1
|
Zhang Y, Ding L, Zhang Z, Shen L, Guo Y, Zhang W, Yu Y, Gu Z, Liu J, Kadier A, Geng J, Mao S, Yao X. An Integrated Approach Utilizing Single-Cell and Bulk RNA-Sequencing for the Identification of a Mitophagy-Associated Genes Signature: Implications for Prognostication and Therapeutic Stratification in Prostate Cancer. Biomedicines 2025; 13:311. [PMID: 40002724 PMCID: PMC11853322 DOI: 10.3390/biomedicines13020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 02/27/2025] Open
Abstract
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: We used single-cell and bulk RNA sequencing data from GEO and TCGA databases. We characterized the tumor microenvironment at the single-cell level, analyzing cell interactions and identifying fibroblasts linked to mitophagy. Target genes were narrowed down at the bulk transcriptome level to construct a PARD prognosis prediction nomogram. Unsupervised consensus clustering classified PARD into subtypes, analyzing differences in clinical features, immune infiltration, and immunotherapy. Furthermore, the cellular functions of the genes of interest were verified in vitro. Results: We identified ten cell types and 160 mitophagy-related single-cell differentially expressed genes (MR-scDEGs). Strong interactions were observed between fibroblasts, endothelial cells, CD8+ T cells, and NK cells. Fibroblasts linked to mitophagy were divided into six subtypes. Intersection of DEGs from three bulk datasets with MR-scDEGs identified 26 key genes clustered into two subgroups. COX regression analysis identified seven prognostic key genes, enabling a prognostic nomogram model. High and low-risk groups showed significant differences in clinical features, immune infiltration, immunotherapy, and drug sensitivity. In prostate cancer cell lines, CAV1, PALLD, and ITGB8 are upregulated, while CLDN7 is downregulated. Knockdown of PALLD significantly inhibits the proliferation and colony-forming ability of PC3 and DU145 cells, suggesting the important roles of this gene in prostate cancer progression. Conclusions: This study analyzed mitophagy-related genes in PARD, predicting prognosis and aiding in subtype identification and immunotherapy response analysis. This approach offers new strategies for treating prostate cancer with specific molecular subtypes and helps develop potential biomarkers for personalized medicine strategies.
Collapse
Affiliation(s)
- Yuke Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Li Ding
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhijin Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Liliang Shen
- Department of Urology, Ningbo Yinzhou People’s Hospital, 251 Baizhang East Road, Ningbo 315100, China;
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Yang Yu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhuoran Gu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Aimaitiaji Kadier
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiang Geng
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Middle Yan Chang Road, Shanghai 200072, China; (Y.Z.); (L.D.); (Z.Z.); (Y.G.); (W.Z.); (Y.Y.); (Z.G.); (J.L.); (A.K.); (J.G.)
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
2
|
Kamath AJ, Chandy AS, Joseph AA, Gorantla JN, Donadkar AD, Nath LR, Sharifi-Rad J, Calina D. Embelin: A multifaceted anticancer agent with translational potential in targeting tumor progression and metastasis. EXCLI JOURNAL 2023; 22:1311-1329. [PMID: 38234968 PMCID: PMC10792175 DOI: 10.17179/excli2023-6590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024]
Abstract
Embelin, a natural para-benzoquinone product, is derived from plants of the Embelia genus, particularly Embelia ribes Burm.f. A staple in traditional medicinal formulations for centuries, Embelin's pharmacological actions are attributed to the hydroxyl benzoquinone present in its structure. Its therapeutic potential is bolstered by unique physical and chemical properties. Recently, Embelin, recognized as a non-peptidic, cell-permeable small inhibitor of the X-linked inhibitor of apoptosis protein (XIAP), has garnered significant attention for its anticancer activity. It demonstrates various anticancer mechanisms, such as apoptosis induction, cell cycle arrest, and autophagy, in different cancer types. Additionally, Embelin modulates several signal transduction pathways, including NF-κB, PI3Kinase/AKT, and STAT3, effectively inhibiting the proliferation of diverse cancer cell lines. This literature review illuminates the anticancer potential of Embelin, detailing its mechanisms of action and prospective clinical applications, based on relevant scientific literature from the past decade sourced from various electronic databases. See also the Graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Adithya Jayaprakash Kamath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Alda Sara Chandy
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Aina Ann Joseph
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Jaggiah N. Gorantla
- Department of Chemistry, Wayne State University, Detroit-48202, Michigan, USA
| | - Asawari Dilip Donadkar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Lekshmi R. Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
3
|
Topal J, Panchal N, Barroeta A, Roppelt A, Mudde A, Gaspar HB, Thrasher AJ, Houghton BC, Booth C. Lentiviral Gene Transfer Corrects Immune Abnormalities in XIAP Deficiency. J Clin Immunol 2023; 43:440-451. [PMID: 36329240 PMCID: PMC9892131 DOI: 10.1007/s10875-022-01389-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND X-linked inhibitor of apoptosis protein (XIAP) deficiency is a severe immunodeficiency with clinical features including hemophagocytic lymphohistiocytosis (HLH) and inflammatory bowel disease (IBD) due to defective NOD2 responses. Management includes immunomodulatory therapies and hematopoietic stem cell transplant (HSCT). However, this cohort is particularly susceptible to the chemotherapeutic regimens and acutely affected by graft-vs-host disease (GvHD), driving poor long-term survival in transplanted patients. Autologous HSC gene therapy could offer an alternative treatment option and would abrogate the risks of alloreactivity. METHODS Hematopoietic progenitor (Lin-ve) cells from XIAPy/- mice were transduced with a lentiviral vector encoding human XIAP cDNA before transplantation into irradiated XIAP y/- recipients. After 12 weeks animals were challenged with the dectin-1 ligand curdlan and recovery of innate immune function was evaluated though analysis of inflammatory cytokines, body weight, and splenomegaly. XIAP patient-derived CD14+ monocytes were transduced with the same vector and functional recovery was demonstrated using in vitro L18-MDP/NOD2 assays. RESULTS In treated XIAPy/- mice, ~40% engraftment of gene-corrected Lin-ve cells led to significant recovery of weight loss, splenomegaly, and inflammatory cytokine responses to curdlan, comparable to wild-type mice. Serum IL-6, IL-10, MCP-1, and TNF were significantly reduced 2-h post-curdlan administration in non-corrected XIAPy/- mice compared to wild-type and gene-corrected animals. Appropriate reduction of inflammatory responses was observed in gene-corrected mice, whereas non-corrected mice developed an inflammatory profile 9 days post-curdlan challenge. In gene-corrected patient CD14+ monocytes, TNF responses were restored following NOD2 activation with L18-MDP. CONCLUSION Gene correction of HSCs recovers XIAP-dependent immune defects and could offer a treatment option for patients with XIAP deficiency.
Collapse
Affiliation(s)
- Joseph Topal
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Neelam Panchal
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Amairelys Barroeta
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Anna Roppelt
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Annelotte Mudde
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - H Bobby Gaspar
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
- Orchard Therapeutics, London, UK
| | - Adrian J Thrasher
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Benjamin C Houghton
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire Booth
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK.
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK.
| |
Collapse
|
4
|
Antrodia salmonea Extracts Regulate p53-AR Signaling and Apoptosis in Human Prostate Cancer LNCaP Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7033127. [DOI: 10.1155/2022/7033127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2022]
Abstract
Antrodia salmonea (AS) is a genus of Antrodia, an epiphyte of Cunninghamia konishii in Taiwan. AS has been reported to have potential therapeutic effects on different diseases, including diarrhea, abdominal pain, and hypertension. AS has been reported to have anticancer effects on numerous cancer types, such as ovarian carcinoma and triple-negative breast cancer. Our previous studies demonstrated that antrocins and triterpenoids are possibly bioactive compositions. However, the effects of AS on prostate cancer remain unknown. Therefore, we investigated the role of AS in prostate cancer growth, apoptosis, and cell cycle regulation. The results showed that AS extracts significantly inhibited the proliferation of prostate cancer LNCaP cells in a dose-dependent manner and increased the levels of apoptotic markers (cleaved PARP and cleaved caspase 3/8/9). In addition, the cell cycle-related proteins CDK1, CDK2, CDK4, and their respective specific regulators Cyclin B1, Cyclin A, and Cyclin D were also affected. Besides, AS treatment increased p53 protein levels and slowed its degradation in LNCaP cells. Interestingly, we found that AS treatment reduced both total protein and Ser-81 phosphorylation levels of the androgen receptor (AR). Notably, the increase of nuclear p53 was accompanied by the down-regulation of AR, suggesting a reverse regulation between p53 and AR in LNCaP cells was triggered by AS treatment. These findings suggest that AS extracts trigger the apoptosis of prostate cancer cells through the reverse regulation of p53 and AR and elucidate that AS extracts might be a potential treatment for androgen-dependent prostate cancer in the near future.
Collapse
|
5
|
Embelin and Its Derivatives: Design, Synthesis, and Potential Delivery Systems for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:ph15091131. [PMID: 36145352 PMCID: PMC9505931 DOI: 10.3390/ph15091131] [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: 08/09/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Embelin is a naturally occurring benzoquinone that inhibits the growth of cancer cells, making it a potent anticancer drug. However, the low water solubility of embelin restricts its clinical applicability. This review provides a concise summary and in-depth analysis of the published literature on the design and synthesis of embelin derivatives possessing increased aqueous solubility and superior therapeutic efficacy. In addition, the potential of drug delivery systems to improve the anticancer capabilities of embelin and its derivatives is discussed.
Collapse
|
6
|
Ho HY, Chen PJ, Lo YS, Lin CC, Chuang YC, Hsieh MJ, Chen MK. Luteolin-7-O-glucoside inhibits cell proliferation and modulates apoptosis through the AKT signaling pathway in human nasopharyngeal carcinoma. ENVIRONMENTAL TOXICOLOGY 2021; 36:2013-2024. [PMID: 34165247 DOI: 10.1002/tox.23319] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/19/2021] [Accepted: 06/15/2021] [Indexed: 06/13/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is an unnoticeable malignant tumor with a high potential of lymphatic metastasis, and its prevalence is high in Asia. Ionizing radiation is the mainstay of treatment for patients with NPC without metastasis. However, patients with metastatic lesions require advanced treatments such as chemotherapy. The present study investigated the apoptotic effect of luteolin-7-O-glucoside on NPC cells and elucidated its underlying signaling mechanisms. The results revealed that luteolin-7-O-glucoside significantly reduced the proliferation of NPC cell lines (NPC-039 and NPC-BM). Flow cytometry and morphological analysis results demonstrated that luteolin-7-O-glucoside treatment induced S and G2 /M cell cycle arrest, chromatin condensation, and apoptosis. In addition, mitochondrial membrane potential was observed to be depolarized with an increasing concentration of luteolin-7-O-glucoside. Proteins involved in the extrinsic and intrinsic pathways of apoptosis, such as death receptor, caspase-3, caspase-8, caspase-9, and Bcl-2 family proteins (Bax, t-Bid, Bcl-2, and Bcl-xL), were downregulated and upregulated after treatment with luteolin-7-O-glucoside, respectively. Moreover, the addition of a PI3K/AKT inhibitor enhanced the activation of poly-ADP-ribose-polymerase (PARP) and attenuated cell viability, indicating that luteolin-7-O-glucoside induced apoptosis in NPC cells through the AKT signaling pathway. These results indicated that the apoptosis of NPC cells modulated by luteolin-7-O-glucoside may be preceded by mitochondrial depolarization, cell cycle arrest, extrinsic and intrinsic apoptosis pathway activation, and AKT signaling modulation. Thus, luteolin-7-O-glucoside can be a promising anticancer agent against human NPC.
Collapse
Affiliation(s)
- Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ping-Ju Chen
- Department of Dentistry, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
7
|
Ansari I, Singh P, Mittal A, Mahato RI, Chitkara D. 2,2-Bis(hydroxymethyl) propionic acid based cyclic carbonate monomers and their (co)polymers as advanced materials for biomedical applications. Biomaterials 2021; 275:120953. [PMID: 34218051 DOI: 10.1016/j.biomaterials.2021.120953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Designing grafted biodegradable polymers with tailored multi-functional properties is one of the most researched fields with extensive biomedical applications. Among many biodegradable polymers, polycarbonates have gained much attention due to their ease of synthesis, high drug loading, and excellent biocompatibility profiles. Among various monomers, 2,2-bis(hydroxymethyl) propionic acid (bis-MPA) derived cyclic carbonate monomers have been extensively explored in terms of their synthesis as well as their polymerization. Since the late 90s, significant advancements have been made in the design of bis-MPA derived cyclic carbonate monomers as well as in their reaction schemes. Currently, bis-MPA derived polycarbonates have taken a form of an entire platform with a multitude of applications, the latest being in the field of nanotechnology, targeted drug, and nucleic acid delivery. The present review outlines an up to date developments that have taken place in the last two decades in the design, synthesis, and biomedical applications of bis-MPA derived cyclic carbonates and their (co)polymers.
Collapse
Affiliation(s)
- Imran Ansari
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India.
| |
Collapse
|
8
|
Cohen L, Livney YD, Assaraf YG. Targeted nanomedicine modalities for prostate cancer treatment. Drug Resist Updat 2021; 56:100762. [PMID: 33857756 DOI: 10.1016/j.drup.2021.100762] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
Prostate cancer (PC) is the second most common cause of death amongst men in the USA. Therapy of PC has been transformed in the past decade by introducing novel therapeutics, advanced functional imaging and diagnostic approaches, next generation sequencing, as well as improved application of existing therapies in localized PC. Treatment of PC at the different stages of the disease may include surgery, androgen deprivation therapy (ADT), chemotherapy and radiation therapy. However, although ADT has proven efficacious in PC treatment, its effectiveness may be temporary, as these tumors frequently develop molecular mechanisms of therapy resistance, which allow them to survive and proliferate even under conditions of testosterone deprivation, inhibition of androgen receptor signaling, or cytotoxic drug treatment. Importantly, ADT was found to induce key alterations which frequently result in the formation of metastatic tumors displaying a therapy refractory phenotype. Hence, to overcome these serious therapeutic impediments, novel PC cell-targeted therapeutic strategies are being developed. These include diverse platforms enabling specific enhanced antitumor drug uptake and increased intracellular accumulation. Studies have shown that these novel treatment modalities lead to enhanced antitumor activity and diminished systemic toxicity due to the use of selective targeting and decreased drug doses. The underlying mechanism of targeting and internalization is based upon the interaction between a selective ligand, conjugated to a drug-loaded nanoparticle or directly to an anti-cancer drug, and a specific plasma membrane biomarker, uniquely overexpressed on the surface of PC cells. Another targeted therapeutic approach is the delivery of unique anti-oncogenic signaling pathway-based therapeutic drugs, which are selectively cytotoxic to PC cells. The current paper reviews PC targeted modalities reported in the past 6 years, and discusses both the advantages and limitations of the various targeted treatment strategies.
Collapse
Affiliation(s)
- Lital Cohen
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yoav D Livney
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
9
|
Yu W, Maynard E, Chiaradia V, Arno MC, Dove AP. Aliphatic Polycarbonates from Cyclic Carbonate Monomers and Their Application as Biomaterials. Chem Rev 2021; 121:10865-10907. [DOI: 10.1021/acs.chemrev.0c00883] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Wei Yu
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Edward Maynard
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Viviane Chiaradia
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Maria C. Arno
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| |
Collapse
|
10
|
Khatoon E, Banik K, Harsha C, Sailo BL, Thakur KK, Khwairakpam AD, Vikkurthi R, Devi TB, Gupta SC, Kunnumakkara AB. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin Cancer Biol 2020; 80:306-339. [DOI: 10.1016/j.semcancer.2020.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
|
11
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
12
|
Rice MA, Malhotra SV, Stoyanova T. Second-Generation Antiandrogens: From Discovery to Standard of Care in Castration Resistant Prostate Cancer. Front Oncol 2019; 9:801. [PMID: 31555580 PMCID: PMC6723105 DOI: 10.3389/fonc.2019.00801] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed cancer affecting men in the United States. The prostate is a hormone-dependent gland in which androgen hormones testosterone and dihydrotestosterone bind to and activate the androgen receptor, initiating nuclear translocation of androgen receptor and a subsequent signaling cascade. Due to the androgen dependency of the prostate, androgen deprivation therapies have emerged as first line treatment for aggressive prostate cancer. Such therapies are effective until the point at which prostate cancer, through a variety of mechanisms including but not limited to generation of ligand-independent androgen receptor splice variants, or intratumoral androgen production, overcome hormone deprivation. These cancers are androgen ablation resistant, clinically termed castration resistant prostate cancer (CRPC) and remain incurable. First-generation antiandrogens established androgen receptor blockade as a therapeutic strategy, but these therapies do not completely block androgen receptor activity. Efficacy and potency have been improved by the development of second-generation antiandrogen therapies, which remain the standard of care for patients with CRPC. Four second-generation anti-androgens are currently approved by the Food and Drug Administration (FDA); abiraterone acetate, enzalutamide, and recently approved apalutamide and darolutamide. This review is intended to provide a thorough overview of FDA approved second-generation antiandrogen discovery, treatment application, strategies for combination therapy to overcome resistance, and an insight for the potential future approaches for therapeutic inhibition of androgen receptor.
Collapse
Affiliation(s)
- Meghan A. Rice
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| | - Sanjay V. Malhotra
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, United States
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
13
|
Prabhu KS, Achkar IW, Kuttikrishnan S, Akhtar S, Khan AQ, Siveen KS, Uddin S. Embelin: a benzoquinone possesses therapeutic potential for the treatment of human cancer. Future Med Chem 2018; 10:961-976. [PMID: 29620447 DOI: 10.4155/fmc-2017-0198] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/18/2017] [Indexed: 08/28/2023] Open
Abstract
Natural products have been gaining recognition and are becoming a significant part of research in the area of drug development and discovery. Phytochemicals derived from these sources have been comprehensively studied and have displayed a wide range of activities against many fatal diseases including cancer. One such product that has gained recognition from its pharmacological properties and nontoxic nature is embelin, obtained from Embelia ribes. Amid all the vivid pharmacological activities, embelin has gained its prominence in the area of cancer research. Embelin binds to the BIR3 domain of XIAP, preventing the association of XIAP and caspase-9 resulting in the suppression of cell growth, proliferation and migration of various types of cancer cells. Furthermore, embelin modulates anti-apoptotic pathways by suppressing the activity of NF-κB, PI3-kinase/AKT, JAK/STAT pathway - among others. The present review summarizes the various reported effects of embelin on different types of cancer cells and highlights the cellular mechanisms of action.
Collapse
Affiliation(s)
- Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Iman W Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kodapully S Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
14
|
Ko JH, Lee SG, Yang WM, Um JY, Sethi G, Mishra S, Shanmugam MK, Ahn KS. The Application of Embelin for Cancer Prevention and Therapy. Molecules 2018. [PMID: 29522451 PMCID: PMC6017120 DOI: 10.3390/molecules23030621] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Embelin is a naturally-occurring benzoquinone compound that has been shown to possess many biological properties relevant to human cancer prevention and treatment, and increasing evidence indicates that embelin may modulate various characteristic hallmarks of tumor cells. This review summarizes the information related to the various oncogenic pathways that mediate embelin-induced cell death in multiple cancer cells. The mechanisms of the action of embelin are numerous, and most of them induce apoptotic cell death that may be intrinsic or extrinsic, and modulate the NF-κB, p53, PI3K/AKT, and STAT3 signaling pathways. Embelin also induces autophagy in cancer cells; however, these autophagic cell-death mechanisms of embelin have been less reported than the apoptotic ones. Recently, several autophagy-inducing agents have been used in the treatment of different human cancers, although they require further exploration before being transferred from the bench to the clinic. Therefore, embelin could be used as a potential agent for cancer therapy.
