Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Cancer Discov ; 13(10): 2131-2149, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37712571

ABSTRACT

Small-molecule drugs have enabled the practice of precision oncology for genetically defined patient populations since the first approval of imatinib in 2001. Scientific and technology advances over this 20-year period have driven the evolution of cancer biology, medicinal chemistry, and data science. Collectively, these advances provide tools to more consistently design best-in-class small-molecule drugs against known, previously undruggable, and novel cancer targets. The integration of these tools and their customization in the hands of skilled drug hunters will be necessary to enable the discovery of transformational therapies for patients across a wider spectrum of cancers. SIGNIFICANCE: Target-centric small-molecule drug discovery necessitates the consideration of multiple approaches to identify chemical matter that can be optimized into drug candidates. To do this successfully and consistently, drug hunters require a comprehensive toolbox to avoid following the "law of instrument" or Maslow's hammer concept where only one tool is applied regardless of the requirements of the task. Combining our ever-increasing understanding of cancer and cancer targets with the technological advances in drug discovery described below will accelerate the next generation of small-molecule drugs in oncology.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Data Science , Precision Medicine , Drug Discovery , Biology
3.
Nat Commun ; 14(1): 3830, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37380628

ABSTRACT

Combination of anti-cancer drugs is broadly seen as way to overcome the often-limited efficacy of single agents. The design and testing of combinations are however very challenging. Here we present a uniquely large dataset screening over 5000 targeted agent combinations across 81 non-small cell lung cancer cell lines. Our analysis reveals a profound heterogeneity of response across the tumor models. Notably, combinations very rarely result in a strong gain in efficacy over the range of response observable with single agents. Importantly, gain of activity over single agents is more often seen when co-targeting functionally proximal genes, offering a strategy for designing more efficient combinations. Because combinatorial effect is strongly context specific, tumor specificity should be achievable. The resource provided, together with an additional validation screen sheds light on major challenges and opportunities in building efficacious combinations against cancer and provides an opportunity for training computational models for synergy prediction.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Drug Combinations
4.
Mol Syst Biol ; 15(3): e8323, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858180

ABSTRACT

Most patients with advanced cancer eventually acquire resistance to targeted therapies, spurring extensive efforts to identify molecular events mediating therapy resistance. Many of these events involve synthetic rescue (SR) interactions, where the reduction in cancer cell viability caused by targeted gene inactivation is rescued by an adaptive alteration of another gene (the rescuer). Here, we perform a genome-wide in silico prediction of SR rescuer genes by analyzing tumor transcriptomics and survival data of 10,000 TCGA cancer patients. Predicted SR interactions are validated in new experimental screens. We show that SR interactions can successfully predict cancer patients' response and emerging resistance. Inhibiting predicted rescuer genes sensitizes resistant cancer cells to therapies synergistically, providing initial leads for developing combinatorial approaches to overcome resistance proactively. Finally, we show that the SR analysis of melanoma patients successfully identifies known mediators of resistance to immunotherapy and predicts novel rescuers.


Subject(s)
Computational Biology , Drug Resistance, Neoplasm/genetics , Drug Synergism , Melanoma/genetics , Female , Gene Expression Profiling , Humans , Immunotherapy , Male , Melanoma/drug therapy , Molecular Targeted Therapy , Synthetic Lethal Mutations
5.
Clin Cancer Res ; 23(16): 4680-4692, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28446504

ABSTRACT

Purpose: Successful development of targeted therapy combinations for cancer patients depends on first discovering such combinations in predictive preclinical models. Stable cell lines and mouse xenograft models can have genetic and phenotypic drift and may take too long to generate to be useful as a personalized medicine tool.Experimental Design: To overcome these limitations, we have used a platform of ultra-high-throughput functional screening of primary biopsies preserving both cancer and stroma cell populations from melanoma patients to nominate such novel combinations from a library of thousands of drug combinations in a patient-specific manner within days of biopsy. In parallel, patient-derived xenograft (PDX) mouse models were created and novel combinations tested for their ability to shrink matched PDXs.Results: The screening method identifies specific drug combinations in tumor cells with patterns that are distinct from those obtained from stable cell lines. Screening results were highly specific to individual patients. For patients with matched PDX models, we confirmed that individualized novel targeted therapy combinations could inhibit tumor growth. In particular, a combination of multi-kinase and PI3K/Akt inhibitors was effective in some BRAF-wild-type melanomas, and the addition of cediranib to the BRAF inhibitor PLX4720 was effective in a PDX model with BRAF mutation.Conclusions: This proof-of-concept study demonstrates the feasibility of using primary biopsies directly for combinatorial drug discovery, complementing stable cell lines and xenografts, but with much greater speed and efficiency. This process could potentially be used in a clinical setting to rapidly identify therapeutic strategies for individual patients. Clin Cancer Res; 23(16); 4680-92. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Discovery/methods , High-Throughput Screening Assays/methods , Melanoma/drug therapy , Animals , Biopsy , Feasibility Studies , Female , Humans , Indoles/administration & dosage , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Quinazolines/administration & dosage , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays
6.
Nat Rev Cancer ; 15(12): 747-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26536825

ABSTRACT

Precision medicine is about matching the right drugs to the right patients. Although this approach is technology agnostic, in cancer there is a tendency to make precision medicine synonymous with genomics. However, genome-based cancer therapeutic matching is limited by incomplete biological understanding of the relationship between phenotype and cancer genotype. This limitation can be addressed by functional testing of live patient tumour cells exposed to potential therapies. Recently, several 'next-generation' functional diagnostic technologies have been reported, including novel methods for tumour manipulation, molecularly precise assays of tumour responses and device-based in situ approaches; these address the limitations of the older generation of chemosensitivity tests. The promise of these new technologies suggests a future diagnostic strategy that integrates functional testing with next-generation sequencing and immunoprofiling to precisely match combination therapies to individual cancer patients.


