1
|
Liu Y, Ge H, Fan ZM, Lu T, He L, Li M, Zhao HR, Leng Q. Simvastatin inhibits proliferation and migration, promotes oxidative stress and ferroptosis in colon cancer. World J Gastrointest Oncol 2025; 17:104522. [DOI: 10.4251/wjgo.v17.i5.104522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/27/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a major cause of cancer-related mortality, with limited therapeutic options for advanced stages. Simvastatin, primarily used to lower cholesterol, has shown potential as an anticancer agent. It may exert its effects by inhibiting SERPINE1, a protein implicated in CRC progression, and activating the cyclic guanosine monophosphate-protein kinase G (cGMP/PKG) signaling pathway. Given these findings, this study hypothesizes that simvastatin inhibits CRC cell proliferation and migration by downregulating SERPINE1 and activating the cGMP/PKG pathway, offering a novel therapeutic strategy for CRC management.
AIM To study the effects of simvastatin on the function of colon cancer cells and to uncover the underlying mechanisms.
METHODS NCM460, HCT-116, and SW620 cell lines were used for in vitro experiments with simvastatin at doses of 20 μM, 40 μM, and 80 μM. The Stitch database was used to analyze the target genes of simvastatin, whereas STRING was used to investigate SERPINE1 and its related pathways. HCT-116 and SW620 cells were transfected with single-cell RNA sequencing reveals SERPINE1 with or without Rp-8-Br-cGMP (a PKG inhibitor). Cell toxicity, proliferation, and migration were evaluated using the cell counting kit-8, colony formation, and Transwell assays, respectively. Apoptosis was analyzed via flow cytometry, and levels of reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and ferrous ion (Fe2+) were detected using commercial kits. Real-time polymerase chain reaction and western blotting were used to analyze gene expression.
RESULTS Simvastatin dose-dependently inhibited the proliferation and migration of HCT-116 and SW620 cells while promoting apoptosis. It downregulated Ki-67, proliferating cell nuclear antigen, MMP2, and MMP9, and upregulated Bax, particularly at higher doses. Simvastatin increased the ROS, MDA, and Fe2+ levels while decreasing the GSH levels. It downregulated SLC7A11 and ferroportin and upregulated TRF1. SERPINE1 was identified as a core target, with related genes enriched in the cGMP/PKG pathway. SERPINE1 knockdown increased GUCY1B1 and PRKG1 levels, decreased cell viability, and altered oxidative stress markers, with the effects being reversed by Rp-8-Br-cGMP.
CONCLUSION Simvastatin effectively inhibited the proliferation and migration of colon cancer cells and promoted apoptosis through the modulation of key targets, such as SERPINE1 and the cGMP/PKG signaling pathway.
Collapse
Affiliation(s)
- Ying Liu
- Department of Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Hao Ge
- Department of Traditional Chinese Medicine Surgery, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210038, Jiangsu Province, China
| | - Zhi-Min Fan
- Department of Anorectal Medicine, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Ting Lu
- Department of Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Lei He
- Department of Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Meng Li
- Department of Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| | - Hao-Ran Zhao
- Department of Traditional Chinese Medicine Surgery, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210038, Jiangsu Province, China
| | - Qiang Leng
- Department of Colon and Rectal Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu Province, China
| |
Collapse
|
2
|
Skriver C, Cronin-Fenton D, Borgquist S, Hansen Viuff J, Alkner S, Rydén L, Lænkholm AV, Manjer J, Bengtsson Y, Frederiksen K, Friis S, Mellemkjær L. Statin use and risk of breast cancer among women with benign breast disease: a Danish nationwide cohort study. Br J Cancer 2025; 132:828-836. [PMID: 40057666 PMCID: PMC12041342 DOI: 10.1038/s41416-025-02974-x] [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: 05/29/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Statins have been suggested to protect against breast cancer risk, but the observational evidence is inconclusive. We examined the association between statin use and breast cancer incidence among women at higher risk of breast cancer due to a history of benign breast disease (BBD). METHODS Using Danish registries, we identified cancer-free women aged ≥50 years during 1996-2018 with a history of BBD and no prior statin prescriptions. Using Cox regression, we estimated multivariable-adjusted hazard ratios (HRs) with 95% confidence intervals (CIs) for invasive breast cancer through 2020 with time-varying statin use defined according to continuity, duration, and intensity (estimated average daily dose), derived from prescription data. RESULTS Among 111,550 women, 7629 were diagnosed with breast cancer during median follow-up of 12.2 years. Overall statin use was not associated with breast cancer incidence (adjusted HR = 0.99; 95% CI, 0.93-1.06), with similar associations observed according to continuity and duration of use. However, long-term (≥10 years), high-intensity statin use was associated with a reduced HR of 0.75 (95% CI, 0.60-0.96). CONCLUSIONS Our findings did not indicate an association for overall statin use with breast cancer incidence among women with BBD. The inverse association with long-term, high-dose statin use requires further evaluation.
Collapse
Affiliation(s)
| | - Deirdre Cronin-Fenton
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Signe Borgquist
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jakob Hansen Viuff
- Danish Cancer Institute, Copenhagen, Denmark
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sara Alkner
- Department of Haematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Lisa Rydén
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Anne-Vibeke Lænkholm
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Jonas Manjer
- Department of Surgery, Skåne University Hospital, Malmö, Sweden
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - Ylva Bengtsson
- Department of Haematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | | | - Søren Friis
- Danish Cancer Institute, Copenhagen, Denmark
| | | |
Collapse
|
3
|
Poswal J, Mandal CC. Lipid metabolism dysregulation for bone metastasis and its prevention. Expert Rev Anticancer Ther 2025:1-17. [PMID: 40219980 DOI: 10.1080/14737140.2025.2492784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Bone metastasis often develops in advanced malignancies. Lipid metabolic dysregulation might play pivotal role in cancer progression and subsequent deterioration of bone health at metastatic condition. In-depth understanding of lipid reprogramming in metastasized cancer cells and other stromal cells including bone marrow adipocyte (BMA) is an urgent need to develop effective therapy. AREA COVERED This paper emphasizes providing an overview of multifaceted role of dysregulated lipids and BMA in cancer cells in association with bone metastasis by utilizing search terms lipid metabolism, lipid and metastasis in PubMed. This study extends to address mechanism linked with lipid metabolism and various crucial genes (e.g. CSF-1, RANKL, NFkB and NFATc1) involved in bone metastasis. This review examines therapeutic strategies targeting lipid metabolism to offer potential avenues to disrupt lipid-driven metastasis. EXPERT OPINION On metastatic condition, dysregulated lipid molecules especially in BMA and other stromal cells not only favors cancer progression but also potentiate lipid reprogramming within cancer cells. Distinct dysregulated lipid-metabolism associated genes may act as biomarker, and targeting these is challenging task for specific treatment. Curbing function of bone resorption associated genes by lipid controlling drugs (e.g. statins, omega-3 FA and metformin) may provide additional support to curtail lipid-associated bone metastasis.
Collapse
Affiliation(s)
- Jyoti Poswal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
4
|
Soni S, Makwana SH, Bansal S, Kumari M, Mandal CC. Lipid metabolism associated PLPP4 gene drives oncogenic and adipogenic potential in breast cancer cells. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159609. [PMID: 40187483 DOI: 10.1016/j.bbalip.2025.159609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/16/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Lipid metabolic reprogramming plays a pivotal role in cancer cell evolution and causing subsequent cancer growth, metastasis and therapy resistance. Cancer associated adipocyte and/or cancer derived adipocyte-like cells often supply fuels and various factors to fulfill the cells bioenergetics to enhance oncogenic potential. This study intends to find out a set of dysregulated genes involved in lipid metabolism in breast cancer studies and uncovers the role of unexplored dysregulated gene in cancer potential. Cancer database analysis determines seven seed signature genes (PLPP2, PLPP4, CDS1, ASAH2, LCLAT1, LPCAT1 and LASS6/CERS6) concluded from relative expression and survival analysis. Furthermore, experimental analysis unveils the gene PLPP4 (Phospholipid Phosphatase 4) as oncogene confirmed by knockdown and overexpression studies in MDA-MB 231 and MCF-7 breast cancer cells. PLPP4 enzyme is involved in regulation of triacyl glycerol metabolism. Lipid accumulation along with other studies documented enhanced lipid droplets, TAG formation and glycerol release with concomitant increased expressions of various adipogenic markers (e.g., PPARγ, perilipin 1 and leptin) in breast cancer cells transfected with PLPP4 gene expressing plasmid whereas downregulation of PLPP4 gene diminished lipid accumulation and adipocyte marker gene expressions. Our findings also revealed that BMP2 induced adipogenic potential in breast cancer cells was mitigated in response to downregulation of PLPP4 gene expression. All these findings together, for first time, demonstrated that BMP2 drives PLPP4 to enhance both oncogenic and adipogenic potential in breast cancer cells. This article uncovers the perturbed lipid metabolism associated PLPP4 acts as oncogene presumably by modulating adipogenic activity in cancer cells.
Collapse
Affiliation(s)
- Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | - Sweta H Makwana
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | - Shivani Bansal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | - Monika Kumari
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India.
| |
Collapse
|
5
|
Eom YS, Kim SJ. Regulatory dynamics of Nanog in chondrocyte dedifferentiation: role of KLF4/p53 and p38/AKT signaling. Funct Integr Genomics 2025; 25:58. [PMID: 40067510 DOI: 10.1007/s10142-025-01572-7] [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: 11/27/2024] [Revised: 02/10/2025] [Accepted: 03/01/2025] [Indexed: 05/13/2025]
Abstract
Homeobox protein Nanog, a member of the transcription factor family, plays a crucial role in maintaining the pluripotency and self-renewal of embryonic stem cells. Due to its diverse activities, Nanog has been identified in multiple cell types, including embryonic stem cells (ESCs) and cancer stem cells (CSCs). However, its molecular mechanism in chondrocytes remains unclear. In this study, we explored the effects of Nanog on chondrocytes and its interaction with chondrocyte-specific proteins. Chondrocytes were transfected with a Nanog cDNA vector, resulting in reduced expression of the chondrogenic markers Type II collagen and SOX9, as confirmed by western blot, RT-PCR, and immunofluorescence. Following siRNA transfection, the dedifferentiation effect of Nanog was reversed, restoring Type II collagen and SOX9 expression. We also discovered that the mechanism by which Nanog affects chondrocytes is closely linked to p53 and KLF4. Overexpression of KLF4 induced the phosphorylation of p53, and phospho-p53 directly inhibited Nanog expression. Moreover, the p53 activator Nutlin-3 A accelerated Nanog degradation, while the p53 inhibitor Pifithrin-α stabilized Nanog. Stabilized Nanog continued to promote chondrocyte dedifferentiation. Additional experiments were performed to identify the signaling pathways involved in Nanog-induced chondrocyte dedifferentiation. Our results showed that Nanog overexpression in chondrocytes significantly impacted the p38 kinase and AKT signaling pathways. Inhibition of p38 and AKT with SB203580 and LY294002 reduced Nanog expression and partially restored Type II collagen levels. Conversely, activation with anisomycin(ANS) and 740 Y-P enhanced Nanog expression, further reducing Type II collagen levels. To investigate Nanog's role in early development in vivo, we injected Nanog expression vectors into zebrafish embryos. The injected zebrafish exhibited structural defects in craniofacial cartilage, confirming Nanog's involvement in chondrocyte differentiation. These findings suggest that Nanog induces chondrocyte dedifferentiation, and this process can be modulated via the p53/KLF4 and p38/AKT pathways.
Collapse
Affiliation(s)
- Young Seok Eom
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, 32588, Republic of Korea.
| |
Collapse
|
6
|
Czechowicz P, Więch-Walów A, Sławski J, Collawn JF, Bartoszewski R. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 2025; 30:27. [PMID: 40038587 PMCID: PMC11881393 DOI: 10.1186/s11658-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Anna Więch-Walów
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland.
| |
Collapse
|
7
|
Atale N, Wells A. Statins as Secondary Preventive Agent to Limit Breast Cancer Metastatic Outgrowth. Int J Mol Sci 2025; 26:1300. [PMID: 39941069 PMCID: PMC11818786 DOI: 10.3390/ijms26031300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is a leading cause of mortality in breast cancer, as metastatic disease is often aggressive and resistant to conventional treatments. Cancer cells that spread to distant organs can enter a dormant phase for extended periods, sometimes years or decades. During this dormant phase, cancer cells avoid immune and pharmacological response. Thus, new approaches are needed to prevent these disseminated cells from becoming lethal cancers. Statins are known inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase that have been extensively used in patients with cardiovascular diseases to lower cholesterol. However, recent research has demonstrated their potential in anticancer therapies. Epidemiological evidence suggests that statins are associated with a reduction in breast cancer-specific mortality, although they do not appear to affect the incidence of primary tumors. In this review, we discuss the role of statins in metastasis and dormancy, their cytocidal and cytostatic effects and their interactions with different cell types in the tumor microenvironment. The exact mechanisms by which statins reduce mortality without influencing primary tumor growth remain unclear, also warranting further investigation into their potential role in metastasis and tumor dormancy, which could ultimately help patients to improve survival and quality of life.