Collapse
Affiliation(s)
- Jeong-Hyeon Ko
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Seok-Geun Lee
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Woong Mo Yang
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Srishti Mishra
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
15
|
Yang WZ, Zhou H, Yan Y. XIAP underlies apoptosis resistance of renal cell carcinoma cells. Mol Med Rep 2017; 17:125-130. [PMID: 29115633 PMCID: PMC5780075 DOI: 10.3892/mmr.2017.7925] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 09/06/2017] [Indexed: 01/28/2023] Open
Abstract
X-linked inhibitor of apoptosis (XIAP), a key member of the inhibitors of apoptosis protein family, can inhibit apoptosis by directly binding to the initiator caspase-9, −3 and −7, thereby promoting tumor cell survival during tumor progression. In the present study, XIAP basal expression levels were investigated and its contribution to the resistance to apoptosis was evaluated, in the RCC cell lines exposed to apoptosis-inducing drugs. This was investigated by histological methods and western blot analysis. Using RNA interference, elimination of XIAP in Caki-1 cells was also studied, and its contribution to the sensitivity to apoptosis induced through the intrinsic pathway was observed. Differences in XIAP expression were detected between ClearCa-2 and ClearCa-6 cell lines. ClearCa-6 cells with lower expression of XIAP were more sensitive to apoptosis-inducing drugs, compared with ClearCa-2 cells. However, the levels of XIAP expression in both cell lines were stable during apoptosis. Furthermore, a Caki-1 cell line with no XIAP expression was used, and was demonstrated to be more sensitive to the apoptosis induced by the mitochondrial pathway. These results suggested that downregulation of XIAP expression could enhance the sensitivity of RCC cells to apoptosis, and the basal expression of XIAP during apoptosis is stable. This may provide novel insight for targeted gene therapy against XIAP, in the clinic.
Collapse
Affiliation(s)
- Wen Zheng Yang
- Department of Anesthesiology, Beijing Shijitan Hospital, Beijing 100038, P.R. China
| | - Haijiang Zhou
- Department of Emergency Medicine, Beijing Chao‑Yang Hospital, Beijing 100038, P.R. China
| | - Yong Yan
- Department of Urology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| |
Collapse
|
16
|
Graça I, Pereira-Silva E, Henrique R, Packham G, Crabb SJ, Jerónimo C. Epigenetic modulators as therapeutic targets in prostate cancer. Clin Epigenetics 2016; 8:98. [PMID: 27651838 PMCID: PMC5025578 DOI: 10.1186/s13148-016-0264-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/07/2016] [Indexed: 01/24/2023] Open
Abstract
Prostate cancer is one of the most common non-cutaneous malignancies among men worldwide. Epigenetic aberrations, including changes in DNA methylation patterns and/or histone modifications, are key drivers of prostate carcinogenesis. These epigenetic defects might be due to deregulated function and/or expression of the epigenetic machinery, affecting the expression of several important genes. Remarkably, epigenetic modifications are reversible and numerous compounds that target the epigenetic enzymes and regulatory proteins were reported to be effective in cancer growth control. In fact, some of these drugs are already being tested in clinical trials. This review discusses the most important epigenetic alterations in prostate cancer, highlighting the role of epigenetic modulating compounds in pre-clinical and clinical trials as potential therapeutic agents for prostate cancer management.
Collapse
Affiliation(s)
- Inês Graça
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; School of Allied Health Sciences (ESTSP), Polytechnic of Porto, Porto, Portugal
| | - Eva Pereira-Silva
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Simon J Crabb
- Cancer Research UK Centre, Cancer Sciences, The Somers Cancer Research Building, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, S016 6YD UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Research Center-LAB 3, F Bdg, 1st floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal ; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
17
|
Shah P, Djisam R, Damulira H, Aganze A, Danquah M. Embelin inhibits proliferation, induces apoptosis and alters gene expression profiles in breast cancer cells. Pharmacol Rep 2016; 68:638-44. [PMID: 27031050 DOI: 10.1016/j.pharep.2016.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 01/21/2023]
Abstract
PURPOSE To investigate effect of embelin on proliferation, apoptosis and gene expression profile changes in breast cancer cells. METHODS Cell viability was determined by MTT assay and apoptosis assayed using flow cytometry. Differential expression of 84 genes commonly involved in breast cancer carcinogenesis was assessed by real-time PCR using the Human Breast Cancer RT(2) Profiler PCR Array. RESULTS MCF-7 and MDA-MB-231 cells were treated with embelin (0-25μM) for 24 and 96h. Embelin exhibited time and dose dependence in both cell lines and was more potent in inhibiting MDA-MB-231 cell proliferation compared to MCF-7 cells. IC50 for embelin in MDA-MB-231 cells was ∼4.45μM and 3.28μM at 24h and 96h, respectively. In contrast, IC50 for embelin in MCF-7 cells was ∼6.04μM and 4.51μM at 24h and 96h, respectively. Embelin (50μM) induced apoptosis and activated caspase 3 activity in both cell lines when exposed for 72h. Treatment of MDA-MB-231 cells with embelin (10μM) for 24h resulted in significant differential expression of 27 genes commonly involved in breast cancer carcinogenesis. CONCLUSIONS Our findings show that embelin inhibits cell proliferation, induces apoptosis and alters expression of breast cancer focused genes in MCF-7 and MDA-MB-231 cells. Based on RT(2)-PCR array analysis, embelin down-regulated expression of pivotal oncogenes. This knowledge could be beneficial in the development of effective embelin-based therapies for treating breast cancer.
