Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 19(6): 1339-1350, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38829020

ABSTRACT

N-Pyridinylthiophene carboxamide (compound 21) displays activity against peripheral nerve sheath cancer cells and mouse xenografts by an unknown mechanism. Through medicinal chemistry, we identified a more active derivative, compound 9, and found that only analogues with structures similar to nicotinamide retained activity. Genetic screens using compound 9 found that both NAMPT and NMNAT1, enzymes in the NAD salvage pathway, are necessary for activity. Compound 9 is metabolized by NAMPT and NMNAT1 into an adenine dinucleotide (AD) derivative in a cell-free system, cultured cells, and mice, and inhibition of this metabolism blocked compound activity. AD analogues derived from compound 9 inhibit IMPDH in vitro and cause cell death by inhibiting IMPDH in cells. These findings nominate these compounds as preclinical candidates for the development of tumor-activated IMPDH inhibitors to treat neuronal cancers.


Subject(s)
NAD , Niacinamide , Thiophenes , Animals , NAD/metabolism , Humans , Mice , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Niacinamide/pharmacology , Niacinamide/chemistry , Thiophenes/pharmacology , Thiophenes/chemistry , Thiophenes/metabolism , Cell Line, Tumor , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors
2.
J Med Chem ; 67(11): 9277-9301, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38804887

ABSTRACT

We recently discovered a novel N-aryl tetracyclic dicarboximide MM0299 (1) with robust activity against glioma stem-like cells that potently and selectively inhibits lanosterol synthase leading to the accumulation of the toxic shunt metabolite 24(S),25-epoxycholesterol. Herein, we delineate a systematic and comprehensive SAR study that explores the structural space surrounding the N-aryl tetracyclic dicarboximide scaffold. A series of 100 analogs were synthesized and evaluated for activity against the murine glioma stem-like cell line Mut6 and for metabolic stability in mouse liver S9 fractions. This study led to several analogs with single-digit nanomolar activity in Mut6 glioblastoma cells that were metabolically stable in S9 fractions. In vivo pharmacokinetic analysis of selected analogs identified compound 52a (IC50 = 63 nM; S9 T1/2 > 240 min) which was orally available (39% plasma; 58% brain) and displayed excellent brain exposure. Chronic oral dosing of 52a during a 2-week tolerability study indicated no adverse effect on body weight nor signs of hematologic, liver, or kidney toxicity.


Subject(s)
Glioma , Neoplastic Stem Cells , Animals , Mice , Structure-Activity Relationship , Glioma/drug therapy , Glioma/pathology , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Drug Discovery , Male , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology
3.
Cell Chem Biol ; 31(1): 139-149.e14, 2024 01 18.
Article in English | MEDLINE | ID: mdl-37967558

ABSTRACT

A novel class of benzoxaboroles was reported to induce cancer cell death but the mechanism was unknown. Using a forward genetics platform, we discovered mutations in cleavage and polyadenylation specific factor 3 (CPSF3) that reduce benzoxaborole binding and confer resistance. CPSF3 is the endonuclease responsible for pre-mRNA 3'-end processing, which is also important for RNA polymerase II transcription termination. Benzoxaboroles inhibit this endonuclease activity of CPSF3 in vitro and also curb transcriptional termination in cells, which results in the downregulation of numerous constitutively expressed genes. Furthermore, we used X-ray crystallography to demonstrate that benzoxaboroles bind to the active site of CPSF3 in a manner distinct from the other known inhibitors of CPSF3. The benzoxaborole compound impeded the growth of cancer cell lines derived from different lineages. Our results suggest benzoxaboroles may represent a promising lead as CPSF3 inhibitors for clinical development.


