-vivo study, we found that ONC201 and L_GEM combination prevented neoplastic proliferation via AKT/ERK blockade to overcome chemoresistance and increased T-cell tumor surveillance. Simultaneous inhibition of the PI3K/AKT and MEK pathways with ONC201 is an attractive approach to potentiate the effect of GEM. Our findings provide insight into rational-directed precision chemo and immunotherapy therapy in PDAC.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bharti Sethi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Dalton W Staller
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prakash Shrestha
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
5
|
Mahato R, Kumar V, Sethi B, Staller D. Gemcitabine elaidate and ONC201 combination therapy inhibits pancreatic cancer in a KRAS mutated syngeneic mouse model. RESEARCH SQUARE 2023:rs.3.rs-3108907. [PMID: 37503215 PMCID: PMC10371096 DOI: 10.21203/rs.3.rs-3108907/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Approximately 90% of pancreatic cancer (PC) contain KRAS mutations. Mutated KRAS activates the downstream oncogenic PI3K/AKT and MEK signaling pathways and induces drug resistance. However, targeting both pathways with different drugs can also lead to access of toxicity. ONC201 targets DR5 to induce apoptosis in several types of cancers and has an excellent safety profile. ONC201 is also a dual PI3K/AKT and MEK pathways inhibitor. Gemcitabine (GEM) is a first-line chemotherapy in PC, but it is metabolically unstable, which can be stabilized by prodrug approach. Here, we used lipid-gemcitabine (L_GEM) conjugate, which is more stable and enters the cells by passive diffusion. We evaluated the efficacy of L_GEM and ONC201 in PanCan cells, and "KrasLSL-G12D; p53LoxP; Pdx1-CreER (KPC) triple mutant xenograft tumor-bearing mice. ONC201, in combination with L_GEM, showed a superior inhibitory effect on the growth of MIA PaCa-2 cells. ONC201 and L_GEM combination prevented neoplastic proliferation via AKT/ERK blockade, to overcome chemoresistance, and increased T-cell tumor surveillance. Simultaneous inhibition of the PI3K/AKT and MEK pathways with ONC201 is an attractive approach to potentiate GEM. Our findings provide insight into rational-directed precision chemo and immunotherapy therapy in PDAC.
Collapse
|
6
|
Abuhijjleh RK, Al Saeedy DY, Ashmawy NS, Gouda AE, Elhady SS, Al-Abd AM. Chemomodulatory Effect of the Marine-Derived Metabolite "Terrein" on the Anticancer Properties of Gemcitabine in Colorectal Cancer Cells. Mar Drugs 2023; 21:md21050271. [PMID: 37233465 DOI: 10.3390/md21050271] [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: 03/25/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Terrein (Terr) is a bioactive marine secondary metabolite that possesses antiproliferative/cytotoxic properties by interrupting various molecular pathways. Gemcitabine (GCB) is an anticancer drug used to treat several types of tumors such as colorectal cancer; however, it suffers from tumor cell resistance, and therefore, treatment failure. METHODS The potential anticancer properties of terrein, its antiproliferative effects, and its chemomodulatory effects on GCB were assessed against various colorectal cancer cell lines (HCT-116, HT-29, and SW620) under normoxic and hypoxic (pO2 ≤ 1%) conditions. Further analysis via flow cytometry was carried out in addition to quantitative gene expression and 1HNMR metabolomic analysis. RESULTS In normoxia, the effect of the combination treatment (GCB + Terr) was synergistic in HCT-116 and SW620 cell lines. In HT-29, the effect was antagonistic when the cells were treated with (GCB + Terr) under both normoxic and hypoxic conditions. The combination treatment was found to induce apoptosis in HCT-116 and SW620. Metabolomic analysis revealed that the change in oxygen levels significantly affected extracellular amino acid metabolite profiling. CONCLUSIONS Terrein influenced GCB's anti-colorectal cancer properties which are reflected in different aspects such as cytotoxicity, cell cycle progression, apoptosis, autophagy, and intra-tumoral metabolism under normoxic and hypoxic conditions.
Collapse
Affiliation(s)
- Reham Khaled Abuhijjleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Dalia Yousef Al Saeedy
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Naglaa S Ashmawy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11591, Egypt
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmed E Gouda
- Life Science Unit, Biomedical Research Division, Nawah Scientific, Al-Mokkatam, Cairo 11571, Egypt
| | - Sameh S Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Mohamed Al-Abd
- Life Science Unit, Biomedical Research Division, Nawah Scientific, Al-Mokkatam, Cairo 11571, Egypt
- National Research Centre, Department of Pharmacology, Medical and Clinical Research Institute, Cairo 12622, Egypt
| |
Collapse
|
7
|
Guo D, Ye L, Wu W, Yu X, Jin K. Novel strategy for oncogenic alteration-induced lipid metabolism reprogramming in pancreatic cancer. Acta Biochim Biophys Sin (Shanghai) 2023; 55:923-937. [PMID: 37021976 PMCID: PMC10326418 DOI: 10.3724/abbs.2023045] [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: 11/21/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
The pathogenesis of pancreatic cancer involves substantial metabolic reprogramming, resulting in abnormal proliferation of tumor cells. This tumorigenic reprogramming is often driven by genetic mutations, such as activating mutations of the KRAS oncogene and inactivating or deletions of the tumor suppressor genes SMAD4, CDKN2A, and TP53, which play a critical role in the initiation and development of pancreatic cancer. As a normal cell gradually develops into a cancer cell, a series of signature characteristics are acquired: activation of signaling pathways that sustain proliferation; an ability to resist growth inhibitory signals and evade apoptosis; and an ability to generate new blood vessels and invade and metastasize. In addition to these features, recent research has revealed that metabolic reprogramming and immune escape are two other novel characteristics of tumor cells. The effect of the interactions between tumor and immune cells on metabolic reprogramming is a key factor determining the antitumor immunotherapy response. Lipid metabolism reprogramming, a feature of many malignancies, not only plays a role in maintaining tumor cell proliferation but also alters the tumor microenvironment by inducing the release of metabolites that in turn affect the metabolism of normal immune cells, ultimately leading to the attenuation of the antitumor immune response and resistance to immunotherapy. Pancreatic cancer has been found to have substantial lipid metabolism reprogramming, but the mechanisms remain elusive. Therefore, this review focuses on the mechanisms regulating lipid metabolism reprogramming in pancreatic cancer cells to provide new therapeutic targets and aid the development of new therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Duancheng Guo
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Longyun Ye
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Weiding Wu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| | - Kaizhou Jin
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Pancreatic Cancer InstituteShanghai200032China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032China
| |
Collapse
|
8
|
Pancreatic stellate cell-induced gemcitabine resistance in pancreatic cancer is associated with LDHA- and MCT4-mediated enhanced glycolysis. Cancer Cell Int 2023; 23:9. [PMID: 36658582 PMCID: PMC9850604 DOI: 10.1186/s12935-023-02852-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Profound resistance to chemotherapy remains a major challenge in achieving better clinical outcomes for patients with pancreatic ductal adenocarcinoma (PDAC). Recent studies indicate that gemcitabine (GEM) resistance is promoted both by pancreatic stellate cells (PSCs) and through increased glycolysis. However, it remains unknown whether PSCs affect GEM sensitivity via glycolytic regulation. METHODS Human pancreatic cancer cell (PCC) lines (BxPC-3, Capan-2, HPAF-II, Mia PaCa-2, Panc-1, SW-1990) were exposed to three different PSC-conditioned media (PSC-CM; PSC-1, PSC-2, HPaSteC), following either pre-treatment with glycolysis inhibitor NV-5440 or transfection for transient silencing of key glycolytic regulators (LDHA and MCT4). Proliferation, glucose transport, extracellular lactate, and GEM sensitivity were assessed. Protein expression was determined by Western blot and immunostaining. Moreover, secreted proteins in PSC-CMs were profiled by mass spectrometry (MS). RESULTS While exposure to PSC-CMs did not affect glucose transport in PCCs, it increased their lactate release and proliferation, and reduced the sensitivity for GEM. Both NV-5440 treatment and transient silencing of LDHA and MCT4 inhibited these PSC-induced changes in PCCs. MS analysis identified 688 unique proteins with differential expression, of which only 87 were common to the three PSC-CMs. Most PSC-secreted proteins were extracellular matrix-related, including SPARC, fibronectin, and collagens. Moreover, exposure to PSC-CMs increased the phosphorylation of ERK in PCCs, but the treatment of PCCs with the MEK/ERK inhibitor PD98059 resulted in a reduction of PSC-CM-induced glycolysis and improved GEM sensitivity. CONCLUSIONS The study findings suggest that PSC-secreted factors promote both glycolysis and GEM resistance in PCCs, and that glycolysis inhibition by NV-5440 and blocking of ERK phosphorylation by PD98059 protect PCCs from PSC-CM-induced loss of GEM sensitivity. Taken together, PSCs appear to promote GEM resistance in PDAC via glycolysis. Thus, targeting glycolysis may improve the effect of chemotherapy in PDAC.
Collapse
|
9
|
Wang CX, Wang TT, Zhang KD, Li MY, Shen QC, Lu SY, Zhang J. Pan-KRAS inhibitors suppress proliferation through feedback regulation in pancreatic ductal adenocarcinoma. Acta Pharmacol Sin 2022; 43:2696-2708. [PMID: 35352018 PMCID: PMC9525295 DOI: 10.1038/s41401-022-00897-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/06/2022] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is currently one of the most lethal cancers worldwide. Several basic studies have confirmed that Kirsten rat sarcoma virus (KRAS) is a key driver gene for the occurrence of PDAC, and KRAS mutations have also been found in most patients in clinical studies. In this study, two pan-KRAS inhibitors, BI-2852 and BAY-293, were chosen as chemical probes to investigate their antitumor potency in PDAC. Their inhibitory effects on KRAS activation were validated in vitro and their antiproliferative potency in PDAC cell lines were profiled, with half-maximal inhibitory concentration (IC50) values of approximately 1 μM, demonstrating the therapeutic potential of pan-KRAS inhibitors in the treatment of PDAC. However, feedback regulation in the KRAS pathway weakened inhibitor activity, which was observed by a 50 times difference in BAY-293 from in vitro activity. Furthermore, pan-KRAS inhibitors effectively inhibited cell proliferation in 3D organoids cultured from PDAC patient samples; however, there were some variations between individuals. These results provide a sufficient theoretical foundation for KRAS as a clinical therapeutic target and for the application of pan-KRAS inhibitors in the treatment of PDAC, with important scientific significance in translational medicine.
Collapse
Affiliation(s)
- Cheng-Xiang Wang
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Ting-Ting Wang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Kun-Dong Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Ming-Yu Li
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Qian-Cheng Shen
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Shao-Yong Lu
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China.
| | - Jian Zhang
- State Key Laboratory of Oncogenes and Related Genes, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China.
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
10
|
Lin Q, Shen S, Qian Z, Rasam SS, Serratore A, Jusko WJ, Kandel ES, Qu J, Straubinger RM. Comparative Proteomic Analysis Identifies Key Metabolic Regulators of Gemcitabine Resistance in Pancreatic Cancer. Mol Cell Proteomics 2022; 21:100409. [PMID: 36084875 PMCID: PMC9582795 DOI: 10.1016/j.mcpro.2022.100409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/21/2022] [Accepted: 09/04/2022] [Indexed: 01/18/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is highly refractory to treatment. Standard-of-care gemcitabine (Gem) provides only modest survival benefits, and development of Gem resistance (GemR) compromises its efficacy. Highly GemR clones of Gem-sensitive MIAPaCa-2 cells were developed to investigate the molecular mechanisms of GemR and implemented global quantitative differential proteomics analysis with a comprehensive, reproducible ion-current-based MS1 workflow to quantify ∼6000 proteins in all samples. In GemR clone MIA-GR8, cellular metabolism, proliferation, migration, and 'drug response' mechanisms were the predominant biological processes altered, consistent with cell phenotypic alterations in cell cycle and motility. S100 calcium binding protein A4 was the most downregulated protein, as were proteins associated with glycolytic and oxidative energy production. Both responses would reduce tumor proliferation. Upregulation of mesenchymal markers was prominent, and cellular invasiveness increased. Key enzymes in Gem metabolism pathways were altered such that intracellular utilization of Gem would decrease. Ribonucleoside-diphosphate reductase large subunit was the most elevated Gem metabolizing protein, supporting its critical role in GemR. Lower Ribonucleoside-diphosphate reductase large subunit expression is associated with better clinical outcomes in PDAC, and its downregulation paralleled reduced MIAPaCa-2 proliferation and migration and increased Gem sensitivity. Temporal protein-level Gem responses of MIAPaCa-2 versus GemR cell lines (intrinsically GemR PANC-1 and acquired GemR MIA-GR8) implicate adaptive changes in cellular response systems for cell proliferation and drug transport and metabolism, which reduce cytotoxic Gem metabolites, in DNA repair, and additional responses, as key contributors to the complexity of GemR in PDAC. These findings additionally suggest targetable therapeutic vulnerabilities for GemR PDAC patients.
