1
|
Zhao J, Sun H, Wang C, Shang D. Breast cancer therapy: from the perspective of glucose metabolism and glycosylation. Mol Biol Rep 2024; 51:546. [PMID: 38642246 DOI: 10.1007/s11033-024-09466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Breast cancer is a leading cause of mortality and the most prevalent form of malignant tumor among women worldwide. Breast cancer cells exhibit an elevated glycolysis and altered glucose metabolism. Moreover, these cells display abnormal glycosylation patterns, influencing invasion, proliferation, metastasis, and drug resistance. Consequently, targeting glycolysis and mitigating abnormal glycosylation represent key therapeutic strategies for breast cancer. This review underscores the importance of protein glycosylation and glucose metabolism alterations in breast cancer. The current research efforts in developing effective interventions targeting glycolysis and glycosylation are further discussed.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Haiting Sun
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Che Wang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China.
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Science, Liaoning Normal University, Dalian, 116081, China.
| | - Dejing Shang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Science, Liaoning Normal University, Dalian, 116081, China.
| |
Collapse
|
2
|
Zhang Y, Yang F, Wu J, Huang J, Li P, Huang G. Idebenone Exerts anti-Triple Negative Breast Cancer Effects via Dual Signaling Pathways of GADD45 and AMPK. Nutr Cancer 2024; 76:379-392. [PMID: 38332562 DOI: 10.1080/01635581.2024.2314320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
Idebenone, a mitochondrial regulator, has exhibited anti-cancer activity in neurogenic and prostate tumor cells; however, its efficacy and specific targets in the treatment of triple-negative breast cancer (TNBC) remain unclear. This study aims to evaluate the potential of Idebenone as a therapeutic agent for TNBC. TNBC cell lines and Xenograft mouse models were used to assess the effect of Idebenone on TNBC both in vitro and in vivo. To investigate the underlying mechanism of Idebenone's effect on TNBC, cell viability assay, transwell invasion assay, cell cycle analysis, apoptosis assay, mitochondrial membrane potential assay, immunofluorescence staining, and transcriptome sequencing were utilized. The results showed that Idebenone impeded the proliferation, colony formation, migration, and invasion of TNBC cells, suppressed apoptosis, and halted the cell cycle in the G2/M phase. The inhibitory effect of Idebenone on TNBC was associated with the GADD45/CyclinB/CDK1 signaling pathway. By disrupting the mitochondrial membrane potential (MMP) and promoting mitophagy, Idebenone promoted cell autophagy through the AMPK/mTOR pathway, thus further suppressing the proliferation of TNBC cells. Furthermore, we found that Idebenone inhibited the development of TNBC in vivo. In conclusion, Idebenone may be a promising therapeutic option for TNBC as it is capable of inducing autophagy and apoptosis.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fan Yang
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahao Wu
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianhong Huang
- Department of General Surgery, Zengcheng District Hospital of Traditional Chinese Medicine, China
| | - Peiqing Li
- Department of General Surgery, Xinyi People's Hospital, Xinyi, China
| | - Guanqun Huang
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
3
|
Kuche K, Yadav V, Patel M, Chaudhari D, Date T, Jain S. Enhancing anti-cancer potential by delivering synergistic drug combinations via phenylboronic acid modified PLGA nanoparticles through ferroptosis-based therapy. BIOMATERIALS ADVANCES 2024; 156:213700. [PMID: 38042001 DOI: 10.1016/j.bioadv.2023.213700] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/11/2023] [Accepted: 11/09/2023] [Indexed: 12/04/2023]
Abstract
In this study, we investigated the potential of the sorafenib (SOR) and simvastatin (SIM) combination to induce ferroptosis-mediated cancer therapy. To enhance targeted drug delivery, we encapsulated the SOR + SIM combination within 4-carboxy phenylboronic acid (CPBA) modified PLGA nanoparticles (CPBA-PLGA(SOR + SIM)-NPs). The developed CPBA-PLGA(SOR + SIM)-NPs exhibited a spherical shape with a size of 213.1 ± 10.9 nm, a PDI of 0.22 ± 0.03, and a Z-potential of -22.9 ± 3.2 mV. Notably, these nanoparticles displayed faster drug release at acidic pH compared to physiological pH. In cellular experiments, CPBA-PLGA(SOR + SIM)-NPs demonstrated remarkable improvements, leading to a 2.51, 2.69, and 2.61-fold decrease in IC50 compared to SOR alone, and a 7.50, 16.71, and 5.11-fold decrease in IC50 compared to SIM alone in MDA-MB-231, A549, and HeLa cells, respectively. Furthermore, CPBA-PLGA(SOR + SIM)-NPs triggered a reduction in glutathione (GSH) levels, an increase in malondialdehyde (MDA) levels, and mitochondrial membrane depolarization in all three cell lines. Pharmacokinetic evaluation revealed a 2.50- and 2.63-fold increase in AUC0-∞, as well as a 1.53- and 2.46-fold increase in mean residence time (MRT) for SOR and SIM, respectively, compared to the free drug groups. Notably, the CPBA-PLGA(SOR + SIM)-NPs group exhibited significant reduction in tumor volume, approximately 9.17, 2.45, and 1.63-fold lower than the control, SOR + SIM, and PLGA(SOR + SIM)-NPs groups, respectively. Histological examination and biomarker analysis showed no significant differences compared to the control group, suggesting the biocompatibility of the developed particles for in-vivo applications. Altogether, our findings demonstrate that CPBA-PLGA(SOR + SIM)-NPs hold tremendous potential as an efficient drug delivery system for inducing ferroptosis, providing a promising therapeutic option for cancer treatment.
Collapse
Affiliation(s)
- Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Vivek Yadav
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Meet Patel
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Dasharath Chaudhari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Tushar Date
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India.
| |
Collapse
|
4
|
Kuche K, Yadav V, Dharshini M, Ghadi R, Chaudhari D, Date T, Jain S. Synergistic anticancer therapy via ferroptosis using modified bovine serum albumin nanoparticles loaded with sorafenib and simvastatin. Int J Biol Macromol 2023; 253:127254. [PMID: 37813219 DOI: 10.1016/j.ijbiomac.2023.127254] [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: 06/06/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023]
Abstract
Ferroptosis is a non-apoptotic cell death pathway characterized by the accumulation of lipid-peroxy radicals within the affected cells. Here, we investigate the synergistic capacity of sorafenib (SOR) and simvastatin (SIM) to trigger ferroptosis for cancer therapy. For precise in-vivo delivery, SOR + SIM was ratiometrically loaded in bovine serum albumin nanoparticles (BSA-NPs) modified with 4-carboxy phenylboronic acid (CPBA). The developed CPBA-BSA(SOR + SIM)-NPs revealed size of 175.2 ± 12.8 nm, with PDI of 0.22 ± 0.03 and Z-potential of -29.6 ± 4.8 mV. Significantly, CPBA-BSA(SOR + SIM)-NPs exhibited > 2 and > 5-fold reduction in IC50 values compared to individual SOR and SIM treatments respectively, in all tested cell lines. Moreover, CPBA-BSA(SOR + SIM)-NPs treated cells exhibited decrease in glutathione levels, increase in malonaldehyde levels and depolarization of mitochondrial membrane potential (JC-1 assay). Pharmacokinetic analysis revealed enhanced AUC0-∞ and MRT levels for SOR and SIM when administered as CPBA-BSA(SOR + SIM)-NPs compared to free drugs. Crucially, in in-vivo experiments, CPBA-BSA(SOR + SIM)-NPs led to a significant reduction in tumor volume compared to various control groups. Histological and biomarker analyses underscore their biocompatibility for clinical applications. In conclusion, this study highlights the potential of CPBA-BSA(SOR + SIM)-NPs as a promising strategy for inducing ferroptosis in cancer cells, concurrently improving drug delivery and therapeutic efficacy. This approach opens new avenues in cancer treatment.
Collapse
Affiliation(s)
- Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Vivek Yadav
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - M Dharshini
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Dasharath Chaudhari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Tushar Date
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab 160062, India.
| |
Collapse
|
5
|
Guo S, Zhang C, Zeng H, Xia Y, Weng C, Deng Y, Wang L, Wang H. Glycolysis maintains AMPK activation in sorafenib-induced Warburg effect. Mol Metab 2023; 77:101796. [PMID: 37696356 PMCID: PMC10550717 DOI: 10.1016/j.molmet.2023.101796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/19/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second deadly cancer in the world and still lacks curative treatment. Aerobic glycolysis, or Warburg effect, is a major resistance mechanism induced by first-line treatment of HCC, sorafenib, and is regulated by the master regulator of metabolism, AMPK. Activation of AMPK is required for resistance; however, activation dynamics of AMPK and its regulation is rarely studied. Engineering cells to express an AMPK activity biosensor, we monitor AMPK activation in single HCC cells in a high throughput manner during sorafenib-induced drug resistance. Sorafenib induces transient activation of AMPK, duration of which is dependent on glucose. Inhibiting glycolysis shortens AMPK activation; whereas increasing glycolysis increases its activation duration. Our data highlight that activation duration of AMPK is important for cancer evasion of therapeutic treatment and glycolysis is a key regulator of activation duration of AMPK.
Collapse
Affiliation(s)
- Sijia Guo
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Chenhao Zhang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Haiou Zeng
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, School of Integrated Circuit, Peking University, Beijing, 100871, China
| | - Yantao Xia
- University of California Los Angeles, Department of Chemical and Biomolecular Engineering, California, 90095, USA
| | - Chenghao Weng
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yichen Deng
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Luda Wang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, School of Integrated Circuit, Peking University, Beijing, 100871, China
| | - Huan Wang
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
6
|
Huang Y, Zhen Y, Chen Y, Sui S, Zhang L. Unraveling the interplay between RAS/RAF/MEK/ERK signaling pathway and autophagy in cancer: From molecular mechanisms to targeted therapy. Biochem Pharmacol 2023; 217:115842. [PMID: 37802240 DOI: 10.1016/j.bcp.2023.115842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
RAS/RAF/MEK/ERK signaling pathway is one of the most important pathways of Mitogen-activated protein kinases (MAPK), which widely participate in regulating cell proliferation, differentiation, apoptosis and signaling transduction. Autophagy is an essential mechanism that maintains cellular homeostasis by degrading aged and damaged organelles. Recently, some studies revealed RAS/RAF/MEK/ERK signaling pathway is closely related to autophagy regulation and has a dual effect in tumor cells. However, the specific mechanism by which RAS/RAF/MEK/ERK signaling pathway participates in autophagy regulation is not fully understood. This article provides a comprehensive review of the research progress with regard to the RAS/RAF/MEK/ERK signaling pathway and autophagy, as well as their interplay in cancer therapy. The impact of small molecule inhibitors that target the RAS/RAF/MEK/ERK signaling pathway on autophagy is discussed in this study. The advantages and limitations of the clinical combination of these small molecule inhibitors with autophagy inhibitors are also explored. The findings from this study may provide additional perspectives for future cancer treatment strategies.
Collapse
Affiliation(s)
- Yunli Huang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yongqi Zhen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanmei Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shaoguang Sui
- Emergency Department, The Second Hospital, Dalian Medical University, Dalian 116000, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
7
|
Beeraka NM, Zhang J, Mandal S, Vikram P. R. H, Liu J, B. M. N, Zhao D, Vishwanath P, B. M. G, Fan R. Screening fructosamine-3-kinase (FN3K) inhibitors, a deglycating enzyme of oncogenic Nrf2: Human FN3K homology modelling, docking and molecular dynamics simulations. PLoS One 2023; 18:e0283705. [PMID: 37910519 PMCID: PMC10619859 DOI: 10.1371/journal.pone.0283705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/14/2023] [Indexed: 11/03/2023] Open
Abstract
Fructosamine-3-kinase (FN3K) is involved in the deglycation of Nrf2, a significant regulator of oxidative stress in cancer cells. However, the intricate functional aspects of FN3K and Nrf2 in breast cancers have not been explored vividly. The objectives of this study are to design the human FN3K protein using homology modeling followed by the screening of several anticancer molecules and examining their efficacy to modulate FN3K activity, Nrf2-mediated antioxidant signalling. Methods pertinent to homology modeling, virtual screening, molecular docking, molecular dynamics simulations, assessment of ADME properties, cytotoxicity assays for anticancer molecules of natural/synthetic origin in breast cancer cells (BT-474, T-47D), and Western blotting were used in this study. The screened anticancer molecules including kinase inhibitors of natural and synthetic origin interacted with the 3-dimensional structure of the catalytic domain in human FN3K protein designed through homology modeling by significant CDOCKER interaction energies. Subsequently, gefitinib, sorafenib, neratinib, tamoxifen citrate, and cyclosporine A enhanced the expression of FN3K in BT-474 cell lines with simultaneous alteration in Nrf2-driven antioxidant signalling. Oxaliplatin significantly downregulated FN3K expression and modulated Nrf2-driven antioxidant signalling when compared to cisplatin and other anticancer drugs. Hence, the study concluded the potential implications of existing anticancer drugs to modulate FN3K activity in breast cancers.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, India
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Subhankar Mandal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Hemanth Vikram P. R.