Subject(s)
Antineoplastic Agents , Boron Compounds , Cleavage And Polyadenylation Specificity Factor , Endonucleases , RNA Precursors , RNA Processing, Post-Transcriptional , Cleavage And Polyadenylation Specificity Factor/antagonists & inhibitors , Cleavage And Polyadenylation Specificity Factor/chemistry , Endonucleases/antagonists & inhibitors , RNA Precursors/genetics , RNA Precursors/metabolism , Boron Compounds/chemistry , Boron Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , RNA Processing, Post-Transcriptional/drug effects , Humans , Cell Line, Tumor
4.
Cell Chem Biol ; 30(11): 1453-1467.e8, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37607550

ABSTRACT

Orphan cytotoxins are small molecules for which the mechanism of action (MoA) is either unknown or ambiguous. Unveiling the mechanism of these compounds may lead to useful tools for biological investigation and new therapeutic leads. In selected cases, the DNA mismatch repair-deficient colorectal cancer cell line, HCT116, has been used as a tool in forward genetic screens to identify compound-resistant mutations, which have ultimately led to target identification. To expand the utility of this approach, we engineered cancer cell lines with inducible mismatch repair deficits, thus providing temporal control over mutagenesis. By screening for compound resistance phenotypes in cells with low or high rates of mutagenesis, we increased both the specificity and sensitivity of identifying resistance mutations. Using this inducible mutagenesis system, we implicate targets for multiple orphan cytotoxins, including a natural product and compounds emerging from a high-throughput screen, thus providing a robust tool for future MoA studies.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Humans , DNA Mismatch Repair , Antineoplastic Agents/pharmacology , Mutagenesis , Cytotoxins
5.
Nat Commun ; 14(1): 3762, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353483

ABSTRACT

Colorectal cancers (CRCs) are prevalent worldwide, yet current treatments remain inadequate. Using chemical genetic screens, we identify that co-inhibition of topoisomerase I (TOP1) and NEDD8 is synergistically cytotoxic in human CRC cells. Combination of the TOP1 inhibitor irinotecan or its bioactive metabolite SN38 with the NEDD8-activating enzyme inhibitor pevonedistat exhibits synergy in CRC patient-derived organoids and xenografts. Mechanistically, we show that pevonedistat blocks the ubiquitin/proteasome-dependent repair of TOP1 DNA-protein crosslinks (TOP1-DPCs) induced by TOP1 inhibitors and that the CUL4-RBX1 complex (CRL4) is a prominent ubiquitin ligase acting on TOP1-DPCs for proteasomal degradation upon auto-NEDD8 modification during replication. We identify DCAF13, a DDB1 and Cullin Associated Factor, as the receptor of TOP1-DPCs for CRL4. Our study not only uncovers a replication-coupled ubiquitin-proteasome pathway for the repair of TOP1-DPCs but also provides molecular and translational rationale for combining TOP1 inhibitors and pevonedistat for CRC and other types of cancers.


Subject(s)
Colorectal Neoplasms , Topoisomerase I Inhibitors , Humans , Topoisomerase I Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ligases/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , RNA-Binding Proteins
6.
bioRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865268

ABSTRACT

Orphan cytotoxins are small molecules for which the mechanism of action (MoA) is either unknown or ambiguous. Unveiling the mechanism of these compounds may lead to useful tools for biological investigation and in some cases, new therapeutic leads. In select cases, the DNA mismatch repair-deficient colorectal cancer cell line, HCT116, has been used as a tool in forward genetic screens to identify compound-resistant mutations, which have ultimately led to target identification. To expand the utility of this approach, we engineered cancer cell lines with inducible mismatch repair deficits, thus providing temporal control over mutagenesis. By screening for compound resistance phenotypes in cells with low or high rates of mutagenesis, we increased both the specificity and sensitivity of identifying resistance mutations. Using this inducible mutagenesis system, we implicate targets for multiple orphan cytotoxins, including a natural product and compounds emerging from a high-throughput screen, thus providing a robust tool for future MoA studies.

7.
Cell Chem Biol ; 30(2): 214-229.e18, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36758549

ABSTRACT

Glioblastoma (GBM) is an aggressive adult brain cancer with few treatment options due in part to the challenges of identifying brain-penetrant drugs. Here, we investigated the mechanism of MM0299, a tetracyclic dicarboximide with anti-glioblastoma activity. MM0299 inhibits lanosterol synthase (LSS) and diverts sterol flux away from cholesterol into a "shunt" pathway that culminates in 24(S),25-epoxycholesterol (EPC). EPC synthesis following MM0299 treatment is both necessary and sufficient to block the growth of mouse and human glioma stem-like cells by depleting cellular cholesterol. MM0299 exhibits superior selectivity for LSS over other sterol biosynthetic enzymes. Critical for its application in the brain, we report an MM0299 derivative that is orally bioavailable, brain-penetrant, and induces the production of EPC in orthotopic GBM tumors but not normal mouse brain. These studies have implications for the development of an LSS inhibitor to treat GBM or other neurologic indications.


Subject(s)
Glioblastoma , Glioma , Adult , Humans , Lanosterol/pharmacology , Lanosterol/metabolism , Brain/metabolism , Glioma/drug therapy , Glioma/metabolism , Cholesterol , Glioblastoma/drug therapy
8.
Mol Cell ; 82(16): 3030-3044.e8, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35764091

ABSTRACT

Characterized by intracellular lipid droplet accumulation, clear cell renal cell carcinoma (ccRCC) is resistant to cytotoxic chemotherapy and is a lethal disease. Through an unbiased siRNA screen of 2-oxoglutarate (2-OG)-dependent enzymes, which play a critical role in tumorigenesis, we identified Jumonji domain-containing 6 (JMJD6) as an essential gene for ccRCC tumor development. The downregulation of JMJD6 abolished ccRCC colony formation in vitro and inhibited orthotopic tumor growth in vivo. Integrated ChIP-seq and RNA-seq analyses uncovered diacylglycerol O-acyltransferase 1 (DGAT1) as a critical JMJD6 effector. Mechanistically, JMJD6 interacted with RBM39 and co-occupied DGAT1 gene promoter with H3K4me3 to induce DGAT1 expression. JMJD6 silencing reduced DGAT1, leading to decreased lipid droplet formation and tumorigenesis. The pharmacological inhibition (or depletion) of DGAT1 inhibited lipid droplet formation in vitro and ccRCC tumorigenesis in vivo. Thus, the JMJD6-DGAT1 axis represents a potential new therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Diacylglycerol O-Acyltransferase , Jumonji Domain-Containing Histone Demethylases , Kidney Neoplasms , Carcinogenesis/genetics , Carcinoma, Renal Cell/genetics , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Epigenesis, Genetic , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kidney Neoplasms/genetics , Lipid Droplets/metabolism
9.
Sci Adv ; 7(47): eabj5405, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34788094

ABSTRACT

Aberrant alternative pre-mRNA splicing plays a critical role in MYC-driven cancers and therefore may represent a therapeutic vulnerability. Here, we show that neuroblastoma, a MYC-driven cancer characterized by splicing dysregulation and spliceosomal dependency, requires the splicing factor RBM39 for survival. Indisulam, a "molecular glue" that selectively recruits RBM39 to the CRL4-DCAF15 E3 ubiquitin ligase for proteasomal degradation, is highly efficacious against neuroblastoma, leading to significant responses in multiple high-risk disease models, without overt toxicity. Genetic depletion or indisulam-mediated degradation of RBM39 induces significant genome-wide splicing anomalies and cell death. Mechanistically, the dependency on RBM39 and high-level expression of DCAF15 determine the exquisite sensitivity of neuroblastoma to indisulam. Our data indicate that targeting the dysregulated spliceosome by precisely inhibiting RBM39, a vulnerability in neuroblastoma, is a valid therapeutic strategy.

10.
Nat Commun ; 12(1): 2954, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34012010

ABSTRACT

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Subject(s)
Breast Neoplasms/metabolism , DNA Replication/physiology , Flap Endonucleases/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , DNA/chemistry , DNA/metabolism , DNA Damage , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA Replication/genetics , Female , Flap Endonucleases/deficiency , Flap Endonucleases/genetics , Gene Knockout Techniques , Genomic Instability , HCT116 Cells , Humans , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleic Acid Conformation , Poly (ADP-Ribose) Polymerase-1/metabolism , S Phase , Substrate Specificity
11.
J Med Chem ; 63(17): 9773-9786, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32787093

ABSTRACT

A series of N-acyl benzothiazoles shows selective and potent cytotoxicity against cancer cell lines expressing cytochrome P450 4F11. A prodrug form is metabolized by cancer cells into an active inhibitor of stearoyl-CoA desaturase (SCD). Substantial variation on the acyl portion of the inhibitors allowed the identification of (R)-27, which balanced potency, solubility, and lipophilicity to allow proof-of-concept studies in mice. The prodrugs were activated inside the tumor, where they can arrest tumor growth. Together, these observations offer promise that a tumor-activated prodrug strategy might exploit the essentiality of SCD for tumor growth, while avoiding toxicity associated with systemic SCD inhibition.


Subject(s)
Benzothiazoles/pharmacology , Enzyme Inhibitors/pharmacology , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Benzothiazoles/pharmacokinetics , Cell Line, Tumor , Cytochrome P450 Family 4/metabolism , Female , Humans , Mice , Prodrugs/metabolism , Tissue Distribution
12.
J Am Chem Soc ; 142(13): 6128-6138, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32163279

ABSTRACT

TASIN (Truncated APC-Selective Inhibitors) compounds are selectively toxic to colorectal cancer cells with APC mutations, although their mechanism of action remains unknown. Here, we found that TASINs inhibit three enzymes in the postsqualene cholesterol biosynthetic pathway including EBP, DHCR7, and DHCR24. Even though all three of these enzymes are required for cholesterol biosynthesis, only inhibition of the most upstream enzyme, EBP, led to cancer cell death via depletion of downstream sterols, an observation that was confirmed by genetic silencing of EBP. Pharmacologic inhibition or genetic silencing of either DHCR7 or DHCR24 had no impact on cell viability. By using photoaffinity probes to generate a relationship between chemical structure and probe competition, we identified compounds that selectively inhibit either EBP or DHCR7. These studies identify EBP, but not downstream enzymes in the cholesterol biosynthetic pathway, as a target in APC mutant colorectal cancer and also have implications for the clinical development of highly selective EBP inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Steroid Isomerases/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/genetics , Antineoplastic Agents/chemistry , Biosynthetic Pathways/drug effects , Cholesterol/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Discovery , Enzyme Inhibitors/chemistry , HCT116 Cells , Humans , Mutation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Steroid Isomerases/metabolism
14.
ACS Chem Biol ; 15(1): 158-170, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31874028

ABSTRACT

Selective toxicity among cancer cells of the same lineage is a hallmark of targeted therapies. As such, identifying compounds that impair proliferation of a subset of non-small-cell lung cancer (NSCLC) cell lines represents one strategy to discover new drugs for lung cancer. Previously, phenotypic screens of 202 103 compounds led to the identification of 208 selective NSCLC toxins ( McMillan , E. A. , Cell , 2018 , 173 , 864 ). The mechanism of action for the majority of these compounds remains unknown. Here, we discovered the target for a series of quinazoline diones (QDC) that demonstrate selective toxicity among 96 NSCLC lines. Using photoreactive probes, we found that the QDC binds to both mitochondrial complex I of the electron transport chain and hydroxyacyl CoA dehydrogenase subunit alpha (HADHA), which catalyzes long-chain fatty acid oxidation. Inhibition of complex I is the on-target activity for QDC, while binding to HADHA is off-target. The sensitivity profile of the QDC across NSCLC lines correlated with the sensitivity profiles of six additional structurally distinct compounds. The antiproliferative activity of these compounds is also the consequence of binding to mitochondrial complex I, reflecting significant structural diversity among complex I inhibitors. Small molecules targeting complex I are currently in clinical development for the treatment of cancer. Our results highlight complex I as a target in NSCLC and report structurally diverse scaffolds that inhibit complex I.


Subject(s)
Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/diet therapy , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Lung Neoplasms/diet therapy , Quinazolinones/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Gene Knockout Techniques , Humans , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Molecular Structure , Molecular Targeted Therapy , Oxidation-Reduction , Oxygen Consumption , Protein Binding , Protein Conformation , Proteomics , Quinazolinones/pharmacology , Structure-Activity Relationship , Substrate Specificity
15.
Cell Rep ; 29(6): 1499-1510.e6, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693891

ABSTRACT

Indisulam and related sulfonamides recruit the splicing factor RBM39 to the CRL4-DCAF15 E3 ubiquitin ligase, resulting in RBM39 ubiquitination and degradation. Here, we used a combination of domain mapping and random mutagenesis to identify domains or residues that are necessary for indisulam-dependent RBM39 ubiquitination. DCAF15 mutations at Q232 or D475 prevent RBM39 recruitment by indisulam. RBM39 is recruited to DCAF15 by its RRM2 (RNA recognition motif 2) and is ubiquitinated on its N terminus. RBM23, which is an RBM39 paralog, is also recruited to the CRL4-DCAF15 ligase through its RRM2 domain and undergoes sulfonamide-dependent degradation. Indisulam alters the expression of more than 3,000 genes and causes widespread intron retention and exon skipping. All of these changes can be attributed to RBM39, and none are the consequence of RBM23 degradation. Our findings demonstrate that indisulam selectively degrades RBM23 and RBM39, the latter of which is critically important for splicing and gene expression.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , RNA Recognition Motif Proteins/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins/metabolism , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lysine , Mutagenesis , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , RNA Recognition Motif Proteins/genetics , RNA Splicing/genetics , RNA-Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics
16.
Structure ; 27(11): 1625-1633.e3, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693911

ABSTRACT

E7820 and indisulam are two examples of aryl sulfonamides that recruit RBM39 to Rbx-Cul4-DDA1-DDB1-DCAF15 E3 ligase complex, leading to its ubiquitination and degradation by the proteasome. To understand their mechanism of action, we performed kinetic analysis on the recruitment of RBM39 to DCAF15 and solved a crystal structure of DDA1-DDB1-DCAF15 in complex with E7820 and the RRM2 domain of RBM39. E7820 packs in a shallow pocket on the surface of DCAF15 and the resulting modified interface binds RBM39 through the α1 helix of the RRM2 domain. Our kinetic studies revealed that aryl sulfonamide and RBM39 bind to DCAF15 in a synergistic manner. The structural and kinetic studies confirm aryl sulfonamides as molecular glues in the recruitment of RBM39 and provide a framework for future efforts to utilize DCAF15 to degrade other proteins of interest.


Subject(s)
Indoles/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , RNA-Binding Proteins/chemistry , Sulfonamides/chemistry , Binding Sites , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Molecular Docking Simulation , Protein Binding , RNA-Binding Proteins/metabolism
17.
Cell Chem Biol ; 26(9): 1315-1321.e3, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31303577

ABSTRACT

Target identification for biologically active small molecules remains a major barrier for drug discovery. Cancer cells exhibiting defective DNA mismatch repair (dMMR) have been used as a forward genetics system to uncover compound targets. However, this approach has been limited by the dearth of cancer cell lines that harbor naturally arising dMMR. Here, we establish a platform for forward genetic screening using CRISPR/Cas9 to engineer dMMR into mammalian cells. We demonstrate the utility of this approach to identify mechanisms of drug action in mouse and human cancer cell lines using in vitro selections against three cellular toxins. In each screen, compound-resistant alleles emerged in drug-resistant clones, supporting the notion that engineered dMMR enables forward genetic screening in mammalian cells.


Subject(s)
Drug Discovery/methods , Genetic Engineering/methods , Genetic Testing/methods , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Mismatch Repair/genetics , Humans , Mice , Neoplasms/genetics
18.
Methods Mol Biol ; 1888: 175-187, 2019.
Article in English | MEDLINE | ID: mdl-30519947

ABSTRACT

Many small molecule compounds with anticancer activities are discovered through phenotype-based screens. However, discovering the targets of these small molecules has been challenging. The gold standard for target identification requires the discovery of mutations in the target protein that block the effects of small molecules in vitro as well as in vivo. Here we describe the procedures for isolating drug resistant clones using the colorectal cancer cell line HCT-116 followed by whole-exome sequencing to identify recurrent mutations associated with compound resistance. Together with downstream in vitro and in vivo validation experiments, this strategy enables rapid target discovery for cytotoxic compounds.


Subject(s)
Drug Discovery/methods , Drug Resistance/genetics , Exome Sequencing , Exome , Cell Line , Cell Survival/drug effects , Computational Biology/methods , HCT116 Cells , Humans , Inhibitory Concentration 50 , Small Molecule Libraries , Software
19.
J Med Chem ; 61(12): 5199-5221, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29869888

ABSTRACT

Stearoyl-CoA desaturase (SCD) catalyzes the first step in the conversion of saturated fatty acids to unsaturated fatty acids. Unsaturated fatty acids are required for membrane integrity and for cell proliferation. For these reasons, inhibitors of SCD represent potential treatments for cancer. However, systemically active SCD inhibitors result in skin toxicity, which presents an obstacle to their development. We recently described a series of oxalic acid diamides that are converted into active SCD inhibitors within a subset of cancers by CYP4F11-mediated metabolism. Herein, we describe the optimization of the oxalic acid diamides and related N-acyl ureas and an analysis of the structure-activity relationships related to metabolic activation and SCD inhibition.


Subject(s)
Cytochrome P450 Family 4/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice, Inbred Strains , Oxalic Acid/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Stearoyl-CoA Desaturase/metabolism , Structure-Activity Relationship
20.
Science ; 356(6336)2017 04 28.
Article in English | MEDLINE | ID: mdl-28302793

ABSTRACT

Indisulam is an aryl sulfonamide drug with selective anticancer activity. Its mechanism of action and the basis for its selectivity have so far been unknown. Here we show that indisulam promotes the recruitment of RBM39 (RNA binding motif protein 39) to the CUL4-DCAF15 E3 ubiquitin ligase, leading to RBM39 polyubiquitination and proteasomal degradation. Mutations in RBM39 that prevent its recruitment to CUL4-DCAF15 increase RBM39 stability and confer resistance to indisulam's cytotoxicity. RBM39 associates with precursor messenger RNA (pre-mRNA) splicing factors, and inactivation of RBM39 by indisulam causes aberrant pre-mRNA splicing. Many cancer cell lines derived from hematopoietic and lymphoid lineages are sensitive to indisulam, and their sensitivity correlates with DCAF15 expression levels. Two other clinically tested sulfonamides, tasisulam and chloroquinoxaline sulfonamide, share the same mechanism of action as indisulam. We propose that DCAF15 expression may be a useful biomarker to guide clinical trials of this class of drugs, which we refer to as SPLAMs (splicing inhibitor sulfonamides).


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Pharmacological/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Nuclear Proteins/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins/metabolism , Sulfonamides/pharmacology , Ubiquitin-Protein Ligase Complexes/metabolism , Amino Acid Substitution , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA-Binding Proteins/genetics , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Ubiquitination , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL