Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(14): 9401-9417, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37439511

ABSTRACT

We report the discovery of sulanemadlin (ALRN-6924), the first cell-permeating, stabilized α-helical peptide to enter clinical trials. ALRN-6924 is a "stapled peptide" that mimics the N-terminal domain of the p53 tumor suppressor protein. It binds with high affinity to both MDM2 and MDMX (also known as MDM4), the endogenous inhibitors of p53, to activate p53 signaling in cells having a non-mutant, or wild-type TP53 genotype (TP53-WT). Iterative structure-activity optimization endowed ALRN-6924 with favorable cell permeability, solubility, and pharmacokinetic and safety profiles. Intracellular proteolysis of ALRN-6924 forms a long-acting active metabolite with potent MDM2 and MDMX binding affinity and slow dissociation kinetics. At high doses, ALRN-6924 exhibits on-mechanism anticancer activity in TP53-WT tumor models. At lower doses, ALRN-6924 transiently arrests the cell cycle in healthy tissues to protect them from chemotherapy without protecting the TP53-mutant cancer cells. These results support the continued clinical evaluation of ALRN-6924 as an anticancer and chemoprotection agent.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Protein Binding , Peptides/chemistry , Antineoplastic Agents/chemistry , Cell Cycle Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 110(36): E3445-54, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23946421

ABSTRACT

Stapled α-helical peptides have emerged as a promising new modality for a wide range of therapeutic targets. Here, we report a potent and selective dual inhibitor of MDM2 and MDMX, ATSP-7041, which effectively activates the p53 pathway in tumors in vitro and in vivo. Specifically, ATSP-7041 binds both MDM2 and MDMX with nanomolar affinities, shows submicromolar cellular activities in cancer cell lines in the presence of serum, and demonstrates highly specific, on-target mechanism of action. A high resolution (1.7-Å) X-ray crystal structure reveals its molecular interactions with the target protein MDMX, including multiple contacts with key amino acids as well as a role for the hydrocarbon staple itself in target engagement. Most importantly, ATSP-7041 demonstrates robust p53-dependent tumor growth suppression in MDM2/MDMX-overexpressing xenograft cancer models, with a high correlation to on-target pharmacodynamic activity, and possesses favorable pharmacokinetic and tissue distribution properties. Overall, ATSP-7041 demonstrates in vitro and in vivo proof-of-concept that stapled peptides can be developed as therapeutically relevant inhibitors of protein-protein interaction and may offer a viable modality for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Binding, Competitive , Cell Line, Tumor , Crystallography, X-Ray , Female , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Models, Molecular , Neoplasms/metabolism , Neoplasms/pathology , Peptides/chemistry , Peptides/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Peptides, Cyclic/therapeutic use , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Rats , Rats, Long-Evans , Xenograft Model Antitumor Assays
3.
J Cell Physiol ; 225(3): 855-64, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20607799

ABSTRACT

TM601 is a synthetic polypeptide with sequence derived from the venom of the scorpion Leiurus quinquestriatus that has anti-neoplastic activity. It has recently been demonstrated to bind annexin A2 on cultured tumor and vascular endothelial cells and to suppress blood vessel growth on chick chorioallantoic membrane. In this study, we investigated the effects of TM601 in models of ocular neovascularization (NV). When administered by intraocular injection, intravenous injections, or periocular injections, TM601 significantly suppressed the development of choroidal NV at rupture sites in Bruch's membrane. Treatment of established choroidal NV with TM601 caused apoptosis of endothelial cells and regression of the NV. TM601 suppressed ischemia-induced and vascular endothelial growth factor-induced retinal NV and reduced excess vascular permeability induced by vascular endothelial growth factor. Immunostaining with an antibody directed against TM601 showed that after intraocular or periocular injection, TM601 selectively bound to choroidal or retinal NV and co-localized with annexin A2, which is undetectable in normal retinal and choroidal vessels, but is upregulated in endothelial cells participating in choroidal or retinal NV. Intraocular injection of plasminogen or tissue plasminogen activator, which like TM601 bind to annexin A2, also suppressed retinal NV. This study supports the hypothesis that annexin A2 is an important target for treatment of neovascular diseases and suggests that TM601, through its interaction with annexin A2, causes suppression and regression of ocular NV and reduces vascular leakage and thus may provide a new treatment for blinding diseases such as neovascular age-related macular degeneration and diabetic retinopathy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Annexin A2/metabolism , Bruch Membrane/blood supply , Choroidal Neovascularization/prevention & control , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Retinopathy of Prematurity/prevention & control , Scorpion Venoms/pharmacology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Capillary Permeability/drug effects , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fibrinolysin/administration & dosage , Humans , Infant, Newborn , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/physiopathology , Rhodopsin/genetics , Scorpion Venoms/administration & dosage , Scorpion Venoms/metabolism , Tissue Plasminogen Activator/administration & dosage , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
4.
Anticancer Res ; 30(1): 39-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20150615

