Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Proc Natl Acad Sci U S A ; 110(50): 20224-9, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277854

ABSTRACT

Wnt signaling is one of the key oncogenic pathways in multiple cancers, and targeting this pathway is an attractive therapeutic approach. However, therapeutic success has been limited because of the lack of therapeutic agents for targets in the Wnt pathway and the lack of a defined patient population that would be sensitive to a Wnt inhibitor. We developed a screen for small molecules that block Wnt secretion. This effort led to the discovery of LGK974, a potent and specific small-molecule Porcupine (PORCN) inhibitor. PORCN is a membrane-bound O-acyltransferase that is required for and dedicated to palmitoylation of Wnt ligands, a necessary step in the processing of Wnt ligand secretion. We show that LGK974 potently inhibits Wnt signaling in vitro and in vivo, including reduction of the Wnt-dependent LRP6 phosphorylation and the expression of Wnt target genes, such as AXIN2. LGK974 is potent and efficacious in multiple tumor models at well-tolerated doses in vivo, including murine and rat mechanistic breast cancer models driven by MMTV-Wnt1 and a human head and neck squamous cell carcinoma model (HN30). We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide both a strategy and tools for targeting Wnt-driven cancers through the inhibition of PORCN.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Pyrazines/pharmacology , Pyridines/pharmacology , Wnt Signaling Pathway/drug effects , Acyltransferases , Animals , Axin Protein/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Cloning, Molecular , High-Throughput Screening Assays , Humans , Mice , Mutagenesis , Phosphorylation/drug effects , Pyrazines/therapeutic use , Pyridines/therapeutic use , Radioligand Assay , Rats , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
Clin Cancer Res ; 27(1): 342-354, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33046519

ABSTRACT

PURPOSE: SHP2 inhibitors offer an appealing and novel approach to inhibit receptor tyrosine kinase (RTK) signaling, which is the oncogenic driver in many tumors or is frequently feedback activated in response to targeted therapies including RTK inhibitors and MAPK inhibitors. We seek to evaluate the efficacy and synergistic mechanisms of combinations with a novel SHP2 inhibitor, TNO155, to inform their clinical development. EXPERIMENTAL DESIGN: The combinations of TNO155 with EGFR inhibitors (EGFRi), BRAFi, KRASG12Ci, CDK4/6i, and anti-programmed cell death-1 (PD-1) antibody were tested in appropriate cancer models in vitro and in vivo, and their effects on downstream signaling were examined. RESULTS: In EGFR-mutant lung cancer models, combination benefit of TNO155 and the EGFRi nazartinib was observed, coincident with sustained ERK inhibition. In BRAFV600E colorectal cancer models, TNO155 synergized with BRAF plus MEK inhibitors by blocking ERK feedback activation by different RTKs. In KRASG12C cancer cells, TNO155 effectively blocked the feedback activation of wild-type KRAS or other RAS isoforms induced by KRASG12Ci and greatly enhanced efficacy. In addition, TNO155 and the CDK4/6 inhibitor ribociclib showed combination benefit in a large panel of lung and colorectal cancer patient-derived xenografts, including those with KRAS mutations. Finally, TNO155 effectively inhibited RAS activation by colony-stimulating factor 1 receptor, which is critical for the maturation of immunosuppressive tumor-associated macrophages, and showed combination activity with anti-PD-1 antibody. CONCLUSIONS: Our findings suggest TNO155 is an effective agent for blocking both tumor-promoting and immune-suppressive RTK signaling in RTK- and MAPK-driven cancers and their tumor microenvironment. Our data provide the rationale for evaluating these combinations clinically.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Mice , Mutation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Xenograft Model Antitumor Assays
3.
Cancer Res ; 80(19): 4278-4287, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32747364

ABSTRACT

Advanced ovarian cancers are a leading cause of cancer-related death in women and are currently treated with surgery and chemotherapy. This standard of care is often temporarily successful but exhibits a high rate of relapse, after which, treatment options are few. Here we investigate whether biomarker-guided use of multiple targeted therapies, including small molecules and antibody-drug conjugates, is a viable alternative. A panel of patient-derived ovarian cancer xenografts (PDX), similar in genetics and chemotherapy responsiveness to human tumors, was exposed to 21 monotherapies and combination therapies. Three monotherapies and one combination were found to be active in different subsets of PDX. Analysis of gene expression data identified biomarkers associated with responsiveness to each of the three targeted therapies, none of which directly inhibits an oncogenic driver. While no single treatment had as high a response rate as chemotherapy, nearly 90% of PDXs were eligible for and responded to at least one biomarker-guided treatment, including tumors resistant to standard chemotherapy. The distribution of biomarker positivity in The Cancer Genome Atlas data suggests the potential for a similar precision approach in human patients. SIGNIFICANCE: This study exploits a panel of patient-derived xenografts to demonstrate that most ovarian tumors can be matched to effective biomarker-guided treatments.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Ovarian Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Antineoplastic Agents/pharmacology , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Molecular Targeted Therapy/methods , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Precision Medicine , Proof of Concept Study
4.
Nat Med ; 21(11): 1318-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26479923

ABSTRACT

Profiling candidate therapeutics with limited cancer models during preclinical development hinders predictions of clinical efficacy and identifying factors that underlie heterogeneous patient responses for patient-selection strategies. We established ∼1,000 patient-derived tumor xenograft models (PDXs) with a diverse set of driver mutations. With these PDXs, we performed in vivo compound screens using a 1 × 1 × 1 experimental design (PDX clinical trial or PCT) to assess the population responses to 62 treatments across six indications. We demonstrate both the reproducibility and the clinical translatability of this approach by identifying associations between a genotype and drug response, and established mechanisms of resistance. In addition, our results suggest that PCTs may represent a more accurate approach than cell line models for assessing the clinical potential of some therapeutic modalities. We therefore propose that this experimental paradigm could potentially improve preclinical evaluation of treatment modalities and enhance our ability to predict clinical trial responses.


Subject(s)
Antineoplastic Agents/therapeutic use , High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Colorectal Neoplasms/drug therapy , Disease Models, Animal , Female , Humans , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Mice , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Reproducibility of Results , Skin Neoplasms/drug therapy , Stomach Neoplasms/drug therapy
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL