Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 66(13): 9095-9119, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37399505

ABSTRACT

The allosteric inhibitor of the mechanistic target of rapamycin (mTOR) everolimus reduces seizures in tuberous sclerosis complex (TSC) patients through partial inhibition of mTOR functions. Due to its limited brain permeability, we sought to develop a catalytic mTOR inhibitor optimized for central nervous system (CNS) indications. We recently reported an mTOR inhibitor (1) that is able to block mTOR functions in the mouse brain and extend the survival of mice with neuronal-specific ablation of the Tsc1 gene. However, 1 showed the risk of genotoxicity in vitro. Through structure-activity relationship (SAR) optimization, we identified compounds 9 and 11 without genotoxicity risk. In neuronal cell-based models of mTOR hyperactivity, both corrected aberrant mTOR activity and significantly improved the survival rate of mice in the Tsc1 gene knockout model. Unfortunately, 9 and 11 showed limited oral exposures in higher species and dose-limiting toxicities in cynomolgus macaque, respectively. However, they remain optimal tools to explore mTOR hyperactivity in CNS disease models.


Subject(s)
MTOR Inhibitors , Sirolimus , Mice , Animals , Syndrome , Central Nervous System/metabolism , Brain/metabolism , TOR Serine-Threonine Kinases , Adenosine Triphosphate
2.
J Med Chem ; 63(3): 1068-1083, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31955578

ABSTRACT

Recent clinical evaluation of everolimus for seizure reduction in patients with tuberous sclerosis complex (TSC), a disease with overactivated mechanistic target of rapamycin (mTOR) signaling, has demonstrated the therapeutic value of mTOR inhibitors for central nervous system (CNS) indications. Given that everolimus is an incomplete inhibitor of the mTOR function, we sought to develop a new mTOR inhibitor that has improved properties and is suitable for CNS disorders. Starting from an in-house purine-based compound, optimization of the physicochemical properties of a thiazolopyrimidine series led to the discovery of the small molecule 7, a potent and selective brain-penetrant ATP-competitive mTOR inhibitor. In neuronal cell-based models of mTOR hyperactivity, 7 corrected the mTOR pathway activity and the resulting neuronal overgrowth phenotype. The new mTOR inhibitor 7 showed good brain exposure and significantly improved the survival rate of mice with neuronal-specific ablation of the Tsc1 gene. These results demonstrate the potential utility of this tool compound to test therapeutic hypotheses that depend on mTOR hyperactivity in the CNS.


Subject(s)
Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Seizures/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thiazoles/therapeutic use , Animals , Anticonvulsants/metabolism , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Binding Sites , Brain/drug effects , Drug Discovery , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Rats , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Thiazoles/metabolism , Thiazoles/pharmacokinetics , Tuberous Sclerosis Complex 1 Protein/genetics
3.
Leukemia ; 33(4): 981-994, 2019 04.
Article in English | MEDLINE | ID: mdl-30185934

ABSTRACT

In chronic myeloid leukemia (CML), tyrosine kinase inhibitor (TKI) treatment induces autophagy that promotes survival and TKI-resistance in leukemic stem cells (LSCs). In clinical studies hydroxychloroquine (HCQ), the only clinically approved autophagy inhibitor, does not consistently inhibit autophagy in cancer patients, so more potent autophagy inhibitors are needed. We generated a murine model of CML in which autophagic flux can be measured in bone marrow-located LSCs. In parallel, we use cell division tracing, phenotyping of primary CML cells, and a robust xenotransplantation model of human CML, to investigate the effect of Lys05, a highly potent lysosomotropic agent, and PIK-III, a selective inhibitor of VPS34, on the survival and function of LSCs. We demonstrate that long-term haematopoietic stem cells (LT-HSCs: Lin-Sca-1+c-kit+CD48-CD150+) isolated from leukemic mice have higher basal autophagy levels compared with non-leukemic LT-HSCs and more mature leukemic cells. Additionally, we present that while HCQ is ineffective, Lys05-mediated autophagy inhibition reduces LSCs quiescence and drives myeloid cell expansion. Furthermore, Lys05 and PIK-III reduced the number of primary CML LSCs and target xenografted LSCs when used in combination with TKI treatment, providing a strong rationale for clinical use of second generation autophagy inhibitors as a novel treatment for CML patients with LSC persistence.


Subject(s)
Aminoquinolines/pharmacology , Autophagy , Drug Resistance, Neoplasm/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Polyamines/pharmacology , Animals , Apoptosis , Cell Proliferation , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured
4.
Nat Commun ; 8(1): 1804, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180704

ABSTRACT

Vps34 PI3K is thought to be the main producer of phosphatidylinositol-3-monophosphate, a lipid that controls intracellular vesicular trafficking. The organismal impact of systemic inhibition of Vps34 kinase activity is not completely understood. Here we show that heterozygous Vps34 kinase-dead mice are healthy and display a robustly enhanced insulin sensitivity and glucose tolerance, phenotypes mimicked by a selective Vps34 inhibitor in wild-type mice. The underlying mechanism of insulin sensitization is multifactorial and not through the canonical insulin/Akt pathway. Vps34 inhibition alters cellular energy metabolism, activating the AMPK pathway in liver and muscle. In liver, Vps34 inactivation mildly dampens autophagy, limiting substrate availability for mitochondrial respiration and reducing gluconeogenesis. In muscle, Vps34 inactivation triggers a metabolic switch from oxidative phosphorylation towards glycolysis and enhanced glucose uptake. Our study identifies Vps34 as a new drug target for insulin resistance in Type-2 diabetes, in which the unmet therapeutic need remains substantial.


Subject(s)
Insulin Resistance , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/physiology , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gene Knock-In Techniques , Glucose/analysis , Glucose/metabolism , Glucose Tolerance Test , Glycolysis/physiology , Hepatocytes , Heterozygote , Humans , Insulin/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Primary Cell Culture
5.
ACS Med Chem Lett ; 7(1): 72-6, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26819669

ABSTRACT

Autophagy is a dynamic process that regulates lysosomal-dependent degradation of cellular components. Until recently the study of autophagy has been hampered by the lack of reliable pharmacological tools, but selective inhibitors are now available to modulate the PI 3-kinase VPS34, which is required for autophagy. Here we describe the discovery of potent and selective VPS34 inhibitors, their pharmacokinetic (PK) properties, and ability to inhibit autophagy in cellular and mouse models.

6.
Nat Cell Biol ; 16(11): 1069-79, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25327288

ABSTRACT

Cells rely on autophagy to clear misfolded proteins and damaged organelles to maintain cellular homeostasis. In this study we use the new autophagy inhibitor PIK-III to screen for autophagy substrates. PIK-III is a selective inhibitor of VPS34 that binds a unique hydrophobic pocket not present in related kinases such as PI(3)Kα. PIK-III acutely inhibits autophagy and de novo lipidation of LC3, and leads to the stabilization of autophagy substrates. By performing ubiquitin-affinity proteomics on PIK-III-treated cells we identified substrates including NCOA4, which accumulates in ATG7-deficient cells and co-localizes with autolysosomes. NCOA4 directly binds ferritin heavy chain-1 (FTH1) to target the iron-binding ferritin complex with a relative molecular mass of 450,000 to autolysosomes following starvation or iron depletion. Interestingly, Ncoa4(-/-) mice exhibit a profound accumulation of iron in splenic macrophages, which are critical for the reutilization of iron from engulfed red blood cells. Taken together, the results of this study provide a new mechanism for selective autophagy of ferritin and reveal a previously unappreciated role for autophagy and NCOA4 in the control of iron homeostasis in vivo.


Subject(s)
Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Ferritins/metabolism , Homeostasis/physiology , Iron/metabolism , Nuclear Receptor Coactivators/metabolism , Animals , Autophagy/drug effects , Cells, Cultured , Humans , Lysosomes/metabolism , Mice , Phagosomes/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...