Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Nat Chem Biol ; 18(6): 615-624, 2022 06.
Article in English | MEDLINE | ID: mdl-35332332

ABSTRACT

The ability to understand and predict variable responses to therapeutic agents may improve outcomes in patients with cancer. We hypothesized that the basal gene-transcription state of cancer cell lines, coupled with cell viability profiles of small molecules, might be leveraged to nominate specific mechanisms of intrinsic resistance and to predict drug combinations that overcome resistance. We analyzed 564,424 sensitivity profiles to identify candidate gene-compound pairs, and validated nine such relationships. We determined the mechanism of a novel relationship, in which expression of the serine hydrolase enzymes monoacylglycerol lipase (MGLL) or carboxylesterase 1 (CES1) confers resistance to the histone lysine demethylase inhibitor GSK-J4 by direct enzymatic modification. Insensitive cell lines could be sensitized to GSK-J4 by inhibition or gene knockout. These analytical and mechanistic studies highlight the potential of integrating gene-expression features with small-molecule response to identify patient populations that are likely to benefit from treatment, to nominate rational candidates for combinations and to provide insights into mechanisms of action.


Subject(s)
Histone Demethylases , Monoacylglycerol Lipases , Biomarkers , Cell Survival , Drug Combinations , Histone Demethylases/metabolism , Humans
2.
BMC Biol ; 18(1): 177, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33234154

ABSTRACT

BACKGROUND: Many biological processes, such as cancer metastasis, organismal development, and acquisition of resistance to cytotoxic therapy, rely on the emergence of rare sub-clones from a larger population. Understanding how the genetic and epigenetic features of diverse clones affect clonal fitness provides insight into molecular mechanisms underlying selective processes. While large-scale barcoding with NGS readout has facilitated cellular fitness assessment at the population level, this approach does not support characterization of clones prior to selection. Single-cell genomics methods provide high biological resolution, but are challenging to scale across large populations to probe rare clones and are destructive, limiting further functional analysis of important clones. RESULTS: Here, we develop CloneSifter, a methodology for tracking and enriching rare clones throughout their response to selection. CloneSifter utilizes a CRISPR sgRNA-barcode library that facilitates the isolation of viable cells from specific clones within the barcoded population using a sequence-specific retrieval reporter. We demonstrate that CloneSifter can measure clonal fitness of cancer cell models in vitro and retrieve targeted clones at abundance as low as 1 in 1883 in a heterogeneous cell population. CONCLUSIONS: CloneSifter provides a means to track and access specific and rare clones of interest across dynamic changes in population structure to comprehensively explore the basis of these changes.


Subject(s)
Cloning, Organism/methods , Clustered Regularly Interspaced Short Palindromic Repeats , RNA/metabolism , Cells, Cultured , Clone Cells
3.
Nature ; 504(7478): 138-42, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24185007

ABSTRACT

Malignant melanomas harbouring point mutations (Val600Glu) in the serine/threonine-protein kinase BRAF (BRAF(V600E)) depend on RAF-MEK-ERK signalling for tumour cell growth. RAF and MEK inhibitors show remarkable clinical efficacy in BRAF(V600E) melanoma; however, resistance to these agents remains a formidable challenge. Global characterization of resistance mechanisms may inform the development of more effective therapeutic combinations. Here we carried out systematic gain-of-function resistance studies by expressing more than 15,500 genes individually in a BRAF(V600E) melanoma cell line treated with RAF, MEK, ERK or combined RAF-MEK inhibitors. These studies revealed a cyclic-AMP-dependent melanocytic signalling network not previously associated with drug resistance, including G-protein-coupled receptors, adenyl cyclase, protein kinase A and cAMP response element binding protein (CREB). Preliminary analysis of biopsies from BRAF(V600E) melanoma patients revealed that phosphorylated (active) CREB was suppressed by RAF-MEK inhibition but restored in relapsing tumours. Expression of transcription factors activated downstream of MAP kinase and cAMP pathways also conferred resistance, including c-FOS, NR4A1, NR4A2 and MITF. Combined treatment with MAPK-pathway and histone-deacetylase inhibitors suppressed MITF expression and cAMP-mediated resistance. Collectively, these data suggest that oncogenic dysregulation of a melanocyte lineage dependency can cause resistance to RAF-MEK-ERK inhibition, which may be overcome by combining signalling- and chromatin-directed therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Melanocytes/drug effects , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Lineage , Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Melanocytes/cytology , Melanocytes/enzymology , Melanoma/enzymology , Melanoma/physiopathology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Trends Genet ; 31(1): 16-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25498789