ABSTRACT

UNLABELLED: Chemically synthesized chlorotoxin (TM601) has been studied as a tumor targeting peptide. In this study, the anti-angiogenic properties of TM601 are reported. MATERIALS AND METHODS: In vitro and in vivo models of angiogenesis and tumor growth were used to characterize the anti-angiogenic effects of TM601. RESULTS: TM601 bound to proliferating vascular endothelial cells, decreased human umbilical vein endothelial cell (HUVEC) invasion, and reduced secretion of bioactive matrix metalloproteinase-2 (MMP-2). Using the chick chorioallantoic membrane assay (CAM), TM601 inhibited angiogenesis stimulated by any of eight pro-angiogenic factors, and when TM601 was co-administered with bevacizumab, the combination was significantly more potent than a ten-fold increase in bevacizumab dose. TM601 did not alter tumor or vascular endothelial cell growth in vitro, but TM601 treatment of tumors grown on the CAM decreased tumor growth and intra-tumoral hemoglobin levels. Intravenously injected TM601 was also shown to significantly decrease new blood vessel growth in mice. CONCLUSION: TM601 inhibits angiogenesis stimulated by many factors and potentiates the anti-angiogenic effect of bevacizumab.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neurotoxins/pharmacology , Scorpion Venoms/pharmacology , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Growth Processes/drug effects , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/blood supply , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Physiologic/drug effects , Neurotoxins/pharmacokinetics , Scorpion Venoms/pharmacokinetics
5.
J Biol Chem ; 285(7): 4366-74, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20018898

ABSTRACT

TM601 is a synthetic form of chlorotoxin, a 36-amino acid peptide derived from the venom of the Israeli scorpion, Leirius quinquestriatus, initially found to specifically bind and inhibit the migration of glioma cells in culture. Subsequent studies demonstrated specific in vitro binding to additional tumor cell lines. Recently, we demonstrated that proliferating human vascular endothelial cells are the only normal cell line tested that exhibits specific binding to TM601. Here, we identify annexin A2 as a novel binding partner for TM601 in multiple human tumor cell lines and human umbilical vein endothelial cell (HUVEC). We demonstrate that the surface binding of TM601 to the pancreatic tumor cell line Panc-1 is dependent on the expression of annexin A2. Identification of annexin A2 as a binding partner for TM601 is also consistent with the anti-angiogenic effects of TM601. Annexin A2 functions in angiogenesis by binding to tissue plasminogen activator and regulating plasminogen activation on vascular endothelial cells. We demonstrate that in HUVECs, TM601 inhibits both vascular endothelial growth factor- and basic fibroblast growth factor-induced tissue plasminogen activator activation, which is required for activation of plasminogen to plasmin. Consistent with inhibition of cell surface protease activity, TM601 also inhibits platelet-derived growth factor-C induced trans-well migration of both HUVEC and U373-MG glioma cells.


Subject(s)
Angiogenesis Inhibitors/metabolism , Annexin A2/metabolism , Antineoplastic Agents/metabolism , Scorpion Venoms/metabolism , Annexin A2/genetics , Biotinylation , Blotting, Western , Cell Line , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Endothelial Cells , Humans , Mass Spectrometry , Protein Binding/genetics , Protein Binding/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology
6.
J Immunol ; 173(6): 3935-44, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15356142

