Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
J Med Chem ; 62(21): 9541-9559, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31593466

ABSTRACT

Novel nanoparticle-drug conjugates (NDCs) containing diverse, clinically relevant anticancer drug payloads (docetaxel, cabazitaxel, and gemcitabine) were successfully generated and tested in drug discovery studies. The NDCs utilized structurally varied linkers that attached the drug payloads to a ß-cyclodextrin-PEG copolymer to form self-assembled nanoparticles. In vitro release studies revealed a diversity of release rates driven by linker structure-activity relationships (SARs). Improved in vivo pharmacokinetics (PK) for the cabazitaxel (CBTX) NDCs with glycinate-containing (1c) and hexanoate-containing linkers (2c) were demonstrated, along with high and sustained tumor levels (>168 h of released drug in tumor tissues). This led to potent efficacy and survival in both taxane- and docetaxel-resistant in vivo anticancer mouse efficacy models. Overall, the CBTX-hexanoate NDC 2c (CRLX522), demonstrated optimal and improved in vivo PK (plasma and tumor) and efficacy profile versus those of the parent drug, and the results support the potential therapeutic use of CRLX522 as a new anticancer agent.


Subject(s)
Drug Carriers/chemistry , Drug Design , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Taxoids/chemistry , Taxoids/pharmacology , beta-Cyclodextrins/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Male , Melanoma, Experimental/pathology , Mice , Taxoids/pharmacokinetics , Tissue Distribution
3.
Bioorg Med Chem Lett ; 27(8): 1670-1680, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28302397

ABSTRACT

The emergence and spread of multidrug-resistant (MDR) Gram negative bacteria presents a serious threat for public health. Novel antimicrobials that could overcome the resistance problems are urgently needed. UDP-3-O-(R-3-hydroxymyristol)-N-acetylglucosamine deacetylase (LpxC) is a cytosolic zinc-based deacetylase that catalyzes the first committed step in the biosynthesis of lipid A, which is essential for the survival of Gram-negative bacteria. Our efforts toward the discovery of novel LpxC inhibitors are presented herein.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Amidohydrolases/metabolism , Drug Discovery , Gram-Negative Bacterial Infections/drug therapy , Humans , Molecular Docking Simulation
4.
ACS Med Chem Lett ; 7(4): 374-8, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27096044

ABSTRACT

The ATPase subunit of DNA gyrase B is an attractive antibacterial target due to high conservation across bacteria and the essential role it plays in DNA replication. A novel class of pyrazolopyridone inhibitors was discovered by optimizing a fragment screening hit scaffold using structure guided design. These inhibitors show potent Gram-positive antibacterial activity and low resistance incidence against clinically important pathogens.

5.
ACS Med Chem Lett ; 6(10): 1080-5, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26487916

ABSTRACT

Antibacterials with a novel mechanism of action offer a great opportunity to combat widespread antimicrobial resistance. Bacterial DNA Gyrase is a clinically validated target. Through physiochemical property optimization of a pyrazolopyridone hit, a novel class of GyrB inhibitors were discovered. Guided by structure-based drug design, indazole derivatives with excellent enzymatic and antibacterial activity as well as great animal efficacy were discovered.

6.
J Med Chem ; 58(21): 8503-12, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26460684

ABSTRACT

The emergence and spread of multidrug resistant bacteria are widely believed to endanger human health. New drug targets and lead compounds exempt from cross-resistance with existing drugs are urgently needed. We report on the discovery of azaindole ureas as a novel class of bacterial gyrase B inhibitors and detail the story of their evolution from a de novo design hit based on structure-based drug design. These inhibitors show potent minimum inhibitory concentrations against fluoroquinolone resistant MRSA and other Gram-positive bacteria.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , DNA Gyrase/metabolism , Indoles/pharmacology , Methicillin-Resistant Staphylococcus aureus/enzymology , Topoisomerase II Inhibitors/pharmacology , Urea/pharmacology , Bacterial Proteins/metabolism , Crystallography, X-Ray , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/enzymology , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Humans , Indoles/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Models, Molecular , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Topoisomerase II Inhibitors/chemistry , Urea/analogs & derivatives
7.
J Med Chem ; 58(12): 5137-42, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25993059