Collapse
Affiliation(s)
- Qingxiang Lin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Zhicheng Qian
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Sailee S Rasam
- Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Andrea Serratore
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Eugene S Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jun Qu
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York, USA.
| | - Robert M Straubinger
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| |
Collapse
|
11
|
Exploration of the System-Level Mechanisms of the Herbal Drug FDY003 for Pancreatic Cancer Treatment: A Network Pharmacological Investigation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7160209. [PMID: 35591866 PMCID: PMC9113891 DOI: 10.1155/2022/7160209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Pancreatic cancer (PC) is the most lethal cancer with the lowest survival rate globally. Although the prescription of herbal drugs against PC is gaining increasing attention, their polypharmacological therapeutic mechanisms are yet to be fully understood. Based on network pharmacology, we explored the anti-PC properties and system-level mechanisms of the herbal drug FDY003. FDY003 decreased the viability of human PC cells and strengthened their chemosensitivity. Network pharmacological analysis of FDY003 indicated the presence of 16 active phytochemical components and 123 PC-related pharmacological targets. Functional enrichment analysis revealed that the PC-related targets of FDY003 participate in the regulation of cell growth and proliferation, cell cycle process, cell survival, and cell death. In addition, FDY003 was shown to target diverse key pathways associated with PC pathophysiology, namely, the PIK3-Akt, MAPK, FoxO, focal adhesion, TNF, p53, HIF-1, and Ras pathways. Our network pharmacological findings advance the mechanistic understanding of the anti-PC properties of FDY003 from a system perspective.
Collapse
|
12
|
Principe DR, Aissa AF, Kumar S, Pham TND, Underwood PW, Nair R, Ke R, Rana B, Trevino JG, Munshi HG, Benevolenskaya EV, Rana A. Calcium channel blockers potentiate gemcitabine chemotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:e2200143119. [PMID: 35476525 PMCID: PMC9170157 DOI: 10.1073/pnas.2200143119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/19/2022] [Indexed: 12/15/2022] Open
Abstract
There is currently no effective treatment for pancreatic ductal adenocarcinoma (PDAC). While palliative chemotherapy offers a survival benefit to most patients, nearly all will eventually progress on treatment and long-term survivability remains poor. Given the lack of subsequent line treatment options, in this study, we sought to identify novel strategies to prevent, delay, or overcome resistance to gemcitabine, one of the most widely used medications in PDAC. Using a combination of single-cell RNA sequencing and high-throughput proteomic analysis, we identified a subset of gemcitabine-resistant tumor cells enriched for calcium/calmodulin signaling. Pharmacologic inhibition of calcium-dependent calmodulin activation led to the rapid loss of drug-resistant phenotypes in vitro, which additional single-cell RNA sequencing identified was due to impaired activation of the RAS/ERK signaling pathway. Consistent with these observations, calcium chelation or depletion of calcium in the culture media also impaired ERK activation in gemcitabine-resistant cells, and restored therapeutic responses to gemcitabine in vitro. We observed similar results using calcium channel blockers (CCBs) such as amlodipine, which inhibited prosurvival ERK signaling in vitro and markedly enhanced therapeutic responses to gemcitabine in both orthotopic xenografts and transgenic models of PDAC. Combined, these results offer insight into a potential means of gemcitabine resistance and suggest that select CCBs may provide a clinical benefit to PDAC patients receiving gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Alexandre F. Aissa
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60612
| | - Sandeep Kumar
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
| | - Patrick W. Underwood
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32611
| | - Rakesh Nair
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Rong Ke
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Basabi Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
| | - Jose G. Trevino
- Division of Surgical Oncology, Department of Surgery, Virginia Commonwealth University, Richmond, VA 23284
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
- Jesse Brown VA Medical Center, Chicago, IL 60612
| | | | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60612
- Jesse Brown VA Medical Center, Chicago, IL 60612
| |
Collapse
|
13
|
An In Vitro Investigation into Cryoablation and Adjunctive Cryoablation/Chemotherapy Combination Therapy for the Treatment of Pancreatic Cancer Using the PANC-1 Cell Line. Biomedicines 2022; 10:biomedicines10020450. [PMID: 35203660 PMCID: PMC8962332 DOI: 10.3390/biomedicines10020450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
As the incidence of pancreatic ductal adenocarcinoma (PDAC) continues to grow, so does the need for new strategies for treatment. One such area being evaluated is cryoablation. While promising, studies remain limited and questions surrounding basic dosing (minimal lethal temperature) coupled with technological issues associated with accessing PDAC tumors and tumor proximity to vasculature and bile ducts, among others, have limited the use of cryoablation. Additionally, as chemotherapy remains the first-line of attack for PDAC, there is limited information on the impact of combining freezing with chemotherapy. As such, this study investigated the in vitro response of a PDAC cell line to freezing, chemotherapy, and the combination of chemotherapy pre-treatment and freezing. PANC-1 cells and PANC-1 tumor models were exposed to cryoablation (freezing insult) and compared to non-frozen controls. Additionally, PANC-1 cells were exposed to varying sub-clinical doses of gemcitabine or oxaliplatin alone and in combination with freezing. The results show that freezing to −10 °C did not affect viability, whereas −15 °C and −20 °C resulted in a reduction in 1 day post-freeze viability to 85% and 20%, respectively, though both recovered to controls by day 7. A complete cell loss was found following a single freeze below −25 °C. The combination of 100 nM gemcitabine (1.1 mg/m2) pre-treatment and a single freeze at −15 °C resulted in near-complete cell death (<5% survival) over the 7-day assessment interval. The combination of 8.8 µM oxaliplatin (130 mg/m2) pre-treatment and a single −15 °C freeze resulted in a similar trend of increased PANC-1 cell death. In summary, these in vitro results suggest that freezing alone to temperatures in the range of −25 °C results in a high degree of PDAC destruction. Further, the data support a potential combinatorial chemo/cryo-therapeutic strategy for the treatment of PDAC. These results suggest that a reduction in chemotherapeutic dose may be possible when offered in combination with freezing for the treatment of PDAC.
Collapse
|
14
|
Cevatemre B, Ulukaya E, Dere E, Dilege S, Acilan C. Pyruvate Dehydrogenase Contributes to Drug Resistance of Lung Cancer Cells Through Epithelial Mesenchymal Transition. Front Cell Dev Biol 2022; 9:738916. [PMID: 35083212 PMCID: PMC8785343 DOI: 10.3389/fcell.2021.738916] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Recently, there has been a growing interest on the role of mitochondria in metastatic cascade. Several reports have shown the preferential utilization of glycolytic pathway instead of mitochondrial respiration for energy production and the pyruvate dehydrogenase (PDH) has been considered to be a contributor to this switch in some cancers. Since epithelial mesenchymal transition (EMT) is proposed to be one of the significant mediators of metastasis, the molecular connections between cancer cell metabolism and EMT may reveal underlying mechanisms and improve our understanding on metastasis. In order to explore a potential role for PDH inhibition on EMT and associated drug resistance, we took both pharmacological and genetic approaches, and selectively inhibited or knocked down PDHA1 by using Cpi613 and shPDHA1, respectively. We found that both approaches triggered morphological changes and characteristics of EMT (increase in mesenchymal markers). This change was accompanied by enhanced wound healing and an increase in migration. Interestingly, cells were more resistant to many of the clinically used chemotherapeutics following PDH inhibition or PDHA1 knockdown. Furthermore, the TGFβRI (known as a major inducer of the EMT) inhibitor (SB-431542) together with the PDHi, was effective in reversing EMT. In conclusion, interfering with PDH induced EMT, and more importantly resulted in chemoresistance. Therefore, our study demonstrates the need for careful consideration of PDH-targeting approaches in cancer treatment.
Collapse
Affiliation(s)
- Buse Cevatemre
- Research Center for Translational Medicine, Koc University, Istanbul, Turkey.,Department of Biology, Uludag University, Bursa, Turkey
| | - Engin Ulukaya
- Department of Clinical Biochemistry, Istinye University Faculty of Medicine, Istanbul, Turkey
| | - Egemen Dere
- Department of Biology, Uludag University, Bursa, Turkey
| | - Sukru Dilege
- School of Medicine, Koc University, Istanbul, Turkey
| | - Ceyda Acilan
- Research Center for Translational Medicine, Koc University, Istanbul, Turkey.,School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
15
|
Wu C, Huang ZH, Meng ZQ, Fan XT, Lu S, Tan YY, You LM, Huang JQ, Stalin A, Ye PZ, Wu ZS, Zhang JY, Liu XK, Zhou W, Zhang XM, Wu JR. A network pharmacology approach to reveal the pharmacological targets and biological mechanism of compound kushen injection for treating pancreatic cancer based on WGCNA and in vitro experiment validation. Chin Med 2021; 16:121. [PMID: 34809653 PMCID: PMC8607619 DOI: 10.1186/s13020-021-00534-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Compound kushen injection (CKI), a Chinese patent drug, is widely used in the treatment of various cancers, especially neoplasms of the digestive system. However, the underlying mechanism of CKI in pancreatic cancer (PC) treatment has not been totally elucidated. METHODS Here, to overcome the limitation of conventional network pharmacology methods with a weak combination with clinical information, this study proposes a network pharmacology approach of integrated bioinformatics that applies a weighted gene co-expression network analysis (WGCNA) to conventional network pharmacology, and then integrates molecular docking technology and biological experiments to verify the results of this network pharmacology analysis. RESULTS The WGCNA analysis revealed 2 gene modules closely associated with classification, staging and survival status of PC. Further CytoHubba analysis revealed 10 hub genes (NCAPG, BUB1, CDK1, TPX2, DLGAP5, INAVA, MST1R, TMPRSS4, TMEM92 and SFN) associated with the development of PC, and survival analysis found 5 genes (TSPOAP1, ADGRG6, GPR87, FAM111B and MMP28) associated with the prognosis and survival of PC. By integrating these results into the conventional network pharmacology study of CKI treating PC, we found that the mechanism of CKI for PC treatment was related to cell cycle, JAK-STAT, ErbB, PI3K-Akt and mTOR signalling pathways. Finally, we found that CDK1, JAK1, EGFR, MAPK1 and MAPK3 served as core genes regulated by CKI in PC treatment, and were further verified by molecular docking, cell proliferation assay, RT-qPCR and western blot analysis. CONCLUSIONS Overall, this study suggests that the optimized network pharmacology approach is suitable to explore the molecular mechanism of CKI in the treatment of PC, which provides a reference for further investigating biomarkers for diagnosis and prognosis of PC and even the clinical rational application of CKI.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhi-Hong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zi-Qi Meng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiao-Tian Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ying-Ying Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Lei-Ming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia-Qi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Antony Stalin
- State Key Laboratory of Subtropical Silviculture, Department of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, 311300, China
| | - Pei-Zhi Ye
- National Cancer Center/National Clinical Research Center for Cancer/Chinese Medicine Department of the Caner Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Shan Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jing-Yuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xin-Kui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wei Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiao-Meng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia-Rui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
16
|
Principe DR, Underwood PW, Korc M, Trevino JG, Munshi HG, Rana A. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma and Barriers to Therapeutic Efficacy. Front Oncol 2021; 11:688377. [PMID: 34336673 PMCID: PMC8319847 DOI: 10.3389/fonc.2021.688377] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, with a median survival time of 10-12 months. Clinically, these poor outcomes are attributed to several factors, including late stage at the time of diagnosis impeding resectability, as well as multi-drug resistance. Despite the high prevalence of drug-resistant phenotypes, nearly all patients are offered chemotherapy leading to modest improvements in postoperative survival. However, chemotherapy is all too often associated with toxicity, and many patients elect for palliative care. In cases of inoperable disease, cytotoxic therapies are less efficacious but still carry the same risk of serious adverse effects, and clinical outcomes remain particularly poor. Here we discuss the current state of pancreatic cancer therapy, both surgical and medical, and emerging factors limiting the efficacy of both. Combined, this review highlights an unmet clinical need to improve our understanding of the mechanisms underlying the poor therapeutic responses seen in patients with PDAC, in hopes of increasing drug efficacy, extending patient survival, and improving quality of life.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Jose G. Trevino
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Hidayatullah G. Munshi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
17
|
Indini A, Rijavec E, Ghidini M, Cortellini A, Grossi F. Targeting KRAS in Solid Tumors: Current Challenges and Future Opportunities of Novel KRAS Inhibitors. Pharmaceutics 2021; 13:pharmaceutics13050653. [PMID: 34064352 PMCID: PMC8147792 DOI: 10.3390/pharmaceutics13050653] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/25/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Activating mutations in RAS family proteins are found in ~25% of all human cancers. Different solid tumors are correlated with mutations in certain isoforms of RAS, with Kirsten RAS (KRAS) being the most frequently mutated isoform. Historically, KRAS has been acknowledged as “undruggable”, largely because the RAS proteins do not appear to present suitable pockets to which small inhibitory molecules can bind. However, this scenario has changed over the last years with the advent of novel KRAS inhibitors. In this review, we describe the role of KRAS mutation across different solid tumors, providing data on novel KRAS inhibitors currently under development and an updated overview of ongoing research in this field. A literature search was performed to select papers, abstracts, and oral presentation on KRAS inhibitory strategies in KRAS mutated solid tumors. Overall, the most promising therapeutic results have been obtained with molecules targeting KRAS G12C, thus paving the way for a significant therapeutic improvement in non-small cell lung cancer. Unfortunately, KRAS G12C mutation is rather uncommon in other solid tumors, namely pancreatic ductal adenocarcinoma and colorectal cancer. Several combination strategies are currently under evaluation in clinical trials, in order to bypass the resistance mechanisms responsible for the intrinsic resistance of mutated KRAS to the main therapeutic strategies adopted to date. Results suggest that the therapeutic scenario of KRAS has started to change, and further research will bring therapeutic results in this field.