ABSTRACT

Over the past decade, tremendous progress in high-throughput small molecule-screening methods has facilitated the rapid expansion of phenotype-based data. Parallel advances in genomic characterization methods have complemented these efforts by providing a growing list of annotated cell line features. Together, these developments have paved the way for feature-based identification of novel, exploitable cellular dependencies, subsequently expanding our therapeutic toolkit in cancer and other diseases. Here, we provide an overview of the evolution of phenotypic small-molecule profiling and discuss the most significant and recent profiling and analytical efforts, their impact on the field, and their clinical ramifications. We additionally provide a perspective for future developments in phenotypic profiling efforts guided by genomic science.


Subject(s)
Drug Discovery , Genetics, Medical , Small Molecule Libraries/pharmacology , Drug Resistance/genetics , Genome, Human/genetics , Humans , Phenotype
5.
Nature ; 468(7326): 968-72, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21107320

ABSTRACT

Oncogenic mutations in the serine/threonine kinase B-RAF (also known as BRAF) are found in 50-70% of malignant melanomas. Pre-clinical studies have demonstrated that the B-RAF(V600E) mutation predicts a dependency on the mitogen-activated protein kinase (MAPK) signalling cascade in melanoma-an observation that has been validated by the success of RAF and MEK inhibitors in clinical trials. However, clinical responses to targeted anticancer therapeutics are frequently confounded by de novo or acquired resistance. Identification of resistance mechanisms in a manner that elucidates alternative 'druggable' targets may inform effective long-term treatment strategies. Here we expressed ∼600 kinase and kinase-related open reading frames (ORFs) in parallel to interrogate resistance to a selective RAF kinase inhibitor. We identified MAP3K8 (the gene encoding COT/Tpl2) as a MAPK pathway agonist that drives resistance to RAF inhibition in B-RAF(V600E) cell lines. COT activates ERK primarily through MEK-dependent mechanisms that do not require RAF signalling. Moreover, COT expression is associated with de novo resistance in B-RAF(V600E) cultured cell lines and acquired resistance in melanoma cells and tissue obtained from relapsing patients following treatment with MEK or RAF inhibitors. We further identify combinatorial MAPK pathway inhibition or targeting of COT kinase activity as possible therapeutic strategies for reducing MAPK pathway activation in this setting. Together, these results provide new insights into resistance mechanisms involving the MAPK pathway and articulate an integrative approach through which high-throughput functional screens may inform the development of novel therapeutic strategies.


Subject(s)
Drug Resistance, Neoplasm , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Allosteric Regulation , Cell Line, Tumor , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Library , Humans , Indoles/pharmacology , Indoles/therapeutic use , MAP Kinase Kinase Kinases/genetics , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Melanoma/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Open Reading Frames/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vemurafenib
6.
Nat Methods ; 8(8): 659-61, 2011 Jun 26.
Article in English | MEDLINE | ID: mdl-21706014

ABSTRACT

Functional characterization of the human genome requires tools for systematically modulating gene expression in both loss-of-function and gain-of-function experiments. We describe the production of a sequence-confirmed, clonal collection of over 16,100 human open-reading frames (ORFs) encoded in a versatile Gateway vector system. Using this ORFeome resource, we created a genome-scale expression collection in a lentiviral vector, thereby enabling both targeted experiments and high-throughput screens in diverse cell types.