ABSTRACT

Novel cyclic lipopeptides with different acyl tails were synthesized via a semisynthetic approach. Structure-activity relationship studies revealed that lipophilicity, chain length, and the location of key aromatic functionalities of the tail modulated activity. The lead compound surotomycin exhibited significantly improved in vitro activity compared with daptomycin (MIC90 0.5 vs 2 µg/mL) against Clostridium difficile including NAP1 epidemic strains. In hamster efficacy studies, surotomycin protected animals at a dose of 0.5 mg/kg, PO.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/drug effects , Diarrhea/drug therapy , Enterocolitis, Pseudomembranous/drug therapy , Lipopeptides/chemistry , Lipopeptides/therapeutic use , Animals , Cricetinae , Diarrhea/microbiology , Enterocolitis, Pseudomembranous/complications , Male , Microbial Sensitivity Tests , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 22(19): 6248-51, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22951041

ABSTRACT

Daptomycin was shown to interact in vitro with pulmonary surfactant leading to reduction of its antibacterial activity. We report herein the preparation and anti-staphylococcal activity of a series of daptomycin analogs with reduced pulmonary surfactant interaction by replacing tryptophan with various amino acids.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Daptomycin/analogs & derivatives , Daptomycin/pharmacology , Pulmonary Surfactants/chemistry , Staphylococcus aureus/drug effects , Tryptophan/metabolism , Daptomycin/chemistry , Microbial Sensitivity Tests , Molecular Conformation
9.
Bioorg Med Chem Lett ; 21(12): 3743-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21561767

ABSTRACT

Ponatinib (AP24534) was previously identified as a pan-BCR-ABL inhibitor that potently inhibits the T315I gatekeeper mutant, and has advanced into clinical development for the treatment of refractory or resistant CML. In this study, we explored a novel series of five and six membered monocycles as alternate hinge-binding templates to replace the 6,5-fused imidazopyridazine core of ponatinib. Like ponatinib, these monocycles are tethered to pendant toluanilides via an ethynyl linker. Several compounds in this series displayed excellent in vitro potency against both native BCR-ABL and the T315I mutant. Notably, a subset of inhibitors exhibited desirable PK and were orally active in a mouse model of T315I-driven CML.


Subject(s)
Alkynes/chemical synthesis , Alkynes/pharmacology , Aniline Compounds/chemical synthesis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Toluene/chemical synthesis , Administration, Oral , Alkynes/chemistry , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Cyclization , Disease Models, Animal , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Models, Molecular , Molecular Structure , Mutation , Rats , Structure-Activity Relationship , Toluene/chemistry , Toluene/pharmacology
10.
J Med Chem ; 53(12): 4701-19, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20513156

ABSTRACT

In the treatment of chronic myeloid leukemia (CML) with BCR-ABL kinase inhibitors, the T315I gatekeeper mutant has emerged as resistant to all currently approved agents. This report describes the structure-guided design of a novel series of potent pan-inhibitors of BCR-ABL, including the T315I mutation. A key structural feature is the carbon-carbon triple bond linker which skirts the increased bulk of Ile315 side chain. Extensive SAR studies led to the discovery of development candidate 20g (AP24534), which inhibited the kinase activity of both native BCR-ABL and the T315I mutant with low nM IC(50)s, and potently inhibited proliferation of corresponding Ba/F3-derived cell lines. Daily oral administration of 20g significantly prolonged survival of mice injected intravenously with BCR-ABL(T315I) expressing Ba/F3 cells. These data, coupled with a favorable ADME profile, support the potential of 20g to be an effective treatment for CML, including patients refractory to all currently approved therapies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Pyridazines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/genetics , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Mice , Mice, SCID , Models, Molecular , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Rats
11.
Cancer Cell ; 16(5): 401-12, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19878872

ABSTRACT

Inhibition of BCR-ABL by imatinib induces durable responses in many patients with chronic myeloid leukemia (CML), but resistance attributable to kinase domain mutations can lead to relapse and a switch to second-line therapy with nilotinib or dasatinib. Despite three approved therapeutic options, the cross-resistant BCR-ABL(T315I) mutation and compound mutants selected on sequential inhibitor therapy remain major clinical challenges. We report design and preclinical evaluation of AP24534, a potent, orally available multitargeted kinase inhibitor active against T315I and other BCR-ABL mutants. AP24534 inhibited all tested BCR-ABL mutants in cellular and biochemical assays, suppressed BCR-ABL(T315I)-driven tumor growth in mice, and completely abrogated resistance in cell-based mutagenesis screens. Our work supports clinical evaluation of AP24534 as a pan-BCR-ABL inhibitor for treatment of CML.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Pyridazines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imidazoles/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, SCID , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Pyridazines/chemistry , Signal Transduction/drug effects
12.
Clin Cancer Res ; 12(20 Pt 2): 6291s-6295s, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17062716

