1
|
Ichikawa K, Johnson HM, Curtis MA, Biswas N, Singh S, Khachatryan HN, Gater AE, Lin SX, Sperry J. Targeting glioma with heteroaromatic alkaloids: A review of potential therapeutics. Bioorg Med Chem 2025; 121:118051. [PMID: 39999647 DOI: 10.1016/j.bmc.2024.118051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 02/27/2025]
Abstract
Glioblastoma multiforme (GBM), classified as a grade IV astrocytoma, is the most aggressive and deadly form of glioma, characterized by rapid progression, extensive genetic heterogeneity, and resistance to conventional therapies. Despite advancements in surgical techniques, radiation therapy, and the frontline chemotherapeutic agent temozolomide, the prognosis for GBM patients remains poor, with a median survival of 15 months and a 5-year survival rate of approximately 7 %. The absence of effective long-term treatments underscores the urgent, unmet clinical need for novel therapeutic strategies to improve patient outcomes. Natural products, particularly alkaloids, have garnered attention as a rich source of bioactive compounds with diverse pharmacological properties. Alkaloids, a structurally diverse group of natural products, are renowned for their chemotherapeutic properties and ability to cross the blood-brain barrier (BBB), making them promising candidates for glioma therapy. This review systematically examines all reported heteroaromatic alkaloids with documented anti-glioma activities, highlighting their mechanisms of action where available. By providing a comprehensive resource, it aims to facilitate the identification and optimisation of alkaloid-based compounds for glioma-targeted drug discovery. Additionally, this review emphasizes the importance of incorporating natural products into the drug development pipeline to address the pressing challenges associated with glioma, particularly GBM treatment.
Collapse
Affiliation(s)
- Karen Ichikawa
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand; Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Hannah M Johnson
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand; Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nandita Biswas
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | - Hasmik N Khachatryan
- Scientific Technological Centre of Organic and Pharmaceutical Chemistry, National Academy of Science of Armenia, Yerevan 0014, Armenia
| | - Anastasia E Gater
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Simon X Lin
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Jonathan Sperry
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand.
| |
Collapse
|
2
|
Han J, Yang S, Byun WS, Jang SC, Bae ES, Park HG, Lee SK. Antitumor activity of selenopsammaplin A analog (SPA-10091-HCl) via histone methyltransferase DOT1L degradation and apoptosis induction in castration-resistant prostate cancer cells. Eur J Pharm Sci 2025; 205:106991. [PMID: 39689820 DOI: 10.1016/j.ejps.2024.106991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/19/2024]
Abstract
Castrate-resistant prostate cancer (CRPC) is one of the most difficult cancers in men and is characterized by a poor prognosis and a high risk of metastasis. The overexpression of the disruptor of telomeric silencing 1-like (DOT1L), which is a specific methyltransferase for histone H3 at lysine residue 79 (H3K79), has been related to poor outcomes in patients with CRPC. Therefore, targeting DOT1L is considered a potential therapeutic approach to overcome the significant medical challenges of CRPC. In our previous study, we designed selenopsammaplin A (SPA) analogs as non-nucleoside DOT1L inhibitors to suppress human breast cancer cell proliferation and metastasis. However, the antitumor activity and the precise underlying mechanism of SPA analogs in PC cells remain unclear. Herein, we administered SPA-10091-HCl, a DOT1L-targeting degrader, to effectively hinder the growth and DOT1L-mediated H3K79 methylation in CRPC (PC3 and DU145) cells. Mechanistically, SPA-10091-HCl selectively degrades DOT1L protein through the nuclear ubiquitin-proteasome pathway, thereby suppressing H3K79 methylation in CRPC cells. SPA-10091-HCl inhibits CRPC cell proliferation, migration, and invasion, with the E-cadherin expression upregulation and N-cadherin and vimentin expression downregulation. Additionally, prolonged SPA-10091-HCl treatment induced apoptosis by regulating apoptosis-associated protein expressions, including Poly (ADP-ribose) polymerase (PARP), caspase-3, caspase-9, and Bcl-2. Moreover, SPA-10091-HCl effectively inhibited tumor growth in the PC3 cells-implanted xenograft mouse model without any overt toxicity. These results indicate SPA-10091-HCl as a potential candidate for further development as a chemotherapeutic agent against CRPC.
Collapse
Affiliation(s)
- Jaeho Han
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehun Yang
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Chul Jang
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeung-Geun Park
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
3
|
Ramli I, Cheriet T, Thuan DTB, Khoi DN, Thu DNK, Posadino AM, Fenu G, Sharifi-Rad J, Pintus G. Potential applications of antofine and its synthetic derivatives in cancer therapy: structural and molecular insights. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8231-8258. [PMID: 38842561 DOI: 10.1007/s00210-024-03180-x] [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: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Cancer is a major global health challenge, being the second leading cause of morbidity and mortality after cardiovascular disease. The growing economic burden and profound psychosocial impact on patients and their families make it urgent to find innovative and effective anticancer solutions. For this reason, interest in using natural compounds to develop new cancer treatments has grown. In this respect, antofine, an alkaloid class found in Apocynaceae, Lauraceae, and Moraceae family plants, exhibits promising biological properties, including anti-inflammatory, anticancer, antiviral, and antifungal activities. Several molecular mechanisms have been identified underlying antofine anti-cancerous effects, including the inhibition of nuclear factor κB (NF-κB) and AKT/mTOR signaling pathways, epigenetic inhibition of protein synthesis, ribosomal targeting, induction of apoptosis, inhibition of DNA synthesis, and cell cycle arrest. This study discusses the molecular structure, sources, photochemistry, and anticancer properties of antofine in relation to its structure-activity relationship and molecular targets. Then, examine in vitro and in vivo studies and analyze the mechanisms of action underpinning antofine efficacy against cancer cells. This review also discusses multidrug resistance in human cancer and the potential of antofine in this context. Safety and toxicity concerns are also addressed as well as current challenges in antofine research, including the need for clinical trials and bioavailability optimization. This review aims to provide comprehensive information for more effective natural compound-based cancer treatments.