Subject(s)
Cloning, Molecular/methods , Genetic Vectors/genetics , Genomic Library , Lentivirus/genetics , Humans , Open Reading Frames
7.
Cancer Cell ; 10(6): 459-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157787

ABSTRACT

Oncogene-induced senescence functions to limit tumor development. However, a complete understanding of the signals that trigger this type of senescence is currently lacking. We found that mutations affecting NF1, Raf, and Ras induce a global negative feedback response that potently suppresses Ras and/or its effectors. Moreover, these signals promote senescence by inhibiting the Ras/PI3K pathway, which can impact the senescence machinery through HDM2 and FOXO. This negative feedback program is regulated in part by RasGEFs, Sprouty proteins, RasGAPs, and MKPs. Moreover, these signals function in vivo in benign human tumors. Thus, the ultimate response to the aberrant activation of the Ras pathway is a multifaceted negative feedback signaling network that terminates the oncogenic signal and participates in the senescence response.


Subject(s)
Cellular Senescence , Genes, ras/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Feedback , Genes, Neurofibromatosis 1/physiology , Genes, Retinoblastoma/physiology , Genes, p53/physiology , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Stem Cells/pathology , raf Kinases/physiology
8.
Nat Commun ; 15(1): 2742, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548752

ABSTRACT

The epidermal growth factor receptor, EGFR, is frequently activated in lung cancer and glioblastoma by genomic alterations including missense mutations. The different mutation spectra in these diseases are reflected in divergent responses to EGFR inhibition: significant patient benefit in lung cancer, but limited in glioblastoma. Here, we report a comprehensive mutational analysis of EGFR function. We perform saturation mutagenesis of EGFR and assess function of ~22,500 variants in a human EGFR-dependent lung cancer cell line. This approach reveals enrichment of erlotinib-insensitive variants of known and unknown significance in the dimerization, transmembrane, and kinase domains. Multiple EGFR extracellular domain variants, not associated with approved targeted therapies, are sensitive to afatinib and dacomitinib in vitro. Two glioblastoma patients with somatic EGFR G598V dimerization domain mutations show responses to dacomitinib treatment followed by within-pathway resistance mutation in one case. In summary, this comprehensive screen expands the landscape of functional EGFR variants and suggests broader clinical investigation of EGFR inhibition for cancers harboring extracellular domain mutations.


Subject(s)
Glioblastoma , Lung Neoplasms , Humans , Glioblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation
9.
Curr Biol ; 18(1): 56-62, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18164202

ABSTRACT

Inactivating mutations in NF1 underlie the prevalent familial cancer syndrome neurofibromatosis type 1 [1]. The NF1-encoded protein is a Ras GTPase-activating protein (RasGAP) [2]. Accordingly, Ras is aberrantly activated in NF1-deficient tumors; however, it is unknown which effector pathways critically function in tumor development. Here we provide in vivo evidence that TORC1/mTOR activity is essential for tumorigenesis. Specifically, we show that the mTOR inhibitor rapamycin potently suppresses the growth of aggressive NF1-associated malignancies in a genetically engineered murine model. However, in these tumors rapamycin does not function via mechanisms generally assumed to mediate tumor suppression, including inhibition of HIF-1alpha and indirect suppression of AKT, but does suppress the mTOR target Cyclin D1 [3]. These results demonstrate that mTOR inhibitors may be an effective targeted therapy for this commonly untreatable malignancy. Moreover, they indicate that mTOR inhibitors do not suppress all tumor types via the same mechanism, suggesting that current biomarkers that rely on HIF-1alpha suppression may not be informative for all cancers. Finally, our results reveal important differences between the effects of mTOR inhibition on the microvasculature in genetically engineered versus xenograft models and indicate that the former may be required for effective preclinical screening with this class of inhibitors.


Subject(s)
Genes, Neurofibromatosis 1 , Neoplasms/genetics , Transcription Factors/physiology , Animals , Cell Line , Cyclin D , Cyclins/genetics , Cyclins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
10.
Mol Cancer Res ; 19(6): 1015-1025, 2021 06.
Article in English | MEDLINE | ID: mdl-33619228

ABSTRACT

FANCJ (BRIP1/BACH1) is a hereditary breast and ovarian cancer (HBOC) gene encoding a DNA helicase. Similar to HBOC genes, BRCA1 and BRCA2, FANCJ is critical for processing DNA inter-strand crosslinks (ICL) induced by chemotherapeutics, such as cisplatin. Consequently, cells deficient in FANCJ or its catalytic activity are sensitive to ICL-inducing agents. Unfortunately, the majority of FANCJ clinical mutations remain uncharacterized, limiting therapeutic opportunities to effectively use cisplatin to treat tumors with mutated FANCJ. Here, we sought to perform a comprehensive screen to identify FANCJ loss-of-function (LOF) mutations. We developed a FANCJ lentivirus mutation library representing approximately 450 patient-derived FANCJ nonsense and missense mutations to introduce FANCJ mutants into FANCJ knockout (K/O) HeLa cells. We performed a high-throughput screen to identify FANCJ LOF mutants that, as compared with wild-type FANCJ, fail to robustly restore resistance to ICL-inducing agents, cisplatin or mitomycin C (MMC). On the basis of the failure to confer resistance to either cisplatin or MMC, we identified 26 missense and 25 nonsense LOF mutations. Nonsense mutations elucidated a relationship between location of truncation and ICL sensitivity, as the majority of nonsense mutations before amino acid 860 confer ICL sensitivity. Further validation of a subset of LOF mutations confirmed the ability of the screen to identify FANCJ mutations unable to confer ICL resistance. Finally, mapping the location of LOF mutations to a new homology model provides additional functional information. IMPLICATIONS: We identify 51 FANCJ LOF mutations, providing important classification of FANCJ mutations that will afford additional therapeutic strategies for affected patients.


Subject(s)
BRCA1 Protein/genetics , DNA Helicases/genetics , DNA Mutational Analysis/methods , Fanconi Anemia Complementation Group Proteins/genetics , Mutation/genetics , Neoplasms/genetics , RNA Helicases/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Codon, Nonsense , Cross-Linking Reagents/pharmacology , Gene Knockout Techniques , HeLa Cells , Humans , Loss of Function Mutation , Mitomycin/pharmacology , Mutation/drug effects , Mutation, Missense , Neoplasms/pathology
11.
Nat Genet ; 53(12): 1664-1672, 2021 12.
Article in English | MEDLINE | ID: mdl-34857952

ABSTRACT

Although single-gene perturbation screens have revealed a number of new targets, vulnerabilities specific to frequently altered drivers have not been uncovered. An important question is whether the compensatory relationship between functionally redundant genes masks potential therapeutic targets in single-gene perturbation studies. To identify digenic dependencies, we developed a CRISPR paralog targeting library to investigate the viability effects of disrupting 3,284 genes, 5,065 paralog pairs and 815 paralog families. We identified that dual inactivation of DUSP4 and DUSP6 selectively impairs growth in NRAS and BRAF mutant cells through the hyperactivation of MAPK signaling. Furthermore, cells resistant to MAPK pathway therapeutics become cross-sensitized to DUSP4 and DUSP6 perturbations such that the mechanisms of resistance to the inhibitors reinforce this mechanism of vulnerability. Together, multigene perturbation technologies unveil previously unrecognized digenic vulnerabilities that may be leveraged as new therapeutic targets in cancer.


Subject(s)
Dual Specificity Phosphatase 6/genetics , Dual-Specificity Phosphatases/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasms/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Enzyme Activation , GTP Phosphohydrolases/genetics , Gene Knockout Techniques , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Membrane Proteins/genetics , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/therapy , Proto-Oncogene Proteins B-raf/genetics
12.
Cell Syst ; 10(1): 52-65.e7, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31668800