Collapse
Affiliation(s)
- Neha Atale
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Alan Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Research and Development Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
- Cell Biology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
Biscetti F, Polito G, Rando MM, Nicolazzi MA, Eraso LH, DiMuzio PJ, Massetti M, Gasbarrini A, Flex A. Residual Traditional Risk in Non-Traditional Atherosclerotic Diseases. Int J Mol Sci 2025; 26:535. [PMID: 39859250 PMCID: PMC11765428 DOI: 10.3390/ijms26020535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Individuals with chronic inflammatory and immune disorders are at an increased risk of atherosclerotic events and premature cardiovascular (CV) disease. Despite extensive literature exploring the relationship between "non-traditional" atherosclerotic conditions and CV risk, many aspects remain unresolved, including the underlying mechanisms promoting the "non-traditional CV risk", the development of an innovative and comprehensive CV risk assessment tool, and recommendations for tailored interventions. This review aims to evaluate the available evidence on key "non-traditional" CV risk-enhancer conditions, with a focus on assessing and managing CV risk factors. We conducted a comprehensive review of 412 original articles, narrative and systematic reviews, and meta-analyses addressing the CV risk associated with "non-traditional" atherosclerotic conditions. The analysis examined the underlying mechanisms of these relationships and identified strategies for assessing and mitigating elevated risk. A major challenge highlighted is the difficulty in quantifying the contribution of individual risk factors and disease-specific elements to CV risk. While evidence supports the cardiovascular benefits of statins beyond lipid lowering, such as pleiotropic and endothelial effects, current guidelines lack specific recommendations for the use of statins or other therapies targeting non-traditional CV risk factors. Additionally, the absence of validated cardiovascular risk scores that incorporate non-traditional risk factors hinders accurate CV risk evaluation and management. The growing prevalence of "non-traditional CV risk-enhancer conditions" underscores the need for improved awareness of CV risk assessment and management. A thorough understanding of all contributing factors, including disease-specific elements, is crucial for accurate prediction of cardiovascular disease (CVD) risk. This represents an essential foundation for informed decision-making in primary and secondary prevention. We advocate for future research to focus on developing innovative, disease-specific CV risk assessment tools that incorporate non-traditional risk factors, recognizing this as a promising avenue for translational and clinical outcome research.
Collapse
Affiliation(s)
- Federico Biscetti
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Giorgia Polito
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Maria Margherita Rando
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Maria Anna Nicolazzi
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Luis H. Eraso
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paul J. DiMuzio
- Division of Vascular and Endovascular Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Massimo Massetti
- Dipartimento di Scienze Cardiovascolari e Pneumologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Department of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Andrea Flex
- Cardiovascular Internal Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
9
|
Alipour B, Veisi Malekshahi Z, Pourjafar F, Faridi-Majidi R, Negahdari B. Anticancer effects of simvastatin-loaded albumin nanoparticles on monolayer and spheroid models of breast cancer. Biochem Biophys Res Commun 2024; 734:150591. [PMID: 39255745 DOI: 10.1016/j.bbrc.2024.150591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/12/2024]
Abstract
Breast cancer is a prominent cause of death among women and is distinguished by a high occurrence of metastasis. From this perspective, apart from conventional therapies, several alternative approaches have been researched and explored in recent years, including the utilization of nano-albumin and statin medications like simvastatin. The objective of this study was to prepare albumin nanoparticles incorporating simvastatin by the self-assembly method and evaluate their impact on breast cancer metastasis and apoptosis. The data showed the prepared nanoparticles have a diameter of 185 ± 24nm and a drug loading capacity of 8.85 %. The findings exhibit improved release in a lysosomal-like environment and under acidic pH conditions. MTT data showed that nanoparticles do not exhibit a dose-dependent effect on cells. Additionally, the results from MTT, flow cytometry, and qPCR analyses demonstrated that nanoparticles have a greater inhibitory and lethal effect on MDA-MB-231 cells compared to normal simvastatin. And cause cells to accumulate in the G0/G1 phase, initiating apoptotic pathways by inhibiting cell cycle progression. Nanoparticles containing simvastatin can prevent cell invasion and migration in both monolayer and spheroid models, as compared to simvastatin alone, at microscopic levels and in gene expression. The obtained data clearly showed that, compared to simvastatin, nanoparticles containing simvastatin demonstrated significant efficacy in suppressing the growth, proliferation, invasion, and migration of cancer cells in monolayer (2D) and spheroid (3D) models.
Collapse
Affiliation(s)
- Behruz Alipour
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Pourjafar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Faridi-Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Makwana SH, Sharma T, Mahapatra MK, Kumari M, Jain A, Shrivastava SK, Mandal CC. Targeting TRIM26: Unveiling an Oncogene and Identification of Plant Metabolites as a Potential Therapeutics for Breast Cancer. J Cell Biochem 2024; 125:e30644. [PMID: 39286999 DOI: 10.1002/jcb.30644] [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: 04/15/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024]
Abstract
Breast cancer is the major cause of cancer-related mortality and frequent malignancies among women worldwide. The TRIM (Tripartite Motif) protein family is a broad and diverse set of proteins that contain a conserved structural motif known as the tripartite motif, which comprises of three different domains, B-box domain, Coiled-coil domain and RBR (Ring-finger, B-box, and coiled-coil) domain. TRIM proteins are involved in regulating cancer growth and metastasis. However, TRIM proteins are still unexplored in cancer cell regulation. In this study, by using a cancer database expression of all TRIM proteins was determined in breast cancer. Out of 77 TRIM genes, 16 genes were upregulated in breast cancer. Here, the upregulated TRIM26 gene's role is not yet explored in breast cancer. Indeed, TRIM26 is upregulated in 21 cancer types out of 33 cancer types. To investigate the role of TRIM26 in breast cancer, siRNA-mediated gene silencing was carried out in MCF-7 and MDA-MB 231 breast cancer cells. Reduced expression of TRIM 26 decreased cancer cell proliferation, migration and invasion with simultaneous reduction of various proliferative, cell cycle and mesenchymal markers and upregulation of epithelial markers. Further, docking studies found potential novel plant metabolites. Thus, targeting TRIM26 may provide a novel therapeutic approach for breast cancer treatment.
Collapse
Affiliation(s)
- Sweta H Makwana
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Tannavi Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Manas K Mahapatra
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Monika Kumari
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Akshat Jain
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sandeep K Shrivastava
- Centre for Innovation, Research & Development, Dr. B. Lal Clinical Laboratory Pvt Ltd, Jaipur, Rajasthan, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
11
|
Pang S, Geng C, Fan Z, Hou M, Mao H, Tao S, Wang J, Wu Y, Wei K, Li Y, Yan L, Yang Q, Chen C, Wang W. Synergistic Effect of Layered Double Hydroxides Nanodosage Form to Induce Apoptosis and Ferroptosis in Breast Cancer. Int J Nanomedicine 2024; 19:4199-4215. [PMID: 38766657 PMCID: PMC11102185 DOI: 10.2147/ijn.s455427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
Background Breast cancer is the most common cancer in women and one of the leading causes of cancer death worldwide. Ferroptosis, a promising mechanism of killing cancer cells, has become a research hotspot in cancer therapy. Simvastatin (SIM), as a potential new anti-breast cancer drug, has been shown to cause ferroptosis of cancer cells and inhibit breast cancer metastasis and recurrence. The purpose of this study is to develop a novel strategy boosting ferroptotic cascade for synergistic cancer therapy. Methods In this paper, iron base form of layered double hydroxide supported simvastatin (LDHs-SIM) was synthesized by hydrothermal co-precipitation method. The characterization of LDHs-SIM were assessed by various analytical techniques, including ultraviolet-visible (UV-vis) spectroscopy, X-ray diffraction (XRD), Fourier transform infrared (FTIR) spectroscopy, and transmission electron microscopy (TEM). Biological activity, ferroptosis mechanism and biocompatibility were analyzed through in vivo and in vitro analysis, so as to evaluate its therapeutic effect on breast cancer. Results The constructed LDHs-SIM nanosystem can not only release SIM through mevalonate (MVA) pathway, inhibit the expression of glutathione peroxidase 4 (GPX4), inhibit the expression of SLC7A11 and reduce the synthesis efficiency of GSH, but also promote the accumulation of Fe2+ in cells through the release of Fe3+, and increase the intracellular ROS content. In addition, LDHs-SIM nanosystem can induce apoptosis of breast cancer cells to a certain extent, and achieve the synergistic effect of apoptosis and ferroptosis. Conclusion In the present study, we demonstrated that nanoparticles of layered double hydroxides (LDHs) loaded with simvastatin were more effective than a free drug at inhibiting breast cancer cell growth, In addition, superior anticancer therapeutic effects were achieved with little systemic toxicity, indicating that LDHs-SIM could serve as a safe and high-performance platform for ferroptosis-apoptosis combined anticancer therapy.
Collapse
Affiliation(s)
- Siyan Pang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Chenchen Geng
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Zihan Fan
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Min Hou
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- School of Basic Courses, Bengbu Medical University, Anhui, People’s Republic of China
| | - Huilan Mao
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Shuang Tao
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Anhui, People’s Republic of China
| | - Jing Wang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Anhui, People’s Republic of China
| | - Yulun Wu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Ke Wei
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Anhui, People’s Republic of China
| | - Yunhao Li
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Liuyang Yan
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
| | - Qingling Yang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Anhui, People’s Republic of China
| | - Changjie Chen
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Anhui, People’s Republic of China
| | - Wenrui Wang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Life Sciences, Bengbu Medical University, Anhui, People’s Republic of China
- Department of Biotechnology, Bengbu Medical University, Anhui, People’s Republic of China
| |
Collapse
|
12
|
Xia Y, Sun M, Huang H, Jin WL. Drug repurposing for cancer therapy. Signal Transduct Target Ther 2024; 9:92. [PMID: 38637540 PMCID: PMC11026526 DOI: 10.1038/s41392-024-01808-1] [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: 02/06/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer, a complex and multifactorial disease, presents a significant challenge to global health. Despite significant advances in surgical, radiotherapeutic and immunological approaches, which have improved cancer treatment outcomes, drug therapy continues to serve as a key therapeutic strategy. However, the clinical efficacy of drug therapy is often constrained by drug resistance and severe toxic side effects, and thus there remains a critical need to develop novel cancer therapeutics. One promising strategy that has received widespread attention in recent years is drug repurposing: the identification of new applications for existing, clinically approved drugs. Drug repurposing possesses several inherent advantages in the context of cancer treatment since repurposed drugs are typically cost-effective, proven to be safe, and can significantly expedite the drug development process due to their already established safety profiles. In light of this, the present review offers a comprehensive overview of the various methods employed in drug repurposing, specifically focusing on the repurposing of drugs to treat cancer. We describe the antitumor properties of candidate drugs, and discuss in detail how they target both the hallmarks of cancer in tumor cells and the surrounding tumor microenvironment. In addition, we examine the innovative strategy of integrating drug repurposing with nanotechnology to enhance topical drug delivery. We also emphasize the critical role that repurposed drugs can play when used as part of a combination therapy regimen. To conclude, we outline the challenges associated with repurposing drugs and consider the future prospects of these repurposed drugs transitioning into clinical application.
Collapse
Affiliation(s)
- Ying Xia
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
- Division of Gastroenterology and Hepatology, Department of Medicine and, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ming Sun
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
| | - Hai Huang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China.
