Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Mol Cancer Ther ; 20(2): 250-262, 2021 02.
Article in English | MEDLINE | ID: mdl-33310762

ABSTRACT

Primary treatment for estrogen receptor-positive (ER+) breast cancer is endocrine therapy. However, substantial evidence indicates a continued role for ER signaling in tumor progression. Selective estrogen receptor degraders (SERD), such as fulvestrant, induce effective ER signaling inhibition, although clinical studies with fulvestrant report insufficient blockade of ER signaling, possibly due to suboptimal pharmaceutical properties. Furthermore, activating mutations in the ER have emerged as a resistance mechanism to current endocrine therapies. New oral SERDs with improved drug properties are under clinical investigation, but the biological profile that could translate to improved therapeutic benefit remains unclear. Here, we describe the discovery of SAR439859, a novel, orally bioavailable SERD with potent antagonist and degradation activities against both wild-type and mutant Y537S ER. Driven by its fluoropropyl pyrrolidinyl side chain, SAR439859 has demonstrated broader and superior ER antagonist and degrader activities across a large panel of ER+ cells, compared with other SERDs characterized by a cinnamic acid side chain, including improved inhibition of ER signaling and tumor cell growth. Similarly, in vivo treatment with SAR439859 demonstrated significant tumor regression in ER+ breast cancer models, including MCF7-ESR1 wild-type and mutant-Y537S mouse tumors, and HCI013, a patient-derived tamoxifen-resistant xenograft tumor. These findings indicate that SAR439859 may provide therapeutic benefit to patients with ER+ breast cancer, including those who have resistance to endocrine therapy with both wild-type and mutant ER.


Subject(s)
Breast Neoplasms/drug therapy , Receptors, Estrogen/therapeutic use , Animals , Disease Models, Animal , Female , Humans , Mice
2.
Clin Cancer Res ; 26(24): 6589-6599, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33046521

ABSTRACT

PURPOSE: Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a glycoprotein that has limited expression in normal adult tissues, but is overexpressed in carcinomas of the gastrointestinal tract, the genitourinary and respiratory systems, and breast cancer. As such, CEACAM5 is an attractive target for antibody-based therapies designed to selectively deliver cytotoxic drugs to certain epithelial tumors. Here, we describe preclinical data for a novel antibody-drug conjugate (ADC), SAR408701, which consists of an anti-CEACAM5 antibody (SAR408377) coupled to a maytansinoid agent DM4 via a cleavable linker. EXPERIMENTAL DESIGN: The specificity and binding affinity of SAR408701 to human and cynomolgus monkey CEACAM5 were tested in vitro. The cytotoxic activity of SAR408701 was assessed in CEACAM5-expressing tumor cell lines and using patient-derived xenograft mouse models of CEACAM5-positive tumors. Pharmacokinetic-pharmacodynamic and pharmacokinetic-efficacy relationships were established. SAR408701 toxicity was evaluated in cynomolgus monkey. RESULTS: SAR408701 bound selectively to human and cynomolgus monkey CEACAM5 with similar apparent Kd values (0.017 nmol/L and 0.024 nmol/L, respectively). Both in vitro and in vivo evaluations showed that SAR408701 has cytotoxic activity, leading to in vivo efficacy in single and repeated dosing. Single doses of SAR408701 induced significant increases in the tumor expression of phosphorylated histone H3, confirming the tubulin-targeting mechanism of action. The overall toxicity profile of SAR408701 in cynomolgus monkey was similar to that observed after intravenous administration of DM4 alone. CONCLUSIONS: On the basis of these preclinical data, the ADC SAR408701 is a promising candidate for development as a potential treatment for patients with CEACAM5-positive tumors.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Maytansine/chemistry , Neoplasms, Glandular and Epithelial/drug therapy , Animals , Antibodies/chemistry , Antibodies/therapeutic use , Antibodies, Monoclonal/immunology , Antineoplastic Agents/chemistry , Apoptosis , Carcinoembryonic Antigen/immunology , Cell Proliferation , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , Humans , Macaca fascicularis , Mice , Mice, SCID , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem Lett ; 30(12): 127202, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32331933

ABSTRACT

The therapeutic potential of interfering with dysregulated proteins by inducing its selective degradation has been pursued using different mechanisms. In the present article, we review representative examples of monovalent protein-degraders that, contrary to the proteolysis targeting chimeras, achieve target degradation without displaying recognition motifs for the recruitment of E3 ubiquitin ligases. We also highlight new technologies and assays that may brought to bear on the discovery of common elements that could predict and enable the selective degradation of pathogenic targets by monovalent protein-degraders. The successful application of these methods would pave the way to the advancement of new drugs with unique efficacy and tolerability properties.


