65 results on '"Cosulich S"'
Search Results
2. Abstract PD4-04: Combined inhibition of mTOR and CDK4/6 is required for optimal blockade of E2F function and long term growth inhibition in estrogen receptor positive breast cancer
- Author
-
Oelmann, E, primary, Michaloglou, C, additional, Crafter, C, additional, Siersbaek, R, additional, Delpuech, O, additional, Curven, J, additional, Carnevalli, L, additional, Staniszweska, A, additional, Polanska, U, additional, Cheraghchi-Bashi, A, additional, Lawson, M, additional, Chernukhin, I, additional, McEwen, R, additional, Carroll, J, additional, and Cosulich, S, additional
- Published
- 2018
- Full Text
- View/download PDF
3. 272 8-(1-Anilino)ethyl)-2-morpholino-4-oxo-4H-chromene-6-carboxamides as PI3Kbeta/delta inhibitors: structure–activity relationships and identification of AZD8186, a clinical candidate for the treatment of PTEN deficient tumours
- Author
-
Barlaam, B., primary, Cosulich, S., additional, Degorce, S., additional, Fitzek, M., additional, Green, S., additional, Hancox, U., additional, Lambert-van der Brempt, C., additional, Lohmann, J.J., additional, Maudet, M., additional, Morgentin, R., additional, Pasquest, M.J., additional, Peru, A., additional, Ple, P., additional, Saleh, T., additional, Vautier, M., additional, Walker, M., additional, Ward, L., additional, and Warin, N., additional
- Published
- 2014
- Full Text
- View/download PDF
4. 275 Modulation of PIP2 levels through small molecule inhibition of PIP5K
- Author
-
Andrews, D., primary, Cosulich, S., additional, Divecha, N., additional, Fitzgerald, D., additional, Flemington, V., additional, Jones, C., additional, Jones, D., additional, Kern, O., additional, MacDonald, E., additional, Maman, S., additional, McKelvie, J., additional, Pike, K., additional, Riddick, M., additional, Robb, G., additional, Roberts, K., additional, Smith, J., additional, Swarbrick, M., additional, Treinies, I., additional, Waring, M., additional, and Wood, R., additional
- Published
- 2014
- Full Text
- View/download PDF
5. Mutant p53 is not fully dominant over endogenous wild type p53 in a colorectal adenoma cell line as demonstrated by induction of MDM2 protein and retention of a p53 dependent G1 arrest after gamma irradiation
- Author
-
Ann Williams, Jc, Miller, Tj, Collard, Ts, Bracey, Cosulich S, and Paraskeva C
- Subjects
Adenoma ,Base Sequence ,Molecular Sequence Data ,G1 Phase ,Nuclear Proteins ,Proto-Oncogene Proteins c-mdm2 ,Genes, p53 ,Transfection ,Neoplasm Proteins ,Gamma Rays ,Transforming Growth Factor beta ,Proto-Oncogene Proteins ,Mutation ,Tumor Cells, Cultured ,Humans ,Colorectal Neoplasms ,Cell Division ,DNA Primers ,Genes, Dominant - Abstract
To determine whether a single mutational event in one p53 gene is sufficient to confer a significant growth advantage on a colonic epithelial cell, the 143(Ala) p53 mutation was previously expressed in the human colonic adenoma derived cell line AA/C1 (which is wild type for p53) and shown to have no effect on it's in vitro or in vivo growth characteristics. In this investigation, by expressing the 175(His), 248(Trp) or 273(His) mutations in the same AA/C1 cell line, we have shown that this failure to affect the growth of the cells was not mutant specific. We have also demonstrated, using induction of MDM2 protein and the ability of the cells to undergo a p53 dependent G1 arrest, that the 143(Ala), 175(His) or 248(Trp) transfected cells retain functional endogenous wild type p53 activity, and suggest that these p53 mutations would not have a fully dominant negative mode of action in vivo. In contrast, one of the two AA/C1 cell lines transfected with the 273(His) mutation did fail to cell cycle arrest after gamma irradiation, indicating that this mutation can act as a dominant negative. However even loss of wild type p53 function in this cell line was insufficient to directly effect the growth rate of the AA/C1 cells, suggesting that acquisition of the 273(His) mutation may contribute to malignant progression through genomic instability (by inhibiting the G1 arrest) and that other mutations are required before outgrowth of the cell population containing the p53 mutation.
- Published
- 1995
6. Role of cytokines in non-genotoxic hepatocarcinogenesis: cause or effect?
- Author
-
Roberts, R.A., primary, James, N.H., additional, Cosulich, S., additional, Hasmall, S.C., additional, and Orphanides, G., additional
- Published
- 2001
- Full Text
- View/download PDF
7. Role of MAP kinase signalling pathways in the mode of action of peroxisome proliferators
- Author
-
Cosulich, S., primary, James, N., additional, and Roberts, R., additional
- Published
- 2000
- Full Text
- View/download PDF
8. Cleavage of Rabaptin-5 blocks endosome fusion during apoptosis
- Author
-
Cosulich, S. C., primary
- Published
- 1997
- Full Text
- View/download PDF
9. Effects of fluorinated inositols on the proliferation of Swiss 3T3 fibroblasts
- Author
-
Cosulich, S C, primary, Offer, J, additional, Smith, G A, additional, Hesketh, R, additional, and Metcalfe, J C, additional
- Published
- 1993
- Full Text
- View/download PDF
10. A dominant negative form of IKK2 prevents suppression of apoptosis by the peroxisome proliferator nafenopin.
- Author
-
Cosulich, S C, James, N H, Needham, M R, Newham, P P, Bundell, K R, and Roberts, R A
- Abstract
Peroxisome proliferators (PPs) are a class of non-genotoxic chemicals that cause rodent liver enlargement and hepatocarcinogenesis. In primary rat hepatocyte cultures, PPs suppress spontaneous apoptosis and that induced by a number of pro-apoptotic stimuli such as transforming growth factor-beta(1). Tumour necrosis factor alpha (TNF-alpha) and the transcription factor NFkappaB have been implicated in the mode of action of PPs. TNF-alpha signalling to NFkappaB is thought to be responsible for many of the effects elicited by this cytokine. NFkappaB regulates gene expression in immunity, stress responses and the inhibition of apoptosis. Activation of NFkappaB requires the successive action of NFkappaB-inducing kinase and the phosphorylation of NFkappaB inhibitory proteins (IkappaB) by an IkappaB kinase (IKK) complex. The IKK2 subunit of IkappaB kinase is thought to be essential for NFkappaB activation and prevention of apoptosis. To determine whether IKK2 plays a role in the suppression of apoptosis by PPs, we expressed a dominant negative form of IKK2 (IKK2dn) in primary rat hepatocyte cultures. Infection with an adenovirus construct expressing IKK2dn caused apoptosis in control primary rat hepatocytes in the absence of exogenous TNF-alpha. Moreover, IKK2dn-induced apoptosis could not be rescued by addition of TNF-alpha or the peroxisome proliferator nafenopin. These results demonstrate a requirement for intracellular signalling pathways mediated by IKK2 in the suppression of apoptosis by the PP class of hepatocarcinogens.
- Published
- 2000
- Full Text
- View/download PDF
11. Cleavage and inactivation of DNA-dependent protein kinase catalytic subunit during apoptosis in Xenopus egg extracts.
- Author
-
Le Romancer, M, Cosulich, S C, Jackson, S P, and Clarke, P R
- Abstract
DNA-dependent protein kinase (DNA-PK) consists of a 460 kDa subunit that contains the catalytic domain (DNA-PKcs) complexed with two polypeptides of 70 kDa and 80 kDa (Ku70 and Ku80) which comprise the Ku autoantigen. DNA-PKcs requires association with DNA via Ku for catalytic activation and is implicated in double strand break repair, V(D)J recombination and transcription. We have utilised a cell-free system of concentrated Xenopus laevis egg extracts to investigate the regulation and possible functions of DNA-PK. Recently, we have shown that this system can reproduce events of apoptosis, including activation of an apoptotic protease that cleaves poly(ADP-ribose) polymerase. Here, we report that DNA-PK is rapidly inactivated with the onset of apoptosis in this system. Loss of activity is concomitant with cleavage of the catalytic subunit, whereas the Ku subunits are stable. Cleavage and inactivation of DNA-PKcs is prevented by prior addition of the anti-apoptotic protein Bcl-2 or inhibition of an apoptotic protease that has characteristics of the CPP-32/Ced-3 family of cysteine proteases that cleave poly(ADP-ribose) polymerase. These results suggest that cleavage and inactivation of DNA-PKcs prevents this factor from functioning in DNA repair, recombination or transcriptional regulation during apoptosis.
- Published
- 1996
12. Bcl-2 regulates activation of apoptotic proteases in a cell-free system
- Author
-
COSULICH, S
- Published
- 1996
- Full Text
- View/download PDF
13. Preclinical Characterization of AZD9574, a Blood-Brain Barrier Penetrant Inhibitor of PARP1.
- Author
-
Staniszewska AD, Pilger D, Gill SJ, Jamal K, Bohin N, Guzzetti S, Gordon J, Hamm G, Mundin G, Illuzzi G, Pike A, McWilliams L, Maglennon G, Rose J, Hawthorne G, Cortes Gonzalez M, Halldin C, Johnström P, Schou M, Critchlow SE, Fawell S, Johannes JW, Leo E, Davies BR, Cosulich S, Sarkaria JN, O'Connor MJ, and Hamerlik P
- Subjects
- Animals, Humans, Mice, Rats, Antineoplastic Agents, Alkylating pharmacology, Blood-Brain Barrier metabolism, Cell Line, Tumor, DNA, O(6)-Methylguanine-DNA Methyltransferase genetics, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Temozolomide pharmacology, Temozolomide therapeutic use, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioma drug therapy, Glioma pathology
- Abstract
Purpose: We evaluated the properties and activity of AZD9574, a blood-brain barrier (BBB) penetrant selective inhibitor of PARP1, and assessed its efficacy and safety alone and in combination with temozolomide (TMZ) in preclinical models., Experimental Design: AZD9574 was interrogated in vitro for selectivity, PARylation inhibition, PARP-DNA trapping, the ability to cross the BBB, and the potential to inhibit cancer cell proliferation. In vivo efficacy was determined using subcutaneous as well as intracranial mouse xenograft models. Mouse, rat, and monkey were used to assess AZD9574 BBB penetration and rat models were used to evaluate potential hematotoxicity for AZD9574 monotherapy and the TMZ combination., Results: AZD9574 demonstrated PARP1-selectivity in fluorescence anisotropy, PARylation, and PARP-DNA trapping assays and in vivo experiments demonstrated BBB penetration. AZD9574 showed potent single agent efficacy in preclinical models with homologous recombination repair deficiency in vitro and in vivo. In an O6-methylguanine-DNA methyltransferase (MGMT)-methylated orthotopic glioma model, AZD9574 in combination with TMZ was superior in extending the survival of tumor-bearing mice compared with TMZ alone., Conclusions: The combination of three key features-PARP1 selectivity, PARP1 trapping profile, and high central nervous system penetration in a single molecule-supports the development of AZD9574 as the best-in-class PARP inhibitor for the treatment of primary and secondary brain tumors. As documented by in vitro and in vivo studies, AZD9574 shows robust anticancer efficacy as a single agent as well as in combination with TMZ. AZD9574 is currently in a phase I trial (NCT05417594). See related commentary by Lynce and Lin, p. 1217., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
14. PLEKHS1 drives PI3Ks and remodels pathway homeostasis in PTEN-null prostate.
- Author
-
Chessa TAM, Jung P, Anwar A, Suire S, Anderson KE, Barneda D, Kielkowska A, Sadiq BA, Lai IW, Felisbino S, Turnham DJ, Pearson HB, Phillips WA, Sasaki J, Sasaki T, Oxley D, Spensberger D, Segonds-Pichon A, Wilson M, Walker S, Okkenhaug H, Cosulich S, Hawkins PT, and Stephens LR
- Subjects
- Animals, Humans, Male, Mice, Homeostasis, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Prostate pathology, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism
- Abstract
The PIP
3 /PI3K network is a central regulator of metabolism and is frequently activated in cancer, commonly by loss of the PIP3 /PI(3,4)P2 phosphatase, PTEN. Despite huge research investment, the drivers of the PI3K network in normal tissues and how they adapt to overactivation are unclear. We find that in healthy mouse prostate PI3K activity is driven by RTK/IRS signaling and constrained by pathway feedback. In the absence of PTEN, the network is dramatically remodeled. A poorly understood YXXM- and PIP3 /PI(3,4)P2 -binding PH domain-containing adaptor, PLEKHS1, became the dominant activator and was required to sustain PIP3 , AKT phosphorylation, and growth in PTEN-null prostate. This was because PLEKHS1 evaded pathway-feedback and experienced enhanced PI3K- and Src-family kinase-dependent phosphorylation of Y258 XXM, eliciting PI3K activation. hPLEKHS1 mRNA and activating Y419 phosphorylation of hSrc correlated with PI3K pathway activity in human prostate cancers. We propose that in PTEN-null cells receptor-independent, Src-dependent tyrosine phosphorylation of PLEKHS1 creates positive feedback that escapes homeostasis, drives PIP3 signaling, and supports tumor progression., Competing Interests: Declaration of interests S.C. is an employee of AZ. We, the authors, have a patent related to this work (patent application number 2304156.9, granted by United Kingdom Patent Office, covering “modulating PLEKHS1 activity in a cell”)., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Discovery of AZD4747, a Potent and Selective Inhibitor of Mutant GTPase KRAS G12C with Demonstrable CNS Penetration.
- Author
-
Kettle JG, Bagal SK, Barratt D, Bodnarchuk MS, Boyd S, Braybrooke E, Breed J, Cassar DJ, Cosulich S, Davies M, Davies NL, Deng C, Eatherton A, Evans L, Feron LJ, Fillery S, Gleave ES, Goldberg FW, Cortés González MA, Guerot C, Haider A, Harlfinger S, Howells R, Jackson A, Johnström P, Kemmitt PD, Koers A, Kondrashov M, Lamont GM, Lamont S, Lewis HJ, Liu L, Mylrea M, Nash S, Niedbala MJ, Peter A, Phillips C, Pike K, Raubo P, Robb GR, Ross S, Sanders MG, Schou M, Simpson I, and Steward O
- Subjects
- Animals, Humans, Proto-Oncogene Proteins p21(ras) genetics, Drug Design, Glycine therapeutic use, Mutation, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Lung Neoplasms drug therapy
- Abstract
The glycine to cysteine mutation at codon 12 of Kirsten rat sarcoma (KRAS) represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 14 , AZD4747, a clinical development candidate for the treatment of KRAS
G12C -positive tumors, including the treatment of central nervous system (CNS) metastases. Building on our earlier discovery of C5-tethered quinazoline AZD4625, excision of a usually critical pyrimidine ring yielded a weak but brain-penetrant start point which was optimized for potency and DMPK. Key design principles and measured parameters that give high confidence in CNS exposure are discussed. During optimization, divergence between rodent and non-rodent species was observed in CNS exposure, with primate PET studies ultimately giving high confidence in the expected translation to patients. AZD4747 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.- Published
- 2023
- Full Text
- View/download PDF
16. Preclinical Characterization of AZD5305, A Next-Generation, Highly Selective PARP1 Inhibitor and Trapper.
- Author
-
Illuzzi G, Staniszewska AD, Gill SJ, Pike A, McWilliams L, Critchlow SE, Cronin A, Fawell S, Hawthorne G, Jamal K, Johannes J, Leonard E, Macdonald R, Maglennon G, Nikkilä J, O'Connor MJ, Smith A, Southgate H, Wilson J, Yates J, Cosulich S, and Leo E
- Subjects
- Humans, Mice, Rats, Animals, Cell Line, Tumor, Xenograft Model Antitumor Assays, Phthalazines pharmacology, Poly (ADP-Ribose) Polymerase-1, DNA Repair, Poly(ADP-ribose) Polymerase Inhibitors, Antineoplastic Agents pharmacology
- Abstract
Purpose: We hypothesized that inhibition and trapping of PARP1 alone would be sufficient to achieve antitumor activity. In particular, we aimed to achieve selectivity over PARP2, which has been shown to play a role in the survival of hematopoietic/stem progenitor cells in animal models. We developed AZD5305 with the aim of achieving improved clinical efficacy and wider therapeutic window. This next-generation PARP inhibitor (PARPi) could provide a paradigm shift in clinical outcomes achieved by first-generation PARPi, particularly in combination., Experimental Design: AZD5305 was tested in vitro for PARylation inhibition, PARP-DNA trapping, and antiproliferative abilities. In vivo efficacy was determined in mouse xenograft and PDX models. The potential for hematologic toxicity was evaluated in rat models, as monotherapy and combination., Results: AZD5305 is a highly potent and selective inhibitor of PARP1 with 500-fold selectivity for PARP1 over PARP2. AZD5305 inhibits growth in cells with deficiencies in DNA repair, with minimal/no effects in other cells. Unlike first-generation PARPi, AZD5305 has minimal effects on hematologic parameters in a rat pre-clinical model at predicted clinically efficacious exposures. Animal models treated with AZD5305 at doses ≥0.1 mg/kg once daily achieved greater depth of tumor regression compared to olaparib 100 mg/kg once daily, and longer duration of response., Conclusions: AZD5305 potently and selectively inhibits PARP1 resulting in excellent antiproliferative activity and unprecedented selectivity for DNA repair deficient versus proficient cells. These data confirm the hypothesis that targeting only PARP1 can retain the therapeutic benefit of nonselective PARPi, while reducing potential for hematotoxicity. AZD5305 is currently in phase I trials (NCT04644068)., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
17. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer.
- Author
-
Martins FC, Couturier DL, de Santiago I, Sauer CM, Vias M, Angelova M, Sanders D, Piskorz A, Hall J, Hosking K, Amirthanayagam A, Cosulich S, Carnevalli L, Davies B, Watkins TBK, Funingana IG, Bolton H, Haldar K, Latimer J, Baldwin P, Crawford R, Eldridge M, Basu B, Jimenez-Linan M, Mcpherson AW, McGranahan N, Litchfield K, Shah SP, McNeish I, Caldas C, Evan G, Swanton C, and Brenton JD
- Subjects
- Humans, Female, DNA Copy Number Variations, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins p21(ras) genetics, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Paclitaxel therapeutic use, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous metabolism
- Abstract
Chromosomal instability is a major challenge to patient stratification and targeted drug development for high-grade serous ovarian carcinoma (HGSOC). Here we show that somatic copy number alterations (SCNAs) in frequently amplified HGSOC cancer genes significantly correlate with gene expression and methylation status. We identify five prevalent clonal driver SCNAs (chromosomal amplifications encompassing MYC, PIK3CA, CCNE1, KRAS and TERT) from multi-regional HGSOC data and reason that their strong selection should prioritise them as key biomarkers for targeted therapies. We use primary HGSOC spheroid models to test interactions between in vitro targeted therapy and SCNAs. MYC chromosomal copy number is associated with in-vitro and clinical response to paclitaxel and in-vitro response to mTORC1/2 inhibition. Activation of the mTOR survival pathway in the context of MYC-amplified HGSOC is statistically associated with increased prevalence of SCNAs in genes from the PI3K pathway. Co-occurrence of amplifications in MYC and genes from the PI3K pathway is independently observed in squamous lung cancer and triple negative breast cancer. In this work, we show that identifying co-occurrence of clonal driver SCNA genes could be used to tailor therapeutics for precision medicine., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. AZD4625 is a Potent and Selective Inhibitor of KRASG12C.
- Author
-
Chakraborty A, Hanson L, Robinson D, Lewis H, Bickerton S, Davies M, Polanski R, Whiteley R, Koers A, Atkinson J, Baker T, Del Barco Barrantes I, Ciotta G, Kettle JG, Magiera L, Martins CP, Peter A, Wigmore E, Underwood Z, Cosulich S, Niedbala M, and Ross S
- Subjects
- Cell Line, Tumor, Glycine pharmacology, Humans, Mutation, Protein Isoforms, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cysteine
- Abstract
AZD4625 is a potent, selective, and orally bioavailable inhibitor of oncogenic KRASG12C as demonstrated in cellular assays and in vivo in preclinical cell line-derived and patient-derived xenograft models. In vitro and cellular assays have shown selective binding and inhibition of the KRASG12C mutant isoform, which carries a glycine to cysteine mutation at residue 12, with no binding and inhibition of wild-type RAS or isoforms carrying non-KRASG12C mutations. The pharmacology of AZD4625 shows that it has the potential to provide therapeutic benefit to patients with KRASG12C mutant cancer as either a monotherapy treatment or in combination with other targeted drug agents., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
19. Acyl chain selection couples the consumption and synthesis of phosphoinositides.
- Author
-
Barneda D, Janardan V, Niewczas I, Collins DM, Cosulich S, Clark J, Stephens LR, and Hawkins PT
- Subjects
- Animals, Glucose, Mammals, Lipogenesis, Phosphatidylinositols metabolism
- Abstract
Phosphoinositides (PIPn) in mammalian tissues are enriched in the stearoyl/arachidonoyl acyl chain species ("C38:4"), but its functional significance is unclear. We have used metabolic tracers (isotopologues of inositol, glucose and water) to study PIPn synthesis in cell lines in which this enrichment is preserved to differing relative extents. We show that PIs synthesised from glucose are initially enriched in shorter/more saturated acyl chains, but then rapidly remodelled towards the C38:4 species. PIs are also synthesised by a distinct 're-cycling pathway', which utilises existing precursors and exhibits substantial selectivity for the synthesis of C38:4-PA and -PI. This re-cycling pathway is rapidly stimulated during receptor activation of phospholipase-C, both allowing the retention of the C38:4 backbone and the close coupling of PIPn consumption to its resynthesis, thus maintaining pool sizes. These results suggest that one property of the specific acyl chain composition of PIPn is that of a molecular code, to facilitate 'metabolic channelling' from PIP2 to PI via pools of intermediates (DG, PA and CDP-DG) common to other lipid metabolic pathways., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
20. Discovery of AZD4625, a Covalent Allosteric Inhibitor of the Mutant GTPase KRAS G12C .
- Author
-
Kettle JG, Bagal SK, Bickerton S, Bodnarchuk MS, Boyd S, Breed J, Carbajo RJ, Cassar DJ, Chakraborty A, Cosulich S, Cumming I, Davies M, Davies NL, Eatherton A, Evans L, Feron L, Fillery S, Gleave ES, Goldberg FW, Hanson L, Harlfinger S, Howard M, Howells R, Jackson A, Kemmitt P, Lamont G, Lamont S, Lewis HJ, Liu L, Niedbala MJ, Phillips C, Polanski R, Raubo P, Robb G, Robinson DM, Ross S, Sanders MG, Tonge M, Whiteley R, Wilkinson S, Yang J, and Zhang W
- Subjects
- Drug Design, Humans, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Quinazolines pharmacology, Structure-Activity Relationship, Antineoplastic Agents pharmacology, Lung Neoplasms drug therapy
- Abstract
KRAS is an archetypal high-value intractable oncology drug target. The glycine to cysteine mutation at codon 12 represents an Achilles heel that has now rendered this important GTPase druggable. Herein, we report our structure-based drug design approach that led to the identification of 21 , AZD4625, a clinical development candidate for the treatment of KRAS
G12C positive tumors. Highlights include a quinazoline tethering strategy to lock out a bio-relevant binding conformation and an optimization strategy focused on the reduction of extrahepatic clearance mechanisms seen in preclinical species. Crystallographic analysis was also key in helping to rationalize unusual structure-activity relationship in terms of ring size and enantio-preference. AZD4625 is a highly potent and selective inhibitor of KRASG12C with an anticipated low clearance and high oral bioavailability profile in humans.- Published
- 2022
- Full Text
- View/download PDF
21. Identification and optimization of a novel series of selective PIP5K inhibitors.
- Author
-
Andrews DM, Cartic S, Cosulich S, Divecha N, Faulder P, Flemington V, Kern O, Kettle JG, MacDonald E, McKelvie J, Pike KG, Roberts B, Rowlinson R, Smith JM, Stockley M, Swarbrick ME, Treinies I, and Waring MJ
- Subjects
- Amides chemistry, Amides metabolism, Animals, Caco-2 Cells, Dose-Response Relationship, Drug, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Humans, Microsomes, Liver chemistry, Microsomes, Liver metabolism, Molecular Structure, Phosphotransferases (Alcohol Group Acceptor) metabolism, Rats, Structure-Activity Relationship, Amides pharmacology, Enzyme Inhibitors pharmacology, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors
- Abstract
Phosphatidyl inositol (4,5)-bisphosphate (PI(4,5)P
2 ) plays several key roles in human biology and the lipid kinase that produces PI(4,5)P2 , PIP5K, has been hypothesized to provide a potential therapeutic target of interest in the treatment of cancers. To better understand and explore the role of PIP5K in human cancers there remains an urgent need for potent and specific PIP5K inhibitor molecules. Following a high throughput screen of the AstraZeneca collection, a novel, moderately potent and selective inhibitor of PIP5K, 1, was discovered. Detailed exploration of the SAR for this novel scaffold resulted in the considerable optimization of both potency for PIP5K, and selectivity over the closely related kinase PI3Kα, as well as identifying several opportunities for the continued optimization of drug-like properties. As a result, several high quality in vitro tool compounds were identified (8, 20 and 25) that demonstrate the desired biochemical and cellular profiles required to aid better understanding of this complex area of biology., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
22. Discovery of 5-{4-[(7-Ethyl-6-oxo-5,6-dihydro-1,5-naphthyridin-3-yl)methyl]piperazin-1-yl}- N -methylpyridine-2-carboxamide (AZD5305): A PARP1-DNA Trapper with High Selectivity for PARP1 over PARP2 and Other PARPs.
- Author
-
Johannes JW, Balazs A, Barratt D, Bista M, Chuba MD, Cosulich S, Critchlow SE, Degorce SL, Di Fruscia P, Edmondson SD, Embrey K, Fawell S, Ghosh A, Gill SJ, Gunnarsson A, Hande SM, Heightman TD, Hemsley P, Illuzzi G, Lane J, Larner C, Leo E, Liu L, Madin A, Martin S, McWilliams L, O'Connor MJ, Orme JP, Pachl F, Packer MJ, Pei X, Pike A, Schimpl M, She H, Staniszewska AD, Talbot V, Underwood E, Varnes JG, Xue L, Yao T, Zhang K, Zhang AX, and Zheng X
- Subjects
- Humans, Crystallography, X-Ray, Substrate Specificity, DNA chemistry, Poly (ADP-Ribose) Polymerase-1 antagonists & inhibitors, Poly(ADP-ribose) Polymerase Inhibitors chemistry, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerases metabolism
- Abstract
Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1-DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.
- Published
- 2021
- Full Text
- View/download PDF
23. Genome-Wide Estrogen Receptor Activity in Breast Cancer.
- Author
-
Farcas AM, Nagarajan S, Cosulich S, and Carroll JS
- Subjects
- Breast Neoplasms genetics, Disease Progression, Enhancer Elements, Genetic, Humans, Transcription Factors metabolism, Breast Neoplasms metabolism, Gene Expression Regulation, Neoplastic, Receptors, Estrogen metabolism
- Abstract
The largest subtype of breast cancer is characterized by the expression and activity of the estrogen receptor alpha (ERalpha/ER). Although several effective therapies have significantly improved survival, the adaptability of cancer cells means that patients frequently stop responding or develop resistance to endocrine treatment. ER does not function in isolation and multiple associating factors have been reported to play a role in regulating the estrogen-driven transcriptional program. This review focuses on the dynamic interplay between some of these factors which co-occupy ER-bound regulatory elements, their contribution to estrogen signaling, and their possible therapeutic applications. Furthermore, the review illustrates how some ER association partners can influence and reprogram the genomic distribution of the estrogen receptor. As this dynamic ER activity enables cancer cell adaptability and impacts the clinical outcome, defining how this plasticity is determined is fundamental to our understanding of the mechanisms of disease progression., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2021
- Full Text
- View/download PDF
24. Structure-Based Design and Pharmacokinetic Optimization of Covalent Allosteric Inhibitors of the Mutant GTPase KRAS G12C .
- Author
-
Kettle JG, Bagal SK, Bickerton S, Bodnarchuk MS, Breed J, Carbajo RJ, Cassar DJ, Chakraborty A, Cosulich S, Cumming I, Davies M, Eatherton A, Evans L, Feron L, Fillery S, Gleave ES, Goldberg FW, Harlfinger S, Hanson L, Howard M, Howells R, Jackson A, Kemmitt P, Kingston JK, Lamont S, Lewis HJ, Li S, Liu L, Ogg D, Phillips C, Polanski R, Robb G, Robinson D, Ross S, Smith JM, Tonge M, Whiteley R, Yang J, Zhang L, and Zhao X
- Subjects
- Allosteric Regulation, Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacokinetics, Caco-2 Cells, Cell Line, Tumor, Drug Design, Humans, Male, Mice, Nude, Molecular Conformation, Mutation, Piperazines chemical synthesis, Piperazines pharmacokinetics, Proto-Oncogene Proteins p21(ras) genetics, Quinazolines chemical synthesis, Quinazolines pharmacokinetics, Quinolones chemical synthesis, Quinolones pharmacokinetics, Rats, Wistar, Structure-Activity Relationship, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Piperazines therapeutic use, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors, Quinazolines therapeutic use, Quinolones therapeutic use
- Abstract
Attempts to directly drug the important oncogene KRAS have met with limited success despite numerous efforts across industry and academia. The KRAS
G12C mutant represents an "Achilles heel" and has recently yielded to covalent targeting with small molecules that bind the mutant cysteine and create an allosteric pocket on GDP-bound RAS, locking it in an inactive state. A weak inhibitor at this site was optimized through conformational locking of a piperazine-quinazoline motif and linker modification. Subsequent introduction of a key methyl group to the piperazine resulted in enhancements in potency, permeability, clearance, and reactivity, leading to identification of a potent KRASG12C inhibitor with high selectivity and excellent cross-species pharmacokinetic parameters and in vivo efficacy.- Published
- 2020
- Full Text
- View/download PDF
25. Discovery and pharmacological characterization of AZD3229, a potent KIT/PDGFRα inhibitor for treatment of gastrointestinal stromal tumors.
- Author
-
Banks E, Grondine M, Bhavsar D, Barry E, Kettle JG, Reddy VP, Brown C, Wang H, Mettetal JT, Collins T, Adeyemi O, Overman R, Lawson D, Harmer AR, Reimer C, Drew L, Packer MJ, Cosulich S, Jones RD, Shao W, Wilson D, Guichard S, Fawell S, and Anjum R
- Subjects
- Animals, Drug Resistance, Neoplasm, Humans, Mutation, Naphthyridines, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-kit genetics, Pyrazoles, Pyrroles, Rats, Receptor, Platelet-Derived Growth Factor alpha genetics, Triazines, Urea analogs & derivatives, Vascular Endothelial Growth Factor A, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Stromal Tumors genetics
- Abstract
Gastrointestinal stromal tumor (GIST) is the most common human sarcoma driven by mutations in KIT or platelet-derived growth factor α ( PDGFR α). Although first-line treatment, imatinib, has revolutionized GIST treatment, drug resistance due to acquisition of secondary KIT / PDGFR α mutations develops in a majority of patients. Second- and third-line treatments, sunitinib and regorafenib, lack activity against a plethora of mutations in KIT/PDGFRα in GIST, with median time to disease progression of 4 to 6 months and inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) causing high-grade hypertension. Patients with GIST have an unmet need for a well-tolerated drug that robustly inhibits a range of KIT/PDGFRα mutations. Here, we report the discovery and pharmacological characterization of AZD3229, a potent and selective small-molecule inhibitor of KIT and PDGFRα designed to inhibit a broad range of primary and imatinib-resistant secondary mutations seen in GIST. In engineered and GIST-derived cell lines, AZD3229 is 15 to 60 times more potent than imatinib in inhibiting KIT primary mutations and has low nanomolar activity against a wide spectrum of secondary mutations. AZD3229 causes durable inhibition of KIT signaling in patient-derived xenograft (PDX) models of GIST, leading to tumor regressions at doses that showed no changes in arterial blood pressure (BP) in rat telemetry studies. AZD3229 has a superior potency and selectivity profile to standard of care (SoC) agents-imatinib, sunitinib, and regorafenib, as well as investigational agents, avapritinib (BLU-285) and ripretinib (DCC-2618). AZD3229 has the potential to be a best-in-class inhibitor for clinically relevant KIT/PDGFRα mutations in GIST., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
26. How is the acyl chain composition of phosphoinositides created and does it matter?
- Author
-
Barneda D, Cosulich S, Stephens L, and Hawkins P
- Subjects
- 1-Phosphatidylinositol 4-Kinase metabolism, Animals, Binding Sites, Signal Transduction, Phosphatidylinositols metabolism
- Abstract
The phosphoinositide (PIPn) family of signalling phospholipids are central regulators in membrane cell biology. Their varied functions are based on the phosphorylation pattern of their inositol ring, which can be recognized by selective binding domains in their effector proteins and be modified by a series of specific PIPn kinases and phosphatases, which control their interconversion in a spatial and temporal manner. Yet, a unique feature of PIPns remains largely unexplored: their unusually uniform acyl chain composition. Indeed, while most phospholipids present a range of molecular species comprising acyl chains of diverse length and saturation, PIPns in several organisms and tissues show the predominance of a single hydrophobic backbone, which in mammals is composed of arachidonoyl and stearoyl chains. Despite evolution having favoured this specific PIPn configuration, little is known regarding the mechanisms and functions behind it. In this review, we explore the metabolic pathways that could control the acyl chain composition of PIPns as well as the potential roles of this selective enrichment. While our understanding of this phenomenon has been constrained largely by the technical limitations in the methods traditionally employed in the PIPn field, we believe that the latest developments in PIPn analysis should shed light onto this old question., (© 2019 The Author(s).)
- Published
- 2019
- Full Text
- View/download PDF
27. Combination of dual mTORC1/2 inhibition and immune-checkpoint blockade potentiates anti-tumour immunity.
- Author
-
Langdon S, Hughes A, Taylor MA, Kuczynski EA, Mele DA, Delpuech O, Jarvis L, Staniszewska A, Cosulich S, Carnevalli LS, and Sinclair C
- Abstract
mTOR inhibition can promote or inhibit immune responses in a context dependent manner, but whether this will represent a net benefit or be contraindicated in the context of immunooncology therapies is less understood. Here, we report that the mTORC1/2 dual kinase inhibitor vistusertib (AZD2014) potentiates anti-tumour immunity in combination with anti-CTLA-4 (αCTLA-4), αPD-1 or αPD-L1 immune checkpoint blockade. Combination of vistusertib and immune checkpoint blocking antibodies led to tumour growth inhibition and improved survival of MC-38 or CT-26 pre-clinical syngeneic tumour models, whereas monotherapies were less effective. Underlying these combinatorial effects, vistusertib/immune checkpoint combinations reduced the occurrence of exhausted phenotype tumour infiltrating lymphocytes (TILs), whilst increasing frequencies of activated Th1 polarized T-cells in tumours. Vistusertib alone was shown to promote a Th1 polarizing proinflammatory cytokine profile by innate primary immune cells. Moreover, vistusertib directly enhanced activation of effector T-cell and survival, an effect that was critically dependent on inhibitor dose. Therefore, these data highlight direct, tumour-relevant immune potentiating benefits of mTOR inhibition that complement immune checkpoint blockade. Together, these data provide a clear rationale to investigate such combinations in the clinic.
- Published
- 2018
- Full Text
- View/download PDF
28. PTEN Regulates PI(3,4)P 2 Signaling Downstream of Class I PI3K.
- Author
-
Malek M, Kielkowska A, Chessa T, Anderson KE, Barneda D, Pir P, Nakanishi H, Eguchi S, Koizumi A, Sasaki J, Juvin V, Kiselev VY, Niewczas I, Gray A, Valayer A, Spensberger D, Imbert M, Felisbino S, Habuchi T, Beinke S, Cosulich S, Le Novère N, Sasaki T, Clark J, Hawkins PT, and Stephens LR
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Epidermal Growth Factor pharmacology, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Mutation, PTEN Phosphohydrolase deficiency, PTEN Phosphohydrolase genetics, Phenotype, Phosphoric Monoester Hydrolases genetics, Phosphoric Monoester Hydrolases metabolism, Phosphorylation, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Time Factors, Breast Neoplasms enzymology, Class I Phosphatidylinositol 3-Kinases metabolism, PTEN Phosphohydrolase metabolism, Phosphatidylinositols metabolism, Prostatic Neoplasms enzymology, Second Messenger Systems drug effects
- Abstract
The PI3K signaling pathway regulates cell growth and movement and is heavily mutated in cancer. Class I PI3Ks synthesize the lipid messenger PI(3,4,5)P
3 . PI(3,4,5)P3 can be dephosphorylated by 3- or 5-phosphatases, the latter producing PI(3,4)P2 . The PTEN tumor suppressor is thought to function primarily as a PI(3,4,5)P3 3-phosphatase, limiting activation of this pathway. Here we show that PTEN also functions as a PI(3,4)P2 3-phosphatase, both in vitro and in vivo. PTEN is a major PI(3,4)P2 phosphatase in Mcf10a cytosol, and loss of PTEN and INPP4B, a known PI(3,4)P2 4-phosphatase, leads to synergistic accumulation of PI(3,4)P2 , which correlated with increased invadopodia in epidermal growth factor (EGF)-stimulated cells. PTEN deletion increased PI(3,4)P2 levels in a mouse model of prostate cancer, and it inversely correlated with PI(3,4)P2 levels across several EGF-stimulated prostate and breast cancer lines. These results point to a role for PI(3,4)P2 in the phenotype caused by loss-of-function mutations or deletions in PTEN., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
29. Discovery of a novel aminopyrazine series as selective PI3Kα inhibitors.
- Author
-
Barlaam B, Cosulich S, Fitzek M, Germain H, Green S, Hanson LL, Harris CS, Hancox U, Hudson K, Lambert-van der Brempt C, Lamorlette M, Magnien F, Ouvry G, Page K, Ruston L, Ward L, and Delouvrié B
- Subjects
- Class I Phosphatidylinositol 3-Kinases, Dose-Response Relationship, Drug, Humans, Molecular Docking Simulation, Molecular Structure, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Pyrazines chemical synthesis, Pyrazines chemistry, Structure-Activity Relationship, Drug Discovery, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology, Pyrazines pharmacology
- Abstract
We report the discovery of a novel aminopyrazine series of PI3Kα inhibitors, designed by hybridizing two known scaffolds of PI3K inhibitors. We describe the progress achieved from the first compounds plagued with poor general kinase selectivity to compounds showing high selectivity for PI3Kα over PI3Kβ and excellent general kinase selectivity. This effort culminated with the identification of compound 5 displaying high potency and selectivity, and suitable physiochemical and pharmacokinetic properties for oral administration. In vivo, compound 5 showed good inhibition of tumour growth (86% tumour growth inhibition at 50mg/kg twice daily orally) in the MCF7 xenograft model in mice., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Discovery of a series of 8-(1-phenylpyrrolidin-2-yl)-6-carboxamide-2-morpholino-4H-chromen-4-one as PI3Kβ/δ inhibitors for the treatment of PTEN-deficient tumours.
- Author
-
Barlaam B, Cosulich S, Degorce S, Ellston R, Fitzek M, Green S, Hancox U, Lambert-van der Brempt C, Lohmann JJ, Maudet M, Morgentin R, Plé P, Ward L, and Warin N
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Benzopyrans pharmacokinetics, Benzopyrans pharmacology, Cell Line, Tumor, Class Ia Phosphatidylinositol 3-Kinase metabolism, Dogs, Gene Deletion, Humans, Male, Mice, Nude, Molecular Docking Simulation, Morpholinos chemistry, Morpholinos pharmacokinetics, Morpholinos pharmacology, Morpholinos therapeutic use, Prostate drug effects, Prostate metabolism, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Benzopyrans chemistry, Benzopyrans therapeutic use, PTEN Phosphohydrolase genetics, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms drug therapy
- Abstract
Attempts to lock the active conformation of compound 4, a PI3Kβ/δ inhibitor (PI3Kβ cell IC
50 0.015μM), led to the discovery of a series of 8-(1-phenylpyrrolidin-2-yl)-6-carboxamide-2-morpholino-4H-chromen-4-ones, which showed high levels of potency and selectivity as PI3Kβ/δ inhibitors. Compound 10 proved exquisitely potent and selective: PI3Kβ cell IC50 0.0011μM in PTEN null MDA-MB-468 cell and PI3Kδ cell IC50 0.014μM in Jeko-1 B-cell, and exhibited suitable physical properties for oral administration. In vivo, compound 10 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-null PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Based on these results, compound 10 was selected as one of our PI3Kβ/δ preclinical candidates., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
31. Targeting mTOR pathway inhibits tumor growth in different molecular subtypes of triple-negative breast cancers.
- Author
-
Hatem R, El Botty R, Chateau-Joubert S, Servely JL, Labiod D, de Plater L, Assayag F, Coussy F, Callens C, Vacher S, Reyal F, Cosulich S, Diéras V, Bièche I, and Marangoni E
- Subjects
- Animals, Cell Growth Processes drug effects, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Mice, Mice, Nude, Molecular Targeted Therapy, Signal Transduction drug effects, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Xenograft Model Antitumor Assays, Everolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, Triple Negative Breast Neoplasms drug therapy
- Abstract
Triple-negative breast cancers (TNBC) are characterized by frequent alterations in the PI3K/AKT/mTOR signaling pathway. In this study, we analyzed PI3K pathway activation in 67 patient-derived xenografts (PDX) of breast cancer and investigated the anti-tumor activity of the mTOR inhibitor everolimus in 15 TNBC PDX with different expression and mutational status of PI3K pathway markers. Expression of the tumor suppressors PTEN and INPP4B was lost in 55% and 76% of TNBC PDX, respectively, while mutations in PIK3CA and AKT1 genes were rare. In 7 PDX treatment with everolimus resulted in a tumor growth inhibition higher than 50%, while 8 models were classified as low responder or resistant. Basal-like, LAR (Luminal AR), mesenchymal and HER2-enriched tumors were present in both responder and resistant groups, suggesting that tumor response to everolimus is not restricted to a specific TNBC subtype. Analysis of treated tumors showed a correlation between tumor response and post-treatment phosphorylation of AKT, increased in responder PDX, while PI3K pathway markers at baseline were not sufficient to predict everolimus response. In conclusion, targeting mTOR decreased tumor growth in 7 out of 15 TNBC PDX tested. Response to everolimus occurred in different TNBC subtypes and was associated with post-treatment increase of P-AKT., Competing Interests: The authors declare that they have no competing interests.
- Published
- 2016
- Full Text
- View/download PDF
32. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor.
- Author
-
Rodrik-Outmezguine VS, Okaniwa M, Yao Z, Novotny CJ, McWhirter C, Banaji A, Won H, Wong W, Berger M, de Stanchina E, Barratt DG, Cosulich S, Klinowska T, Rosen N, and Shokat KM
- Subjects
- Animals, Binding Sites drug effects, Cell Line, Tumor, Female, Humans, Mice, Mutation drug effects, Neoplasms drug therapy, Neoplasms enzymology, Neoplasms genetics, Neoplasms pathology, Protein Kinase Inhibitors classification, Protein Structure, Tertiary genetics, Signal Transduction drug effects, TOR Serine-Threonine Kinases chemistry, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, Drug Resistance drug effects, Drug Resistance genetics, Mutation genetics, Protein Kinase Inhibitors pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics
- Abstract
Precision medicines exert selective pressure on tumour cells that leads to the preferential growth of resistant subpopulations, necessitating the development of next-generation therapies to treat the evolving cancer. The PIK3CA-AKT-mTOR pathway is one of the most commonly activated pathways in human cancers, which has led to the development of small-molecule inhibitors that target various nodes in the pathway. Among these agents, first-generation mTOR inhibitors (rapalogs) have caused responses in 'N-of-1' cases, and second-generation mTOR kinase inhibitors (TORKi) are currently in clinical trials. Here we sought to delineate the likely resistance mechanisms to existing mTOR inhibitors in human cell lines, as a guide for next-generation therapies. The mechanism of resistance to the TORKi was unusual in that intrinsic kinase activity of mTOR was increased, rather than a direct active-site mutation interfering with drug binding. Indeed, identical drug-resistant mutations have been also identified in drug-naive patients, suggesting that tumours with activating MTOR mutations will be intrinsically resistant to second-generation mTOR inhibitors. We report the development of a new class of mTOR inhibitors that overcomes resistance to existing first- and second-generation inhibitors. The third-generation mTOR inhibitor exploits the unique juxtaposition of two drug-binding pockets to create a bivalent interaction that allows inhibition of these resistant mutants.
- Published
- 2016
- Full Text
- View/download PDF
33. Discovery of a series of 8-(2,3-dihydro-1,4-benzoxazin-4-ylmethyl)-2-morpholino-4-oxo-chromene-6-carboxamides as PI3Kβ/δ inhibitors for the treatment of PTEN-deficient tumours.
- Author
-
Barlaam B, Cosulich S, Degorce S, Fitzek M, Green S, Hancox U, Lambert-van der Brempt C, Lohmann JJ, Maudet M, Morgentin R, Péru A, Plé P, Saleh T, Ward L, and Warin N
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Phosphorylation, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Isoenzymes antagonists & inhibitors, Morpholinos chemistry, Morpholinos pharmacology, PTEN Phosphohydrolase genetics, Phosphoinositide-3 Kinase Inhibitors
- Abstract
We report the discovery and optimisation of a series of 8-(2,3-dihydro-1,4-benzoxazin-4-ylmethyl)-2-morpholino-4-oxo-chromene-6-carboxamides, leading to compound 16 as a potent and selective PI3Kβ/δ inhibitor: PI3Kβ cell IC50 0.012 μM (in PTEN null MDA-MB-468 cell) and PI3Kδ cell IC50 0.047 μM (in Jeko-1 B-cell), with good pharmacokinetics and physical properties. In vivo, 16 showed profound pharmacodynamic modulation of AKT phosphorylation in a mouse PTEN-deficient PC3 prostate tumour xenograft after a single oral dose and gave excellent tumour growth inhibition in the same model after chronic oral dosing. Compound 16 was selected as a preclinical candidate for the treatment of PTEN-deficient tumours., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. Discovery of 1-(4-(5-(5-amino-6-(5-tert-butyl-1,3,4-oxadiazol-2-yl)pyrazin-2-yl)-1-ethyl-1,2,4-triazol-3-yl)piperidin-1-yl)-3-hydroxypropan-1-one (AZD8835): A potent and selective inhibitor of PI3Kα and PI3Kδ for the treatment of cancers.
- Author
-
Barlaam B, Cosulich S, Delouvrié B, Ellston R, Fitzek M, Germain H, Green S, Hancox U, Harris CS, Hudson K, Lambert-van der Brempt C, Lebraud H, Magnien F, Lamorlette M, Le Griffon A, Morgentin R, Ouvry G, Page K, Pasquet G, Polanska U, Ruston L, Saleh T, Vautier M, and Ward L
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacokinetics, Cell Line, Tumor, Dogs, Humans, Mice, Mice, Nude, Mice, SCID, Molecular Docking Simulation, Oxadiazoles chemical synthesis, Piperidines chemical synthesis, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors pharmacokinetics, Rats, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Oxadiazoles pharmacology, Phosphoinositide-3 Kinase Inhibitors, Piperidines pharmacology, Protein Kinase Inhibitors pharmacology
- Abstract
Starting from potent inhibitors of PI3Kα having poor general kinase selectivity (e.g., 1 and 2), optimisation of this series led to the identification of 25, a potent inhibitor of PI3Kα (wild type, E545K and H1047R mutations) and PI3Kδ, selective versus PI3Kβ and PI3Kγ, with excellent general kinase selectivity. Compound 25 displayed low metabolic turnover and suitable physical properties for oral administration. In vivo, compound 25 showed pharmacodynamic modulation of AKT phosphorylation and near complete inhibition of tumour growth (93% tumour growth inhibition) in a murine H1047R PI3Kα mutated SKOV-3 xenograft tumour model after chronic oral administration at 25mg/kg b.i.d. Compound 25, also known as AZD8835, is currently in phase I clinical trials., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Design of selective PI3Kα inhibitors starting from a promiscuous pan kinase scaffold.
- Author
-
Barlaam B, Cosulich S, Fitzek M, Green S, Harris CS, Hudson K, Lambert-van der Brempt C, Ouvry G, Page K, Ruston L, Ward L, and Delouvrié B
- Subjects
- Amino Acid Sequence, Binding Sites, Biomarkers, Tumor antagonists & inhibitors, Biomarkers, Tumor chemistry, Biomarkers, Tumor genetics, Cell Line, Tumor, Class I Phosphatidylinositol 3-Kinases, Drug Design, Enzyme Inhibitors chemical synthesis, Humans, Models, Molecular, Mutant Proteins antagonists & inhibitors, Mutant Proteins chemistry, Mutant Proteins genetics, Phosphatidylinositol 3-Kinases chemistry, Phosphatidylinositol 3-Kinases genetics, Pyrazoles chemical synthesis, Pyrazoles chemistry, Pyrazoles pharmacology, Structure-Activity Relationship, Triazoles chemical synthesis, Triazoles chemistry, Triazoles pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Phosphoinositide-3 Kinase Inhibitors
- Abstract
Starting from compound 1, a potent PI3Kα inhibitor having poor general kinase selectivity, we used structural data and modelling to identify key exploitable differences between PI3Kα and the other kinases. This approach led us to design chemical modifications of the central pyrazole, which solved the poor kinase selectivity seen as a strong liability for the initial compound 1. Amongst the modifications explored, a 1,3,4-triazole ring (as in compound 4) as a replacement of the initial pyrazole provided good potency against PI3Kα, with excellent kinase selectivity., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Novel Role for p110β PI 3-Kinase in Male Fertility through Regulation of Androgen Receptor Activity in Sertoli Cells.
- Author
-
Guillermet-Guibert J, Smith LB, Halet G, Whitehead MA, Pearce W, Rebourcet D, León K, Crépieux P, Nock G, Strömstedt M, Enerback M, Chelala C, Graupera M, Carroll J, Cosulich S, Saunders PT, Huhtaniemi I, and Vanhaesebroeck B
- Subjects
- Animals, Blastocyst cytology, Cells, Cultured, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Class I Phosphatidylinositol 3-Kinases genetics, Female, Homeodomain Proteins genetics, Infertility, Female genetics, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Morula cytology, Receptors, Androgen genetics, Signal Transduction genetics, Spermatogenesis genetics, Transcription Factors genetics, Transcription, Genetic genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Fertility physiology, Infertility, Male genetics, Receptors, Androgen metabolism, Sertoli Cells metabolism
- Abstract
The organismal roles of the ubiquitously expressed class I PI3K isoform p110β remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110β, we document that full inactivation of p110β leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110β kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110β results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110β was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110β also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110β inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110β inactivation. In line with a crucial role for p110β in SCs, selective inactivation of p110β in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110β and AR have previously been reported to functionally interact.
- Published
- 2015
- Full Text
- View/download PDF
37. Discovery of (R)-8-(1-(3,5-difluorophenylamino)ethyl)-N,N-dimethyl-2-morpholino-4-oxo-4H-chromene-6-carboxamide (AZD8186): a potent and selective inhibitor of PI3Kβ and PI3Kδ for the treatment of PTEN-deficient cancers.
- Author
-
Barlaam B, Cosulich S, Degorce S, Fitzek M, Green S, Hancox U, Lambert-van der Brempt C, Lohmann JJ, Maudet M, Morgentin R, Pasquet MJ, Péru A, Plé P, Saleh T, Vautier M, Walker M, Ward L, and Warin N
- Subjects
- Aniline Compounds pharmacology, Animals, Chromones pharmacology, Dogs, Drug Discovery, Humans, Male, Mice, Neoplasms, Experimental chemistry, Structure-Activity Relationship, Aniline Compounds chemical synthesis, Chromones chemical synthesis, Neoplasms, Experimental drug therapy, PTEN Phosphohydrolase deficiency, Phosphoinositide-3 Kinase Inhibitors
- Abstract
Several studies have highlighted the dependency of PTEN deficient tumors to PI3Kβ activity and specific inhibition of PI3Kδ has been shown activity against human B-cell cancers. We describe the discovery and optimization of a series of 8-(1-anilino)ethyl)-2-morpholino-4-oxo-4H-chromene-6-carboxamides as PI3Kβ/δ inhibitors, which led to the discovery of the clinical candidate 13, also known as AZD8186. On the basis of the lower lipophilicity of the chromen-4-one core compared to the previously utilized pyrido[1,2-a]pyrimid-4-one core, this series of compounds displayed high metabolic stability and suitable physical properties for oral administration. Compound 13 showed profound pharmacodynamic modulation of p-Akt in PTEN-deficient PC3 prostate tumor bearing mice after oral administration and showed complete inhibition of tumor growth in the mouse PTEN-deficient PC3 prostate tumor xenograft model. 13 was selected as a clinical candidate for treatment of PTEN-deficient cancers and has recently entered phase I clinical trials.
- Published
- 2015
- Full Text
- View/download PDF
38. Inhibition of PI3Kβ signaling with AZD8186 inhibits growth of PTEN-deficient breast and prostate tumors alone and in combination with docetaxel.
- Author
-
Hancox U, Cosulich S, Hanson L, Trigwell C, Lenaghan C, Ellston R, Dry H, Crafter C, Barlaam B, Fitzek M, Smith PD, Ogilvie D, D'Cruz C, Castriotta L, Wedge SR, Ward L, Powell S, Lawson M, Davies BR, Harrington EA, Foster E, Cumberbatch M, Green S, and Barry ST
- Subjects
- Aniline Compounds pharmacology, Animals, Antineoplastic Agents pharmacology, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cell Line, Tumor, Cell Proliferation drug effects, Chromones pharmacology, Docetaxel, Drug Synergism, Female, Humans, Male, Mice, PTEN Phosphohydrolase deficiency, Prostatic Neoplasms metabolism, Triple Negative Breast Neoplasms metabolism, Xenograft Model Antitumor Assays, Aniline Compounds administration & dosage, Antineoplastic Agents administration & dosage, Chromones administration & dosage, Phosphoinositide-3 Kinase Inhibitors, Prostatic Neoplasms drug therapy, Signal Transduction drug effects, Taxoids administration & dosage, Triple Negative Breast Neoplasms drug therapy
- Abstract
Loss of PTEN protein results in upregulation of the PI3K/AKT pathway, which appears dependent on the PI3Kβ isoform. Inhibitors of PI3Kβ have potential to reduce growth of tumors in which loss of PTEN drives tumor progression. We have developed a small-molecule inhibitor of PI3Kβ and PI3Kδ (AZD8186) and assessed its antitumor activity across a panel of cell lines. We have then explored the antitumor effects as single agent and in combination with docetaxel in triple-negative breast (TNBC) and prostate cancer models. In vitro, AZD8186 inhibited growth of a range of cell lines. Sensitivity was associated with inhibition of the AKT pathway. Cells sensitive to AZD8186 (GI50 < 1 μmol/L) are enriched for, but not exclusively associated with, PTEN deficiency. In vivo, AZD8186 inhibits PI3K pathway biomarkers in prostate and TNBC tumors. Scheduling treatment with AZD8186 shows antitumor activity required only intermittent exposure, and that increased tumor control is achieved when AZD8186 is used in combination with docetaxel. AZD8186 is a potent inhibitor of PI3Kβ with activity against PI3Kδ signaling, and has potential to reduce growth of tumors dependent on dysregulated PTEN for growth. Moreover, AZD8186 can be combined with docetaxel, a chemotherapy commonly used to treat advanced TBNC and prostate tumors. The ability to schedule AZD8186 and maintain efficacy offers opportunity to combine AZD8186 more effectively with other drugs., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
39. Discovery of 9-(1-anilinoethyl)-2-morpholino-4-oxo-pyrido[1,2-a]pyrimidine-7-carboxamides as PI3Kβ/δ inhibitors for the treatment of PTEN-deficient tumours.
- Author
-
Barlaam B, Cosulich S, Degorce S, Fitzek M, Giordanetto F, Green S, Inghardt T, Hennequin L, Hancox U, Lambert-van der Brempt C, Morgentin R, Pass S, Plé P, Saleh T, and Ward L
- Subjects
- Administration, Oral, Amides administration & dosage, Amides chemistry, Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Mice, Molecular Structure, Neoplasms, Experimental enzymology, Neoplasms, Experimental pathology, PTEN Phosphohydrolase metabolism, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors chemistry, Structure-Activity Relationship, Amides pharmacology, Antineoplastic Agents pharmacology, Drug Discovery, Neoplasms, Experimental drug therapy, PTEN Phosphohydrolase deficiency, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
Starting from TGX-221, we designed a series of 9-(1-anilinoethyl)-2-morpholino-4-oxo-pyrido[1,2-a]pyrimidine-7-carboxamides as potent and selective PI3Kβ/δ inhibitors. Structure-activity relationships and structure-property relationships around the aniline and the amide substituents are discussed. We identified compounds 17 and 18, which showed profound pharmacodynamic modulation of phosphorylated Akt in the PC3 prostate tumour xenograft, after a single oral dose. Compound 17 also gave significant inhibition of tumour growth in the PC3 prostate tumour xenograft model after chronic oral dosing., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. Identification of a subset of human non-small cell lung cancer patients with high PI3Kβ and low PTEN expression, more prevalent in squamous cell carcinoma.
- Author
-
Cumberbatch M, Tang X, Beran G, Eckersley S, Wang X, Ellston RP, Dearden S, Cosulich S, Smith PD, Behrens C, Kim ES, Su X, Fan S, Gray N, Blowers DP, Wistuba II, and Womack C
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Squamous Cell genetics, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Lung Neoplasms genetics, Tissue Array Analysis, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Squamous Cell metabolism, Lung Neoplasms metabolism, PTEN Phosphohydrolase biosynthesis, Phosphatidylinositol 3-Kinases biosynthesis
- Abstract
Purpose: The phosphoinositide 3-kinase (PI3K) pathway is a major oncogenic signaling pathway and an attractive target for therapeutic intervention. Signaling through the PI3K pathway is moderated by the tumor suppressor PTEN, which is deficient or mutated in many human cancers. Molecular characterization of the PI3K signaling network has not been well defined in lung cancer; in particular, the role of PI3Kβ and its relation to PTEN in non-small cell lung cancer NSCLC remain unclear., Experimental Design: Antibodies directed against PI3Kβ and PTEN were validated and used to examine, by immunohistochemistry, expression in 240 NSCLC resection tissues [tissue microarray (TMA) set 1]. Preliminary observations were extended to an independent set of tissues (TMA set 2) comprising 820 NSCLC patient samples analyzed in a separate laboratory applying the same validated antibodies and staining protocols. The staining intensities for PI3Kβ and PTEN were explored and colocalization of these markers in individual tumor cores were correlated., Results: PI3Kβ expression was elevated significantly in squamous cell carcinomas (SCC) compared with adenocarcinomas. In contrast, PTEN loss was greater in SCC than in adenocarcinoma. Detailed correlative analyses of individual patient samples revealed a significantly greater proportion of SCC in TMA set 1 with higher PI3Kβ and lower PTEN expression when compared with adenocarcinoma. These findings were reinforced following independent analyses of TMA set 2., Conclusions: We identify for the first time a subset of NSCLC more prevalent in SCC, with elevated expression of PI3Kβ accompanied by a reduction/loss of PTEN, for whom selective PI3Kβ inhibitors may be predicted to achieve greater clinical benefit., (©2013 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
41. Signaling via class IA Phosphoinositide 3-kinases (PI3K) in human, breast-derived cell lines.
- Author
-
Juvin V, Malek M, Anderson KE, Dion C, Chessa T, Lecureuil C, Ferguson GJ, Cosulich S, Hawkins PT, and Stephens LR
- Subjects
- Cell Line, Cell Line, Tumor, Epidermal Growth Factor pharmacology, Female, Humans, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Phosphatidylinositol 3-Kinases genetics, Phosphorylation drug effects, Signal Transduction genetics, Signal Transduction physiology, Phosphatidylinositol 3-Kinases metabolism
- Abstract
We have addressed the differential roles of class I Phosphoinositide 3-kinases (PI3K) in human breast-derived MCF10a (and iso-genetic derivatives) and MDA-MB 231 and 468 cells. Class I PI3Ks are heterodimers of p110 catalytic (α, β, δ and γ) and p50-101 regulatory subunits and make the signaling lipid, phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) that can activate effectors, eg protein kinase B (PKB), and responses, eg migration. The PtdIns(3,4,5)P3-3-phosphatase and tumour-suppressor, PTEN inhibits this pathway. p110α, but not other p110s, has a number of onco-mutant variants that are commonly found in cancers. mRNA-seq data shows that MCF10a cells express p110β>>α>δ with undetectable p110γ. Despite this, EGF-stimulated phosphorylation of PKB depended upon p110α-, but not β- or δ- activity. EGF-stimulated chemokinesis, but not chemotaxis, was also dependent upon p110α, but not β- or δ- activity. In the presence of single, endogenous alleles of onco-mutant p110α (H1047R or E545K), basal, but not EGF-stimulated, phosphorylation of PKB was increased and the effect of EGF was fully reversed by p110α inhibitors. Cells expressing either onco-mutant displayed higher basal motility and EGF-stimulated chemokinesis.This latter effect was, however, only partially-sensitive to PI3K inhibitors. In PTEN(-/-) cells, basal and EGF-stimulated phosphorylation of PKB was substantially increased, but the p110-dependency was variable between cell types. In MDA-MB 468s phosphorylation of PKB was significantly dependent on p110β, but not α- or δ- activity; in PTEN(-/-) MCF10a it remained, like the parental cells, p110α-dependent. Surprisingly, loss of PTEN suppressed basal motility and EGF-stimulated chemokinesis. These results indicate that; p110α is required for EGF signaling to PKB and chemokinesis, but not chemotaxis; onco-mutant alleles of p110α augment signaling in the absence of EGF and may increase motility, in part, via acutely modulating PI3K-activity-independent mechanisms. Finally, we demonstrate that there is not a universal mechanism that up-regulates p110β function in the absence of PTEN.
- Published
- 2013
- Full Text
- View/download PDF
42. Stable isotope-labelling analysis of the impact of inhibition of the mammalian target of rapamycin on protein synthesis.
- Author
-
Huo Y, Iadevaia V, Yao Z, Kelly I, Cosulich S, Guichard S, Foster LJ, and Proud CG
- Subjects
- Carbon Isotopes, Eukaryotic Initiation Factor-4E metabolism, HeLa Cells, Humans, Indoles pharmacology, Isotope Labeling, Morpholines pharmacology, Nitrogen Isotopes, Protein Biosynthesis genetics, Purines pharmacology, RNA, Messenger metabolism, Ribosomes metabolism, Sirolimus pharmacology, Protein Biosynthesis drug effects, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
mTORC1 [mTOR (mammalian target of rapamycin) complex 1] regulates diverse cell functions. mTORC1 controls the phosphorylation of several proteins involved in mRNA translation and the translation of specific mRNAs, including those containing a 5'-TOP (5'-terminal oligopyrimidine). To date, most of the proteins encoded by known 5'-TOP mRNAs are proteins involved in mRNA translation, such as ribosomal proteins and elongation factors. Rapamycin inhibits some mTORC1 functions, whereas mTOR-KIs (mTOR kinase inhibitors) interfere with all of them. mTOR-KIs inhibit overall protein synthesis more strongly than rapamycin. To study the effects of rapamycin or mTOR-KIs on synthesis of specific proteins, we applied pSILAC [pulsed SILAC (stable isotope-labelling with amino acids in cell culture)]. Our results reveal, first, that mTOR-KIs and rapamycin differentially affect the synthesis of many proteins. Secondly, mTOR-KIs inhibit the synthesis of proteins encoded by 5'-TOP mRNAs much more strongly than rapamycin does, revealing that these mRNAs are controlled by rapamycin-insensitive outputs from mTOR. Thirdly, the synthesis of certain other proteins shows a similar pattern of inhibition. Some of them appear to be encoded by 'novel' 5'-TOP mRNAs; they include proteins which, like known 5'-TOP mRNA-encoded proteins, are involved in protein synthesis, whereas others are enzymes involved in intermediary or anabolic metabolism. These results indicate that mTOR signalling may promote diverse biosynthetic processes through the translational up-regulation of specific mRNAs. Lastly, a SILAC-based approach revealed that, although rapamycin and mTOR-KIs have little effect on general protein stability, they stabilize proteins encoded by 5'-TOP mRNAs.
- Published
- 2012
- Full Text
- View/download PDF
43. Preclinical pharmacology of AZD5363, an inhibitor of AKT: pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic background.
- Author
-
Davies BR, Greenwood H, Dudley P, Crafter C, Yu DH, Zhang J, Li J, Gao B, Ji Q, Maynard J, Ricketts SA, Cross D, Cosulich S, Chresta CC, Page K, Yates J, Lane C, Watson R, Luke R, Ogilvie D, and Pass M
- Subjects
- Animals, Breast Neoplasms drug therapy, Breast Neoplasms enzymology, Cell Line, Tumor, Cell Proliferation, Female, Humans, Male, Mice, Mice, Nude, Mice, SCID, Prostatic Neoplasms drug therapy, Prostatic Neoplasms enzymology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Xenograft Model Antitumor Assays, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Pyrimidines pharmacology, Pyrroles pharmacology
- Abstract
AKT is a key node in the most frequently deregulated signaling network in human cancer. AZD5363, a novel pyrrolopyrimidine-derived compound, inhibited all AKT isoforms with a potency of 10 nmol/L or less and inhibited phosphorylation of AKT substrates in cells with a potency of approximately 0.3 to 0.8 μmol/L. AZD5363 monotherapy inhibited the proliferation of 41 of 182 solid and hematologic tumor cell lines with a potency of 3 μmol/L or less. Cell lines derived from breast cancers showed the highest frequency of sensitivity. There was a significant relationship between the presence of PIK3CA and/or PTEN mutations and sensitivity to AZD5363 and between RAS mutations and resistance. Oral dosing of AZD5363 to nude mice caused dose- and time-dependent reduction of PRAS40, GSK3β, and S6 phosphorylation in BT474c xenografts (PRAS40 phosphorylation EC(50) ~ 0.1 μmol/L total plasma exposure), reversible increases in blood glucose concentrations, and dose-dependent decreases in 2[18F]fluoro-2-deoxy-D-glucose ((18)F-FDG) uptake in U87-MG xenografts. Chronic oral dosing of AZD5363 caused dose-dependent growth inhibition of xenografts derived from various tumor types, including HER2(+) breast cancer models that are resistant to trastuzumab. AZD5363 also significantly enhanced the antitumor activity of docetaxel, lapatinib, and trastuzumab in breast cancer xenografts. It is concluded that AZD5363 is a potent inhibitor of AKT with pharmacodynamic activity in vivo, has potential to treat a range of solid and hematologic tumors as monotherapy or a combinatorial agent, and has potential for personalized medicine based on the genetic status of PIK3CA, PTEN, and RAS. AZD5363 is currently in phase I clinical trials., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
44. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity.
- Author
-
Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE, Vincent JP, Ellston R, Jones D, Sini P, James D, Howard Z, Dudley P, Hughes G, Smith L, Maguire S, Hummersone M, Malagu K, Menear K, Jenkins R, Jacobsen M, Smith GC, Guichard S, and Pass M
- Subjects
- Animals, Cell Proliferation drug effects, Female, Humans, Mice, Mice, Nude, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, TOR Serine-Threonine Kinases, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Morpholines pharmacology, Neoplasms, Experimental drug therapy, Protein Kinases drug effects, Signal Transduction drug effects
- Abstract
The mammalian target of rapamycin (mTOR) kinase forms two multiprotein complexes, mTORC1 and mTORC2, which regulate cell growth, cell survival, and autophagy. Allosteric inhibitors of mTORC1, such as rapamycin, have been extensively used to study tumor cell growth, proliferation, and autophagy but have shown only limited clinical utility. Here, we describe AZD8055, a novel ATP-competitive inhibitor of mTOR kinase activity, with an IC50 of 0.8 nmol/L. AZD8055 showed excellent selectivity (approximately 1,000-fold) against all class I phosphatidylinositol 3-kinase (PI3K) isoforms and other members of the PI3K-like kinase family. Furthermore, there was no significant activity against a panel of 260 kinases at concentrations up to 10 micromol/L. AZD8055 inhibits the phosphorylation of mTORC1 substrates p70S6K and 4E-BP1 as well as phosphorylation of the mTORC2 substrate AKT and downstream proteins. The rapamycin-resistant T37/46 phosphorylation sites on 4E-BP1 were fully inhibited by AZD8055, resulting in significant inhibition of cap-dependent translation. In vitro, AZD8055 potently inhibits proliferation and induces autophagy in H838 and A549 cells. In vivo, AZD8055 induces a dose-dependent pharmacodynamic effect on phosphorylated S6 and phosphorylated AKT at plasma concentrations leading to tumor growth inhibition. Notably, AZD8055 results in significant growth inhibition and/or regression in xenografts, representing a broad range of human tumor types. AZD8055 is currently in phase I clinical trials.
- Published
- 2010
- Full Text
- View/download PDF
45. PPAR alpha and the regulation of cell division and apoptosis.
- Author
-
Roberts RA, Chevalier S, Hasmall SC, James NH, Cosulich SC, and Macdonald N
- Subjects
- Antigens, CD genetics, Apoptosis drug effects, Cell Division drug effects, Cytokines physiology, Humans, Receptors, Tumor Necrosis Factor genetics, Receptors, Tumor Necrosis Factor, Type I, Apoptosis physiology, Cell Division physiology, Peroxisome Proliferators pharmacology, Receptors, Cytoplasmic and Nuclear physiology, Transcription Factors physiology
- Abstract
Peroxisome proliferators (PPs) such as the hypolipidaemic drug, nafenopin and the phthalate plasticiser 2-diethylhexylphthalate induce rodent hepatocyte cell proliferation and suppress apoptosis leading to tumours. PPs act via the nuclear hormone receptor peroxisome proliferator activated receptor alpha (PPAR alpha) which directly regulates genes implicated in the response to PPs such as the peroxisomal gene acyl CoA oxidase. As expected for xenobiotics that perturb proliferation, PPs alter expression of cell cycle regulatory proteins. However, the ability to alter expression of cyclins and cyclin-dependent kinases is shared by physiological hepatic mitogens such as epidermal growth factor and is thus unlikely to be specific to the PP-induced aberrant growth associated with hepatocarcinogenesis. Recent evidence suggests that the response of hepatocytes to PPs is not only dependent upon PPAR alpha but also on the trophic environment provided by nonparenchymal cells and by cytokines such as tumour necrosis factor alpha. Additionally, the ability of PPs to suppress apoptosis and induce proliferation depends upon survival signalling mediated by p38 mitogen activated protein kinase. The cross talk between PPAR alpha-mediated transcription, survival signalling and cell cycle will be discussed with particular emphasis on relevance to toxicology.
- Published
- 2002
- Full Text
- View/download PDF
46. The role of protein kinase B and mitogen-activated protein kinase in epidermal growth factor and tumor necrosis factor alpha-mediated rat hepatocyte survival and apoptosis.
- Author
-
Roberts RA, James NH, and Cosulich SC
- Subjects
- Animals, Cell Survival drug effects, Cells, Cultured, Enzyme Activation, Liver cytology, Liver enzymology, Mitogen-Activated Protein Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt, Rats, p38 Mitogen-Activated Protein Kinases, Apoptosis drug effects, Epidermal Growth Factor pharmacology, Liver physiology, Mitogen-Activated Protein Kinases physiology, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins physiology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Perturbation of hepatocyte growth regulation is associated with a number of liver diseases such as fibrosis and cancer. These diseases are mediated by a network of growth factors and cytokines that regulate the induction of hepatocyte proliferation and apoptosis. In this study, we have investigated the role of signaling pathways activated by tumor necrosis factor alpha (TNF-alpha) and epidermal growth factor (EGF) in the regulation of apoptosis induced by transforming growth factor beta(1) (TGF-beta(1)), because this physiological factor is believed to regulate spontaneous apoptosis in the liver. We show that pretreatment with (10 ng/mL) EGF or (25 ng/mL) TNF-alpha can suppress TGF-beta(1)-induced apoptosis by 73% and 50%, respectively, in isolated rat hepatocytes. However, suppression of TGF-beta(1)-induced apoptosis by EGF and TNF-alpha occurs via different protein kinase signaling pathways. Using specific inhibitors, we show that suppression of apoptosis by EGF is dependent on activation of phosphoinositide 3-kinase (PI 3-kinase) and the extracellular signal regulated kinase (ERK) mitogen-activated protein (MAP) kinase pathways, but not p38 MAP kinase. In contrast, suppression of TGF-beta(1)-induced apoptosis by TNF-alpha does not require PI 3-kinase and protein kinase B (PKB or Akt)-mediated pathways, but is dependent on ERK and p38 MAP kinase activity. These data contribute to our understanding of the intracellular survival signals that play a role in normal liver homeostasis and in diverse pathological conditions.
- Published
- 2000
- Full Text
- View/download PDF
47. Role for tumor necrosis factor alpha receptor 1 and interleukin-1 receptor in the suppression of mouse hepatocyte apoptosis by the peroxisome proliferator nafenopin.
- Author
-
West DA, James NH, Cosulich SC, Holden PR, Brindle R, Rolfe M, and Roberts RA
- Subjects
- Animals, Antibodies, Antigens, CD chemistry, Antigens, CD classification, Cell Nucleus drug effects, Cell Nucleus metabolism, Cell Survival drug effects, Cells, Cultured, DNA biosynthesis, DNA genetics, DNA metabolism, Interleukin-1 antagonists & inhibitors, Interleukin-1 pharmacology, Liver drug effects, Male, Mice, NF-kappa B metabolism, Oxidation-Reduction drug effects, Peroxisome Proliferators pharmacology, Peroxisomes drug effects, Peroxisomes metabolism, Receptors, Interleukin-1 antagonists & inhibitors, Receptors, Tumor Necrosis Factor chemistry, Receptors, Tumor Necrosis Factor classification, Receptors, Tumor Necrosis Factor, Type I, S Phase drug effects, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta pharmacology, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha physiology, Antigens, CD physiology, Apoptosis drug effects, Liver cytology, Nafenopin antagonists & inhibitors, Nafenopin pharmacology, Peroxisome Proliferators antagonists & inhibitors, Receptors, Interleukin-1 physiology, Receptors, Tumor Necrosis Factor physiology
- Abstract
Peroxisome proliferators (PPs) cause rodent liver enlargement and tumors. In vitro, PPs induce rat and mouse hepatocyte DNA synthesis and suppress apoptosis, a response mimicked by exogenous tumor necrosis factor alpha (TNFalpha). Here, we determine the role of TNF receptor 1 (TNFR1), TNF receptor 2 (TNFR2), and nuclear factor kappa beta (NFkappaB) in the response of mouse hepatocytes to the PP, nafenopin. Nafenopin (50 micromol/L) induced DNA synthesis as measured by bromodeoxyuridine (BrdU) incorporation, suppressed cell death as measured by Hoechst 33258 staining, induced peroxisomal beta-oxidation as measured by cyanide insensitive palmitoyl CoA oxidation (PCO) and caused activation of nuclear factor kappa beta (NFkappaB) as determined by electrophoretic mobility gel shift assay (EMSA). The induction of DNA synthesis and the suppression of apoptosis in response to nafenopin was abrogated completely by blocking antibodies to TNFR1 but not to TNFR2. In contrast, the induction of peroxisomal beta-oxidation by nafenopin was not blocked by the anti-TNFR1 antibody. Next, we evaluated the response of hepatocytes to interleukin-1 (IL-1), another proinflammatory cytokine. IL-1alpha (2.5 ng/mL) and, to a lesser extent, IL-1beta (5 ng/mL), shared the ability of TNFalpha to induce DNA synthesis and suppress apoptosis. In addition, anti-IL-1 receptor, type 1/p80 (IL-1R) antibodies were able to abrogate the response to nafenopin. IL-1alpha was still able to perturb hepatocyte growth in the presence of the anti-TNFR1 antibody suggesting that IL-1alpha acts independently rather than by elaborating TNFalpha. In summary, these data provide additional evidence for a role for hepatic cytokines in the perturbation of hepatocyte growth by PPs such as nafenopin.
- Published
- 1999
- Full Text
- View/download PDF
48. Suppression of apoptosis and induction of DNA synthesis in vitro by the phthalate plasticizers monoethylhexylphthalate (MEHP) and diisononylphthalate (DINP): a comparison of rat and human hepatocytes in vitro.
- Author
-
Hasmall SC, James NH, Macdonald N, West D, Chevalier S, Cosulich SC, and Roberts RA
- Subjects
- Animals, DNA Replication drug effects, Diethylhexyl Phthalate toxicity, Humans, Liver cytology, Liver drug effects, Male, Rats, Rats, Inbred F344, Transforming Growth Factor beta pharmacology, Apoptosis drug effects, Carcinogens toxicity, DNA biosynthesis, Diethylhexyl Phthalate analogs & derivatives, Liver metabolism, Phthalic Acids toxicity, Plasticizers toxicity
- Abstract
Diethylhexylphthalate (DEHP) and diisononylphthalate (DINP) are plasticizers with many important commercial, industrial and medical applications. However, both DEHP and DINP are rodent peroxisome proliferators (PPs), a class of compounds that cause rodent liver tumours associated with peroxisome proliferation, induction of hepatic DNA synthesis and the suppression of apoptosis. Despite these effects in the rodent, humans appear to be nonresponsive to the adverse effects of PPs. Previously, we have shown that the fibrate hypolipidaemic peroxisome proliferator, nafenopin, induced DNA synthesis and suppressed apoptosis in rat but not in human hepatocytes. In this work, we have examined species differences in the response of rat and human hepatocytes to DEHP and DINP in vitro. In rat hepatocytes in vitro, both DINP and MEHP (a principle metabolite of DEHP and the proximal peroxisome proliferator) caused a concentration-dependent induction of DNA synthesis and suppression of both spontaneous and transforming growth factor beta1 (TGFbeta1)-induced apoptosis. Similarly, both MEHP and DINP caused a concentration-dependent induction of peroxisomal beta-oxidation although the response to DINP was less robust. In contrast to the pleiotropic response noted in rat hepatocytes, neither DINP nor MEHP caused an induction of beta-oxidation, stimulation of DNA synthesis and suppression of apoptosis in human hepatocytes cultured from three separate donors. These data provide evidence for species differences in the hepatic response to the phthalates DEHP and DINP, confirming that human hepatocytes appear to be refractory to the hepatocarcinogenic effects of PPs first noted in rodents.
- Published
- 1999
- Full Text
- View/download PDF
49. Bcl-2 regulates a caspase-3/caspase-2 apoptotic cascade in cytosolic extracts.
- Author
-
Swanton E, Savory P, Cosulich S, Clarke P, and Woodman P
- Subjects
- Caspase 2, Caspase 3, Cell Extracts, Cytosol enzymology, Enzyme Activation, HeLa Cells, Humans, Apoptosis, Caspases metabolism, Enzyme Precursors metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
Apoptosis is accompanied by the activation of a number of apoptotic proteases (caspases) which selectively cleave specific cellular substrates. Caspases themselves are zymogens which are activated by proteolysis. It is widely believed that 'initiator' caspases are recruited to and activated within apoptotic signalling complexes, and then cleave and activate downstream 'effector' caspases. While activation of the effector caspase, caspase-3, has indeed been observed as distal to activation of several different initiator caspases, evidence for a further downstream proteolytic cascade is limited. In particular, there is little evidence that cellular levels of caspase-3 that are activated via one pathway are sufficient to cleave and activate other initiator caspases. To address this issue, the ability of caspase-3, activated upon addition to cytosolic extracts of cytochrome c, to cause cleavage of caspase-2 was investigated. It was demonstrated that cleavage of caspase-2 follows, and is dependent upon, activation of caspase-3. Moreover, the activation of both caspases was inhibited by Bcl-2. Together, these data indicate that Bcl-2 can protect cells from apoptosis by acting at a point downstream from release of mitochondrial cytochrome c, thereby preventing a caspase-3 dependent proteolytic cascade.
- Published
- 1999
- Full Text
- View/download PDF
50. Bcl-2 regulates amplification of caspase activation by cytochrome c.
- Author
-
Cosulich SC, Savory PJ, and Clarke PR
- Subjects
- Animals, Apoptosis, Caspase 3, Cell-Free System, Cytochrome c Group physiology, Enzyme Activation drug effects, Feedback, HeLa Cells, Humans, Mitochondria enzymology, Oligopeptides pharmacology, Oocytes, Proto-Oncogene Proteins c-bcl-2 physiology, Recombinant Fusion Proteins pharmacology, Xenopus Proteins, Xenopus laevis, bcl-X Protein, Caspases metabolism, Cytochrome c Group pharmacology, Proto-Oncogene Proteins c-bcl-2 pharmacology
- Abstract
Caspases, a family of specific proteases, have central roles in apoptosis [1]. Caspase activation in response to diverse apoptotic stimuli involves the relocalisation of cytochrome c from mitochondria to the cytoplasm where it stimulates the proteolytic processing of caspase precursors. Cytochrome c release is controlled by members of the Bcl-2 family of apoptosis regulators [2] [3]. The anti-apoptotic members Bcl-2 and Bcl-xL may also control caspase activation independently of cytochrome c relocalisation or may inhibit a positive feedback mechanism [4] [5] [6] [7]. Here, we investigate the role of Bcl-2 family proteins in the regulation of caspase activation using a model cell-free system. We found that Bcl-2 and Bcl-xL set a threshold in the amount of cytochrome c required to activate caspases, even in soluble extracts lacking mitochondria. Addition of dATP (which stimulates the procaspase-processing factor Apaf-1 [8] [9]) overcame inhibition of caspase activation by Bcl-2, but did not prevent the control of cytochrome c release from mitochondria by Bcl-2. Cytochrome c release was accelerated by active caspase-3 and this positive feedback was negatively regulated by Bcl-2. These results provide evidence for a mechanism to amplify caspase activation that is suppressed at several distinct steps by Bcl-2, even after cytochrome c is released from mitochondria.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.