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Junqi Liu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Namitha B. M.
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Di Zhao
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Prashanth Vishwanath
- Department of Biochemistry, Center of Excellence in Molecular Biology and Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Gurupadayya B. M.
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Ruitai Fan
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Kuche K, Yadav V, Patel M, Ghadi R, Jain S. Exploring Sorafenib and Simvastatin Combination for Ferroptosis-Induced Cancer Treatment: Cytotoxicity Screening, In Vivo Efficacy, and Safety Assessment. AAPS PharmSciTech 2023; 24:180. [PMID: 37697085 DOI: 10.1208/s12249-023-02639-z] [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: 06/09/2023] [Accepted: 08/13/2023] [Indexed: 09/13/2023] Open
Abstract
Ferroptosis, a pathway dependent on oxygen and iron catalysts, holds promise as a therapeutic approach for cancer treatment due to its manageable regulation, direct control, and immunogenic properties. The sensitivity of cancer cells to ferroptosis induction varies based on their metabolic, genetic, and signalling pathways, prompting the use of combination therapy. In this study, we conducted a screening of drug combinations, including sorafenib (SOR) with simvastatin (SIM), phenethyl isothiocyanate, and trigonelline, in MDA-MB-231, A549, and HeLa cells to assess their cytotoxicity. The SOR-SIM combination exhibited a synergistic effect in MDA-MB-231, A549, and HeLa cells, with calculated CI values of ~ 0.66, 0.53, and 0.59, respectively. Furthermore, co-treatment with ferrostatin-1 resulted in a concentration-dependent increase in the IC50 values. Additionally, SOR + SIM demonstrated a significant reduction in GSH levels, an increase in MDA levels, and mitochondrial membrane depolarization across all three cell lines, indicating their ferroptosis inducing potential. In-vivo studies showed a significant reduction in tumor volume by 3.53-, 2.55-, and 1.47-fold compared to control, SIM, and SOR, respectively. Toxicity assessments revealed insignificant changes in biomarker levels and no observable deformations in isolated organs, except for erythrocyte shrinkage and membrane scrambling effects caused by the SOR + SIM combination. Overall, our findings highlight the potential of the SOR + SIM combination as an effective strategy for cancer treatment, emphasizing the importance of further research in targeted drug delivery systems to ensure its safety.
Collapse
Affiliation(s)
- Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab, India, 160062
| | - Vivek Yadav
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab, India, 160062
| | - Meet Patel
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab, India, 160062
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab, India, 160062
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali, Punjab, India, 160062.
| |
Collapse
|
9
|
Li RZ, Guan XX, Wang XR, Bao WQ, Lian LR, Choi SW, Zhang FY, Yan PY, Leung ELH, Pan HD, Liu L. Sinomenine hydrochloride bidirectionally inhibits progression of tumor and autoimmune diseases by regulating AMPK pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154751. [PMID: 37004400 DOI: 10.1016/j.phymed.2023.154751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Chronic diseases such as tumors and autoimmune disorders are closely linked to metabolism and immunity and require conflicting treatment methods. AMPK can regulate cell growth and inflammation through energy metabolism. Sinomenine is a compound extracted from the traditional Chinese herb sinomenium acutum (Thunb.) Rehd. et Wils. It has been used to treat NSCLC (non-small-cell lung cancer) and RA (rheumatoid arthritis) in some studies, but with limited understanding of its mechanisms. OBJECTIVE This study aims to examine the inhibitory effect of sinomenine hydrochloride (SH) on NSCLC and RA and to understand the underlying joint mechanisms. RESULTS The results indicate that SH has a cytotoxic effect specifically on tumor cells, but not on normal cells. SH was found to induce cell apoptosis by activating the AMPK-mTOR pathway. Additionally, in autoimmune disease cell models, SH was shown to reduce the growth of RA-FLS cells by inhibiting the phosphorylation of AMPK, while having no effect on normal macrophages. Moreover, in vivo studies also showed that SH could reduce the production of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 and slow the development of adjuvant arthritis in rats. Furthermore, SH was found to significantly suppress tumor growth in a tumor xenograft experiment in mice. CONCLUSIONS This study provides new insights into the treatment of tumors and autoimmune diseases by demonstrating that SH can selectively inhibit the growth of NSCLC cells and the progression of RA through activation of the AMPK pathway.
Collapse
Affiliation(s)
- Run Ze Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China
| | - Xiao Xiang Guan
- State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute of Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa, Macau (SAR), China
| | - Xuan Run Wang
- State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute of Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa, Macau (SAR), China
| | - Wei-Qian Bao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China
| | - Li-Rong Lian
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China
| | - Seong Wang Choi
- State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute of Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa, Macau (SAR), China
| | - Fang Yuan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute of Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa, Macau (SAR), China
| | - Pei-Yu Yan
- State Key Laboratory of Quality Research in Chinese Medicine/ Macau Institute of Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Macao, Taipa, Macau (SAR), China
| | - Elaine Lai Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR), China, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China
| | - Hu-Dan Pan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China.
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Caligiuri I, Vincenzo C, Asano T, Kumar V, Rizzolio F. The metabolic crosstalk between PIN1 and the tumour microenvironment. Semin Cancer Biol 2023; 91:143-157. [PMID: 36871635 DOI: 10.1016/j.semcancer.2023.03.001] [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: 09/12/2022] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) is a member of a family of peptidyl-prolyl isomerases that specifically recognizes and binds phosphoproteins, catalyzing the rapid cis-trans isomerization of phosphorylated serine/threonine-proline motifs, which leads to changes in the structures and activities of the targeted proteins. Through this complex mechanism, PIN1 regulates many hallmarks of cancer including cell autonomous metabolism and the crosstalk with the cellular microenvironment. Many studies showed that PIN1 is largely overexpressed in cancer turning on a set of oncogenes and abrogating the function of tumor suppressor genes. Among these targets, recent evidence demonstrated that PIN1 is involved in lipid and glucose metabolism and accordingly, in the Warburg effect, a characteristic of tumor cells. As an orchestra master, PIN1 finely tunes the signaling pathways allowing cancer cells to adapt and take advantage from a poorly organized tumor microenvironment. In this review, we highlight the trilogy among PIN1, the tumor microenvironment and the metabolic program rewiring.
Collapse
Affiliation(s)
- Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Canzonieri Vincenzo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Tomochiro Asano
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| | - Vinit Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida 201313, Uttar Pradesh, India.
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 30123 Venezia, Italy.
| |
Collapse
|
11
|
Novel Anti-Cancer Products Targeting AMPK: Natural Herbal Medicine against Breast Cancer. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020740. [PMID: 36677797 PMCID: PMC9863744 DOI: 10.3390/molecules28020740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Breast cancer is a common cancer in women worldwide. The existing clinical treatment strategies have been able to limit the progression of breast cancer and cancer metastasis, but abnormal metabolism, immunosuppression, and multidrug resistance involving multiple regulators remain the major challenges for the treatment of breast cancer. Adenosine 5'-monophosphate (AMP)-Activated Protein Kinase (AMPK) can regulate metabolic reprogramming and reverse the "Warburg effect" via multiple metabolic signaling pathways in breast cancer. Previous studies suggest that the activation of AMPK suppresses the growth and metastasis of breast cancer cells, as well as stimulating the responses of immune cells. However, some other reports claim that the development and poor prognosis of breast cancer are related to the overexpression and aberrant activation of AMPK. Thus, the role of AMPK in the progression of breast cancer is still controversial. In this review, we summarize the current understanding of AMPK, particularly the comprehensive bidirectional functions of AMPK in cancer progression; discuss the pharmacological activators of AMPK and some specific molecules, including the natural products (including berberine, curcumin, (-)-epigallocatechin-3-gallate, ginsenosides, and paclitaxel) that influence the efficacy of these activators in cancer therapy; and elaborate the role of AMPK as a potential therapeutic target for the treatment of breast cancer.
Collapse
|
12
|
Disorders of cancer metabolism: The therapeutic potential of cannabinoids. Biomed Pharmacother 2023; 157:113993. [PMID: 36379120 DOI: 10.1016/j.biopha.2022.113993] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
Abnormal energy metabolism, as one of the important hallmarks of cancer, was induced by multiple carcinogenic factors and tumor-specific microenvironments. It comprises aerobic glycolysis, de novo lipid biosynthesis, and glutamine-dependent anaplerosis. Considering that metabolic reprogramming provides various nutrients for tumor survival and development, it has been considered a potential target for cancer therapy. Cannabinoids have been shown to exhibit a variety of anticancer activities by unclear mechanisms. This paper first reviews the recent progress of related signaling pathways (reactive oxygen species (ROS), AMP-activated protein kinase (AMPK), mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), hypoxia-inducible factor-1alpha (HIF-1α), and p53) mediating the reprogramming of cancer metabolism (including glucose metabolism, lipid metabolism, and amino acid metabolism). Then we comprehensively explore the latest discoveries and possible mechanisms of the anticancer effects of cannabinoids through the regulation of the above-mentioned related signaling pathways, to provide new targets and insights for cancer prevention and treatment.
Collapse
|
13
|
Zheng X, Ma H, Wang J, Huang M, Fu D, Qin L, Yin Q. Energy metabolism pathways in breast cancer progression: The reprogramming, crosstalk, and potential therapeutic targets. Transl Oncol 2022; 26:101534. [PMID: 36113343 PMCID: PMC9482139 DOI: 10.1016/j.tranon.2022.101534] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/14/2022] [Accepted: 09/04/2022] [Indexed: 11/19/2022] Open
Abstract
Breast cancer (BC) is a malignant tumor that seriously endangers health in women. BC, like other cancers, is accompanied by metabolic reprogramming. Among energy metabolism-related pathways, BC exhibits enhanced glycolysis, tricarboxylic acid (TCA) cycle, pentose phosphate pathway (PPP), glutamate metabolism, and fatty acid metabolism activities. These pathways facilitate the proliferation, growth and migration of BC cells. The progression of BC is closely related to the alterations in the activity or expression level of several metabolic enzymes, which are regulated by the intrinsic factors such as the key signaling and transcription factors. The metabolic reprogramming in the progression of BC is attributed to the aberrant expression of the signaling and transcription factors associated with the energy metabolism pathways. Understanding the metabolic mechanisms underlying the development of BC will provide a druggable potential for BC treatment and drug discovery.