Subject(s)
Proteolysis/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Biosensing Techniques , Cinnamates/pharmacology , Drug Discovery , Fulvestrant/pharmacology , Humans , Indoles/pharmacology , Ligands , Protein Binding , Receptors, Estrogen/metabolism , Stilbenes/pharmacology , Tamoxifen/pharmacology
4.
J Med Chem ; 63(2): 512-528, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31721572

ABSTRACT

More than 75% of breast cancers are estrogen receptor alpha (ERα) positive (ER+), and resistance to current hormone therapies occurs in one-third of ER+ patients. Tumor resistance is still ERα-dependent, but mutations usually confer constitutive activation to the hormone receptor, rendering ERα modulator drugs such as tamoxifen and aromatase inhibitors ineffective. Fulvestrant is a potent selective estrogen receptor degrader (SERD), which degrades the ERα receptor in drug-resistant tumors and has been approved for the treatment of hormone-receptor-positive metastatic breast cancer following antiestrogen therapy. However, fulvestrant shows poor pharmacokinetic properties in human, low solubility, weak permeation, and high metabolism, limiting its administration to inconvenient intramuscular injections. This Drug Annotation describes the identification and optimization of a new series of potent orally available SERDs, which led to the discovery of 6-(2,4-dichlorophenyl)-5-[4-[(3S)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7H-benzo[7]annulene-2-carboxylic acid (43d), showing promising antitumor activity in breast cancer mice xenograft models and whose properties warranted clinical evaluation.


Subject(s)
Breast Neoplasms/drug therapy , Drug Discovery/methods , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/therapeutic use , Animals , Breast Neoplasms/metabolism , Crystallography, X-Ray , Dogs , Drug Resistance, Neoplasm , Female , Half-Life , High-Throughput Screening Assays , Humans , Ligands , Mice , Models, Molecular , Rats , Receptors, Estrogen/drug effects , Selective Estrogen Receptor Modulators/pharmacokinetics , Selective Estrogen Receptor Modulators/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
Cell Chem Biol ; 25(6): 705-717.e11, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29628435

ABSTRACT

Activating KRAS mutations are major oncogenic drivers in multiple tumor types. Synthetic lethal screens have previously been used to identify targets critical for the survival of KRAS mutant cells, but their application to drug discovery has proven challenging, possibly due in part to a failure of monolayer cultures to model tumor biology. Here, we report the results of a high-throughput synthetic lethal screen for small molecules that selectively inhibit the growth of KRAS mutant cell lines in soft agar. Chemoproteomic profiling identifies the target of the most KRAS-selective chemical series as dihydroorotate dehydrogenase (DHODH). DHODH inhibition is shown to perturb multiple metabolic pathways. In vivo preclinical studies demonstrate strong antitumor activity upon DHODH inhibition in a pancreatic tumor xenograft model.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Dihydroorotate Dehydrogenase , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, SCID , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
6.
Mol Cancer Ther ; 15(7): 1460-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27196754

ABSTRACT

Class IA PI3K pathway activation resulting from PTEN deficiency has been associated with lack of sensitivity of melanoma to BRAF kinase inhibitors. Although previous studies have shown synergistic activity when pan-PI3K inhibitors were combined with MAPK inhibitors in the treatment of melanoma exhibiting concurrent genetic abnormalities, overlapping adverse events in patients limit optimal dosing and clinical application. With the aim of specifically targeting PTEN-deficient cancers and minimizing the potential for on-target toxicity when inhibiting multiple PI3K isoforms, we developed a program to discover PI3Kß-selective kinase inhibitors and identified SAR260301 as a potent PI3Kß-selective, orally available compound, which is now in clinical development. Herein, we provide a detailed biological characterization of SAR260301, and show that this compound has outstanding biochemical and cellular selectivity for the PI3Kß isoform versus the α, δ, and γ isoforms and a large panel of protein and lipid kinases. We demonstrate that SAR260301 blocks PI3K pathway signaling preferentially in PTEN-deficient human tumor models, and has synergistic antitumor activity when combined with vemurafenib (BRAF inhibitor) or selumetinib (MEK inhibitor) in PTEN-deficient/BRAF-mutated human melanoma tumor models. Combination treatments were very well tolerated, suggesting the potential for a superior safety profile at optimal dosing using selective compounds to inhibit multiple signaling pathways. Together, these experiments provide a preclinical proof-of-concept for safely combining inhibitors of PI3Kß and BRAF or MEK kinase modulators to improve antitumor activity in PTEN-deficient/BRAF-mutant melanoma, and support the evaluation of SAR260301-based combinations in clinical studies. Mol Cancer Ther; 15(7); 1460-71. ©2016 AACR.


