Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(4): 2566-2588, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36749735

ABSTRACT

The development of orally bioavailable, furanopyrimidine-based double-mutant (L858R/T790M) EGFR inhibitors is described. First, selectivity for mutant EGFR was accomplished by replacing the (S)-2-phenylglycinol moiety of 12 with either an ethanol or an alkyl substituent. Then, the cellular potency and physicochemical properties were optimized through insights from molecular modeling studies by implanting various solubilizing groups in phenyl rings A and B. Optimized lead 52 shows 8-fold selective inhibition of H1975 (EGFRL858R/T790M overexpressing) cancer cells over A431 (EGFRWT overexpressing) cancer cells; western blot analysis further confirmed EGFR mutant-selective target modulation inside the cancer cells by 52. Notably, 52 displayed in vivo antitumor effects in two different mouse xenograft models (BaF3 transfected with mutant EGFR and H1975 tumors) with TGI = 74.9 and 97.5% after oral administration (F = 27%), respectively. With an extraordinary kinome selectivity (S(10) score of 0.017), 52 undergoes detailed preclinical development.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Lung Neoplasms , Protein Kinase Inhibitors , Pyrimidines , Animals , Humans , Mice , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Mutation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Pyrimidines/administration & dosage , Pyrimidines/pharmacology
2.
Int J Mol Sci ; 23(19)2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36233031

ABSTRACT

CXCR4 antagonists have been claimed to reduce mortality after myocardial infarction in myocardial infarction (MI) animals, presumably due to suppressing inflammatory responses caused by myocardial ischemia-reperfusion injury, thus, subsequently facilitating tissue repair and cardiac function recovery. This study aims to determine whether a newly designed CXCR4 antagonist DBPR807 could exert better vascular-protective effects than other clinical counterparts (e.g., AMD3100) to alleviate cardiac damage further exacerbated by reperfusion. Consequently, we find that instead of traditional continuous treatment or multiple-dose treatment at different intervals of time, a single-dose treatment of DBPR807 before reperfusion in MI animals could attenuate inflammation via protecting oxidative stress damage and preserve vascular/capillary density and integrity via mobilizing endothelial progenitor cells, leading to a desirable fibrosis reduction and recovery of cardiac function, as evaluated with the LVEF (left ventricular ejection fraction) in infarcted hearts in rats and mini-pigs, respectively. Thus, it is highly suggested that CXCR4 antagonists should be given at a single high dose prior to reperfusion to provide the maximal cardiac functional improvement. Based on its favorable efficacy and safety profiles indicated in tested animals, DBPR807 has a great potential to serve as an adjunctive medicine for percutaneous coronary intervention (PCI) therapies in acute MI patients.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Percutaneous Coronary Intervention , Receptors, CXCR4 , Animals , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/etiology , Rats , Receptors, CXCR4/antagonists & inhibitors , Stroke Volume , Swine , Swine, Miniature , Ventricular Function, Left
3.
J Med Chem ; 65(6): 4767-4782, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35234475

ABSTRACT

Chemotherapy-induced neurotoxicity is a common adverse effect of cancer treatment. No medication has been shown to be effective in the prevention or treatment of chemotherapy-induced neurotoxicity. Using minoxidil as an initial template for structural modifications in conjunction with an in vitro neurite outgrowth assay, an image-based high-content screening platform, and mouse behavior models, an effective neuroprotective agent CN016 was discovered. Our results showed that CN016 could inhibit paclitaxel-induced inflammatory responses and infiltration of immune cells into sensory neurons significantly. Thus, the suppression of proinflammatory factors elucidates, in part, the mechanism of action of CN016 on alleviating paclitaxel-induced peripheral neuropathy. Based on excellent efficacy in improving behavioral functions, high safety profiles (MTD > 500 mg/kg), and a large therapeutic window (MTD/MED > 50) in mice, CN016 might have great potential to become a peripherally neuroprotective agent to prevent neurotoxicity caused by chemotherapeutics as typified by paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic , Antineoplastic Agents , Neuroprotective Agents , Peripheral Nervous System Diseases , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/toxicity , Ganglia, Spinal , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control
4.
Life Sci ; 278: 119574, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33961850

ABSTRACT

AIMS: Dipeptidyl peptidase 4 (DPP-4) is a valid molecular drug target from which its inhibitors have been developed as medicines for treating diabetes. The present study evaluated a new synthetic DPP-4-specific inhibitor of small molecule DBPR108 for pharmacology and pharmacokinetic profiles. MAIN METHODS: DBPR108 of various doses was orally administered to rats, diabetic mice, and dogs and the systemic circulating DPP-4 activities in the animals were measured to demonstrate the pharmacological mechanisms of action via DPP-4 inhibition. Upon an oral administration of DBPR108, the serum active GLP-1 and insulin levels of the rats challenged with an oral glucose ingestion were measured. Oral glucose tolerance test in diet-induced obese mice was performed to examine if DBPR108 increases the glucose tolerability in animals. KEY FINDINGS: Orally administered DBPR108 inhibited the systemic plasma DPP-4 activities in rats, dogs and diabetic mice in a dose-dependent manner. DBPR108 caused elevated serum levels of active GLP-1 and insulin in the rats. DBPR108 dose-dependently increased the glucose tolerability in diet-induced obese (DIO) mice and, furthermore, DIO mice treated with DBPR108 (0.1 mg/kg) in combination with metformin (50 or 100 mg/kg) showed a prominently strong increase in the glucose tolerability. SIGNIFICANCE: DBPR108 is a novel DPP-4-selective inhibitor of small molecule that demonstrated potent in vivo pharmacological effects and good safety profiles in animals. DBPR108 is now a drug candidate being further developed in the clinical studies as therapeutics for treating diabetes.


Subject(s)
Butanes/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hyperglycemia/drug therapy , Hypoglycemic Agents/pharmacology , Nitriles/pharmacology , Pyrrolidines/pharmacology , Administration, Oral , Animals , Area Under Curve , Body Weight , Butanes/pharmacokinetics , Diabetes Mellitus, Experimental/drug therapy , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dogs , Glucose Tolerance Test , Hypoglycemic Agents/pharmacokinetics , Insulin/metabolism , Jugular Veins/pathology , Male , Metformin , Mice , Mice, Inbred C57BL , Mice, Obese , Nitriles/pharmacokinetics , Pyrrolidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Species Specificity
5.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753481

ABSTRACT

The CXC chemokine receptor type 4 (CXCR4) receptor and its ligand, CXCL12, are overexpressed in various cancers and mediate tumor progression and hypoxia-mediated resistance to cancer therapy. While CXCR4 antagonists have potential anticancer effects when combined with conventional anticancer drugs, their poor potency against CXCL12/CXCR4 downstream signaling pathways and systemic toxicity had precluded clinical application. Herein, BPRCX807, known as a safe, selective, and potent CXCR4 antagonist, has been designed and experimentally realized. In in vitro and in vivo hepatocellular carcinoma mouse models it can significantly suppress primary tumor growth, prevent distant metastasis/cell migration, reduce angiogenesis, and normalize the immunosuppressive tumor microenvironment by reducing tumor-associated macrophages (TAMs) infiltration, reprogramming TAMs toward an immunostimulatory phenotype and promoting cytotoxic T cell infiltration into tumor. Although BPRCX807 treatment alone prolongs overall survival as effectively as both marketed sorafenib and anti-PD-1, it could synergize with either of them in combination therapy to further extend life expectancy and suppress distant metastasis more significantly.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Drug Synergism , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Molecular Docking Simulation , Rats , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Sorafenib/pharmacology , Sorafenib/therapeutic use , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Xenograft Model Antitumor Assays
6.
Pharmacol Res ; 161: 105183, 2020 11.
Article in English | MEDLINE | ID: mdl-32896579

ABSTRACT

Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related death worldwide. NSCLC patients with overexpressed or mutated epidermal growth factor receptor (EGFR) related to disease progression are treated with EGFR-tyrosine kinase inhibitors (EGFR-TKIs). Acquired drug resistance after TKI treatments has been a major focus for development of NSCLC therapies. This study aimed to establish afatinib-resistant cell lines from which afatinib resistance-associated genes are identified and the underlying mechanisms of multiple-TKI resistance in NSCLC can be further investigated. Nude mice bearing subcutaneous NSCLC HCC827 tumors were administered with afatinib at different dose intensities (5-100 mg/kg). We established three HCC827 sublines resistant to afatinib (IC50 > 1 µM) with cross-resistance to gefitinib (IC50 > 5 µM). cDNA microarray revealed several of these sublines shared 27 up- and 13 down-regulated genes. The mRNA expression of selective novel genes - such as transmembrane 4 L six family member 19 (TM4SF19), suppressor of cytokine signaling 2 (SOCS2), and quinolinate phosphoribosyltransferase (QPRT) - are responsive to afatinib treatments only at high concentrations. Furthermore, c-MET amplification and activations of a subset of tyrosine kinase receptors were observed in all three resistant cells. PHA665752, a c-MET inhibitor, remarkably increased the sensitivity of these resistant cells to afatinib (IC50 = 12-123 nM). We established afatinib-resistant lung cancer cell lines and here report genes associated with afatinib resistance in human NSCLC. These cell lines and the identified genes serve as useful investigational tools, prognostic biomarkers of TKI therapies, and promising molecule targets for development of human NSCLC therapeutics.