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
13
|
McKechnie T, Brown Z, Lovrics O, Yang S, Kazi T, Eskicioglu C, Parvez E. Concurrent Use of Statins in Patients Undergoing Curative Intent Treatment for Triple Negative Breast Cancer: A Systematic Review and Meta-Analysis. Clin Breast Cancer 2024; 24:e103-e115. [PMID: 38296737 DOI: 10.1016/j.clbc.2023.12.001] [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: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 02/02/2024]
Abstract
Pre-clinical studies in triple negative breast cancer (TNBC) suggest that statins may inhibit cell proliferation, promote cell-cycle arrest, induce apoptosis, change the tumor microenvironment, and improve effectiveness of other therapies. Observational studies have demonstrated variable effects from statin therapy on oncologic outcomes in these patients. As such, we aimed to pool previous data via a systematic review and meta-analysis to elucidate the impact of concurrent statin use on oncologic outcomes for patients with TNBC. Medline, EMBASE, CENTRAL, and PubMed were systematically searched from inception through to June 2022. Studies were included if they compared patients with TNBC receiving and not receiving statin therapy concurrently with oncologic treatment for curative intent in terms of recurrence and survival in a non-metastatic setting. The primary outcomes were 5-year disease-free survival (DFS) and 5-year overall survival (OS). A pairwise meta-analyses was performed using inverse variance random effects. Risk of bias was assessed with the ROBINS-I and the GRADE approach was conducted to assess quality of evidence. From 4014 citations, 5 studies with 625 patients on statin therapy and 2707 patients not on statin therapy were included. There was a significant increase in 5-year DFS for patients on statin therapy compared to patients not on statin therapy (OR 1.44, 95% CI 1.04-1.98, P = .03). No significant difference was noted in 5-year OS between the 2 groups (OR 1.12, 95% CI 0.86-1.47, P = .40). Included studies were at moderate-to-high risk of bias. The GRADE quality of evidence was very low. This review presents very low-quality evidence that concurrent use of statins with oncologic treatment may potentially improve long-term DFS for patients with TNBC undergoing curative intent therapy. Future research by way of large, prospective study is required to further clarify the clinical utility of statins on patients undergoing treatment for TNBC.
Collapse
Affiliation(s)
- Tyler McKechnie
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Zachary Brown
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Olivia Lovrics
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Shuling Yang
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Tania Kazi
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Cagla Eskicioglu
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Elena Parvez
- Division of General Surgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada; Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
14
|
Ou Y, Chu GCY, Lyu J, Yin L, Lim A, Zhai N, Cui X, Lewis MS, Edderkaoui M, Pandol SJ, Wang R, Zhang Y. Overcoming Resistance in Prostate Cancer Therapy Using a DZ-Simvastatin Conjugate. Mol Pharm 2024; 21:873-882. [PMID: 38229228 PMCID: PMC11025579 DOI: 10.1021/acs.molpharmaceut.3c00993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Prostate cancer (PC), particularly its metastatic castration-resistant form (mCRPC), is a leading cause of cancer-related deaths among men in the Western world. Traditional systemic treatments, including hormonal therapy and chemotherapy, offer limited effectiveness due to tumors' inherent resistance to these therapies. Moreover, they often come with significant side effects. We have developed a delivery method using a tumor-cell-specific heptamethine carbocyanine dye (DZ) designed to transport therapeutic agents directly to tumor cells. This research evaluated simvastatin (SIM) as the antitumor payload because of its demonstrated chemopreventive effects on human cancers and its well-documented safety profile. We designed and synthesized a DZ-SIM conjugate for tumor cell targeting. PC cell lines and xenograft tumor models were used to assess tumor-cell targeting specificity and killing activity and to investigate the corresponding mechanisms. DZ-SIM treatment effectively killed PC cells regardless of their androgen receptor status or inherent therapeutic resistance. The conjugate targeted and suppressed xenograft tumor formation without harming normal cells of the host. In cancer cells, DZ-SIM was enriched in subcellular organelles, including mitochondria, where the conjugate formed adducts with multiple proteins and caused the loss of transmembrane potential, promoting cell death. The DZ-SIM specifically targets PC cells and their mitochondria, resulting in a loss of mitochondrial function and cell death. With a unique subcellular targeting strategy, the conjugate holds the potential to outperform existing chemotherapeutic drugs. It presents a novel strategy to circumvent therapeutic resistance, offering a more potent treatment for mCRPC.
Collapse
Affiliation(s)
- Yan Ou
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Gina Chia-Yi Chu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ji Lyu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Liyuan Yin
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ning Zhai
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Xiaojiang Cui
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Michael S. Lewis
- Department of Pathology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Los Angeles, CA 90073, United States
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ruoxiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Yi Zhang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| |
Collapse
|
15
|
Krishnan J, Symington A, Kernohan N, Bray S, Robertson A, Nabi G. HMG co-reductase expression and response to intravesical Bacillus Calmette-Guérin in patients with high grade non-muscle invasive urinary bladder cancer receiving statins. Scott Med J 2024; 69:3-9. [PMID: 37960856 DOI: 10.1177/00369330231213935] [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] [Indexed: 11/15/2023]
Abstract
BACKGROUND Cardiovascular disease affects over 7 million people in the UK and statins are often prescribed to mitigate cardiovascular risks. The effect of statins on a number of cancers is debated and their effect on Bacillus Calmette-Guérin (BCG) responsiveness in non-muscle invasive urinary bladder cancer (NMIBC) is not fully understood. AIMS This study aims to explore the difference in HMG Co-A reductase (HMGCR) expression in NMIBC on immunochemistry in BCG responders and non-responders while on statins. METHOD Three hundred and thirty-two cases of intravesical BCG treatment for high-risk NMIBC between November 2003 and December 2017 were identified. Patients taking statins for at least 12 months before the diagnosis of NIMBC and with a follow-up of at least 5 years were included. They were divided into BCG responders and non-responders. Tumour tissue from these patients was immunohistochemically stained and quantitative image analysis carried out to assess and compare HMGCR expression in the groups. RESULTS & CONCLUSION This study showed a differential expression of HMGCR in responders vs. non-responders to BCG for high-risk NMIBC on statins. This data should form the basis of a further research and multi-centre study in a larger cohort, using HMGCR as a biomarker of response in patients on statins.
Collapse
Affiliation(s)
- Jamie Krishnan
- Urology, Ninewells Hospital and Medical School, Dundee, UK
| | | | - Neil Kernohan
- Urology, Ninewells Hospital and Medical School, Dundee, UK
| | - Suan Bray
- Urology, Ninewells Hospital and Medical School, Dundee, UK
| | | | - Ghulam Nabi
- Urology, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
16
|
Xiao Q, Xia M, Tang W, Zhao H, Chen Y, Zhong J. The lipid metabolism remodeling: A hurdle in breast cancer therapy. Cancer Lett 2024; 582:216512. [PMID: 38036043 DOI: 10.1016/j.canlet.2023.216512] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Abstract
Lipids, as one of the three primary energy sources, provide energy for all cellular life activities. Lipids are also known to be involved in the formation of cell membranes and play an important role as signaling molecules in the intracellular and microenvironment. Tumor cells actively or passively remodel lipid metabolism, using the function of lipids in various important cellular life activities to evade therapeutic attack. Breast cancer has become the leading cause of cancer-related deaths in women, which is partly due to therapeutic resistance. It is necessary to fully elucidate the formation and mechanisms of chemoresistance to improve breast cancer patient survival rates. Altered lipid metabolism has been observed in breast cancer with therapeutic resistance, indicating that targeting lipid reprogramming is a promising anticancer strategy.
Collapse
Affiliation(s)
- Qian Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Min Xia
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Weijian Tang
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, 330031, PR China
| | - Hu Zhao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Yajun Chen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Jing Zhong
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
17
|
Jaiswal V, Agrawal V, Ang SP, Saleeb M, Ishak A, Hameed M, Rajak K, Kalra K, Jaiswal A. Post-diagnostic statin use and its association with cancer recurrence and mortality in breast cancer patients: a systematic review and meta-analysis. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2023; 9:731-740. [PMID: 37562940 DOI: 10.1093/ehjcvp/pvad057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Statins are widely acknowledged for their application in patients with hypercholesterolemia to reduce cardiovascular morbidity and mortality. More recently, their potential to exert pleiotropic effects, particularly in impeding the proliferation of neoplastic cells, has attracted considerable attention. Prior studies have demonstrated that statins may mitigate cancer progression and micrometastasis. However, the benefits of statins in breast cancer have been inconclusive. OBJECTIVE The aim of this meta-analysis was to evaluate the impact of statin use following a breast cancer diagnosis on breast cancer recurrence and mortality. METHODS We performed a systematic literature search using PubMed, Embase, and Scopus for relevant articles from inception until 30th May 2023. Hazard ratios (HR) were pooled using a random-effect model. The primary outcome of interest was the risk of breast cancer recurrence. The secondary outcomes included breast cancer-specific mortality and all-cause mortality. RESULTS A total of 15 studies with 156 448 patients were included in the final analysis. The mean age of patients between statin users and non-users was 64.59 and 59.15 years, respectively. Statin use was associated with a reduction in the recurrence of breast cancer [HR 0.76, 95% confidence interval (CI): 0.67-0.87] compared with non-statin users. This trend was similar among lipophilic statin users (HR 0.73, 95% CI: 0.63-0.85) but not for hydrophilic statin users (HR 1.17, 95% CI: 0.82-1.68). Furthermore, statin users exhibited a lower risk of breast cancer mortality (HR 0.80, 95% CI: 0.66-0.96) but all-cause mortality (HR 0.82, 95% CI: 0.66-1.02) was comparable among both groups of patients. Conversely, lipophilic statins demonstrated a reduction in both all-cause mortality (HR 0.84, 95% CI: 0.75-0.93) and breast cancer mortality (HR 0.85, 95% CI: 0.74-0.99) compared to non-statin users. CONCLUSION Among patients with breast cancer, statin use post-diagnosis decreases the risk of breast cancer recurrence and breast cancer mortality. Furthermore, lipophilic statins exhibit an additional advantage of reduction in all-cause mortality.PROSPERO registration: CRD42022362011.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Cardiovascular Research, Larkin Community Hospital, South Miami, FL, 33143, USA
- JCCR Cardiology Research, Varanasi, 221005, India
| | - Vibhor Agrawal
- Department of Medicine, King George's Medical University, Lucknow, 226003, India
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, NJ 08755, USA
| | - Marina Saleeb
- Public Health Institute, Faculty of Health, Liverpool John Moores University, Liverpool L2 2QP, UK
| | - Angela Ishak
- Department of Internal Medicine, Henry Ford Hospital, Detroit, 48202, USA
| | - Maha Hameed
- Department of Internal Medicine, Florida State University/Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Kripa Rajak
- Department of Internal Medicine, UPMC, Harrisburg, PA 17101, USA
| | - Kriti Kalra
- Department of Cardiology, MedStar Washington Hospital Center, WD 20010, USA
| | - Akash Jaiswal
- Department of Geriatric Medicine, All India Institute of Medical Science, New Delhi, 110608, India
| |
Collapse
|
18
|
Yadav P, Makwana S, Bansal S, Soni S, Mahapatra MK, Bandyopadhayaya S, Tailor R, Shrivastava SK, Sharma LK, Mandal CC. Metformin prevents osteoblast-like potential and calcification in lung cancer A549 cells. J Biochem Mol Toxicol 2023; 37:e23454. [PMID: 37409753 DOI: 10.1002/jbt.23454] [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: 01/14/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
In spite of recent advances made in understanding its progression, cancer is still a leading cause of death across the nations. Molecular pathophysiology of these cancer cells largely differs depending on cancer types and even within the same tumor. Pathological mineralization/calcification is seen in various tissues including breast, prostate, and lung cancer. Osteoblast-like cells derived after trans-differentiation of mesenchymal cells usually drive calcium deposition in various tissues. This study aims to explore the presence of osteoblast-like potential in lung cancer cells and its prevention. ALP assay, ALP staining, nodule formation, RT-PCR, RT-qPCR, and western blot analysis experiments were carried out in lung cancer A549 cells to achieve said objective. Expressions of various osteoblast markers (e.g., ALP, OPN, RUNX2, and Osterix) along with osteoinducer genes (BMP-2 and BMP-4) were observed in A549 cells. Moreover, ALP activity and ability leading to nodule formation revealed the presence of osteoblast-like potential in lung cancer cells. Here, BMP-2 treatment increased expressions of osteoblast transcription factors such as RUNX2 and Osterix, enhanced ALP activity, and augmented calcification in this cell line. It was also observed that antidiabetic metformin inhibited BMP-2 mediated increase in osteoblast-like potential and calcification in these cancer cells. The current study noted that metformin blocked BMP-2 mediated increase in epithelial to mesenchymal transition (EMT) in A549 cells. The above findings for the first time unravel that A549 cells possess osteoblast-like potential which drives lung cancer calcification. Metformin might prevent BMP-2 induced osteoblast-like phenotype of the lung cancer cells with concomitant inhibition of EMT to inhibit lung cancer tissue calcification.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sweta Makwana
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Shivani Bansal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Manas K Mahapatra
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Rashmi Tailor
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sandeep K Shrivastava
- Centre for Innovation, Research & Development, Dr. B. Lal Clinical Laboratory Pvt Ltd., Jaipur, Rajasthan, India
| | - Lokendra K Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
19
|
Yadav P, Bandyopadhayaya S, Soni S, Saini S, Sharma LK, Shrivastava SK, Mandal CC. Simvastatin prevents BMP-2 driven cell migration and invasion by suppressing oncogenic DNMT1 expression in breast cancer cells. Gene 2023; 882:147636. [PMID: 37442305 DOI: 10.1016/j.gene.2023.147636] [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: 01/14/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Both epigenetic and genetic changes in the cancer genome act simultaneously to promote tumor development and metastasis. Aberrant DNA methylation, a prime epigenetic event, is often observed in various cancer types. The elevated DNA methyltransferase 1 (DNMT1) enzyme creates DNA hypermethylation at CpG islands to drive oncogenic potential. This study emphasized to decipher the molecular mechanism of endogenous regulation of DNMT1 expression for finding upstream signaling molecules. Cancer database analyses found an upregulated DNMT1 expression in most cancer types including breast cancer. Overexpression of DNMT1 showed an increased cell migration, invasion, and stemness potential whereas 5-azacytidine (DNMT1 inhibitor) and siRNA mediated knockdown of DNMT1 exhibited inhibition of such cancer activities in breast cancer MDA-MB-231 and MCF-7 cells. Infact, cancer database analyses further found a positive correlation of DNMT1 transcript with both cholesterol pathway regulatory genes and BMP signaling molecules. Experimental observations documented that the cholesterol-lowering drug, simvastatin decreased DNMT1 transcript as well as protein, whereas BMP-2 treatment increased DNMT1 expression in breast cancer cells. In addition, expression of various key cholesterol regulatory genes was found to be upregulated in response to BMP-2 treatment. Moreover, simvastatin inhibited BMP-2 induced DNMT1 expression in breast cancer cells. Thus, this study for the first time reveals that both BMP-2 signaling and cholesterol pathways could regulate endogenous DNMT1 expression in cancer cells.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Sunil Saini
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Lokendra K Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, U.P., India
| | - Sandeep K Shrivastava
- Centre for Innovation, Research & Development, Dr. B. Lal Clinical Laboratory Pvt Ltd. Jaipur, Rajasthan, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India.
