Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 34: 102055, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37928443

ABSTRACT

Insulin-like growth factor I (IGF-I) is a growth-promoting anabolic hormone that fosters cell growth and tissue homeostasis. IGF-I deficiency is associated with several diseases, including growth disorders and neurological and musculoskeletal diseases due to impaired regeneration. Despite the vast regenerative potential of IGF-I, its unfavorable pharmacokinetic profile has prevented it from being used therapeutically. In this study, we resolved these challenges by the local administration of IGF-I mRNA, which ensures desirable homeostatic kinetics and non-systemic, local dose-dependent expression of IGF-I protein. Furthermore, IGF-I mRNA constructs were sequence engineered with heterologous signal peptides, which improved in vitro protein secretion (2- to 6-fold) and accelerated in vivo functional regeneration (16-fold) over endogenous IGF-I mRNA. The regenerative potential of engineered IGF-I mRNA was validated in a mouse myotoxic muscle injury and rabbit spinal disc herniation models. Engineered IGF-I mRNA had a half-life of 17-25 h in muscle tissue and showed dose-dependent expression of IGF-I over 2-3 days. Animal models confirm that locally administered IGF-I mRNA remained at the site of injection, contributing to the safety profile of mRNA-based treatment in regenerative medicine. In summary, we demonstrate that engineered IGF-I mRNA holds therapeutic potential with high clinical translatability in different diseases.

2.
J Med Chem ; 63(22): 13595-13617, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33166139

ABSTRACT

The mechanistic target of rapamycin (mTOR) pathway is hyperactivated in cancer and neurological disorders. Rapalogs and mTOR kinase inhibitors (TORKi) have recently been applied to alleviate epileptic seizures in tuberous sclerosis complex (TSC). Herein, we describe a pharmacophore exploration to identify a highly potent, selective, brain penetrant TORKi. An extensive investigation of the morpholine ring engaging the mTOR solvent exposed region led to the discovery of PQR626 (8). 8 displayed excellent brain penetration and was well-tolerated in mice. In mice with a conditionally inactivated Tsc1 gene in glia, 8 significantly reduced the loss of Tsc1-induced mortality at 50 mg/kg p.o. twice a day. 8 overcomes the metabolic liabilities of PQR620 (52), the first-in-class brain penetrant TORKi showing efficacy in a TSC mouse model. The improved stability in human hepatocytes, excellent brain penetration, and efficacy in Tsc1GFAPCKO mice qualify 8 as a potential therapeutic candidate for the treatment of neurological disorders.


Subject(s)
Brain/metabolism , Morpholines/administration & dosage , Morpholines/metabolism , Nervous System Diseases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Administration, Oral , Animals , Brain/drug effects , Dogs , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Morpholines/chemistry , Nervous System Diseases/drug therapy , Rats , Rats, Sprague-Dawley
3.
Neuropharmacology ; 180: 108297, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32890589

ABSTRACT

Mechanistic target of rapamycin (mTOR) regulates cell proliferation, growth and survival, and is activated in cancer and neurological disorders, including epilepsy. The rapamycin derivative ("rapalog") everolimus, which allosterically inhibits the mTOR pathway, is approved for the treatment of partial epilepsy with spontaneous recurrent seizures (SRS) in individuals with tuberous sclerosis complex (TSC). In contrast to the efficacy in TSC, the efficacy of rapalogs on SRS in other types of epilepsy is equivocal. Furthermore, rapalogs only poorly penetrate into the brain and are associated with peripheral adverse effects, which may compromise their therapeutic efficacy. Here we compare the antiseizure efficacy of two novel, brain-permeable ATP-competitive and selective mTORC1/2 inhibitors, PQR620 and PQR626, and the selective dual pan-PI3K/mTORC1/2 inhibitor PQR530 in two mouse models of chronic epilepsy with SRS, the intrahippocampal kainate (IHK) mouse model of acquired temporal lobe epilepsy and Tsc1GFAP CKO mice, a well-characterized mouse model of epilepsy in TSC. During prolonged treatment of IHK mice with rapamycin, everolimus, PQR620, PQR626, or PQR530; only PQR620 exerted a transient antiseizure effect on SRS, at well tolerated doses whereas the other compounds were ineffective. In contrast, all of the examined compounds markedly suppressed SRS in Tsc1GFAP CKO mice during chronic treatment at well tolerated doses. Thus, against our expectation, no clear differences in antiseizure efficacy were found across the three classes of mTOR inhibitors examined in mouse models of genetic and acquired epilepsies. The main advantage of the novel 1,3,5-triazine derivatives is their excellent tolerability compared to rapalogs, which would favor their development as new therapies for TORopathies such as TSC.