Collapse
Affiliation(s)
- Alice Indini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.I.); (E.R.); (M.G.)
| | - Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.I.); (E.R.); (M.G.)
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.I.); (E.R.); (M.G.)
| | - Alessio Cortellini
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, Du Cane Road, London W120NN, UK
| | - Francesco Grossi
- Medical Oncology Unit, Department of Medicine and Surgery, University of Insubria, ASST dei Sette Laghi, 21100 Varese, Italy
- Correspondence: or
| |
Collapse
|
18
|
Inhibition of the Receptor for Advanced Glycation End Products Enhances the Cytotoxic Effect of Gemcitabine in Murine Pancreatic Tumors. Biomolecules 2021; 11:biom11040526. [PMID: 33915939 PMCID: PMC8067004 DOI: 10.3390/biom11040526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a very difficult cancer to treat. Recent in vitro and in vivo studies suggest that the activation of the receptor for advanced glycation end products (RAGE) by its ligands stimulates pancreatic cancer cell proliferation and tumor growth. Additional studies show that, in the RAGE ligand, the high mobility group box 1 (HMGB1) protein plays an important role in chemoresistance against the cytotoxic agent gemcitabine by promoting cell survival through increased autophagy. We hypothesized that blocking the RAGE/HMGB1 interaction would enhance the cytotoxic effect of gemcitabine by reducing cell survival and autophagy. Using a preclinical mouse model of PDAC and a monoclonal antibody (IgG 2A11) as a RAGE inhibitor, we demonstrate that RAGE inhibition concurrent with gemcitabine treatment enhanced the cytotoxic effect of gemcitabine. The combination of IgG 2A11 and gemcitabine resulted in decreased autophagy compared to treatment with gemcitabine combined with control antibodies. Notably, we also observed that RAGE inhibition protected against excessive weight loss during treatment with gemcitabine. Our data suggest that the combination of gemcitabine with a RAGE inhibitor could be a promising therapeutic approach for the treatment of pancreatic cancer and needs to be further investigated.
Collapse
|
19
|
Wang H, Ren R, Yang Z, Cai J, Du S, Shen X. The COL11A1/Akt/CREB signaling axis enables mitochondrial-mediated apoptotic evasion to promote chemoresistance in pancreatic cancer cells through modulating BAX/BCL-2 function. J Cancer 2021; 12:1406-1420. [PMID: 33531986 PMCID: PMC7847647 DOI: 10.7150/jca.47032] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 12/05/2020] [Indexed: 12/27/2022] Open
Abstract
Collagen XI, a member of the collagen family, is present in the extracellular matrix (ECM), and high collagen XI/αI (COL11A1) expression in tumor tissue is reportedly correlated with the clinicopathological parameters of pancreatic ductal adenocarcinoma (PDAC). However, the function of COL11A1 in the development of pancreatic cancer cells remains unclear. In the current study, we assessed mRNA expression of COL11A1 and its receptors and created a testing-model of both a COL11A1-overexpressing tumor microenvironment and/or altered-COL11A1 expression in pancreatic cancer cell lines. Next, we investigated the mechanism by which COL11A1 affects growth, gemcitabine (GEM) resistance and apoptosis in pancreatic cancer cells. We demonstrated that COL11A1 phosphorylated AktSer473, promoting proliferation of cancer cells and inhibiting their apoptosis. Additionally, our data showed that COL11A1/Akt/CREB altered the balance between BCL-2 and BAX and mediated their mitochondrial translocation in pancreatic cancer cells. The COL11A1/Akt axis induced disruption of mitochondrial transmembrane function, enabling mitochondria-mediated apoptotic evasion to promote chemoresistance. We also explored the regulatory effect of COL11A1/Akt on molecular signaling in the mitochondria-mediated apoptotic program. COL11A1/Akt disturbed the BCL-2/BAX balance, inhibiting cytochrome c (Cyt-C) release and binding of Apaf-1/procaspase-9/Cyt-C, which suppressed the apoptotic program and induced GEM resistance in pancreatic cancer cells. In conclusion, COL11A1 modulates apoptotic inhibition and chemoresistance in pancreatic cancer cells by activating the Akt/CREB/BCL-2/BAX signaling pathway. COL11A1 may represent a distinct prognostic indicator and may be an attractive therapeutic target for PDAC.
Collapse
Affiliation(s)
- Hui Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Runling Ren
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Zizhong Yang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jun Cai
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shaoxia Du
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaohong Shen
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
20
|
Ray P, Dutta D, Haque I, Nair G, Mohammed J, Parmer M, Kale N, Orr M, Jain P, Banerjee S, Reindl KM, Mallik S, Kambhampati S, Banerjee SK, Quadir M. pH-Sensitive Nanodrug Carriers for Codelivery of ERK Inhibitor and Gemcitabine Enhance the Inhibition of Tumor Growth in Pancreatic Cancer. Mol Pharm 2020; 18:87-100. [PMID: 33231464 DOI: 10.1021/acs.molpharmaceut.0c00499] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a metabolic disorder, remains one of the leading cancer mortality sources worldwide. An initial response to treatments, such as gemcitabine (GEM), is often followed by emergent resistance reflecting an urgent need for alternate therapies. The PDAC resistance to GEM could be due to ERK1/2 activity. However, successful ERKi therapy is hindered due to low ligand efficiency, poor drug delivery, and toxicity. In this study, to overcome these limitations, we have designed pH-responsive nanoparticles (pHNPs) with a size range of 100-150 nm for the simultaneous delivery of ERKi (SCH 772984) and GEM with tolerable doses. These pHNPs are polyethylene glycol (PEG)-containing amphiphilic polycarbonate block copolymers with tertiary amine side chains. They are systemically stable and capable of improving in vitro and in vivo drug delivery at the cellular environment's acidic pH. The functional analysis indicates that the nanomolar doses of ERKi or GEM significantly decreased the 50% growth inhibition (IC50) of PDAC cells when encapsulated in pHNPs compared to free drugs. The combination of ERKi with GEM displayed a synergistic inhibitory effect. Unexpectedly, we uncover that the minimum effective dose of ERKi significantly promotes GEM activities on PDAC cells. Furthermore, we found that pHNP-encapsulated combination therapy of ERKi with GEM was superior to unencapsulated combination drug therapy. Our findings, thus, reveal a simple, yet efficient, drug delivery approach to overcome the limitations of ERKi for clinical applications and present a new model of sensitization of GEM by ERKi with no or minimal toxicity.
Collapse
Affiliation(s)
- Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Inamul Haque
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Gauthami Nair
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Jiyan Mohammed
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Meredith Parmer
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Narendra Kale
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Megan Orr
- Department of Statistics, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Pooja Jain
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Katie M Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Suman Kambhampati
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| |
Collapse
|
21
|
Li M, Song SW, Ge Y, Jin JY, Li XY, Tan XD. The Ras-ERK signaling pathway regulates acetylated activating transcription factor 2 via p300 in pancreatic cancer cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1234. [PMID: 33178766 PMCID: PMC7607129 DOI: 10.21037/atm-20-5880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Activating transcription factor 2 (ATF2) regulates the expression of downstream target genes and is phosphorylated by the Ras-extracellular-signal-regulated kinase (ERK) pathway. Acetylation of ATF2 is necessary for this type of regulation. However, the molecular mechanism by which the Ras-ERK pathway mediates the regulation of acetylated ATF2 is unknown. This study investigates the mechanism of Ras-ERK pathway-mediated regulation of acetylated ATF2 in maintaining the characteristic phenotype of pancreatic cancer cells. Methods This study was carried out using ASPC-1 and BXPC-3 pancreatic cancer cell lines transfected with the double mutant RasG12V/T35S. The levels of phosphorylated ERK1/2 were measured to establish the activated Ras-ERK pathway. The regulation of acetylated ATF2 was examined by detecting the protein level using western blotting, and the effects on cancer cell phenotype were measured using cell viability, proliferation, migration, and apoptosis assays. Also, chromatin immunoprecipitation (ChIP) assays were used to measure the effect on respective downstream target genes. Results The results showed that RasG12V/T35S reduced the level of acetylated ATF2 in ASPC-1 and BXPC-3 cells. Compared to wild-type ATF2, the mutant ATF2K357Q (which mimics the irreversible acetylated form of ATF2) reduced the cancer cell phenotype and showed decreased enrichment on target genes upon transfection with Ras. Moreover, the level of acetylated ATF2 was regulated by the degradation of p300 through E3 ubiquitin ligase mouse double minute 2 homolog (MDM2). Conclusions Activation of the Ras-ERK pathway regulates acetylated ATF2 through degradation of p300 via a proteasome-dependent pathway, which alters the transcription of downstream target genes responsible for the cancer cell phenotype.