Subject(s)
Indoles/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/genetics , Melanoma/metabolism , Mutation , PTEN Phosphohydrolase/deficiency , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Pyrimidinones/pharmacology , Administration, Oral , Animals , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Disease Models, Animal , Drug Synergism , Female , Humans , Indoles/administration & dosage , Indoles/chemistry , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/pathology , Mice , Models, Molecular , Molecular Conformation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidinones/administration & dosage , Pyrimidinones/chemistry , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Drug Discov Today ; 20(8): 978-87, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25814036

ABSTRACT

Herein, we report a novel and general method, lead optimization attrition analysis (LOAA), to benchmark two distinct small-molecule lead series using a relatively unbiased, simple technique and commercially available software. We illustrate this approach with data collected during lead optimization of two independent oncology programs as a case study. Easily generated graphics and attrition curves enabled us to calibrate progress and support go/no go decisions on each program. We believe that this data-driven technique could be used broadly by medicinal chemists and management to guide strategic decisions during drug discovery.


Subject(s)
Algorithms , Antineoplastic Agents/pharmacology , Chemistry, Pharmaceutical/methods , Decision Support Techniques , Drug Discovery/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Biological Assay , Humans , Models, Biological , Models, Statistical , Software
8.
Mol Cancer Res ; 13(6): 1009-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25758165

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) remains a significant clinical challenge with few therapeutic options available to cancer patients. MicroRNA 21-5p (miR-21) has been shown to be upregulated in HCC, but the contribution of this oncomiR to the maintenance of tumorigenic phenotype in liver cancer remains poorly understood. We have developed potent and specific single-stranded oligonucleotide inhibitors of miR-21 (anti-miRNAs) and used them to interrogate dependency on miR-21 in a panel of liver cancer cell lines. Treatment with anti-miR-21, but not with a mismatch control anti-miRNA, resulted in significant derepression of direct targets of miR-21 and led to loss of viability in the majority of HCC cell lines tested. Robust induction of caspase activity, apoptosis, and necrosis was noted in anti-miR-21-treated HCC cells. Furthermore, ablation of miR-21 activity resulted in inhibition of HCC cell migration and suppression of clonogenic growth. To better understand the consequences of miR-21 suppression, global gene expression profiling was performed on anti-miR-21-treated liver cancer cells, which revealed striking enrichment in miR-21 target genes and deregulation of multiple growth-promoting pathways. Finally, in vivo dependency on miR-21 was observed in two separate HCC tumor xenograft models. In summary, these data establish a clear role for miR-21 in the maintenance of tumorigenic phenotype in HCC in vitro and in vivo. IMPLICATIONS: miR-21 is important for the maintenance of the tumorigenic phenotype of HCC and represents a target for pharmacologic intervention.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Gene Regulatory Networks/drug effects , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Oligoribonucleotides, Antisense/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Heterografts , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , Neoplasm Invasiveness , Oligoribonucleotides, Antisense/therapeutic use
9.
Nucleic Acids Res ; 43(2): 1204-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25550434

ABSTRACT

Mechanisms of unassisted delivery of RNA therapeutics, including inhibitors of microRNAs, remain poorly understood. We observed that the hepatocellular carcinoma cell line SKHEP1 retains productive free uptake of a miR-21 inhibitor (anti-miR-21). Uptake of anti-miR-21, but not a mismatch (MM) control, induces expression of known miR-21 targets (DDAH1, ANKRD46) and leads to dose-dependent inhibition of cell growth. To elucidate mechanisms of SKHEP1 sensitivity to anti-miR-21, we conducted an unbiased shRNA screen that revealed tumor susceptibility gene 101 (TSG101), a component of the endosomal sorting complex required for transport (ESCRT-I), as an important determinant of anti-proliferative effects of anti-miR-21. RNA interference-mediated knockdown of TSG101 and another ESCRT-I protein, VPS28, improved uptake of anti-miR-21 in parental SKHEP1 cells and restored productive uptake to SKHEP1 clones with acquired resistance to anti-miR-21. Depletion of ESCRT-I in several additional cancer cell lines with inherently poor uptake resulted in improved activity of anti-miR-21. Finally, knockdown of TSG101 increased uptake of anti-miR-21 by cancer cells in vivo following systemic delivery. Collectively, these data support an important role for the ESCRT-I complex in the regulation of productive free uptake of anti-miRs and reveal potential avenues for improving oligonucleotide free uptake by cancer cells.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , MicroRNAs/antagonists & inhibitors , Neoplasms/metabolism , Oligonucleotides/metabolism , Animals , Biological Transport , Cell Line, Tumor , DNA-Binding Proteins/physiology , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/physiology , Female , Humans , Mice, SCID , MicroRNAs/metabolism , Neoplasms/genetics , Transcription Factors/physiology
10.
J Med Chem ; 58(1): 376-400, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25402320

ABSTRACT

Vps34 (the human class III phosphoinositide 3-kinase) is a lipid kinase involved in vesicle trafficking and autophagy and therefore constitutes an interesting target for cancer treatment. Because of the lack of specific Vps34 kinase inhibitors, we aimed to identify such compounds to further validate the role of this lipid kinase in cancer maintenance and progression. Herein, we report the discovery of a series of tetrahydropyrimidopyrimidinone derivatives. Starting with hit compound 1a, medicinal chemistry optimization led to compound 31. This molecule displays potent activity, an exquisite selectivity for Vps34 with excellent properties. The X-ray crystal structure of compound 31 in human Vps34 illustrates how the unique molecular features of the morpholine synthon bestows selectivity against class I PI3Ks. This molecule exhibits suitable in vivo mouse PK parameters and induces a sustained inhibition of Vps34 upon acute administration. Compound 31 constitutes an optimized Vps34 inhibitor that could be used to investigate human cancer biology.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Pyrimidinones/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Area Under Curve , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Caco-2 Cells , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/metabolism , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , HeLa Cells , Humans , Male , Mice, SCID , Models, Chemical , Models, Molecular , Molecular Sequence Data , Molecular Structure , Neoplasms/pathology , Protein Binding , Protein Structure, Tertiary , Pyrimidinones/chemistry , Pyrimidinones/pharmacokinetics , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Thermodynamics , Xenograft Model Antitumor Assays
11.
Bioorg Med Chem Lett ; 24(23): 5357-63, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25455482

ABSTRACT

Antibody-drug conjugates (ADCs) consist of cytotoxic drugs covalently linked to monoclonal antibodies directed to antigens differentially overexpressed in tumor cells. These loaded antibodies are expected to selectively deliver lethal cargoes to tumor cells and provide sustained clinical benefit to pre-selected cancer patients while, at the same time, minimizing systemic toxicity. Although on-target adverse events are not completely avoided and the true efficacy of these innovative agents still requires further clarification, proof-of-concept has already been achieved in clinical settings with immunoconjugates containing calicheamicin, auristatin or maytansine-based cytotoxic payloads. In this present article we review the characteristics of the preceding cytotoxic platforms and their chemical conjugation approaches.


Subject(s)
Antibodies, Monoclonal/metabolism , Neoplasms/drug therapy , Biological Products , Humans , Molecular Structure
12.
Nat Chem Biol ; 10(12): 1013-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25326666

ABSTRACT

Vps34 is a phosphoinositide 3-kinase (PI3K) class III isoform that has attracted major attention over the recent years because of its role in autophagy. Herein we describe the biological characterization of SAR405, which is a low-molecular-mass kinase inhibitor of Vps34 (KD 1.5 nM). This compound has an exquisite protein and lipid kinase selectivity profile that is explained by its unique binding mode and molecular interactions within the ATP binding cleft of human Vps34. To the best of our knowledge, this is the first potent and specific Vps34 inhibitor described so far. Our results demonstrate that inhibition of Vps34 kinase activity by SAR405 affects both late endosome-lysosome compartments and prevents autophagy. Moreover, we show that the concomitant inhibition of Vps34 and mTOR, with SAR405 and the US Food and Drug Administration-approved mTOR inhibitor everolimus, results in synergistic antiproliferative activity in renal tumor cell lines, indicating a potential clinical application in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidinones/pharmacology , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemical synthesis , Autophagy/genetics , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/genetics , Drug Synergism , Endosomes/drug effects , Endosomes/metabolism , Everolimus , Gene Expression , Humans , Kidney/enzymology , Kidney/pathology , Kinetics , Lysosomes/drug effects , Lysosomes/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Pyridines/chemical synthesis , Pyrimidinones/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Signal Transduction , Sirolimus/chemical synthesis , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/genetics
13.
J Med Chem ; 57(19): 7888-9, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25220827

ABSTRACT

The field of antibody-drug conjugates (ADCs) has gained significant momentum after the recent regulatory approval of two ADCs, and significant research efforts are directed to identify more effective payloads and simplify current manufacturing challenges.