Subject(s)
Epilepsies, Partial/drug therapy , Everolimus/therapeutic use , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Sirolimus/therapeutic use , Tuberous Sclerosis/drug therapy , Animals , Disease Models, Animal , Epilepsies, Partial/physiopathology , Everolimus/pharmacology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Knockout , Treatment Outcome , Tuberous Sclerosis/physiopathology
4.
Neuropharmacology ; 162: 107812, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31622602

ABSTRACT

One of the pathological hallmarks of Huntington disease (HD) is accumulation of the disease-causing mutant huntingtin (mHTT), which leads to the disruption of a variety of cellular functions, ultimately resulting in cell death. Induction of autophagy, for example by the inhibition of mechanistic target of rapamycin (mTOR) signaling, has been shown to reduce HTT levels and aggregates. While rapalogs like rapamycin allosterically inhibit the mTOR complex 1 (TORC1), ATP-competitive mTOR inhibitors suppress activities of TORC1 and TORC2 and have been shown to be more efficient in inducing autophagy and reducing protein levels and aggregates than rapalogs. The ability to cross the blood-brain barrier of first generation catalytic mTOR inhibitors has so far been limited, and therefore sufficient target coverage in the brain could not be reached. Two novel, brain penetrant compounds - the mTORC1/2 inhibitor PQR620, and the dual pan-phosphoinositide 3-kinase (PI3K) and mTORC1/2 kinase inhibitor PQR530 - were evaluated by assessing their potential to induce autophagy and reducing mHTT levels. For this purpose, expression levels of autophagic markers and well-defined mTOR targets were analyzed in STHdh cells and HEK293T cells and in mouse brains. Both compounds potently inhibited mTOR signaling in cell models as well as in mouse brain. As proof of principle, reduction of aggregates and levels of soluble mHTT were demonstrated upon treatment with both compounds. Originally developed for cancer treatment, these second generation mTORC1/2 and PI3K/mTOR inhibitors show brain penetrance and efficacy in cell models of HD, making them candidate molecules for further investigations in HD.


Subject(s)
Azabicyclo Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Morpholines/pharmacology , Neurons/drug effects , Protein Aggregates/drug effects , Pyridines/pharmacology , Triazines/pharmacology , Animals , Autophagy/drug effects , Blood-Brain Barrier , Cell Line , Corpus Striatum/cytology , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mice , Neurons/metabolism , Phosphatidylinositol 3-Kinases , Phosphoinositide-3 Kinase Inhibitors/pharmacology
5.
Neuropharmacology ; 162: 107817, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31654704

ABSTRACT

Dysregulation of the PI3K/Akt/mTOR pathway has been implicated in several brain disorders, including epilepsy. Rapamycin and similar compounds inhibit mTOR. complex 1 and have been reported to decrease seizures, delay seizure development, or prevent epileptogenesis in different animal models of genetic or acquired epilepsies. However, data for acquired epilepsy are inconsistent, which, at least in part, may be due to the poor brain penetration and long brain persistence of rapamycin and the fact that it blocks only one of the two cellular mTOR complexes. Here we examined the antiepileptogenic or disease-modifying effects of two novel, brain-permeable and well tolerated 1,3,5-triazine derivatives, the ATP-competitive mTORC1/2 inhibitor PQR620 and the dual pan-PI3K/mTORC1/2 inhibitor PQR530 in the intrahippocampal kainate mouse model, in which spontaneous seizures develop after status epilepticus (SE). Following kainate injection, the two compounds were administered over 2 weeks at doses previously been shown to block mTORC1/2 or PI3K/mTORC1/2 in the mouse brain. When spontaneous seizures were recorded by continuous (24/7) video-EEG recording starting 6 weeks after termination of treatment, no effects on incidence or frequency of seizures were observed. Drug treatment suppressed the epilepsy-induced activation of the PI3K/Akt/mTOR pathway in the hippocampus, but granule cell dispersion in the dentate gyrus was not prevented. When epilepsy-associated behavioral alterations were determined 12-14 weeks after kainate, mice pretreated with PQR620 or PQR530 exhibited reduced anxiety-related behavior in the light-dark box, indicating a disease-modifying effect. Overall, the data indicate that mTORC1/C2 or PI3K/mTORC1/C2 inhibition may not be an antiepileptogenic strategy for SE-induced epilepsy.


