Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 64(5): 2501-2520, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33631934

ABSTRACT

SUMOylation is a reversible post-translational modification that regulates protein function through covalent attachment of small ubiquitin-like modifier (SUMO) proteins. The process of SUMOylating proteins involves an enzymatic cascade, the first step of which entails the activation of a SUMO protein through an ATP-dependent process catalyzed by SUMO-activating enzyme (SAE). Here, we describe the identification of TAK-981, a mechanism-based inhibitor of SAE which forms a SUMO-TAK-981 adduct as the inhibitory species within the enzyme catalytic site. Optimization of selectivity against related enzymes as well as enhancement of mean residence time of the adduct were critical to the identification of compounds with potent cellular pathway inhibition and ultimately a prolonged pharmacodynamic effect and efficacy in preclinical tumor models, culminating in the identification of the clinical molecule TAK-981.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Sulfonic Acids/therapeutic use , Sumoylation/drug effects , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Humans , Mice , Molecular Structure , Protein Binding , Protein Processing, Post-Translational/drug effects , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/metabolism , Ubiquitin-Activating Enzymes/metabolism , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 19(10): 2079-2088, 2020 10.
Article in English | MEDLINE | ID: mdl-32788205

ABSTRACT

Guanylyl cyclase C (GCC) is a unique therapeutic target with expression restricted to the apical side of epithelial cell tight junctions thought to be only accessible by intravenously administered agents on malignant tissues where GCC expression is aberrant. In this study, we sought to evaluate the therapeutic potential of a second-generation investigational antibody-dug conjugate (ADC), TAK-164, comprised of a human anti-GCC mAb conjugated via a peptide linker to the highly cytotoxic DNA alkylator, DGN549. The in vitro binding, payload release, and in vitro activity of TAK-164 was characterized motivating in vivo evaluation. The efficacy of TAK-164 and the relationship to exposure, pharmacodynamic marker activation, and biodistribution was evaluated in xenograft models and primary human tumor xenograft (PHTX) models. We demonstrate TAK-164 selectively binds to, is internalized by, and has potent cytotoxic effects against GCC-expressing cells in vitro A single intravenous administration of TAK-164 (0.76 mg/kg) resulted in significant growth rate inhibition in PHTX models of metastatic colorectal cancer. Furthermore, imaging studies characterized TAK-164 uptake and activity and showed positive relationships between GCC expression and tumor uptake which correlated with antitumor activity. Collectively, our data suggest that TAK-164 is highly active in multiple GCC-positive tumors including those refractory to TAK-264, a GCC-targeted auristatin ADC. A strong relationship between uptake of 89Zr-labeled TAK-164, levels of GCC expression and, most notably, response to TAK-164 therapy in GCC-expressing xenografts and PHTX models. These data supported the clinical development of TAK-164 as part of a first-in-human clinical trial (NCT03449030).


Subject(s)
Immunoconjugates/therapeutic use , Animals , Female , HEK293 Cells , Humans , Immunoconjugates/pharmacology , Mice , Mice, Nude , Tissue Distribution , Xenograft Model Antitumor Assays
3.
PLoS One ; 13(1): e0191046, 2018.
Article in English | MEDLINE | ID: mdl-29370189

ABSTRACT

Guanylyl cyclase C (GCC) is a cell-surface protein that is expressed by normal intestinal epithelial cells, more than 95% of metastatic colorectal cancers (mCRC), and the majority of gastric and pancreatic cancers. Due to strict apical localization, systemically delivered GCC-targeting agents should not reach GCC in normal intestinal tissue, while accessing antigen in tumor. We generated an investigational antibody-drug conjugate (TAK-264, formerly MLN0264) comprising a fully human anti-GCC monoclonal antibody conjugated to monomethyl auristatin E via a protease-cleavable peptide linker. TAK-264 specifically bound, was internalized by, and killed GCC-expressing cells in vitro in an antigen-density-dependent manner. In GCC-expressing xenograft models with similar GCC expression levels/patterns observed in human mCRC samples, TAK-264 induced cell death, leading to tumor regressions and long-term tumor growth inhibition. TAK-264 antitumor activity was generally antigen-density-dependent, although some GCC-expressing tumors were refractory to TAK-264-targeted high local concentrations of payload. These data support further evaluation of TAK-264 in the treatment of GCC-expressing tumors.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoconjugates/pharmacology , Oligopeptides/metabolism , Receptors, Enterotoxin/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Blotting, Western , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , HEK293 Cells , Humans , Intestinal Mucosa/enzymology , Mice , Mice, SCID , Receptors, Enterotoxin/genetics , Receptors, Enterotoxin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
4.
Nat Chem Biol ; 13(11): 1164-1171, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28892090