ABSTRACT

Signaling through the Notch1 receptor is essential for T cell development in the thymus. Stromal OP9 cells ectopically expressing the Notch ligand Delta-like1 mimic the thymic environment by inducing hemopoietic stem cells to undergo in vitro T cell development. Notch1 is also expressed on Pax5-/- pro-B cells, which are clonable lymphoid progenitors with a latent myeloid potential. In this study, we demonstrate that Pax5-/- progenitors efficiently differentiate in vitro into CD4+CD8+ alphabeta and gammadelta T cells upon coculture with OP9-Delta-like1 cells. In vitro T cell development of Pax5-/- progenitors strictly depends on Notch1 function and progresses through normal developmental stages by expressing T cell markers and rearranging TCRbeta, gamma, and delta loci in the correct temporal sequence. Notch-stimulated Pax5-/- progenitors efficiently down-regulate the expression of B cell-specific genes, consistent with a role of Notch1 in preventing B lymphopoiesis in the thymus. At the same time, Notch signaling rapidly induces cell surface expression of the c-Kit receptor and transcription of the target genes Deltex1 and pre-Talpha concomitant with the activation of TCR Vbeta germline transcription and the regulatory genes GATA3 and Tcf1. These data suggest that Notch1 acts upstream of GATA3 and Tcf1 in early T cell development and regulates Vbeta-DJbeta rearrangements by controlling the chromatin accessibility of Vbeta genes at the TCRbeta locus.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Receptors, Cell Surface/physiology , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Lineage/genetics , Cell Lineage/immunology , Clone Cells , Coculture Techniques , DNA-Binding Proteins/physiology , Down-Regulation/genetics , Down-Regulation/immunology , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , PAX5 Transcription Factor , Receptor, Notch1 , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/physiology , Stromal Cells/physiology , T-Lymphocyte Subsets/metabolism
7.
J Cell Physiol ; 199(1): 140-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14978743

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are regulated by MAPK kinases (MKKs), which are in turn regulated by MKK kinases (MKKKs). While a single MKKK can regulate several different MAPK family members, and several MKKKs can often activate the same MAPK, emerging evidence indicates a unique role for individual MKKKs in acting as signaling nodes to coordinately activate different subsets of MAPKs in response to specific cellular stimuli. Thus, while there is much apparent overlap in MAPK regulation by different MKKKs, each MKKK serves a specific purpose in regulation of unique cellular functions. The purpose of this study was to define the specific role of MEKK2, an MKKK, in MAPK regulation and cell function. MEKK2 coordinately activates the ERK5 and JNK pathways. Targeted disruption of MEKK2 expression causes loss of ERK5 and JNK activation in response to FGF-2 in mouse embryonic fibroblasts (MEFs). FGF-2 receptor signaling requires MEKK2 for induction of mRNA for c-Jun, Fra-1, and Fra-2, components of the AP-1 transcription complex. In FGF-2-stimulated MEKK2-/- fibroblasts, c-Jun phosphorylation is inhibited, consistent with a loss of JNK activation. Thus, MEKK2 regulates AP-1 activity at two levels, by regulating both expression of AP-1 components and c-Jun N-terminal phosphorylation. One function of the AP-1 transcription complex is to regulate cytokine gene expression. Expression of IL-1alpha, IL-1beta, IL-6, and TNFalpha is inhibited in MEKK2-/- fibroblasts. Bacterial lipopolysaccharide (LPS) and TNFalpha neither activate ERK5 nor require MEKK2 for JNK activation, demonstrating specificity of MEKK2 in FGF-2 receptor signaling and control of cytokine gene expression.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Fibroblasts/drug effects , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/drug effects , Cytokines/genetics , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Fibroblasts/enzymology , Fluorescent Antibody Technique , Gene Expression Regulation , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinase 2 , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinases/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor AP-1/physiology , Transcription, Genetic
8.
EMBO J ; 22(13): 3346-55, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12839996

ABSTRACT

Herein, we define how MEKK1, a MAPK kinase kinase, regulates cell migration. MEKK1 is associated with actin fibers and focal adhesions, localizing MEKK1 to sites critical in the control of cell adhesion and migration. EGF-induced ERK1/2 activation and chemotaxis are inhibited in MEKK1-/- fibroblasts. MEKK1 deficiency causes loss of vinculin in focal adhesions of migrating cells, increased cell adhesion and impeded rear-end detachment. MEKK1 is required for activation of the cysteine protease calpain and cleavage of spectrin and talin, proteins linking focal adhesions to the cytoskeleton. Inhibition of ERK1/2 or calpain, but not of JNK, mimics MEKK1 deficiency. Therefore, MEKK1 regulates calpain-mediated substratum release of migrating fibroblasts.