Collapse
Affiliation(s)
- Mu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shao-Wei Song
- Department of General Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Ge
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun-Yi Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Ying Li
- Department of General Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
22
|
Das M, Li J, Bao M, Huang L. Nano-delivery of Gemcitabine Derivative as a Therapeutic Strategy in a Desmoplastic KRAS Mutant Pancreatic Cancer. AAPS J 2020; 22:88. [PMID: 32572645 DOI: 10.1208/s12248-020-00467-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma remains one of the challenging malignancies to treat, and chemotherapy is the primary treatment strategy available to most patients. Gemcitabine, one of the oldest chemotherapeutic drugs approved for pancreatic cancer, has limited efficacy, due to low drug distribution to the tumor and chemoresistance following therapy. In this study, we delivered gemcitabine monophosphate using lipid calcium phosphate nanoparticles, to desmoplastic pancreatic tumors. Monophosphorylation is a critical, rate-limiting step following cellular uptake of gemcitabine and precursor of the pharmacologically active gemcitabine triphosphate. Our drug delivery strategy enabled us to achieve robust tumor regression with a low parenteral dose in a clinically relevant, KRAS mutant, syngeneic orthotopic allograft, lentivirus-transfected KPC cell line-derived model of pancreatic cancer. Treatment with gemcitabine monophosphate significantly increased apoptosis of cancer cells, enabled reduction in the proportion of immunosuppressive tumor-associated macrophages and myeloid-derived suppressor cells, and did not increase expression of cancer stem cell markers. Overall, we could trigger a strong antitumor response in a treatment refractory PDAC model, while bypassing critical hallmarks of gemcitabine chemoresistance.
Collapse
Affiliation(s)
- Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Jun Li
- Qualiber Inc., Durham, North Carolina, 27705, USA
- ZY Therapeutics, Research Triangle Park, North Carolina, 27709, USA
| | - Michelle Bao
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
- North Carolina School of Science and Mathematics, Durham, North Carolina, 27705, USA
- Stanford University, Stanford, California, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA.
| |
Collapse
|
23
|
(3E,5E)-3,5-Bis(pyridin-3-methylene)-tetrahydrothiopyran-4-one enhances the inhibitory effect of gemcitabine on pancreatic cancer cells. Bioorg Chem 2020; 101:104022. [PMID: 32599367 DOI: 10.1016/j.bioorg.2020.104022] [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: 03/24/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Gemcitabine (GEM) is a commonly used treatment for advanced pancreatic cancer. However, chemoresistance and toxic side effect limits its clinical success. In an earlier study, our laboratory found that the curcumin analogue, (3E,5E)-3,5-Bis(pyridin-3-methylene)-tetrahydrothiopyran-4-one (FN2) had strong inhibitory effect on human pancreatic cancer cells. In the present study, we investigated the effects of FN2 in combination with GEM on growth inhibition and apoptosis in human pancreatic cancer Panc-1 cells. The results showed that the combination of FN2 and GEM synergistically inhibited the growth of Panc-1 cells. Panc-1 cells survived the GEM treatment became partially resistant to the drug. Treatment with FN2 in combination with GEM strongly inhibited the growth and stimulated apoptosis in the GEM resistant Panc-1 cells. Mechanistic studies showed that inhibition of cell growth and induction of apoptosis in the GEM resistant Panc-1 cells were associated with decreases in activation of NF-κB and Akt. FN2 in combination with GEM also decreased the level of Bcl-2 and increased the level of Bax. Results of the present study indicate that GEM in combination with FN2 may represent an effective strategy for improving the efficacy of GEM and decreasing the resistance of pancreatic cancer to GEM chemotherapy.
Collapse
|
24
|
The Ras-ERK1/2 signaling pathway regulates H3K9ac through PCAF to promote the development of pancreatic cancer. Life Sci 2020; 256:117936. [PMID: 32531376 DOI: 10.1016/j.lfs.2020.117936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 05/21/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
AIMS The regulation of the Ras-ERK pathway is the crucial point in pancreatic carcinogenesis, and the Ras kinase is an essential regulatory upstream signal molecule of the ERK1/2 pathway. H3K9ac is a vital histone modification, but its specific role in pancreatic cancer remains unclear. This research aims to study whether the modification level of H3K9ac can regulate the characteristic phenotype of the pancreatic cancer cells by affecting the downstream expression, proliferation, migration, and other related genes. MAIN METHODS The RasG12V/T35S were used to transfect pancreatic cancer cells, and the levels of phosphorylated ERK1/2 and H3K9ac were detected by western blotting. The colony formation assay, transwell assay, and chromatin immunoprecipitation assay were used to study cell viability, migration, and the downstream genes of the ERK1/2 pathway. KEY FINDINGS The results showed that Ras ERK1/2 reduced H3K9ac expression in ASPC-1 cells, and H3K9ac significantly repressed the viability of cells, colony formation, and ASPC-1 cell movement induced by Ras ERK1/2. Besides, HDAC1 silencing increased H3K9ac expression, and changed the effect of Ras ERK1/2 on ASPC-1 cells proliferation, its movement, and mRNAs of ERK1/2 downstream genes. Moreover, Ras ERK1/2 inhibited H3K9ac expression by the degradation of PCAF via MDM2. SIGNIFICANCE Ras ERK1/2 promotes pancreatic carcinogenesis cell movement, through down-regulating H3K9ac via MDM2 mediated PCAF degradation.
Collapse
|
25
|
An EJ, Kim Y, Lee SH, Ko HM, Chung WS, Jang HJ. Anti-Cancer Potential of Oxialis obtriangulata in Pancreatic Cancer Cell through Regulation of the ERK/Src/STAT3-Mediated Pathway. Molecules 2020; 25:molecules25102301. [PMID: 32422890 PMCID: PMC7288118 DOI: 10.3390/molecules25102301] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 01/09/2023] Open
Abstract
As a plant medicine, Oxalidaceae has been used to treat various diseases in Korea. However, there is little data on the anti-cancer efficacy of Oxalidaceae, particularly O. obtriangulata. This study aimed to investigate the anti-cancer effect of O. obtriangulata methanol extract (OOE) and its regulatory actions on pancreatic carcinoma. OOE showed anti-proliferative effects and induced cell death in the colony formation and cell viability assays, respectively. The Fluorescence-activated cell sorting (FACS) data confirmed that OOE significantly induced cell cycle accumulation at the G2/M phase and apoptotic effects. Additionally, OOE inhibited the activated ERK (extracellular-signal-regulated kinase)/Src (Proto-oncogene tyrosine-protein kinase Src)/STAT3 (signal transducers and activators of transcription 3) pathways including nuclear translocation of STAT3. Furthermore, suppression of Ki67, PARP(Poly ADP-ribose polymerase), caspase-3, P27(Cyclin-dependent kinase inhibitor 1B), and c-Myc as well as the STAT3 target genes CDK(cyclin-dependent kinase)1, CDK2, Cyclin B1, VEGF-1(vascular endothelial growth factor-1), MMP-9(Matrix metallopeptidase 9), and Survivin by OOE was observed in BxPC3. We speculate that these molecular actions might support an anti-cancer effect of OOE. In this study, we demonstrated that OOE may be a promising anti-cancer material and may serve as a natural therapy and alternative remedy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Eun-Jin An
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Yumi Kim
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Seung-Hyeon Lee
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Hyun Min Ko
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Won-Seok Chung
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Correspondence: (W.-S.C.); (H.-J.J.)
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- College of Korean Medicine and College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: (W.-S.C.); (H.-J.J.)
| |
Collapse
|
26
|
Afrin F, Chi M, Eamens AL, Duchatel RJ, Douglas AM, Schneider J, Gedye C, Woldu AS, Dun MD. Can Hemp Help? Low-THC Cannabis and Non-THC Cannabinoids for the Treatment of Cancer. Cancers (Basel) 2020; 12:cancers12041033. [PMID: 32340151 PMCID: PMC7226605 DOI: 10.3390/cancers12041033] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Cannabis has been used to relieve the symptoms of disease for thousands of years. However, social and political biases have limited effective interrogation of the potential benefits of cannabis and polarised public opinion. Further, the medicinal and clinical utility of cannabis is limited by the psychotropic side effects of ∆9-tetrahydrocannabinol (∆9-THC). Evidence is emerging for the therapeutic benefits of cannabis in the treatment of neurological and neurodegenerative diseases, with potential efficacy as an analgesic and antiemetic for the management of cancer-related pain and treatment-related nausea and vomiting, respectively. An increasing number of preclinical studies have established that ∆9-THC can inhibit the growth and proliferation of cancerous cells through the modulation of cannabinoid receptors (CB1R and CB2R), but clinical confirmation remains lacking. In parallel, the anti-cancer properties of non-THC cannabinoids, such as cannabidiol (CBD), are linked to the modulation of non-CB1R/CB2R G-protein-coupled receptors, neurotransmitter receptors, and ligand-regulated transcription factors, which together modulate oncogenic signalling and redox homeostasis. Additional evidence has also demonstrated the anti-inflammatory properties of cannabinoids, and this may prove relevant in the context of peritumoural oedema and the tumour immune microenvironment. This review aims to document the emerging mechanisms of anti-cancer actions of non-THC cannabinoids.
Collapse
Affiliation(s)
- Farjana Afrin
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Mengna Chi
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Andrew L. Eamens
- Centre for Plant Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia;
| | - Ryan J. Duchatel
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Alicia M. Douglas
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
| | - Jennifer Schneider
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Priority Research Centre for Chemical Biology and Clinical Pharmacology, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Craig Gedye
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Calvary Mater Newcastle, Waratah, NSW 2298, Australia
| | - Ameha S. Woldu
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Correspondence: (A.S.W.); (M.D.D.); Tel.: +61-02-4921-7807 (A.S.W.); +61-02-4921-5693 (M.D.D.)
| | - Matthew D. Dun
- Cancer Signalling Research Group, Medical Biochemistry, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; (F.A.); (M.C.); (R.J.D.); (A.M.D.); (C.G.)
- Priority Research Centre for Cancer Research, Innovation & Translation, Faculty of Health and Medicine, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia;
- Correspondence: (A.S.W.); (M.D.D.); Tel.: +61-02-4921-7807 (A.S.W.); +61-02-4921-5693 (M.D.D.)
| |
Collapse
|
27
|
Buscail L, Bournet B, Cordelier P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev Gastroenterol Hepatol 2020; 17:153-168. [PMID: 32005945 DOI: 10.1038/s41575-019-0245-4] [Citation(s) in RCA: 448] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is predicted to be the second most common cause of death within the next 10 years. The prognosis for this disease is poor despite diagnostic progress and new chemotherapeutic regimens. The oncogenic KRAS mutation is the major event in pancreatic cancer; it confers permanent activation of the KRAS protein, which acts as a molecular switch to activate various intracellular signalling pathways and transcription factors inducing cell proliferation, migration, transformation and survival. Several laboratory methods have been developed to detect KRAS mutations in biological samples, including digital droplet PCR (which displays high sensitivity). Clinical studies have revealed that a KRAS mutation assay in fine-needle aspiration material combined with cytopathology increases the sensitivity, accuracy and negative predictive value of cytopathology for a positive diagnosis of pancreatic cancer. In addition, the presence of KRAS mutations in serum and plasma (liquid biopsies) correlates with a worse prognosis. The presence of mutated KRAS can also have therapeutic implications, whether at the gene level per se, during its post-translational maturation, interaction with nucleotides and after activation of the various oncogenic signals. Further pharmacokinetic and toxicological studies on new molecules are required, especially small synthetic molecules, before they can be used in the therapeutic arsenal for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Louis Buscail
- Department of Gastroenterology, University of Toulouse III, Rangueil Hospital, Toulouse, France. .,INSERM UMR 1037, Toulouse Centre for Cancer Research, University of Toulouse III, Toulouse, France.
| | - Barbara Bournet
- Department of Gastroenterology, University of Toulouse III, Rangueil Hospital, Toulouse, France.,INSERM UMR 1037, Toulouse Centre for Cancer Research, University of Toulouse III, Toulouse, France
| | - Pierre Cordelier
- INSERM UMR 1037, Toulouse Centre for Cancer Research, University of Toulouse III, Toulouse, France
| |
Collapse
|
28
|
Steckiewicz KP, Barcinska E, Sobczak K, Tomczyk E, Wojcik M, Inkielewicz-Stepniak I. Assessment of Anti-Tumor potential and safety of application of Glutathione stabilized Gold Nanoparticles conjugated with Chemotherapeutics. Int J Med Sci 2020; 17:824-833. [PMID: 32218704 PMCID: PMC7085271 DOI: 10.7150/ijms.40827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/08/2020] [Indexed: 12/24/2022] Open
Abstract
Due to the high toxicity of currently used chemotherapeutics, novel methods of cancer treatment are needed. Gold nanoparticles (AuNPs) seem to be an interesting alternative due to penetration through biological membranes and systemic barriers. AuNPs as carriers of chemotherapeutics allow for reduced concentrations whilst maintaining the expected effect, and thus reducing the costs of therapy and adverse effects. We synthesized AuNPs stabilized with reduced glutathione (GSH) and conjugated with doxorubicin (DOX), gemcitabine (GEM) or cytarabine (CTA). This is the first study in which cytarabine-AuNPs were synthesized and characterized. Transmission electron microscopy (TEM), thermogravimetric analysis (TGA), nuclear magnetic resonance spectroscopy (NMR) and high-performance liquid chromatography (HPLC) were used to chemically characterize obtained nanoparticles. Antitumor activity and safety of application were assessed by MTT assay in in vitro model (human osteosarcoma cells -143B, human osteoblast- hFOB1.19, breast cancer cells - MCF7, breast epithelial cells - MCF10A, pancreatic cancer cells - PANC-1, and pancreatic cells - hTERT-HPNE cells). We have shown that cellular response varies according to the type and concentration of AuNPs. At some concentrations, we were able to show selective cytotoxicity of our AuNPs conjugates only to cancer cell lines. Synthesized nanoparticles were more cytotoxic to tumor cell lines than chemotherapeutics alone.