Subject(s)
Afatinib/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Nude , Oligonucleotide Array Sequence Analysis , Transcriptome , Xenograft Model Antitumor Assays
7.
J Med Chem ; 62(22): 10108-10123, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31560541

ABSTRACT

Epidermal growth factor receptor (EGFR)-targeted therapy in non-small cell lung cancer represents a breakthrough in the field of precision medicine. Previously, we have identified a lead compound, furanopyrimidine 2, which contains a (S)-2-phenylglycinol structure as a key fragment to inhibit EGFR. However, compound 2 showed high clearance and poor oral bioavailability in its pharmacokinetics studies. In this work, we optimized compound 2 by scaffold hopping and exploiting the potent inhibitory activity of various warhead groups to obtain a clinical candidate, 78 (DBPR112), which not only displayed a potent inhibitory activity against EGFRL858R/T790M double mutations but also exhibited tenfold potency better than the third-generation inhibitor, osimertinib, against EGFR and HER2 exon 20 insertion mutations. Overall, pharmacokinetic improvement through lead-to-candidate optimization yielded fourfold oral AUC better that afatinib along with F = 41.5%, an encouraging safety profile, and significant antitumor efficacy in in vivo xenograft models. DBPR112 is currently undergoing phase 1 clinical trial in Taiwan.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Exons , Humans , Male , Mice, Inbred ICR , Mice, Nude , Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Pyrimidines/chemistry , Rats , Receptor, ErbB-2 , Structure-Activity Relationship , Xenograft Model Antitumor Assays
8.
J Med Chem ; 61(3): 818-833, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29314840

ABSTRACT

The function of the CXCR4/CXCL12 axis accounts for many disease indications, including tissue/nerve regeneration, cancer metastasis, and inflammation. Blocking CXCR4 signaling with its antagonists may lead to moving out CXCR4+ cell types from bone marrow to peripheral circulation. We have discovered a novel series of pyrimidine-based CXCR4 antagonists, a representative (i.e., 16) of which was tolerated at a higher dose and showed better HSC-mobilizing ability at the maximal response dose relative to the approved drug 1 (AMD3100), and thus considered a potential drug candidate for PBSCT indication. Docking compound 16 into the X-ray crystal structure of CXCR4 receptor revealed that it adopted a spider-like conformation striding over both major and minor subpockets. This putative binding mode provides a new insight into CXCR4 receptor-ligand interactions for further structural modifications.


Subject(s)
Peripheral Blood Stem Cell Transplantation , Receptors, CXCR4/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Animals , Benzylamines , Cyclams , HEK293 Cells , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , Male , Mice , Molecular Docking Simulation , Protein Conformation , Receptors, CXCR4/chemistry
9.
Cell Transplant ; 26(4): 571-583, 2017 04 13.
Article in English | MEDLINE | ID: mdl-27938478

ABSTRACT

C-X-C chemokine receptor type 4 (CXCR4) is a receptor for a pleiotropic chemokine CXCL12. Previous studies have shown that the acute administration of the CXCR4 antagonist AMD3100 reduced neuroinflammation in stroke brain and mobilized bone marrow hematopoietic stem cells (HSCs). The purpose of this study was to characterize the neuroprotective and neurotrophic effect of a novel CXCR4 antagonist CX549. We demonstrated that CX549 had a higher affinity for CXCR4 and was more potent than AMD3100 to inhibit CXCL12-mediated chemotaxis in culture. CX549 effectively reduced the activation of microglia and improved neuronal survival after injury in neuron/microglia cocultures. Early poststroke treatment with CX549 significantly improved behavioral function, reduced brain infarction, and suppressed the expression of inflammatory markers. Compared to AMD3100, CX549 has a higher affinity for CXCR4, is more efficient to mobilize HSCs for transplantation, and induces behavioral improvement. Our data support that CX549 is a potent anti-inflammatory agent, is neuroprotective against ischemic brain injury, and may have clinical implications for the treatment of stroke.