ABSTRACT

Src tyrosine kinase was the first gene product shown to have an essential function in bone using recombinant DNA technology after its expression was knocked out in mice approximately 15 years ago. Since then, our understanding of the regulation of bone catabolism has advanced significantly with the identification of other key enzymes that regulate osteoclast formation, activation, and survival after their knockout in mice or recognition of mutations in them in humans. This led to the discovery or development of specific inhibitors of some of these key enzymes, including Src, as proof-of-concept lead compounds or potential clinical candidates for the prevention of diseases associated with increased bone resorption, such as osteoporosis and metastatic bone disease. Although bisphosphonates have been prescribed with proven and improving efficacy for the prevention of bone loss for >30 years, adverse effects, such as upper gastrointestinal tract symptoms, and the requirement to take them at least 2 hours before food have limited patient compliance. Thus, with growing knowledge of the pathways regulating osteoclast function and the appreciation that some of these are active also in tumor cells, drug companies have made efforts to identify small-molecular lead compounds for development into new therapeutic agents for the prevention of bone loss with efficacy that matches or supersedes that of bisphosphonates. In this article, we review our current understanding of the signaling pathways that regulate osteoclast formation, activation, and survival with specific reference to the role of Src tyrosine kinase and downstream signaling and highlight in a variety of models of increased bone resorption the effects of Src kinase inhibitors that have been targeted to bone to limit potential adverse effects on other cells.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Bone Resorption/prevention & control , Osteoclasts/drug effects , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/metabolism , Animals , Cell Differentiation , Humans , Mice , Osteoclasts/cytology , Osteoclasts/enzymology , Protein Kinase Inhibitors/chemistry , Signal Transduction/physiology , Structure-Activity Relationship , src-Family Kinases/drug effects
13.
Cancer Res ; 66(17): 8633-9, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951177

ABSTRACT

Src, a nonreceptor tyrosine kinase, is a key mediator for multiple signaling pathways that regulate critical cellular functions and is often aberrantly activated in a number of solid tumors, including ovarian carcinoma. The purpose of this study was to determine the role of activated Src inhibition on tumor growth in an orthotopic murine model of ovarian carcinoma. In vitro studies on HeyA8 and SKOV3ip1 cell lines revealed that Src inhibition by the Src-selective inhibitor, AP23846, occurred within 1 hour and responded in a dose-dependent manner. Furthermore, Src inhibition enhanced the cytotoxicity of docetaxel in both chemosensitive and chemoresistant ovarian cancer cell lines, HeyA8 and HeyA8-MDR, respectively. In vivo, Src inhibition by AP23994, an orally bioavailable analogue of AP23846, significantly decreased tumor burden in HeyA8 (P = 0.02), SKOV3ip1 (P = 0.01), as well as HeyA8-MDR (P < 0.03) relative to the untreated controls. However, the greatest effect on tumor reduction was observed in combination therapy with docetaxel (P < 0.001, P = 0.002, and P = 0.01, for the above models, respectively). Proliferating cell nuclear antigen staining showed that Src inhibition alone (P = 0.02) and in combination with docetaxel (P = 0.007) significantly reduced tumor proliferation. In addition, Src inhibition alone and in combination with docetaxel significantly down-regulated tumoral production of vascular endothelial growth factor and interleukin 8, whereas combination therapy decreased the microvessel density (P = 0.02) and significantly affected vascular permeability (P < 0.05). In summary, Src inhibition with AP23994 has potent antiangiogenic effects and significantly reduces tumor burden in preclinical ovarian cancer models. Thus, Src inhibition may be an attractive therapeutic approach for patients with ovarian carcinoma.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Ovarian Neoplasms/pathology , src-Family Kinases/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Cell Line, Tumor , Edetic Acid/pharmacology , Female , Humans
14.
Ann N Y Acad Sci ; 1068: 447-57, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16831942

ABSTRACT

Understanding of the regulation of bone catabolism has advanced significantly over the past two decades with the identification of key enzymes that regulate osteoclast formation, activation, and survival following their knockout in mice or recognition of mutations in humans. This led to the discovery of specific inhibitors of some of these key enzymes as proof-of-concept lead compounds or potential clinical candidates for the prevention of osteoporosis and other diseases associated with increased bone resorption. Bisphosphonates have been the major therapeutic agents prescribed for the prevention of bone loss in a variety of pathologic conditions for over 30 years. More potent amino bisphosphonates have increased efficacy than earlier drugs, but side effects such as upper gastrointestinal symptoms and the requirement to take them at least 2 h before food have limited patient compliance. This, coupled with the growing knowledge of the pathways regulating osteoclast function, has driven efforts to identify small molecular lead compounds that could be developed into new therapeutic agents with efficacy that matches or supersedes that of bisphosphonates for the prevention of bone loss. In this article, we review briefly the effects of specific inhibitors of bone resorption that have been developed to date and highlight in a variety of models of increased bone resorption the effects of Src kinase inhibitors that have been targeted to bone to limit potential unwanted side effects on other cells.


Subject(s)
Bone Diseases/drug therapy , Bone and Bones/metabolism , Enzyme Inhibitors/therapeutic use , Bone Resorption , Cathepsin K , Cathepsins/antagonists & inhibitors , Humans , Osteoclasts/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Sirolimus/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
15.
Proc Natl Acad Sci U S A ; 103(24): 9244-9, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16754879

ABSTRACT

Mutation in the ABL kinase domain is the principal mechanism of imatinib resistance in patients with chronic myelogenous leukemia. Many mutations favor active kinase conformations that preclude imatinib binding. Because the active forms of ABL and SRC resemble one another, we tested two dual SRC-ABL kinase inhibitors, AP23464 and PD166326, against 58 imatinib-resistant (IM(R)) BCR/ABL kinase variants. Both compounds potently inhibit most IM(R) variants, and in vitro drug selection demonstrates that active (AP23464) and open (PD166326) conformation-specific compounds are less susceptible to resistance than imatinib. Combinations of inhibitors suppressed essentially all resistance mutations, with the notable exception of T315I. Guided by mutagenesis studies and molecular modeling, we designed a series of AP23464 analogues to target T315I. The analogue AP23846 inhibited both native and T315I variants of BCR/ABL with submicromolar potency but showed nonspecific cellular toxicity. Our data illustrate how conformational dynamics of the ABL kinase accounts for the activity of dual SRC-ABL inhibitors against IM(R)-mutants and provides a rationale for combining conformation specific inhibitors to suppress resistance.


Subject(s)
Drug Resistance/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Benzamides , Fusion Proteins, bcr-abl , Humans , Imatinib Mesylate , Models, Molecular , Molecular Structure , Mutation , Piperazines/chemistry , Piperazines/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-abl/genetics , Pyridines/chemistry , Pyridines/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , src-Family Kinases/genetics
16.
Chem Biol Drug Des ; 67(1): 46-57, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16492148

ABSTRACT

The tyrosine kinase pp60src (Src) is the prototypical member of a family of proteins that participate in a broad array of cellular signal transduction processes, including cell growth, differentiation, survival, adhesion, and migration. Abnormal Src family kinase (SFK) signaling has been linked to several disease states, including osteoporosis and cancer metastases. Src has thus emerged as a molecular target for the discovery of small-molecule inhibitors that regulate Src kinase activity by binding to the ATP pocket within the catalytic domain. Here, we present crystal structures of the kinase domain of Src in complex with two purine-based inhibitors: AP23451, a small-molecule inhibitor designed to inhibit Src-dependent bone resorption, and AP23464, a small-molecule inhibitor designed to inhibit the Src-dependent metastatic spread of cancer. In each case, a trisubstituted purine template core was elaborated using structure-based drug design to yield a potent Src kinase inhibitor. These structures represent early examples of high affinity purine-based Src family kinase-inhibitor complexes, and they provide a detailed view of the specific protein-ligand interactions that lead to potent inhibition of Src. In particular, the 3-hydroxyphenethyl N9 substituent of AP23464 forms unique interactions with the protein that are critical to the picomolar affinity of this compound for Src. The comparison of these new structures with two relevant kinase-inhibitor complexes provides a structural basis for the observed kinase inhibitory selectivity. Further comparisons reveal a concerted induced-fit movement between the N- and C-terminal lobes of the kinase that correlates with the affinity of the ligand. Binding of the most potent inhibitor, AP23464, results in the largest induced-fit movement, which can be directly linked to interactions of the hydrophenethyl N9 substituent with a region at the interface between the two lobes. A less pronounced induced-fit movement is also observed in the Src-AP23451 complex. These new structures illustrate how the combination of structural, computational, and medicinal chemistry can be used to rationalize the process of developing high affinity, selective tyrosine kinase inhibitors as potential therapeutic agents.