Collapse
Affiliation(s)
- Iman Ramli
- Laboratory of Applied Biochemistry, Faculté Des Sciences de La Nature Et de La Vie, Université Frères Mentouri Constantine 1, 25000, Constantine, Algeria
| | - Thamere Cheriet
- Unité de Valorisation Des Ressources Naturelles, Molécules Bioactives Et Analyses Physicochimiques Et Biologiques, Université Des Frères Mentouri Constantine, 25000, Constantine, Algeria
- Département Science de La Nature Et de La Vie, Faculté Des Sciences Exactes Et Science de La Natute Et de La Vie, Université Mohammed Boudiaf-Oum El-Bouaghi, 04000, Oum El-Bouaghi, Algeria
| | | | - Dang Ngoc Khoi
- College of Health Sciences, VinUniversity, Gia Lam, 100000, Hanoi, Vietnam
| | | | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy
| | - Grazia Fenu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy.
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
4
|
Kausar MA, Parveen S, Anwar S, Sadaf, Massey S, El-Horany HES, Khan FH, Shahein M, Husain SA. Cytotoxic potential and metabolomic profiling of alkaloid rich fraction of Tylophora indica leaves. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159505. [PMID: 38729236 DOI: 10.1016/j.bbalip.2024.159505] [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: 10/06/2023] [Revised: 03/28/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Tylophora indica (Burm f.) Merrill, belong to family Asclepiadaceae, is considered to be a natural remedy with high medicinal benefits. The objective of this work is to assess the metabolomic profile of T. indica leaves enriched in alkaloids, as well as to evaluate the in vitro cytotoxicity of these leaves using the MTT assay on human breast MCF-7 and liver HepG2 cancer cell lines. Dried leaves of T. indica were extracted by sonication, using methanol containing 2 % (v/v) of acetic acid and obtained fraction was characterized by HPTLC and UPLC-MS. The UPLC-MS study yielded a preliminary identification of 32 metabolites, with tylophorine, tylophorine B, tylophorinine, and tylophorinidine being the predominant metabolites. The cytotoxicity of the extract of T. indica was evaluated on HepG2 and MCF-7 cell lines, yielding inhibitory concentration (IC50) values of 75.71 μg/mL and 69.60 μg/mL, respectively. Data suggested that the phytochemical screening clearly showed presence of numerous secondary metabolites with moderate cytotoxic efficacy. In conclusion, the future prospects of T. indica appear promising for the advancement of phytopharmaceutical-based anticancer medications, as well as for the design of contemporary pharmaceuticals in the field of cancer chemotherapy.
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha'il, Hail 81411, Saudi Arabia; Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia.
| | - Shabana Parveen
- Department of Biosciences, Jamia Millia Islamia, Okhla, New Delhi 110025, India.
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha'il, Hail 81411, Saudi Arabia; Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia.
| | - Sadaf
- Department of Biotechnology, Jamia Millia Islamia, Okhla, New Delhi 110025, India
| | - Sheersh Massey
- Department of Biosciences, Jamia Millia Islamia, Okhla, New Delhi 110025, India.
| | - Hemat El-Sayed El-Horany
- Department of Biochemistry, College of Medicine, University of Ha'il, Hail 81411, Saudi Arabia; Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia; Medical Biochemistry Department, Faculty of Medicine, Tanta University, Egypt.
| | - Farida Habib Khan
- Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia; Department of Community and Family Medicine, College of Medicine, University of Ha'il, Hail 81411, Saudi Arabia.
| | - Mona Shahein
- Department of Pediatrics, College of Medicine, University of Ha'il, Hail 81411, Saudi Arabia
| | - Syed Akhtar Husain
- Department of Biosciences, Jamia Millia Islamia, Okhla, New Delhi 110025, India.
| |
Collapse
|
5
|
Gross F, Mancini A, Breton B, Kobayashi H, Pereira PHS, Le Gouill C, Bouvier M, Schann S, Leroy X, Sabbagh L. EGFR signaling and pharmacology in oncology revealed with innovative BRET-based biosensors. Commun Biol 2024; 7:250. [PMID: 38429428 PMCID: PMC10907714 DOI: 10.1038/s42003-024-05965-5] [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: 06/05/2023] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
Mutations of receptor tyrosine kinases (RTKs) are associated with the development of many cancers by modifying receptor signaling and contributing to drug resistance in clinical settings. We present enhanced bystander bioluminescence resonance energy transfer-based biosensors providing new insights into RTK biology and pharmacology critical for the development of more effective RTK-targeting drugs. Distinct SH2-specific effector biosensors allow for real-time and spatiotemporal monitoring of signal transduction pathways engaged upon RTK activation. Using EGFR as a model, we demonstrate the capacity of these biosensors to differentiate unique signaling signatures, with EGF and Epiregulin ligands displaying differences in efficacy, potency, and responses within different cellular compartments. We further demonstrate that EGFR single point mutations found in Glioblastoma or non-small cell lung cancer, impact the constitutive activity of EGFR and response to tyrosine kinase inhibitor. The BRET-based biosensors are compatible with microscopy, and more importantly characterize the next generation of therapeutics directed against RTKs.