Collapse
Affiliation(s)
- Priyank Shah
- Department of Pharmaceutical Sciences, Chicago State University, Chicago, USA
| | - Ransford Djisam
- Department of Pharmaceutical Sciences, Chicago State University, Chicago, USA
| | - Hamidah Damulira
- Department of Pharmaceutical Sciences, Chicago State University, Chicago, USA
| | - Alice Aganze
- Department of Pharmaceutical Sciences, Chicago State University, Chicago, USA
| | - Michael Danquah
- Department of Pharmaceutical Sciences, Chicago State University, Chicago, USA.
| |
Collapse
|
18
|
Lu H, Wang J, Wang Y, Qiao L, Zhou Y. Embelin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016:397-418. [DOI: 10.1007/978-3-319-41334-1_16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
19
|
Brooke GN, Gamble SC, Hough MA, Begum S, Dart DA, Odontiadis M, Powell SM, Fioretti FM, Bryan RA, Waxman J, Wait R, Bevan CL. Antiandrogens act as selective androgen receptor modulators at the proteome level in prostate cancer cells. Mol Cell Proteomics 2015; 14:1201-16. [PMID: 25693800 PMCID: PMC4424393 DOI: 10.1074/mcp.m113.036764] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Indexed: 11/06/2022] Open
Abstract
Current therapies for prostate cancer include antiandrogens, inhibitory ligands of the androgen receptor, which repress androgen-stimulated growth. These include the selective androgen receptor modulators cyproterone acetate and hydroxyflutamide and the complete antagonist bicalutamide. Their activity is partly dictated by the presence of androgen receptor mutations, which are commonly detected in patients who relapse while receiving antiandrogens, i.e. in castrate-resistant prostate cancer. To characterize the early proteomic response to these antiandrogens we used the LNCaP prostate cancer cell line, which harbors the androgen receptor mutation most commonly detected in castrate-resistant tumors (T877A), analyzing alterations in the proteome, and comparing these to the effect of these therapeutics upon androgen receptor activity and cell proliferation. The majority are regulated post-transcriptionally, possibly via nongenomic androgen receptor signaling. Differences detected between the exposure groups demonstrate subtle changes in the biological response to each specific ligand, suggesting a spectrum of agonistic and antagonistic effects dependent on the ligand used. Analysis of the crystal structures of the AR in the presence of cyproterone acetate, hydroxyflutamide, and DHT identified important differences in the orientation of key residues located in the AF-2 and BF-3 protein interaction surfaces. This further implies that although there is commonality in the growth responses between androgens and those antiandrogens that stimulate growth in the presence of a mutation, there may also be influential differences in the growth pathways stimulated by the different ligands. This therefore has implications for prostate cancer treatment because tumors may respond differently dependent upon which mutation is present and which ligand is activating growth, also for the design of selective androgen receptor modulators, which aim to elicit differential proteomic responses dependent upon cellular context.
