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










Publication year range
1.
Clin Cancer Res ; 30(12): 2659-2671, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38619278

ABSTRACT

PURPOSE: The inherent genetic heterogeneity of acute myeloid leukemia (AML) has challenged the development of precise and effective therapies. The objective of this study was to elucidate the genomic basis of drug resistance or sensitivity, identify signatures for drug response prediction, and provide resources to the research community. EXPERIMENTAL DESIGN: We performed targeted sequencing, high-throughput drug screening, and single-cell genomic profiling on leukemia cell samples derived from patients with AML. Statistical approaches and machine learning models were applied to identify signatures for drug response prediction. We also integrated large public datasets to understand the co-occurring mutation patterns and further investigated the mutation profiles in the single cells. The features revealed in the co-occurring or mutual exclusivity pattern were further subjected to machine learning models. RESULTS: We detected genetic signatures associated with sensitivity or resistance to specific agents, and identified five co-occurring mutation groups. The application of single-cell genomic sequencing unveiled the co-occurrence of variants at the individual cell level, highlighting the presence of distinct subclones within patients with AML. Using the mutation pattern for drug response prediction demonstrates high accuracy in predicting sensitivity to some drug classes, such as MEK inhibitors for RAS-mutated leukemia. CONCLUSIONS: Our study highlights the importance of considering the gene mutation patterns for the prediction of drug response in AML. It provides a framework for categorizing patients with AML by mutations that enable drug sensitivity prediction.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute , Mutation , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Drug Resistance, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Single-Cell Analysis/methods , Machine Learning , High-Throughput Nucleotide Sequencing , Male
2.
bioRxiv ; 2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38559080

ABSTRACT

Diffuse Midline Gliomas (DMGs) are universally fatal, primarily pediatric malignancies affecting the midline structures of the central nervous system. Despite decades of clinical trials, treatment remains limited to palliative radiation therapy. A major challenge is the coexistence of molecularly distinct malignant cell states with potentially orthogonal drug sensitivities. To address this challenge, we leveraged established network-based methodologies to elucidate Master Regulator (MR) proteins representing mechanistic, non-oncogene dependencies of seven coexisting subpopulations identified by single-cell analysis-whose enrichment in essential genes was validated by pooled CRISPR/Cas9 screens. Perturbational profiles of 372 clinically relevant drugs helped identify those able to invert the activity of subpopulation-specific MRs for follow-up in vivo validation. While individual drugs predicted to target individual subpopulations-including avapritinib, larotrectinib, and ruxolitinib-produced only modest tumor growth reduction in orthotopic models, systemic co-administration induced significant survival extension, making this approach a valuable contribution to the rational design of combination therapy.

3.
Article in English | MEDLINE | ID: mdl-34250400

ABSTRACT

Multiple myeloma (MM) is a genetically heterogeneous malignancy characterized by variable treatment responses. Although numerous drugs have been approved in recent years, the ability to predict treatment response and tailor individual therapy is limited by the absence of robust predictive biomarkers. The goal of this clinical trial was to use ex vivo, high-throughput screening (HTS) of 170 compounds to predict response among patients with relapsed or refractory MM and inform the next treatment decisions. Additionally, we integrated HTS with multi-omic analysis to uncover novel associations between in vitro drug sensitivity and gene expression and mutation profiles. MATERIALS AND METHODS: Twenty-five patients with relapsed or refractory MM underwent a screening bone marrow or soft tissue biopsy. Sixteen patients were found to have sufficient plasma cells for HTS. Targeted next-generation sequencing was performed on plasma cell-free DNA from all patients who underwent HTS. RNA and whole-exome sequencing of bone marrow plasma cells were performed on eight and seven patients, respectively. RESULTS: Results of HTS testing were made available to treating physicians within a median of 5 days from the biopsy. An actionable treatment result was identified in all 16 patients examined. Among the 13 patients who received assay-guided therapy, 92% achieved stable disease or better. The expression of 105 genes and mutations in 12 genes correlated with in vitro cytotoxicity. CONCLUSION: In patients with relapsed or refractory MM, we demonstrate the feasibility of ex vivo drug sensitivity testing on isolated plasma cells from patient bone marrow biopsies or extramedullary plasmacytomas to inform the next line of therapy.