Collapse
Affiliation(s)
- Florence Gross
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada
| | - Arturo Mancini
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada
| | - Billy Breton
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Hiroyuki Kobayashi
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Pedro Henrique Scarpelli Pereira
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, and Department of Biochemistry and Molecular Medicine, University of Montreal, 2950 Chemin de Polytechnique, Montreal, Quebec, H3T 1J4, Canada
| | - Stephan Schann
- Domain Therapeutics SA, 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg-Illkirch, France
| | - Xavier Leroy
- Domain Therapeutics SA, 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg-Illkirch, France
| | - Laurent Sabbagh
- Domain Therapeutics North America Inc., 7171 Frederick-Banting, Saint-Laurent, Quebec, H4S 1Z9, Canada.
| |
Collapse
|
6
|
St Pierre M, Kempthorne CJ, Liscombe DK, McNulty J. Rapid entry to phenanthroindolizidine alkaloids via an acid-catalysed acyliminium ion-electrocyclization cascade. Org Biomol Chem 2023; 21:8075-8078. [PMID: 37665044 DOI: 10.1039/d3ob01359f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
A rapid total synthesis of seco-phenanthroindolizidine alkaloids was achieved involving a one-pot acid catalyzed deprotection- condensation-electrocyclization strategy. This synthetic route provided a concise synthesis of (±)-seco-antofine and (±)-septicine in only 4 steps with an overall yield of 22% and 17%, respectively.
Collapse
Affiliation(s)
- Max St Pierre
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada.
| | - Christine J Kempthorne
- Vineland Research and Innovation Centre, 4890 Victoria Ave North, Box 4000, Vineland Station, Ontario L0R 2E0, Canada
| | - David K Liscombe
- Vineland Research and Innovation Centre, 4890 Victoria Ave North, Box 4000, Vineland Station, Ontario L0R 2E0, Canada
| | - James McNulty
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4M1, Canada.
| |
Collapse
|
7
|
Periplocin exerts antitumor activity by regulating Nrf2-mediated signaling pathway in gemcitabine-resistant pancreatic cancer cells. Biomed Pharmacother 2023; 157:114039. [PMID: 36423542 DOI: 10.1016/j.biopha.2022.114039] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022] Open
Abstract
Although gemcitabine-based chemotherapy is common and effective for pancreatic cancer (PC), acquired drug resistance is one of the major reasons for treatment failure. Therefore, a novel therapeutic approach for gemcitabine-resistant PC is required. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an oxidative stress-responsive transcription factor regulating antioxidant responses and plays a crucial role in chemoresistance. In the present study, the antitumor activity of periplocin, a natural cardiac glycoside, was evaluated in an established gemcitabine-resistant PC cell line (PANC-GR). Nrf2 was overexpressed in gemcitabine-resistant cells, and Nrf2 knockdown recovered gemcitabine sensitivity in PANC-GR cells. The antiproliferative activity of periplocin was highly associated with Nrf2 downregulation and Nrf2-mediated signaling pathways in PANC-GR cells. Periplocin also increased reactive oxygen species production inducing G0/G1 cell cycle arrest and apoptosis in PANC-GR cells. Periplocin and gemcitabine combined significantly inhibited tumor growth in a PANC-GR cells-implanted xenograft mouse model via Nrf2 downregulation. Overall, these findings suggest that periplocin might be a novel therapeutic agent against gemcitabine resistance, as it could recover sensitivity to gemcitabine by regulating Nrf2-mediated signaling pathways in gemcitabine-resistant PC cells.
Collapse
|
8
|
Bajalia EM, Azzouz FB, Chism DA, Giansiracusa DM, Wong CG, Plaskett KN, Bishayee A. Phytochemicals for the Prevention and Treatment of Renal Cell Carcinoma: Preclinical and Clinical Evidence and Molecular Mechanisms. Cancers (Basel) 2022; 14:3278. [PMID: 35805049 PMCID: PMC9265746 DOI: 10.3390/cancers14133278] [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: 05/23/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Renal cell carcinoma (RCC) is associated with about 90% of renal malignancies, and its incidence is increasing globally. Plant-derived compounds have gained significant attention in the scientific community for their preventative and therapeutic effects on cancer. To evaluate the anticancer potential of phytocompounds for RCC, we compiled a comprehensive and systematic review of the available literature. Our work was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses criteria. The literature search was performed using scholarly databases such as PubMed, Scopus, and ScienceDirect and keywords such as renal cell carcinoma, phytochemicals, cancer, tumor, proliferation, apoptosis, prevention, treatment, in vitro, in vivo, and clinical studies. Based on in vitro results, various phytochemicals, such as phenolics, terpenoids, alkaloids, and sulfur-containing compounds, suppressed cell viability, proliferation and growth, showed cytotoxic activity, inhibited invasion and migration, and enhanced the efficacy of chemotherapeutic drugs in RCC. In various animal tumor models, phytochemicals suppressed renal tumor growth, reduced tumor size, and hindered angiogenesis and metastasis. The relevant antineoplastic mechanisms involved upregulation of caspases, reduction in cyclin activity, induction of cell cycle arrest and apoptosis via modulation of a plethora of cell signaling pathways. Clinical studies demonstrated a reduced risk for the development of kidney cancer and enhancement of the efficacy of chemotherapeutic drugs. Both preclinical and clinical studies displayed significant promise of utilizing phytochemicals for the prevention and treatment of RCC. Further research, confirming the mechanisms and regulatory pathways, along with randomized controlled trials, are needed to establish the use of phytochemicals in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (E.M.B.); (F.B.A.); (D.A.C.); (D.M.G.); (C.G.W.); (K.N.P.)