Subject(s)
Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Molecular Diagnostic Techniques/methods , Neoplasms/diagnosis , Neoplasms/genetics , Precision Medicine , Humans
7.
PLoS One ; 10(10): e0140310, 2015.
Article in English | MEDLINE | ID: mdl-26461489

ABSTRACT

A newer generation of anti-cancer drugs targeting underlying somatic genetic driver events have resulted in high single-agent or single-pathway response rates in selected patients, but few patients achieve complete responses and a sizeable fraction of patients relapse within a year. Thus, there is a pressing need for identification of combinations of targeted agents which induce more complete responses and prevent disease progression. We describe the results of a combination screen of an unprecedented scale in mammalian cells performed using a collection of targeted, clinically tractable agents across a large panel of melanoma cell lines. We find that even the most synergistic drug pairs are effective only in a discrete number of cell lines, underlying a strong context dependency for synergy, with strong, widespread synergies often corresponding to non-specific or off-target drug effects such as multidrug resistance protein 1 (MDR1) transporter inhibition. We identified drugs sensitizing cell lines that are BRAFV600E mutant but intrinsically resistant to BRAF inhibitor PLX4720, including the vascular endothelial growth factor receptor/kinase insert domain receptor (VEGFR/KDR) and platelet derived growth factor receptor (PDGFR) family inhibitor cediranib. The combination of cediranib and PLX4720 induced apoptosis in vitro and tumor regression in animal models. This synergistic interaction is likely due to engagement of multiple receptor tyrosine kinases (RTKs), demonstrating the potential of drug- rather than gene-specific combination discovery approaches. Patients with elevated biopsy KDR expression showed decreased progression free survival in trials of mitogen-activated protein kinase (MAPK) kinase pathway inhibitors. Thus, high-throughput unbiased screening of targeted drug combinations, with appropriate library selection and mechanistic follow-up, can yield clinically-actionable drug combinations.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Molecular Targeted Therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , High-Throughput Screening Assays , Humans , Indoles/pharmacology , Indoles/therapeutic use , Melanoma/pathology , Mice , Proto-Oncogene Proteins B-raf/metabolism , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays
9.
Chem Commun (Camb) ; 48(1): 55-7, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22083295

ABSTRACT

A convenient approach toward polycyclic frameworks containing fused 1,2,3-triazoles is described. The synthesis consists of a Cu-catalyzed cycloaddition and an intramolecular Pd-catalyzed direct arylation or Heck reaction, and affords the products in good to excellent yields.


Subject(s)
Chemistry Techniques, Synthetic/methods , Halogenation , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/chemical synthesis , Palladium/chemistry , Triazoles/chemistry , Catalysis , Cyclization
10.
Sci Signal ; 4(196): rs10, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22028469

ABSTRACT

Characterizing the extent and logic of signaling networks is essential to understanding specificity in such physiological and pathophysiological contexts as cell fate decisions and mechanisms of oncogenesis and resistance to chemotherapy. Cell-based RNA interference (RNAi) screens enable the inference of large numbers of genes that regulate signaling pathways, but these screens cannot provide network structure directly. We describe an integrated network around the canonical receptor tyrosine kinase (RTK)-Ras-extracellular signal-regulated kinase (ERK) signaling pathway, generated by combining parallel genome-wide RNAi screens with protein-protein interaction (PPI) mapping by tandem affinity purification-mass spectrometry. We found that only a small fraction of the total number of PPI or RNAi screen hits was isolated under all conditions tested and that most of these represented the known canonical pathway components, suggesting that much of the core canonical ERK pathway is known. Because most of the newly identified regulators are likely cell type- and RTK-specific, our analysis provides a resource for understanding how output through this clinically relevant pathway is regulated in different contexts. We report in vivo roles for several of the previously unknown regulators, including CG10289 and PpV, the Drosophila orthologs of two components of the serine/threonine-protein phosphatase 6 complex; the Drosophila ortholog of TepIV, a glycophosphatidylinositol-linked protein mutated in human cancers; CG6453, a noncatalytic subunit of glucosidase II; and Rtf1, a histone methyltransferase.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Genomics/methods , MAP Kinase Signaling System , Proteomics/methods , Algorithms , Animals , Blotting, Western , Cell Line , Drosophila/cytology , Drosophila/genetics , Drosophila/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Regulatory Networks , Immunoprecipitation , Models, Genetic , Protein Binding , Protein Interaction Mapping/methods , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...