Subject(s)
Calpain/physiology , Cell Movement , MAP Kinase Kinase Kinase 1 , Protein Serine-Threonine Kinases/physiology , Vinculin/metabolism , Animals , Cells, Cultured , Enzyme Activation , Epidermal Growth Factor/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescence , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Hydrolysis , Mice , Papillomaviridae/physiology , Protein-Tyrosine Kinases/metabolism
9.
Mol Cell Biol ; 23(7): 2298-308, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12640115

ABSTRACT

Lad is an SH2 domain-containing adaptor protein that binds MEK kinase 2 (MEKK2), a mitogen-activated protein kinase (MAPK) kinase kinase for the extracellular signal-regulated kinase 5 (ERK5) and JNK pathways. Lad and MEKK2 are in a complex in resting cells. Antisense knockdown of Lad expression and targeted gene disruption of MEKK2 expression results in loss of epidermal growth factor (EGF) and stress stimuli-induced activation of ERK5. Activation of MEKK2 and the ERK5 pathway by EGF and stress stimuli is dependent on Src kinase activity. The Lad-binding motif is encoded within amino acids 228 to 282 in the N terminus of MEKK2, and expression of this motif blocks Lad-MEKK2 interaction, resulting in inhibition of Src-dependent activation of MEKK2 and ERK5. JNK activation by EGF is similarly inhibited by loss of Lad or MEKK2 expression and by blocking the interaction of MEKK2 and Lad. Our studies demonstrate that Src kinase activity is required for ERK5 activation in response to EGF, MEKK2 expression is required for ERK5 activation by Src, Lad and MEKK2 association is required for Src activation of ERK5, and EGF and Src stimulation of ERK5-regulated MEF2-dependent promoter activity requires a functional Lad-MEKK2 signaling complex.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Epidermal Growth Factor/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism , Amino Acid Motifs/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Kidney/cytology , Kidney/metabolism , MAP Kinase Kinase Kinase 2 , MAP Kinase Kinase Kinases/genetics , Macromolecular Substances , Mice , Mink , Mitogen-Activated Protein Kinase 7 , Osmolar Concentration , Oxidative Stress/physiology , Protein Binding/physiology , Signal Transduction/physiology , Transfection , Two-Hybrid System Techniques
10.
J Biol Chem ; 277(17): 14666-73, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-11847223

ABSTRACT

The lymphocyte-specific protein-tyrosine kinase Lck plays a critical role in T cell activation. In response to T cell antigen receptor binding Lck undergoes phosphorylation on serine residues that include serines 59 and 194. Serine 59 is phosphorylated by ERK mitogen-activated protein kinase. Recently, we showed that in mitotic T cells Lck becomes hyper-phosphorylated on serine residues. In this report, using one-dimensional phosphopeptide mapping analysis, we identify serine 59 as a site of in vivo mitotic phosphorylation in Lck. The mitotic phosphorylation of serine 59 did not require either the catalytic activity or functional SH2 or SH3 domains of Lck. In addition, the presence of ZAP-70 also was dispensable for the phosphorylation of serine 59. Although previous studies demonstrated that serine 59 is a substrate for the ERK MAPK pathway, inhibitors of this pathway did not block the mitotic phosphorylation of serine 59. These results identify serine 59 as a site of mitotic phosphorylation in Lck and suggest that a pathway distinct from that induced by antigen receptor signaling is responsible for its phosphorylation. Thus, the phosphorylation of serine 59 is the result of two distinct signaling pathways, differentially activated in response to the physiological state of the T cell.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mitosis , Serine/metabolism , Animals , Catalysis , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , MAP Kinase Signaling System , Mice , Peptide Mapping , Phosphorylation , Protein-Tyrosine Kinases/metabolism , ZAP-70 Protein-Tyrosine Kinase
11.
J Biol Chem ; 277(7): 5683-91, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11741956

ABSTRACT

Lck is a member of the Src family of protein-tyrosine kinases and is essential for T cell development and function. Lck is localized to the inner surface of the plasma membrane and partitions into lipid rafts via dual acylation on its N terminus. We have tested the role of Lck binding domains in regulating Lck localization to lipid rafts. A form of Lck containing a point mutation inactivating the SH3 domain (W97ALck) was preferentially localized to lipid rafts compared with wild type or SH2 domain-inactive (R154K) Lck when expressed in Lck-deficient J.CaM1 cells. W97ALck incorporated more of the radioiodinated version of palmitic acid, 16-[(125)I]iodohexadecanoic acid. Overexpression of c-Cbl, a ligand of the Lck SH3 domain, depleted Lck from lipid rafts in Jurkat cells. Additionally, Lck localization to lipid rafts was enhanced in c-Cbl-deficient T cells. The association of Lck with c-Cbl in vivo required a functional SH3 domain. These results suggest a model whereby the SH3 domain negatively regulates basal localization of Lck to lipid rafts via association with c-Cbl.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Membrane Microdomains/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases , Animals , Cell Line , Chickens , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Green Fluorescent Proteins , Humans , Jurkat Cells , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Point Mutation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-cbl , Transfection , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...