| |
Collapse
|
9
|
Byun WS, Bae ES, Kim WK, Lee SK. Antitumor Activity of Rutaecarpine in Human Colorectal Cancer Cells by Suppression of Wnt/β-Catenin Signaling. JOURNAL OF NATURAL PRODUCTS 2022; 85:1407-1418. [PMID: 35544614 DOI: 10.1021/acs.jnatprod.2c00224] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alkaloids derived from natural products have been traditionally used to treat various diseases, including cancers. Rutaecarpine (1), a β-carboline-type alkaloid obtained from Evodia rutaecarpa, has been previously reported as an anti-inflammatory agent. Nonetheless, its anticancer activity and the underlying molecular mechanisms remain to be explored. In the procurement of Wnt/β-catenin inhibitors from natural alkaloids, 1 was found to exhibit activity against the Wnt/β-catenin-response reporter gene. Since the abnormal activation of Wnt/β-catenin signaling is highly involved in colon carcinogenesis, the antitumor activity and molecular mechanisms of 1 were investigated in colorectal cancer (CRC) cells. The antiproliferative activity of 1 was associated with the suppression of the Wnt/β-catenin-mediated signaling pathway and its target gene expression in human CRC cells. 1 also induced G0/G1 cell cycle arrest and apoptotic cell death, and the antimigration and anti-invasion potential of 1 was confirmed through epithelial-mesenchymal transition biomarker inhibition by the regulation of Wnt signaling. The antitumor activity of 1 was supported in an Ls174T-implanted xenograft mouse model via Wnt target gene regulation. Overall, these findings suggest that targeting the Wnt/β-catenin signaling pathway by 1 is a promising therapeutic option for the treatment of human CRC harboring β-catenin mutation.
Collapse
Affiliation(s)
- Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
10
|
Li J, Tan G, Cai Y, Liu R, Xiong X, Gu B, He W, Liu B, Ren Q, Wu J, Chi B, Zhang H, Zhao Y, Xu Y, Zou Z, Kang F, Xu K. A novel Apigenin derivative suppresses renal cell carcinoma via directly inhibiting wild-type and mutant MET. Biochem Pharmacol 2021; 190:114620. [PMID: 34043966 DOI: 10.1016/j.bcp.2021.114620] [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: 02/23/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022]
Abstract
MET, the receptor of hepatocyte growth factor (HGF), is a driving factor in renal cell carcinoma (RCC) and also a proven drug target for cancer treatment. To improve the activity and to investigate the mechanisms of action of Apigenin (APG), novel derivatives of APG with improved properties were synthesized and their activities against Caki-1 human renal cancer cell line were evaluated. It was found that compound 15e exhibited excellent potency against the growth of multiple RCC cell lines including Caki-1, Caki-2 and ACHN and is superior to APG and Crizotinib. Subsequent investigations demonstrated that compound 15e can inhibit Caki-1 cell proliferation, migration and invasion. Mechanistically, 15e directly targeted the MET kinase domain, decreased its auto-phosphorylation at Y1234/Y1235 and inhibited its kinase activity and downstream signaling. Importantly, 15e had inhibitory activity against mutant MET V1238I and Y1248H which were resistant to approved MET inhibitors Cabozantinib, Crizotinib or Capmatinib. In vivo tumor graft study confirmed that 15e repressed RCC growth through inhibition of MET activation. These results indicate that compound 15e has the potential to be developed as a treatment for RCC, and especially against drug-resistant MET mutations.
Collapse
Affiliation(s)
- Jing Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Guishan Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Xiangya Hospital of Central South University, Changsha 410008, China
| | - Yabo Cai
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Ruihuan Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Zhuzhou Qianjin Pharmaceutical Co. Ltd, Zhuzhou, 412007, China
| | - Xiaolin Xiong
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Baohua Gu
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Wei He
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Bing Liu
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Qingyun Ren
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Jianping Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bo Chi
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Hang Zhang
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Yanzhong Zhao
- The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yangrui Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Zhenxing Zou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Fenghua Kang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Kangping Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
11
|
Jia XH, Zhao HX, Du CL, Tang WZ, Wang XJ. Possible pharmaceutical applications can be developed from naturally occurring phenanthroindolizidine and phenanthroquinolizidine alkaloids. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2020; 20:845-868. [PMID: 32994757 PMCID: PMC7517060 DOI: 10.1007/s11101-020-09723-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/14/2020] [Indexed: 06/11/2023]
Abstract
Naturally occurring phenanthroindolizidine and phenanthroquinolizidine alkaloids (PIAs and PQAs) are two small groups of herbal metabolites sharing a similar pentacyclic structure with a highly oxygenated phenanthrene moiety fused with a saturated or an unsaturated N-heterocycle (indolizidine/quinolizidine moieties). Natural PIAs and PQAs only could be obtained from finite plant families (such as Asclepiadaceae, Lauraceae and Urticaceae families, etc.). Up to date, more than one hundred natural PIAs, while only nine natural PQAs had been described. PIA and PQA analogues have been applied to the development of potent anticancer agents all along because of their excellent cytotoxic activity. However, in the last two decades, other great biological properties, such as anti-inflammatory and antiviral activities were revealed successively by different pharmacological assays. Especially because of their potent antiviral activity against coronavirus (TGEV, SARS CoV and MHV) and tobacco mosaic virus, PIA and PQA analogues have attracted much pharmaceutical attention again, some of them have been used to present interesting targets for total or semi synthesis, and structure-activity relationship (SAR) study for the development of antiviral agents. In this review, natural PIA and PQA analogues obtained in the last two decades with their herbal origins, key spectroscopic characteristics for structural identification, biological activity with possible SARs and application prospects were systematically summarized. We hope this paper can stimulate further investigations on PIA and PQA analogues as an important source for potential drug discovery.
Collapse
Affiliation(s)
- Xian-hui Jia
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250062 People’s Republic of China
| | - Huan-xin Zhao
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250062 People’s Republic of China
| | - Cheng-lin Du
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250062 People’s Republic of China
| | - Wen-zhao Tang
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250062 People’s Republic of China
| | - Xiao-jing Wang
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250062 People’s Republic of China
| |
Collapse
|
12
|
Renner J, Thakur A, Rutz PM, Cowley JM, Evangelista JL, Kumar P, Prater MB, Stolley RM, Louie J. Total Synthesis of Indolizidine Alkaloids via Nickel-Catalyzed (4 + 2) Cyclization. Org Lett 2020; 22:924-928. [DOI: 10.1021/acs.orglett.9b04479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jonas Renner
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Ashish Thakur
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Philipp M. Rutz
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Jacob M. Cowley
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Judah L. Evangelista
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Puneet Kumar
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Matthew B. Prater
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Ryan M. Stolley
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Janis Louie
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| |
Collapse
|
13
|
Bach DH, Lee SK. The Potential Impacts of Tylophora Alkaloids and their Derivatives in Modulating Inflammation, Viral Infections, and Cancer. Curr Med Chem 2019; 26:4709-4725. [PMID: 30047325 DOI: 10.2174/0929867325666180726123339] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022]
Abstract
Cancer chemotherapies or antitumor agents mainly remain the backbone of current treatment based on killing the rapidly dividing cancer cell such as tylophora alkaloids and their analogues which have also demonstrated anticancer potential through diverse biological pathways including regulation of the immune system. The introduction of durable clinically effective monoclonal antibodies, however, unmasked a new era of cancer immunotherapies. Therefore, the understanding of cancer pathogenesis will provide new possible treatment options, including cancer immunotherapy and targeted agents. Combining cytotoxic agents and immunotherapies may offer several unique advantages that are complementary to and potentially synergistic with biologic modalities. Herein, we highlight the dynamic mechanism of action of immune modulation in cancer and the immunological aspects of the orally active antitumor agents tylophora alkaloids and their analogues. We also suggest that future cancer treatments will rely on the development of combining tumor-targeted agents and biologic immunotherapies.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
14
|
Mondal A, Gandhi A, Fimognari C, Atanasov AG, Bishayee A. Alkaloids for cancer prevention and therapy: Current progress and future perspectives. Eur J Pharmacol 2019; 858:172472. [PMID: 31228447 DOI: 10.1016/j.ejphar.2019.172472] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Alkaloids are important chemical compounds that serve as a rich source for drug discovery. Numerous alkaloids screened from medicinal plants and herbs showed antiproliferative and anticancer effects on wide category of cancers both in vitro and in vivo. Vinblastine, vinorelbine, vincristine, and vindesine have already been successfully developed as anticancer drugs. The available and up-to-date information on the ethnopharmacological uses in traditional medicine, phytochemistry, pharmacology and clinical utility of alkaloids were collected using various resources (PubMed, ScienceDirect, Google Scholar and Springerlink). In this article, we provide a comprehensive and critical overview on naturally-occurring alkaloids with anticancer activities and highlight the molecular mechanisms of action of these secondary metabolites. Furthermore, this review also presents a summary of synthetic derivatives and pharmacological profiles useful to researchers for the therapeutic development of alkaloids. Based on the literature survey compiled in this review, alkaloids represent an important group of anticancer drugs of plant origin with enormous potential for future development of drugs for cancer therapy and management.
Collapse
Affiliation(s)
- Arijit Mondal
- Department of Pharmacy, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, 700 053, West Bengal, India.
| | - Arijit Gandhi
- Department of Pharmaceutics, Bengal College of Pharmaceutical Science and Research, Durgapur, 713 212, West Burdwan, West Bengal, India
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto 237, 47921, Rimini, Italy
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, 1090, Vienna, Austria; Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev Street, Sofia, 1113, Bulgaria
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
15
|
Genomic Identification of the TOR Signaling Pathway as a Target of the Plant Alkaloid Antofine in the Phytopathogen Fusarium graminearum. mBio 2019; 10:mBio.00792-19. [PMID: 31186319 PMCID: PMC6561021 DOI: 10.1128/mbio.00792-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Fusarium head blight caused by the fungal pathogen Fusarium graminearum is a devastating disease of cereal crops worldwide, with limited effective chemical treatments available. Here we show that the natural alkaloid compound antofine can inhibit fusarium head blight in wheat. Using yeast genomic screening, we identified the TOR pathway component RRD2 as a target of antofine that is also required for F. graminearum pathogenicity. Antofine, a phenanthroindolizidine alkaloid, is a bioactive natural product isolated from milkweeds that exhibits numerous biological activities, including anticancer, antimicrobial, antiviral, and anti-inflammatory properties. However, the direct targets and mode of action of antofine have not been determined. In this report, we show that antofine displays antifungal properties against the phytopathogen Fusarium graminearum, the cause of Fusarium head blight disease (FHB). FHB does devastating damage to agriculture, causing billions of dollars in economic losses annually. We therefore sought to understand the mode of action of antofine in F. graminearum using insights from yeast chemical genomic screens. We used haploinsufficiency profiling (HIP) to identify putative targets of antofine in yeast and identified three candidate targets, two of which had homologs in F. graminearum. The Fusarium homologues of two targets, glutamate dehydrogenase (FgGDH) and resistance to rapamycin deletion 2 (FgRRD2), can bind antofine. Of the two genes, only the Fgrrd2 knockout displayed a loss of virulence in wheat, indicating that RRD2 is an antivirulence target of antofine in F. graminearum. Mechanistically, we demonstrate that antofine disrupts the interaction between FgRRD2 and FgTap42, which is part of the Tap42-phosphatase complex in the target of rapamycin (TOR) signaling pathway, a central regulator of cell growth in eukaryotes and a pathway of extensive study for controlling numerous pathologies.