| |
Collapse
|
20
|
Liu C, Chen H, Hu B, Shi J, Chen Y, Huang K. New insights into the therapeutic potentials of statins in cancer. Front Pharmacol 2023; 14:1188926. [PMID: 37484027 PMCID: PMC10359995 DOI: 10.3389/fphar.2023.1188926] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
The widespread clinical use of statins has contributed to significant reductions of cardiovascular morbidity and mortality. Increasing preclinical and epidemiological evidences have revealed that dyslipidemia is an important risk factor for carcinogenesis, invasion and metastasis, and that statins as powerful inhibitor of HMG-CoA reductase can exert prevention and intervention effects on cancers, and promote sensitivity to anti-cancer drugs. The anti-cancer mechanisms of statins include not only inhibition of cholesterol biosynthesis, but also their pleiotropic effects in modulating angiogenesis, apoptosis, autophagy, tumor metastasis, and tumor microenvironment. Moreover, recent clinical studies have provided growing insights into the therapeutic potentials of statins and the feasibility of combining statins with other anti-cancer agents. Here, we provide an updated review on the application potential of statins in cancer prevention and treatment and summarize the underneath mechanisms, with focuses on data from clinical studies.
Collapse
Affiliation(s)
- Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Bicheng Hu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajian Shi
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Lee YCG, Chou FN, Tung SY, Chou HC, Ko TL, Fann YC, Juan SH. Tumoricidal Activity of Simvastatin in Synergy with RhoA Inactivation in Antimigration of Clear Cell Renal Cell Carcinoma Cells. Int J Mol Sci 2023; 24:ijms24119738. [PMID: 37298689 DOI: 10.3390/ijms24119738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Among kidney cancers, clear cell renal cell carcinoma (ccRCC) has the highest incidence rate in adults. The survival rate of patients diagnosed as having metastatic ccRCC drastically declines even with intensive treatment. We examined the efficacy of simvastatin, a lipid-lowering drug with reduced mevalonate synthesis, in ccRCC treatment. Simvastatin was found to reduce cell viability and increase autophagy induction and apoptosis. In addition, it reduced cell metastasis and lipid accumulation, the target proteins of which can be reversed through mevalonate supplementation. Moreover, simvastatin suppressed cholesterol synthesis and protein prenylation that is essential for RhoA activation. Simvastatin might also reduce cancer metastasis by suppressing the RhoA pathway. A gene set enrichment analysis (GSEA) of the human ccRCC GSE53757 data set revealed that the RhoA and lipogenesis pathways are activated. In simvastatin-treated ccRCC cells, although RhoA was upregulated, it was mainly restrained in the cytosolic fraction and concomitantly reduced Rho-associated protein kinase activity. RhoA upregulation might be a negative feedback effect owing to the loss of RhoA activity caused by simvastatin, which can be restored by mevalonate. RhoA inactivation by simvastatin was correlated with decreased cell metastasis in the transwell assay, which was mimicked in dominantly negative RhoA-overexpressing cells. Thus, owing to the increased RhoA activation and cell metastasis in the human ccRCC dataset analysis, simvastatin-mediated Rho inactivation might serve as a therapeutic target for ccRCC patients. Altogether, simvastatin suppressed the cell viability and metastasis of ccRCC cells; thus, it is a potentially effective ccRCC adjunct therapy after clinical validation for ccRCC treatment.
Collapse
Affiliation(s)
- Yuan-Chii Gladys Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Fang-Ning Chou
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Szu-Yu Tung
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsui-Ling Ko
- College of Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yang C Fann
- Intramural IT and Bioinformatics Program, Division of Intramural, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
22
|
Marwaha D, Gautam S, Singh N, Rai N, Sharma M, Tiwari P, Shukla RP, Urandur S, Banala VT, Mugale MN, Kumar A, Mishra PR. Synergistic delivery of Imatinib through multifunctional nano-crystalline capsules, in response to redox environment for improved breast cancer therapy. Colloids Surf B Biointerfaces 2023; 226:113316. [PMID: 37086687 DOI: 10.1016/j.colsurfb.2023.113316] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023]
Abstract
Chondroitin anchored crystalline nano-capsules bearing Imatinib (IMT), and simvastatin (SMV) was developed using Poly (L-lactic acid) (PLLA) by two-step method, i.e., firstly, by synthesizing chondroitin (CSA) anchored simvastatin (SMV) using cystamine as a spacer (SMV-SS-CSA) for disulfide triggered glutathione (GSH) sensitive release and secondly, by developing phenyl boronic ester grafted Pluronic F68 (PEPF) for H2O2 responsive release. By combining these conjugates, we have prepared crystalline nano-capsules (CNs) for preferential targeting of CD44 receptors. The developed CNs were spherical when characterized through SEM, TEM, and AFM for surface morphology, while changes in particle size and crystalline structure were confirmed through Quasi-Elastic light scattering (QELS) and Wide Angle X-ray Scattering (WAXS). The enhanced cellular uptake was noted in chondroitin-modified nano-capsules IMT/SMV-SS-CSA@CNs compared to unmodified nano-capsules IMT+SMV@CNs. IMT/SMV-SS-CSA@CNs displayed significantly higher G2/M phase arrest (76.9%) than unmodified nano-capsules. The prototype formulation (IMT/SMV-SS-CSA@CNs) showed an overall improved pharmacokinetic profile in terms of both half-life and AUC0-α. When tested in the 4T1 subcutaneously injected tumor-bearing Balb/c mice model, the tumor growth inhibition rate of IMT/SMV-SS-CSA@CNs was significantly higher (91%) than the IMT+SMV combination. Overall, the findings suggest that the proposed dual responsive chondroitin-modified drug delivery could have a step forward in achieving spatial and temporal targeting at the tumor site.
Collapse
Affiliation(s)
- Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Venkatesh Teja Banala
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | | | - Akhilesh Kumar
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
23
|
Takada K, Kashiwagi S, Iimori N, Kouhashi R, Yabumoto A, Goto W, Asano Y, Tauchi Y, Morisaki T, Ogisawa K, Shibutani M, Tanaka H, Maeda K. Impact of oral statin therapy on clinical outcomes in patients with cT1 breast cancer. BMC Cancer 2023; 23:224. [PMID: 36894884 PMCID: PMC9999569 DOI: 10.1186/s12885-023-10631-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/09/2023] [Indexed: 03/11/2023] Open
Abstract
PURPOSE A previous meta-analysis examining the relationship between statin use and breast cancer reported that the inhibitory effect of statins on breast cancer may be more pronounced in early-stage cases. In this study, we aimed to investigate the effects of hyperlipidemia treatment at the time of breast cancer diagnosis and to examine its correlation with metastasis to axillary lymph nodes among patients with so-called cT1 breast cancer whose primary lesion was 2 cm or less and was pathologically evaluated by sentinel lymph node biopsy or axillary lymph node dissection. We also investigated the effects of hyperlipidemic drugs on the prognosis of patients with early-stage breast cancer. METHODS After excluding cases that did not meet the criteria, we analyzed data from 719 patients who were diagnosed with breast cancer, with a primary lesion of 2 cm or less identified by preoperative imaging, and who underwent surgery without preoperative chemotherapy. RESULTS Regarding hyperlipidemia drugs, no correlation was found between statin use and lymph node metastasis (p = 0.226), although a correlation was found between lipophilic statin use and lymph node metastasis (p = 0.042). Also, the disease-free survival periods were prolonged following treatment of hyperlipidemia (p = 0.047, hazard ratio: 0.399) and statin administration (p = 0.028, hazard ratio: 0.328). CONCLUSION In cT1 breast cancer, the results suggest that oral statin therapy may contribute to favorable outcomes.
Collapse
Affiliation(s)
- Koji Takada
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Nozomi Iimori
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Rika Kouhashi
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Akimichi Yabumoto
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Wataru Goto
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yuka Asano
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yukie Tauchi
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tamami Morisaki
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kana Ogisawa
- Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masatsune Shibutani
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Hiroaki Tanaka
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kiyoshi Maeda
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
24
|
Kuldeep S, Soni S, Srivastava A, Mishra A, Sharma LK, Mandal CC. Dysregulated cholesterol regulatory genes as a diagnostic biomarker for cancer. J Gene Med 2023; 25:e3475. [PMID: 36670344 DOI: 10.1002/jgm.3475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/04/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND A dysregulation of cholesterol homeostasis is often seen in various cancer cell types, and elevated cholesterol content and that of its metabolites appears to be crucial for cancer progression and metastasis. Cholesterol is a precursor of various steroid hormones and a key plasma membrane component especially in lipid-rafts, also modulating many intracellular signaling pathways. METHODS To provide an insight of dysregulated cholesterol regulatory genes, their transcript levels were analyzed in different cancers and their influence was correlated with the overall survival of cancer patients using cancer database analysis. RESULTS This analysis found a set of genes (e.g., ACAT1, RXRA, SOAT1 and SQLE) that were not only often dysregulated, but also had been associated with poorer overall survival in most cancer types. Quantitative reverse transcriptase-polymerase chain reaction analysis revealed elevated SQLE and SOAT1 transcript levels and downregulated expression of RXRA and ACAT1 genes in triple negative breast cancer tissues compared to adjacent control tissues, indicating that this dysregulated expression of the gene signature is a diagnostic marker for breast cancer. CONCLUSION For the first time, the present study identified a gene signature associated with the dysregulation of cholesterol homeostasis in cancer cells that may not only be used as a diagnostic marker, but also comprise a promising drug target for the advancement of cancer therapy.