Subject(s)
High-Throughput Screening Assays , Multiple Myeloma/drug therapy , Aged , Drug Evaluation, Preclinical , Female , Humans , Male , Middle Aged , Multiple Myeloma/genetics , Treatment Outcome
5.
Cell Stem Cell ; 22(6): 929-940.e4, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29779890

ABSTRACT

Organoids derived from human pluripotent stem cells are a potentially powerful tool for high-throughput screening (HTS), but the complexity of organoid cultures poses a significant challenge for miniaturization and automation. Here, we present a fully automated, HTS-compatible platform for enhanced differentiation and phenotyping of human kidney organoids. The entire 21-day protocol, from plating to differentiation to analysis, can be performed automatically by liquid-handling robots, or alternatively by manual pipetting. High-content imaging analysis reveals both dose-dependent and threshold effects during organoid differentiation. Immunofluorescence and single-cell RNA sequencing identify previously undetected parietal, interstitial, and partially differentiated compartments within organoids and define conditions that greatly expand the vascular endothelium. Chemical modulation of toxicity and disease phenotypes can be quantified for safety and efficacy prediction. Screening in gene-edited organoids in this system reveals an unexpected role for myosin in polycystic kidney disease. Organoids in HTS formats thus establish an attractive platform for multidimensional phenotypic screening.


Subject(s)
Cell Differentiation , High-Throughput Screening Assays , Kidney/cytology , Organoids/cytology , Phenotype , Pluripotent Stem Cells/cytology , Automation , Cell Culture Techniques , Humans , Sequence Analysis, RNA
6.
Stem Cell Reports ; 10(5): 1610-1624, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29681544

ABSTRACT

Connecting specific cancer genotypes with phenotypes and drug responses constitutes the central premise of precision oncology but is hindered by the genetic complexity and heterogeneity of primary cancer cells. Here, we use patient-derived induced pluripotent stem cells (iPSCs) and CRISPR/Cas9 genome editing to dissect the individual contributions of two recurrent genetic lesions, the splicing factor SRSF2 P95L mutation and the chromosome 7q deletion, to the development of myeloid malignancy. Using a comprehensive panel of isogenic iPSCs-with none, one, or both genetic lesions-we characterize their relative phenotypic contributions and identify drug sensitivities specific to each one through a candidate drug approach and an unbiased large-scale small-molecule screen. To facilitate drug testing and discovery, we also derive SRSF2-mutant and isogenic normal expandable hematopoietic progenitor cells. We thus describe here an approach to dissect the individual effects of two cooperating mutations to clinically relevant features of malignant diseases.


Subject(s)
Antineoplastic Agents/therapeutic use , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Alternative Splicing/genetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasms/pathology , Phenotype , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Small Molecule Libraries/pharmacology
7.
Nat Commun ; 9(1): 42, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29298978

ABSTRACT

Cancers that appear pathologically similar often respond differently to the same drug regimens. Methods to better match patients to drugs are in high demand. We demonstrate a promising approach to identify robust molecular markers for targeted treatment of acute myeloid leukemia (AML) by introducing: data from 30 AML patients including genome-wide gene expression profiles and in vitro sensitivity to 160 chemotherapy drugs, a computational method to identify reliable gene expression markers for drug sensitivity by incorporating multi-omic prior information relevant to each gene's potential to drive cancer. We show that our method outperforms several state-of-the-art approaches in identifying molecular markers replicated in validation data and predicting drug sensitivity accurately. Finally, we identify SMARCA4 as a marker and driver of sensitivity to topoisomerase II inhibitors, mitoxantrone, and etoposide, in AML by showing that cell lines transduced to have high SMARCA4 expression reveal dramatically increased sensitivity to these agents.