Collapse
|
16
|
Antitumor Activity of DFX117 by Dual Inhibition of c-Met and PI3Kα in Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11050627. [PMID: 31060329 PMCID: PMC6562471 DOI: 10.3390/cancers11050627] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
Aberrant activation of hepatocyte growth factor (HGF)/c-Met signaling pathway caused by gene amplification or mutation plays an important role in tumorigenesis. Therefore, c-Met is considered as an attractive target for cancer therapy and c-Met inhibitors have been developed with great interests. However, cancers treated with c-Met inhibitors inevitably develop resistance commonly caused by the activation of PI3K/Akt signal transduction pathway. Therefore, the combination of c-Met and PI3Kα inhibitors showed synergistic activities, especially, in c-Met hyperactivated and PIK3CA-mutated cells. In our previous study, we rationally designed and synthesized DFX117(6-(5-(2,4-difluorophenylsulfonamido)-6-methoxypyridin-3-yl)-N-(2-morpholinoethyl) imidazo[1,2-a]pyridine-3-carboxamide) as a novel PI3Kα selective inhibitor. Herein, the antitumor activity and underlying mechanisms of DFX117 against non-small cell lung cancer (NSCLC) cells were evaluated in both in vitro and in vivo animal models. Concurrent targeted c-Met and PI3Kα by DFX117 dose-dependent inhibited the cell growth of H1975 cells (PIK3CA mutation and c-Met amplification) and A549 cells (KRAS mutation). DFX117 subsequently induced G0/G1 cell cycle arrest and apoptosis. These data highlight the significant potential of DFX117 as a feasible and efficacious agent for the treatment of NSCLC patients.
Collapse
|
17
|
Li S, Zhao H, Yin Z, Deng S, Gao Y, Li Y, Dai T. Synthesis and Antitumor Activity Evaluation of New Phenanthrene-Based Tylophorine Derivatives. LETT ORG CHEM 2019. [DOI: 10.2174/1570178615666181025115513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A series of new phenanthrene-based tylophorine derivatives (PBTs) were synthesized in
good yield and their structures were characterized by 1H-NMR spectroscopy and ESI MS. In vitro antitumor
activity of these compounds against five human carcinoma cell lines, including HCT116 (colorectal),
BGC-823 (gastric), HepG-2 (hepatic), Hela (cervical) and H460 (lung) cells, was evaluated by
MTT assay. Among these PBTs, compound 6b showed the highest antitumor activities against
HCT116 and HepG-2 cell lines with IC50 values of 6.1 and 6.4 μM, respectively, which were comparable
to that of adriamycin hydrochloride. The structure-activity relationship of these compounds was
also discussed based on the results of their antitumor activity.
Collapse
Affiliation(s)
- Songtao Li
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Hongling Zhao
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Zhifeng Yin
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Shuhua Deng
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Yang Gao
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Yunfeng Li
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| | - Tao Dai
- Hebei Province Key Laboratory of Research and Development of Traditional Chinese Medicine, Institute of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, China
| |
Collapse
|
18
|
Ansari A, Ramapanicker R. Enantioselective Synthesis of (R)-Antofine and (R)-Cryptopleurine. ChemistrySelect 2018. [DOI: 10.1002/slct.201803529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Anas Ansari
- Department of Chemistry; Indian Institute of Technology Kanpur Kanpur; Uttar Pradesh- 208016 India
| | - Ramesh Ramapanicker
- Department of Chemistry; Indian Institute of Technology Kanpur Kanpur; Uttar Pradesh- 208016 India
| |
Collapse
|
19
|
Byun WS, Jin M, Yu J, Kim WK, Song J, Chung HJ, Jeong LS, Lee SK. A novel selenonucleoside suppresses tumor growth by targeting Skp2 degradation in paclitaxel-resistant prostate cancer. Biochem Pharmacol 2018; 158:84-94. [PMID: 30292755 DOI: 10.1016/j.bcp.2018.10.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/02/2018] [Indexed: 01/03/2023]
Abstract
Prostate cancer (PC) is the most common disease in men over age 50, and its prevalence rate has been gradually increasing since 1980. Taxane-derived anticancer agents are the primary agents used to treat metastatic prostate cancer patients; however, the side effects and acquired drug resistance limit the success of these therapies. Because there is no specific treatment for paclitaxel-resistant prostate cancer, it is necessary to develop new targets and therapeutic strategies to overcome the acquired resistance. In this study, the antitumor activity of a novel selenonucleoside (4'-selenofuranosyl-2,6-dichloropurine, LJ-2618), a third-generation nucleoside, and its plausible mechanisms of action in paclitaxel-resistant prostate cancer (PC-3-Pa) cells were investigated. The established PC-3-Pa cells exhibited over 100-fold resistance against paclitaxel compared to the paclitaxel-sensitive PC-3 cells. LJ-2618, however, effectively inhibited the proliferation of both cell lines with similar IC50 values in vitro. In PC-3-Pa cells, the activated PI3K/Akt signaling pathway was suppressed by LJ-2618 treatment. In addition, Skp2 was found to be over-expressed in paclitaxel-resistant cells, and the transfection of Skp2 siRNA recovered the sensitivity of paclitaxel in PC-3-Pa cells. Furthermore, LJ-2618 significantly down-regulated Skp2 expression in PC-3-Pa cells by promoting degradation and inducing destabilization of Skp2, which triggers G2/M cell cycle arrest. In a xenograft mouse model implanted with PC-3-Pa cells, LJ-2618 (3 or 10 mg/kg) effectively inhibited tumor growth with the enhancement of Skp2 degradation and induction of p27 expression in tumor tissues. These findings suggest that LJ-2618 may have potential for overcoming paclitaxel resistance via promoting Skp2 degradation and stabilizing p27 expression in PC-3-Pa cells. Therefore, the novel selenonucleoside LJ-2618 may lead to the development of a new treatment strategy for patients with paclitaxel-resistant, castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Woong Sub Byun