Subject(s)
Adenine/analogs & derivatives , Drug Design , Enzyme Inhibitors/chemistry , Organophosphonates/chemistry , Purines/chemistry , Structure-Activity Relationship , src-Family Kinases/chemistry , Adenine/chemistry , Adenine/metabolism , Adenine/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Bone and Bones/metabolism , Carbon/chemistry , Catalytic Domain/drug effects , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Ligands , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Models, Molecular , Neoplasms/drug therapy , Nitrogen/chemistry , Organophosphonates/metabolism , Organophosphonates/pharmacology , Protein Conformation/drug effects , Purines/chemical synthesis , Purines/pharmacology , Pyrimidines/chemistry , Substrate Specificity , src-Family Kinases/antagonists & inhibitors
17.
Mol Cancer Ther ; 4(12): 1900-11, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16373705

ABSTRACT

c-Src is frequently activated in human malignancies, including colon, breast, and pancreatic carcinomas. Several recent studies have shown that activation of Src family kinases leads to tumor progression and metastasis by increasing cellular migration and invasion, promoting cell growth and survival, and deregulating expression of proangiogenic molecules. Therefore, selective inhibitors of Src are being developed for cancer therapy. In this study, we characterize the biological effects of the novel ATP-based Src family kinase inhibitor, AP23846, in tumor cells with high Src activity. As a lead compound, AP23846 is a potent c-Src kinase inhibitor (IC50 approximately 0.5 nmol/L in vitro, approximately 10-fold more potent than PP2, the most widely used commercially available Src family kinase inhibitor). At concentrations of 1 micromol/L, AP23846 led to complete Src inhibition for 48 hours in cells. No cytotoxicity was observed under these conditions, although proliferation rates were slower. Therefore, this was an excellent inhibitor to examine Src-regulated signaling pathways in tumor cells. AP23846 reduced cellular migration, vascular endothelial growth factor, and interleukin-8 in a dose-dependent fashion in pancreatic adenocarcinoma cells grown in vitro. Correspondingly, cell culture supernatants from L3.6pl pancreatic adenocarcinoma cells pretreated with AP23846 failed to promote migration of hepatic endothelial cells in vitro and failed to support angiogenesis into gel foams implanted s.c. in mice in vivo. These results suggest that Src inhibitors affect biological properties of tumor progression and may be useful as cancer therapeutic agents in more advanced disease.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Enzyme Inhibitors/pharmacology , Interleukin-8/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/antagonists & inhibitors , Adenosine Triphosphate/pharmacology , Animals , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Movement , DNA Primers , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Mice , Mice, Inbred C3H , Neoplasms/blood supply , Phosphorylation , RNA, Small Interfering
18.
Blood ; 106(1): 227-34, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15746079

ABSTRACT

Oncogenic mutations of the Kit receptor tyrosine kinase occur in several types of malignancy. Juxtamembrane domain mutations are common in gastrointestinal stromal tumors, whereas mutations in the kinase activation loop, most commonly D816V, are seen in systemic mastocytosis and acute myelogenous leukemia. Kit activation-loop mutants are insensitive to imatinib mesylate and have been largely resistant to targeted inhibition. We determined the sensitivities of both Kit mutant classes to the adenosine triphosphate (ATP)-based inhibitors AP23464 and AP23848. In cell lines expressing activation-loop mutants, low-nM concentrations of AP23464 inhibited phosphorylation of Kit and its downstream targets Akt and signal transducer and activator of transcription 3 (STAT3). This was associated with cell-cycle arrest and apoptosis. Wild-type Kit-and juxtamembrane-mutant-expressing cell lines required considerably higher concentrations for equivalent inhibition, suggesting a therapeutic window in which cells harboring D816V Kit could be eliminated without interfering with normal cellular function. Additionally, AP23464 did not disrupt normal hematopoietic progenitor-cell growth at concentrations that inhibited activation-loop mutants of Kit. In a murine model, AP23848 inhibited activation-loop mutant Kit phosphorylation and tumor growth. Thus, AP23464 and AP23848 potently and selectively target activation-loop mutants of Kit in vitro and in vivo and could have therapeutic potential against D816V-expressing malignancies.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Adenosine Triphosphate/metabolism , Animals , B-Lymphocytes/cytology , Cell Division/drug effects , Cell Division/immunology , Cell Line, Tumor , Gene Expression Regulation, Leukemic , Humans , In Vitro Techniques , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Mice , Mutagenesis , Phosphorylation/drug effects , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/chemistry , Signal Transduction/drug effects
19.
Cancer Res ; 65(4): 1335-42, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15735019