ABSTRACT

Small ubiquitin-like modifier (SUMO) family proteins regulate target-protein functions by post-translational modification. However, a potent and selective inhibitor targeting the SUMO pathway has been lacking. Here we describe ML-792, a mechanism-based SUMO-activating enzyme (SAE) inhibitor with nanomolar potency in cellular assays. ML-792 selectively blocks SAE enzyme activity and total SUMOylation, thus decreasing cancer cell proliferation. Moreover, we found that induction of the MYC oncogene increased the ML-792-mediated viability effect in cancer cells, thus indicating a potential application of SAE inhibitors in treating MYC-amplified tumors. Using ML-792, we further explored the critical roles of SUMOylation in mitotic progression and chromosome segregation. Furthermore, expression of an SAE catalytic-subunit (UBA2) S95N M97T mutant rescued SUMOylation loss and the mitotic defect induced by ML-792, thus confirming the selectivity of ML-792. As a potent and selective SAE inhibitor, ML-792 provides rapid loss of endogenously SUMOylated proteins, thereby facilitating novel insights into SUMO biology.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Sumoylation , Cell Proliferation/drug effects , Chromosome Segregation/drug effects , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Humans , Mitosis/drug effects , Neoplasms/genetics , Neoplasms/pathology , Protein Processing, Post-Translational , Tumor Cells, Cultured
5.
Mol Cancer Ther ; 6(1): 262-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237285

ABSTRACT

Poh1 deubiquitinase activity is required for proteolytic processing of polyubiquitinated substrates by the 26S proteasome, linking deubiquitination to complete substrate degradation. Poh1 RNA interference (RNAi) in HeLa cells resulted in a reduction in cell viability and an increase in polyubiquitinated protein levels, supporting the link between Poh1 and the ubiquitin proteasome pathway. To more specifically test for any requirement of the zinc metalloproteinase motif of Poh1 to support cell viability and proteasome function, we developed a RNAi complementation strategy. Effects on cell viability and proteasome activity were assessed in cells with RNAi of endogenous Poh1 and induced expression of wild-type Poh1 or a mutant form of Poh1, in which two conserved histidines of the proposed catalytic site were replaced with alanines. We show that an intact zinc metalloproteinase motif is essential for cell viability and 26S proteasome function. As a required enzymatic component of the proteasome, Poh1 is an intriguing therapeutic drug target for cancer.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Ubiquitin/metabolism , Amino Acid Motifs , Cell Survival , HeLa Cells , Humans , Mutant Proteins/metabolism , Proteasome Endopeptidase Complex/deficiency , RNA Interference , Trans-Activators/deficiency
6.
J Leukoc Biol ; 72(3): 492-502, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12223517

ABSTRACT

Gangliosides of macrophages have immunoregulatory and structural attributes, distinct from neural gangliosides. We previously produced a monoclonal antibody to human macrophage gangliosides (HMG; mAb25F4), which inhibited macrophage migration and recognized a surface-accessible epitope. We investigated expanded immunoregulatory properties and molecular domains for antibody recognition. mAb25F4 directly induced human macrophage production of proinflammatory cytokines, interleukin-1beta, and tumor necrosis factor alpha. Conditions were established for selective, reversible depletion of HMG with D-threo-(R,R)-1-phenyl-2-decanoyl-amino-3-morpholine-1-propanol. mAb25F4 had diminished recognition for ganglioside-depleted macrophages, which was restored with regeneration of gangliosides. Although desialylation of HMG did not impair mAb25F4 recognition, enzymatic cleavage of ceramide abolished antibody binding. Antibody recognition was specific for the ceramide fraction, with preferential recognition for ceramide of HMG and murine macrophage gangliosides and limited recognition for neural tissue ceramide and gangliosides. This study underscores the importance of structurally distinct ceramide of macrophage gangliosides as a critical domain for ganglioside-mediated activation of human macrophages.


Subject(s)
Ceramides/pharmacology , Gangliosides/immunology , Macrophage Activation/physiology , Animals , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Cytokines/biosynthesis , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Female , Gangliosides/metabolism , Glycolipids/analysis , Glycoside Hydrolases/pharmacology , Humans , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C3H , Morpholines/pharmacology , Neuraminidase/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...