Collapse
Affiliation(s)
- Xuewei Zheng
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Haodi Ma
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jingjing Wang
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Mengjiao Huang
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Dongliao Fu
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Qinan Yin
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
14
|
Orlistat Resensitizes Sorafenib-Resistance in Hepatocellular Carcinoma Cells through Modulating Metabolism. Int J Mol Sci 2022; 23:ijms23126501. [PMID: 35742944 PMCID: PMC9223797 DOI: 10.3390/ijms23126501] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
Sorafenib is one of the options for advanced hepatocellular carcinoma treatment and has been shown to extend median overall survival. However, sorafenib resistance often develops a few months after treatment. Hence, developing various strategies to overcome sorafenib resistance and understand the possible mechanisms is urgently needed. We first established sorafenib-resistant hepatocellular carcinoma (HCC) cells. Then, we found that sorafenib-resistant Huh7 cells (Huh7/SR) exhibit higher glucose uptakes and express elevated fatty acid synthesis and glucose metabolism-related proteins than their parental counterparts (Huh7). The current study investigated whether sorafenib resistance could be reversed by suppressing fatty acid synthesis, using a fatty acid synthase (FASN) inhibitor, orlistat, in HCC cells. FASN inhibition-caused changes in protein expressions and cell cycle distribution were analyzed by Western blot and flow cytometry, and changes in glucose uptakes were also evaluated by 18F-FDG uptake. Orlistat remarkably enhanced the cytotoxicity of sorafenib in both Huh7 and Huh7/SR cells, and flow cytometry showed that combination treatment significantly increased the sub-G1 population in both cell lines. Western blot revealed that the combination treatment effectively increased the ratio of Bax/Bcl-2 and decreased expressions of pERK; additionally, the combination treatment also strongly suppressed fatty acid synthesis-related proteins (e.g., FASN and SCD) in both cell lines. Lastly, the 18F-FDG uptake was repressed by the combination treatment in both cell lines. Our results indicated that orlistat-mediated FASN inhibition could overcome sorafenib resistance and enhance cell killing in HCC by changing cell metabolism.
Collapse
|
15
|
Metformin Protects Against Sunitinib-induced Cardiotoxicity: Investigating the Role of AMPK. J Cardiovasc Pharmacol 2022; 79:799-807. [PMID: 35266920 DOI: 10.1097/fjc.0000000000001256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 02/25/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Sunitinib is associated with cardiotoxicity through inhibition of AMP-protein kinase (AMPK) signaling. By contrast, the common antidiabetic agent metformin has demonstrated cardioprotection through indirect AMPK activation. In this study, we investigate the effects of metformin during sunitinib-induced cytotoxicity. Left ventricular developed pressure, coronary flow, heart rate, and infarct size were measured in Langendorff-perfused rat hearts treated with 1 µM sunitinib ±50 µM metformin ±1 µM human equilibrative nucleoside transporter inhibitor S-(4-Nitrobenzyl)-6-thioinosine (NBTI). Western blot analysis was performed for p-AMPKα levels. Primary isolated cardiac myocytes from the left ventricular tissue were used to measure live cell population levels. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assess adjunctive treatment of and metformin in human hepatoma G2 and promyelocytic leukemia (HL-60) cells treated with 0.1-100 µM sunitinib ±50 µM metformin. In the perfused hearts, coadministration of metformin attenuated the sunitinib-induced changes to left ventricular developed pressure, infarct size, and cardiac myocyte population. Western blot analysis revealed a significant decrease in p-AMPKα during sunitinib treatment, which was attenuated after coadministration with metformin. All metformin-induced effects were attenuated, and NBTI was coadministered. The MTT assay demonstrated an increase in the EC50 value during coadministration of metformin with sunitinib compared with sunitinib monotherapy in hepatoma G2 and HL-60 cell lines, demonstrating the impact and complexity of metformin coadministration and the possible role of AMPK signaling. This study highlights the novel cardioprotective properties of metformin and AMPK activation during sunitinib-induced cardiotoxicity when administered together in the Langendorff heart model.
Collapse
|
16
|
Jiang D, Xu J, Liu S, Nasser MI, Wei W, Mao T, Liu X, Zou X, Li J, Li X. Rosmanol induces breast cancer cells apoptosis by regulating PI3K/AKT and STAT3/JAK2 signaling pathways. Oncol Lett 2021; 22:631. [PMID: 34267823 PMCID: PMC8258625 DOI: 10.3892/ol.2021.12892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is one of the most frequently diagnosed cancers amongst women; however, there is currently no effective treatment. Natural compounds are considered to contribute to cancer prevention and have a pivotal role in modulating apoptosis. Rosmanol is a phenolic diterpene compound with antioxidant and anti-inflammatory properties. In the present study, the effects of Rosmanol on breast cancer cell proliferation/apoptosis were investigated, and it was demonstrated that it inhibited the proliferation of MCF-7 and MDA-MB 231 cells but did not have a significant effect on normal human breast MCF-10A cells. In addition, the apoptotic process was accelerated by Rosmanol, through mitochondrial pathways and reactive oxygen species (ROS) production caused by DNA damage, which function further demonstrated by the attenuation and addition of the ROS inhibitor, N-acetyl-cysteine. It was also demonstrated that Rosmanol accelerated cell apoptosis, and arrested breast cancer cells in the S phase. Moreover, Rosmanol inhibited proliferation and promoted apoptosis of cancer cells via the inhibition of ERK and STAT3 signals, attributable to the increase in p-p38, the overexpression of protein inhibitor of activated STAT3, and the decrease in PI3K/AKT, ERK and JAK2/STAT3.
Collapse
Affiliation(s)
- Dongjun Jiang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiaqi Xu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Sitong Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Moussa Ide Nasser
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Wei Wei
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Tianjiao Mao
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xintong Liu
- Bioprobe Application Research Unit, Chemical Biology Department, RIKEN-Max Planck Joint Research Division, RIkagaku KENkyusho/Institute of Physical and Chemical Research (RIKEN) Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Xiaopan Zou
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiang Li
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
17
|
Morikawa A, Grkovski M, Patil S, Jhaveri KL, Tang K, Humm JL, Holodny A, Beal K, Schöder H, Seidman AD. A phase I trial of sorafenib with whole brain radiotherapy (WBRT) in breast cancer patients with brain metastases and a correlative study of FLT-PET brain imaging. Breast Cancer Res Treat 2021; 188:415-425. [PMID: 34109515 PMCID: PMC11557212 DOI: 10.1007/s10549-021-06209-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Sorafenib has demonstrated anti-tumor efficacy and radiosensitizing activity preclinically and in breast cancer. We examined sorafenib in combination with whole brain radiotherapy (WBRT) and explored the [18F] 3'deoxy-3'-fluorothymidine (FLT)-PET as a novel brain imaging modality in breast cancer brain metastases. METHODS A phase I trial of WBRT + sorafenib was conducted using a 3 + 3 design with safety-expansion cohort. Sorafenib was given daily at the start of WBRT for 21 days. The primary endpoints were to determine a maximum tolerated dose (MTD) and to evaluate safety and toxicity. The secondary endpoint was CNS progression-free survival (CNS-PFS). MacDonald Criteria were used for response assessment with a correlative serial FLT-PET imaging study. RESULTS 13 pts were evaluable for dose-limiting toxicity (DLT). DLTs were grade 4 increased lipase at 200 mg (n = 1) and grade 3 rash at 400 mg (n = 3). The MTD was 200 mg. The overall response rate was 71%. Median CNS-PFS was 12.8 months (95%CI: 6.7-NR). A total of 15 pts (10 WBRT + sorafenib and 5 WBRT) were enrolled in the FLT-PET study: baseline (n = 15), 7-10 days post WBRT (FU1, n = 14), and an additional 12 week (n = 9). A decline in average SUVmax of ≥ 25% was seen in 9/10 (90%) of WBRT + sorafenib patients and 2/4 (50%) of WBRT only patients. CONCLUSIONS Concurrent WBRT and sorafenib appear safe at 200 mg daily dose with clinical activity. CNS response was favorable compared to historical controls. This combination should be considered for further efficacy evaluation. FLT-PET may be useful as an early response imaging tool for brain metastases. TRIAL AND CLINICAL REGISTRY Trial registration numbers and dates: NCT01724606 (November 12, 2012) and NCT01621906 (June 18, 2012).
Collapse
Affiliation(s)
- Aki Morikawa
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sujata Patil
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Komal L Jhaveri
- Breast Cancer Medicine Service, Evelyn Lauder Breast Center, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA
| | - Kendrick Tang
- Breast Cancer Medicine Service, Evelyn Lauder Breast Center, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrei Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kathryn Beal
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew D Seidman
- Breast Cancer Medicine Service, Evelyn Lauder Breast Center, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY, 10065, USA.
| |
Collapse
|
18
|
Chen Y, Liu R, Wang W, Wang C, Zhang N, Shao X, He Q, Ying M. Advances in targeted therapy for osteosarcoma based on molecular classification. Pharmacol Res 2021; 169:105684. [PMID: 34022396 DOI: 10.1016/j.phrs.2021.105684] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/03/2021] [Accepted: 05/15/2021] [Indexed: 02/08/2023]
Abstract
Osteosarcoma, a highly malignant tumor, is characterized by widespread and recurrent chromosomal and genetic abnormalities. In recent years, a number of elaborated sequencing analyses have made it possible to cluster the osteosarcoma based on the identification of candidate driver genes and develop targeted therapy. Here, we reviewed recent next-generation genome sequencing studies and advances in targeted therapies for osteosarcoma based on molecular classification. First, we stratified osteosarcomas into ten molecular subtypes based on genetic changes. And we analyzed potential targeted therapies for osteosarcoma based on the identified molecular subtypes. Finally, the development of targeted therapies for osteosarcoma investigated in clinical trials were further summarized and discussed. Therefore, we indicated the importance of molecular classification on the targeted therapy for osteosarcoma. And the stratification of patients based on the genetic characteristics of osteosarcoma will help to obtain a better therapeutic response to targeted therapies, bringing us closer to the era of personalized medicine.
Collapse
Affiliation(s)
- Yingqian Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Runzhi Liu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chen Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, China
| | - Xuejing Shao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
19
|
Sharaky M, Kamel M, Aziz MA, Omran M, Rageh MM, Abouzid KAM, Shouman SA. Design, synthesis and biological evaluation of a new thieno[2,3- d]pyrimidine-based urea derivative with potential antitumor activity against tamoxifen sensitive and resistant breast cancer cell lines. J Enzyme Inhib Med Chem 2021; 35:1641-1656. [PMID: 32781854 PMCID: PMC7470147 DOI: 10.1080/14756366.2020.1804383] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Breast cancer (BC) and endocrine resistance to chemotherapy are challenging problems where angiogenesis plays fundamental roles. Thus, targeting of VEGFR-2 signalling pathway has been an attractive approach. In this study, we synthesised a new sorafenib analogue, thieno[2,3-d]pyrimidine based urea derivative, KM6. It showed 65% inhibition of VEGF2 tyrosine kinase activity and demonstrated a potential antitumor activity in TAM-resistant, LCC2, and its parental MCF7 BC cells. KM6 retained the sensitivity of LCC2 through upregulation of key enzymes of apoptosis and proteins of cell death including caspases 3, 8, 9, P53, BAX/BCL-2 ratio and LDH in media. It downregulated mRNA expression of Ki-67, survivin, Akt, and reduced levels of ROS and glucose uptake. Moreover, KM6 reduced the levels of inflammation markers PGE2, COX2, IL-1β and IL6 and metastasis markers MMP-2 and MMP-9. In conclusion, KM6 is a promising compound for ER + and TAM-resistant BC with many potential antitumor and polypharmacological mechanisms.
Collapse
Affiliation(s)
- Marwa Sharaky
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa Kamel
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa A Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Mervat Omran
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Monira M Rageh
- Department of Biophysics, Faculty of Science, Cairo University, Giza, Egypt
| | - Khaled A M Abouzid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt.,Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| | - Samia A Shouman
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Silva BR, Rebelo R, Rodrigues JM, Xavier CPR, Vasconcelos MH, Queiroz MJRP. Synthesis of Novel Methyl 3-(hetero)arylthieno[3,2- b]pyridine-2-carboxylates and Antitumor Activity Evaluation: Studies In Vitro and In Ovo Grafts of Chick Chorioallantoic Membrane (CAM) with a Triple Negative Breast Cancer Cell Line. Molecules 2021; 26:molecules26061594. [PMID: 33805741 PMCID: PMC7999514 DOI: 10.3390/molecules26061594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 11/26/2022] Open
Abstract
A series of novel functionalized methyl 3-(hetero)arylthieno[3,2-b]pyridine-2-carboxylates 2a–2h were synthesized by C-C Pd-catalyzed Suzuki-Miyaura cross-coupling of methyl 3-bromothieno[3,2-b]pyridine-2-carboxylate with (hetero)aryl pinacol boranes, trifluoro potassium boronate salts or boronic acids. Their antitumoral potential was evaluated in two triple negative breast cancer (TNBC) cell lines—MDA-MB-231 and MDA-MB-468, by sulforhodamine B assay. Their effects on the non-tumorigenic MCF-12A cells were also evaluated. The results demonstrated that three compounds caused growth inhibition in both TNBC cell lines, with little or no effect against the non-tumorigenic cells. The most promising compound was further studied concerning possible effects on cell viability (by trypan blue exclusion assay), cell proliferation (by bromodeoxyuridine assay) and cell cycle profile (by flow cytometry). The results demonstrated that the GI50 concentration of compound 2e (13 μM) caused a decreased in MDA-MB-231 cell number, which was correlated with a decreased in the % of proliferating cells. Moreover, this compound increased G0/G1 phase and decreased S phases, when compared to control cells (although was not statistic significant). Interestingly, compound 2e also reduced tumor size using an in ovo CAM (chick chorioallantoic membrane) model. This work highlights the potential antitumor effect of a novel methyl 3-arylthieno[3,2-b]pyridine-2-carboxylate derivative.