Subject(s)
Brain/pathology , Neuroprotection , Neuroprotective Agents/therapeutic use , Quinazolines/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Stroke/drug therapy , Triazoles/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Behavior, Animal/drug effects , Benzylamines , Brain/drug effects , Brain Infarction/complications , Brain Infarction/drug therapy , Brain Infarction/pathology , Chemotaxis/drug effects , Cyclams , HEK293 Cells , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Humans , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Quinazolines/pharmacology , Rats, Sprague-Dawley , Receptors, CXCR4/metabolism , Stroke/complications , Stroke/pathology , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism
10.
J Med Chem ; 58(5): 2315-25, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25686267

ABSTRACT

We have discovered a novel series of quinazoline-based CXCR4 antagonists. Of these, compound 19 mobilized CXCR4(+) cell types, including hematopoietic stem cells and endothelial progenitor cells, more efficiently than the marketed 1 (AMD3100) with subcutaneous administration at the same dose (6 mg/kg) in mice. This series of compounds thus provides a set of valuable tools to study diseases mediated by the CXCR4/SDF-1 axis, including myocardial infarction, ischemic stroke, and cancer metastasis. More importantly, treatment with compound 19 significantly lowered levels of blood urea nitrogen and serum creatinine in rats with renal ischemia-reperfusion injury, providing evidence for its therapeutic potential in preventing ischemic acute kidney injury. CXCR4 antagonists such as 19 might also be useful to increase circulating levels of adult stem cells, thereby exerting beneficial effects on damaged and/or inflamed tissues in diseases that currently are not treated by standard approaches.


Subject(s)
Acute Kidney Injury/prevention & control , Chemotaxis/drug effects , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Quinazolines/chemistry , Quinazolines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Reperfusion Injury/prevention & control , Triazoles/chemistry , Triazoles/pharmacology , Animals , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Signal Transduction
12.
J Med Chem ; 53(20): 7316-26, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20961149

ABSTRACT

HTS hit 7 was modified through hybrid design strategy to introduce a chiral side chain followed by introduction of Michael acceptor group to obtain potent EGFR kinase inhibitors 11 and 19. Both 11 and 19 showed over 3 orders of magnitude enhanced HCC827 antiproliferative activity compared to HTS hit 7 and also inhibited gefitinib-resistant double mutant (DM, T790M/L858R) EGFR kinase at nanomolar concentration. Moreover, treatment with 19 shrinked tumor in nude mice xenograft model.


Subject(s)
Antineoplastic Agents/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Drug Design , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , ErbB Receptors/genetics , Gefitinib , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Male , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
13.
Anticancer Res ; 30(7): 2813-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683017

ABSTRACT

BACKGROUND: Antimitotic tubulin-binding BPR0L075 is structurally analogous to the vascular-disrupting combretastatin A-4. MATERIALS AND METHODS: In vitro/in vivo models of endothelial cells cultures, Matrigel plug assay, tumor-bearing nude mice, and murine leukemia cells-inoculated mice were utilized to evaluate BPR0L075 for antiangiogenic and antitumoral activity spectra. RESULTS: BPR0L075 concentration-dependently inhibited proliferation and migration of human umbilical vein endothelial cells (HUVECs), disrupted capillary tube formations of HUVECs and rat aorta endothelial cells, and suppressed in vivo VEGF-mediated angiogenesis in Matrigel plugs in mice. Besides inhibiting the colony growth of cancer cells, BPR0L075 suppressed growth of subcutaneously-xenografted human lung, colorectal, and cervical solid tumors in nude mice. Combination treatments of BPR0L075 plus cisplatin, compared to either agent alone, demonstrated a stronger growth inhibition against the tumor xenografts in nude mice and longer lifespan in the leukemia mice. CONCLUSION: BPR0L075 is an antitumoral and antiangiogenic agent and potentiates the anticancer activity of cisplatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Growth Processes/drug effects , Cisplatin/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Endothelial Cells/drug effects , Female , HCT116 Cells , Humans , Indoles/administration & dosage , Leukemia P388/drug therapy , Male , Mice , Mice, Inbred DBA , Mice, Nude , Neoplasms/pathology , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 20(12): 3596-600, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483603