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Minkyung Jin
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinha Yu
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jayoung Song
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Jin Chung
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Lak Shin Jeong
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
20
|
Multiplicity of acquired cross-resistance in paclitaxel-resistant cancer cells is associated with feedback control of TUBB3 via FOXO3a-mediated ABCB1 regulation. Oncotarget 2018; 7:34395-419. [PMID: 27284014 PMCID: PMC5085164 DOI: 10.18632/oncotarget.9118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
Acquired drug resistance is a primary obstacle for effective cancer therapy. The correlation of point mutations in class III β-tubulin (TUBB3) and the prominent overexpression of ATP-binding cassette P-glycoprotein (ABCB1), a multidrug resistance gene, have been protruding mechanisms of resistance to microtubule disruptors such as paclitaxel (PTX) for many cancers. However, the precise underlying mechanism of the rapid onset of cross-resistance to an array of structurally and functionally unrelated drugs in PTX-resistant cancers has been poorly understood. We determined that our established PTX-resistant cancer cells display ABCB1/ABCC1-associated cross-resistance to chemically different drugs such as 5-fluorouracil, docetaxel, and cisplatin. We found that feedback activation of TUBB3 can be triggered through the FOXO3a-dependent regulation of ABCB1, which resulted in the accentuation of induced PTX resistance and encouraged multiplicity in acquired cross-resistance. FOXO3a-directed regulation of P-glycoprotein (P-gp) function suggests that control of ABCB1 involves methylation-dependent activation. Consistently, transcriptional overexpression or downregulation of FOXO3a directs inhibitor-controlled protease-degradation of TUBB3. The functional PI3K/Akt signaling is tightly responsive to FOXO3a activation alongside doxorubicin treatment, which directs FOXO3a arginine hypermethylation. In addition, we found that secretome factors from PTX-resistant cancer cells with acquired cross-resistance support a P-gp-dependent association in multidrug resistance (MDR) development, which assisted the FOXO3a-mediated control of TUBB3 feedback. The direct silencing of TUBB3 reverses induced multiple cross-resistance, reduces drug-resistant tumor mass, and suppresses the impaired microtubule stability status of PTX-resistant cells with transient cross-resistance. These findings highlight the control of the TUBB3 response to ABCB1 genetic suppressors as a mechanism to reverse the profuse development of multidrug resistance in cancer.
Collapse
|
21
|
Antofine, a natural phenanthroindolizidine alkaloid, suppresses angiogenesis via regulation of AKT/mTOR and AMPK pathway in endothelial cells and endothelial progenitor cells derived from mouse embryonic stem cells. Food Chem Toxicol 2017; 107:201-207. [DOI: 10.1016/j.fct.2017.06.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/18/2017] [Accepted: 06/22/2017] [Indexed: 11/17/2022]
|
22
|
Liu J, He Y, Zhang D, Cai Y, Zhang C, Zhang P, Zhu H, Xu N, Liang S. In vitro anticancer effects of two novel phenanthroindolizidine alkaloid compounds on human colon and liver cancer cells. Mol Med Rep 2017; 16:2595-2603. [PMID: 28677760 PMCID: PMC5548052 DOI: 10.3892/mmr.2017.6879] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/07/2017] [Indexed: 02/05/2023] Open
Abstract
Malignant cancer is one of the most serious diseases threatening the health of human beings. Natural plant alkaloids exhibit multiple biological functions, including inhibition of cell proliferation, and may have potential anticancer activity. However, most natural alkaloids may not be suitable for human therapies owing to instability, poor dissolubility and potential side effects. To improve their anticancer activity and drug effect, the present study aimed to develop new alkaloid derivatives, the phenanthroindolizidine alkaloid compounds, and evaluated their potential antitumor effects on human cancer cells in vitro. Among the several newly synthesized analogues of phenanthroindolizidine alkaloids (PAs), the compounds YS306 and YS206 exhibited an increased growth inhibition activity on HepG2 liver cancer cells and on HCT116 and HT29 colon cancer cells, with half‑maximal inhibitory concentrations in the micromolar range. YS206 and YS306 (5 µg/ml) both significantly induced cell cycle arrest at the G2/M phase and notably decreased cell distribution at the G0/G1 and S phase. In addition, these two molecules significantly inhibited cancer cell migration, as analyzed by the wound‑healing and Transwell assays. However, neither YS306 nor YS206 exhibited observable effects on apoptosis. Therefore, chemical structure modifications of natural PAs based on anticancer activity assessments may be feasible in the development of new cancer chemotherapeutic agents.