Collapse
Affiliation(s)
- Bruna R. Silva
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Rebelo
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Juliana M. Rodrigues
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence: (C.P.R.X.); (M.-J.R.P.Q.)
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (M.H.V.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria-João R. P. Queiroz
- Centre of Chemistry, University of Minho, 4710-057 Braga, Portugal; (B.R.S.); (J.M.R.)
- Correspondence: (C.P.R.X.); (M.-J.R.P.Q.)
| |
Collapse
|
21
|
Digiacomo G, Fumarola C, La Monica S, Bonelli MA, Cretella D, Alfieri R, Cavazzoni A, Galetti M, Bertolini P, Missale G, Petronini PG. Simultaneous Combination of the CDK4/6 Inhibitor Palbociclib With Regorafenib Induces Enhanced Anti-tumor Effects in Hepatocarcinoma Cell Lines. Front Oncol 2020; 10:563249. [PMID: 33072590 PMCID: PMC7539564 DOI: 10.3389/fonc.2020.563249] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/20/2020] [Indexed: 12/28/2022] Open
Abstract
Advanced hepatocarcinoma (HCC) is an aggressive malignancy with poor prognosis and limited treatment options. Alterations of the cyclin D-CDK4/6-Rb pathway occur frequently in HCC, providing the rationale for its targeting at least in a molecular subset of HCC. In a panel of HCC cell lines, we investigated whether the CDK4/6 inhibitor palbociclib might improve the efficacy of regorafenib, a powerful multi-kinase inhibitor approved as second-line treatment for advanced HCC after sorafenib failure and currently under clinical investigation as first-line therapy in combination with immunotherapy. In Rb-proficient cells, the simultaneous drug combination, but not the sequential schedules, inhibited cell proliferation, either in short or in long-term experiments, and induced cell death more strongly than individual treatments. Moreover, the combination significantly reduced spheroid cell growth and inhibited cell migration/invasion. The superior efficacy of palbociclib plus regorafenib emerged also under hypoxia and was associated with a significant down-regulation of CDK4/6-Rb-myc and mTORC1/p70S6K signaling. Moreover, regorafenib suppressed palbociclib-induced expression of cyclin D1 contributing to the cytotoxic effects of the combination. Besides these inhibitory effects on cell viability/proliferation, palbociclib and regorafenib reduced glucose uptake, although this effect was dependent on the cell model and on the oxygen availability (normoxia or hypoxia). Palbociclib and regorafenib combination impaired glucose uptake and utilization, down-regulating basal and hypoxia-induced expression of HIF-1α, HIF-2α, GLUT-1, and MCT4 proteins as well as the activity/expression of glycolytic enzymes (HK2, PFKP, aldolase A, PKM2). In addition, regorafenib alone reduced mitochondrial respiration. The combined treatment impaired glucose metabolism and respiration without enhancing the effects of the single agents. Our findings provide pre-clinical evidence for the effectiveness of palbociclib and regorafenib combination in HCC cell models.
Collapse
Affiliation(s)
| | - Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mara A Bonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Daniele Cretella
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maricla Galetti
- Istituto Nazionale per l'Assicurazione contro gli Infortuni sul Lavoro (INAIL) Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Rome, Italy
| | - Patrizia Bertolini
- Paediatric Hematology Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Gabriele Missale
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Unit of Infectious Diseases and Hepatology, University Hospital of Parma, Parma, Italy
| | | |
Collapse
|
22
|
Bonelli M, Terenziani R, Zoppi S, Fumarola C, La Monica S, Cretella D, Alfieri R, Cavazzoni A, Digiacomo G, Galetti M, Petronini PG. Dual Inhibition of CDK4/6 and PI3K/AKT/mTOR Signaling Impairs Energy Metabolism in MPM Cancer Cells. Int J Mol Sci 2020; 21:ijms21145165. [PMID: 32708306 PMCID: PMC7403976 DOI: 10.3390/ijms21145165] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive malignancy associated to asbestos exposure. One of the most frequent genetic alteration in MPM patients is CDKN2A/ARF loss, leading to aberrant activation of the Rb pathway. In MPM cells, we previously demonstrated the therapeutic efficacy of targeting this signaling with the CDK4/6 inhibitor palbociclib in combination with PI3K/mTOR inhibitors. Here, we investigated whether such combination may have an impact on cell energy metabolism. Methods: The study was performed in MPM cells of different histotypes; metabolic analyses were conducted by measuring GLUT-1 expression and glucose uptake/consumption, and by SeaHorse technologies. Results: MPM cell models differed for their ability to adapt to metabolic stress conditions, such as glucose starvation and hypoxia. Independently of these differences, combined treatments with palbociclib and PI3K/mTOR inhibitors inhibited cell proliferation more efficaciously than single agents. The drugs alone reduced glucose uptake/consumption as well as glycolysis, and their combination further enhanced these effects under both normoxic and hypoxic conditions. Moreover, the drug combinations significantly impaired mitochondrial respiration as compared with individual treatments. These metabolic effects were mediated by the concomitant inhibition of Rb/E2F/c-myc and PI3K/AKT/mTOR signaling. Conclusions: Dual blockade of glycolysis and respiration contributes to the anti-tumor efficacy of palbociclib-PI3K/mTOR inhibitors combination.
Collapse
Affiliation(s)
- Mara Bonelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Rita Terenziani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Silvia Zoppi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
- Correspondence: ; Tel.: +39-0521-903762
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Daniele Cretella
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Graziana Digiacomo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| | - Maricla Galetti
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, 00078 Monte Porzio Catone (Rome), Italy;
| | - Pier Giorgio Petronini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.B.); (R.T.); (S.Z.); (S.L.M.); (D.C.); (R.A.); (A.C.); (G.D.); (P.G.P.)
| |
Collapse
|
23
|
Therapeutic aspects of AMPK in breast cancer: Progress, challenges, and future directions. Biochim Biophys Acta Rev Cancer 2020; 1874:188379. [PMID: 32439311 DOI: 10.1016/j.bbcan.2020.188379] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most ubiquitous type of neoplasms among women worldwide. Molecular aberrations associated with breast development and progressions have been extensively investigated in recent years. An AMP-activated kinase (AMPK) initially identified as a cellular energy sensor that plays a crucial role in cellular energy homeostasis. Intensive research over the last decade about the molecular mechanisms of AMPK has demonstrated that AMPK mediated diverse biological functions are achieved through phosphorylation and regulation of multiple downstream signaling molecules in normal tissue. Downregulation of AMPK activity or decreased level involved in the promotion of breast tumorigenesis, and thus activation of AMPK found to oppose tumor progression. In this review, we epitomize the recent advances in exploring the tumor suppressor function of AMPK pathways. Besides, we discuss the developments in the area of AMPK activator and its molecular mechanisms for breast cancer treatment.
Collapse
|
24
|
Ocaña MC, Martínez-Poveda B, Marí-Beffa M, Quesada AR, Medina MÁ. Fasentin diminishes endothelial cell proliferation, differentiation and invasion in a glucose metabolism-independent manner. Sci Rep 2020; 10:6132. [PMID: 32273578 PMCID: PMC7145862 DOI: 10.1038/s41598-020-63232-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
The synthetic compound fasentin has been described as a modulator of GLUT-1 and GLUT-4 transporters, thus inhibiting glucose uptake in some cancer cells. Endothelial glucose metabolism has been recently connected to angiogenesis and it is now an emerging topic in scientific research. Indeed, certain compounds with a known effect on glucose metabolism have also been shown to inhibit angiogenesis. In this work we tested the capability of fasentin to modulate angiogenesis in vitro and in vivo. We show that fasentin inhibited tube formation in endothelial cells by a mechanism that involves a negative effect on endothelial cell proliferation and invasion, without affecting other steps related to the angiogenic process. However, fasentin barely decreased glucose uptake in human dermal microvascular endothelial cells and the GLUT-1 inhibitor STF-31 failed to inhibit tube formation in these cells. Therefore, this modulatory capacity on endothelial cells function exerted by fasentin is most likely independent of a modulation of glucose metabolism. Taken together, our results show a novel biological activity of fasentin, which could be evaluated for its utility in cancer and other angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Mª Carmen Ocaña
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain
| | - Beatriz Martínez-Poveda
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain
| | - Manuel Marí-Beffa
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, E-29071, Málaga, Spain
| | - Ana R Quesada
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain
| | - Miguel Ángel Medina
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071, Málaga, Spain.
- IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain.
- CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain.
| |
Collapse
|
25
|
Antunes F, Pereira GJS, Saito RF, Buri MV, Gagliardi M, Bincoletto C, Chammas R, Fimia GM, Piacentini M, Corazzari M, Smaili SS. Effective Synergy of Sorafenib and Nutrient Shortage in Inducing Melanoma Cell Death through Energy Stress. Cells 2020; 9:E640. [PMID: 32155825 PMCID: PMC7140454 DOI: 10.3390/cells9030640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
Skin melanoma is one of the most aggressive and difficult-to-treat human malignancies, characterized by poor survival rates, thus requiring urgent novel therapeutic approaches. Although metabolic reprogramming has represented so far, a cancer hallmark, accumulating data indicate a high plasticity of cancer cells in modulating cellular metabolism to adapt to a heterogeneous and continuously changing microenvironment, suggesting a novel therapeutic approach for dietary manipulation in cancer therapy. To this aim, we exposed melanoma cells to combined nutrient-restriction/sorafenib. Results indicate that cell death was efficiently induced, with apoptosis representing the prominent feature. In contrast, autophagy was blocked in the final stage by this treatment, similarly to chloroquine, which also enhanced melanoma cell sensitization to combined treatment. Energy stress was evidenced by associated treatment with mitochondrial dysfunction and glycolysis impairment, suggesting metabolic stress determining melanoma cell death. A reduction of tumor growth after cycles of intermittent fasting together with sorafenib treatment was also observed in vivo, reinforcing that the nutrient shortage can potentiate anti-melanoma therapy. Our findings showed that the restriction of nutrients by intermittent fasting potentiates the effects of sorafenib due to the modulation of cellular metabolism, suggesting that it is possible to harness the energy of cancer cells for the treatment of melanoma.
Collapse
Affiliation(s)
- Fernanda Antunes
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Gustavo J. S. Pereira
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Renata F. Saito
- Center for Translational Research in Oncology, Department of Radiology and Oncology, Faculty of Medicine of the University of São Paulo and Cancer Institute of the State of São Paulo, São Paulo 04021-001, Brazil; (R.F.S.); (R.C.)
| | - Marcus V. Buri
- Department of Molecular Biology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil;
| | - Mara Gagliardi
- Department of Health Sciences (DISS), University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), 28100 Novara, Italy
| | - Claudia Bincoletto
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Roger Chammas
- Center for Translational Research in Oncology, Department of Radiology and Oncology, Faculty of Medicine of the University of São Paulo and Cancer Institute of the State of São Paulo, São Paulo 04021-001, Brazil; (R.F.S.); (R.C.)
| | - Gian Maria Fimia
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘Lazzaro Spallanzani’, 00149 Rome, Italy; (G.M.F.); (M.P.)