Subject(s)
DNA Helicases/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/genetics , Machine Learning , Nuclear Proteins/genetics , Precision Medicine/methods , Transcription Factors/genetics , Algorithms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Cell Line , Datasets as Topic , Etoposide/pharmacology , Etoposide/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use
8.
J Natl Compr Canc Netw ; 14(1): 8-17, 2016 01.
Article in English | MEDLINE | ID: mdl-26733551

ABSTRACT

Accelerating cancer research is expected to require new types of clinical trials. This report describes the Intensive Trial of OMics in Cancer (ITOMIC) and a participant with triple-negative breast cancer metastatic to bone, who had markedly elevated circulating tumor cells (CTCs) that were monitored 48 times over 9 months. A total of 32 researchers from 14 institutions were engaged in the patient's evaluation; 20 researchers had no prior involvement in patient care and 18 were recruited specifically for this patient. Whole-exome sequencing of 3 bone marrow samples demonstrated a novel ROS1 variant that was estimated to be present in most or all tumor cells. After an initial response to cisplatin, a hypothesis of crizotinib sensitivity was disproven. Leukapheresis followed by partial CTC enrichment allowed for the development of a differential high-throughput drug screen and demonstrated sensitivity to investigational BH3-mimetic inhibitors of BCL-2 that could not be tested in the patient because requests to the pharmaceutical sponsors were denied. The number and size of CTC clusters correlated with clinical status and eventually death. Focusing the expertise of a distributed network of investigators on an intensively monitored patient with cancer can generate high-resolution views of the natural history of cancer and suggest new opportunities for therapy. Optimization requires access to investigational drugs.


Subject(s)
Community Networks , Research Personnel , Triple Negative Breast Neoplasms/diagnosis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/secondary , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Expert Testimony , Female , Follow-Up Studies , Humans , Leukapheresis , Longitudinal Studies , Middle Aged , Neoplasm Metastasis , Neoplastic Cells, Circulating , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
9.
PLoS One ; 7(12): e50457, 2012.
Article in English | MEDLINE | ID: mdl-23227175

ABSTRACT

The Wnt/ß-catenin signaling pathway controls important cellular events during development and often contributes to disease when dysregulated. Using high throughput screening we have identified a new small molecule inhibitor of Wnt/ß-catenin signaling, WIKI4. WIKI4 inhibits expression of ß-catenin target genes and cellular responses to Wnt/ß-catenin signaling in cancer cell lines as well as in human embryonic stem cells. Furthermore, we demonstrate that WIKI4 mediates its effects on Wnt/ß-catenin signaling by inhibiting the enzymatic activity of TNKS2, a regulator of AXIN ubiquitylation and degradation. While TNKS has previously been shown to be the target of small molecule inhibitors of Wnt/ß-catenin signaling, WIKI4 is structurally distinct from previously identified TNKS inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Naphthalimides/pharmacology , Signal Transduction/drug effects , Tankyrases/antagonists & inhibitors , Triazoles/pharmacology , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Cell Line , High-Throughput Screening Assays , Humans , Ubiquitination , Wnt Proteins/metabolism , beta Catenin/metabolism
10.
Oncotarget ; 3(10): 1124-36, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23165409

ABSTRACT

Glioblastoma Multiforme (GBM) continues to have a poor patient prognosis despite optimal standard of care. Glioma stem cells (GSCs) have been implicated as the presumed cause of tumor recurrence and resistance to therapy. With this in mind, we screened a diverse chemical library of 2,000 compounds to identify therapeutic agents that inhibit GSC proliferation and therefore have the potential to extend patient survival. High-throughput screens (HTS) identified 78 compounds that repeatedly inhibited cellular proliferation, of which 47 are clinically approved for other indications and 31 are experimental drugs. Several compounds (such as digitoxin, deguelin, patulin and phenethyl caffeate) exhibited high cytotoxicity, with half maximal inhibitory concentrations (IC50) in the low nanomolar range. In particular, the FDA approved drug for the treatment of alcoholism, disulfiram (DSF), was significantly potent across multiple patient samples (IC50 of 31.1 nM). The activity of DSF was potentiated by copper (Cu), which markedly increased GSC death. DSF-Cu inhibited the chymotrypsin-like proteasomal activity in cultured GSCs, consistent with inactivation of the ubiquitin-proteasome pathway and the subsequent induction of tumor cell death. Given that DSF is a relatively non-toxic drug that can penetrate the blood-brain barrier, we suggest that DSF should be tested (as either a monotherapy or as an adjuvant) in pre-clinical models of human GBM. Data also support targeting of the ubiquitin-proteasome pathway as a therapeutic approach in the treatment of GBM.


Subject(s)
Alcohol Deterrents/pharmacology , Brain Neoplasms/drug therapy , Disulfiram/pharmacology , Glioblastoma/drug therapy , High-Throughput Screening Assays , Neoplastic Stem Cells/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL