Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 23(6): 766-779, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38592383

ABSTRACT

Aurora kinase inhibitors, such as alisertib, can destabilize MYC-family oncoproteins and have demonstrated compelling antitumor efficacy. In this study, we report 6K465, a novel pyrimidine-based Aurora A inhibitor, that reduces levels of c-MYC and N-MYC oncoproteins more potently than alisertib. In an analysis of the antiproliferative effect of 6K465, the sensitivities of small cell lung cancer (SCLC) and breast cancer cell lines to 6K465 were strongly associated with the protein levels of c-MYC and/or N-MYC. We also report DBPR728, an acyl-based prodrug of 6K465 bearing fewer hydrogen-bond donors, that exhibited 10-fold improved oral bioavailability. DBPR728 induced durable tumor regression of c-MYC- and/or N-MYC-overexpressing xenografts including SCLC, triple-negative breast cancer, hepatocellular carcinoma, and medulloblastoma using a 5-on-2-off or once-a-week dosing regimen on a 21-day cycle. A single oral dose of DBPR728 at 300 mg/kg induced c-MYC reduction and cell apoptosis in the tumor xenografts for more than 7 days. The inhibitory effect of DBPR728 at a reduced dosing frequency was attributed to its uniquely high tumor/plasma ratio (3.6-fold within 7 days) and the long tumor half-life of active moiety 6K465. Furthermore, DBPR728 was found to synergize with the mTOR inhibitor everolimus to suppress c-MYC- or N-MYC-driven SCLC. Collectively, these results suggest DBPR728 has the potential to treat cancers overexpressing c-MYC and/or N-MYC.


Subject(s)
Aurora Kinase A , Everolimus , Proto-Oncogene Proteins c-myc , Xenograft Model Antitumor Assays , Humans , Animals , Aurora Kinase A/antagonists & inhibitors , Mice , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Everolimus/pharmacology , Everolimus/pharmacokinetics , Everolimus/administration & dosage , Cell Line, Tumor , Female , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
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.
Eur J Med Chem ; 224: 113673, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34303872

ABSTRACT

Rare oncogenic NTRK gene fusions result in uncontrolled TRK signaling leading to various adult and pediatric solid tumors. Based on the architecture of our multi-targeted clinical candidate BPR1K871 (10), we designed and synthesized a series of quinazoline compounds as selective and orally bioavailable type II TRK inhibitors. Property-driven and lead optimization strategies informed by structure-activity relationship studies led to the identification of 39, which showed higher (about 15-fold) selectivity for TRKA over AURA and AURB, as well as potent cellular activity (IC50 = 56.4 nM) against the KM12 human colorectal cancer cell line. 39 also displayed good AUC and oral bioavailability (F = 27%), excellent in vivo efficacy (TGI = 64%) in a KM12 xenograft model, and broad-spectrum anti-TRK mutant potency (IC50 = 3.74-151.4 nM), especially in the double-mutant TRKA enzymatic assays. 39 is therefore proposed for further development as a next-generation, selective, and orally-administered type II TRK inhibitor.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemistry , Receptor, trkA/antagonists & inhibitors , Administration, Oral , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/metabolism , Binding Sites , Cell Line, Tumor , Half-Life , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use , Rats , Receptor, trkA/metabolism , Structure-Activity Relationship , Transplantation, Heterologous
4.
Transl Oncol ; 14(1): 100897, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33069101

ABSTRACT

Zinc(II)-dipicolylamine (Zn-DPA) has been shown to specifically identify and bind to phosphatidylserine (PS), which exists in bulk in the tumor microenvironment. BPRDP056, a Zn-DPA-SN38 conjugate was designed to provide PS-targeted drug delivery of a cytotoxic SN38 to the tumor microenvironment, thereby allowing a lower dosage of SN38 that induces apoptosis in cancer cells. Micro-Western assay showed that BPRDP056 exhibited apoptotic signal levels similar to those of CPT-11 in the treated tumors growing in mice. Pharmacokinetic study showed that BPRDP056 has excellent systemic stability in circulation in mice and rats. BPRDP056 is accumulated in tumors and thus increases the cytotoxic effects of SN38. The in vivo antitumor activities of BPRDP056 have been shown to be significant in subcutaneous pancreas, prostate, colon, liver, breast, and glioblastoma tumors, included an orthotopic pancreatic tumor, in mice. BPRDP056 shrunk tumors at a lower (~20% only) dosing intensity of SN38 compared to that of SN38 conjugated in CPT-11 in all tumor models tested. A wide spectrum of antitumor activities is expected to treat all cancer types of PS-rich tumor microenvironments. BPRDP056 is a first-in-class small molecule drug conjugate for cancer therapy.

5.
J Med Chem ; 62(24): 11135-11150, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31721578

ABSTRACT

Gastrointestinal stromal tumors (GISTs) are prototypes of stem cell factor receptor (c-KIT)-driven cancer. Two receptor tyrosine kinases, c-KIT and fms-tyrosine kinase (FLT3), are frequently mutated in acute myeloid leukemia (AML) patients, and these mutations are associated with poor prognosis. In this study, we discovered a multitargeted tyrosine kinase inhibitor, compound 15a, with potent inhibition against single or double mutations of c-KIT developed in GISTs. Moreover, crystal structure analysis revealed the unique binding mode of 15a with c-KIT and may elucidate its high potency in inhibiting c-KIT kinase activity. Compound 15a inhibited cell proliferation and induced apoptosis by targeting c-KIT in c-KIT-mutant GIST cell lines. The antitumor effects of 15a were also demonstrated in GIST430 and GIST patient-derived xenograft models. Further studies demonstrated that 15a inhibited the proliferation of c-KIT- and FLT3-driven AML cells in vitro and in vivo. The results of this study suggest that 15a may be a potential anticancer drug for the treatment of GISTs and AML.


Subject(s)
Antineoplastic Agents/pharmacology , Gastrointestinal Stromal Tumors/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Proliferation , Female , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/enzymology , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/enzymology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred ICR , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphorylation , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/chemistry , Rats, Sprague-Dawley , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/genetics
6.
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
7.
Bioconjug Chem ; 28(7): 1878-1892, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28581724

ABSTRACT

A series of zinc(II) dipicolylamine (ZnDPA)-based drug conjugates have been synthesized to probe the potential of phosphatidylserine (PS) as a new antigen for small molecule drug conjugate (SMDC) development. Using in vitro cytotoxicity and plasma stability studies, PS-binding assay, in vivo pharmacokinetic studies, and maximum tolerated dose profiles, we provided a roadmap and the key parameters required for the development of the ZnDPA based drug conjugate. In particular, conjugate 24 induced tumor regression in the COLO 205 xenograft model and exhibited a more potent antitumor effect with a 70% reduction of cytotoxic payload compared to that of the marketed irinotecan when dosed at the same regimen. In addition to the validation of PS as an effective pharmacodelivery target for SMDC, our work also provided the foundation that, if applicable, a variety of therapeutic agents could be conjugated in the same manner to treat other PS-associated diseases.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Immunoconjugates/therapeutic use , Molecular Targeted Therapy/methods , Organometallic Compounds/immunology , Phosphatidylserines/immunology , Picolinic Acids/immunology , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Phosphatidylserines/metabolism , Picolinic Acids/chemical synthesis , Picolinic Acids/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
8.
Oncotarget ; 7(52): 86239-86256, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27863392

ABSTRACT

The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents. BPR1K871 showed potent anti-proliferative activities in MOLM-13 and MV4-11 AML cells (EC50 ~ 5 nM). Moreover, kinase profiling and cell-line profiling revealed BPR1K871 to be a potential multi-kinase inhibitor. Functional studies using western blot and DNA content analysis in MV4-11 and HCT-116 cell lines revealed FLT3 and AURKA/B target modulation inside the cells. In vivo efficacy in AML xenograft models (MOLM-13 and MV4-11), as well as in solid tumor models (COLO205 and Mia-PaCa2), led to the selection of BPR1K871 as a preclinical development candidate for anti-cancer therapy. Further detailed studies could help to investigate the full potential of BPR1K871 as a multi-kinase inhibitor.