Collapse
Affiliation(s)
- Seema Kuldeep
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Anubhav Srivastava
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences-, Lucknow, Uttar Pradesh, India
| | - Anjali Mishra
- Department of Endocrine and Breast Surgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences-, Lucknow, Uttar Pradesh, India
| | - Lokendra Kumar Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences-, Lucknow, Uttar Pradesh, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
25
|
Goto W, Kashiwagi S, Takada K, Asano Y, Ogisawa K, Morisaki T, Shibutani M, Tanaka H, Maeda K. Clinical verification of the relationship between serum lipid metabolism and immune activity in breast cancer patients treated with neoadjuvant chemotherapy. Eur J Med Res 2023; 28:2. [PMID: 36593486 PMCID: PMC9806883 DOI: 10.1186/s40001-022-00964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lipid metabolism has been recently reported to affect the prognosis and tumor immune activity in cancer patients. However, the effect of lipid metabolism on chemosensitivity in patients with breast cancer treated with neoadjuvant chemotherapy (NAC) remains unclear. METHODS We examined 327 patients with breast cancer who were treated with NAC followed by curative surgery. The correlations between the serum levels of total cholesterol (TC) and triglyceride (TG) and the clinicopathological features, including the efficacy of NAC, neutrophil-to-lymphocyte ratio (NLR), and absolute lymphocyte count (ALC), were evaluated retrospectively. RESULTS Serum TG levels were increased after NAC in all the subtypes, and the rate of change was the highest, especially in triple-negative breast cancer (TNBC) (21.0% → 48.1%). In addition, only TNBC patients with an objective response (OR) had significantly higher TG levels after NAC than those without (P = 0.049). Patients with a high ALC before NAC had significantly higher TG levels after NAC than patients with all breast cancer (P = 0.001), HER2-enriched breast cancer (P = 0.021), and TNBC (P = 0.008). Patients with a low NLR before NAC had significantly higher TG levels after NAC only among patients with TNBC (P = 0.025). In patients with human epidermal growth factor receptor 2-enriched breast cancer, the group with normal TC levels before NAC had significantly better OS than those with high TC levels (P = 0.013, log-rank test), and in patients with TNBC, the group with high TC levels after NAC had significantly better OS than those with normal TC levels (P = 0.014, log-rank test). CONCLUSIONS Good systemic immune activity and chemosensitivity may be associated with lipid metabolism regulated by NAC in TNBC patients.
Collapse
Affiliation(s)
- Wataru Goto
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Shinichiro Kashiwagi
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Koji Takada
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Yuka Asano
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Kana Ogisawa
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Tamami Morisaki
- grid.258799.80000 0004 0372 2033Department of Breast Surgical Oncology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Masatsune Shibutani
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Hiroaki Tanaka
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| | - Kiyoshi Maeda
- grid.258799.80000 0004 0372 2033Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-Ku, Osaka, 545-8585 Japan
| |
Collapse
|
26
|
Watson R, Tulk A, Erdrich J. The Link Between Statins and Breast Cancer in Mouse Models: A Systematic Review. Cureus 2022; 14:e31893. [PMID: 36579200 PMCID: PMC9790759 DOI: 10.7759/cureus.31893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 11/27/2022] Open
Abstract
Statins, a class of cholesterol-lowering drugs, have consistently demonstrated pleiotropic effects in both preclinical and clinical studies. Outside of inhibiting the production of cholesterol in cells, statins have shown antineoplastic properties most commonly in breast cancer. Clinical and epidemiological studies, however, are less definitive than preclinical studies regarding statins as potential adjuvant oncologic therapy. Our objective is to summarize mouse model studies that investigate the link between statins and breast cancer using a cancer care continuum framework to provide a clinically relevant picture of the potential use of statins in breast cancer. A systematic review of the PubMed database was performed to identify studies published between January 2007 and July 2022 that investigated the effects of statins on breast cancer prevention, treatment, and survivorship in mouse models. Overall, 58 studies were identified using our search strategy. Based on our inclusion and exclusion criteria, 26 mouse model studies were eligible to be included in our systematic review. In breast cancer mouse models, statins alone and in combination with anti-cancer therapies demonstrate proven antineoplastic effects across the cancer care continuum. The antineoplastic benefit of statins as single agents in mouse model studies helps inform their synergistic benefit that future clinical studies can test. Parameters such as statin timing, dose, and breast cancer subtype are key stepping stones in defining how statins could be used in the treatment of breast cancer.
Collapse
Affiliation(s)
- Raj Watson
- Department of Surgery, University of Arizona College of Medicine - Tucson, Tucson, USA
| | - Angela Tulk
- Department of Surgery, University of Arizona College of Medicine - Tucson, Tucson, USA
| | - Jennifer Erdrich
- Department of Surgery, University of Arizona College of Medicine - Tucson, Tucson, USA
| |
Collapse
|
27
|
Cao L, Fang H, Yan D, Wu XM, Zhang J, Chang MX. CD44a functions as a regulator of p53 signaling, apoptosis and autophagy in the antibacterial immune response. Commun Biol 2022; 5:889. [PMID: 36042265 PMCID: PMC9427754 DOI: 10.1038/s42003-022-03856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/17/2022] [Indexed: 11/26/2022] Open
Abstract
The cell adhesion molecule CD44 has been implicated in diverse biological functions including the pathological responses to infections and inflammatory diseases. The variable forms of CD44 contribute to functional variations, which are not yet defined in teleost. Here, we show that zebrafish CD44a plays a protective role in the host defense against Edwardsiella piscicida infection. Zebrafish CD44a deficiency inhibits cell growth and proliferation, impairs cell growth and death pathways, and regulates the expression levels of many genes involved in p53 signaling, apoptosis and autophagy. In addition, CD44a gene disruption in zebrafish leads to inhibition of apoptosis and induction of autophagy, with the increased susceptibility to E. piscicida infection. Furthermore, we show that zebrafish CD44a variants including CD44a_tv1 and CD44a_tv2 promote the translocation of p53 from the nucleus to the cytoplasm and interact with p53 in the cytoplasm. Mechanistically, zebrafish CD44a_tv1 mediates the beneficial effect for larvae survival infected with E. piscicida is depending on the CASP8-mediated apoptosis. However, the antibacterial effect of zebrafish CD44a_tv2 depends on the cytoplasmic p53-mediated inhibition of autophagy. Collectively, our results identify that different mechanisms regulate CD44a variants-mediated antibacterial responses.
Collapse
Affiliation(s)
- Lu Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hong Fang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Dong Yan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xiao Man Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
28
|
Cholesterol Synthesis Is Important for Breast Cancer Cell Tumor Sphere Formation and Invasion. Biomedicines 2022; 10:biomedicines10081908. [PMID: 36009455 PMCID: PMC9405659 DOI: 10.3390/biomedicines10081908] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer has a high risk of recurrence and distant metastasis after remission. Controlling distant metastasis is important for reducing breast cancer mortality, but accomplishing this goal remains elusive. In this study, we investigated the molecular pathways underlying metastasis using cells that mimic the breast cancer distant metastasis process. HCC1143 breast cancer cells were cultured under two-dimensional (2D)-adherent, tumor sphere (TS), and reattached (ReA) culture conditions to mimic primary tumors, circulating tumor cells, and metastasized tumors, respectively. ReA cells demonstrated increased TS formation and enhanced invasion capacity compared to the original 2D-cultured parental cells. In addition, ReA cells had a higher frequency of ESA+CD44+CD24− population, which represents a stem-cell-like cell population. RNA sequencing identified the cholesterol synthesis pathway as one of the most significantly increased pathways in TS and ReA cells compared to parental cells, which was verified by measuring intracellular cholesterol levels. Furthermore, the pharmacological inhibition of the cholesterol synthesis pathway decreased the ability of cancer cells to form TSs and invade. Our results suggest that the cholesterol synthesis pathway plays an important role in the distant metastasis of breast cancer cells by augmenting TS formation and invasion capacity.
Collapse
|
29
|
The mevalonate pathway in breast cancer biology. Cancer Lett 2022; 542:215761. [DOI: 10.1016/j.canlet.2022.215761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023]
|
30
|
Minichsdorfer C, Fuereder T, Leutner M, Singer CF, Kacerovsky-Strobl S, Egle D, Greil R, Balic M, Fitzal F, Pfeiler G, Frantal S, Bartsch R, Gnant M. Effect of concomitant statin treatment in postmenopausal patients with hormone receptor-positive early-stage breast cancer receiving adjuvant denosumab or placebo: a post hoc analysis of ABCSG-18. ESMO Open 2022; 7:100426. [PMID: 35334418 PMCID: PMC9058905 DOI: 10.1016/j.esmoop.2022.100426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 11/24/2022] Open
Abstract
Background Statins are cholesterol-lowering drugs prescribed for the prevention and treatment of cardiovascular disease. Moreover, statins may possess anticancer properties and interact with receptor activator of nuclear factor κB ligand expression. We aimed at evaluating a hypothetical synergistic effect of statins with denosumab in early-stage breast cancer (BC) patients from the Austrian Breast and Colorectal Cancer Study Group (ABCSG) trial 18. Patients and methods ABCSG-18 (NCT00556374) is a prospective, randomized, double-blind, phase III study; postmenopausal patients with hormone receptor-positive BC receiving a nonsteroidal aromatase inhibitor were randomly assigned to denosumab or placebo. In this post hoc analysis, we investigated the effects of concomitant statin therapy on recurrence risk (RR) of BC, fracture risk and bone mineral density (BMD). Results In the study population (n = 3420), statin therapy (n = 824) was associated with worse disease-free survival (DFS) [hazard ratio (HR) 1.35, 95% confidence interval (CI) 1.04-1.75; P = 0.023]. While no significant effect of lipophilic statins (n = 710) on RR was observed (HR 1.30, 95% CI 0.99-1.72; P = 0.062), patients on hydrophilic statins (n = 87) had worse DFS compared with patients not receiving any statins (HR 2.00, 95% CI 1.09-3.66; P = 0.026). This finding was mainly driven by the effect of hydrophilic statins on DFS in the denosumab arm (HR 2.63, 95% CI 1.21-5.68; P = 0.014). However, this effect subsided after correction for confounders in the sensitivity analysis. No association between statin use and fracture risk or osteoporosis was observed. Conclusion According to this analysis, hydrophilic statins showed a detrimental effect on DFS in the main model, which was attenuated after correction for confounders. Our data need to be interpreted with caution due to their retrospective nature and the low number of patients receiving hydrophilic statins. Statin co-medication was initially associated with a worse DFS in hormone receptor-positive early-stage BC patients. This effect was mainly driven by patients on hydrophilic statins. However, this effect subsided after correction for confounders in the sensitivity analysis. No association between statin use and fracture risk or osteoporosis was observed.
Collapse
Affiliation(s)
- C Minichsdorfer
- Departments of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - T Fuereder
- Departments of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - M Leutner
- Departments of Medicine 3, Clinical Division of Endocrinology, Medical University of Vienna, Vienna, Austria
| | - C F Singer
- Departments of Gynaecology, Medical University of Vienna, Vienna, Austria
| | | | - D Egle
- Department of Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - R Greil
- Department of Medicine 3, Paracelsus University Salzburg, Salzburg Cancer Research Institute-CCCIT, Cancer Cluster Salzburg, Salzburg, Austria
| | - M Balic
- Department of Medicine, Clinical Division of Oncology, Medical University of Graz, Graz, Austria
| | - F Fitzal
- General Surgery, Medical University of Vienna, Vienna, Austria
| | - G Pfeiler
- Departments of Gynaecology, Medical University of Vienna, Vienna, Austria
| | - S Frantal
- Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria
| | - R Bartsch
- Departments of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria.
| | - M Gnant
- Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria; Comprehensive Cancer Centre, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Relationship between serum lipid levels and the immune microenvironment in breast cancer patients: a retrospective study. BMC Cancer 2022; 22:167. [PMID: 35164691 PMCID: PMC8842971 DOI: 10.1186/s12885-022-09234-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Therapeutic agents for dyslipidaemia, in particular statins, have been recently reported to suppress growth and metastasis of breast cancer. However, the predictive value of lipid control in breast cancer patients has not been discussed sufficiently. In addition, though immunometabolism is a relatively novel approach for tumour immunotherapy, the relationship between lipid metabolism and immune status has not been well documented. We therefore investigated the effects of lipid metabolism on antitumour immune response and cancer prognosis. METHODS Except for patients with ductal carcinoma in situ, 938 patients treated with curative surgery were examined. The correlation between treatment for dyslipidaemia or serum lipid levels and clinicopathological features, including the prognosis, was evaluated retrospectively. Also, we stratified these results by intrinsic subtype of breast cancer, menopause, and type of therapeutic agents for dyslipidaemia. Moreover, neutrophil- to-lymphocyte ratio (NLR) and tumour-infiltrating lymphocytes (TILs) were used as indicators of systemic and local immune status, respectively. RESULTS Of 194 patients treated for dyslipidaemia, recurrence-free survival (RFS) and overall survival (OS) did not differ significantly between users of drugs for dyslipidaemia and non-users (p = 0.775 and p = 0.304, log-rank, respectively). Among postmenopausal, hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer patients treated for dyslipidaemia, the good serum lipid control group had significantly better RFS (p = 0.014, log-rank), lower postoperative NLR (p = 0.012), and higher TILs in resected tissues (p = 0.024) than the poor control group. Multivariate analysis showed that postoperative serum lipid levels were a risk factor for recurrence (hazard ratio = 4.722, 95% confidence interval 1.006-22.161, p = 0.049). CONCLUSIONS Good control of serum lipid metabolism may improve the tumour immune microenvironment and prognosis in postmenopausal HR-positive/HER2-negative breast cancer patients.