- Department of Molecular Medicine, University of Rome La Sapienza, 00185 Rome, Italy
| | - Mauro Piacentini
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘Lazzaro Spallanzani’, 00149 Rome, Italy; (G.M.F.); (M.P.)
- Institute of Cytology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Marco Corazzari
- Department of Health Sciences (DISS), University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), 28100 Novara, Italy
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| |
Collapse
|
26
|
Barbosa AM, Martel F. Targeting Glucose Transporters for Breast Cancer Therapy: The Effect of Natural and Synthetic Compounds. Cancers (Basel) 2020; 12:cancers12010154. [PMID: 31936350 PMCID: PMC7016663 DOI: 10.3390/cancers12010154] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Reprogramming of cellular energy metabolism is widely accepted to be a cancer hallmark. The deviant energetic metabolism of cancer cells-known as the Warburg effect-consists in much higher rates of glucose uptake and glycolytic oxidation coupled with the production of lactic acid, even in the presence of oxygen. Consequently, cancer cells have higher glucose needs and thus display a higher sensitivity to glucose deprivation-induced death than normal cells. So, inhibitors of glucose uptake are potential therapeutic targets in cancer. Breast cancer is the most commonly diagnosed cancer and a leading cause of cancer death in women worldwide. Overexpression of facilitative glucose transporters (GLUT), mainly GLUT1, in breast cancer cells is firmly established, and the consequences of GLUT inhibition and/or knockout are under investigation. Herein we review the compounds, both of natural and synthetic origin, found to interfere with uptake of glucose by breast cancer cells, and the consequences of interference with that mechanism on breast cancer cell biology. We will also present data where the interaction with GLUT is exploited in order to increase the efficiency or selectivity of anticancer agents, in breast cancer cells.
Collapse
Affiliation(s)
- Ana M. Barbosa
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4169-007 Porto, Portugal;
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Correspondence: ; Tel.: +351-22-042-6654
| |
Collapse
|
27
|
Covalent triazine-based polyimine framework as a biocompatible pH-dependent sustained-release nanocarrier for sorafenib: An in vitro approach. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2019.111898] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
28
|
Transferrin-Modified Nanoliposome Codelivery Strategies for Enhancing the Cancer Therapy. J Pharm Sci 2019; 109:2426-2436. [PMID: 31760084 DOI: 10.1016/j.xphs.2019.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/22/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
Chemotherapy remains one of the most effective treatments for many cancers in a clinic. At present, various targets have been used to modify the PEGylated liposomes for doxorubicin (Dox) delivery, but the antitumor effect of Dox is not satisfactory. Therefore, combination chemotherapeutics has been considered as a promising method to improve tumor treatment. These years, RAF/MEK/ERK-mediated cell signaling pathway has been discovered to inhibit the growth of tumors. Thus, Sorafenib tosylate (Sor) was used in this study, which directly inhibited tumor cell proliferation through blocking RAF/MEK/ERK-mediated cell signaling pathway and indirectly inhibited tumor cell growth through blocking angiogenesis by VEGFR and PDGF. In this article, we develop a "combination delivery system" to deliver the hydrophobic drug (Sor) in phospholipid bilayer and hydrophilic drug (Dox) in inner cores for enhancing the antitumor effect. Moreover, in vitro experiments verified whether the physicochemical properties of carriers were stable and transferrin-modified liposomes displayed the highest uptake. The results of in vivo experiments showed that the codelivery system inhibited the tumor growth more effectively than monotherapy. Overall, this combination delivery system for delivering the hydrophobic and hydrophilic drugs simultaneously may offer a novel strategy for breast cancer treatment and provide a reference for the possibility of clinical usage.
Collapse
|
29
|
Ma W, Liu M, Liang F, Zhao L, Gao C, Jiang X, Zhang X, Zhan H, Hu H, Zhao Z. Cardiotoxicity of sorafenib is mediated through elevation of ROS level and CaMKII activity and dysregulation of calcium homoeostasis. Basic Clin Pharmacol Toxicol 2019; 126:166-180. [DOI: 10.1111/bcpt.13318] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Wenzhuo Ma
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Mei Liu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Fanfan Liang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Lili Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Chenying Gao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xixi Jiang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xin Zhang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Heqin Zhan
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
- Department of Pharmacology College of Pharmacy Xinxiang Medical University Xinxiang Henan 453003 China
| | - Hao Hu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Zhenghang Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| |
Collapse
|
30
|
Zhang Y, Zhou X, Cheng L, Wang X, Zhang Q, Zhang Y, Sun S. PRKAA1 Promotes Proliferation and Inhibits Apoptosis of Gastric Cancer Cells Through Activating JNK1 and Akt Pathways. Oncol Res 2019; 28:213-223. [PMID: 31558185 PMCID: PMC7851536 DOI: 10.3727/096504019x15668125347026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PRKAA1 (protein kinase AMP-activated catalytic subunit α 1) is a catalytic subunit of AMP-activated protein kinase (AMPK), which plays a key role in regulating cellular energy metabolism through phosphorylation, and genetic variations in the PRKAA1 have been found to be associated with gastric cancer risk. However, the effect and underlying molecular mechanism of PRKAA1 on gastric cancer tumorigenesis, especially the proliferation and apoptosis, are not fully understood. Our data showed that PRKAA1 is highly expressed in BGC-823 and MKN45 cells and is expressed low in SGC-7901 and MGC-803 cells in comparison with the other gastric cancer cells. PRKAA1 downregulation by shRNA or treatment of AMPK inhibitor compound C significantly inhibited proliferation as well as promoted cell cycle arrest and apoptosis of BGC-823 and MKN45 cells. Moreover, the expression of PCNA and Bcl-2 and the activity of JNK1 and Akt signaling were also reduced in BGC-823 and MKN45 cells after PRKAA1 downregulation. In vivo experiments demonstrated that tumor growth in nude mice was significantly inhibited after PRKAA1 silencing. Importantly, inactivation of JNK1 or Akt signaling pathway significantly inhibited PRKAA1 overexpression-induced increased cell proliferation and decreased cell apoptosis in MGC-803 cells. In conclusion, our findings suggest that PRKAA1 increases proliferation and restrains apoptosis of gastric cancer cells through activating JNK1 and Akt pathways.
Collapse
Affiliation(s)
- Yangmei Zhang
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Xichang Zhou
- Department of Intervention, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Long Cheng
- Department of Intervention, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Xiang Wang
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Qinglin Zhang
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Youwei Zhang
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| | - Sanyuan Sun
- Department of Oncology, Xuzhou Central Hospital, Xuzhou Medical UniversityXuzhouP.R. China
| |
Collapse
|
31
|
Hu M, Chen X, Ma L, Ma Y, Li Y, Song H, Xu J, Zhou L, Li X, Jiang Y, Kong B, Huang P. AMPK Inhibition Suppresses the Malignant Phenotype of Pancreatic Cancer Cells in Part by Attenuating Aerobic Glycolysis. J Cancer 2019; 10:1870-1878. [PMID: 31205544 PMCID: PMC6547975 DOI: 10.7150/jca.28299] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is a highly aggressive tumor characterized by enhanced aerobic glycolysis. AMP-activated protein kinase (AMPK), which is identified as a well-known regulator of glycolysis, plays an essential role in tumorigenesis. In the present study, we aim to explore the function of AMPK in pancreatic cancer cells and attempt to clarify the possible underlying mechanism. The Cancer Genome Atlas (TCGA) data showed that elevated AMPK expression highly correlated with lower median survival time. In an in vitro study, inhibition of AMPK blocked the proliferation, migration, and invasion ability of four cell lines under normoxia and hypoxia. Additionally, AMPK suppression led to cell cycle arrest and remarkably induced apoptosis. Furthermore, the lactic acid content, ATP content, and the glucose consumption rate were significantly reduced in all four cell lines under different conditions, accompanied by down-regulation of glycolytic biomarkers including phosphorylated mammalian target of rapamycin (p-mTOR)/total mTOR (t-mTOR), Pyruvate kinase M2 (Pkm2), and Hexokinase 2 (Hk2). Collectively, our data showed that AMPK activation is highly involved in pancreatic cancer progression and exerts its pro-tumorigenic functions partly by sustaining glycolytic activity. Hence, AMPK is expected to be a potential therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Mingyue Hu
- Medical School of Southeast University, Nanjing 210009, China
| | - Xiangxu Chen
- Medical School of Southeast University, Nanjing 210009, China
| | - Li Ma
- Medical School of Southeast University, Nanjing 210009, China
| | - Yu Ma
- Medical School of Southeast University, Nanjing 210009, China
| | - Yuan Li
- Medical School of Southeast University, Nanjing 210009, China
| | - Huihui Song
- Medical School of Southeast University, Nanjing 210009, China
| | - Jiajia Xu
- Medical School of Southeast University, Nanjing 210009, China
| | - Lingna Zhou
- Medical School of Southeast University, Nanjing 210009, China
| | - Xiaoxue Li
- Medical School of Southeast University, Nanjing 210009, China
| | - Yuhui Jiang
- Medical School of Southeast University, Nanjing 210009, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich 81675, Germany.,Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Peilin Huang
- Medical School of Southeast University, Nanjing 210009, China
| |
Collapse
|
32
|
Chenlo M, Rodriguez-Gomez IA, Serramito R, Garcia-Rendueles AR, Villar-Taibo R, Fernandez-Rodriguez E, Perez-Romero S, Suarez-Fariña M, Garcia-Allut A, Cabezas-Agricola JM, Rodriguez-Garcia J, Lear PV, Alvarez-San Martin RM, Alvarez-Escola C, Bernabeu I, Alvarez CV. Unmasking a new prognostic marker and therapeutic target from the GDNF-RET/PIT1/p14ARF/p53 pathway in acromegaly. EBioMedicine 2019; 43:537-552. [PMID: 30975543 PMCID: PMC6562173 DOI: 10.1016/j.ebiom.2019.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/29/2022] Open
Abstract
Background Acromegaly is produced by excess growth hormone secreted by a pituitary adenoma of somatotroph cells (ACRO). First-line therapy, surgery and adjuvant therapy with somatostatin analogs, fails in 25% of patients. There is no predictive factor of resistance to therapy. New therapies are investigated using few dispersed tumor cells in acute primary cultures in standard conditions where the cells do not grow, or using rat pituitary cell lines that do not maintain the full somatotroph phenotype. The RET/PIT1/p14ARF/p53 pathway regulates apoptosis in normal pituitary somatotrophs whereas the RET/GDNF pathway regulates survival, controlling PIT1 levels and blocking p14ARF (ARF) and p53 expression. Methods We investigated these two RET pathways in a prospective series of 32 ACRO and 63 non-functioning pituitary adenomas (NFPA), studying quantitative RNA and protein gene expression for molecular-clinical correlations and how the RET pathway might be implicated in therapeutic success. Clinical data was collected during post-surgical follow-up. We also established new'humanized’ pituitary cultures, allowing 20 repeated passages and maintaining the pituitary secretory phenotype, and tested five multikinase inhibitors (TKI: Vandetanib, Lenvatinib, Sunitinib, Cabozantinib and Sorafenib) potentially able to act on the GDNF-induced RET dimerization/survival pathway. Antibody arrays investigated intracellular molecular pathways. Findings In ACRO, there was specific enrichment of all genes in both RET pathways, especially GDNF. ARF and GFRA4 gene expression were found to be opposing predictors of response to first-line therapy. ARF cut-off levels, calculated categorizing by GNAS mutation, were predictive of good response (above) or resistance (below) to therapy months later. Sorafenib, through AMPK, blocked the GDNF/AKT survival action without altering the RET apoptotic pathway. Interpretation Tumor ARF mRNA expression measured at the time of the surgery is a prognosis factor in acromegaly. The RET inhibitor, Sorafenib, is proposed as a potential treatment for resistant ACRO. Fund This project was supported by national grants from Agencia Estatal de Investigación (AEI) and Instituto Investigación Carlos III, with participation of European FEDER funds, to IB (PI150056) and CVA (BFU2016-76973-R). It was also supported initially by a grant from the Investigator Initiated Research (IIR) Program (WI177773) and by a non-restricted Research Grant from Pfizer Foundation to IB. Some of the pituitary acromegaly samples were collected in the framework of the Spanish National Registry of Acromegaly (REMAH), partially supported by an unrestricted grant from Novartis to the Spanish Endocrine Association (SEEN). CVA is also supported from a grant of Medical Research Council UK MR/M018539/1.
