1
|
Lin F, Li J, Zhou L, Yi R, Chen Y, He S. Targeting vulnerability in tumor therapy: Dihydroorotate dehydrogenase. Life Sci 2025; 371:123612. [PMID: 40187643 DOI: 10.1016/j.lfs.2025.123612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine biosynthetic pathway and a recognized therapeutic target in various diseases. In oncology research, DHODH has gained increasing importance and become a hot target for various tumor therapy studies. This review highlights three key points: (1) DHODH enables its diverse biological functions through its unique structural features and dominates the regulation of tumor metabolism and cell fate; (2) DHODH activates oncogenic signals, drives metastatic adaptation, and remodels drug resistance networks in tumors, making it a metabolic-signaling dual hub; and (3) DHODH inhibitors have shown significant efficacy in preclinical models of various tumors but face multiple challenges in clinical trials, including drug-related limitations and external constraints. Given these challenges, future research should explore DHODH inhibitors as a foundation for overcoming technological and translational barriers while establishing a systematic framework for the clinical application of DHODH-targeted tumor therapies.
Collapse
Affiliation(s)
- Fu Lin
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Jiaxin Li
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Zhou
- Laboratory of Pathogen Biology and Immunology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Rigui Yi
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Yingge Chen
- School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Shuai He
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China.
| |
Collapse
|
2
|
Martins M, Veiga F, Paiva-Santos AC, Pires PC. Drug Repurposing and Nanotechnology for Topical Skin Cancer Treatment: Redirecting toward Targeted and Synergistic Antitumor Effects. ACS Pharmacol Transl Sci 2025; 8:308-338. [PMID: 39974652 PMCID: PMC11833728 DOI: 10.1021/acsptsci.4c00679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Skin cancer represents a major health concern due to its rising incidence and limited treatment options. Current treatments (surgery, chemotherapy, radiotherapy, immunotherapy, and targeted therapy) often entail high costs, patient inconvenience, significant adverse effects, and limited therapeutic efficacy. The search for novel treatment options is also marked by the high capital investment and extensive development involved in the drug discovery process. In response to these challenges, repurposing existing drugs for topical application and optimizing their delivery through nanotechnology could be the answer. This innovative strategy aims to combine the advantages of the known pharmacological background of commonly used drugs to expedite therapeutic development, with nanosystem-based formulations, which among other advantages allow for improved skin permeation and retention and overall higher therapeutic efficacy and safety. The present review provides a critical analysis of repurposed drugs such as doxycycline, itraconazole, niclosamide, simvastatin, leflunomide, metformin, and celecoxib, formulated into different nanosystems, namely, nanoemulsions and nanoemulgels, nanodispersions, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanoparticles, hybrid lipid-polymer nanoparticles, hybrid electrospun nanofibrous scaffolds, liposomes and liposomal gels, ethosomes and ethosomal gels, and aspasomes, for improved outcomes in the battle against skin cancer. Enhanced antitumor effects on melanoma and nonmelanoma research models are highlighted, with some nanoparticles even showing intrinsic anticancer properties, leading to synergistic effects. The explored research findings highly evidence the potential of these approaches to complement the currently available therapeutic strategies in the hope that these treatments might one day reach the pharmaceutical market.
Collapse
Affiliation(s)
- Maria Martins
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- RISE-Health,
Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
3
|
Niwata C, Nakagawa T, Naruse T, Sakuma M, Yamakado N, Akagi M, Ono S, Tobiume K, Gao J, Jimi E, Ohta K, Aikawa T. Anticancer effect of the antirheumatic drug leflunomide on oral squamous cell carcinoma by the inhibition of tumor angiogenesis. Discov Oncol 2025; 16:53. [PMID: 39815040 PMCID: PMC11735718 DOI: 10.1007/s12672-025-01763-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVES Leflunomide (LEF) is a conventional synthetic disease-modifying antirheumatic drug and suppresses T-cell proliferation and activity by inhibiting pyrimidine synthesis using dihydroorotase dehydrogenase (DHODH); however, several studies have demonstrated that LEF possesses anticancer and antiangiogenic effects in some malignant tumors. Therefore, we investigated the anticancer and antiangiogenic effects of LEF on oral squamous cell carcinoma (OSCC). METHODS To evaluate the inhibitory effect of LEF on OSCC, cell proliferation and wound-healing assays using human OSCC cell lines were performed. The DHODH inhibitory effect of LEF was evaluated by Western blot. To assess the suppression of pyrimidine biosynthesis induced by LEF on OSCC, cell proliferation assays with or without uridine supplementation were performed. The antiangiogenic effect of LEF was evaluated by in vitro tube formation assay using immortalized human umbilical vein endothelial cells, which were electroporatically transfected with hTERT. The tumor-suppressive effect of LEF in vivo was examined in both immunodeficient and syngeneic mice by implanting mouse OSCC cells. Tumor vascularization was evaluated by immunohistochemistry of the tumor extracted from syngeneic mice. RESULTS LEF dose-dependently inhibited OSCC proliferation and migration. LEF significantly inhibited DHODH expression, and uridine supplementation rescued the inhibitory effect of LEF. LEF dose-dependently suppressed endothelial tube formation. In the animal study, LEF significantly suppressed tumor growth in both immunodeficient and syngeneic mice. Histologically, LEF decreased DHODH expression and tumor vascularization. CONCLUSION LEF is a potent anticancer agent with antiangiogenic effects on OSCC and might be clinically applicable to OSCC by drug repositioning.
Collapse
Affiliation(s)
- Chieko Niwata
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Takayuki Nakagawa
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Takako Naruse
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Miyuki Sakuma
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Nao Yamakado
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Misaki Akagi
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Shigehiro Ono
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Kei Tobiume
- Graduate School of Biomedical & Health Sciences (Dentistry & Oral Health Sciences), Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Jing Gao
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eijiro Jimi
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kouji Ohta
- Department of Public Oral Health, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Tomonao Aikawa
- Department of Oral and Maxillofacial Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
4
|
Zewail M. Leflunomide nanocarriers: a new prospect of therapeutic applications. J Microencapsul 2024; 41:715-738. [PMID: 39320955 DOI: 10.1080/02652048.2024.2407373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Leflunomide (LEF) is a well-known disease-modifying anti-rheumatic agent (DMARDs) that was approved in 1998 for rheumatoid arthritis (RA) management. It is enzymatically converted into active metabolite teriflunomide (TER) inside the body. LEF and TER possess several pharmacological effects in a variety of diseases including multiple sclerosis, cancer, viral infections and neurobehavioral brain disorders. Despite the aforementioned pharmacological effects exploring these effects in nanomedicine applications has been focused mainly on RA and cancer treatment. This review summarises the main pharmacological, and pharmacokinetic effects of LEF along with highlighting the applications of nanoencapsulation of LEF and its metabolite in different diseases.
Collapse
Affiliation(s)
- Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| |
Collapse
|
5
|
Kabir MZ, Tayyab H, Erkmen C, Mohamad SB, Uslu B. Comprehensive views toward the biomolecular recognition of an anticancer drug, leflunomide with human serum albumin. J Biomol Struct Dyn 2024; 42:7257-7271. [PMID: 37529911 DOI: 10.1080/07391102.2023.2239931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/16/2023] [Indexed: 08/03/2023]
Abstract
Biomolecular association of an anticancer drug, leflunomide (LEF) with human serum albumin (HSA), the leading ligands carrier in human circulation was characterized using biophysical (i.e., fluorescence, absorption and voltammetric) methods and computational (i.e., molecular docking and molecular dynamics simulation) techniques. Evaluations of fluorescence, absorption and voltammetric findings endorsed the complex formation between LEF and HSA. An inverse relationship of Stern-Volmer constant-temperature and hyperchromic shift of the protein's absorption signal with addition of LEF confirmed the LEF quenched the HSA fluorescence through static process. Moderate nature of binding strength (binding constant = 2.76-4.77 × 104 M-1) was detected towards the LEF-HSA complexation, while the association process was naturally driven via hydrophobic interactions, van der Waals interactions and hydrogen bonds, as evident from changes in entropy (ΔS= + 19.91 J mol-1 K-1) and enthalpy (ΔH = - 20.09 kJ mol-1), and molecular docking assessments. Spectral analyses of synchronous and three-dimensional fluorescence validated microenvironmental fluctuations near Trp and Tyr residues upon LEF binding to the protein. LEF association with HSA significantly defended temperature-induced destabilization of the protein. Although LEF was found to attach to HSA at Sudlow's sites I and II, but exhibited greater preference toward its site I, as detected by the investigations of competitive site-marker displacement. Molecular dynamics simulation assessment revealed that the complex attained equilibrium throughout simulations, showing the LEF-HSA complex constancy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Md Zahirul Kabir
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| | - Hafsa Tayyab
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Cem Erkmen
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| | - Saharuddin B Mohamad
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
- Centre of Research for Computational Sciences and Informatics for Biology, Bioindustry, Environment, Agriculture and Healthcare, University of Malaya, Kuala Lumpur, Malaysia
| | - Bengi Uslu
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| |
Collapse
|
6
|
Yousaf S, Shahzadi K. Utilizing topological indices in QSPR modeling to identify non-cancer medications with potential anti-cancer properties: a promising strategy for drug repurposing. Front Chem 2024; 12:1410882. [PMID: 39176073 PMCID: PMC11338857 DOI: 10.3389/fchem.2024.1410882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024] Open
Abstract
The exploration of non-cancer medications with potential anti-cancer activity offers a promising avenue for drug repurposing, accelerating the development of new oncological therapies. This study employs Quantitative Structure-Property Relationship (QSPR) modeling to identify and predict the anti-cancer efficacy of various non-cancer drugs, utilizing topological indices as key descriptors. Topological indices, which capture the molecular structure's geometric and topological characteristics, provide critical insights into the pharmacological interactions relevant to anti-cancer activity. By analyzing a comprehensive dataset of non-cancer medications, this research establishes robust QSPR models that correlate topological indices with anti-cancer activity. The models demonstrate significant predictive power, highlighting several non-cancer drugs with potential anti-cancer properties. Further, we will use linear, quadratic and logarithmic regression to understand the structures of anti-cancer drugs and strengthen our ability to manipulate the molecular structures. The findings underscore the utility of topological indices in drug repurposing strategies and pave the way for further experimental validation and clinical trials. This integrative approach enhances our understanding of drug action mechanisms and offers a cost-effective strategy for expanding the repertoire of anti-cancer agents.
