Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Discov ; 7(9): 1030-1045, 2017 09.
Article in English | MEDLINE | ID: mdl-28526733

ABSTRACT

Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.


Subject(s)
Antineoplastic Agents/therapeutic use , Cadherins/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cadherins/genetics , Cadherins/metabolism , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Macaca fascicularis , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Rats , Xenograft Model Antitumor Assays
2.
MAbs ; 6(6): 1560-70, 2014.
Article in English | MEDLINE | ID: mdl-25484045

ABSTRACT

Multiple therapeutic agonists of death receptor 5 (DR5) have been developed and are under clinical evaluation. Although these agonists demonstrate significant anti-tumor activity in preclinical models, the clinical efficacy in human cancer patients has been notably disappointing. One possible explanation might be that the current classes of therapeutic molecules are not sufficiently potent to elicit significant response in patients, particularly for dimeric antibody agonists that require secondary cross-linking via Fcγ receptors expressed on immune cells to achieve optimal clustering of DR5. To overcome this limitation, a novel multivalent Nanobody approach was taken with the goal of generating a significantly more potent DR5 agonist. In the present study, we show that trivalent DR5 targeting Nanobodies mimic the activity of natural ligand, and furthermore, increasing the valency of domains to tetramer and pentamer markedly increased potency of cell killing on tumor cells, with pentamers being more potent than tetramers in vitro. Increased potency was attributed to faster kinetics of death-inducing signaling complex assembly and caspase-8 and caspase-3 activation. In vivo, multivalent Nanobody molecules elicited superior anti-tumor activity compared to a conventional DR5 agonist antibody, including the ability to induce tumor regression in an insensitive patient-derived primary pancreatic tumor model. Furthermore, complete responses to Nanobody treatment were obtained in up to 50% of patient-derived primary pancreatic and colon tumor models, suggesting that multivalent DR5 Nanobodies may represent a significant new therapeutic modality for targeting death receptor signaling.


Subject(s)
Caspases/immunology , Neoplasms/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Single-Domain Antibodies/immunology , Animals , Antibody Affinity/immunology , Blotting, Western , Caspases/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , HCT116 Cells , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Protein Multimerization , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
3.
Sci Transl Med ; 2(51): 51ra70, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20881279

ABSTRACT

The malignant brain cancer medulloblastoma is characterized by mutations in Hedgehog (Hh) signaling pathway genes, which lead to constitutive activation of the G protein (heterotrimeric guanosine triphosphate-binding protein)-coupled receptor Smoothened (Smo). The Smo antagonist NVP-LDE225 inhibits Hh signaling and induces tumor regression in animal models of medulloblastoma. However, evidence of resistance was observed during the course of treatment. Molecular analysis of resistant tumors revealed several resistance mechanisms. We noted chromosomal amplification of Gli2, a downstream effector of Hh signaling, and, more rarely, point mutations in Smo that led to reactivated Hh signaling and restored tumor growth. Analysis of pathway gene expression signatures also, unexpectedly, identified up-regulation of phosphatidylinositol 3-kinase (PI3K) signaling in resistant tumors as another potential mechanism of resistance. Probing the relevance of increased PI3K signaling, we demonstrated that addition of the PI3K inhibitor NVP-BKM120 or the dual PI3K-mTOR (mammalian target of rapamycin) inhibitor NVP-BEZ235 to the initial treatment with the Smo antagonist markedly delayed the development of resistance. Our findings may be useful in informing treatment strategies for medulloblastoma.


Subject(s)
Aminopyridines/pharmacology , Drug Resistance, Neoplasm/drug effects , Medulloblastoma/enzymology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Aminopyridines/therapeutic use , Animals , Cell Proliferation/drug effects , Gene Amplification/drug effects , Hedgehog Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Kruppel-Like Transcription Factors/metabolism , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Morpholines/therapeutic use , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Zinc Finger Protein Gli2
4.
Proc Natl Acad Sci U S A ; 107(35): 15473-8, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713706

ABSTRACT

Disregulated Wnt/beta-catenin signaling has been linked to various human diseases, including cancers. Inhibitors of oncogenic Wnt signaling are likely to have a therapeutic effect in cancers. LRP5 and LRP6 are closely related membrane coreceptors for Wnt proteins. Using a phage-display library, we identified anti-LRP6 antibodies that either inhibit or enhance Wnt signaling. Two classes of LRP6 antagonistic antibodies were discovered: one class specifically inhibits Wnt proteins represented by Wnt1, whereas the second class specifically inhibits Wnt proteins represented by Wnt3a. Epitope-mapping experiments indicated that Wnt1 class-specific antibodies bind to the first propeller and Wnt3a class-specific antibodies bind to the third propeller of LRP6, suggesting that Wnt1- and Wnt3a-class proteins interact with distinct LRP6 propeller domains. This conclusion is further supported by the structural functional analysis of LRP5/6 and the finding that the Wnt antagonist Sclerostin interacts with the first propeller of LRP5/6 and preferentially inhibits the Wnt1-class proteins. We also show that Wnt1 or Wnt3a class-specific anti-LRP6 antibodies specifically block growth of MMTV-Wnt1 or MMTV-Wnt3 xenografts in vivo. Therapeutic application of these antibodies could be limited without knowing the type of Wnt proteins expressed in cancers. This is further complicated by our finding that bivalent LRP6 antibodies sensitize cells to the nonblocked class of Wnt proteins. The generation of a biparatopic LRP6 antibody blocks both Wnt1- and Wnt3a-mediated signaling without showing agonistic activity. Our studies provide insights into Wnt-induced LRP5/6 activation and show the potential utility of LRP6 antibodies in Wnt-driven cancer.


Subject(s)
Antibodies/pharmacology , LDL-Receptor Related Proteins/immunology , Ligands , Wnt Proteins/metabolism , Animals , Antibodies/immunology , Cell Line , Cell Transformation, Viral , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6 , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Protein Binding/drug effects , Signal Transduction/drug effects , Tumor Burden/drug effects , Wnt Proteins/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Wnt3 Protein , Wnt3A Protein , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...