Collapse
Affiliation(s)
- Jingjing Liu
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Yu He
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Dan Zhang
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Ying Cai
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Chenggang Zhang
- Department of Chemistry and Materials, Sichuan Normal University, Chengdu, Sichuan 610066, P.R. China
| | - Peng Zhang
- Department of Urinary Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 100034, P.R. China
| | - Ningzhi Xu
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Shufang Liang
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
23
|
Aldonza MBD, Son YS, Sung HJ, Ahn JM, Choi YJ, Kim YI, Cho S, Cho JY. Paraoxonase-1 (PON1) induces metastatic potential and apoptosis escape via its antioxidative function in lung cancer cells. Oncotarget 2017; 8:42817-42835. [PMID: 28467805 PMCID: PMC5522108 DOI: 10.18632/oncotarget.17069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/20/2017] [Indexed: 01/19/2023] Open
Abstract
Paraoxonase-1 (PON1) gene polymorphisms have been closely associated with the development of advanced cancers while PON1 secretion to the serum is linked with inhibition of oxidized high-density lipoprotein by its antioxidative function. Our group previously demonstrated that post-translational modification of serum PON1 in form of fucosylated PON1 is a potential biomarker of small cell lung cancer. Here, we interrogated the role of PON1 in the pathobiology of lung cancer (LC) by addressing cell-autonomous mechanisms using gain-of-function and loss-of-function approaches and protein expression profiling of tissue samples in our clinical biobank. PON1 expression in LC patient tissues varied between overexpression in squamous cell carcinoma and minimal loss in adenocarcinoma sub-types. Simultaneous overexpression of PON1 both at the gene and protein stability levels induced pro-oncogenic characteristics in LC cells and xenografts. PON1 overexpression supported metastatic progression of LC by decreasing G1/S ratio and LC cell senescence involving p21Waf1/Cip1. PON1 suppressed drug- and ligand-induced cell death and protected LC cells from genotoxic damages with maintained ATP levels, requiring p53-directed signals. PON1 promoted ROS deregulation protecting the mitochondria from dysregulation. PON1 knockdown resulted in the blockage of its antioxidant function in LC cells through Akt signaling with reduced invasive signature as a consequence of scant expression. Targeted glycolysis stimulated PON1 antioxidant activity regulating phosphorylation of AMPK-α. The functional data imply that exploitation of the antioxidative function of PON1 is consequential in driving LC pathogenesis at the cell-autonomous mechanistic level with consequences on tumor growth.
Collapse
Affiliation(s)
- Mark Borris D. Aldonza
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yeon Sung Son
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jung Mo Ahn
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: Bio Center, Incheon Technopark, Incheon, Republic of Korea
| | - Young-Jin Choi
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoungnam-Si, Gyeonggi-Do, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Anti-Melanogenic Activity of Gagunin D, a Highly Oxygenated Diterpenoid from the Marine Sponge Phorbas sp., via Modulating Tyrosinase Expression and Degradation. Mar Drugs 2016; 14:md14110212. [PMID: 27869664 PMCID: PMC5128755 DOI: 10.3390/md14110212] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 12/19/2022] Open
Abstract
Tyrosinase is the rate-limiting enzyme critical for melanin synthesis and controls pigmentation in the skin. The inhibition of tyrosinase is currently the most common approach for the development of skin-whitening cosmetics. Gagunin D (GD), a highly oxygenated diterpenoid isolated from the marine sponge Phorbas sp., has exhibited cytotoxicity toward human leukemia cells. However, the effect of GD on normal cells and the molecular mechanisms remain to be elucidated. In the present study, we identified for the first time the anti-melanogenic activity of GD and its precise underlying mechanisms in mouse melan-a cells. GD significantly inhibited melanin synthesis in the melan-a cells and a reconstructed human skin model. Further analysis revealed that GD suppressed the expression of tyrosinase and increased the rate of tyrosinase degradation. GD also inhibited tyrosinase enzymatic activity. In addition, GD effectively suppressed the expression of proteins associated with melanosome transfer. These findings suggest that GD is a potential candidate for cosmetic formulations due to its multi-functional properties.
Collapse
|
25
|
Kwon Y, Song J, Lee H, Kim EY, Lee K, Lee SK, Kim S. Design, Synthesis, and Biological Activity of Sulfonamide Analogues of Antofine and Cryptopleurine as Potent and Orally Active Antitumor Agents. J Med Chem 2015; 58:7749-62. [PMID: 26393416 DOI: 10.1021/acs.jmedchem.5b00764] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Due to their profound antiproliferative activity and unique mode of action, phenanthroindolizidine and phenanthroquinolizidine alkaloids, represented by antofine and cryptopleurine, have attracted attention recently as potential therapeutic agents. We have designed, synthesized, and evaluated the methanesulfonamide analogues of these natural alkaloids with the hope of improving their druglikeness. The analogues showed enhanced growth inhibition of human cancer cells compared with the parent natural products. In particular, a methanesulfonamide analogue of cryptopleurine (5b) exhibited improved bioavailability and significant antitumor activity, which suggests that 5b is a promising new anticancer agent. Our studies suggest that the inhibition of cancer cell growth by 5b is associated with the induction of G0/G1 cell cycle arrest via nicotinamide N-methyltransferase-dependent JNK activation in Caki-1 renal cancer cells. In addition, compound 5b significantly inhibited the migration and invasion of Caki-1 cancer cells by modulating the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Yongseok Kwon
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Jayoung Song
- Natural Products Research Institute, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Honggu Lee
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Eun-Yeong Kim
- College of Pharmacy, Korea University , Sejong 339-700, Korea
| | - Kiho Lee
- College of Pharmacy, Korea University , Sejong 339-700, Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Sanghee Kim
- Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| |
Collapse
|