Subject(s)
Azabicyclo Compounds/pharmacology , Epilepsy, Temporal Lobe/prevention & control , Hippocampus/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Triazines/pharmacology , Animals , Anxiety , Behavior, Animal/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Disease Models, Animal , Electroencephalography , Enzyme Inhibitors/pharmacology , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/etiology , Excitatory Amino Acid Agonists/toxicity , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Mice , Phosphatidylinositol 3-Kinases , Seizures , Signal Transduction , Status Epilepticus/chemically induced , Status Epilepticus/complications
6.
Int J Mol Sci ; 20(22)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752127

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway has been implicated as a cancer target. Big pharma players and small companies have been developing small molecule inhibitors of PI3K and/or mTOR since the 1990s. Although four inhibitors have been approved, many open questions regarding tolerability, patient selection, sensitivity markers, development of resistances, and toxicological challenges still need to be addressed. Besides clear oncological indications, PI3K and mTOR inhibitors have been suggested for treating a plethora of different diseases. In particular, genetically induced PI3K/mTOR pathway activation causes rare disorders, known as overgrowth syndromes, like PTEN (phosphatase and tensin homolog) hamartomas, tuberous sclerosis complex (TSC), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA)-related overgrowth spectrum (PROS), and activated PI3-Kinase delta syndrome (PI3KCD, APDS). Some of those disorders likeTSC or hemimegalencephaly, which are one of the PROS disorders, also belong to a group of diseases called mTORopathies. This group of syndromes presents with additional neurological manifestations associated with epilepsy and other neuropsychiatric symptoms induced by neuronal mTOR pathway hyperactivation. While PI3K and mTOR inhibitors have been and still are intensively tested in oncology indications, their use in genetically defined syndromes and mTORopathies appear to be promising avenues for a pharmacological intervention.


Subject(s)
Neoplasms/drug therapy , Rare Diseases/drug therapy , Signal Transduction/drug effects , Small Molecule Libraries/therapeutic use , Animals , Humans , Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rare Diseases/metabolism , Small Molecule Libraries/pharmacology , TOR Serine-Threonine Kinases/metabolism
7.
ACS Med Chem Lett ; 10(10): 1473-1479, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31620236

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is a critical regulator of cell growth and is frequently hyperactivated in cancer. Therefore, PI3K inhibitors represent a valuable asset in cancer therapy. Herein we have developed a novel anticancer agent, the potent pan-PI3K inhibitor PQR514 (4), which is a follow-up compound for the phase-II clinical compound PQR309 (1). Compound 4 has an improved potency both in vitro and in cellular assays with respect to its predecessor compounds. It shows superiority in the suppression of cancer cell proliferation and demonstrates significant antitumor activity in an OVCAR-3 xenograft model at concentrations approximately eight times lower than PQR309 (1). The favorable pharmacokinetic profile and a minimal brain penetration promote PQR514 (4) as an optimized candidate for the treatment of systemic tumors.

8.
J Med Chem ; 62(18): 8609-8630, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31465220

ABSTRACT

The mechanistic target of rapamycin (mTOR) plays a pivotal role in growth and tumor progression and is an attractive target for cancer treatment. ATP-competitive mTOR kinase inhibitors (TORKi) have the potential to overcome limitations of rapamycin derivatives in a wide range of malignancies. Herein, we exploit a conformational restriction approach to explore a novel chemical space for the generation of TORKi. Structure-activity relationship (SAR) studies led to the identification of compound 12b with a ∼450-fold selectivity for mTOR over class I PI3K isoforms. Pharmacokinetic studies in male Sprague Dawley rats highlighted a good exposure after oral dosing and a minimum brain penetration. CYP450 reactive phenotyping pointed out the high metabolic stability of 12b. These results identify the tricyclic pyrimido-pyrrolo-oxazine moiety as a novel scaffold for the development of highly selective mTOR inhibitors for cancer treatment.