Collapse
Affiliation(s)
- Greg N Brooke
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK; §Molecular Oncology, School of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Simon C Gamble
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK
| | - Michael A Hough
- §Molecular Oncology, School of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Shajna Begum
- ¶Kennedy Institute of Rheumatology, Imperial College London, London W6 8LH, UK
| | - D Alwyn Dart
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK; ‖Cardiff University Peking University Cancer Institute, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Michael Odontiadis
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK
| | - Sue M Powell
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK
| | - Flavia M Fioretti
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK
| | - Rosie A Bryan
- §Molecular Oncology, School of Biological Sciences, University of Essex, Colchester CO4 3SQ, UK
| | - Jonathan Waxman
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK
| | - Robin Wait
- ¶Kennedy Institute of Rheumatology, Imperial College London, London W6 8LH, UK
| | - Charlotte L Bevan
- From the ‡Androgen Signalling Laboratory, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
20
|
Hu R, Li J, Liu Z, Miao M, Yao K. GDC-0152 induces apoptosis through down-regulation of IAPs in human leukemia cells and inhibition of PI3K/Akt signaling pathway. Tumour Biol 2014; 36:577-84. [PMID: 25273171 DOI: 10.1007/s13277-014-2648-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/18/2014] [Indexed: 11/29/2022] Open
Abstract
The inhibitor of apoptosis proteins (IAPs) is closely related to leukemia apoptosis. The present study was undertaken to determine the molecular mechanisms by which GDC-0152, an IAP inhibitor, induces apoptosis in human leukemia cells (K562 and HL60 cells). GDC-0152 inhibited the proliferation of K562 and HL60 cells in a dose- and time-dependent manner, which was largely attributed to intrinsic apoptosis. GDC-0152 down-regulated the IAPs including X-linked inhibitor of apoptosis protein (XIAP), cellular inhibitor of apoptosis protein-1 (cIAP1), and cellular inhibitor of apoptosis protein-2 (cIAP2) expression and induced the activation of caspase-9 and caspase-3. GDC-0152-induced cell proliferation inhibition in K562 cells was prevented by pan-caspase inhibitor. GDC-0152 also inhibited PI3K and Akt expression in K562 and HL60 cells. Taken together, these findings suggest that GDC-0152 results in human leukemia apoptosis through caspase-dependent mechanisms involving down-regulation of IAPs and inhibition of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Rong Hu
- Department of Hematology, Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | | | | | | | | |
Collapse
|
21
|
Poojari RJ. Embelin, a small molecule quinone with a co-clinical power for castrate-resistant prostate cancer. Front Pharmacol 2014; 5:184. [PMID: 25152733 PMCID: PMC4126039 DOI: 10.3389/fphar.2014.00184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/21/2014] [Indexed: 12/02/2022] Open
Affiliation(s)
- Radhika J Poojari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay Mumbai, India
| |
Collapse
|
22
|
Wen D, Chitkara D, Wu H, Danquah M, Patil R, Miller DD, Mahato RI. LHRH-Conjugated Micelles for Targeted Delivery of Antiandrogen to Treat Advanced Prostate Cancer. Pharm Res 2014; 31:2784-95. [DOI: 10.1007/s11095-014-1375-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/21/2014] [Indexed: 01/08/2023]
|
23
|
Abstract
Clinical application of anticancer drugs is limited by problems such as low water solubility, lack of tissue-specificity and toxicity. Formulation development represents an important approach to these problems. Among the many delivery systems studied, polymeric micelles have gained considerable attention owing to ease in preparation, small sizes (10-100 nm), and ability to solubilize water-insoluble anticancer drugs and accumulate specifically at the tumors. This article provides a brief review of several promising micellar systems and their applications in tumor therapy. The emphasis is placed on the discussion of the authors' recent work on several nanomicellar systems that have both a delivery function and antitumor activity, named dual-function drug carriers.
Collapse
|
24
|
Wang H, Li F, Du C, Wang H, Mahato RI, Huang Y. Doxorubicin and Lapatinib Combination Nanomedicine for Treating Resistant Breast Cancer. Mol Pharm 2014; 11:2600-11. [DOI: 10.1021/mp400687w] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Huiyuan Wang
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke
Rd, Shanghai 201203, China
| | - Feng Li
- Department
of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, Virginia 23668, United States
| | - Chengan Du
- Department
of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, Virginia 23668, United States
| | - Huixin Wang
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke
Rd, Shanghai 201203, China
| | - Ram I. Mahato
- Department
of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yongzhuo Huang
- Shanghai
Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke
Rd, Shanghai 201203, China
| |
Collapse
|
25
|
Poojari R. Embelin – a drug of antiquity: shifting the paradigm towards modern medicine. Expert Opin Investig Drugs 2014; 23:427-44. [DOI: 10.1517/13543784.2014.867016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
26
|
Gryder BE, Akbashev MJ, Rood MK, Raftery ED, Meyers WM, Dillard P, Khan S, Oyelere AK. Selectively targeting prostate cancer with antiandrogen equipped histone deacetylase inhibitors. ACS Chem Biol 2013; 8:2550-60. [PMID: 24004176 DOI: 10.1021/cb400542w] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Diverse cellular processes relevant to cancer progression are regulated by the acetylation status of proteins. Among such processes is chromatin remodeling via histone proteins, controlled by opposing histone deacetylase (HDAC) and histone acetyltransferase (HAT) enzymes. Histone deacetylase inhibitors (HDACi) show great promise in preclinical cancer models, but clinical trials treating solid tumors have failed to improve patient survival. This is due in part to an inability of HDACi to effectively accumulate in cancerous cells. To address this problem we designed HDACi with secondary pharmacophores to facilitate selective accumulation in malignant cells. We present the first example of HDACi compounds targeted to prostate tumors by equipping them with the additional ability to bind the androgen receptor (AR) with nonsteroidal antiandrogen moieties. Leads among these new dual-acting molecules bind to the AR and halt AR transcriptional activity at lower concentrations than clinical antiandrogens. They inhibit key isoforms of HDAC with low nanomolar potency. Fluorescent microscopy reveals varying degrees of AR nuclear localization in response to these compounds that correlates with their HDAC activity. These biological properties translate into potent anticancer activity against hormone-dependent (AR+) LNCaP and to a lesser extent against hormone-independent (AR-) DU145 prostate cancer, while having greatly reduced toxicity in noncancerous cells. This illustrates that engaging multiple biological targets with a single chemical probe can achieve both potent and cell-type-selective responses.