Collapse
|
32
|
Günaçar DN, Yemenoğlu H, Ustaoğlu G, Arıöz Ö. Effects of hyperlipidemia on trabecular and cortical structures of the mandible. Dentomaxillofac Radiol 2022; 51:20210214. [PMID: 34283651 PMCID: PMC8802705 DOI: 10.1259/dmfr.20210214] [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: 04/30/2021] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES To evaluate the mandibular bone structure of patients with hyperlipidemia by fractal dimension (FD) analysis and panoramic radiomorphometric indices including mandibular cortical thickness measurement, panoramic mandibular index (PMI), mandibular cortical index (MCI) and to compare with the healthy group. METHODS In total, 60 panoramic radiographs were included, including panoramic radiographs of 30 individuals with hyperlipidemia and 30 individuals with systemically healthy. FD analysis in the mandibular condyle, angle of the mandible, the distal side of the second premolar and anterior to mental foramen, PMI, MCI, and mandibular cortical thickness measurements were evaluated on radiographs. Independent samples t-test was used for differences between healthy and hyperlipidemia groups with regard to age and PMI. Repeated measurement of variance with one within and one between factors in the comparison of four regions and two groups in terms of FD and cortical thickness measurements. Following this analysis, significant differences were detected by post-hoc Sidak test. Fisher-Freeman-Halton analysis was applied to determine the relationship between categorical variables. RESULTS FD values of the hyperlipidemic patients were found to be lower than the healthy group. Between the hyperlipidemic and healthy groups, there was a difference in the angle of the mandible FD values (p = 0.020). There were no differences in the cortical thickness measurements and PMI between the groups (p > 0.05). There was a difference in MCI values between the groups (p < 0.05). CONCLUSION The trabecular structure of the angle of the mandible and the cortical bone structure of the mandible were found to be negatively affected by hyperlipidemia.
Collapse
Affiliation(s)
- Dilara Nil Günaçar
- Oral and Maxillofacial Radiology Department, Recep Tayyip Erdoğan University, Faculty of Dentistry, Rize, Turkey
| | - Hatice Yemenoğlu
- Periodontology Department, Recep Tayyip Erdoğan University, Faculty of Dentistry, Rize, Turkey
| | - Gülbahar Ustaoğlu
- Periodontology Department, Abant İzzet Baysal University, Faculty of Dentistry, Bolu, Turkey
| | - Özkan Arıöz
- Periodontology Department, Abant İzzet Baysal University, Faculty of Dentistry, Bolu, Turkey
| |
Collapse
|
33
|
Xiao Y, Liu Q, Peng N, Li Y, Qiu D, Yang T, Kang R, Usmani A, Amadasu E, Borlongan CV, Yu G. Lovastatin Inhibits RhoA to Suppress Canonical Wnt/β-Catenin Signaling and Alternative Wnt-YAP/TAZ Signaling in Colon Cancer. Cell Transplant 2022; 31:9636897221075749. [PMID: 35168393 PMCID: PMC8855423 DOI: 10.1177/09636897221075749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/25/2021] [Accepted: 01/08/2022] [Indexed: 12/14/2022] Open
Abstract
Statins are first-line drugs used to control patient lipid levels, but there is recent evidence that statin treatment can lower colorectal cancer (CRC) incidence by 50% and prolong CRC patient survival through mechanisms that are poorly understood. In this study, we found that the treatment of APCmin mice by the mevalonate pathway inhibitor lovastatin significantly reduced the number of colonic masses and improved hypersplenism and peripheral anemia. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) analysis of colonic mass tissues showed a potent inhibitory effect in both Wnt/β-catenin signaling and YAP/TAZ signaling in the lovastatin treatment group. The results of our transcriptomic analyses in RKO indicated that lovastatin regulated several proliferation-related signaling pathways. Moreover, lovastatin suppressed important genes and proteins related to the canonical Wnt/β-catenin and alternative Wnt-YAP/TAZ signaling pathways in RKO and SW480 cells, and these effects were rescued by mevalonic acid (MVA), as confirmed through a series of Western blotting, RT-PCR, and reporter assays. Given that statins suppress oncogenic processes primarily through the inhibition of Rho GTPase in the mevalonate pathway, we speculate that lovastatin can inhibit certain Rho GTPases to suppress both canonical Wnt/β-catenin signaling and alternative Wnt-YAP/TAZ signaling. In RKO cells, lovastatin showed similar inhibitory properties as the RhoA inhibitor CCG1423, being able to inhibit β-catenin, TAZ, and p-LATS1 protein activity. Our results revealed that lovastatin inhibited RhoA activity, thereby suppressing the downstream canonical Wnt/β-catenin and alternative Wnt-YAP/TAZ pathways in colon cancer cells. These inhibitory properties suggest the promise of statins as a treatment for CRC. Altogether, the present findings support the potential clinical use of statins in non-cardiovascular contexts and highlight novel targets for anticancer treatments.
Collapse
Affiliation(s)
- Yi Xiao
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Qin Liu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Nanyin Peng
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Yuzhang Li
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Danyang Qiu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Tianlun Yang
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Richard Kang
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Ahsan Usmani
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Efosa Amadasu
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Cesario V. Borlongan
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Guolong Yu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Nowakowska MK, Lei X, Thompson MT, Shaitelman SF, Wehner MR, Woodward WA, Giordano SH, Nead KT. Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer 2021; 127:4142-4150. [PMID: 34342892 PMCID: PMC11912801 DOI: 10.1002/cncr.33797] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/20/2021] [Accepted: 06/24/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Previous studies have examined the association of statin therapy and breast cancer outcomes with mixed results. The objective of this study was to investigate the clinical effects of incident statin use among individuals with triple-negative breast cancer (TNBC). METHODS Data from the Surveillance, Epidemiology, and End Results-Medicare and Texas Cancer Registry-Medicare databases were used, and women aged ≥66 years who had stage I, II, and III breast cancer were identified. Multivariable Cox proportional hazards regression models were used to examine the association of new statin use in the 12 months after a breast cancer diagnosis with overall survival (OS) and breast cancer-specific survival (BCSS). RESULTS When examining incident statin use, defined as the initiation of statin therapy in the 12 months after breast cancer diagnosis, a significant association was observed between statin use and improved BCSS (standardized hazard ratio, 0.42; 95% confidence interval [CI], 0.20-0.88; P = .022) and OS (hazard ratio, 0.70; 95% CI, 0.50-0.99; P = .046) among patients with TNBC (n = 1534). No association was observed with BCSS (standardized hazard ratio, 0.99; 95% CI, 0.71-1.39; P = .97) or OS (hazard ratio, 1.04; 95% CI, 0.92-1.17; P = .55) among those without TNBC (n = 15,979). The results were consistent when examining statin exposure as a time-varying variable. CONCLUSIONS Among women with I, II, and III TNBC, initiation of statin therapy in the 12 months after breast cancer diagnosis was associated with an OS and BCSS benefit. Statins may have a role in select patients with breast cancer, and further investigation is warranted.
Collapse
Affiliation(s)
| | - Xiudong Lei
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mikayla T Thompson
- School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Simona F Shaitelman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mackenzie R Wehner
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendy A Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sharon H Giordano
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kevin T Nead
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
35
|
Arnold S, Kortland J, Maltseva DV, Nersisyan SA, Samatov TR, Lezius S, Tonevitsky AG, Milde-Langosch K, Wicklein D, Schumacher U, Stürken C. Fra-2 overexpression upregulates pro-metastatic cell-adhesion molecules, promotes pulmonary metastasis, and reduces survival in a spontaneous xenograft model of human breast cancer. J Cancer Res Clin Oncol 2021; 148:1525-1542. [PMID: 34693476 PMCID: PMC9114065 DOI: 10.1007/s00432-021-03812-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/22/2021] [Indexed: 11/28/2022]
Abstract
Purpose The transcription factor Fra-2 affects the invasive potential of breast cancer cells by dysregulating adhesion molecules in vitro. Previous results suggested that it upregulates the expression of E- and P-selectin ligands. Such selectin ligands are important members of the leukocyte adhesion cascade, which govern the adhesion and transmigration of cancer cells into the stroma of the host organ of metastasis. As so far, no in vivo data are available, this study was designed to elucidate the role of Fra-2 expression in a spontaneous breast cancer metastasis xenograft model. Methods The effect of Fra-2 overexpression in two stable Fra-2 overexpressing clones of the human breast cancer cell line MDA MB231 on survival and metastatic load was studied after subcutaneous injection into scid and E- and P-selectin-deficient scid mice. Results Fra-2 overexpression leads to a significantly shorter overall survival and a higher amount of spontaneous lung metastases not only in scid mice, but also in E- and P-deficient mice, indicating that it regulates not only selectin ligands, but also selectin-independent adhesion processes. Conclusion Thus, Fra-2 expression influences the metastatic potential of breast cancer cells by changing the expression of adhesion molecules, resulting in increased adherence to endothelial cells in a breast cancer xenograft model. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03812-2.
Collapse
Affiliation(s)
- Sabrina Arnold
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jan Kortland
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Diana V Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia
| | - Stepan A Nersisyan
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia
| | - Timur R Samatov
- Evotec International GmbH, Marie-Curie-Str. 7, 37079, Göttingen, Germany
| | - Susanne Lezius
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Alexander G Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia.,Scientific Research Center Bioclinicum, Ugreshskaya Str. 2/85, 115088, Moscow, Russia
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christine Stürken
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
36
|
Yao X, Xie R, Cao Y, Tang J, Men Y, Peng H, Yang W. Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology 2021; 19:311. [PMID: 34627266 PMCID: PMC8502296 DOI: 10.1186/s12951-021-01058-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/22/2021] [Indexed: 01/21/2023] Open
Abstract
Triple-negative breast cancer (TNBC), a management of aggressive breast cancer, remains an unmet medical challenge. Although a wave of efforts had spurred to design novel therapeutic method of TNBC, unpredictable prognosis with lacking effective therapeutic targets along with the resistance to apoptosis seriously limited survival benefits. Ferroptosis is a non-apoptotic form of cell death that is induced by excessive lipid peroxidation, which provide an innovative way to combat cancer. Emerging evidence suggests that ferroptosis plays an important role in the treatment of TNBC cells. Herein, a novel ferroptosis nanomedicine was prepared by loading simvastatin (SIM), a ferroptosis drug, into zwitterionic polymer coated magnetic nanoparticles (Fe3O4@PCBMA) to improve the therapeutic effect of TNBC. The as-obtained Fe3O4@PCBMA-SIM nanoparticles demonstrated more cytotoxicity against MDA-MB-231 than MCF-7 due to the higher expression of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), which demonstrated that statins could effectively kill TNBC. Further experiments showed that SIM could inhibit the expression of HMGCR to downregulate the mevalonate (MVA) pathway and glutathione peroxidase 4 (GPX4), thereby inducing cancer cell ferroptosis. What's more, PCBMA endows Fe3O4@PCBMA longer blood circulation performance to enhance their accumulation at tumor sites. Given that Fe3O4 have proven for clinical applications by the U.S. Food and Drug Administration (FDA) and SIM could induce cancer cell ferroptosis, the developed Fe3O4@PCBMA-SIM nanosystem would have great potential in clinics for overcoming the drug resistance brought about by apoptotic drugs to cancer cells.
