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










Publication year range
1.
EMBO J ; 43(5): 666-694, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38279026

ABSTRACT

The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.


Subject(s)
Anaphase-Promoting Complex-Cyclosome , Neoplasms , Humans , Anaphase-Promoting Complex-Cyclosome/genetics , Dyneins , Kinesins/genetics , Kinetochores , Mitosis , Neoplasms/genetics
2.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38151625

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Subject(s)
Kinesins , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Kinesins/genetics , Kinesins/metabolism , Mitosis/genetics , Cell Line , M Phase Cell Cycle Checkpoints
4.
Blood Cancer J ; 13(1): 57, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37088806

ABSTRACT

TP53-mutant acute myeloid leukemia (AML) respond poorly to currently available treatments, including venetoclax-based drug combinations and pose a major therapeutic challenge. Analyses of RNA sequencing and reverse phase protein array datasets revealed significantly lower BAX RNA and protein levels in TP53-mutant compared to TP53-wild-type (WT) AML, a finding confirmed in isogenic CRISPR-generated TP53-knockout and -mutant AML. The response to either BCL-2 (venetoclax) or MCL-1 (AMG176) inhibition was BAX-dependent and much reduced in TP53-mutant compared to TP53-WT cells, while the combination of two BH3 mimetics effectively activated BAX, circumventing survival mechanisms in cells treated with either BH3 mimetic, and synergistically induced cell death in TP53-mutant AML and stem/progenitor cells. The BH3 mimetic-driven stress response and cell death patterns after dual inhibition were largely independent of TP53 status and affected by apoptosis induction. Co-targeting, but not individual targeting of BCL-2 and MCL-1 in mice xenografted with TP53-WT and TP53-R248W Molm13 cells suppressed both TP53-WT and TP53-mutant cell growth and significantly prolonged survival. Our results demonstrate that co-targeting BCL-2 and MCL-1 overcomes BAX deficiency-mediated resistance to individual BH3 mimetics in TP53-mutant cells, thus shifting cell fate from survival to death in TP53-deficient and -mutant AML. This concept warrants clinical evaluation.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Animals , Mice , Myeloid Cell Leukemia Sequence 1 Protein , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/pharmacology , bcl-2-Associated X Protein/therapeutic use , Apoptosis , Cell Line, Tumor , Proto-Oncogene Proteins c-bcl-2 , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Antineoplastic Agents/therapeutic use
5.
Mol Cancer Ther ; 21(6): 879-889, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35364607

ABSTRACT

MCL-1 is known to play a major role in resistance to BCL-2 inhibition, but the contribution of other BCL-2 family proteins has not been fully explored. We, here, demonstrate the ineffectiveness of MCL-1 inhibitor AMG176 in venetoclax-resistant, and conversely, of venetoclax in AMG176-resistant acute myelogenous leukemia (AML). Like cells with acquired resistance to venetoclax, cells with acquired resistance to AMG176 express increased MCL-1. Both cells with acquired resistance to venetoclax and to AMG176 express increased levels of BCL-2 and BCL-2A1, decreased BAX, and/or altered levels of other BCL-2 proteins. Cotargeting BCL-2 and MCL-1 was highly synergistic in AML cell lines with intrinsic or acquired resistance to BH3 mimetics or engineered to genetically overexpress BCL-2 or BCL-2A1 or downregulate BAX. The combination effectively eliminated primary AML blasts and stem/progenitor cells resistant to or relapsed after venetoclax-based therapy irrespective of mutations and cytogenetic abnormalities. Venetoclax and AMG176 combination markedly suppressed antiapoptotic BCL-2 proteins and AML stem/progenitor cells and dramatically extended mouse survival (median 336 vs. control 126 days; P < 0.0001) in a patient-derived xenograft (PDX) model developed from a venetoclax/hypomethylating agent therapy-resistant patient with AML. However, decreased BAX levels in the bone marrow residual leukemia cells after 4-week combination treatment may represent a resistance mechanism that contributed to their survival. Enhanced antileukemia activity was also observed in a PDX model of monocytic AML, known to be resistant to venetoclax therapy. Our results support codependence on multiple antiapoptotic BCL-2 proteins and suppression of BAX as mechanisms of AML resistance to individual BH3 mimetics. Cotargeting of MCL-1 and BCL-2 eliminates otherwise apoptosis-resistant cells.


Subject(s)
Apoptosis Regulatory Proteins , Biomimetic Materials , Leukemia, Myeloid, Acute , Animals , Apoptosis , Biomimetic Materials/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Proto-Oncogene Proteins c-bcl-2 , Stem Cells/metabolism , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/pharmacology
6.
Clin Cancer Res ; 27(5): 1526-1537, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33203642

ABSTRACT

PURPOSE: Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a high relapse rate, limited therapeutic options, and poor prognosis. We investigated the antitumor activity of AMG 757, a half-life extended bispecific T-cell engager molecule targeting delta-like ligand 3 (DLL3)-a target that is selectively expressed in SCLC tumors, but with minimal normal tissue expression. EXPERIMENTAL DESIGN: AMG 757 efficacy was evaluated in SCLC cell lines and in orthotopic and patient-derived xenograft (PDX) mouse SCLC models. Following AMG 757 administration, changes in tumor volume, pharmacodynamic changes in tumor-infiltrating T cells (TILs), and the spatial relationship between the appearance of TILs and tumor histology were examined. Tolerability was assessed in nonhuman primates (NHPs). RESULTS: AMG 757 showed potent and specific killing of even those SCLC cell lines with very low DLL3 expression (<1,000 molecules per cell). AMG 757 effectively engaged systemically administered human T cells, induced T-cell activation, and redirected T cells to lyse tumor cells to promote significant tumor regression and complete responses in PDX models of SCLC and in orthotopic models of established primary lung SCLC and metastatic liver lesions. AMG 757 was well tolerated with no AMG 757-related adverse findings up to the highest tested dose (4.5 mg/kg weekly) in NHP. AMG 757 exhibits an extended half-life in NHP, which is projected to enable intermittent administration in patients. CONCLUSIONS: AMG 757 has a compelling safety and efficacy profile in preclinical studies making it a viable option for targeting DLL3-expressing SCLC tumors in the clinical setting.


Subject(s)
Antibodies, Bispecific , Antibodies, Monoclonal , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , Lung Neoplasms , Membrane Proteins , Small Cell Lung Carcinoma , T-Lymphocytes , Animals , Female , Humans , Mice , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Mice, Inbred NOD , Mice, SCID , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Med Chem ; 62(22): 10258-10271, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31736296

ABSTRACT

Overexpression of the antiapoptotic protein Mcl-1 provides a survival advantage to some cancer cells, making inhibition of this protein an attractive therapeutic target for the treatment of certain types of tumors. Herein, we report our efforts toward the identification of a novel series of macrocyclic Mcl-1 inhibitors featuring an α-hydroxy phenylacetic acid pharmacophore or bioisostere. This work led to the discovery of 1, a potent Mcl-1 inhibitor (IC50 = 19 nM in an OPM-2 cell viability assay) with good pharmacokinetic properties and excellent in vivo efficacy in an OPM-2 multiple myeloma xenograft model.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Phenylacetates/chemistry , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Drug Stability , Female , Humans , Hydrogen Bonding , Mice, Nude , Multiple Myeloma/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
8.
Animals (Basel) ; 9(9)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31491961

ABSTRACT

Alternative lactation housing could reduce aggression when sows are mixed. We aimed to compare the effects of mixing sows in lactation (with or without piglets), at weaning or after insemination, and determine the effects of lactation housing on the piglet. This study used 120 multiparous Large White × Landrace sows and 54 focal litters. The sows were mixed into groups of six and allocated to multisuckle from day 21 lactation (MS), separated from litter and housed in groups, with piglets left in the crate for seven hours daily from day 21 lactation (SEP), mixed at weaning (day 28 lactation) (WEAN) and mixed after artificial insemination (AI) (MAI; 4 ± 1 day after last AI). Behaviour, saliva for free salivary cortisol concentration and injury counts were taken on M-1 (before mixing), M0 (mixing), M1 and M6. Piglets were weighed, injury-scored and bloods taken for cortisol. There was reduced aggression, seen as fights, bites and knocks in MS compared to the other treatments on all days (p < 0.05). MS sows had no fights on M1 and M6 and had more piglets born in the subsequent farrowing. Piglet weight, cortisol and mortality were unaffected by treatment (p > 0.05). MS piglets had greater injury scores immediately after moving to multisuckle and lower injuries around weaning (p > 0.001). Multisuckle housing could decrease aggression and stress at mixing in sows, with changes in the time of peak piglet injury (at mixing rather than at weaning) but overall no negative effects on the piglets.

9.
Animals (Basel) ; 9(1)2018 Dec 22.
Article in English | MEDLINE | ID: mdl-30583570

ABSTRACT

When sows are mixed into groups, hierarchies form and resulting aggression and stress can affect production and welfare. This study determined the effect of providing point-source materials on aggressive and play behaviors in gestating sows. Large white cross Landrace sows were mixed after insemination; six pens of 12 sows were housed in 'standard' pens, and six pens of 12 sows were housed in 'enhanced' pens. The 'enhanced' pens each contained two rubber mats, eight strands of 24 mm-thick sisal rope and two yellow plastic disks, suspended from the roof. The sows remained in these pens until pregnancy confirmation. Salivary cortisol concentration, injury counts, and sow behaviors were recorded the day before mixing (day 1), mixing (day 0) and post-mixing day 1, day 4, day 7 and day 20. At farrowing, reproductive outcomes were obtained. Play was observed (including locomotor and object play) in the 'enhanced' pen, and percentage of time spent playing was greater on d4 (1.48 ± 0.3 Square root transformed data (2.84% non-transformed adjusted mean)), d7 (1.43 ± 0.3 (2.97%)) and d20 (1.64 ± 0.3 (3.84%)), compared to d0 (0.56 ± 0.3 (0.70%)) and d1 (0.87 ± 0.3 (1.67%) (p < 0.05)). No play was observed in standard housing. Aggression, salivary free cortisol concentrations and injuries were unaffected (p > 0.05). The provision of materials had no impact on aggression, although their presence maintained sow interest and play behavior, suggesting a positive effect.

10.
Cancer Discov ; 8(12): 1598-1613, 2018 12.
Article in English | MEDLINE | ID: mdl-30254092

ABSTRACT

BH3 mimetic drugs, which inhibit prosurvival BCL2 family proteins, have limited single-agent activity in solid tumor models. The potential of BH3 mimetics for these cancers may depend on their ability to potentiate the apoptotic response to chemotherapy and targeted therapies. Using a novel class of potent and selective MCL1 inhibitors, we demonstrate that concurrent MEK + MCL1 inhibition induces apoptosis and tumor regression in KRAS-mutant non-small cell lung cancer (NSCLC) models, which respond poorly to MEK inhibition alone. Susceptibility to BH3 mimetics that target either MCL1 or BCL-xL was determined by the differential binding of proapoptotic BCL2 proteins to MCL1 or BCL-xL, respectively. The efficacy of dual MEK + MCL1 blockade was augmented by prior transient exposure to BCL-xL inhibitors, which promotes the binding of proapoptotic BCL2 proteins to MCL1. This suggests a novel strategy for integrating BH3 mimetics that target different BCL2 family proteins for KRAS-mutant NSCLC. SIGNIFICANCE: Defining the molecular basis for MCL1 versus BCL-xL dependency will be essential for effective prioritization of BH3 mimetic combination therapies in the clinic. We discover a novel strategy for integrating BCL-xL and MCL1 inhibitors to drive and subsequently exploit apoptotic dependencies of KRAS-mutant NSCLCs treated with MEK inhibitors.See related commentary by Leber et al., p. 1511.This article is highlighted in the In This Issue feature, p. 1494.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , A549 Cells , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Knockout , Mice, Nude , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
11.
Cancer Discov ; 8(12): 1582-1597, 2018 12.
Article in English | MEDLINE | ID: mdl-30254093

ABSTRACT

The prosurvival BCL2 family member MCL1 is frequently dysregulated in cancer. To overcome the significant challenges associated with inhibition of MCL1 protein-protein interactions, we rigorously applied small-molecule conformational restriction, which culminated in the discovery of AMG 176, the first selective MCL1 inhibitor to be studied in humans. We demonstrate that MCL1 inhibition induces a rapid and committed step toward apoptosis in subsets of hematologic cancer cell lines, tumor xenograft models, and primary patient samples. With the use of a human MCL1 knock-in mouse, we demonstrate that MCL1 inhibition at active doses of AMG 176 is tolerated and correlates with clear pharmacodynamic effects, demonstrated by reductions in B cells, monocytes, and neutrophils. Furthermore, the combination of AMG 176 and venetoclax is synergistic in acute myeloid leukemia (AML) tumor models and in primary patient samples at tolerated doses. These results highlight the therapeutic promise of AMG 176 and the potential for combinations with other BH3 mimetics. SIGNIFICANCE: AMG 176 is a potent, selective, and orally bioavailable MCL1 inhibitor that induces a rapid commitment to apoptosis in models of hematologic malignancies. The synergistic combination of AMG 176 and venetoclax demonstrates robust activity in models of AML at tolerated doses, highlighting the promise of BH3-mimetic combinations in hematologic cancers.See related commentary by Leber et al., p. 1511.This article is highlighted in the In This Issue feature, p. 1494.

12.
Mol Reprod Dev ; 84(9): 883-896, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28628264

ABSTRACT

Lactation anoestrus limits the flexibility of modern pig production systems such that any increase in lactation length reduces farrowing frequency, and thus profit. This review focuses on post-partum development of the sow's reproductive system, the physiology of lactation anoestrus and how it can be overcome, as well as the fertility of sows mated while lactating. The propensity for sows to ovulate spontaneously while lactating is high (24-31%), and a high proportion of sows will ovulate rapidly and synchronously in response to combinations of altered suckling (split weaning, interrupted suckling), daily boar contact, exogenous gonadotrophins, and group housing. The apparent ease with which lactation anoestrus can be overcome represents an opportunity to uncouple sow mating from weaning, thus reducing the impact of lactation length on productivity. This is especially true when considering the benefits of the described stimulation methods on the reproductive performance (i.e., shorter weaning to oestrus intervals and higher litter sizes) of the low proportion of sows that maintain lactation anoestrus.


Subject(s)
Estrus Synchronization/methods , Estrus , Lactation , Animals , Female , Male , Swine
13.
Oncotarget ; 8(11): 17795-17809, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147313

ABSTRACT

Therapeutic resistance is a major obstacle to achieving durable clinical responses with targeted therapies, highlighting a need to elucidate the underlying mechanisms responsible for resistance and identify strategies to overcome this challenge. An emerging body of data implicates the tyrosine kinase MET in mediating resistance to BRAF inhibitors in BRAFV600E mutant melanoma. In this study we observed a dominant role for the HGF/MET axis in mediating resistance to BRAF and MEK inhibitors in models of BRAFV600E and NRAS mutant melanoma. In addition, we showed that MAPK pathway inhibition induced rapid increases in MET and GAB1 levels, providing novel mechanistic insight into how BRAFV600E mutant melanoma is primed for HGF-mediated rescue. We also determined that tumor-derived HGF, not systemic HGF, may be required to convey resistance to BRAF inhibition in vivo and that resistance could be reversed following treatment with AMG 337, a selective MET inhibitor. In summary, these findings support the clinical evaluation of MET-directed targeted therapy to circumvent resistance to BRAF and MEK inhibitors in BRAFV600E mutant melanoma. In addition, the induction of MET following treatment with BRAF and MEK inhibitors has the potential to serve as a predictive biomarker for identifying patients best suited for MET inhibitor combination therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Cell Line, Tumor , Dipeptides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Indoles/pharmacology , Melanoma/genetics , Mice , Mice, Nude , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/metabolism , Pyridones/pharmacology , Sulfonamides/pharmacology , Triazoles/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays
14.
Trends Immunol ; 37(7): 462-476, 2016 07.
Article in English | MEDLINE | ID: mdl-27216414

ABSTRACT

Many advances in the treatment of cancer have been driven by the development of targeted therapies that inhibit oncogenic signaling pathways and tumor-associated angiogenesis, as well as by the recent development of therapies that activate a patient's immune system to unleash antitumor immunity. Some targeted therapies can have effects on host immune responses, in addition to their effects on tumor biology. These immune-modulating effects, such as increasing tumor antigenicity or promoting intratumoral T cell infiltration, provide a rationale for combining these targeted therapies with immunotherapies. Here, we discuss the immune-modulating effects of targeted therapies against the MAPK and VEGF signaling pathways, and how they may synergize with immunomodulatory antibodies that target PD1/PDL1 and CTLA4. We critically examine the rationale in support of these combinations in light of the current understanding of the underlying mechanisms of action of these therapies. We also discuss the available preclinical and clinical data for these combination approaches and their implications regarding mechanisms of action. Insights from these studies provide a framework for considering additional combinations of targeted therapies and immunotherapies for the treatment of cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Combined Modality Therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes/drug effects , Vascular Endothelial Growth Factor A/metabolism
15.
Mol Cancer Ther ; 15(7): 1568-79, 2016 07.
Article in English | MEDLINE | ID: mdl-27196782

ABSTRACT

The MET receptor tyrosine kinase is involved in cell growth, survival, and invasion. Clinical studies with small molecule MET inhibitors have shown the role of biomarkers in identifying patients most likely to benefit from MET-targeted therapy. AMG 337 is an oral, small molecule, ATP-competitive, highly selective inhibitor of the MET receptor. Herein, we describe AMG 337 preclinical activity and mechanism of action in MET-dependent tumor models. These studies suggest MET is the only therapeutic target for AMG 337. In an unbiased tumor cell line proliferation screen (260 cell lines), a closely related analogue of AMG 337, Compound 5, exhibited activity in 2 of 260 cell lines; both were MET-amplified. Additional studies examining the effects of AMG 337 on the proliferation of a limited panel of cell lines with varying MET copy numbers revealed that high-level focal MET amplification (>12 copies) was required to confer MET oncogene addiction and AMG 337 sensitivity. One MET-amplified cell line, H1573 (>12 copies), was AMG 337 insensitive, possibly because of a downstream G12A KRAS mutation. Mechanism-of-action studies in sensitive MET-amplified cell lines demonstrated that AMG 337 inhibited MET and adaptor protein Gab-1 phosphorylation, subsequently blocking the downstream PI3K and MAPK pathways. AMG 337 exhibited potency in pharmacodynamic assays evaluating MET signaling in tumor xenograft models; >90% inhibition of Gab-1 phosphorylation was observed at 0.75 mg/kg. These findings describe the preclinical activity and mechanism of action of AMG 337 in MET-dependent tumor models and indicate its potential as a novel therapeutic for the treatment of MET-dependent tumors. Mol Cancer Ther; 15(7); 1568-79. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Amplification , Humans , MAP Kinase Signaling System/drug effects , Mice , Necrosis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
Bioorg Med Chem Lett ; 25(19): 4136-42, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26298499

ABSTRACT

Based on lead compound 1, which was discovered from a high-throughput screen, a series of PI3Kα/mTOR inhibitors were evaluated that contained an imidazo[1,2-a]pyridine as a core replacement for the benzimidazole contained in 1. By exploring various ring systems that occupy the affinity pocket, two fragments containing a methoxypyridine were identified that gave <100 nM potency toward PI3Kα in enzyme and cellular assays with moderate stability in rat and human liver microsomes. With the two methoxypyridine groups selected to occupy the affinity pocket, analogs were prepared with various fragments intended to occupy the ribose pocket of PI3Kα and mTOR. From these analogs, tertiary alcohol 18 was chosen for in vivo pharmacodynamic evaluation based on its potency in the PI3Kα cellular assay, microsomal stability, and in vivo pharmacokinetic properties. In a mouse liver pharmacodynamic assay, compound 18 showed 56% inhibition of HFG-induced AKT (Ser473) phosphorylation at a 30 mg/kg dose.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemical synthesis , Rats , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
17.
Bioorg Med Chem Lett ; 24(24): 5630-5634, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25466188

ABSTRACT

Replacement of the piperazine sulfonamide portion of the PI3Kα inhibitor AMG 511 (1) with a range of aliphatic alcohols led to the identification of a truncated gem-dimethylbenzylic alcohol analog, 2-(5-(4-amino-6-methyl-1,3,5-triazin-2-yl)-6-((5-fluoro-6-methoxypyridin-3-yl)amino)pyridin-3-yl)propan-2-ol (7). This compound possessed good in vitro efficacy and pharmacokinetic parameters and demonstrated an EC50 of 239 ng/mL in a mouse liver pharmacodynamic model measuring the inhibition of hepatocyte growth factor (HGF)-induced Akt Ser473 phosphorylation in CD1 nude mice 6 h post-oral dosing.