ABSTRACT

Cancer evolution poses a central obstacle to cure, as resistant clones expand under therapeutic selection pressures. Genome sequencing of relapsed disease can nominate genomic alterations conferring resistance but sample collection lags behind, limiting therapeutic innovation. Genome-wide screens offer a complementary approach to chart the compendium of escape genotypes, anticipating clinical resistance. We report genome-wide open reading frame (ORF) resistance screens for first- and third-generation epidermal growth factor receptor (EGFR) inhibitors and a MEK inhibitor. Using serial sampling, dose gradients, and mathematical modeling, we generate genotype-fitness maps across therapeutic contexts and identify alterations that escape therapy. Our data expose varying dose-fitness relationship across genotypes, ranging from complete dose invariance to paradoxical dose dependency where fitness increases in higher doses. We predict fitness with combination therapy and compare these estimates to genome-wide fitness maps of drug combinations, identifying genotypes where combination therapy results in unexpected inferior effectiveness. These data are applied to nominate combination optimization strategies to forestall resistant disease.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Acrylamides/administration & dosage , Acrylamides/pharmacology , Adenocarcinoma of Lung/enzymology , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/pharmacology , Genetic Fitness , Genotype , Humans , Lung Neoplasms/enzymology , MAP Kinase Signaling System
13.
Clin Cancer Res ; 26(18): 4995-5006, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32631955

ABSTRACT

PURPOSE: Rhabdoid tumors are devastating pediatric cancers in need of improved therapies. We sought to identify small molecules that exhibit in vitro and in vivo efficacy against preclinical models of rhabdoid tumor. EXPERIMENTAL DESIGN: We screened eight rhabdoid tumor cell lines with 481 small molecules and compared their sensitivity with that of 879 other cancer cell lines. Genome-scale CRISPR-Cas9 inactivation screens in rhabdoid tumors were analyzed to confirm target vulnerabilities. Gene expression and CRISPR-Cas9 data were queried across cell lines and primary rhabdoid tumors to discover biomarkers of small-molecule sensitivity. Molecular correlates were validated by manipulating gene expression. Subcutaneous rhabdoid tumor xenografts were treated with the most effective drug to confirm in vitro results. RESULTS: Small-molecule screening identified the protein-translation inhibitor homoharringtonine (HHT), an FDA-approved treatment for chronic myelogenous leukemia (CML), as the sole drug to which all rhabdoid tumor cell lines were selectively sensitive. Validation studies confirmed the sensitivity of rhabdoid tumor to HHT was comparable with that of CML cell lines. Low expression of the antiapoptotic gene BCL2L1, which encodes Bcl-XL, was the strongest predictor of HHT sensitivity, and HHT treatment consistently depleted Mcl-1, the synthetic-lethal antiapoptotic partner of Bcl-XL. Rhabdoid tumor cell lines and primary-tumor samples expressed low BCL2L1, and overexpression of BCL2L1 induced resistance to HHT in rhabdoid tumor cells. Furthermore, HHT treatment inhibited rhabdoid tumor cell line and patient-derived xenograft growth in vivo. CONCLUSIONS: Rhabdoid tumor cell lines and xenografts are highly sensitive to HHT, at least partially due to their low expression of BCL2L1. HHT may have therapeutic potential against rhabdoid tumors.


Subject(s)
Homoharringtonine/pharmacology , Protein Biosynthesis/drug effects , Rhabdoid Tumor/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Homoharringtonine/therapeutic use , Humans , Mice , Rhabdoid Tumor/pathology , Xenograft Model Antitumor Assays , bcl-X Protein/genetics
14.
Cancer Res ; 79(9): 2352-2366, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30819666

ABSTRACT

Combinatorial inhibition of MEK1/2 and CDK4/6 is currently undergoing clinical investigation in NRAS-mutant melanoma. To prospectively map the landscape of resistance to this investigational regimen, we utilized a series of gain- and loss-of-function forward genetic screens to identify modulators of resistance to clinical inhibitors of MEK1/2 and CDK4/6 alone and in combination. First, we identified NRAS-mutant melanoma cell lines that were dependent on NRAS for proliferation and sensitive to MEK1/2 and CDK4/6 combination treatment. We then used a genome-scale ORF overexpression screen and a CRISPR knockout screen to identify modulators of resistance to each inhibitor alone or in combination. These orthogonal screening approaches revealed concordant means of achieving resistance to this therapeutic modality, including tyrosine kinases, RAF, RAS, AKT, and PI3K signaling. Activated KRAS was sufficient to cause resistance to combined MEK/CDK inhibition and to replace genetic depletion of oncogenic NRAS. In summary, our comprehensive functional genetic screening approach revealed modulation of resistance to the inhibition of MEK1/2, CDK4/6, or their combination in NRAS-mutant melanoma. SIGNIFICANCE: These findings reveal that NRAS-mutant melanomas can acquire resistance to genetic ablation of NRAS or combination MEK1/2 and CDK4/6 inhibition by upregulating activity of the RTK-RAS-RAF and RTK-PI3K-AKT signaling cascade.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , GTP Phosphohydrolases/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Membrane Proteins/genetics , Mutation , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Checkpoints , Cell Proliferation , Humans , Melanoma/genetics , Melanoma/pathology , Phosphorylation , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
J Clin Invest ; 129(11): 5005-5019, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31437130

ABSTRACT

The interleukin-3 receptor α subunit, CD123, is expressed in many hematologic malignancies including acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Tagraxofusp (SL-401) is a CD123-targeted therapy consisting of interleukin-3 fused to a truncated diphtheria toxin payload. Factors influencing response to tagraxofusp other than CD123 expression are largely unknown. We interrogated tagraxofusp resistance in patients and experimental models and found that it was not associated with CD123 loss. Rather, resistant AML and BPDCN cells frequently acquired deficiencies in the diphthamide synthesis pathway, impairing tagraxofusp's ability to ADP-ribosylate cellular targets. Expression of DPH1, encoding a diphthamide pathway enzyme, was reduced by DNA CpG methylation in resistant cells. Treatment with the DNA methyltransferase inhibitor azacitidine restored DPH1 expression and tagraxofusp sensitivity. We also developed a drug-dependent ADP-ribosylation assay in primary cells that correlated with tagraxofusp activity and may represent an additional novel biomarker. As predicted by these results and our observation that resistance also increased mitochondrial apoptotic priming, we found that the combination of tagraxofusp and azacitidine was effective in patient-derived xenografts treated in vivo. These data have important implications for clinical use of tagraxofusp and led to a phase 1 study combining tagraxofusp and azacitidine in myeloid malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dendritic Cells/metabolism , Drug Delivery Systems , Hematologic Neoplasms , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute , Neoplasm Proteins/metabolism , Animals , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation , Dendritic Cells/pathology , Female , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Nude , Minor Histocompatibility Antigens/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
16.
Mol Cancer Res ; 17(11): 2281-2293, 2019 11.
Article in English | MEDLINE | ID: mdl-31462500

ABSTRACT

High-grade serous ovarian cancer (HGSOC) is often sensitive to initial treatment with platinum and taxane combination chemotherapy, but most patients relapse with chemotherapy-resistant disease. To systematically identify genes modulating chemotherapy response, we performed pooled functional genomic screens in HGSOC cell lines treated with cisplatin, paclitaxel, or cisplatin plus paclitaxel. Genes in the intrinsic pathway of apoptosis were among the top candidate resistance genes in both gain-of-function and loss-of-function screens. In an open reading frame overexpression screen, followed by a mini-pool secondary screen, anti-apoptotic genes including BCL2L1 (BCL-XL) and BCL2L2 (BCL-W) were associated with chemotherapy resistance. In a CRISPR-Cas9 knockout screen, loss of BCL2L1 decreased cell survival whereas loss of proapoptotic genes promoted resistance. To dissect the role of individual anti-apoptotic proteins in HGSOC chemotherapy response, we evaluated overexpression or inhibition of BCL-2, BCL-XL, BCL-W, and MCL1 in HGSOC cell lines. Overexpression of anti-apoptotic proteins decreased apoptosis and modestly increased cell viability upon cisplatin or paclitaxel treatment. Conversely, specific inhibitors of BCL-XL, MCL1, or BCL-XL/BCL-2, but not BCL-2 alone, enhanced cell death when combined with cisplatin or paclitaxel. Anti-apoptotic protein inhibitors also sensitized HGSOC cells to the poly (ADP-ribose) polymerase inhibitor olaparib. These unbiased screens highlight anti-apoptotic proteins as mediators of chemotherapy resistance in HGSOC, and support inhibition of BCL-XL and MCL1, alone or combined with chemotherapy or targeted agents, in treatment of primary and recurrent HGSOC. IMPLICATIONS: Anti-apoptotic proteins modulate drug resistance in ovarian cancer, and inhibitors of BCL-XL or MCL1 promote cell death in combination with chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Drug Resistance, Neoplasm , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Ovarian Neoplasms/genetics , bcl-X Protein/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/pharmacology , Female , Genomics , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , bcl-X Protein/genetics , bcl-X Protein/metabolism
18.
Cancer Cell ; 36(4): 369-384.e13, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31543463

ABSTRACT

Mitochondrial apoptosis can be effectively targeted in lymphoid malignancies with the FDA-approved B cell lymphoma 2 (BCL-2) inhibitor venetoclax, but resistance to this agent is emerging. We show that venetoclax resistance in chronic lymphocytic leukemia is associated with complex clonal shifts. To identify determinants of resistance, we conducted parallel genome-scale screens of the BCL-2-driven OCI-Ly1 lymphoma cell line after venetoclax exposure along with integrated expression profiling and functional characterization of drug-resistant and engineered cell lines. We identified regulators of lymphoid transcription and cellular energy metabolism as drivers of venetoclax resistance in addition to the known involvement by BCL-2 family members, which were confirmed in patient samples. Our data support the implementation of combinatorial therapy with metabolic modulators to address venetoclax resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Clonal Evolution/drug effects , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Middle Aged , Mitochondria/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/therapeutic use , Treatment Outcome , Xenograft Model Antitumor Assays
19.
Mol Cell Biol ; 25(14): 5834-45, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988001

ABSTRACT

The tuberous sclerosis tumor suppressors TSC1 and TSC2 regulate the mTOR pathway to control translation and cell growth in response to nutrient and growth factor stimuli. We have recently identified the stress response REDD1 gene as a mediator of tuberous sclerosis complex (TSC)-dependent mTOR regulation by hypoxia. Here, we demonstrate that REDD1 inhibits mTOR function to control cell growth in response to energy stress. Endogenous REDD1 is induced following energy stress, and REDD1-/- cells are highly defective in dephosphorylation of the key mTOR substrates S6K and 4E-BP1 following either ATP depletion or direct activation of the AMP-activated protein kinase (AMPK). REDD1 likely acts on the TSC1/2 complex, as regulation of mTOR substrate phosphorylation by REDD1 requires TSC2 and is blocked by overexpression of the TSC1/2 downstream target Rheb but is not blocked by inhibition of AMPK. Tetracycline-inducible expression of REDD1 triggers rapid dephosphorylation of S6K and 4E-BP1 and significantly decreases cellular size. Conversely, inhibition of endogenous REDD1 by short interfering RNA increases cell size in a rapamycin-sensitive manner, and REDD1-/- cells are defective in cell growth regulation following ATP depletion. These results define REDD1 as a critical transducer of the cellular response to energy depletion through the TSC-mTOR pathway.


Subject(s)
Energy Metabolism/physiology , Protein Kinases/metabolism , Repressor Proteins/metabolism , Transcription Factors/physiology , Tumor Suppressor Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Cell Size , Energy Metabolism/genetics , Enzyme Activation , Humans , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphorylation , Protein Kinases/physiology , RNA Interference , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Tuberous Sclerosis Complex 2 Protein
20.
Cancer Cell ; 33(5): 801-815, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29763622

ABSTRACT

Despite advances in cancer biology and therapeutics, drug resistance remains problematic. Resistance is often multifactorial, heterogeneous, and prone to undersampling. Nonetheless, many individual mechanisms of targeted therapy resistance may coalesce into a smaller number of convergences, including pathway reactivation (downstream re-engagement of original effectors), pathway bypass (recruitment of a parallel pathway converging on the same downstream output), and pathway indifference (development of a cellular state independent of the initial therapeutic target). Similar convergences may also underpin immunotherapy resistance. Such parsimonious, convergence-based frameworks may help explain resistance across tumor types and therapeutic categories and may also suggest strategies to overcome it.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Neoplasms/metabolism , Humans , Immunotherapy , Molecular Targeted Therapy , Neoplasms/drug therapy , Precision Medicine
SELECTION OF CITATIONS
SEARCH DETAIL