Subject(s)
Drug Discovery , Immunoconjugates/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Humans , Trastuzumab
14.
Blood ; 124(11): 1777-89, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25006129

ABSTRACT

Postchemotherapy relapse presents a major unmet medical need in acute myeloid leukemia (AML), where treatment options are limited. CD25 is a leukemic stem cell marker and a conspicuous prognostic marker for overall/relapse-free survival in AML. Rare occurrence of genetic alterations among PIM family members imposes a substantial hurdle in formulating a compelling patient stratification strategy for the clinical development of selective PIM inhibitors in cancer. Here we show that CD25, a bona fide STAT5 regulated gene, is a mechanistically relevant predictive biomarker for sensitivity to PIM kinase inhibitors. Alone or in combination with tyrosine kinase inhibitors, PIM inhibitors can suppress STAT5 activation and significantly shorten the half-life of MYC to achieve substantial growth inhibition of high CD25-expressing AML cells. Our results highlight the importance of STAT5 and MYC in rendering cancer cells sensitive to PIM inhibitors. Because the presence of a CD25-positive subpopulation in leukemic blasts correlates with poor overall or relapse-free survival, our data suggest that a combination of PIM inhibitors with chemotherapy and tyrosine kinase inhibitors could improve long-term therapeutic outcomes in CD25-positive AML.


Subject(s)
Antineoplastic Agents/pharmacology , Blast Crisis , Gene Expression Regulation, Leukemic/drug effects , Interleukin-2 Receptor alpha Subunit/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Antineoplastic Agents/chemistry , Blast Crisis/drug therapy , Blast Crisis/genetics , Blast Crisis/metabolism , Blast Crisis/pathology , Female , HL-60 Cells , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , STAT5 Transcription Factor/genetics
15.
Oncotarget ; 5(10): 3362-74, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24830942

ABSTRACT

Inhibitors of JAK2 kinase are emerging as an important treatment modality for myeloproliferative neoplasms (MPN). However, similar to other kinase inhibitors, resistance to JAK2 inhibitors may eventually emerge through a variety of mechanisms. Effective drug combination is one way to enhance therapeutic efficacy and combat resistance against JAK2 inhibitors. To identify potential combination partners for JAK2 compounds in MPN cell lines, we performed pooled shRNA screen targeting 5,000 genes in the presence or absence of JAK2 blockade. One of the top hits identified was MYC, an oncogenic transcription factor that is difficult to inhibit directly, but could be targeted by modulation of upstream regulatory elements such as kinases. We demonstrate herein that PIM kinase inhibitors efficiently suppress MYC protein levels in MPN cell lines. Importantly, overexpression of MYC restores the viability of PIM inhibitor-treated cells, revealing causal relationship between MYC down-regulation and cell growth inhibition by PIM compounds. Combination of various PIM inhibitors with a JAK2 inhibitor results in significant synergistic growth inhibition of multiple MPN cancer cell lines and induction of apoptosis. Mechanistic studies revealed strong downregulation of phosphorylated forms of S6 and 4EBP1 by JAK2/PIM inhibitor combination treatment. Finally, such combination was effective in eradicating in vitro JAK2 inhibitor-resistant MPN clones, where MYC is consistently up-regulated. These findings demonstrate that simultaneous suppression of JAK2 and PIM kinase activity by small molecule inhibitors is more effective than either agent alone in suppressing MPN cell growth. Our data suggest that JAK2 and PIM combination might warrant further investigation for the treatment of JAK2-driven hematologic malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Janus Kinase 2/antagonists & inhibitors , Myeloproliferative Disorders/enzymology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunoblotting , Protein Kinase Inhibitors/pharmacology
16.
Mol Cancer Ther ; 13(5): 1117-29, 2014 May.
Article in English | MEDLINE | ID: mdl-24608574

