Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 147: 107412, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696845

ABSTRACT

The development of novel topoisomerase I (TOP1) inhibitors is crucial for overcoming the drawbacks and limitations of current TOP1 poisons. Here, we identified two potential TOP1 inhibitors, namely, FTY720 (a sphingosine 1-phosphate antagonist) and COH29 (a ribonucleotide reductase inhibitor), through experimental screening of known active compounds. Biological experiments verified that FTY720 and COH29 were nonintercalative TOP1 catalytic inhibitors that did not induce the formation of DNA-TOP1 covalent complexes. Molecular docking revealed that FTY720 and COH29 interacted favorably with TOP1. Molecular dynamics simulations revealed that FTY720 and COH29 could affect the catalytic domain of TOP1, thus resulting in altered DNA-binding cavity size. The alanine scanning and interaction entropy identified Arg536 as a hotspot residue. In addition, the bioinformatics analysis predicted that FTY720 and COH29 could be effective in treating malignant breast tumors. Biological experiments verified their antitumor activities using MCF-7 breast cancer cells. Their combinatory effects with TOP1 poisons were also investigated. Further, FTY720 and COH29 were found to cause less DNA damage compared with TOP1 poisons. The findings provide reliable lead compounds for the development of novel TOP1 catalytic inhibitors and offer new insights into the potential clinical applications of FTY720 and COH29 in targeting TOP1.


Subject(s)
Antineoplastic Agents , DNA Topoisomerases, Type I , Fingolimod Hydrochloride , Molecular Docking Simulation , Topoisomerase I Inhibitors , Humans , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/chemical synthesis , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/chemistry , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/chemical synthesis , Molecular Structure , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Molecular Dynamics Simulation , MCF-7 Cells
2.
J Antibiot (Tokyo) ; 73(10): 666-678, 2020 10.
Article in English | MEDLINE | ID: mdl-32681100

ABSTRACT

Fingolimod is a first-in-class of sphingosine-1-phosphate (S1P) receptor modulator and is widely used a therapeutic drug for multiple sclerosis (MS), autoimmune disease in the central nervous system. About 25 year ago, a natural product, myriocin was isolated from culture broths of the fungus Isaria sinclairii. Myriocin, a rather complex amino acid having three successive asymmetric centers, was found to show a potent immunosuppressive activity in vitro; however, it induced a strong toxicity in vivo. To find out a less toxic immunosuppressive candidate, the chemical structure of myriocin was simplified to a nonchiral symmetric 2-substituted-2-aminoproane-1,3-diol framework. Finally, a highly potent immunosuppressant, fingolimod was found by the extensive chemical modification and pharmacological evaluation using skin allograft model in vivo. Throughout the analyses of the mechanism action of fingolimod, it is revealed that S1P receptor 1 (S1P1) plays an essential role in lymphocyte circulation and that the molecular target of fingolimod is S1P1. Phosphorylated fingolimod acts as a "functional" antagonist at S1P1, modulates lymphocyte circulation, and shows a potent immunosuppressive activity. Fingolimod significantly reduced the relapse rate of MS in the clinical studies and has been approved as a new therapeutic drug for MS in more than 80 countries.


Subject(s)
Fingolimod Hydrochloride/chemical synthesis , Hypocreales/chemistry , Animals , Drug Discovery , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/isolation & purification , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Lymphocytes/drug effects , Multiple Sclerosis/drug therapy , Structure-Activity Relationship
3.
J Mater Chem B ; 8(28): 6148-6158, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32568342

ABSTRACT

The addition of osteoimmunology drugs to bone repair materials is beneficial to bone regeneration by regulating the local immune microenvironment. Fingolimod (FTY720) has been reported to be an osteoimmunology drug that promotes osteogenesis. However, there is no ideal biomaterial for the sustained release of FTY720 in the bone defect areas. In the present work, FTY720 loaded mesoporous bioactive glass (FTY720@MBGs) was successfully prepared based on the mesoporous properties of MBGs and electrostatic attraction. FTY720 achieved a sustained release for 7 days. The in vitro study found that FTY720@MBGs could synergistically promote osteogenesis and inhibit osteoclastogenesis due to their ability to promote macrophages toward the M2 phenotype. The in vivo study confirmed that FTY720@MBGs could significantly improve bone regeneration. This study provides new strategies for designing smart cell-instructive biomaterials that can play a role in all bone healing processes from early inflammation to bone reconstruction.


Subject(s)
Bone Regeneration/drug effects , Drug Delivery Systems , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Osteoclasts/drug effects , Osteogenesis/drug effects , Animals , Cells, Cultured , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/chemistry , Glass/chemistry , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Macrophages/drug effects , Mesenchymal Stem Cells/drug effects , Mice , Particle Size , Porosity , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Surface Properties
4.
Chem Pharm Bull (Tokyo) ; 66(10): 1015-1018, 2018.
Article in English | MEDLINE | ID: mdl-30270236

ABSTRACT

FTY720 is employed for the treatment of multiple sclerosis and exerts apoptotic effects on various cancers through protein phosphatase 2A (PP2A) activation. In compound 4, the dihydroxy head group of FTY720 was modified into dihydroxy phenyl group. The cell survival in compound 4 treated colorectal and gastric cancer cells was significantly reduced as compared with control, 34.6 and 25.1%, respectively. The docking study of compound 4 showed that the aromatic head group effectively binds to PP2A.


Subject(s)
Antineoplastic Agents/pharmacology , Fingolimod Hydrochloride/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Molecules ; 23(11)2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30355990

ABSTRACT

FTY720 inhibits various cancers through PP2A activation. The structure of FTY720 is also used as a basic structure for the design of sphingosine kinase (SK) inhibitors. We have synthesized derivatives using an amide chain in FTY720 with a phenyl backbone, and then compounds were screened by an MTT cell viability assay. The PP2A activity of compound 7 was examined. The phosphorylation levels of AKT and ERK, downstream targets of PP2A, in the presence of compound 7, were determined. Compound 7 may exhibit anticancer effects through PP2A activation rather than the mechanism by inhibition of SK1 in cancer cells. In the docking study of compound 7 and PP2A, the amide chain of compound 7 showed an interaction with Asn61 that was different from FTY720, which is expected to affect the activity of the compound.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Molecular Structure , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
6.
Chem Commun (Camb) ; 54(85): 12002-12005, 2018 Oct 23.
Article in English | MEDLINE | ID: mdl-30221278

ABSTRACT

The direct, catalytic vicinal difluorination of terminal alkenes via an I(i)/I(iii) manifold was exploited to install a chiral, hybrid bioisostere of the CF3 and Et groups (BITE) in Gilenya®; the first orally available drug for the clinical management of Multiple Sclerosis (MS). This subtle fluorination pattern allows lipophilicity (log D) to be tempered compared to the corresponding CF3 and Et derivatives (CH2CH3 > CH2CF3 > CHFCH2F).


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/chemistry , Immunosuppressive Agents/chemistry , Animals , Drug Stability , Fingolimod Hydrochloride/chemical synthesis , Halogenation , Hepatocytes/drug effects , Humans , Immunosuppressive Agents/chemical synthesis , Microsomes, Liver/metabolism , Multiple Sclerosis/drug therapy , Rats
7.
Bioorg Med Chem ; 25(2): 483-495, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27913115

ABSTRACT

Multiple sclerosis (MS) is a very common disease of vital importance. In the MS treatment, some drugs such as fingolimod which help to protect nerves from damage are used. The main goal of the drug therapy in MS is to take control of the inflammation which leads to the destruction of myelin and axons in nerve cell and thus prevent and stop the progression of the disease. Fingolimod (FTY720) is an orally active immunomodulatory drug that has been used for the treatment of relapsing-remitting multiple sclerosis. It is a sphingosine-1-phosphate receptor modulator which prevents lymphocytes from contributing to an autoimmune reaction by inhibiting egress of lymphocytes them from lymph nodes. In this study, we have computer designed, synthesized and characterized two novel derivatives of FTY720, F1-12h and F2-9, and have determined their underlying mechanism of their beneficial effect in SH-SY5Y, SK-N-SH, and U-118 MG cell lines. For this purpose, we first determined the regulation of the cAMP response element (CRE) activity and cAMP concentration by F1-12h and F2-9 together with FTY720 using pGL4.29 luciferase reporter assay and cAMP immunoassay, respectively. Then, we have determined their effect on MS- and GPCR-related gene expression profiles using custom arrays along with FTY720 treatment at non-toxic doses (EC10). It was found that both derivatives significantly activate CRE and increase cAMP concentration in all three cell lines, indicating that they activate cAMP pathway through cell surface receptors as FTY720 does. Furthermore, F1-12h and F2-9 modulate the expression of the pathway related genes that are important in inflammatory signaling, cAMP signaling pathway, cell migration as well as diverse receptor and transcription factors. Expression of the genes involved in myelination was also increased by the treatment with F1-12h and F2-9. In summary, our data demonstrate that the two novel FTY720 derivatives act as anti-inflammatory ultimately by influencing the gene expression via the cAMP and downstream transcription factor CRE pathway. In conclusion, F1-12h and F2-9 might contribute future therapies for autoimmune diseases such as multiple sclerosis.


Subject(s)
Computer-Aided Design , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Multiple Sclerosis/drug therapy , Cell Survival/drug effects , Dose-Response Relationship, Drug , Fingolimod Hydrochloride/therapeutic use , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Org Biomol Chem ; 14(20): 4605-16, 2016 May 18.
Article in English | MEDLINE | ID: mdl-27102578

ABSTRACT

AAL(S), the chiral deoxy analog of the FDA approved drug FTY720, has been shown to inhibit proliferation and apoptosis in several cancer cell lines. It has been suggested that it does this by activating protein phosphatase 2A (PP2A). Here we report the synthesis of new cytotoxic analogs of AAL(S) and the evaluation of their cytotoxicity in two myeloid cell lines, one of which is sensitive to PP2A activation. We show that these analogs activate PP2A in these cells supporting the suggested mechanism for their cytotoxic properties. Our findings identify key structural motifs required for anti-cancer effects.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Protein Phosphatase 2/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Enzyme Activation/drug effects , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/therapeutic use , Leukemia, Myeloid, Acute/enzymology
9.
Org Biomol Chem ; 13(48): 11593-6, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26535908

ABSTRACT

A convergent synthesis to access hydrophobic tail analogs and head group modifications of AAL(S) is described. The analogs synthesised were evaluated for their ability to inhibit ceramide synthase 1 and for their cytotoxicity in K562 cells. Our results have identified inhibitors which are non-cytotoxic yet maintain CerS1 inhibition.