Collapse
Affiliation(s)
- Miguel Chenlo
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Iria A Rodriguez-Gomez
- Servicio de Endocrinología y Nutrición, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Servicio de Endocrinología y Nutrición, Hospital HM Modelo, A Coruña, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Ramon Serramito
- Servicio de Neurocirugía, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Angela R Garcia-Rendueles
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Rocío Villar-Taibo
- Servicio de Endocrinología y Nutrición, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Eva Fernandez-Rodriguez
- Servicio de Endocrinología y Nutrición, Complejo Hospitalario Universitario de Ourense, Spain
| | - Sihara Perez-Romero
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Maria Suarez-Fariña
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Alfredo Garcia-Allut
- Servicio de Neurocirugía, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Jose M Cabezas-Agricola
- Servicio de Endocrinología y Nutrición, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Javier Rodriguez-Garcia
- Servicio de Análisis Clínicos, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Pamela V Lear
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom
| | | | | | - Ignacio Bernabeu
- Servicio de Endocrinología y Nutrición, Complejo Hospitalario Universitario de Santiago de Compostela (CHUS)-SERGAS, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain.
| | - Clara V Alvarez
- Neoplasia & Endocrine Differentiation P0L5, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela (USC), Spain; Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
33
|
Fumarola C, Bozza N, Castelli R, Ferlenghi F, Marseglia G, Lodola A, Bonelli M, La Monica S, Cretella D, Alfieri R, Minari R, Galetti M, Tiseo M, Ardizzoni A, Mor M, Petronini PG. Expanding the Arsenal of FGFR Inhibitors: A Novel Chloroacetamide Derivative as a New Irreversible Agent With Anti-proliferative Activity Against FGFR1-Amplified Lung Cancer Cell Lines. Front Oncol 2019; 9:179. [PMID: 30972293 PMCID: PMC6443895 DOI: 10.3389/fonc.2019.00179] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fibroblast Growth Factor Receptors (FGFR1-4) have a critical role in the progression of several human cancers, including Squamous Non-Small-Cell Lung Cancer (SQCLC). Both non-selective and selective reversible FGFR inhibitors are under clinical investigation for the treatment of patients with tumors harboring FGFR alterations. Despite their potential efficacy, the clinical development of these drugs has encountered several challenges, including toxicity, and the appearance of drug resistance. Recent efforts have been directed at development of irreversible FGFR inhibitors, which have the potential to exert superior anti-proliferative activity in tumors carrying FGFR alterations. With this in mind, we synthetized, and investigated a set of novel inhibitors possessing a warhead potentially able to covalently bind a cysteine in the P-loop of FGFR. Among them, the chloroacetamide UPR1376 resulted able to irreversible inhibit FGFR1 phosphorylation in FGFR1 over-expressing cells generated from SQCLC SKMES-1 cells. In addition, this compound inhibited cell proliferation in FGFR1-amplified H1581 cells with a potency higher than the reversible inhibitor BGJ398 (infigratinib), while sparing FGFR1 low-expressing cells. The anti-proliferative effects of UPR1376 were demonstrated in both 2D and 3D systems and were associated with the inhibition of MAPK and AKT/mTOR signaling pathways. UPR1376 inhibited cell proliferation also in two BGJ398-resistant cell clones generated from H1581 by chronic exposure to BGJ398, although at concentrations higher than those effective in the parental cells, likely due to the persistent activation of the MAPK pathway associated to NRAS amplification. Combined blockade of FGFR1 and MAPK signaling, by UPR1376 and trametinib respectively, significantly enhanced the efficacy of UPR1376, providing a means of circumventing resistance to FGFR1 inhibition. Our findings suggest that the insertion of a chloroacetamide warhead on a suitable scaffold, as exemplified by UPR1376, is a valuable strategy to develop a novel generation of FGFR inhibitors for the treatment of SQCLC patients with FGFR alterations.
Collapse
Affiliation(s)
- Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicole Bozza
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | | | | | - Alessio Lodola
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Daniele Cretella
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Maricla Galetti
- Italian Workers' Compensation Authority (INAIL) Research Center, Parma, Italy.,Department of Medicine and Surgery, Center of Excellence for Toxicological Research, University of Parma, Parma, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Andrea Ardizzoni
- Division of Medical Oncology, Sant'Orsola-Malpighi University Hospital and Alma Mater University of Bologna, Bologna, Italy
| | - Marco Mor
- Department of Food and Drug, University of Parma, Parma, Italy
| | | |
Collapse
|
34
|
Dai J, Huang Q, Niu K, Wang B, Li Y, Dai C, Chen Z, Tao K, Dai J. Sestrin 2 confers primary resistance to sorafenib by simultaneously activating AKT and AMPK in hepatocellular carcinoma. Cancer Med 2018; 7:5691-5703. [PMID: 30311444 PMCID: PMC6247041 DOI: 10.1002/cam4.1826] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/04/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the malignancy derived from normal hepatocytes with increasing incidence and extremely poor prognosis worldwide. The only approved first‐line systematic treatment agent for HCC, sorafenib, is capable to effectively improve advanced HCC patients’ survival. However, it is gradually recognized that the therapeutic response to sorafenib could be drastically diminished after short‐term treatment, defined as primary resistance. The present study is aimed to explore the role of stress‐inducible protein Sestrin2 (SESN2), one of the most important sestrins family members, in sorafenib primary resistance. Herein, we initially found that SESN2 expression was significantly up‐regulated in both HCC cell lines and tissues compared to normal human hepatocytes and corresponding adjacent liver tissues, respectively. In addition, SESN2 expression was highly correlated with sorafenib IC50 of HCC cell lines. Thereafter, we showed that sorafenib treatment resulted in an increase of SESN2 expression and the knockdown of SESN2 exacerbated sorafenib‐induced proliferation inhibition and cell apoptosis. Further mechanistic study uncovered that SESN2 deficiency impaired both AKT and AMPK phosphorylation and activation after sorafenib treatment. Moreover, the correlations between SESN2 expression and both phosphor‐AKT and phosphor‐AMPK expression were illustrated in HCC tissues. Taken together, our study demonstrates that SESN2 activates AKT and AMPK signaling as a novel mechanism to induce sorafenib primary resistance in HCC.
Collapse
Affiliation(s)
- Jimin Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China.,The Cadet Team 6 (Regiment 6) of School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qichao Huang
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Kunwei Niu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yijie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Chen Dai
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Air Force Medical University, Xi'an, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jingyao Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China.,Department of Cell Biology, National Translational Science Center for Molecular Medicine, Air Force Medical University, Xi'an, China
| |
Collapse
|
35
|
Poulose N, Mills IG, Steele RE. The impact of transcription on metabolism in prostate and breast cancers. Endocr Relat Cancer 2018; 25:R435-R452. [PMID: 29760165 DOI: 10.1530/erc-18-0048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
Abstract
Metabolic dysregulation is regarded as an important driver in cancer development and progression. The impact of transcriptional changes on metabolism has been intensively studied in hormone-dependent cancers, and in particular, in prostate and breast cancer. These cancers have strong similarities in the function of important transcriptional drivers, such as the oestrogen and androgen receptors, at the level of dietary risk and epidemiology, genetics and therapeutically. In this review, we will focus on the function of these nuclear hormone receptors and their downstream impact on metabolism, with a particular focus on lipid metabolism. We go on to discuss how lipid metabolism remains dysregulated as the cancers progress. We conclude by discussing the opportunities that this presents for drug repurposing, imaging and the development and testing of new therapeutics and treatment combinations.
Collapse
Affiliation(s)
- Ninu Poulose
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
| | - Ian G Mills
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
- Nuffield Department of Surgical SciencesJohn Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Rebecca E Steele
- Centre for Cancer Research and Cell BiologyQueen's University of Belfast, Belfast, UK
| |
Collapse
|
36
|
Damaraju VL, Kuzma M, Cass CE, Putman CT, Sawyer MB. Multitargeted kinase inhibitors imatinib, sorafenib and sunitinib perturb energy metabolism and cause cytotoxicity to cultured C2C12 skeletal muscle derived myotubes. Biochem Pharmacol 2018; 155:162-171. [PMID: 29983397 DOI: 10.1016/j.bcp.2018.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/01/2018] [Indexed: 12/12/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) have advanced cancer treatment and prognosis but have also resulted in adverse effects such as fatigue, diarrhea, hypothyroidism, and other toxicities. We investigated TKI effects on skeletal muscle as a possible explanation of TKI induced fatigue. Changes in mitochondrial function due to inhibition of oxidative phosphorylation complexes, generation of superoxides, and inhibition of key transporters involved in uptake of glucose and/or nucleosides may result in alteration of energy metabolism and/or mitochondrial function. We investigated effects of imatinib, sorafenib and sunitinib on these processes in cultured C2C12 murine skeletal muscle cells. Imatinib, sorafenib and sunitinib were cytotoxic to C2C12 cells with IC50 values of 20, 8 and 8 µM, respectively. Imatinib stimulated glucose uptake and inhibited complex V activity by 35% at 50 µM. Sorafenib inhibited complex II/III and V with IC50 values of 32 and 28 µM, respectively. Sorafenib caused activation of caspase 3/7 and depolarization of mitochondrial membranes occurred very rapidly with complete loss at 5-10 µM. Sunitinib inhibited Complex I with an IC50 value of 38 µM and caused ATP depletion, caspase 3/7 activation, an increase in reactive oxygen species (ROS), and decreased nucleoside and glucose uptake. In conclusion, imatinib, sunitinib and sorafenib caused changes in mitochondrial complex activities, glucose and nucleoside uptake leading to decreased energy production and mitochondrial function in a skeletal muscle cell model, suggesting that these changes may play a role in fatigue, one of the most common adverse effects of TKIs.
Collapse
Affiliation(s)
- Vijaya L Damaraju
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave NW, Edmonton, Alberta T6G1Z2, Canada.
| | - Michelle Kuzma
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave NW, Edmonton, Alberta T6G1Z2, Canada.
| | - Carol E Cass
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave NW, Edmonton, Alberta T6G1Z2, Canada.
| | - Charles T Putman
- Faculty of Kinesiology, Sport, and Recreation
- Medicine & Dentistry, University of Alberta, Edmonton, Alberta T6G2H9, Canada.
| | - Michael B Sawyer
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave NW, Edmonton, Alberta T6G1Z2, Canada; Department of Medical Oncology, Cross Cancer Institute, 11560 University Avenue NW, Edmonton, Alberta T6G1Z2, Canada.
| |
Collapse
|
37
|
Chhipa RR, Fan Q, Anderson J, Muraleedharan R, Huang Y, Ciraolo G, Chen X, Waclaw R, Chow LM, Khuchua Z, Kofron M, Weirauch MT, Kendler A, McPherson C, Ratner N, Nakano I, Dasgupta N, Komurov K, Dasgupta B. AMP kinase promotes glioblastoma bioenergetics and tumour growth. Nat Cell Biol 2018; 20:823-835. [PMID: 29915361 PMCID: PMC6113057 DOI: 10.1038/s41556-018-0126-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/18/2018] [Indexed: 12/19/2022]
Abstract
Stress is integral to tumour evolution, and cancer cell survival depends on stress management. We found that cancer-associated stress chronically activates the bioenergetic sensor AMP kinase (AMPK) and, to survive, tumour cells hijack an AMPK-regulated stress response pathway conserved in normal cells. Analysis of The Cancer Genome Atlas data revealed that AMPK isoforms are highly expressed in the lethal human cancer glioblastoma (GBM). We show that AMPK inhibition reduces viability of patient-derived GBM stem cells (GSCs) and tumours. In stressed (exercised) skeletal muscle, AMPK is activated to cooperate with CREB1 (cAMP response element binding protein-1) and promote glucose metabolism. We demonstrate that oncogenic stress chronically activates AMPK in GSCs that coopt the AMPK-CREB1 pathway to coordinate tumour bioenergetics through the transcription factors HIF1α and GABPA. Finally, we show that adult mice tolerate systemic deletion of AMPK, supporting the use of AMPK pharmacological inhibitors in the treatment of GBM.