Collapse
Affiliation(s)
- Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Ruihong Xie
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Yongbin Cao
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Jing Tang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Yongzhi Men
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haibao Peng
- Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
37
|
Kumar N, Mandal CC. Cholesterol-Lowering Drugs on Akt Signaling for Prevention of Tumorigenesis. Front Genet 2021; 12:724149. [PMID: 34603386 PMCID: PMC8483559 DOI: 10.3389/fgene.2021.724149] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Cholesterol has been reported to be accumulated in cancer cells. The metabolic dysregulation of the cholesterol is associated with tumor development and progression. The cholesterol-lowering drugs have been found to be involved in the prevention and treatment of various cancers. Akt, a serine/threonine kinase, can modulate the role of several downstream proteins involved in cell proliferation, migration, invasion, metabolism, and apoptosis. Since its involvement in several signaling pathways, its dysregulation is commonly reported in several cancers. Thus, targeting Akt could be an effective approach for cancer prevention and therapy. Cholesterol-lowering drugs have been found to affect the expression of Akt, and its activation in the cancer cells and thus have shown anticancer activity in different type of cancers. These drugs act on various signaling pathways such as PTEN/Akt, PI3k/Akt, Akt/NF-κB, Akt/FOXO1, Akt/mTOR, etc., which will be discussed in this article. This review article will discuss the significance of cholesterol in cancer cells, cholesterol-lowering drugs, the role of Akt in cancer cells, and the effects of cholesterol-lowering drugs on Akt in the prevention of therapy resistance and metastasis.
Collapse
Affiliation(s)
- Navneet Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
38
|
Laskus-Zakrzewska A, Kazimierczak P, Kolmas J. Porous Composite Granules with Potential Function of Bone Substitute and Simvastatin Releasing System: A Preliminary Study. MATERIALS (BASEL, SWITZERLAND) 2021; 14:5068. [PMID: 34501158 PMCID: PMC8434560 DOI: 10.3390/ma14175068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/19/2022]
Abstract
In this work, 3D porous granules based on Zn and Se-containing calcium phosphates (CaPs) were fabricated using a droplet-extrusion technique. The composite beads varied in composition and contained two different natural polymers: sodium alginate (SA) and gelatin (GEL). To analyse and compare their physicochemical properties, such as porosity and morphology, different techniques were applied, including scanning electron microscopy (SEM), sorption of N2 and mercury porosimetry. Prior to the fabrication of the granules, the properties of CaPs materials, (the bioceramic base of the beads), selenium (IV)-substituted hydroxyapatite (Se-HA) and zinc-substituted dicalcium phosphate dihydrate (Zn-DCPD), were also investigated. The results of cell viability assessment showed that Se-HA powder was non-toxic to human osteoblasts (hFOB 1.19) and simultaneously exhibited high toxicity to tumour cells (Saos-2). Once the cytotoxicity assay was completed, Se-HA and Zn-DCPD were used to prepare 3D materials. The prepared porous granules were used as matrices to deliver simvastatin to bones. Simvastatin was applied in either the lipophilic form or hydrophilic form. The release kinetics of simvastatin from granules of different composition was then assessed and compared.
Collapse
Affiliation(s)
- Aleksandra Laskus-Zakrzewska
- Department of Analytical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, ul. Banacha 1, 02-097 Warsaw, Poland;
| | - Paulina Kazimierczak
- Independent Unit of Tissue Engineering and Regenerative Medicine, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland;
| | - Joanna Kolmas
- Department of Analytical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, ul. Banacha 1, 02-097 Warsaw, Poland;
| |
Collapse
|
39
|
Malviya R, Raj S, Fuloria S, Subramaniyan V, Sathasivam K, Kumari U, Unnikrishnan Meenakshi D, Porwal O, Hari Kumar D, Singh A, Chakravarthi S, Kumar Fuloria N. Evaluation of Antitumor Efficacy of Chitosan-Tamarind Gum Polysaccharide Polyelectrolyte Complex Stabilized Nanoparticles of Simvastatin. Int J Nanomedicine 2021; 16:2533-2553. [PMID: 33824590 PMCID: PMC8018389 DOI: 10.2147/ijn.s300991] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The present study was intended to fabricate chitosan (Ch)-tamarind gum polysaccharide (TGP) polyelectrolyte complex stabilized cubic nanoparticles of simvastatin and evaluate their potential against human breast cancer cell lines. MATERIALS AND METHODS The antisolvent precipitation method was used for formulation of nanoparticles. Factorial design (32) was utilized as a tool to analyze the effect of Ch and TGP concentration on particle size and entrapment efficiency of nanoparticles. RESULTS Formulated nanoparticles showed high entrapment efficiency (67.19±0.42-83.36±0.23%) and small size (53.3-383.1 nm). The present investigation involved utilization of two biological membranes (egg and tomato) as biological barriers for drug release. The study revealed that drug release from tomato membranes was retarded (as compared to egg membranes) but the release pattern matched that of egg membranes. All formulations followed the Baker-Lansdale model of drug release irrespective of the two different biological barriers. Stability studies were carried out for 45 days and exhibited less variation in particle size as well as a reduction in entrapment efficiency. Simvastatin loaded PEC stabilized nanoparticles exhibited better control on growth of human breast cancer cell lines than simple simvastatin. An unusual anticancer effect of simvastatin nanoparticles is also supported by several other research studies. CONCLUSION The present study involves first-time synthesis of Ch-TGP polyelectrolyte complex stabilized nanoparticles of simvastatin against MCF-7 cells. It recommends that, in future, theoretical modeling and IVIVC should be carried out for perfect designing of delivery systems.
Collapse
Affiliation(s)
- Rishabha Malviya
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida, U.P., India
| | - Shakshi Raj
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida, U.P., India
| | - Shivkanya Fuloria
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Kedah, 08100, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Pharmacology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur, 42610, Malaysia
| | - Kathiresan Sathasivam
- Department of Biotechnology, Faculty of Applied Science, AIMST University, Kedah, 08100, Malaysia
| | - Usha Kumari
- Department of Physiology, Faculty of Medicine, AIMST University, Kedah, 08100, Malaysia
| | | | - Omji Porwal
- Department of Pharmacognosy, Faculty of Pharmacy, Tishk International University, Erbil, 44001, KRG, Iraq
| | - Darnal Hari Kumar
- Department of Pathology, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Johor Bahru, 80200, Malaysia
| | - Amit Singh
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida, U.P., India
| | - Srikumar Chakravarthi
- Department of Pathology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur, 42610, Malaysia
| | - Neeraj Kumar Fuloria
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AIMST University, Kedah, 08100, Malaysia
| |
Collapse
|
40
|
Woollam M, Wang L, Grocki P, Liu S, Siegel AP, Kalra M, Goodpaster JV, Yokota H, Agarwal M. Tracking the Progression of Triple Negative Mammary Tumors over Time by Chemometric Analysis of Urinary Volatile Organic Compounds. Cancers (Basel) 2021; 13:1462. [PMID: 33806757 PMCID: PMC8004946 DOI: 10.3390/cancers13061462] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that volatile organic compounds (VOCs) are potential biomarkers of breast cancer. An unanswered question is how urinary VOCs change over time as tumors progress. To explore this, BALB/c mice were injected with 4T1.2 triple negative murine tumor cells in the tibia. This typically causes tumor progression and osteolysis in 1-2 weeks. Samples were collected prior to tumor injection and from days 2-19. Samples were analyzed by headspace solid phase microextraction coupled to gas chromatography-mass spectrometry. Univariate analysis identified VOCs that were biomarkers for breast cancer; some of these varied significantly over time and others did not. Principal component analysis was used to distinguish Cancer (all Weeks) from Control and Cancer Week 1 from Cancer Week 3 with over 90% accuracy. Forward feature selection and linear discriminant analysis identified a unique panel that could identify tumor presence with 94% accuracy and distinguish progression (Cancer Week 1 from Cancer Week 3) with 97% accuracy. Principal component regression analysis also demonstrated that a VOC panel could predict number of days since tumor injection (R2 = 0.71 and adjusted R2 = 0.63). VOC biomarkers identified by these analyses were associated with metabolic pathways relevant to breast cancer.
Collapse
Affiliation(s)
- Mark Woollam
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Luqi Wang
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Paul Grocki
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Shengzhi Liu
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Amanda P. Siegel
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Maitri Kalra
- Hematology and Oncology, Ball Memorial Hospital, Indiana University Health, Muncie, IN 47303, USA;
| | - John V. Goodpaster
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
| | - Hiroki Yokota
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
- Biomechanics and Biomaterials Research Center, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Mangilal Agarwal
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Mechanical & Energy Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| |
Collapse
|
41
|
Bandyopadhayaya S, Akimov MG, Verma R, Sharma A, Sharma D, Kundu GC, Gretskaya NM, Bezuglov VV, Mandal CC. N-arachidonoyl dopamine inhibits epithelial-mesenchymal transition of breast cancer cells through ERK signaling and decreasing the cellular cholesterol. J Biochem Mol Toxicol 2021; 35:e22693. [PMID: 33393692 DOI: 10.1002/jbt.22693] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/04/2020] [Accepted: 12/11/2020] [Indexed: 01/02/2023]
Abstract
N-acyl dopamines (NADAs) are bioactive lipids of the endovanilloid family with known cytotoxicity for the cancer cells; however, the available data on the participation of the endovanilloids in epithelial-mesenchymal transition (EMT) and cancer stemness are controversial. This study unveils the inhibitory role of N-arachidonoyl dopamine (AA-DA), a typical representative of the NADA family, in breast cancer cell migration, EMT, and stemness. AA-DA treatment also led to a decrease in cholesterol biosynthesis gene expressions, and addition of exogenous cholesterol reverted these AA-DA-mediated inhibitory effects. Notably, AA-DA treatment inhibited the key regulatory gene of the cholesterol biosynthesis pathway, sterol regulatory element-binding protein 1 (SREBP1), with concurrent repression of the endoplasmic reticulum kinase 1/2 (ERK1/2) pathway. Furthermore, U0126, an ERK inhibitor, inhibited SREBP1 and decreased cellular cholesterol level, unwinding the molecular mechanism behind AA-DA-mediated anticancer activity. Thus, we, for the first time, revealed that AA-DA counteracts breast cancer EMT via inhibition of ERK signaling and cholesterol content.
Collapse
Affiliation(s)
- Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Mikhail G Akimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Ranjeet Verma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Ankit Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Divya Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Gopal C Kundu
- School of Biotechnology, Institute of Eminence, KIIT Deemed to be University, Bhubaneswar, India
| | - Natalia M Gretskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Vladimir V Bezuglov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS, Moscow, Russia
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
42
|
Bytautaite M, Petrikaite V. Comparative Study of Lipophilic Statin Activity in 2D and 3D in vitro Models of Human Breast Cancer Cell Lines MDA-MB-231 and MCF-7. Onco Targets Ther 2020; 13:13201-13209. [PMID: 33380809 PMCID: PMC7769197 DOI: 10.2147/ott.s283033] [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: 09/21/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Statins are a type of drugs that are used to lower cholesterol level in blood. Since the early 1990s, it has been known that statins could be beneficial in cancer therapy. However, data remain controversial, especially regarding estrogen receptors status. Despite many studies in breast cancer models in vitro, the correlations of effects of separate statins in various model systems remain unclear. Aim Our aim was to evaluate the anticancer activity of lovastatin, mevastatin, pitavastatin and simvastatin on different subtypes of human breast cancer (MDA-MB-231 and MCF-7 cell lines) in spatially different 2D and 3D cultures in vitro. Materials and Methods Cell viability was tested using MTT assay. Effect of statins on cell colony formation was evaluated by calculating breast cancer cell colony area and number. The effect on cell migration was estimated by “wound healing” assay. The activity of compounds in 3D cultures was evaluated by measuring the spheroid size changes during incubation. Results Among the tested statins, pitavastatin had the greatest effect on the viability of breast cancer MDA-MB-231 and MCF-7 cell lines. The mevastatin and pitavastatin mostly decreased the MDA-MB-231 and MCF-7 cell colony formation. All statins at 90% of their estimated effective concentration (EC50) and simvastatin at 10% of its EC50 concentration suppressed the MCF-7 cells migration in “wound healing” assay. Only higher concentrations of mevastatin and pitavastatin slowed down the MDA-MB-231 cell migration. Statins showed different activity on 3D cell cultures growth. Lovastatin and simvastatin delayed the growth of MDA-MB-231 cell spheroids, while mevastatin and pitavastatin reduced the growth of MCF-7 spheroids. Conclusion Statins possess different anticancer activity in human breast cancer MDA-MB-231 and MCF-7 cell cultures. Pitavastatin and simvastatin showed the highest activity in most tested assays, especially against MCF-7 cell line.