Subject(s)
Amino Alcohols/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride/chemical synthesis , Oxidoreductases/antagonists & inhibitors , Amino Alcohols/chemistry , Amino Alcohols/pharmacology , Cell Survival/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , K562 Cells , Models, Biological , Molecular Structure
10.
Bioorg Med Chem ; 23(5): 1011-26, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25656338

ABSTRACT

Sphingosine-1-phosphate (S1P) influences various fundamental biological processes by interacting with a family of five G protein-coupled receptors (S1P1-5). FTY720, a sphingosine analogue, which was approved for treatment of relapsing forms of multiple sclerosis, is phosphorylated in vivo and acts as an agonist of four of the five S1P receptor subtypes. Starting from these lead structures we developed new agonists for the S1P1 receptor. The biological activity was tested in vivo and promising ligands were fluorinated at different positions to identify candidates for positron emission tomography (PET) imaging after [(18)F]-labelling. The radioligands shall enable the imaging of S1P1 receptor expression in vivo and thus may serve as novel imaging markers of S1P-related diseases.


Subject(s)
Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Receptors, Lysosphingolipid/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Fingolimod Hydrochloride/chemistry , Humans , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Ligands , Mice , Receptors, Lysosphingolipid/metabolism
11.
Yao Xue Xue Bao ; 49(6): 896-904, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-25212038

ABSTRACT

A novel series of fingolimod analogues containing diphenyl ether moiety were designed and synthesized based on the modification of immunosuppressive agent fingolimod used in the treatment of multiple sclerosis. Compounds were evaluated in vivo for lymphopenic activity and heart rate affection. Most compounds showed moderate lymphopenic activity. It is worth noting that compounds 6c, 6d and 14c-14e showed considerable immunosuppressive activities comparable to fingolimod. And compound 14e had no effect on heart rate.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Animals , Fingolimod Hydrochloride/chemical synthesis , Heart Rate/drug effects , Immunosuppressive Agents/chemistry , Lymphopenia/pathology , Phenyl Ethers/chemistry , Structure-Activity Relationship
12.
J Pharmacol Exp Ther ; 331(1): 54-64, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19592667

ABSTRACT

Novel therapies are needed to address the vascular endothelial cell (EC) barrier disruption that occurs in inflammatory diseases such as acute lung injury (ALI). We previously demonstrated the potent barrier-enhancing effects of both sphingosine 1-phosphate (S1P) and the structurally similar compound FTY720 [2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] in inflammatory lung injury. In this study, we examined the therapeutic potential of several novel FTY720 analogs to reduce vascular leak. Similar to S1P and FTY720, the (R)- and (S)-enantiomers of FTY720 phosphonate and enephosphonate analogs produce sustained EC barrier enhancement in vitro, as seen by increases in transendothelial electrical resistance (TER). In contrast, the (R)- and (S)-enantiomers of FTY720-regioisomeric analogs disrupt EC barrier integrity in a dose-dependent manner. Barrier-enhancing FTY720 analogs demonstrate a wider protective concentration range in vitro (1-50 microM) and greater potency than either S1P or FTY720. In contrast to FTY720-induced EC barrier enhancement, S1P and the FTY720 analogs dramatically increase TER within minutes in association with cortical actin ring formation. Unlike S1P, these FTY720 analogs exhibit differential phosphorylation effects without altering the intracellular calcium level. Inhibitor studies indicate that barrier enhancement by these analogs involves signaling via G(i)-coupled receptors, tyrosine kinases, and lipid rafts. Consistent with these in vitro responses, the (S)-phosphonate analog of FTY720 significantly reduces multiple indices of alveolar and vascular permeability in a lipopolysaccharide-mediated murine model of ALI (without significant alterations in leukocyte counts). These results demonstrate the capacity for FTY720 analogs to significantly decrease pulmonary vascular leakage and inflammation in vitro and in vivo.


Subject(s)
Capillary Permeability/drug effects , Capillary Permeability/physiology , Fingolimod Hydrochloride/analogs & derivatives , Inflammation Mediators/chemical synthesis , Inflammation Mediators/pharmacology , Organophosphonates/chemical synthesis , Organophosphonates/pharmacology , Propylene Glycols/chemical synthesis , Propylene Glycols/pharmacology , Pulmonary Artery/drug effects , Sphingosine/analogs & derivatives , Animals , Cell Line , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Humans , Lung/blood supply , Lung/drug effects , Lung/pathology , Mice , Mice, Inbred C57BL , Pulmonary Artery/pathology , Sphingosine/chemical synthesis , Sphingosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...