Subject(s)
Antineoplastic Agents/chemical synthesis , Aurora Kinase A/antagonists & inhibitors , Drug Discovery , Leukemia, Myeloid, Acute/drug therapy , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Quinazolines/chemical synthesis , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Humans , Male , Models, Molecular , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Eur J Med Chem ; 100: 151-61, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26081023

ABSTRACT

Numerous FLT3 inhibitors have been explored as a viable therapy for the treatment of acute myeloid leukemia (AML). However, clinical data have been underwhelming due to incomplete inhibition of FLT3 or the emergence of resistant mutations treated with these older agents. We previously developed a series of 3-phenyl-1H-5-pyrazolylamine derivatives as highly potent and selective FLT3 inhibitors with good in vivo efficacy using an intravenous (IV) route. However, the poor bioavailability of these pyrazole compounds limits the development of these promising antileukemic compounds for clinical use. Herein, we describe a novel class of 5-phenyl-thiazol-2-ylamine compounds that are multi-targeted FLT3 inhibitors. From this class of compounds, compound 7h was very potent against AML cell lines and exhibited excellent oral efficacy in AML xenograft models. In addition, further studies demonstrated that compound 7h exhibited potent in vitro and in vivo activities against clinically relevant AC220 (3)-resistant kinase domain mutants of FLT3-ITD.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms, Experimental/drug therapy , Point Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Inbred ICR , Mice, Nude , Molecular Structure , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
10.
Invest New Drugs ; 30(1): 164-75, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20890633

ABSTRACT

Designed from a high throughput screened hit compound, novel 2-amino-1-thiazolyl imidazoles were synthesized and demonstrated cytotoxicity against human cancer cells. 1-(4-Phenylthiazol-2-yl)-4-(thiophen-2-yl)-1H-imidazol-2-amine (compound 2), a 2-amino-1-thiazolyl imidazole, inhibited tubulin polymerization, interacted with the colchicine-binding sites of tubulins, and caused cell cycle arrest at the G(2)/M phase in human gastric cancer cells. Disruption of the microtubule structure in cancer cells by compound 2 was also observed. Compound 2 concentration-dependently inhibited the proliferation of cancer cells in histocultured human gastric and colorectal tumors. Given orally, compound 2 prolonged the lifespans of leukemia mice intraperitoneally inoculated with the murine P388 leukemic cells. We report 2-amino-1-thiazolyl imidazoles as a novel class of orally active microtubule-destabilizing anticancer agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Neoplasms, Experimental/drug therapy , Thiazoles/administration & dosage , Tubulin Modulators/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding, Competitive , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Dose-Response Relationship, Drug , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred DBA , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/metabolism , Time Factors , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
11.
Cancer Sci ; 102(1): 182-91, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21040217

ABSTRACT

BPR0C261 is a synthetic small molecule compound cytotoxic against human cancer cells and active prolonging the lifespan of leukemia mice. In the present study, we further investigated the mechanisms of its anticancer action and found that BPR0C261 inhibited microtubule polymerization through interacting with the colchicine binding sites on tubulins, disrupted microtubule arrangement and caused cell cycle arrest at G(2)/M phase in cancer cells. BPR0C261 also inhibited the clonogenic growths of cancer cells and showed cytotoxicity against human cervical cancer cells of multidrug-resistant phenotype. In addition, BPR0C261 concentration-dependently inhibited the proliferation and migration of HUVECs and disrupted the endothelial capillary-like tube formations in HUVEC and rat aorta ring cultures. Given orally, BPR0C261 inhibited angiogenesis in s.c. implanted Matrigel plugs in mice. Notably, its IC(50) values against the endothelial cell growths were approximately 10-fold lower than those against the cancer cells. It was found orally absorbable in mice and showed a good oral bioavailability (43%) in dogs. BPR0C261 permeated through the human intestinal Caco-2 cell monolayer, suggesting oral availability in humans. Orally absorbed BPR0C261 distributed readily into the s.c. xenografted tumors in nude mice in which the tumor tissue levels of BPR0C261 were found oral dose-dependent. BPR0C261 showed in vivo activities against human colorectal, gastric, and nasopharyngeal tumors in nude mice. Most interestingly, the combination of BPR0C261 plus cisplatin synergistically prolonged the lifespans of mice inoculated with murine leukemia cells. Thus, BPR0C261 is a novel orally active tubulin-binding antitumor agent with antimitotic, apoptosis-inducing, and vasculature disrupting activities.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antimitotic Agents/pharmacology , Antineoplastic Agents/pharmacology , Indoles/pharmacology , Thiazoles/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Dogs , Humans , Leukemia, Experimental/drug therapy , Male , Mice , Mice, Inbred Strains , Microtubules/chemistry , Microtubules/drug effects , Rats , Rats, Sprague-Dawley
12.
J Med Chem ; 53(6): 2409-17, 2010 Mar 25.
Article in English | MEDLINE | ID: mdl-20170097

ABSTRACT

2-Amino-1-arylidenaminoimidazoles, a novel class of orally (po) active microtubule-destabilizing anticancer agents, were synthesized. The compounds were designed from a hit compound identified in a drug discovery platform by using cancer cell-based high throughput screening assay. Selective synthesized compounds exerted cell cytotoxicity against human cancer cells. The underlying mechanisms for the anticancer activity were demonstrated as interacting with the tubulins and inhibiting microtubule assembly, leading to proliferation inhibition and apoptosis induction in the human tumor cells. Furthermore, two compounds showed in vivo anticancer activities in both po and intravenously (iv) administered routes and prolonged the life spans of murine leukemic P388 cells-inoculated mice. These new po active antimitotic anticancer agents are to be further examined in preclinical studies and developed for clinical uses.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Neoplasms/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/chemistry , Inhibitory Concentration 50 , Leukemia P388/drug therapy , Leukemia P388/pathology , Mice , Mice, Inbred DBA , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Models, Chemical , Molecular Structure , Neoplasms/pathology , Structure-Activity Relationship , Tubulin/metabolism , Xenograft Model Antitumor Assays
13.
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
14.
Pharmacol Res ; 61(2): 108-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19643180

ABSTRACT

DB-67 and its lactone homolog DB-91 are derivatives of topoisomerase I inhibitor camptothecin (CPT) with silyl moiety, which may exhibit a slower inactivation process by changed kinetics of protein binding and/or hydrolysis of its lactone ring and result in increased antitumor activity and decreased toxicity. Pharmacokinetic properties and antitumor activities of the two silatecans were studied and compared. The lactone ring of DB-91 is more stable than those of all the other CPT derivatives in mouse plasma. Both silatecans were metabolized faster than CPT in mouse and human liver microsomes. Pharmacokinetic study revealed a plasma elimination half-life (t(1/2)) of 33 and 94min for DB-67 and DB-91, respectively; similar systemic exposure in plasma between DB-67 and DB-91; and similar volume of distribution at the steady state between DB-67 and DB-91, approximately 15-fold smaller than that of CPT. While DB-91 showed limited activities, DB-67 exhibited activities against the growth of in vivo-like histocultured human tumors and s.c. xenografted human tumors in nude mice. In conclusion, DB-67 is more effective, compared to DB-91, against human tumor growth in in vitro, in vivo-like and in vivo systems. Further pre-clinical and clinical investigations of DB-67 are warranted.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Organosilicon Compounds/pharmacology , Topoisomerase I Inhibitors , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/blood , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Drug Resistance, Neoplasm , Drug Stability , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Half-Life , Humans , Injections, Intravenous , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes, Liver/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Organosilicon Compounds/administration & dosage , Organosilicon Compounds/blood , Organosilicon Compounds/pharmacokinetics , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
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
16.
J Biomed Sci ; 14(2): 233-44, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17206490