Subject(s)
Alcohols/chemistry , Phosphoinositide-3 Kinase Inhibitors , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Sulfonamides/chemistry , Triazines/chemical synthesis , Animals , Female , Half-Life , Liver/metabolism , Male , Mice , Mice, Nude , Molecular Conformation , Phosphatidylinositol 3-Kinases/metabolism , Piperazine , Piperazines/metabolism , Piperazines/pharmacokinetics , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/metabolism , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Triazines/metabolism , Triazines/pharmacokinetics
18.
Oncotarget ; 5(8): 2030-43, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24810962

ABSTRACT

While MDM2 inhibitors hold great promise as cancer therapeutics, drug resistance will likely limit their efficacy as single agents. To identify drug combinations that might circumvent resistance, we screened for agents that could synergize with MDM2 inhibition in the suppression of cell viability. We observed broad and robust synergy when combining MDM2 antagonists with either MEK or PI3K inhibitors. Synergy was not limited to cell lines harboring MAPK or PI3K pathway mutations, nor did it depend on which node of the PI3K axis was targeted. MDM2 inhibitors also synergized strongly with BH3 mimetics, BCR-ABL antagonists, and HDAC inhibitors. MDM2 inhibitor-mediated synergy with agents targeting these mechanisms was much more prevalent than previously appreciated, implying that clinical translation of these combinations could have far-reaching implications for public health. These findings highlight the importance of combinatorial drug targeting and provide a framework for the rational design of MDM2 inhibitor clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Gene Expression/drug effects , Humans
19.
Anim Reprod Sci ; 142(1-2): 48-55, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24051168

ABSTRACT

This study evaluated the effect of split weaning and fence-line boar exposure during lactation on the incidence of lactation oestrus. Large White and Large White × Landrace sows (parity 2.9 ± 0.17; mean ± SEM) were housed in conventional farrowing crates from day -4 to 30 post-parturition. Four treatments (n = 18) were used: control (SPW0): continuous lactation of 10 piglets with all piglets weaned on day 30 of lactation; and three split wean (SPW) treatments with 3 (SPW3), 5 (SPW5) or 7 (SPW7) of the heaviest piglets removed from the sow on day 18 lactation. From day 18 lactation all sows received 15 min daily, fence-line boar exposure in a detection mating area. Fewer sows in the SPW0 treatment (56% (10/18)) expressed a lactation oestrus compared to the SPW3, SPW5, and SPW7 treatments (83%; 89%; 94%, respectively). SPW0 sows had a lower subsequent total born compared to SPW5 or SPW7 sows (8.9 ± 1.1 vs. 12.5 ± 1.0 and 13.1 ± 1.1, respectively). Between day 18 and 30 of lactation, sows in SPW5 and SPW7 gained weight (4.5 ± 1.4 and 1.9 ± 1.4 kg, respectively) whereas SPW0 and SPW3 sows lost weight (4.9 ± 1.4 and 2.9 ± 1.4 kg, respectively) (P<0.05). Split weaned piglets were heavier at day 17 of age by 1.0 kg however by day 40 of age no weight differences were observed between piglets weaned on day 18 compared to day 30 (P<0.05). In conclusion, split weaning coupled with fence-line boar exposure in late lactation induced lactation oestrus in a higher proportion of sows compared to those suckling a normal litter size.


Subject(s)
Estrus/physiology , Lactation/physiology , Swine/physiology , Weaning , Adipose Tissue , Animal Husbandry , Animals , Body Weight , Female , Male , Swine/growth & development , Time Factors
20.
J Med Chem ; 55(17): 7796-816, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22897589

ABSTRACT

The phosphoinositide 3-kinase family catalyzes the phosphorylation of phosphatidylinositol-4,5-diphosphate to phosphatidylinositol-3,4,5-triphosphate, a secondary messenger which plays a critical role in important cellular functions such as metabolism, cell growth, and cell survival. Our efforts to identify potent, efficacious, and orally available phosphatidylinositol 3-kinase (PI3K) inhibitors as potential cancer therapeutics have resulted in the discovery of 4-(2-((6-methoxypyridin-3-yl)amino)-5-((4-(methylsulfonyl)piperazin-1-yl)methyl)pyridin-3-yl)-6-methyl-1,3,5-triazin-2-amine (1). In this paper, we describe the optimization of compound 1, which led to the design and synthesis of pyridyltriazine 31, a potent pan inhibitor of class I PI3Ks with a superior pharmacokinetic profile. Compound 31 was shown to potently block the targeted PI3K pathway in a mouse liver pharmacodynamic model and inhibit tumor growth in a U87 malignant glioma glioblastoma xenograft model. On the basis of its excellent in vivo efficacy and pharmacokinetic profile, compound 31 was selected for further evaluation as a clinical candidate and was designated AMG 511.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Triazines/pharmacology , Crystallography, X-Ray , Models, Molecular , Protein Kinase Inhibitors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...