ABSTRACT

Src tyrosine kinase expression and activity are elevated during colon cancer progression. How this contributes to the malignant phenotype is not fully understood. We show that in KM12C colon carcinoma cells, expression of kinase-deficient Src proteins (SrcMF and Src251) does not alter cell growth. Src kinase activity is required for turnover of cell-matrix adhesions and, in particular, the Src-dependent phosphorylation of focal adhesion kinase (FAK) is required for their disassembly. Surprisingly, we found that expression of SrcMF or Src251 resulted in increased tyrosine phosphorylation of FAK on Tyr(407), Tyr(576), Tyr(577), and Tyr(861), which are considered to be Src kinase substrates. This Src kinase-independent phosphorylation of FAK required an intact Src SH2 domain that mediates association of Src and FAK at peripheral adhesions. Use of a novel highly potent and selective Src kinase inhibitor AP23464 combined with experiments in Src/Fyn/Yes-deficient fibroblasts showed that increased phosphorylation of FAK in cells expressing SrcMF did not require Src-like kinases. However, specific phosphorylation on Tyr(925) of FAK was not evident in SrcMF- or Src251-expressing cells, and lack of Src kinase-dependent phosphorylation on this site was associated with impaired adhesion turnover. Our data show that Src kinase activity is required for adhesion turnover associated with cell migration in cancer cells and that, in addition to the catalytic activity, Src also acts as an adaptor to recruit other kinases that can phosphorylate key substrates including FAK. These studies have implications for tumor progression with respect to the use of Src kinase inhibitors.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Animals , Binding Sites , Catalysis , Cell Adhesion/physiology , Cell Movement/physiology , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mice , Mice, Nude , Phosphorylation , Signal Transduction , Substrate Specificity , Tyrosine/metabolism , src Homology Domains
20.
Blood ; 104(8): 2532-9, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15256422

ABSTRACT

The deregulated, oncogenic tyrosine kinase Bcr-Abl causes chronic myeloid leukemia (CML). Imatinib mesylate (Gleevec, STI571), a Bcr-Abl kinase inhibitor, selectively inhibits proliferation and promotes apoptosis of CML cells. Despite the success of imatinib mesylate in the treatment of CML, resistance is observed, particularly in advanced disease. The most common imatinib mesylate resistance mechanism involves Bcr-Abl kinase domain mutations that impart varying degrees of drug insensitivity. AP23464, a potent adenosine 5'-triphosphate (ATP)-based inhibitor of Src and Abl kinases, displays antiproliferative activity against a human CML cell line and Bcr-Abl-transduced Ba/F3 cells (IC(50) = 14 nM; imatinib mesylate IC(50) = 350 nM). AP23464 ablates Bcr-Abl tyrosine phosphorylation, blocks cell cycle progression, and promotes apoptosis of Bcr-Abl-expressing cells. Biochemical assays with purified glutathione S transferase (GST)-Abl kinase domain confirmed that AP23464 directly inhibits Abl activity. Importantly, the low nanomolar cellular and biochemical inhibitory properties of AP23464 extend to frequently observed imatinib mesylate-resistant Bcr-Abl mutants, including nucleotide binding P-loop mutants Q252H, Y253F, E255K, C-terminal loop mutant M351T, and activation loop mutant H396P. AP23464 was ineffective against mutant T315I, an imatinib mesylate contact residue. The potency of AP23464 against imatinib mesylate-refractory Bcr-Abl and its distinct binding mode relative to imatinib mesylate warrant further investigation of AP23464 for the treatment of CML.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Mutation/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/chemistry , Amino Acids/genetics , Amino Acids/metabolism , Apoptosis/drug effects , Benzamides , Cell Cycle/drug effects , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/chemistry , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Milk Proteins/metabolism , Models, Molecular , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Piperazines/chemistry , Piperazines/pharmacology , Protein Structure, Tertiary , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , STAT5 Transcription Factor , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...