ABSTRACT

Cancer has been the leading cause of death in Taiwan over the past two decades and liver cancer is the leading cause of all cancer deaths in Taiwan with a trend of increase in incidence. Therapeutic options and efficacy for liver cancer have been limited and the 5-year survival rate is less than 7% in the Unite States. The study was conducted to establish a histoculture system of human hepatocellular carcinomas (HCC) for biological and pharmacological studies and to determine the efficacy of anticancer drugs with the established HCC histocultures. Patient HCC tissues freshly obtained after surgeries were prepared and histocultured. The histocultured HCC were treated with doxorubicin and paclitaxel of various concentrations for 96-h. Upon drug treatments, the activity of tumor cell proliferation and extent of cell death induction were measured and changes of the alpha-fetoprotein levels in the culture medium were determined. We demonstrated that human HCC can be successfully cultured in a 3-dimensional histoculture system and used for pharmacological studies. Doxorubicin and paclitaxel showed concentration-dependent activities in anti-proliferation and cell death induction against the human HCC. Inhibitory effects of both drugs on alpha-fetoprotein production of the cultured HCC were in agreement with their anti-proliferative effects. Exposure time-dependent antitumoral effects of paclitaxel treatments at 3-, 24-, and 96-h against the histocultured HCC PLC/PRF/5 xenograft tumors were also observed. In conclusion, we have demonstrated a histoculture system for patient HCC and it can be utilized in selection of active drugs prior to treatments in patients and in evaluation of new agents against HCC, for which therapeutic agents are in desperate needs worldwide.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Liver Neoplasms , Paclitaxel/pharmacology , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , Liver Neoplasms/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Multidrug Resistance-Associated Proteins/analysis , Multidrug Resistance-Associated Proteins/metabolism , Time Factors , alpha-Fetoproteins/metabolism
17.
Anticancer Res ; 23(1A): 205-10, 2003.
Article in English | MEDLINE | ID: mdl-12680214

ABSTRACT

BACKGROUND: Gastric cancer is one of the prevalent cancer types in the Far East region. In order to discover new prognostic and diagnostic biomarkers for gastric cancer progression, we have used degenerated PCR primers designed to amplify all expressed protein tyrosine kinase molecules from human cancer tissues. In previous studies, we have shown the clinical significance of arg tyrosine kinases in a colorectal cancer progression model. MATERIALS AND METHODS: We further investigated the protein expression of arg tyrosine kinase by immunohistochemistry and analyzed the clinicopathological association with human cancers. RESULTS AND CONCLUSION: Specimens from 79 patients were examined and this demonstrated that the expression of arg kinase showed no statistically significant correlation with the patients' overall survival. However, higher levels of arg kinase immunoreactivity did show a statistically significant association with vessel invasion in gastric cancer tissues examined. This indicates a potential involvement of arg in gastric cancer invasion and progression.


Subject(s)
Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Protein-Tyrosine Kinases/biosynthesis , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Aged , Amino Acid Sequence , Female , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Sequence Data , Neoplasm Invasiveness
18.
Anticancer Res ; 22(2B): 1071-8, 2002.
Article in English | MEDLINE | ID: mdl-12168903

ABSTRACT

BACKGROUND: We have used degenerated PCR primers designed according to the consensus kinase motifs in order to amplify expressed protein tyrosine kinase molecules from human gastric cancers. From such kinase expression profiles, we have identified more than fifty different tyrosine and serine/threonine kinases from two matched pairs of gastric cancer tissue and normal mucosa. In previous studies, we have shown the clinical significance of two tyrosine kinases identified in gastric cancer, tie-1 and mkk4. MATERIALS AND METHODS: In this report, we further investigate the protein expression of the whole axl receptor tyrosine-kinase family (axl/ufo, nyk/mer and sky/rse) by immunohistochemistry and their clinicopathological associations. RESULTS: On examination of 96 patients specimens, expression of the axl kinase family alone showed no statistical significance with respect to the patients' survivaL However, the combination of nyk/mer expression with axl/ufo expression correlated inversely with the patients' prognosis result. CONCLUSION: This finding indicates that a co-operative relationship exists between axllufo and nyk/mer protein kinases and this seems to play an important role in gastric cancer progression and metastasis.


Subject(s)
Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Stomach Neoplasms/enzymology , Aged , Antibodies/chemistry , Antibodies/immunology , Antibody Specificity , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Middle Aged , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Proto-Oncogene Proteins , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...