ABSTRACT

A series of 2-[3-[2-[(2S)-2-cyano-1-pyrrolidinyl]-2-oxoethylamino]-3-methyl-1-oxobutyl]-based DPP-IV inhibitors with various monocyclic amines were synthesized. The structure-activity relationships (SAR) led to the discovery of potent DPP-IV inhibitors, having IC(50) values of <100 nM with excellent selectivity over the closely related enzymes, DPP-II, DPP8, DPP9 and FAP (IC(50)>20 microM). Of these compounds, the analogues 12a, 12h and 12i exhibited a long-lasting ex vivo DPP-IV inhibition in rats.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Nitriles/chemical synthesis , Pyrrolidines/chemical synthesis , Administration, Oral , Amines , Animals , Biological Availability , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Inhibitory Concentration 50 , Nitriles/pharmacology , Pyrrolidines/pharmacology , Rats , Structure-Activity Relationship
15.
Cancer Chemother Pharmacol ; 66(4): 773-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20044750

ABSTRACT

PURPOSE: Gastric cancer is one of the leading cancerous diseases worldwide. It is diagnosed often at the advanced stage for which chemotherapy is the main treatment option. The prognosis remains poor for metastatic, especially the diffuse type, gastric cancers. We investigated the efficacy of intravenously administered paclitaxel treating metastases of locally disseminated gastric tumors of diffuse type. METHODS: Transfection of green fluorescent proteins (GFP)-expressing plasmid into human gastric cancer MKN45 cells of diffuse type was performed, and MKN45-GFP cells constitutively expressing GFP were isolated. The MKN45-GFP cells were orthotopically inoculated into the mouse peritoneal cavity, and tumor growth and organ metastases were monitored. Liver metastases were harvested, re-inoculated, monitored for liver metastases again, and harvested for further inoculation. This in vivo selection procedure was repeated to isolate a subline with high metastatic abilities demonstrated by in vitro invasion abilities using Transwell((R)) system. By visualizing the GFP-expressing tumors, the effects of intravenously administered paclitaxel against the growing peritoneally disseminated and metastasized tumors in nude mice without laparotomy were measured. RESULTS: An in vivo selected gastric cancer cell line MKN45-GFP-ip4 with high metastatic ability was established. Its invasion ability was inhibited by paclitaxel treatments in vitro. The growths of metastatic and intraperitoneally disseminated MKN45-GFP-ip4 tumors were significantly suppressed by intravenous paclitaxel treatments in nude mice. CONCLUSIONS: We found that intravenous paclitaxel is active against the metastases of human gastric cancer of peritoneal diffuse type, which warrants further investigations on optimizing the perioperative regimens with intravenous paclitaxel therapy for gastric cancer in patients.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Paclitaxel/therapeutic use , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Green Fluorescent Proteins/metabolism , Humans , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Paclitaxel/administration & dosage , Peritoneum/pathology , Stomach Neoplasms/pathology , Transfection
16.
Bioorg Med Chem ; 17(6): 2388-99, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19261480

ABSTRACT

A series of trans-2-aryl-cyclopropylamine derived compounds were synthesized and evaluated their biological activities against DPP-IV. The structure-activity relationships (SAR) led to the discovery of novel series of DPP-IV inhibitors, having IC(50) values of <100 nM with excellent selectivity over the closely related enzymes, DPP8, DPP-II and FAP. The studies identified a potent and selective DPP-IV inhibitor 24b, which exhibited the ability to both significantly inhibit plasma DPP-IV activity in rats and improve glucose tolerance in lean mice and diet induced obese mice.


Subject(s)
Cyclopropanes/chemistry , Cyclopropanes/pharmacology , Dipeptidyl-Peptidase IV Inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Animals , Dipeptidyl Peptidase 4/blood , Glucose Tolerance Test , Magnetic Resonance Spectroscopy , Mice , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 19(7): 1908-12, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19269819

ABSTRACT

A series of (2S)-cyanopyrrolidines with glutamic acid derivatives at the P2 site have been prepared and evaluated as inhibitors of dipeptidyl peptidase IV (DPP-IV). The structure-activity relationships (SAR) led to the discovery of potent 3-substituted glutamic acid analogues, providing enhanced chemical stability and excellent selectivity over the closely related enzymes, DPP8, DPP-II and FAP. Compound 13f exhibited the ability to both significantly decrease the glucose excursion and inhibit plasma DPP-IV activity.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Glutamic Acid/chemistry , Pyrrolidines/chemical synthesis , Administration, Oral , Animals , Dipeptidyl Peptidase 4/blood , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Glucose/metabolism , Mice , Mice, Inbred C57BL , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...