Collapse
Affiliation(s)
- Berkley E. Gryder
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Michelle J. Akbashev
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Michael K. Rood
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Eric D. Raftery
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| | - Warren M. Meyers
- Department of Cellular
and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Paulette Dillard
- Center
for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Shafiq Khan
- Center
for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Adegboyega K. Oyelere
- Parker H. Petit Institute for Bioengineering & Biosciences, Department of Chemistry and Biochemistry, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332-0230, United States
| |
Collapse
|
27
|
Lunardi A, Ala U, Epping MT, Salmena L, Clohessy JG, Webster KA, Wang G, Mazzucchelli R, Bianconi M, Stack EC, Lis R, Patnaik A, Cantley LC, Bubley G, Cordon-Cardo C, Gerald WL, Montironi R, Signoretti S, Loda M, Nardella C, Pandolfi PP. A co-clinical approach identifies mechanisms and potential therapies for androgen deprivation resistance in prostate cancer. Nat Genet 2013; 45:747-55. [PMID: 23727860 PMCID: PMC3787876 DOI: 10.1038/ng.2650] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/01/2013] [Indexed: 12/14/2022]
Abstract
Here we report an integrated analysis that leverages data from treatment of genetic mouse models of prostate cancer along with clinical data from patients to elucidate new mechanisms of castration resistance. We show that castration counteracts tumor progression in a Pten-loss driven mouse model of prostate cancer through the induction of apoptosis and proliferation block. Conversely, this response is bypassed upon deletion of either Trp53 or Lrf together with Pten, leading to the development of castration resistant prostate cancer (CRPC). Mechanistically, the integrated acquisition of data from mouse models and patients identifies the expression patterns of XAF1-XIAP/SRD5A1 as a predictive and actionable signature for CRPC. Importantly, we show that combined inhibition of XIAP, SRD5A1, and AR pathways overcomes castration resistance. Thus, our co-clinical approach facilitates stratification of patients and the development of tailored and innovative therapeutic treatments.
Collapse
Affiliation(s)
- Andrea Lunardi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lu J, Huang Y, Zhao W, Marquez RT, Meng X, Li J, Gao X, Venkataramanan R, Wang Z, Li S. PEG-derivatized embelin as a nanomicellar carrier for delivery of paclitaxel to breast and prostate cancers. Biomaterials 2012. [PMID: 23182923 DOI: 10.1016/j.biomaterials.2012.10.073] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Paclitaxel (PTX) is one of the most effective chemotherapeutic agents for a wide spectrum of cancers, but its therapeutic benefit is often limited by severe side effects. We have developed a micelle-based PTX formulation based on a simple conjugate derived from polyethylene glycol 5000 (PEG(5K)) and embelin (EB). Embelin is a natural product and exhibits antitumor activity through blocking the activity of X-linked inhibitor of apoptosis protein (XIAP). PEG(5K)-EB₂ conjugate self-assembles to form stable micelles in aqueous solution and efficiently encapsulates hydrophobic drugs such as PTX. PEG(5K)-EB₂ micelles have a relatively low CMC of 0.002 mg/mL (0.35 μM) with sizes in the range of 20 ∼ 30 nm with or without loaded PTX. In vitro cell uptake study showed that the PEG(5K)-EB₂ micelles were efficiently taken up by tumor cells. In vitro release study showed that PTX formulated in PEG(5K)-EB₂ micelles was slowly released over 5 days with much slower release kinetics than that of Taxol formulation. PTX formulated in PEG(5K)-EB₂ micelles exhibited more potent cytotoxicity than Taxol in several cultured tumor cell lines. Total body near infrared fluorescence (NIRF) imaging showed that PEG(5K)-EB₂ micelles were selectively accumulated at tumor site with minimal uptake in major organs including liver and spleen. PTX-loaded PEG(5K)-EB₂ micelles demonstrated an excellent safety profile with a maximum tolerated dose (MTD) of 100-120 mg PTX/kg in mice, which was significantly higher than that for Taxol (15-20 mg PTX/kg). Finally, PTX formulated in PEG(5K)-EB₂ micelles showed superior antitumor activity compared to Taxol in murine models of breast and prostate cancers.
Collapse
Affiliation(s)
- Jianqin Lu
- Center for Pharmacogenetics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Danquah M, Fujiwara T, Mahato RI. Lactic acid- and carbonate-based crosslinked polymeric micelles for drug delivery. ACTA ACUST UNITED AC 2012. [DOI: 10.1002/pola.26392] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|