Collapse
Affiliation(s)
- Shamaila Yousaf
- Department of Mathematics, University of Gujrat, Gujrat, Pakistan
| | | |
Collapse
|
7
|
Mirzapoiazova T, Tseng L, Mambetsariev B, Li H, Lou CH, Pozhitkov A, Ramisetty SK, Nam S, Mambetsariev I, Armstrong B, Malhotra J, Arvanitis L, Nasser MW, Batra SK, Rosen ST, Wheeler DL, Singhal SS, Kulkarni P, Salgia R. Teriflunomide/leflunomide synergize with chemotherapeutics by decreasing mitochondrial fragmentation via DRP1 in SCLC. iScience 2024; 27:110132. [PMID: 38993482 PMCID: PMC11237869 DOI: 10.1016/j.isci.2024.110132] [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: 12/15/2023] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 07/13/2024] Open
Abstract
Although up to 80% small cell lung cancer (SCLC) patients' response is good for first-line chemotherapy regimen, most patients develop recurrence of the disease within weeks to months. Here, we report cytostatic effect of leflunomide (Leflu) and teriflunomide (Teri) on SCLC cell proliferation through inhibition of DRP1 phosphorylation at Ser616 and decreased mitochondrial fragmentation. When administered together, Teri and carboplatin (Carbo) act synergistically to significantly inhibit cell proliferation and DRP1 phosphorylation, reduce abundance of intermediates in pyrimidine de novo pathway, and increase apoptosis and DNA damage. Combination of Leflu&Carbo has anti-tumorigenic effect in vivo. Additionally, lurbinectedin (Lur) and Teri potently and synergistically inhibited spheroid growth and depleted uridine and DRP1 phosphorylation in mouse tumors. Our results suggest combinations of Carbo and Lur with Teri or Leflu are efficacious and underscore how the relationship between DRP1/DHODH and mitochondrial plasticity serves as a potential therapeutic target to validate these treatment strategies in SCLC clinical trials.
Collapse
Affiliation(s)
- Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Liz Tseng
- Department of Shared Resources, Light Microscopy Digital Imaging Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Bolot Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Haiqing Li
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Chih-Hong Lou
- Genome Editing Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Alex Pozhitkov
- Division of Research Informatics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Sravani Keerthi Ramisetty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sangkil Nam
- Department of Shared Resources, Molecular Pathology Core, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Brian Armstrong
- Department of Shared Resources, Light Microscopy Digital Imaging Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jyoti Malhotra
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Steven T. Rosen
- Hematology Malignancies and Stem Cell Transplantation Institute, Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Deric L. Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institute for Medical Research, Madison, WI, USA
| | - Sharad S. Singhal
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
8
|
Zhao Y, Chen C, Xiao X, Fang L, Cheng X, Chang Y, Peng F, Wang J, Shen S, Wu S, Huang Y, Cai W, Zhou L, Qiu W. Teriflunomide Promotes Blood-Brain Barrier Integrity by Upregulating Claudin-1 via the Wnt/β-catenin Signaling Pathway in Multiple Sclerosis. Mol Neurobiol 2024; 61:1936-1952. [PMID: 37819429 DOI: 10.1007/s12035-023-03655-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/10/2023] [Indexed: 10/13/2023]
Abstract
The blood-brain barrier (BBB) and tight junction (TJ) proteins maintain the homeostasis of the central nervous system (CNS). The dysfunction of BBB allows peripheral T cells infiltration into CNS and contributes to the pathophysiology of multiple sclerosis (MS). Teriflunomide is an approved drug for the treatment of MS by suppressing lymphocytes proliferation. However, whether teriflunomide has a protective effect on BBB in MS is not understood. We found that teriflunomide restored the injured BBB in the EAE model. Furthermore, teriflunomide treatment over 6 months improved BBB permeability and reduced peripheral leakage of CNS proteins in MS patients. Teriflunomide increased human brain microvascular endothelial cell (HBMEC) viability and promoted BBB integrity in an in vitro cell model. The TJ protein claudin-1 was upregulated by teriflunomide and responsible for the protective effect on BBB. Furthermore, RNA sequencing revealed that the Wnt signaling pathway was affected by teriflunomide. The activation of Wnt signaling pathway increased claudin-1 expression and reduced BBB damage in cell model and EAE rats. Our study demonstrated that teriflunomide upregulated the expression of the tight junction protein claudin-1 in endothelial cells and promoted the integrity of BBB through Wnt signaling pathway.