Subject(s)
Oxazines/chemistry , Protein Kinase Inhibitors/chemistry , Pyrimidinones/chemistry , Pyrroles/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Dogs , Drug Design , Humans , Inhibitory Concentration 50 , Kinetics , Male , Mice , Molecular Conformation , Neoplasms/drug therapy , Oxazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidinones/pharmacokinetics , Pyrroles/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , TOR Serine-Threonine Kinases/chemistry
9.
Cancers (Basel) ; 11(6)2019 Jun 04.
Article in English | MEDLINE | ID: mdl-31167506

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling cascade is an important therapeutic target for lymphomas. Rapamycin-derivates as allosteric mTOR complex 1 (TORC1) inhibitors have shown moderate preclinical and clinical anti-lymphoma activity. Here, we assessed the anti-tumor activity of PQR620, a novel brain penetrant dual TORC1/2 inhibitor, in 56 lymphoma cell lines. We observed anti-tumor activity across 56 lymphoma models with a median IC50 value of 250 nM after 72 h of exposure. PQR620 was largely cytostatic, but the combination with the BCL2 inhibitor venetoclax led to cytotoxicity. Both the single agent and the combination data were validated in xenograft models. The data support further evaluation of PQR620 as a single agent or in combination with venetoclax.

10.
J Med Chem ; 62(13): 6241-6261, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31244112

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/mechanistic target of rapamycin (mTOR) pathway is frequently overactivated in cancer, and drives cell growth, proliferation, survival, and metastasis. Here, we report a structure-activity relationship study, which led to the discovery of a drug-like adenosine 5'-triphosphate-site PI3K/mTOR kinase inhibitor: (S)-4-(difluoromethyl)-5-(4-(3-methylmorpholino)-6-morpholino-1,3,5-triazin-2-yl)pyridin-2-amine (PQR530, compound 6), which qualifies as a clinical candidate due to its potency and specificity for PI3K and mTOR kinases, and its pharmacokinetic properties, including brain penetration. Compound 6 showed excellent selectivity over a wide panel of kinases and an excellent selectivity against unrelated receptor enzymes and ion channels. Moreover, compound 6 prevented cell growth in a cancer cell line panel. The preclinical in vivo characterization of compound 6 in an OVCAR-3 xenograft model demonstrated good oral bioavailability, excellent brain penetration, and efficacy. Initial toxicity studies in rats and dogs qualify 6 for further development as a therapeutic agent in oncology.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyridines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazines/pharmacology , Aminopyridines/chemical synthesis , Aminopyridines/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Brain/metabolism , Cell Line, Tumor , Dogs , Female , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Morpholines/chemical synthesis , Morpholines/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors/metabolism , Protein Binding , Pyridines/chemical synthesis , Pyridines/metabolism , Rats, Wistar , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Triazines/chemical synthesis , Triazines/metabolism , Xenograft Model Antitumor Assays
12.
J Med Chem ; 61(22): 10084-10105, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30359003

ABSTRACT

Mechanistic target of rapamycin (mTOR) promotes cell proliferation, growth, and survival and is overactivated in many tumors and central nervous system disorders. PQR620 (3) is a novel, potent, selective, and brain penetrable inhibitor of mTORC1/2 kinase. PQR620 (3) showed excellent selectivity for mTOR over PI3K and protein kinases and efficiently prevented cancer cell growth in a 66 cancer cell line panel. In C57BL/6J and Sprague-Dawley mice, maximum concentration ( Cmax) in plasma and brain was reached after 30 min, with a half-life ( t1/2) > 5 h. In an ovarian carcinoma mouse xenograft model (OVCAR-3), daily dosing of PQR620 (3) inhibited tumor growth significantly. Moreover, PQR620 (3) attenuated epileptic seizures in a tuberous sclerosis complex (TSC) mouse model. In conclusion, PQR620 (3) inhibits mTOR kinase potently and selectively, shows antitumor effects in vitro and in vivo, and promises advantages in CNS indications due to its brain/plasma distribution ratio.


Subject(s)
Azabicyclo Compounds/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Pyridines/pharmacology , Seizures/drug therapy , Triazines/pharmacology , Animals , Azabicyclo Compounds/metabolism , Azabicyclo Compounds/therapeutic use , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Models, Molecular , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Conformation , Pyridines/metabolism , Pyridines/therapeutic use , Rats , Triazines/metabolism , Triazines/therapeutic use
13.
Neuropharmacology ; 140: 107-120, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30081001

ABSTRACT

The mTOR signaling pathway has emerged as a possible therapeutic target for epilepsy. Clinical trials have shown that mTOR inhibitors such as everolimus reduce seizures in tuberous sclerosis complex patients with intractable epilepsy. Furthermore, accumulating preclinical data suggest that mTOR inhibitors may have anti-seizure or anti-epileptogenic actions in other types of epilepsy. However, the chronic use of rapalogs such as everolimus is limited by poor tolerability, particularly by immunosuppression, poor brain penetration and induction of feedback loops which might contribute to their limited therapeutic efficacy. Here we describe two novel, brain-permeable and well tolerated small molecule 1,3,5-triazine derivatives, the catalytic mTORC1/C2 inhibitor PQR620 and the dual pan-PI3K/mTOR inhibitor PQR530. These derivatives were compared with the mTORC1 inhibitors rapamycin and everolimus as well as the anti-seizure drugs phenobarbital and levetiracetam. The anti-seizure potential of these compounds was determined by evaluating the electroconvulsive seizure threshold in normal and epileptic mice. Rapamycin and everolimus only poorly penetrated into the brain (brain:plasma ratio 0.0057 for rapamycin and 0.016 for everolimus). In contrast, the novel compounds rapidly entered the brain, reaching brain:plasma ratios of ∼1.6. Furthermore, they significantly decreased phosphorylation of S6 ribosomal protein in the hippocampus of normal and epileptic mice, demonstrating effective mTOR inhibition. PQR620 and PQR530 significantly increased seizure threshold at tolerable doses. The effect of PQR620 was more marked in epileptic vs. nonepileptic mice, matching the efficacy of levetiracetam. Overall, the novel compounds described here have the potential to overcome the disadvantages of rapalogs for treatment of epilepsy and mTORopathies directly connected to mutations in the mTOR signaling cascade.


Subject(s)
Anticonvulsants , Azabicyclo Compounds , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Enzyme Inhibitors/pharmacology , Epilepsy/complications , Epilepsy/drug therapy , Morpholines , Pyridines , Seizures/complications , Seizures/prevention & control , Triazines , Animals , Anticonvulsants/blood , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Azabicyclo Compounds/blood , Azabicyclo Compounds/pharmacokinetics , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Catalysis/drug effects , Electroshock , Everolimus/blood , Everolimus/pharmacokinetics , Everolimus/pharmacology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Levetiracetam/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mice , Morpholines/blood , Morpholines/pharmacokinetics , Morpholines/pharmacology , Morpholines/therapeutic use , Phenobarbital/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pyridines/blood , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , Ribosomal Proteins/metabolism , Sirolimus/blood , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Triazines/blood , Triazines/pharmacokinetics , Triazines/pharmacology , Triazines/therapeutic use
14.
Clin Cancer Res ; 24(1): 120-129, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29066507

ABSTRACT

Purpose: Activation of the PI3K/mTOR signaling pathway is recurrent in different lymphoma types, and pharmacologic inhibition of the PI3K/mTOR pathway has shown activity in lymphoma patients. Here, we extensively characterized the in vitro and in vivo activity and the mechanism of action of PQR309 (bimiralisib), a novel oral selective dual PI3K/mTOR inhibitor under clinical evaluation, in preclinical lymphoma models.Experimental Design: This study included preclinical in vitro activity screening on a large panel of cell lines, both as single agent and in combination, validation experiments on in vivo models and primary cells, proteomics and gene-expression profiling, and comparison with other signaling inhibitors.Results: PQR309 had in vitro antilymphoma activity as single agent and in combination with venetoclax, panobinostat, ibrutinib, lenalidomide, ARV-825, marizomib, and rituximab. Sensitivity to PQR309 was associated with specific baseline gene-expression features, such as high expression of transcripts coding for the BCR pathway. Combining proteomics and RNA profiling, we identified the different contribution of PQR309-induced protein phosphorylation and gene expression changes to the drug mechanism of action. Gene-expression signatures induced by PQR309 and by other signaling inhibitors largely overlapped. PQR309 showed activity in cells with primary or secondary resistance to idelalisib.Conclusions: On the basis of these results, PQR309 appeared as a novel and promising compound that is worth developing in the lymphoma setting. Clin Cancer Res; 24(1); 120-9. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphoma/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm , Humans , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Purines , Quinazolinones , Xenograft Model Antitumor Assays
15.
J Med Chem ; 60(17): 7524-7538, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28829592

ABSTRACT

Phosphoinositide 3-kinase (PI3K) is deregulated in a wide variety of human tumors and triggers activation of protein kinase B (PKB/Akt) and mammalian target of rapamycin (mTOR). Here we describe the preclinical characterization of compound 1 (PQR309, bimiralisib), a potent 4,6-dimorpholino-1,3,5-triazine-based pan-class I PI3K inhibitor, which targets mTOR kinase in a balanced fashion at higher concentrations. No off-target interactions were detected for 1 in a wide panel of protein kinase, enzyme, and receptor ligand assays. Moreover, 1 did not bind tubulin, which was observed for the structurally related 4 (BKM120, buparlisib). Compound 1 is orally available, crosses the blood-brain barrier, and displayed favorable pharmacokinetic parameters in mice, rats, and dogs. Compound 1 demonstrated efficiency in inhibiting proliferation in tumor cell lines and a rat xenograft model. This, together with the compound's safety profile, identifies 1 as a clinical candidate with a broad application range in oncology, including treatment of brain tumors or CNS metastasis. Compound 1 is currently in phase II clinical trials for advanced solid tumors and refractory lymphoma.


Subject(s)
Aminopyridines/therapeutic use , Antineoplastic Agents/therapeutic use , Morpholines/therapeutic use , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Aminopyridines/administration & dosage , Aminopyridines/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Brain/drug effects , Brain/metabolism , Cell Proliferation/drug effects , Dogs , Humans , Mice , Models, Molecular , Morpholines/administration & dosage , Morpholines/pharmacokinetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Rats , Rats, Nude , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
16.
Nature ; 491(7424): 458-462, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23064226

ABSTRACT

Cancer cells exhibit several unique metabolic phenotypes that are critical for cell growth and proliferation. Specifically, they overexpress the M2 isoform of the tightly regulated enzyme pyruvate kinase (PKM2), which controls glycolytic flux, and are highly dependent on de novo biosynthesis of serine and glycine. Here we describe a new rheostat-like mechanistic relationship between PKM2 activity and serine biosynthesis. We show that serine can bind to and activate human PKM2, and that PKM2 activity in cells is reduced in response to serine deprivation. This reduction in PKM2 activity shifts cells to a fuel-efficient mode in which more pyruvate is diverted to the mitochondria and more glucose-derived carbon is channelled into serine biosynthesis to support cell proliferation.


Subject(s)
Ligands , Pyruvate Kinase/metabolism , Serine/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Glucose/metabolism , Glycine/metabolism , Glycine/pharmacology , Humans , Pyruvate Kinase/genetics , Pyruvic Acid/metabolism , Recombinant Proteins/metabolism , Serine/pharmacology
17.
Cell Cycle ; 10(21): 3731-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22045127

ABSTRACT

Addition of short (6 to 16 amino acids) peptide sequences to the N-terminus of p110α induces a gain of function. Such sequences include the common Flag, His, and VSV tags as well as random sequences. An N-terminal myristylation signal generally believed to activate p110α by providing a constitutive membrane address is also activating, if myristylation is mutationally abolished. The gain of function seen with N-terminally tagged (NTT) p110α constructs extends to signaling, oncogenic transformation and stimulation of cell growth. The activating effect of N-terminal tags requires a functional Ras-binding domain in p110α. Mutations in that domain (T208D and K227A) abolish the gains of function in oncogenicity and signaling. The dominant negative mutant of Ras, RasN17, interferes with transformation induced by NTT p110α. In contrast, binding to p85 activity is not required for cellular transformation and enhanced signaling by NTT p110α.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/physiology , Signal Transduction , Amino Acid Sequence , Animals , Catalytic Domain , Cell Transformation, Neoplastic , Chick Embryo , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/metabolism , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Protein Subunits/physiology , ras Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 107(35): 15547-52, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713702

ABSTRACT

Cancer-specific mutations in the iSH2 (inter-SH2) and nSH2 (N-terminal SH2) domains of p85alpha, the regulatory subunit of phosphatidylinositide 3-kinase (PI3K), show gain of function. They induce oncogenic cellular transformation, stimulate cellular proliferation, and enhance PI3K signaling. Quantitative determinations of oncogenic activity reveal large differences between individual mutants of p85alpha. The mutant proteins are still able to bind to the catalytic subunits p110alpha and p110beta. Studies with isoform-specific inhibitors of p110 suggest that expression of p85 mutants in fibroblasts leads exclusively to an activation of p110alpha, and p110alpha is the sole mediator of p85 mutant-induced oncogenic transformation. The characteristics of the p85 mutants are in agreement with the hypothesis that the mutations weaken an inhibitory interaction between p85alpha and p110alpha while preserving the stabilizing interaction between p85alpha iSH2 and the adapter-binding domain of p110alpha.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Catalytic Domain/genetics , Cell Line , Cell Proliferation , Cells, Cultured , Chick Embryo , Class I Phosphatidylinositol 3-Kinases , Dioxoles/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoprecipitation , Morpholines/pharmacology , Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Binding , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Pyrimidinones/pharmacology , Quinazolines/pharmacology , Thiazolidinediones/pharmacology , Transfection
19.
J Med Chem ; 52(9): 2762-75, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19419204

ABSTRACT

The P2Y(2) receptor, which is activated by UTP, ATP, and dinucleotides, was studied as a prototypical nucleotide-activated GPCR. A combination of receptor mutagenesis, determination of its effects on potency and efficacy of agonists and antagonists, homology modeling, and chemical experiments was applied. R272 (extracellular loop EL3) was found to play a gatekeeper role, presumably responsible for recognition and orientation of the nucleotides. R272 is also directly involved in binding of dinucleotides, which behaved as partial agonists. Y118A (3.37) mutation led to dramatically reduced efficacy of agonists; it is part of the entry channel as well as the triphosphate binding site. While the Y114A (3.33) mutation did not have any effect on agonist activities, the antagonist Reactive Blue 2 (6) was completely inactive at that mutant. The disulfide bridge Cys25-Cys278 was found to be important for agonist potency but neither for agonist efficacy nor for antagonist potency.


Subject(s)
Models, Molecular , Mutagenesis, Site-Directed , Nucleotides/pharmacology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Amino Acids/metabolism , Animals , Cell Line , Disulfides/chemistry , Drug Design , Enzyme-Linked Immunosorbent Assay , Extracellular Space/metabolism , Gene Expression , Humans , Ligands , Molecular Sequence Data , Oxidation-Reduction , Protein Structure, Secondary , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2Y2
20.
Toxicol Appl Pharmacol ; 227(1): 36-47, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18061226

ABSTRACT

High blood levels of ammonium/ammonia (NH(4)(+)/NH(3)) are associated with severe neurotoxicity as observed in hepatic encephalopathy (HE). Astrocytes are the main targets of ammonium toxicity, while neuronal cells are less vulnerable. In the present study, an astrocytoma cell line 1321N1 and a neuroblastoma glioma hybrid cell line NG108-15 were used as model systems for astrocytes and neuronal cells, respectively. Ammonium salts evoked a transient increase in intracellular calcium concentrations ([Ca(2+)](i)) in astrocytoma (EC(50)=6.38 mM), but not in NG108-15 cells. The ammonium-induced increase in [Ca(2+)](i) was due to an intracellular effect of NH(4)(+)/NH(3) and was independent of extracellular calcium. Acetate completely inhibited the ammonium effect. Ammonium potently reduced calcium signaling by G(q) protein-coupled receptors (H(1) and M3) expressed on the cells. Ammonium (5 mM) also significantly inhibited the proliferation of 1321N1 astrocytoma cells. While mRNA for the mammalian ammonium transporters RhBG and RhCG could not be detected in 1321N1 astrocytoma cells, both transporters were expressed in NG108-15 cells. RhBG and RhBC in brain may promote the excretion of NH(3)/NH(4)(+) from neuronal cells. Cellular uptake of NH(4)(+)/NH(3) was mainly by passive diffusion of NH(3). Human 1321N1 astrocytoma cells appear to be an excellent, easily accessible human model for studying HE, which can substitute animal studies, while NG108-15 cells may be useful for investigating the role of the recently discovered Rhesus family type ammonium transporters in neuronal cells. Our findings may contribute to the understanding of pathologic ammonium effects in different brain cells, and to the treatment of hyperammonemia.


Subject(s)
Astrocytoma/metabolism , Calcium/metabolism , Quaternary Ammonium Compounds/pharmacology , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Potassium Chloride/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...