Collapse
Affiliation(s)
- Rishi Raj Chhipa
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, PA, USA
| | - Qiang Fan
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jane Anderson
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Yan Huang
- Division of Molecular and Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Georgianne Ciraolo
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Chen
- Division of Center for Autoimmune Genomics and Etiology and Biomedical Informatics and Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ronald Waclaw
- Division of Experimental Hematology and Cancer Biology, Cincinnati, OH, USA
| | - Lionel M Chow
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zaza Khuchua
- Division of Molecular and Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Sechenov University, Department of Biochemistry, Moscow, Russian Federation
| | - Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Center for Autoimmune Genomics and Etiology and Biomedical Informatics and Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ady Kendler
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Christopher McPherson
- Department of Neurosurgery, Brain Tumor Center, University of Cincinnati Neuroscience Institute and Mayfield Clinic, Cincinnati, OH, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati, OH, USA
| | - Ichiro Nakano
- Department of Neurosurgery, University of Alabama, Cincinnati, OH, USA
| | - Nupur Dasgupta
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kakajan Komurov
- Division of Experimental Hematology and Cancer Biology, Cincinnati, OH, USA
| | - Biplab Dasgupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
38
|
Zanotto-Filho A, Rajamanickam S, Loranc E, Masamsetti VP, Gorthi A, Romero JC, Tonapi S, Gonçalves RM, Reddick RL, Benavides R, Kuhn J, Chen Y, Bishop AJR. Sorafenib improves alkylating therapy by blocking induced inflammation, invasion and angiogenesis in breast cancer cells. Cancer Lett 2018; 425:101-115. [PMID: 29608984 DOI: 10.1016/j.canlet.2018.03.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022]
Abstract
Molecular targeted compounds are emerging as a strategy to improve classical chemotherapy. Herein, we describe that using low dose of the multikinase inhibitor sorafenib improves cyclophosphamide antitumor activity by inhibiting angiogenesis, metastasis and promoting tumor healing in MDA-MB231 xenografts and the 4T1-12B syngeneic breast cancer metastasis model. Mechanistic studies in MDA-MB231 cells revealed that alkylation upregulates inflammatory genes/proteins such as COX-2, IL8, CXCL2 and MMP1 in a MEK1/2-ERK1/2-dependent manner. These proteins enrich the secretome of cancer cells, stimulating cell invasion and angiogenesis via autocrine and paracrine mechanisms. Sorafenib inhibits MEK1/2-ERK1/2 pathway thereby decreasing inflammatory genes and mitigating cell invasion and angiogenesis at basal and alkylation-induced conditions whereas NRF2 and ER stress pathways involved in alkylation survival are not affected. In non-invasive/non-angiogenic breast cancer cells (SKBR3 and MCF7), alkylation did not elicit inflammatory responses with the only sorafenib effect being ERK1/2-independent ROS-dependent cytotoxicity when using higher drug concentrations. In summary, our data show that alkylating agents may elicit inflammatory responses that seems to contribute to malignant progression in specific breast cancer cells. Identifying and targeting drivers of this phenotype may offer opportunities to optimize combined drug regimens between classical chemotherapeutics and targeted agents.
Collapse
Affiliation(s)
- Alfeu Zanotto-Filho
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Departamento de Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Subapriya Rajamanickam
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Eva Loranc
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - V Pragathi Masamsetti
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Aparna Gorthi
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - July Carolina Romero
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Sonal Tonapi
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Rosangela Mayer Gonçalves
- Departamento de Farmacologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Robert L Reddick
- Department of Pathology, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Raymond Benavides
- Department of Pathology, University of Texas College of Pharmacy, Austin, TX, USA
| | - John Kuhn
- Department of Pathology, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Pathology, University of Texas College of Pharmacy, Austin, TX, USA
| | - Yidong Chen
- Department of Epidemiology and Biostatistics, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Alexander J R Bishop
- Greehey Children's Cancer Research Institute, University of Texas Health at San Antonio, San Antonio, TX, USA; Department of Cell Systems and Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
39
|
Cretella D, Ravelli A, Fumarola C, La Monica S, Digiacomo G, Cavazzoni A, Alfieri R, Biondi A, Generali D, Bonelli M, Petronini PG. The anti-tumor efficacy of CDK4/6 inhibition is enhanced by the combination with PI3K/AKT/mTOR inhibitors through impairment of glucose metabolism in TNBC cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:72. [PMID: 29587820 PMCID: PMC5872523 DOI: 10.1186/s13046-018-0741-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Daniele Cretella
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Ravelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alessandra Biondi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy.,U.O. Multidisciplinare di Patologia Mammaria, U.S Terapia Molecolare e Farmacogenomica, ASST Cremona, Cremona, Italy
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
40
|
Fumarola C, Petronini PG, Alfieri R. Impairing energy metabolism in solid tumors through agents targeting oncogenic signaling pathways. Biochem Pharmacol 2018. [PMID: 29530507 DOI: 10.1016/j.bcp.2018.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell metabolic reprogramming is one of the main hallmarks of cancer and many oncogenic pathways that drive the cancer-promoting signals also drive the altered metabolism. This review focuses on recent data on the use of oncogene-targeting agents as potential modulators of deregulated metabolism in different solid cancers. Many drugs, originally designed to inhibit a specific target, then have turned out to have different effects involving also cell metabolism, which may contribute to the mechanisms underlying the growth inhibitory activity of these drugs. Metabolic reprogramming may also represent a way by which cancer cells escape from the selective pressure of targeted drugs and become resistant. Here we discuss how targeting metabolism could emerge as a new effective strategy to overcome such resistance. Finally, accumulating evidence indicates that cancer metabolic rewiring may have profound effects on tumor-infiltrating immune cells. Modulating cancer metabolic pathways through oncogene-targeting agents may not only restore more favorable conditions for proper lymphocytes activation, but also increase the persistence of memory T cells, thereby improving the efficacy of immune-surveillance.
Collapse
Affiliation(s)
- Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | | | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
41
|
Zheng M, Xu H, Liao XH, Chen CP, Zhang AL, Lu W, Wang L, Yang D, Wang J, Liu H, Zhou XZ, Lu KP. Inhibition of the prolyl isomerase Pin1 enhances the ability of sorafenib to induce cell death and inhibit tumor growth in hepatocellular carcinoma. Oncotarget 2018; 8:29771-29784. [PMID: 28404959 PMCID: PMC5444702 DOI: 10.18632/oncotarget.15967] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer, but is the second leading cause of cancer deaths, partially due to its heterogeneity and drug resistance. Sorafenib is the only medical treatment with a proven efficacy against advanced HCC, but its overall clinical efficacy is still modest. Therefore, a major challenge is how to improve its therapeutic efficacy. The unique prolyl isomerase Pin1 regulates numerous cancer-driving pathways. Notably, Pin1 is overexpressed in about 70% HBV-positive HCC patients and contributes to HCC tumorigenesis. However, the role of Pin1 in the efficacy of sorafenib against HCC is unknown. Here we found that sorafenib down-regulated Pin1 mRNA and protein expression, likely through inhibition of Pin1 transcription by the Rb/E2F pathway. Importantly, Pin1 knockdown potently enhanced the ability of sorafenib to induce cell death in HCC, which was further supported by the findings that Pin1 knockdown led to stabilization of Fbxw7 and destabilization of Mcl-1. Furthermore, all-trans retinoic acid (ATRA), a known anticancer drug that inhibits and ultimately induces degradation of active Pin1 in cancer cells, also potently sensitized HCC cells to sorafenib-induced cell death at least in part through a caspase-dependent manner. Moreover, ATRA also synergistically enhanced the ability of sorafenib to reduce Pin1 and inhibit tumor growth of HCC in mouse xenograft models. Collectively, these results not only demonstrate that Pin1 down-regulation is a key event underlying the anti-tumor effects of sorafenib, but also uncover that Pin1 inhibitors offer a novel approach to enhance the therapeutic efficacy of sorafenib against HCC.
Collapse
Affiliation(s)
- Min Zheng
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Huijuan Xu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Xin-Hua Liao
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Champ Peng Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Arina Li Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Wenxian Lu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Dayun Yang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Jichuang Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Xiao Zhen Zhou
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China.,Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kun Ping Lu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China.,Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
42
|
Rodríguez-Hernández MA, González R, de la Rosa ÁJ, Gallego P, Ordóñez R, Navarro-Villarán E, Contreras L, Rodríguez-Arribas M, González-Gallego J, Álamo-Martínez JM, Marín-Gómez LM, Del Campo JA, Quiles JL, Fuentes JM, de la Cruz J, Mauriz JL, Padillo FJ, Muntané J. Molecular characterization of autophagic and apoptotic signaling induced by sorafenib in liver cancer cells. J Cell Physiol 2018; 234:692-708. [PMID: 30132846 DOI: 10.1002/jcp.26855] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Sorafenib is the unique accepted molecular targeted drug for the treatment of patients in advanced stage of hepatocellular carcinoma. The current study evaluated cell signaling regulation of endoplasmic reticulum (ER) stress, c-Jun-N-terminal kinase (JNK), Akt, and 5'AMP-activated protein kinase (AMPK) leading to autophagy and apoptosis induced by sorafenib. Sorafenib induced early (3-12 hr) ER stress characterized by an increase of Ser51 P-eIF2α/eIF2α, C/EBP homologous protein (CHOP), IRE1α, and sXBP1, but a decrease of activating transcription factor 6 expression, overall temporally associated with the increase of Thr183,Tyr185 P-JNK1/2/JNK1/2, Thr172 P-AMPKα, Ser413 P-Foxo3a, Thr308 P-AKt/AKt and Thr32 P-Foxo3a/Foxo3a ratios, and reduction of Ser2481 P-mammalian target of rapamycin (mTOR)/mTOR and protein translation. This pattern was related to a transient increase of tBid, Bim EL , Beclin-1, Bcl-xL, Bcl-2, autophagy markers, and reduction of myeloid cell leukemia-1 (Mcl-1) expression. The progressive increase of CHOP expression, and reduction of Thr308 P-AKt/AKt and Ser473 P-AKt/AKt ratios were associated with the reduction of autophagic flux and an additional upregulation of Bim EL expression and caspase-3 activity (24 hr). Small interfering-RNA (si-RNA) assays showed that Bim, but not Bak and Bax, was involved in the induction of caspase-3 in sorafenib-treated HepG2 cells. Sorafenib increased autophagic and apoptotic markers in tumor-derived xenograft model. In conclusion, the early sorafenib-induced ER stress and regulation of JNK and AMPK-dependent signaling were related to the induction of survival autophagic process. The sustained drug treatment induced a progressive increase of ER stress and PERK-CHOP-dependent rise of Bim EL , which was associated with the shift from autophagy to apoptosis. The kinetic of Bim EL expression profile might also be related to the tight balance between AKt- and AMPK-related signaling leading to Foxo3a-dependent BIM EL upregulation.
Collapse
Affiliation(s)
- María A Rodríguez-Hernández
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Raúl González
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Ángel J de la Rosa
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Paloma Gallego
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Seville, Spain
| | - Raquel Ordóñez
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Elena Navarro-Villarán
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
| | - Laura Contreras
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of Genetics, University of Seville, Seville, Spain
| | - Mario Rodríguez-Arribas
- Department of Biochemistry, Molecular Biology and Genetics, Faculty of Nursery and Occupational Therapy, University of Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - José M Álamo-Martínez
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Luís M Marín-Gómez
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - José A Del Campo
- Unit for the Clinical Management of Digestive Diseases, Hospital University "Nuestra Señora de Valme", Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - José L Quiles
- Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, Department of Physiology, University of Granada, Granada, Spain
| | - José M Fuentes
- Department of Biochemistry, Molecular Biology and Genetics, Faculty of Nursery and Occupational Therapy, University of Extremadura, Cáceres, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús de la Cruz
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Department of Genetics, University of Seville, Seville, Spain
| | - José L Mauriz
- Institute of Biomedicine (IBIOMED), Department of Biomedical Sciences, University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Francisco J Padillo
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| | - Jordi Muntané
- Institute of Biomedicine of Seville (IBiS), Hospital University "Virgen del Rocío"/CSIC/University of Seville, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of General Surgery, Hospital University "Virgen del Rocío"/CSIC/University of Seville/IBiS/CSIC/University of Seville, Spain
| |
Collapse
|
43
|
Allison SE, Chen Y, Petrovic N, Zhang J, Bourget K, Mackenzie PI, Murray M. Activation of ALDH1A1 in MDA-MB-468 breast cancer cells that over-express CYP2J2 protects against paclitaxel-dependent cell death mediated by reactive oxygen species. Biochem Pharmacol 2017; 143:79-89. [DOI: 10.1016/j.bcp.2017.07.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/24/2017] [Indexed: 01/26/2023]
|
44
|
Abstract
Orthologues of AMP-activated protein kinase (AMPK) occur in essentially all eukaryotes as heterotrimeric complexes comprising catalytic α subunits and regulatory β and γ subunits. The canonical role of AMPK is as an energy sensor, monitoring levels of the nucleotides AMP, ADP, and ATP that bind competitively to the γ subunit. Once activated, AMPK acts to restore energy homeostasis by switching on alternate ATP-generating catabolic pathways while switching off ATP-consuming anabolic pathways. However, its ancestral role in unicellular eukaryotes may have been in sensing of glucose rather than energy. In this article, we discuss a few interesting recent developments in the AMPK field. Firstly, we review recent findings on the canonical pathway by which AMPK is regulated by adenine nucleotides. Secondly, AMPK is now known to be activated in mammalian cells by glucose starvation by a mechanism that occurs in the absence of changes in adenine nucleotides, involving the formation of complexes with Axin and LKB1 on the surface of the lysosome. Thirdly, in addition to containing the nucleotide-binding sites on the γ subunits, AMPK heterotrimers contain a site for binding of allosteric activators termed the allosteric drug and metabolite (ADaM) site. A large number of synthetic activators, some of which show promise as hypoglycaemic agents in pre-clinical studies, have now been shown to bind there. Fourthly, some kinase inhibitors paradoxically activate AMPK, including one (SU6656) that binds in the catalytic site. Finally, although downstream targets originally identified for AMPK were mainly concerned with metabolism, recently identified targets have roles in such diverse areas as mitochondrial fission, integrity of epithelial cell layers, and angiogenesis.
Collapse
Affiliation(s)
- David Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Sheng-Cai Lin
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiang’an Campus, Xiamen, China
| |
Collapse
|
45
|
Ross FA, Hawley SA, Auciello FR, Gowans GJ, Atrih A, Lamont DJ, Hardie DG. Mechanisms of Paradoxical Activation of AMPK by the Kinase Inhibitors SU6656 and Sorafenib. Cell Chem Biol 2017; 24:813-824.e4. [PMID: 28625738 PMCID: PMC5522529 DOI: 10.1016/j.chembiol.2017.05.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/28/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023]
Abstract
SU6656, a Src kinase inhibitor, was reported to increase fat oxidation and reduce body weight in mice, with proposed mechanisms involving AMP-activated protein kinase (AMPK) activation via inhibition of phosphorylation of either LKB1 or AMPK by the Src kinase, Fyn. However, we report that AMPK activation by SU6656 is independent of Src kinases or tyrosine phosphorylation of LKB1 or AMPK and is not due to decreased cellular energy status or binding at the ADaM site on AMPK. SU6656 is a potent AMPK inhibitor, yet binding at the catalytic site paradoxically promotes phosphorylation of Thr172 by LKB1. This would enhance phosphorylation of downstream targets provided the lifetime of Thr172 phosphorylation was sufficient to allow dissociation of the inhibitor and subsequent catalysis prior to its dephosphorylation. By contrast, sorafenib, a kinase inhibitor in clinical use, activates AMPK indirectly by inhibiting mitochondrial metabolism and increasing cellular AMP:ADP and/or ADP:ATP ratios.
Collapse
Affiliation(s)
- Fiona A Ross
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Simon A Hawley
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - F Romana Auciello
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Graeme J Gowans
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Abdelmadjid Atrih
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Douglas J Lamont
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK.
| |
Collapse
|
46
|
Fumarola C, Cretella D, La Monica S, Bonelli MA, Alfieri R, Caffarra C, Quaini F, Madeddu D, Falco A, Cavazzoni A, Digiacomo G, Mazzaschi G, Vivo V, Barocelli E, Tiseo M, Petronini PG, Ardizzoni A. Enhancement of the anti-tumor activity of FGFR1 inhibition in squamous cell lung cancer by targeting downstream signaling involved in glucose metabolism. Oncotarget 2017; 8:91841-91859. [PMID: 29190880 PMCID: PMC5696146 DOI: 10.18632/oncotarget.19279] [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: 03/03/2017] [Accepted: 06/18/2017] [Indexed: 12/26/2022] Open
Abstract
Fibroblast Growth Factor Receptor (FGFR) signaling is a complex pathway which controls several processes, including cell proliferation, survival, migration, and metabolism. FGFR1 signaling is frequently deregulated via amplification/over-expression in NSCLC of squamous histotype (SQCLC), however its inhibition has not been successfully translated in clinical setting. We determined whether targeting downstream signaling implicated in FGFR1 effects on glucose metabolism potentiates the anti-tumor activity of FGFR1 inhibition in SQCLC. In FGFR1 amplified/over-expressing SQCLC cell lines, FGF2-mediated stimulation of FGFR1 under serum-deprivation activated both MAPK and AKT/mTOR pathways and increased glucose uptake, glycolysis, and lactate production, through AKT/mTOR-dependent HIF-1α accumulation and up-regulation of GLUT-1 glucose transporter. These effects were hindered by PD173074 and NVP-BGJ398, selective FGFR inhibitors, as well as by dovitinib, a multi-kinase inhibitor. Glucose metabolism was hampered by the FGFR inhibitors also under hypoxic conditions, with consequent inhibition of cell proliferation and viability. In presence of serum, glucose metabolism was impaired only in cell models in which FGFR1 inhibition was associated with AKT/mTOR down-regulation. When the activation of the AKT/mTOR pathway persisted despite FGFR1 down-regulation, the efficacy of NVP-BGJ398 could be significantly improved by the combination with NVP-BEZ235 or other inhibitors of this signaling cascade, both in vitro and in xenotransplanted nude mice. Collectively our results indicate that inhibition of FGFR1 signaling impacts on cancer cell growth also by affecting glucose energy metabolism. In addition, this study strongly suggests that the therapeutic efficacy of FGFR1 targeting molecules in SQCLC may be implemented by combined treatments tackling on glucose metabolism.
Collapse
Affiliation(s)
- Claudia Fumarola
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Daniele Cretella
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Silvia La Monica
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mara A Bonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Cristina Caffarra
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Valentina Vivo
- Food and Drug Department, University of Parma, Parma, Italy
| | | | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Andrea Ardizzoni
- Division of Medical Oncology, Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
47
|
Bronte G, Andreis D, Bravaccini S, Maltoni R, Cecconetto L, Schirone A, Farolfi A, Fedeli A, Serra P, Donati C, Amadori D, Rocca A. Sorafenib for the treatment of breast cancer. Expert Opin Pharmacother 2017; 18:621-630. [PMID: 28335647 DOI: 10.1080/14656566.2017.1309024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Breast cancer treatment includes many options depending on the tumor clinicopathological profile, which groups breast cancer into various subtypes. Bevacizumab is currently the only drug capable of targeting angiogenesis in breast cancer. Sorafenib has also been studied in combination with other agents. Areas covered: Pharmacological aspects of sorafenib, including results from preclinical studies on breast cancer cells; findings about clinical efficacy and safety in both single-arm and randomized clinical trials; ongoing trials. Expert opinion: Since sorafenib as a single agent has shown limited efficacy in breast cancer, its combination with other drugs is under investigation. Dose reduction is the main challenge when sorafenib is combined with chemotherapy or endocrine therapy. Although randomized phase-II trials on sorafenib plus chemotherapy versus chemotherapy alone have shown potential benefits in progression-free survival, preliminary results from a phase-III study in combination with capecitabine are negative. The definitive results of this trial and results from other ongoing phase-II trials will determine further developments of sorafenib in breast cancer. Although these additional data could help determine the most appropriate dose, drug combination and patient settings, a confirmation of the preliminary negative results reported in the phase-III trial are likely to discourage further use of sorafenib in breast cancer, given its non-negligible toxicity, lack of predicting markers, and the number of more promising drugs for breast cancer.
Collapse
Affiliation(s)
- Giuseppe Bronte
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Daniele Andreis
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Sara Bravaccini
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Roberta Maltoni
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Lorenzo Cecconetto
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alessio Schirone
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alberto Farolfi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Anna Fedeli
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Patrizia Serra
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Caterina Donati
- d Oncology Pharmacy , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Dino Amadori
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Andrea Rocca
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| |
Collapse
|
48
|
Zatula A, Dikic A, Mulder C, Sharma A, Vågbø CB, Sousa MML, Waage A, Slupphaug G. Proteome alterations associated with transformation of multiple myeloma to secondary plasma cell leukemia. Oncotarget 2017; 8:19427-19442. [PMID: 28038447 PMCID: PMC5386695 DOI: 10.18632/oncotarget.14294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Plasma cell leukemia is a rare and aggressive plasma cell neoplasm that may either originate de novo (primary PCL) or by leukemic transformation of multiple myeloma (MM) to secondary PCL (sPCL). The prognosis of sPCL is very poor, and currently no standard treatment is available due to lack of prospective clinical studies. In an attempt to elucidate factors contributing to transformation, we have performed super-SILAC quantitative proteome profiling of malignant plasma cells collected from the same patient at both the MM and sPCL stages of the disease. 795 proteins were found to be differentially expressed in the MM and sPCL samples. Gene ontology analysis indicated a metabolic shift towards aerobic glycolysis in sPCL as well as marked down-regulation of enzymes involved in glycan synthesis, potentially mediating altered glycosylation of surface receptors. There was no significant change in overall genomic 5-methylcytosine or 5-hydroxymethylcytosine at the two stages, indicating that epigenetic dysregulation was not a major driver of transformation to sPCL. The present study constitutes the first attempt to provide a comprehensive map of the altered protein expression profile accompanying transformation of MM to sPCL in a single patient, identifying several candidate proteins that can be targeted by currently available small molecule drugs. Our dataset furthermore constitutes a reference dataset for further proteomic analysis of sPCL transformation.
Collapse
Affiliation(s)
- Alexey Zatula
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Aida Dikic
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Celine Mulder
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Present address: University of Utrecht, Utrecht, Holland
| | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Cathrine B Vågbø
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Mirta M L Sousa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Department of Hematology, Department of Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| |
Collapse
|
49
|
Rogolino D, Cavazzoni A, Gatti A, Tegoni M, Pelosi G, Verdolino V, Fumarola C, Cretella D, Petronini PG, Carcelli M. Anti-proliferative effects of copper(II) complexes with hydroxyquinoline-thiosemicarbazone ligands. Eur J Med Chem 2017; 128:140-153. [DOI: 10.1016/j.ejmech.2017.01.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/31/2016] [Accepted: 01/21/2017] [Indexed: 01/21/2023]
|
50
|
ROS homeostasis and metabolism: a critical liaison for cancer therapy. Exp Mol Med 2016; 48:e269. [PMID: 27811934 PMCID: PMC5133371 DOI: 10.1038/emm.2016.119] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/17/2022] Open
Abstract
Evidence indicates that hypoxia and oxidative stress can control metabolic reprogramming of cancer cells and other cells in tumor microenvironments and that the reprogrammed metabolic pathways in cancer tissue can also alter the redox balance. Thus, important steps toward developing novel cancer therapy approaches would be to identify and modulate critical biochemical nodes that are deregulated in cancer metabolism and determine if the therapeutic efficiency can be influenced by changes in redox homeostasis in cancer tissues. In this review, we will explore the molecular mechanisms responsible for the metabolic reprogramming of tumor microenvironments, the functional modulation of which may disrupt the effects of or may be disrupted by redox homeostasis modulating cancer therapy.
Collapse
|