Collapse
Affiliation(s)
- Yipeng Zhao
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
- The Center of Mental and Neurological Disorders Study, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Xiuqing Xiao
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Ling Fang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Xi Cheng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Yanyu Chang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Jingqi Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Shishi Shen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Shilin Wu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Yiying Huang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Wei Cai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
- The Center of Mental and Neurological Disorders Study, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Linli Zhou
- The Center of Mental and Neurological Disorders Study, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
- The Center of Mental and Neurological Disorders Study, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510000, China.
| |
Collapse
|
9
|
Gehlot P, Vyas VK. A Patent Review of Human Dihydroorotate Dehydrogenase (hDHODH) Inhibitors as Anticancer Agents and their Other Therapeutic Applications (1999-2022). Recent Pat Anticancer Drug Discov 2024; 19:280-297. [PMID: 37070439 DOI: 10.2174/1574892818666230417094939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 04/19/2023]
Abstract
Highly proliferating cells, such as cancer cells, are in high demand of pyrimidine nucleotides for their proliferation, accomplished by de novo pyrimidine biosynthesis. The human dihydroorotate dehydrogenase (hDHODH) enzyme plays a vital role in the rate-limiting step of de novo pyrimidine biosynthesis. As a recognised therapeutic target, hDHODH plays a significant role in cancer and other illness. In the past two decades, small molecules as inhibitors hDHODH enzyme have drawn much attention as anticancer agents, and their role in rheumatoid arthritis (RA), and multiple sclerosis (MS). In this patent review, we have compiled patented hDHODH inhibitors published between 1999 and 2022 and discussed the development of hDHODH inhibitors as anticancer agents. Therapeutic potential of small molecules as hDHODH inhibitors for the treatment of various diseases, such as cancer, is very well recognised. Human DHODH inhibitors can rapidly cause intracellular uridine monophosphate (UMP) depletion to produce starvation of pyrimidine bases. Normal cells can better endure a brief period of starvation without the side effects of conventional cytotoxic medication and resume synthesis of nucleic acid and other cellular functions after inhibition of de novo pathway using an alternative salvage pathway. Highly proliferative cells such as cancer cells do not endure starvation because they are in high demand of nucleotides for cell differentiation, which is fulfilled by de novo pyrimidine biosynthesis. In addition, hDHODH inhibitors produce their desired activity at lower doses rather than a cytotoxic dose of other anticancer agents. Thus, inhibition of de novo pyrimidine biosynthesis will create new prospects for the development of novel targeted anticancer agents, which ongoing preclinical and clinical experiments define. Our work brings together a comprehensive patent review of the role of hDHODH in cancer, as well as various patents related to the hDHODH inhibitors and their anticancer and other therapeutic potential. This compiled work on patented DHODH inhibitors will guide researchers in pursuing the most promising drug discovery strategies against the hDHODH enzyme as anticancer agents.
Collapse
Affiliation(s)
- Pinky Gehlot
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujrat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujrat, India
| |
Collapse
|
10
|
Sivanesan E, Sanchez KR, Zhang C, He SQ, Linderoth B, Stephens KE, Raja SN, Guan Y. Spinal Cord Stimulation Increases Chemoefficacy and Prevents Paclitaxel-Induced Pain via CX3CL1. Neuromodulation 2023; 26:938-949. [PMID: 37045646 PMCID: PMC10330336 DOI: 10.1016/j.neurom.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/19/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Despite increasing utilization of spinal cord stimulation (SCS), its effects on chemoefficacy, cancer progression, and chemotherapy-induced peripheral neuropathy (CIPN) pain remain unclear. Up to 30% of adults who are cancer survivors may suffer from CIPN, and there are currently no effective preventative treatments. MATERIALS AND METHODS Through a combination of bioluminescent imaging, behavioral, biochemical, and immunohistochemical approaches, we investigated the role of SCS and paclitaxel (PTX) on tumor growth and PTX-induced peripheral neuropathy (PIPN) pain development in T-cell-deficient male rats (Crl:NIH-Foxn1rnu) with xenograft human non-small cell lung cancer. We hypothesized that SCS can prevent CIPN pain and enhance chemoefficacy partially by modulating macrophages, fractalkine (CX3CL1), and inflammatory cytokines. RESULTS We show that preemptive SCS enhanced the antitumor efficacy of PTX and prevented PIPN pain. Without SCS, rats with and without tumors developed robust PIPN pain-related mechanical hypersensitivity, but only those with tumors developed cold hypersensitivity, suggesting T-cell dependence for different PIPN pain modalities. SCS increased soluble CX3CL1 and macrophages and decreased neuronal and nonneuronal insoluble CX3CL1 expression and inflammation in dorsal root ganglia. CONCLUSION Collectively, our findings suggest that preemptive SCS is a promising strategy to increase chemoefficacy and prevent PIPN pain via CX3CL1-macrophage modulation.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Karla R Sanchez