Collapse
Affiliation(s)
- Karol P Steckiewicz
- Chair and Department of Medical Chemistry, Medical University of Gdansk, Debinki street 1, 80-211 Gdansk, Poland
| | - Ewelina Barcinska
- Chair and Department of Medical Chemistry, Medical University of Gdansk, Debinki street 1, 80-211 Gdansk, Poland
| | - Katarzyna Sobczak
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Ewelina Tomczyk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Michał Wojcik
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Iwona Inkielewicz-Stepniak
- Chair and Department of Medical Chemistry, Medical University of Gdansk, Debinki street 1, 80-211 Gdansk, Poland
| |
Collapse
|
29
|
Chen CH, Hsieh TH, Lin YC, Liu YR, Liou JP, Yen Y. Targeting Autophagy by MPT0L145, a Highly Potent PIK3C3 Inhibitor, Provides Synergistic Interaction to Targeted or Chemotherapeutic Agents in Cancer Cells. Cancers (Basel) 2019; 11:cancers11091345. [PMID: 31514441 PMCID: PMC6770340 DOI: 10.3390/cancers11091345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Anticancer therapies reportedly promote pro-survival autophagy in cancer cells that confers drug resistance, rationalizing the concept to combine autophagy inhibitors to increase their therapeutic potential. We previously identified that MPT0L145 is a PIK3C3/FGFR inhibitor that not only increases autophagosome formation due to fibroblast growth factor receptor (FGFR) inhibition but also perturbs autophagic flux via PIK3C3 inhibition in bladder cancer cells harboring FGFR activation. In this study, we hypothesized that combined-use of MPT0L145 with agents that induce pro-survival autophagy may provide synthetic lethality in cancer cells without FGFR activation. The results showed that MPT0L145 synergistically sensitizes anticancer effects of gefitinib and gemcitabine in non-small cell lung cancer A549 cells and pancreatic cancer PANC-1 cells, respectively. Mechanistically, drug combination increased incomplete autophagy due to impaired PIK3C3 function by MPT0L145 as evidenced by p62 accumulation and no additional apoptotic cell death was observed. Meanwhile, drug combination perturbed survival pathways and increased vacuolization and ROS production in cancer cells. In conclusion, the data suggest that halting pro-survival autophagy by targeting PIK3C3 with MPT0L145 significantly sensitizes cancer cells to targeted or chemotherapeutic agents, fostering rational combination strategies for cancer therapy in the future.
Collapse
Affiliation(s)
- Chun-Han Chen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Chen Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Yun Yen
- The Ph.D. Program for Cancer Molecular Biology and drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University Taipei 110, Taiwan.
| |
Collapse
|
30
|
Amrutkar M, Aasrum M, Verbeke CS, Gladhaug IP. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer 2019; 19:596. [PMID: 31208372 PMCID: PMC6580453 DOI: 10.1186/s12885-019-5803-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
Background Gemcitabine remains a cornerstone in chemotherapy of pancreatic ductal adenocarcinoma (PDAC) despite suboptimal clinical effects that are partly due to the development of chemoresistance. Pancreatic stellate cells (PSCs) of the tumor stroma are known to interact with pancreatic cancer cells (PCCs) and influence the progression of PDAC through a complex network of signaling molecules that involve extracellular matrix (ECM) proteins. To understand tumor-stroma interactions regulating chemosensitivity, the role of PSC-secreted fibronectin (FN) in the development of gemcitabine resistance in PDAC was examined. Methods PSC cultures obtained from ten different human PDAC tumors were co-cultured with PCC lines (AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990) either directly, or indirectly via incubation with PSC-conditioned medium (PSC-CM). Gemcitabine dose response cytotoxicity was determined using MTT based cell viability assays. Protein expression was assessed by western blotting and immunofluorescence. PSC-CM secretome analysis was performed by proteomics-based LC-MS/MS, and FN content in PSC-CM was determined with ELISA. Radiolabeled gemcitabine was used to determine the capacity of PCCs to uptake the drug. Results In both direct and indirect co-culture, PSCs induced varying degrees of resistance to the cytotoxic effects of gemcitabine among all cancer cell lines examined. A variable degree of increased phosphorylation of ERK1/2 was observed across all PCC lines upon incubation with PSC-CM, while activation of AKT was not detected. Secretome analysis of PSC-CM identified 796 different proteins, including several ECM-related proteins such as FN and collagens. Soluble FN content in PSC-CM was detected in the range 175–350 ng/ml. Neither FN nor PSC-CM showed any effect on PCC uptake capacity of gemcitabine. PCCs grown on FN-coated surface displayed higher resistance to gemcitabine compared to cells grown on non-coated surface. Furthermore, a FN inhibitor, synthetic Arg-Gly-Asp-Ser (RGDS) peptide significantly inhibited PSC-CM-induced chemoresistance in PCCs via downregulation of ERK1/2 phosphorylation. Conclusions The findings of this study suggest that FN secreted by PSCs in the ECM plays a key role in the development of resistance to gemcitabine via activation of ERK1/2. FN-blocking agents added to gemcitabine-based chemotherapy might counteract chemoresistance in PDAC and provide better clinical outcomes. Electronic supplementary material The online version of this article (10.1186/s12885-019-5803-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway. .,Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway.
| | - Monica Aasrum
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway.,Department of Pathology, Oslo University Hospital Rikshospitalet, Nydalen, 0424, Oslo, Norway
| | - Ivar P Gladhaug
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway.,Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital Rikshospitalet, Nydalen, 0424, Oslo, Norway
| |
Collapse
|
31
|
Ferro R, Adamska A, Lattanzio R, Mavrommati I, Edling CE, Arifin SA, Fyffe CA, Sala G, Sacchetto L, Chiorino G, De Laurenzi V, Piantelli M, Sansom OJ, Maffucci T, Falasca M. GPR55 signalling promotes proliferation of pancreatic cancer cells and tumour growth in mice, and its inhibition increases effects of gemcitabine. Oncogene 2018; 37:6368-6382. [PMID: 30061636 DOI: 10.1038/s41388-018-0390-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 04/19/2018] [Accepted: 06/02/2018] [Indexed: 12/28/2022]
Abstract
The life expectancy for pancreatic cancer patients has seen no substantial changes in the last 40 years as very few and mostly just palliative treatments are available. As the five years survival rate remains around 5%, the identification of novel pharmacological targets and development of new therapeutic strategies are urgently needed. Here we demonstrate that inhibition of the G protein-coupled receptor GPR55, using genetic and pharmacological approaches, reduces pancreatic cancer cell growth in vitro and in vivo and we propose that this may represent a novel strategy to inhibit pancreatic ductal adenocarcinoma (PDAC) progression. Specifically, we show that genetic ablation of Gpr55 in the KRASWT/G12D/TP53WT/R172H/Pdx1-Cre+/+ (KPC) mouse model of PDAC significantly prolonged survival. Importantly, KPC mice treated with a combination of the GPR55 antagonist Cannabidiol (CBD) and gemcitabine (GEM, one of the most used drugs to treat PDAC), survived nearly three times longer compared to mice treated with vehicle or GEM alone. Mechanistically, knockdown or pharmacologic inhibition of GPR55 reduced anchorage-dependent and independent growth, cell cycle progression, activation of mitogen-activated protein kinase (MAPK) signalling and protein levels of ribonucleotide reductases in PDAC cells. Consistent with this, genetic ablation of Gpr55 reduced proliferation of tumour cells, MAPK signalling and ribonucleotide reductase M1 levels in KPC mice. Combination of CBD and GEM inhibited tumour cell proliferation in KPC mice and it opposed mechanisms involved in development of resistance to GEM in vitro and in vivo. Finally, we demonstrate that the tumour suppressor p53 regulates GPR55 protein expression through modulation of the microRNA miR34b-3p. Our results demonstrate the important role played by GPR55 downstream of p53 in PDAC progression. Moreover our data indicate that combination of CBD and GEM, both currently approved for medical use, might be tested in clinical trials as a novel promising treatment to improve PDAC patients' outcome.
Collapse
Affiliation(s)
- R Ferro
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - A Adamska
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia
| | - R Lattanzio
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti, 66100, Italy
| | - I Mavrommati
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - C E Edling
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - S A Arifin
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - C A Fyffe
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - G Sala
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti, 66100, Italy
| | - L Sacchetto
- Cancer Genomics Laboratory, Fondazione Edo and Elvo Tempia, Biella, Italy
| | - G Chiorino
- Cancer Genomics Laboratory, Fondazione Edo and Elvo Tempia, Biella, Italy
| | - V De Laurenzi
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti, 66100, Italy
| | - M Piantelli
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Centro Studi sull'Invecchiamento, CeSI-MeT, Chieti, 66100, Italy
| | - O J Sansom
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| | - T Maffucci
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK
| | - M Falasca
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, 4 Newark Street, London, E1 2AT, UK.
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia.
| |
Collapse
|
32
|
Qian CJ, Qi YX, Zhong S, Zeng JP, Chen XY, Yao J. Mitogen-activated protein kinase inhibition enhances the antitumor effects of sporamin in human pancreatic cancer cells. Oncol Lett 2018; 16:1237-1242. [PMID: 30061945 DOI: 10.3892/ol.2018.8746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/17/2018] [Indexed: 01/07/2023] Open
Abstract
Sporamin, a sweet potato tuber storage protein, is a Kunitz-type trypsin inhibitor (TI) that has exhibited antitumor activity through poorly defined mechanisms in a number of types of tumor cells. The present study aimed to analyze the combined effects of sporamin and three mitogen-activated protein kinase (MAPK) inhibitors, PD98059, SP600125 and SB203580, on the pancreatic cancer cell line, PANC-1. Cell proliferation activity was assessed using a 3H-thymidine incorporation assay, and cell viability was analyzed using an MTT assay. Apoptosis was assayed by flow cytometry and fluorescence microscopy. Protein expression levels in PANC-1 cells were determined by western blotting. The results of this analysis demonstrated that sporamin induced a temporary increase in the phosphorylation of MAPKs, including phosphorylated extracellular signal regulated-kinase 1/2, phosphorylated c-Jun amino-terminal protein kinase 1/2 and phosphorylated p38-MAPK, in a concentration-dependent manner. However, treatment with MAPK inhibitors promoted the inhibition of cell proliferation and viability, and the induction of apoptosis in sporamin-treated PANC-1 cells. In conclusion, the present study demonstrated that MAPK inhibition enhanced the antitumor activity of sporamin in PANC-1 cells.
Collapse
Affiliation(s)
- Cui-Juan Qian
- Department of Gastroenterology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang 318000, P.R. China.,Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Yong-Xiao Qi
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Sheng Zhong
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Ju-Ping Zeng
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xiao-Ying Chen
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jun Yao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
33
|
Adamska A, Elaskalani O, Emmanouilidi A, Kim M, Abdol Razak NB, Metharom P, Falasca M. Molecular and cellular mechanisms of chemoresistance in pancreatic cancer. Adv Biol Regul 2018; 68:77-87. [PMID: 29221990 DOI: 10.1016/j.jbior.2017.11.007] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 02/07/2023]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the most chemoresistant cancers, and current therapies targeting cancer-associated molecular pathways have not given satisfactory results, owing in part to rapid upregulation of alternative compensatory pathways. Most of the available treatments are palliative, focussing on improving the quality of life. At present, available options are surgery, embolization, radiation, chemotherapy, immunotherapy and use of other more targeted drugs. In this review, we describe the cellular and molecular effects of current chemotherapy drugs such as gemcitabine, FOLFIRINOX (5-fluorouracil [5-FU], oxaliplatin, irinotecan, and leucovorin) and ABRAXANE (nab-Paclitaxel), which have shown a survival benefit, although modest, for pancreatic cancer patients. Nevertheless, gemcitabine remains the standard first-line option for advanced-stage pancreatic cancer patients and, as resistance to the drug has attracted an increasing scientific interest, we deliberate on the main intracellular processes and proteins vital in acquired chemoresistance to gemcitabine. Lastly, our review examines various microenvironmental factors capable of instigating PDAC to develop resistance to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Aleksandra Adamska
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Omar Elaskalani
- Platelet Research Laboratory, Curtin Health Innovation and Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Aikaterini Emmanouilidi
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Minkyoung Kim
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Norbaini Binti Abdol Razak
- Platelet Research Laboratory, Curtin Health Innovation and Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Pat Metharom
- Platelet Research Laboratory, Curtin Health Innovation and Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
34
|
Schambach A, Schott JW, Morgan MA. Uncoupling the Oncogenic Engine. Cancer Res 2017; 77:6060-6064. [PMID: 29097608 DOI: 10.1158/0008-5472.can-17-2362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 08/31/2017] [Accepted: 09/13/2017] [Indexed: 11/16/2022]
Abstract
Inhibition of oncogenic signaling and correction of aberrant metabolic processes may be key paradigms to eliminate cancer cells. The high incidence of activating RAS mutations and hyperactivated ERK1/2 signaling observed in many human tumors and the lack of effective targeted therapies to elicit long-term inhibition of the RAS-ERK1/2 signaling pathway add to the importance of discovering novel strategies to treat malignancies characterized by elevated RAS-ERK1/2 signaling. In this review, we describe connections between oncogenic signaling and cancer cell metabolism and how these links may be exploited for novel modern molecular medicine approaches. Cancer Res; 77(22); 6060-4. ©2017 AACR.
Collapse
Affiliation(s)
- Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Juliane W Schott
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Michael A Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany. .,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
35
|
Elaskalani O, Falasca M, Moran N, Berndt MC, Metharom P. The Role of Platelet-Derived ADP and ATP in Promoting Pancreatic Cancer Cell Survival and Gemcitabine Resistance. Cancers (Basel) 2017; 9:cancers9100142. [PMID: 29064388 PMCID: PMC5664081 DOI: 10.3390/cancers9100142] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022] Open
Abstract
Platelets have been demonstrated to be vital in cancer epithelial-mesenchymal transition (EMT), an important step in metastasis. Markers of EMT are associated with chemotherapy resistance. However, the association between the development of chemoresistance, EMT, and the contribution of platelets to the process, is still unclear. Here we report that platelets regulate the expression of (1) human equilibrative nucleoside transporter 1 (hENT1) and (2) cytidine deaminase (CDD), markers of gemcitabine resistance in pancreatic cancer. Human ENT1 (hENT1) is known to enable cellular uptake of gemcitabine while CDD deactivates gemcitabine. Knockdown experiments demonstrate that Slug, a mesenchymal transcriptional factor known to be upregulated during EMT, regulates the expression of hENT1 and CDD. Furthermore, we demonstrate that platelet-derived ADP and ATP regulate Slug and CDD expression in pancreatic cancer cells. Finally, we demonstrate that pancreatic cancer cells express the purinergic receptor P2Y12, an ADP receptor found mainly on platelets. Thus ticagrelor, a P2Y12 inhibitor, was used to examine the potential therapeutic effect of an ADP receptor antagonist on cancer cells. Our data indicate that ticagrelor negated the survival signals initiated in cancer cells by platelet-derived ADP and ATP. In conclusion, our results demonstrate a novel role of platelets in modulating chemoresistance in pancreatic cancer. Moreover, we propose ADP/ATP receptors as additional potential drug targets for treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Omar Elaskalani
- Platelet Research Laboratory, School of Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia.
| | - Marco Falasca
- Metabolic Signalling Group, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia.
| | - Niamh Moran
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Michael C Berndt
- Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia.
| | - Pat Metharom
- Platelet Research Laboratory, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia.
| |
Collapse
|
36
|
Engineered Resistant-Starch (ERS) Diet Shapes Colon Microbiota Profile in Parallel with the Retardation of Tumor Growth in In Vitro and In Vivo Pancreatic Cancer Models. Nutrients 2017; 9:nu9040331. [PMID: 28346394 PMCID: PMC5409670 DOI: 10.3390/nu9040331] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 12/15/2022] Open
Abstract
Background/aims: Pancreatic cancer (PC) is ranked as the fourth leading cause of cancer-related deaths worldwide. Despite recent advances in treatment options, a modest impact on the outcome of the disease is observed so far. We have previously demonstrated that short-term fasting cycles have the potential to improve the efficacy of chemotherapy against PC. The aim of this study was to assess the effect of an engineered resistant-starch (ERS) mimicking diet on the growth of cancer cell lines in vitro, on the composition of fecal microbiota, and on tumor growth in an in vivo pancreatic cancer mouse xenograft model. Materials and Methods: BxPC-3, MIA PaCa-2 and PANC-1 cells were cultured in the control, and in the ERS-mimicking diet culturing condition, to evaluate tumor growth and proliferation pathways. Pancreatic cancer xenograft mice were subjected to an ERS diet to assess tumor volume and weight as compared to mice fed with a control diet. The composition and activity of fecal microbiota were further analyzed in growth experiments by isothermal microcalorimetry. Results: Pancreatic cancer cells cultured in an ERS diet-mimicking medium showed decreased levels of phospho-ERK1/2 (extracellular signal-regulated kinase proteins) and phospho-mTOR (mammalian target of rapamycin) levels, as compared to those cultured in standard medium. Consistently, xenograft pancreatic cancer mice subjected to an ERS diet displayed significant retardation in tumor growth. In in vitro growth experiments, the fecal microbial cultures from mice fed with an ERS diet showed enhanced growth on residual substrates, higher production of formate and lactate, and decreased amounts of propionate, compared to fecal microbiota from mice fed with the control diet. Conclusion: A positive effect of the ERS diet on composition and metabolism of mouse fecal microbiota shown in vitro is associated with the decrease of tumor progression in the in vivo PC xenograft mouse model. These results suggest that engineered dietary interventions could be supportive as a synergistic approach to enhance the efficacy of existing cancer treatments in pancreatic cancer patients.
Collapse
|
37
|
Yang X, Xu Y, Wang T, Shu D, Guo P, Miskimins K, Qian SY. Inhibition of cancer migration and invasion by knocking down delta-5-desaturase in COX-2 overexpressed cancer cells. Redox Biol 2017; 11:653-662. [PMID: 28157665 PMCID: PMC5288391 DOI: 10.1016/j.redox.2017.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
We recently reported that knockdown of delta-5-desaturase (a key enzyme that converts dihomo-γ-linolenic acid, DGLA, to the downstream ω-6 arachidonic acid) promotes formation of an anti-cancer byproduct 8-hydroxyoctanoic acid from cyclooxygenase (COX)-catalyzed DGLA peroxidation. 8-hydroxyoctanoic acid can exert its growth inhibitory effect on cancer cells (e.g. colon and pancreatic cancer) by serving as a histone deacetylase inhibitor. Since histone deacetylase inhibitors have been well-known to suppress cancer cell migration and invasion, we thus tested whether knockdown of delta-5-desaturase and DGLA treatment could also be used to inhibit cancer migration and invasion of colon cancer and pancreatic cancer cells. Wound healing assay, transwell assay and western blot were used to assess cell migration and invasion as well as the associated molecular mechanisms. Formation of threshold level of 8-hydroxyoctanoic acid was quantified from COX-catalyzed DGLA peroxidation in the cancer cells that overexpress COX-2 and their delta-5-desaturases were knocked down by shRNA transfection. Our results showed that knockdown of delta-5-desaturase along with DGLA supplement not only significantly inhibited cell migration, but also improved the efficacies of 5-flurouracil and gemcitabine, two frontline chemotherapy drugs currently used in the treatment of colon and pancreatic cancer, respectively. The molecular mechanism behind these observations is that 8-hydroxyoctanoic acid inhibits histone deacetylase, resulting in downregulation of cancer metastasis promotors, e.g., MMP-2 and MMP-9 as well as upregulation of cancer metastasis suppressor, e.g. E-cadherin. For the first time, we demonstrated that we could take the advantage of the common phenomenon of COX-2 overexpression in cancers to inhibit cancer cell migration and invasion. With the shifting paradigm of COX-2 cancer biology, our research outcome may provide us a novel cancer treatment strategy.
High level of COX-2 could be used to inhibit cancer cell migration and invasion. 8-hydroxyoctanoic acid suppresses cancer migration and invasion via inhibiting HDAC. D5D knockdown and DGLA improves efficacy of chemotherapy to inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Yi Xu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Tao Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Dan Shu
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, and College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, and College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Keith Miskimins
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA.
| |
Collapse
|
38
|
Elaskalani O, Razak NBA, Falasca M, Metharom P. Epithelial-mesenchymal transition as a therapeutic target for overcoming chemoresistance in pancreatic cancer. World J Gastrointest Oncol 2017; 9:37-41. [PMID: 28144398 PMCID: PMC5241525 DOI: 10.4251/wjgo.v9.i1.37] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/25/2016] [Accepted: 11/22/2016] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer has one of the worst prognoses among all cancers due to the late manifestation of identifiable symptoms and high resistance to chemo- and radiation therapies. In recent years, a cancer development phase termed epithelial-mesenchymal transition (EMT) has gained increasing research focus. The process is implicated in tumour metastasis, and emerging evidence suggests EMT also contributes or induces chemoresistance in several cancers. Nevertheless, the applicability of therapeutic targeting of EMT faces many challenges. In this mini-review, we summarise the evidence supporting the role of EMT in pancreatic cancer progression, focusing particularly on its association with chemoresistance.
Collapse
|
39
|
The cornerstone K-RAS mutation in pancreatic adenocarcinoma: From cell signaling network, target genes, biological processes to therapeutic targeting. Crit Rev Oncol Hematol 2017; 111:7-19. [PMID: 28259298 DOI: 10.1016/j.critrevonc.2017.01.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/15/2016] [Accepted: 01/05/2017] [Indexed: 01/17/2023] Open
Abstract
RAS belongs to the super family of small G proteins and plays crucial roles in signal transduction from membrane receptors in the cell. Mutations of K-RAS oncogene lead to an accumulation of GTP-bound proteins that maintains an active conformation. In the pancreatic ductal adenocarcinoma (PDAC), one of the most deadly cancers in occidental countries, mutations of the K-RAS oncogene are nearly systematic (>90%). Moreover, K-RAS mutation is the earliest genetic alteration occurring during pancreatic carcinogenetic sequence. In this review, we discuss the central role of K-RAS mutations and their tremendous diversity of biological properties by the interconnected regulation of signaling pathways (MAPKs, NF-κB, PI3K, Ral…). In pancreatic ductal adenocarcinoma, transcriptome analysis and preclinical animal models showed that K-RAS mutation alters biological behavior of PDAC cells (promoting proliferation, migration and invasion, evading growth suppressors, regulating mucin pattern, and miRNA expression). K-RAS also impacts tumor microenvironment and PDAC metabolism reprogramming. Finally we discuss therapeutic targeting strategies of K-RAS that have been developed without significant clinical success so far. As K-RAS is considered as the undruggable target, targeting its multiple effectors and target genes should be considered as potential alternatives.