Collapse
Affiliation(s)
- Meda Bytautaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| | - Vilma Petrikaite
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, LT-50162, Lithuania
| |
Collapse
|
43
|
Schweiger MW, Li M, Giovanazzi A, Fleming RL, Tabet EI, Nakano I, Würdinger T, Chiocca EA, Tian T, Tannous BA. Extracellular Vesicles Induce Mesenchymal Transition and Therapeutic Resistance in Glioblastomas through NF-κB/STAT3 Signaling. ADVANCED BIOSYSTEMS 2020; 4:e1900312. [PMID: 32519463 PMCID: PMC7718424 DOI: 10.1002/adbi.201900312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 02/05/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor and despite optimal treatment, long-term survival remains uncommon. GBM can be roughly divided into three different molecular subtypes, each varying in aggressiveness and treatment resistance. Recent evidence shows plasticity between these subtypes in which the proneural (PN) glioma stem-like cells (GSCs) undergo transition into the more aggressive mesenchymal (MES) subtype, leading to therapeutic resistance. Extracellular vesicles (EVs) are membranous structures secreted by nearly every cell and are shown to play a key role in GBM progression by acting as multifunctional signaling complexes. Here, it is shown that EVs derived from MES cells educate PN cells to increase stemness, invasiveness, cell proliferation, migration potential, aggressiveness, and therapeutic resistance by inducing mesenchymal transition through nuclear factor-κB/signal transducer and activator of transcription 3 signaling. The findings could potentially help explore new treatment strategies for GBM and indicate that EVs may also play a role in mesenchymal transition of different tumor types.
Collapse
Affiliation(s)
- Markus W. Schweiger
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
- Department of Neurosurgery, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Mao Li
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Alberta Giovanazzi
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
- Department of Neurosurgery, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Renata L. Fleming
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
| | - Elie I. Tabet
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
- Department of Biomedical Engineering, University of South Dakota, 4800 N. Career Ave, Suite 221, Sioux Falls, SD USA
| | - Ichiro Nakano
- Department of Neurosurgery and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL USA
| | - Thomas Würdinger
- Department of Neurosurgery, Cancer Center Amsterdam, Brain Tumor Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - E. Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, Massachusetts 02115, United States
| | - Tian Tian
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
- Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bakhos A. Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA 02129, USA
- Neuroscience Program, Harvard Medical School, Boston MA 02129, USA
| |
Collapse
|
44
|
Mandal CC. Osteolytic metastasis in breast cancer: effective prevention strategies. Expert Rev Anticancer Ther 2020; 20:797-811. [PMID: 32772585 DOI: 10.1080/14737140.2020.1807950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Breast cancer is the most common cancer in women throughout the world. Patients who are diagnosed early generally have better prognosis and survivability. Indeed, advanced stage breast cancer often develops osteolytic metastases, leading to bone destruction. Although there are select drugs available to treat bone metastatic disease, these drugs have shown limited success. AREA COVERED This paper emphasizes updated mechanisms of bone remodeling and osteolytic bone metastases of breast cancer. This article also aims to explore the potential of novel natural and synthetic therapeutics in the effective prevention of breast cancer-induced osteolysis and osteolytic metastases of breast cancer. EXPERT OPINION Targeting TGFβ and BMP signaling pathways, along with osteoclast activity, appears to be a promising therapeutic strategy in the prevention of breast cancer-induced osteolytic bone destruction and metastatic growth at bone metastatic niches. Pilot studies in animal models suggest various natural and synthetic compounds and monoclonal antibodies as putative therapeutics in the prevention of breast cancer stimulated osteolytic activity. However, comprehensive pre-clinical studies demonstrating the PK/PD and in-depth understanding of molecular mechanism(s) by which these potential molecules exhibit anti-tumor growth and anti-osteolytic activity are still required to develop effective therapies against breast cancer-induced osteolytic bone disease.
Collapse
Affiliation(s)
- Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan , Ajmer, India
| |
Collapse
|
45
|
Liu SH, Yu J, Creeden JF, Sutton JM, Markowiak S, Sanchez R, Nemunaitis J, Kalinoski A, Zhang JT, Damoiseaux R, Erhardt P, Brunicardi FC. Repurposing metformin, simvastatin and digoxin as a combination for targeted therapy for pancreatic ductal adenocarcinoma. Cancer Lett 2020; 491:97-107. [PMID: 32829010 DOI: 10.1016/j.canlet.2020.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/23/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Patients with pancreatic adenocarcinoma (PDAC) have a 5-year survival rate of 8%, the lowest of any cancer in the United States. Traditional chemotherapeutic regimens, such as gemcitabine- and fluorouracil-based regimens, often only prolong survival by months. Effective precision targeted therapy is therefore urgently needed to substantially improve survival. In an effort to expedite approval and delivery of targeted therapy to patients, we utilized a platform to develop a novel combination of FDA approved drugs that would target pancreaticoduodenal homeobox1 (PDX1) and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) utilizing super-promoters of the target genes to interrogate an FDA approved drug library. We identified and selected metformin, simvastatin and digoxin (C3) as a novel combination of FDA approved drugs, which were shown to effectively target PDX1 and BIRC5 in human PDAC tumors in mice with no toxicity.
Collapse
Affiliation(s)
- Shi-He Liu
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| | - Juehua Yu
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Justin F Creeden
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jeffrey M Sutton
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Stephen Markowiak
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robbi Sanchez
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Andrea Kalinoski
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jian-Ting Zhang
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Paul Erhardt
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, 43614, USA
| | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| |
Collapse
|
46
|
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8:30. [PMID: 32793401 PMCID: PMC7391760 DOI: 10.1038/s41413-020-00105-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
As one of the most common metastatic sites of malignancies, bone has a unique microenvironment that allows metastatic tumor cells to grow and flourish. The fenestrated capillaries in the bone, bone matrix, and bone cells, including osteoblasts and osteoclasts, together maintain the homeostasis of the bone microenvironment. In contrast, tumor-derived factors act on bone components, leading to subsequent bone resorption or excessive bone formation. The various pathways involved also provide multiple targets for therapeutic strategies against bone metastases. In this review, we summarize the current understanding of the mechanism of bone metastases. Based on the general process of bone metastases, we specifically highlight the complex crosstalk between tumor cells and the bone microenvironment and the current management of cancer bone metastases.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| |
Collapse
|
47
|
Sharma T, Mandal CC. Omega-3 fatty acids in pathological calcification and bone health. J Food Biochem 2020; 44:e13333. [PMID: 32548903 DOI: 10.1111/jfbc.13333] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/19/2023]
Abstract
Omega-3 fatty acids (ω-3FAs) such as Docosahexaenoic acid (DHA) and Eicosapentanoic acid (EPA), are active ingredient of fish oil, which have larger health benefits against various diseases including cardiovascular, neurodegenerative, cancers and bone diseases. Substantial studies documented a preventive role of omega-3 fatty acids in pathological calcification like vascular calcification and microcalcification in cancer tissues. In parallel, these fatty acids improve bone quality probably by preventing bone decay and augmenting bone mineralization. This study also addresses that the functions of ω-3FAs not only depend on tissue types, but also work through different molecular mechanisms for preventing pathological calcification in various tissues and improving bone health. PRACTICAL APPLICATIONS: Practical applications of the current study are to improve the knowledge about the supplementation of omega-3 fatty acids. This study infers that supplementation of omega-3 fatty acids aids in bone preservation in elder females at the risk of osteoporosis and also, on the contrary, omega-3 fatty acids interfere with pathological calcification of vascular cells and cancer cells. Omega-3 supplementation should be given to the cardiac patients because of its cardio protective role. In line with this, omega-3 supplementation should be included with chemotherapy for cancer patients as it can prevent osteoblastic potential of breast cancer patients, responsible for pathological mineralization, and blocks off target toxicities. Administration of omega-3 fatty acid with chemotherapy will not only improve survival of cancer patients, but also improve the bone quality. Thus, this study allows a better understanding on omega-3 fatty acids in combating pathological complications such as osteoporosis, vascular calcification, and breast microcalcification.
Collapse
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
48
|
Bandyopadhayaya S, Ford B, Mandal CC. Cold-hearted: A case for cold stress in cancer risk. J Therm Biol 2020; 91:102608. [PMID: 32716858 DOI: 10.1016/j.jtherbio.2020.102608] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/25/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
A negative correlation exists between environmental temperature and cancer risk based on both epidemiological and statistical analyses. Previously, cold stress was reported to be an effective cause of tumorigenesis. Several studies have demonstrated that cold temperature serves as a potential risk factor in cancer development. Most recently, a link was demonstrated between the effects of extreme cold climate on cancer incidence, pinpointing its impact on tumour suppressor genes by causing mutation. The underlying mechanism behind cold stress and its association with tumorigenesis is not well understood. Hence, this review intends to shed light on the role of associated factors, genetic and/or non-genetic, which are modulated by cold temperature, and eventually influence tumorigenic potential. While scrutinizing the effect of cold exposure on the body, the expression of certain genes, e.g. uncoupled proteins and heat-shock proteins, were elevated. Biological chemicals such as norepinephrine, thyroxine, and cholesterol were also elevated. Brown adipose tissue, which plays an essential role in thermogenesis, displayed enhanced activity upon cold exposure. Adaptive measures are utilized by the body to tolerate the cold, and in doing so, invites both epigenetic and genetic changes. Unknowingly, these adaptive strategies give rise to a lethal outcome i.e., genesis of cancer. Concisely, this review attempts to draw a link between cold stress, genetic and epigenetic changes, and tumorigenesis and aspires to ascertain the mechanism behind cold temperature-mediated cancer risk.
Collapse
Affiliation(s)
| | - Bridget Ford
- Department of Biology, University of the Incarnate Word, San Antonio, TX, 78209, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, 305817, India.
| |
Collapse
|
49
|
Cholesterol and beyond - The role of the mevalonate pathway in cancer biology. Biochim Biophys Acta Rev Cancer 2020; 1873:188351. [PMID: 32007596 DOI: 10.1016/j.bbcan.2020.188351] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer is a multifaceted global disease. Transformation of a normal to a malignant cell takes several steps, including somatic mutations, epigenetic alterations, metabolic reprogramming and loss of cell growth control. Recently, the mevalonate pathway has emerged as a crucial regulator of tumor biology and a potential therapeutic target. This pathway controls cholesterol production and posttranslational modifications of Rho-GTPases, both of which are linked to several key steps of tumor progression. Inhibitors of the mevalonate pathway induce pleiotropic antitumor-effects in several human malignancies, identifying the pathway as an attractive candidate for novel therapies. In this review, we will provide an overview about the role and regulation of the mevalonate pathway in certain aspects of cancer initiation and progression and its potential for therapeutic intervention in oncology.
Collapse
|
50
|
Fichtali K, Bititi A, Elghanmi A, Ghazi B. Serum Lipidomic Profiling in Breast Cancer to Identify Screening, Diagnostic, and Prognostic Biomarkers. Biores Open Access 2020; 9:1-6. [PMID: 32042507 PMCID: PMC6945794 DOI: 10.1089/biores.2018.0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the major mortality cause of women worldwide. In the course of management of breast cancer, the identification of a biomarker is important in enhancing our knowledge on cancer pathology, predicting the response to treatment, and selecting the patients who are more favorable to receive certain treatments. These biomarkers have a prognostic value. In addition to traditional breast cancer prognosis factors such as the tumor size and grade, the axillary lymph node micrometastasis, and biomarkers such as HER2/neu, newly discovered biomarkers have been discovered. Some of these factors are genetic signature in tissue or in peripheral blood. Lipid profil, a simple and accessible biological examination, has been a novel path on the prediction of breast cancer risk of occurrence and recurrence in many studies. The main goal of our review is to evaluate lipid profile and breast cancer risk with an emphasis on the prognosis value of lipid profiles in breast cancer patient management.
Collapse
Affiliation(s)
- Karima Fichtali
- Cheikh Khalifa International Hospital, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Amine Bititi
- Cheikh Khalifa International Hospital, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Adil Elghanmi
- Cheikh Khalifa International Hospital, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Bouchra Ghazi
- National Laboratory of Reference, Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| |
Collapse
|