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kimberly E Stephens
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
11
|
Sankowski B, Michorowska S, Raćkowska E, Sikora M, Giebułtowicz J. Saliva as Blood Alternative in Therapeutic Monitoring of Teriflunomide-Development and Validation of the Novel Analytical Method. Int J Mol Sci 2022; 23:ijms23179544. [PMID: 36076939 PMCID: PMC9455247 DOI: 10.3390/ijms23179544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/13/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Therapeutic drug monitoring (TDM) is extremely helpful in individualizing dosage regimen of drugs with narrow therapeutic ranges. It may also be beneficial in the case of drugs characterized by serious side effects and marked interpatient pharmacokinetic variability observed with leflunomide and its biologically active metabolite, teriflunomide. One of the most popular matrices used for TDM is blood. A more readily accessible body fluid is saliva, which can be collected in a much safer way comparing to blood. This makes it especially advantageous alternative to blood during life-threatening SARS-CoV-2 pandemic. However, drug’s saliva concentration is not always a good representation of its blood concentration. The aim of this study was to verify whether saliva can be used in TDM of teriflunomide. We also developed and validated the first reliable and robust LC-MS/MS method for quantification of teriflunomide in saliva. Additionally, the effect of salivary flow and swab absorptive material from the collector device on teriflunomide concentration in saliva was evaluated. Good linear correlation was obtained between the concentration of teriflunomide in plasma and resting saliva (p < 0.000016, r = 0.88), and even better between plasma and the stimulated saliva concentrations (p < 0.000001, r = 0.95) confirming the effectiveness of this non-invasive method of teriflunomide’s TDM. The analyzed validation criteria were fulfilled. No significant influence of salivary flow (p = 0.198) or type of swab in the Salivette device on saliva’s teriflunomide concentration was detected. However, to reduce variability the use of stimulated saliva and synthetic swabs is advised.
Collapse
Affiliation(s)
- Bartłomiej Sankowski
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Sylwia Michorowska
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Emilia Raćkowska
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drug Analysis, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
- Correspondence:
| |
Collapse
|
12
|
Zhu Z, Cao C, Zhang D, Zhang Z, Liu L, Wu D, Sun J. UBE2T-mediated Akt ubiquitination and Akt/β-catenin activation promotes hepatocellular carcinoma development by increasing pyrimidine metabolism. Cell Death Dis 2022; 13:154. [PMID: 35169125 PMCID: PMC8847552 DOI: 10.1038/s41419-022-04596-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 12/16/2021] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
The oncogene protein ubiquitin-conjugating enzyme E2T (UBE2T) is reported to be upregulated in hepatocellular carcinoma (HCC) and correlated with poor clinical outcomes of HCC patients. However, the underlying mechanism by which UBE2T exerts its oncogenic function in HCC remains largely unexplored. In this study, in vitro and in vivo experiments suggested that UBE2T promoted HCC development including proliferation and metastasis. GSEA analysis indicated that UBE2T was positively correlated with pyrimidine metabolism, and LC/MS-MS metabolomics profiling revealed that the key products of pyrimidine metabolism were significantly increased in UBE2T-overexpressing cells. UBE2T overexpression led to the upregulation of several key enzymes catalyzing de novo pyrimidine synthesis, including CAD, DHODH, and UMPS. Moreover, the utilization of leflunomide, a clinically approved DHODH inhibitor, blocked the effect of UBE2T in promoting HCC progression. Mechanistically, UBE2T increased Akt K63-mediated ubiquitination and Akt/β-catenin signaling pathway activation. The disruption of UBE2T-mediated ubiquitination on Akt, including E2-enzyme-deficient mutation (C86A) of UBE2T and ubiquitination-site-deficient mutation (K8/14 R) of Akt impaired UBE2T’s effect in upregulating CAD, DHODH, and UMPS. Importantly, we demonstrated that UBE2T was positively correlated with p-Akt, β-catenin, CAD, DHODH, and UMPS in HCC tumor tissues. In summary, our study indicates that UBE2T increases pyrimidine metabolism by promoting Akt K63-linked ubiquitination, thus contributing to HCC development. This work provides a novel insight into HCC development and a potential therapeutic strategy for HCC patients.