Collapse
|
40
|
Illiano M, Sapio L, Caiafa I, Chiosi E, Spina A, Naviglio S. Forskolin sensitizes pancreatic cancer cells to gemcitabine via Stat3 and Erk1/2 inhibition. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.2.224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
41
|
Yang X, Xu Y, Brooks A, Guo B, Miskimins KW, Qian SY. Knockdown delta-5-desaturase promotes the formation of a novel free radical byproduct from COX-catalyzed ω-6 peroxidation to induce apoptosis and sensitize pancreatic cancer cells to chemotherapy drugs. Free Radic Biol Med 2016; 97:342-350. [PMID: 27368132 PMCID: PMC5807006 DOI: 10.1016/j.freeradbiomed.2016.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 11/26/2022]
Abstract
Recent research has demonstrated that colon cancer cell proliferation can be suppressed in the cells that overexpress COX-2 via generating 8-hydroxyoctanoic acid (a free radical byproduct) during dihomo-γ-linolenic acid (DGLA, an ω-6 fatty acid) peroxidation from knocking down cellular delta-5-desaturase (D5D, the key enzyme for converting DGLA to the downstream ω-6, arachidonic acid). Here, this novel research finding is extended to pancreatic cancer growth, as COX-2 is also commonly overexpressed in pancreatic cancer. The pancreatic cancer cell line, BxPC-3 (with high COX-2 expression and mutated p53), was used to assess not only the inhibitory effects of the enhanced formation of 8-hydroxyoctanoic acid from cellular COX-2-catalyzed DGLA peroxidation but also its potential synergistic and/or additive effect on current chemotherapy drugs. This work demonstrated that, by inducing DNA damage through inhibition of histone deacetylase, a threshold level of 8-hydroxyoctanoic acid achieved in DGLA-treated and D5D-knockdown BxPC-3 cells subsequently induce cancer cell apoptosis. Furthermore, it was shown that a combination of D5D knockdown along with DGLA treatment could also significantly sensitize BxPC-3 cells to various chemotherapy drugs, likely via a p53-independent pathway through downregulating of anti-apoptotic proteins (e.g., Bcl-2) and activating pro-apoptotic proteins (e.g., caspase 3, -9). This study reinforces the supposition that using commonly overexpressed COX-2 for molecular targeting, a strategy conceptually distinct from the prevailing COX-2 inhibition strategy used in cancer treatment, is an important as well as viable alternative to inhibit cancer cell growth. Based on the COX-2 metabolic cascade, the outcomes presented here could guide the development of a novel ω-6-based dietary care strategy in combination with chemotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Yi Xu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Amanda Brooks
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Bin Guo
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States
| | - Keith W Miskimins
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, United States
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND 58108, United States.
| |
Collapse
|
42
|
Yang N, Cui H, Han F, Zhang L, Huang T, Zhou Y, Zhou J. Paeoniflorin inhibits human pancreatic cancer cell apoptosis via suppression of MMP-9 and ERK signaling. Oncol Lett 2016; 12:1471-1476. [PMID: 27446455 DOI: 10.3892/ol.2016.4761] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/22/2016] [Indexed: 12/12/2022] Open
Abstract
Paeoniflorin exhibits anticancer, anti-inflammatory and antioxidation effects, as well as specific pharmacological effects on smooth muscle and the immune, cardiovascular and central nervous systems. The present study aimed to investigate the anticancer effects of paeoniflorin on pancreatic cancer cells and to elucidate the mechanisms by which these effects occur. In the present study, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays were performed to assess cell viability and cell cytotoxicity of BXPC-3 human pancreatic cancer cells, respectively. Cellular apoptosis and caspase-3/9 activities were analyzed using an Annexin V-fluorescein isothiocyanate/propidium iodide Apoptosis Detection kit, a DAPI staining assay and colorimetric kits, respectively. Matrix metalloproteinase-9 (MMP-9) and extracellular signal-regulated kinases (ERK) protein expression in BXPC-3 cells were also investigated using gelatin zymography assays and western blot analysis, respectively. In the present study, paeoniflorin was found to inhibit the cell viability and increase cell cytotoxicity of BXPC-3 cells in a dose- and time-dependent manner. In addition, cellular apoptosis, as well as caspase-3 and -9 activity of BXPC-3 cells was increased following paeoniflorin treatment. Notably, paeoniflorin reduced MMP-9 and ERK protein expression in BXPC-3 cells. These results indicate that paeoniflorin exhibits a potential anticancer effect by enhancing human pancreatic cancer cell apoptosis via the suppression of MMP-9 and ERK signaling.
Collapse
Affiliation(s)
- Nanmu Yang
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Hong Cui
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Feng Han
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Ling Zhang
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Tao Huang
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Yi Zhou
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Jinxue Zhou
- Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| |
Collapse
|
43
|
He X, Wang J, Wei W, Shi M, Xin B, Zhang T, Shen X. Hypoxia regulates ABCG2 activity through the activivation of ERK1/2/HIF-1α and contributes to chemoresistance in pancreatic cancer cells. Cancer Biol Ther 2016; 17:188-98. [PMID: 26785721 DOI: 10.1080/15384047.2016.1139228] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a drug resistant hypovascular tumor. Although there are many studies on the mechanism of chemoresistance in pancreatic cancers, studies on the relationship between ABCG2 and chemoresistance during hypoxia of pancreatic cancer are rare. Hypoxia-inducible factor-1 (HIF-1α) is a master regulator of the transcriptional response to oxygen deprivation in cancer cells. The aim of this study was to examine the role of ABCG2 and HIF-1α in mediating chemoresistance during hypoxia in pancreatic cancer. In this study, we detected the expression levels of ABCG2, ERK/phosphorylated-ERK (p-ERK) and HIF-1α by immunohistochemistry in fresh pancreatic cancer and paracarcinoma tissues obtained from 25 patients. The mechanism by which p-ERK1/2 and HIF-1α affect ABCG2s expression was analyzed in the hypoxic cultured human pancreatic cancer cell line Capan-2. ABCG2-mediatedregulation of gemcitabine response under hypoxic conditions in pancreatic cancer cells was observed. It was found that ABCG2, ERK/p-ERK and HIF-1α were overexpressed in cancer tissues. ABCG2, HIF-1α and p-ERK levels were demonstrated to be high during hypoxic conditions in pancreatic cancer cells. Hypoxia induced phosphorylation of ERK1/2 to activate HIF-1α and contribute the ABCG2 expression and mediated gemcitabine chemoresistance in pancreatic cancer cells. Hypoxic conditions induced HIF-1α binding to target gene sequences in the ABCG2 promoter, resulting in increased transcription in pancreatic cancer cells. We demonstrated that hypoxia-induced chemoresistance is due to the regulation of ABCG2 through the activation of ERK1/2/HIF-1α. ABCG2 could serve as a predictor of gemcitabine response and, potentially, as a chemotherapeutic target in pancreatic cancer. Inhibition of ERK1/2 and HIF-1αcould result in increased gemcitabine sensitization in pancreatic cancer with highly expressed ABCG2 cell member protein.
Collapse
Affiliation(s)
- Xiaodan He
- a School of Medicine, Nankai University , Tianjin , China
| | - Juan Wang
- a School of Medicine, Nankai University , Tianjin , China
| | - Wei Wei
- b Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Meiyan Shi
- a School of Medicine, Nankai University , Tianjin , China
| | - Beibei Xin
- a School of Medicine, Nankai University , Tianjin , China
| | - Ti Zhang
- b Tianjin Medical University Cancer Institute and Hospital , Tianjin , China
| | - Xiaohong Shen
- a School of Medicine, Nankai University , Tianjin , China
| |
Collapse
|
44
|
Miyashita T, Miki K, Kamigaki T, Makino I, Nakagawara H, Tajima H, Takamura H, Kitagawa H, Fushida S, Ahmed AK, Duncan MD, Harmon JW, Ohta T. Low-dose gemcitabine induces major histocompatibility complex class I-related chain A/B expression and enhances an antitumor innate immune response in pancreatic cancer. Clin Exp Med 2015; 17:19-31. [PMID: 26449615 DOI: 10.1007/s10238-015-0394-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/12/2015] [Indexed: 02/06/2023]
Abstract
We investigated the effect of gemcitabine (GEM), a key drug for pancreatic cancer treatment, on the expression of cell surface MICA/B in pancreatic cancer cells and resulting cytotoxicity of γδ T cells. We assessed the effect of GEM on the upregulation of cell surface MICA/B expression by flow cytometry, utilizing six pancreatic cancer cell lines. MICA and CD16 expressions from resected pancreatic cancer patient specimens, which received neoadjuvant chemotherapy (NAC) with GEM, were analyzed by immunohistochemistry. GEM could increase MICA/B expression on cell surface in pancreatic cancer cell lines (in 2 of 6 cell lines). This effect was most effectively at concentration not affecting cell growth of GEM (0.001 μM), because MICA/B negative population was appeared at concentration at cytostatic and cytotoxic effect to cell growth (0.1 and 10 μM). The cytotoxic activity of γδ T cells against PANC-1 was detected and functions through interactions between NKG2D and MICA/B. However, the enhancement of NKG2D-dependent cytotoxicity with increased MICA/B expression, by GEM treatment, was not observed. In addition, soluble MIC molecules were released from pancreatic cancer cell lines in culture supernatant with GEM treatment. Immunohistochemical staining demonstrated that MICA expression in tumor cells and CD16 positive cells surrounding tumors were significantly higher in the NAC group compared to that of the control group. There was a significant correlation between NAC and MICA expression, as well as NAC and CD16 positive cell expression. The present results indicate that low-dose GEM-induced MICA/B expression enhances innate immune function rather than cytotoxicity in pancreatic cancer. In addition, our result suggests that the inhibition of cleavage and release of MIC molecules from the tumor surface could potentially improve NKG2D-dependent cytotoxicity.
Collapse
Affiliation(s)
- Tomoharu Miyashita
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Kenji Miki
- Medinet Medical Institute, MEDINET Co., Ltd., 2-2-8 Tamagawadai, Setagaya-ku, Tokyo, 158-0096, Japan
| | - Takashi Kamigaki
- Medinet Medical Institute, MEDINET Co., Ltd., 2-2-8 Tamagawadai, Setagaya-ku, Tokyo, 158-0096, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hisatoshi Nakagawara
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hirohisa Kitagawa
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Ali K Ahmed
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - Mark D Duncan
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - John W Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, 4940 Eastern Avenue, Baltimore, MD, 21224, USA
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
45
|
Song B, Bian Q, Zhang YJ, Shao CH, Li G, Liu AA, Jing W, Liu R, Zhou YQ, Jin G, Hu XG. Downregulation of ASPP2 in pancreatic cancer cells contributes to increased resistance to gemcitabine through autophagy activation. Mol Cancer 2015; 14:177. [PMID: 26438046 PMCID: PMC4594892 DOI: 10.1186/s12943-015-0447-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/21/2015] [Indexed: 01/05/2023] Open
Abstract
Background Apoptosis-stimulating of p53 protein 2 (ASPP2) is one of the ASPP family members and it has been reported to be associated with human cancer. However, the role of it in pancreatic cancer is still not clear. Methods We analyzed the expression level of ASPP2 in cancer tissue samples with RT-qPCR, Western Blotting assay and immunohistochemistry staining. We studied the biological function of ASPP2 and its mechanism with gene overexpression and gene silencing technologies. We determined the sensitivity of pancreatic cells with differential ASPP2 level to gemcitabine and whether autophagy inhibition affected the gemcitabine resistance, both in vitro and in vivo. Results Expression of ASPP2 was downregulated in cancerous tissues in comparison with para-cancerous tissues. ASPP2 expression was linked to clinical outcomes in patients and down-regulation of ASPP2 increased cell proliferation, autophagic flux, the activity of AMP Kinase of pancreatic cancer cells and vice versa. Knockdown of ASPP2 results in increased resistance to gemcitabine, which was attributed to the enhanced autophagy. Conclusions ASSP2 expression is lower in cancerous tissues and decreased ASPP2 lead to higher cancer cells proliferation and autophagic flux, which contribute to the gemcitabine resistance. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0447-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bin Song
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Qi Bian
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| | - Yi-Jie Zhang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Cheng-Hao Shao
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Gang Li
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - An-An Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Wei Jing
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Rui Liu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Ying-Qi Zhou
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Gang Jin
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China. .,Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| | - Xian-Gui Hu
- Department of General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China. .,Department of Nephrology, Changhai Hospital, Second Military Medical University, Changhai Road No.168, Shanghai, China.
| |
Collapse
|
46
|
Manuel ER, Chen J, D'Apuzzo M, Lampa MG, Kaltcheva TI, Thompson CB, Ludwig T, Chung V, Diamond DJ. Salmonella-Based Therapy Targeting Indoleamine 2,3-Dioxygenase Coupled with Enzymatic Depletion of Tumor Hyaluronan Induces Complete Regression of Aggressive Pancreatic Tumors. Cancer Immunol Res 2015; 3:1096-107. [PMID: 26134178 PMCID: PMC4561205 DOI: 10.1158/2326-6066.cir-14-0214] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 06/02/2015] [Indexed: 12/18/2022]
Abstract
Bacterial-based therapies are emerging as effective cancer treatments and hold promise for refractory neoplasms, such as pancreatic ductal adenocarcinoma (PDAC), which has not shown significant improvement in therapy for more than 25 years. Using a novel combination of shIDO-ST, a Salmonella-based therapy targeting the immunosuppressive molecule indoleamine 2,3-dioxygenase (IDO), with an enzyme, PEGPH20, which depletes extracellular matrix hyaluronan, we observed extended survival with frequent total regression of autochthonous and orthotopic PDAC tumors. This observation was associated with migration and accumulation of activated polymorphonuclear neutrophils (PMN) from spleens into tumors, which was not seen using a scrambled control (shScr-ST). Purified splenic PMNs from PEGPH20/shIDO-ST-treated mice exhibited significant IDO knockdown and were able to kill tumor targets ex vivo through mechanisms involving FasL and serine proteases. In addition, CD8(+) T cells were observed to contribute to late control of pancreatic tumors. Collectively, our data demonstrate that entry of shIDO-ST and PMNs into otherwise impermeable desmoplastic tumors is facilitated by PEGPH20-mediated HA removal, further highlighting an important component of effective treatment for PDAC.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/therapeutic use
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Cell Adhesion Molecules/therapeutic use
- Combined Modality Therapy
- Gene Knockdown Techniques/methods
- Genetic Therapy/methods
- Hyaluronic Acid/deficiency
- Hyaluronic Acid/metabolism
- Hyaluronoglucosaminidase/therapeutic use
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice, Inbred C57BL
- Molecular Targeted Therapy/methods
- Neoplasm Transplantation
- Neutrophils/immunology
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Recombinant Proteins/therapeutic use
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Edwin R Manuel
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California.
| | - Jeremy Chen
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California
| | - Massimo D'Apuzzo
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Melanie G Lampa
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California
| | - Teodora I Kaltcheva
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California
| | | | - Thomas Ludwig
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University, Columbus, Ohio
| | - Vincent Chung
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Don J Diamond
- Department of Experimental Therapeutics, Beckman Research Institute of City of Hope, Duarte, California.
| |
Collapse
|
47
|
Yadav V, Varshney P, Sultana S, Yadav J, Saini N. Moxifloxacin and ciprofloxacin induces S-phase arrest and augments apoptotic effects of cisplatin in human pancreatic cancer cells via ERK activation. BMC Cancer 2015; 15:581. [PMID: 26260159 PMCID: PMC4531397 DOI: 10.1186/s12885-015-1560-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 07/15/2015] [Indexed: 01/05/2023] Open
Abstract
Background Pancreatic cancer, one of the most dreadful gastrointestinal tract malignancies, with the current chemotherapeutic drugs has posed a major impediment owing to poor prognosis and chemo-resistance thereby suggesting critical need for additional drugs as therapeutics in combating the situation. Fluoroquinolones have shown promising and significant anti-tumor effects on several carcinoma cell lines. Methods Previously, we reported growth inhibitory effects of fourth generation fluoroquinolone Gatifloxacin, while in the current study we have investigated the anti-proliferative and apoptosis-inducing mechanism of older generation fluoroquinolones Moxifloxacin and Ciprofloxacin on the pancreatic cancer cell-lines MIA PaCa-2 and Panc-1. Cytotoxicity was measured by MTT assay. Apoptosis induction was evaluated using annexin assay, cell cycle assay and activation of caspase-3, 8, 9 were measured by western blotting and enzyme activity assay. Results Herein, we found that both the fluoroquinolones suppressed the proliferation of pancreatic cancer cells by causing S-phase arrest and apoptosis. Blockade in S-phase of cell cycle was associated with decrease in the levels of p27, p21, CDK2, cyclin-A and cyclin-E. Herein we also observed triggering of extrinsic as well as intrinsic mitochondrial apoptotic pathway as suggested by the activation of caspase-8, 9, 3, and Bid respectively. All this was accompanied by downregulation of antiapoptotic protein Bcl-xL and upregulation of proapoptotic protein Bak. Our results strongly suggest the role of extracellular-signal-regulated kinases (ERK1/2), but not p53, p38 and c-JUN N-terminal kinase (JNK) in fluoroquinolone induced growth inhibitory effects in both the cell lines. Additionally, we also found both the fluoroquinolones to augment the apoptotic effects of broad spectrum anticancer drug Cisplatin via ERK. Conclusion The fact that these fluoroquinolones synergize the effect of cisplatin opens new insight into therapeutic index in treatment of pancreatic cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1560-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vikas Yadav
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India. .,Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi, India.
| | - Pallavi Varshney
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India.
| | - Sarwat Sultana
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi, India.
| | - Jyoti Yadav
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India.
| | - Neeru Saini
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, India.
| |
Collapse
|
48
|
Blockade of dual-specificity phosphatase 28 decreases chemo-resistance and migration in human pancreatic cancer cells. Sci Rep 2015. [PMID: 26212664 PMCID: PMC4515742 DOI: 10.1038/srep12296] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer remains one of the most deadly cancers, with a grave prognosis. Despite numerous endeavors to improve treatment of the neoplasm, limited progress has been made. In the present study, we investigated the role of dual specificity phosphatase 28 (DUSP28) in relation to anti-cancer drug sensitivity and migratory activity in human pancreatic cancer cells for the first time. Analysis using Universal exPress Codes (UPCs) with the GEO database showed significantly higher DUSP28 mRNA expression in pancreatic cancers. We found that DUSP28 was highly expressed in several human pancreatic cancer cell lines that showed resistance to anti-cancer drugs. Overexpression of DUSP28 decreased anti-cancer drug-sensitivity and enhanced cellular migration via the ERK1/2 pathway in DUSP28-negative cell lines. Knockdown of DUSP28 re-sensitized cells to anti-cancer drugs even at sublethal doses by inducing an apoptotic pathway and significantly reduced migration in DUSP28-positive human pancreatic cancer cell lines. Furthermore, DUSP28-positive cell line (Panc-1) xenograft models were more resistant to gemcitabine treatment than DUSP28-negative cell line (SNU-213) xenograft models. Collectively, these results indicate that DUSP28 plays a key role in drug resistance and migratory activity in human pancreatic cells, and suggest that targeting DUSP28 might have clinical relevance in eradicating malignant pancreatic cancers.
Collapse
|
49
|
RLN2 Is a Positive Regulator of AKT-2-Induced Gene Expression Required for Osteosarcoma Cells Invasion and Chemoresistance. BIOMED RESEARCH INTERNATIONAL 2015; 2015:147468. [PMID: 26229955 PMCID: PMC4503584 DOI: 10.1155/2015/147468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 01/09/2023]
Abstract
The aim of the study was to determine the effect of H2 relaxin (RLN2) on invasion, migration, and chemosensitivity to cisplatin in human osteosarcoma U2-OS and MG-63 cells and then to investigate the effect of RLN2 on the AKT/NF-κB signaling pathway. The expression of RLN2, p-AKT (Ser473), and p-ERK1/2 (Phospho-Thr202/Tyr204) proteins was detected by western blot in OS tissues from 21 patients with pulmonary metastatic disease, and the correlation between RLN2 and p-AKT or RLN2 and p-ERK1/2 expression was investigated. RLN2 expression was inhibited by RLN2 siRNA transfection in the MG-63 cells. RLN2 was overexpressed in the U2-OS cells by treatment with recombinant relaxin. The results showed that positive relation was found between RLN2 and p-AKT expression in tissues of OS. Silencing RLN2 inhibited cell migratory and invasive ability and angiogenesis formation and increased the chemosensitivity to cisplatin in MG-63 cells. RLN2 overexpression promoted migratory and invasive ability and angiogenesis and increased the chemoresistance to cisplatin in U2-OS cells. Silencing RLN2 inhibited the activity of AKT/NF-κB signaling pathway in MG-63 cells, and vice versa. Blockage of both pathways by specific inhibitors abrogated RLN2-induced survival and invasion of OS cells, and vice versa. Our results indicated RLN2 confers to migratory and invasive ability, angiogenesis, and chemoresistance to cisplatin via modulating the AKT/NF-κB signaling pathway in vitro.
Collapse
|
50
|
Wang M, Lu X, Dong X, Hao F, Liu Z, Ni G, Chen D. pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol 2015; 13:66. [PMID: 25880226 PMCID: PMC4337256 DOI: 10.1186/s12957-015-0451-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/08/2015] [Indexed: 11/10/2022] Open
Abstract
Background Our previous study has demonstrated that knockdown of activated ERK1/2(pERK1/2) sensitizes pancreatic cancer cells to chemotherapeutic drug gemcitabine (Gem) treatment. However, the details of this survival mechanism remain undefined. It has also shown that Bcl-2 confers resistance and Bax sensitizes to gemcitabine-induced apoptosis in pancreatic cancer cells. Furthermore, the extracellular signaling-regulated kinase (ERK) signaling pathway regulates Bcl-2/Bax expression ratio. We therefore tested the hypothesis that pancreatic cancer cells are resistant to gemcitabine and this resistance is due to activation of ERK1/2 and subsequent upregulation of Bcl-2 and downregulation of Bax. Methods Pancreatic cancer cell BXPC-3 was used in the study. The effect of pharmacological inhibition of ERK1/2 on resistance of pancreatic cancer cells to apoptosis induced by treatment with gemcitabine was analyzed. The following methods were utilized: TUNEL and ELISA were used to detect apoptosis. Western blot was used to detect the protein expression. Results Gemcitabine treatment enhanced the activity of ERK1/2 in the BXPC-3 cells. Inhibition of the ERK1/2 by PD98059 could downregulate Bcl-2 and upregulate Bax and was associated with restoration of sensitivity to gemcitabine in BXPC-3 cells. Depletion of endogenous Bcl-2 expression by specific small interfering RNA transfection significantly increased gemcitabine-induced cell apoptosis. Combined treatment with PD98059 and Bax siRNA transfection could decrease gemcitabine-induced ERK1/2 and Bax activation, which subsequently resulted in decreased apoptosis. Conclusions The upregulation of ERK1/2-dependent Bcl-2 and downregulation of ERK1/2-dependent Bax can protect human pancreatic cancer cells from gemcitabine-induced apoptosis. Targeting the ERK1/2-Bax/Bcl-2 pathway may in part lead to sensitization of pancreatic cancer to gemcitabine.
Collapse
Affiliation(s)
- Min Wang
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China.
| | - Xingjiao Lu
- Department of Internal Neurology, People's Hospital of Zhangqiu, ZhangQiu, Shangdong, China.
| | - Xueguang Dong
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China.
| | - Fengyun Hao
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China. .,Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shangdong, China.
| | - Zimin Liu
- Department of Clinical Laboratory, People's Hospital of Laiwu, Laiwu, Shangdong, China. .,Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Guangzhen Ni
- Department of Clinical Laboratory, People's Hospital of Weifang, Weifang, Shangdong, China.
| | - Dong Chen
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|