ABSTRACT

Somatic PIK3CA mutations are frequently found in solid tumors, raising the hypothesis that selective inhibition of PI3Kα may have robust efficacy in PIK3CA-mutant cancers while sparing patients the side-effects associated with broader inhibition of the class I phosphoinositide 3-kinase (PI3K) family. Here, we report the biologic properties of the 2-aminothiazole derivative NVP-BYL719, a selective inhibitor of PI3Kα and its most common oncogenic mutant forms. The compound selectivity combined with excellent drug-like properties translates to dose- and time-dependent inhibition of PI3Kα signaling in vivo, resulting in robust therapeutic efficacy and tolerability in PIK3CA-dependent tumors. Novel targeted therapeutics such as NVP-BYL719, designed to modulate aberrant functions elicited by cancer-specific genetic alterations upon which the disease depends, require well-defined patient stratification strategies in order to maximize their therapeutic impact and benefit for the patients. Here, we also describe the application of the Cancer Cell Line Encyclopedia as a preclinical platform to refine the patient stratification strategy for NVP-BYL719 and found that PIK3CA mutation was the foremost positive predictor of sensitivity while revealing additional positive and negative associations such as PIK3CA amplification and PTEN mutation, respectively. These patient selection determinants are being assayed in the ongoing NVP-BYL719 clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Thiazoles/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Female , Humans , Inhibitory Concentration 50 , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Rats , Thiazoles/pharmacokinetics , Xenograft Model Antitumor Assays
17.
Bioorg Med Chem Lett ; 24(6): 1506-10, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24560540
18.
J Med Chem ; 57(3): 903-20, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24387221

ABSTRACT

Compelling molecular biology publications have reported the implication of phosphoinositide kinase PI3Kß in PTEN-deficient cell line growth and proliferation. These findings supported a scientific rationale for the development of PI3Kß-specific inhibitors for the treatment of PTEN-deficient cancers. This paper describes the discovery of 2-[2-(2,3-dihydro-indol-1-yl)-2-oxo-ethyl]-6-morpholin-4-yl-3H-pyrimidin-4-one (7) and the optimization of this new series of active and selective pyrimidone indoline amide PI3Kß inhibitors. 2-[2-(2-Methyl-2,3-dihydro-indol-1-yl)-2-oxo-ethyl]-6-morpholin-4-yl-3H-pyrimidin-4-one (28), identified following a carefully designed methyl scan, displayed improved physicochemical and in vitro pharmacokinetic properties. Structural biology efforts enabled the acquisition of the first X-ray cocrystal structure of p110ß with the selective inhibitor compound 28 bound to the ATP site. The nonplanar binding mode described herein is consistent with observed structure-activity relationship for the series. Compound 28 demonstrated significant in vivo activity in a UACC-62 xenograft model in mice, warranting further preclinical investigation. Following successful development, compound 28 entered phase I/Ib clinical trial in patients with advanced cancer.


Subject(s)
Antineoplastic Agents/chemistry , Indoles/chemistry , Neoplasms/drug therapy , PTEN Phosphohydrolase/deficiency , Phosphoinositide-3 Kinase Inhibitors , Pyrimidinones/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Cell Line, Tumor , Cell Membrane Permeability , Crystallography, X-Ray , Dogs , Drug Screening Assays, Antitumor , Female , Heterografts , Humans , Indoles/pharmacokinetics , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Microsomes, Liver/metabolism , Molecular Conformation , Molecular Docking Simulation , Neoplasm Transplantation , Neoplasms/enzymology , PTEN Phosphohydrolase/genetics , Protein Binding , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology , Rats , Rats, Nude , Solubility , Stereoisomerism , Structure-Activity Relationship
19.
J Biol Chem ; 288(42): 30125-30138, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24003220

ABSTRACT

PRP4 kinase is known for its roles in regulating pre-mRNA splicing and beyond. Therefore, a wider spectrum of PRP4 kinase substrates could be expected. The role of PRP4 kinase in cancer is also yet to be fully elucidated. Attaining specific and potent PRP4 inhibitors would greatly facilitate the study of PRP4 biological function and its validation as a credible cancer target. In this report, we verified the requirement of enzymatic activity of PRP4 in regulating cancer cell growth and identified an array of potential novel substrates through orthogonal proteomics approaches. The ensuing effort in structural biology unveiled for the first time unique features of PRP4 kinase domain and its potential mode of interaction with a low molecular weight inhibitor. These results provide new and important information for further exploration of PRP4 kinase function in cancer.


Subject(s)
Neoplasm Proteins , Neoplasms , Protein Kinase Inhibitors , Ribonucleoprotein, U4-U6 Small Nuclear , Cell Line, Tumor , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Ribonucleoprotein, U4-U6 Small Nuclear/antagonists & inhibitors , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...