Collapse
|
13
|
Zhang L, Zhang J, Wang J, Ren C, Tang P, Ouyang L, Wang Y. Recent advances of human dihydroorotate dehydrogenase inhibitors for cancer therapy: Current development and future perspectives. Eur J Med Chem 2022; 232:114176. [DOI: 10.1016/j.ejmech.2022.114176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
14
|
Phan T, Nguyen VH, Buettner R, Morales C, Yang L, Wong P, Tsai W, Salazar MD, Gil Z, Diamond DJ, Rabinowitz JD, Rosen S, Melstrom LG. Inhibition of de novo pyrimidine synthesis augments Gemcitabine induced growth inhibition in an immunocompetent model of pancreatic cancer. Int J Biol Sci 2021; 17:2240-2251. [PMID: 34239352 PMCID: PMC8241727 DOI: 10.7150/ijbs.60473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Leflunomide (Lef) is an agent used in autoimmune disorders that interferes with DNA synthesis. De Novo pyrimidine synthesis is a mechanism of Gemcitabine (Gem) resistance in pancreatic cancer. This study aims to assess the efficacy and changes in the tumor microenvironment of Lef monotherapy and in combination with Gem, in a syngeneic mouse model of pancreatic cancer. Methods: MTS proliferation assays were conducted to assess growth inhibition by Gem (0-20 nM), Lef (0-40 uM) and Gem+Lef in KPC (KrasLSL.G12D/+;p53R172H/+; PdxCretg/+) cells in vitro. An in vivo heterotopic KPC model was used and cohorts were treated with: PBS (control), Gem (75 mg/kg/q3d), Lef (40 mg/kg/d), or Gem+Lef. At d28 post-treatment, tumor burden, proliferation index (Ki67), and vascularity (CD31) were measured. Changes in the frequency of peripheral and intratumoral immune cell subsets were evaluated via FACS. Liquid chromatography-mass spectrometry was used for metabolomics profiling. Results: Lef inhibits KPC cell growth and synergizes with Gem in vitro (P<0.05; Combination Index 0.44 (<1 indicates synergy). In vivo, Lef alone and in combination with Gem delays KPC tumor progression (P<0.001). CTLA-4+T cells are also significantly decreased in tumors treated with Lef, Gem or in combination (Gem+Lef) compared to controls (P<0.05). Combination therapy also decreased the Ki67 and vascularity (P<0.01). Leflunomide inhibits de novo pyrimidine synthesis both in vitro (p<0.0001) and in vivo (p<0.05). Conclusions: In this study, we demonstrated that Gem+Lef inhibits pancreatic cancer growth, decrease T cell exhaustion, vascularity and as proof of principle inhibits de novo pyrimidine synthesis. Further characterization of changes in adaptive immunity are necessary to characterize the mechanism of tumor growth inhibition and facilitate translation to a clinical trial.
Collapse
Affiliation(s)
- Thuy Phan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Vu H. Nguyen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Ralf Buettner
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Corey Morales
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Lifeng Yang
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Paul Wong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
| | - Weiman Tsai
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | | | - Ziv Gil
- Rambam Medical Center, Israel
| | - Don J Diamond
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Joshua D. Rabinowitz
- Lewis Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544
| | - Steven Rosen
- Department of Hematology, City of Hope National Medical Center, Duarte, CA 91010
| | - Laleh G. Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010
- ✉ Corresponding author: Laleh Melstrom MD, City of Hope National Medical Center, Department of Surgery and Immuno-oncology, 1500 E Duarte Road, Duarte, CA 91010. E-mail: ; Phone: 626 218 0282; Fax: 626 218 1113
| |
Collapse
|
15
|
Selective Binding of Cyclodextrins with Leflunomide and Its Pharmacologically Active Metabolite Teriflunomide. Int J Mol Sci 2020; 21:ijms21239102. [PMID: 33265979 PMCID: PMC7730839 DOI: 10.3390/ijms21239102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
The selectivity of encapsulation of leflunomide and teriflunomide by native α-, β- and γ-cyclodextrins was investigated through 1H NMR and molecular modeling. Thermodynamic analysis revealed the main driving forces involved in the binding. For α-cyclodextrin, the partial encapsulation was obtained while deep penetration was characterized for the other two cyclodextrins, where the remaining polar fragment of the molecule is located outside the macrocyclic cavity. The interactions via hydrogen bonding are responsible for high negative enthalpy and entropy changes accompanying the complexation of cyclodextrins with teriflunomide. These results were in agreement with the molecular modeling calculations, which provide a clearer picture of the involved interactions at the atomic level.
Collapse
|
16
|
Narayanan D, Ma S, Özcelik D. Targeting the Redox Landscape in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12071706. [PMID: 32605023 PMCID: PMC7407119 DOI: 10.3390/cancers12071706] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are produced predominantly by the mitochondrial electron transport chain and by NADPH oxidases in peroxisomes and in the endoplasmic reticulum. The antioxidative defense counters overproduction of ROS with detoxifying enzymes and molecular scavengers, for instance, superoxide dismutase and glutathione, in order to restore redox homeostasis. Mutations in the redox landscape can induce carcinogenesis, whereas increased ROS production can perpetuate cancer development. Moreover, cancer cells can increase production of antioxidants, leading to resistance against chemo- or radiotherapy. Research has been developing pharmaceuticals to target the redox landscape in cancer. For instance, inhibition of key players in the redox landscape aims to modulate ROS production in order to prevent tumor development or to sensitize cancer cells in radiotherapy. Besides the redox landscape of a single cell, alternative strategies take aim at the multi-cellular level. Extracellular vesicles, such as exosomes, are crucial for the development of the hypoxic tumor microenvironment, and hence are explored as target and as drug delivery systems in cancer therapy. This review summarizes the current pharmaceutical and experimental interventions of the cancer redox landscape.
Collapse
Affiliation(s)
- Dilip Narayanan
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Sana Ma
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Dennis Özcelik
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
- current address: Chemistry | Biology | Pharmacy Information Center, ETH Zürich, Vladimir-Prelog-Weg 10, 8093 Zürich, Switzerland
- Correspondence:
| |
Collapse
|
17
|
Nasal Drug Delivery of Anticancer Drugs for the Treatment of Glioblastoma: Preclinical and Clinical Trials. Molecules 2019; 24:molecules24234312. [PMID: 31779126 PMCID: PMC6930669 DOI: 10.3390/molecules24234312] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal form of brain tumor, being characterized by the rapid growth and invasion of the surrounding tissue. The current standard treatment for glioblastoma is surgery, followed by radiotherapy and concurrent chemotherapy, typically with temozolomide. Although extensive research has been carried out over the past years to develop a more effective therapeutic strategy for the treatment of GBM, efforts have not provided major improvements in terms of the overall survival of patients. Consequently, new therapeutic approaches are urgently needed. Overcoming the blood–brain barrier (BBB) is a major challenge in the development of therapies for central nervous system (CNS) disorders. In this context, the intranasal route of drug administration has been proposed as a non-invasive alternative route for directly targeting the CNS. This route of drug administration bypasses the BBB and reduces the systemic side effects. Recently, several formulations have been developed for further enhancing nose-to-brain transport, mainly with the use of nano-sized and nanostructured drug delivery systems. The focus of this review is to provide an overview of the strategies that have been developed for delivering anticancer compounds for the treatment of GBM while using nasal administration. In particular, the specific properties of nanomedicines proposed for nose-to-brain delivery will be critically evaluated. The preclinical and clinical data considered supporting the idea that nasal delivery of anticancer drugs may represent a breakthrough advancement in the fight against GBM.
Collapse
|
18
|
Hongell K, Kurki S, Sumelahti ML, Soilu-Hänninen M. Risk of cancer among Finnish multiple sclerosis patients. Mult Scler Relat Disord 2019; 35:221-227. [DOI: 10.1016/j.msard.2019.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/29/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022]
|
19
|
Buettner R, Morales C, Wu X, Sanchez JF, Li H, Melstrom LG, Rosen ST. Leflunomide Synergizes with Gemcitabine in Growth Inhibition of PC Cells and Impairs c-Myc Signaling through PIM Kinase Targeting. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:149-158. [PMID: 31211245 PMCID: PMC6562366 DOI: 10.1016/j.omto.2019.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023]
Abstract
The immunosuppressive agent leflunomide has been used in the treatment of over 300,000 patients with rheumatoid arthritis. Its active metabolite, teriflunomide (Ter), directly inhibits dihydroorotate dehydrogenase (DHODH), an enzyme involved in nucleoside synthesis. We report that Ter not only shows in vitro anti-proliferative activity in pancreatic cancer (PC) cells as a single agent but also synergizes with the chemotherapeutic gemcitabine (Gem) in growth inhibition of PC cells. The growth-inhibitory effects of Ter are not solely caused by inhibition of DHODH. Through a kinase screening approach, we identified the PIM-3 serine-threonine kinase as a novel direct target. Subsequent dose-response kinase assays showed that Ter directly inhibited all three PIM family members, with the highest activities against PIM-3 and -1. The PIM-3 kinase was the PIM family member most often associated with PC oncogenesis and was also the kinase inhibited the most by Ter among more than 600 kinases investigated. Ter in PC cells induced changes in phosphorylation and expression of PIM downstream targets, consistent with the effects achieved by overexpression or downregulation of PIM-3. Finally, pharmacological inhibition of PIM proteins not only diminished PC cell proliferation, but also small-molecule pan-PIM and PIM-3 inhibitors synergized with Gem in growth inhibition of PC cells.
Collapse
Affiliation(s)
- Ralf Buettner
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Corey Morales
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, Department of Molecular Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - James F Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hongzhi Li
- Department of Computational Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Laleh G Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Steven T Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
20
|
Madak JT, Bankhead A, Cuthbertson CR, Showalter HD, Neamati N. Revisiting the role of dihydroorotate dehydrogenase as a therapeutic target for cancer. Pharmacol Ther 2018; 195:111-131. [PMID: 30347213 DOI: 10.1016/j.pharmthera.2018.10.012] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Identified as a hallmark of cancer, metabolic reprogramming allows cancer cells to rapidly proliferate, resist chemotherapies, invade, metastasize, and survive a nutrient-deprived microenvironment. Rapidly growing cells depend on sufficient concentrations of nucleotides to sustain proliferation. One enzyme essential for the de novo biosynthesis of pyrimidine-based nucleotides is dihydroorotate dehydrogenase (DHODH), a known therapeutic target for multiple diseases. Brequinar, leflunomide, and teriflunomide, all of which are potent DHODH inhibitors, have been clinically evaluated but failed to receive FDA approval for the treatment of cancer. Inhibition of DHODH depletes intracellular pyrimidine nucleotide pools and results in cell cycle arrest in S-phase, sensitization to current chemotherapies, and differentiation in neural crest cells and acute myeloid leukemia (AML). Furthermore, DHODH is a synthetic lethal susceptibility in several oncogenic backgrounds. Therefore, DHODH-targeted therapy has potential value as part of a combination therapy for the treatment of cancer. In this review, we focus on the de novo pyrimidine biosynthesis pathway as a target for cancer therapy, and in particular, DHODH. In the first part, we provide a comprehensive overview of this pathway and its regulation in cancer. We further describe the relevance of DHODH as a target for cancer therapy using bioinformatic analyses. We then explore the preclinical and clinical results of pharmacological strategies to target the de novo pyrimidine biosynthesis pathway, with an emphasis on DHODH. Finally, we discuss potential strategies to harness DHODH as a target for the treatment of cancer.
Collapse
Affiliation(s)
- Joseph T Madak
- Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Armand Bankhead
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christine R Cuthbertson
- Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Hollis D Showalter
- Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Rogel Cancer Center, Ann Arbor, MI 48109, USA.
| | - Nouri Neamati
- Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Rogel Cancer Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
21
|
Göttle P, Manousi A, Kremer D, Reiche L, Hartung HP, Küry P. Teriflunomide promotes oligodendroglial differentiation and myelination. J Neuroinflammation 2018; 15:76. [PMID: 29534752 PMCID: PMC5851312 DOI: 10.1186/s12974-018-1110-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/28/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a neuroinflammatory autoimmune disease of the central nervous system (CNS) which in most cases initially presents with episodes of transient functional deficits (relapsing-remitting MS; RRMS) and eventually develops into a secondary progressive form (SPMS). Aside from neuroimmunological activities, MS is also characterized by neurodegenerative and regenerative processes. The latter involve the restoration of myelin sheaths-electrically insulating structures which are the primary targets of autoimmune attacks. Spontaneous endogenous remyelination takes place even in the adult CNS and is primarily mediated by activation, recruitment, and differentiation of resident oligodendroglial precursor cells (OPCs). However, the overall efficiency of remyelination is limited and further declines with disease duration and progression. From a therapeutic standpoint, it is therefore key to understand how oligodendroglial maturation can be modulated pharmacologically. Teriflunomide has been approved as a first-line treatment for RRMS in the USA and the European Union. As the active metabolite of leflunomide, an established disease-modifying anti-rheumatic drug, it mainly acts via an inhibition of de novo pyrimidine synthesis exerting a cytostatic effect on proliferating B and T cells. METHODS We investigated teriflunomide-dependent effects on primary rat oligodendroglial homeostasis, proliferation, and differentiation related to cellular processes important for myelin repair hence CNS regeneration in vitro. To this end, several cellular parameters, including specific oligodendroglial maturation markers, in vitro myelination, and p53 family member signaling, were examined by means of gene/protein expression analyses. The rate of myelination was determined using neuron-oligodendrocyte co-cultures. RESULTS Low teriflunomide concentrations resulted in cell cycle exit while higher doses led to decreased cell survival. Short-term teriflunomide pulses can efficiently promote oligodendroglial cell differentiation suggesting that young, immature cells could benefit from such stimulation. In vitro myelination can be boosted by means of an early stimulation window with teriflunomide. p73 signaling is functionally involved in promoting OPC differentiation and myelination. CONCLUSION Our findings indicate a critical window of opportunity during which regenerative oligodendroglial activities including myelination of CNS axons can be stimulated by teriflunomide.
Collapse
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| |
Collapse
|
22
|
Zhang C, Chu M. Leflunomide: A promising drug with good antitumor potential. Biochem Biophys Res Commun 2018; 496:726-730. [PMID: 29357281 DOI: 10.1016/j.bbrc.2018.01.107] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 01/01/2023]
Abstract
Leflunomide, an inhibitor of dihydroorotase dehydrogenase and thereby pyrimidine synthesis, was approved for treatment of rheumatoid arthritis in 1998. During the following years, leflunomide was used in various preclinical studies as a potential cancer treatment; at the same time, more mechanisms underlying the anticancer effect of leflunomide were identified. Thus, leflunomide has been identified as a potent anticancer drug. This article summarizes the mechanisms as well as results of leflunomide in the evolving field of cancer therapy.
Collapse
Affiliation(s)
- Chunying Zhang
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, 246 Xuefu St., Nan Gang District, Harbin, China
| | - Maolin Chu
- Department of Urology, The Second Affiliated Hospital, Harbin Medical University, 246 Xuefu St., Nan Gang District, Harbin, China.
| |
Collapse
|
23
|
Lai Z, Li Z, Liu Y, Yang P, Fang X, Zhang W, Liu B, Chang H, Xu H, Xu Y. Iron-Mediated Synthesis of Isoxazoles from Alkynes: Using Iron(III) Nitrate as a Nitration and Cyclization Reagent. J Org Chem 2017; 83:145-153. [DOI: 10.1021/acs.joc.7b02483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Zhenzhen Lai
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Zhenxing Li
- State
Key Laboratory of Heavy Oil Processing, China University of Petroleum-Beijing, Beijing 102249, P. R. China
| | - Yawei Liu
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Pengkun Yang
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Xiaomin Fang
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Wenkai Zhang
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Baoying Liu
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Haibo Chang
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| | - Hao Xu
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
- State
Key Laboratory of Heavy Oil Processing, China University of Petroleum-Beijing, Beijing 102249, P. R. China
| | - Yuanqing Xu
- College
of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, P. R. China
| |
Collapse
|