58 results on '"Nicola Colclough"'
Search Results
2. Expanding the Armory: Predicting and Tuning Covalent Warhead Reactivity.
- Author
-
Richard Lonsdale, Jonathan Burgess, Nicola Colclough, Nichola L. Davies, Eva M. Lenz, Alexandra L. Orton, and Richard A. Ward
- Published
- 2017
- Full Text
- View/download PDF
3. Unbound Brain-to-Plasma Partition Coefficient, Kp,uu,brain—a Game Changing Parameter for CNS Drug Discovery and Development
- Author
-
Irena Loryan, Andreas Reichel, Bo Feng, Christoffer Bundgaard, Christopher Shaffer, Cory Kalvass, Dallas Bednarczyk, Denise Morrison, Dominique Lesuisse, Edmund Hoppe, Georg C. Terstappen, Holger Fischer, Li Di, Nicola Colclough, Scott Summerfield, Stephen T. Buckley, Tristan S. Maurer, and Markus Fridén
- Subjects
CNS drug development ,Pharmacology ,drug transport ,unbound brain-to-plasma partition coefficient ,neuropharmacokinetics ,Organic Chemistry ,Pharmaceutical Science ,K-p uu brain ,blood-brain barrier ,Farmaceutiska vetenskaper ,Pharmaceutical Sciences ,Molecular Medicine ,Pharmacology (medical) ,Biotechnology - Abstract
Purpose More than 15 years have passed since the first description of the unbound brain-to-plasma partition coefficient (Kp,uu,brain) by Prof. Margareta Hammarlund-Udenaes, which was enabled by advancements in experimental methodologies including cerebral microdialysis. Since then, growing knowledge and data continue to support the notion that the unbound (free) concentration of a drug at the site of action, such as the brain, is the driving force for pharmacological responses. Towards this end, Kp,uu,brain is the key parameter to obtain unbound brain concentrations from unbound plasma concentrations. Methods To understand the importance and impact of the Kp,uu,brain concept in contemporary drug discovery and development, a survey has been conducted amongst major pharmaceutical companies based in Europe and the USA. Here, we present the results from this survey which consisted of 47 questions addressing: 1) Background information of the companies, 2) Implementation, 3) Application areas, 4) Methodology, 5) Impact and 6) Future perspectives. Results and conclusions From the responses, it is clear that the majority of the companies (93%) has established a common understanding across disciplines of the concept and utility of Kp,uu,brain as compared to other parameters related to brain exposure. Adoption of the Kp,uu,brain concept has been mainly driven by individual scientists advocating its application in the various companies rather than by a top-down approach. Remarkably, 79% of all responders describe the portfolio impact of Kp,uu,brain implementation in their companies as ‘game-changing’. Although most companies (74%) consider the current toolbox for Kp,uu,brain assessment and its validation satisfactory for drug discovery and early development, areas of improvement and future research to better understand human brain pharmacokinetics/pharmacodynamics translation have been identified.
- Published
- 2022
- Full Text
- View/download PDF
4. Supplementary Information from Orally Bioavailable and Blood–Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Kristoffer Valerie, Stephen T. Durant, Martin Pass, Kan Chen, Yingchun Wang, Tianwei Zhang, Li Zheng, Ian P. Barrett, Aaron Smith, Joanne Wilson, Nicola Colclough, Jason Kettle, Sebastien L. Degorce, Bernard Barlaam, Nitai Mukhopadhyay, Jason M. Beckta, Jenna Kahn, Laura Biddlestone-Thorpe, Nicholas C.K. Valerie, Amrita Sule, Thomas A. Hunt, Kurt G. Pike, Alan Lau, Jennifer Vincent, Bhavika Patel, Andrew G. Thomason, Antonio Garcia-Trinidad, Lucy C. Riches, Elaine B. Cadogan, Paul Farrington, Rajesh Odedra, Victoria Sheridan, Gareth Hughes, Syed F. Ahmad, Jasmine Allen, and Jeremy Karlin
- Abstract
Supplementary Information
- Published
- 2023
- Full Text
- View/download PDF
5. Video 2 - ATMi increases the rate of mitotic catastrophe in glioma cells when p53 is knocked down. from Orally Bioavailable and Blood–Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Kristoffer Valerie, Stephen T. Durant, Martin Pass, Kan Chen, Yingchun Wang, Tianwei Zhang, Li Zheng, Ian P. Barrett, Aaron Smith, Joanne Wilson, Nicola Colclough, Jason Kettle, Sebastien L. Degorce, Bernard Barlaam, Nitai Mukhopadhyay, Jason M. Beckta, Jenna Kahn, Laura Biddlestone-Thorpe, Nicholas C.K. Valerie, Amrita Sule, Thomas A. Hunt, Kurt G. Pike, Alan Lau, Jennifer Vincent, Bhavika Patel, Andrew G. Thomason, Antonio Garcia-Trinidad, Lucy C. Riches, Elaine B. Cadogan, Paul Farrington, Rajesh Odedra, Victoria Sheridan, Gareth Hughes, Syed F. Ahmad, Jasmine Allen, and Jeremy Karlin
- Abstract
U87/Centrin2-EGFP/H2B-mCherry/puro cells were seeded on dishes with cover slip bottoms, treated with AZ32 (3 uM), and irradiated (5 Gy). Time-lapse videos were recorded intermittently (every 7 min) over 16 hrs. Treatment results in 11% aberrant mitoses (see Fig. 3F).
- Published
- 2023
- Full Text
- View/download PDF
6. Video 3 - ATMi increases the rate of mitotic catastrophe in glioma cells when p53 is knocked down. from Orally Bioavailable and Blood–Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Kristoffer Valerie, Stephen T. Durant, Martin Pass, Kan Chen, Yingchun Wang, Tianwei Zhang, Li Zheng, Ian P. Barrett, Aaron Smith, Joanne Wilson, Nicola Colclough, Jason Kettle, Sebastien L. Degorce, Bernard Barlaam, Nitai Mukhopadhyay, Jason M. Beckta, Jenna Kahn, Laura Biddlestone-Thorpe, Nicholas C.K. Valerie, Amrita Sule, Thomas A. Hunt, Kurt G. Pike, Alan Lau, Jennifer Vincent, Bhavika Patel, Andrew G. Thomason, Antonio Garcia-Trinidad, Lucy C. Riches, Elaine B. Cadogan, Paul Farrington, Rajesh Odedra, Victoria Sheridan, Gareth Hughes, Syed F. Ahmad, Jasmine Allen, and Jeremy Karlin
- Abstract
U87/shp53/Centrin2-EGFP/H2B-mCherry/shp53 cells were seeded on dishes with cover slip bottoms and irradiated (5 Gy). Time-lapse videos were recorded intermittently (every 7 min) over 16 hrs. Treatment results in 12% aberrant mitoses (see Fig. 3F).
- Published
- 2023
- Full Text
- View/download PDF
7. Video 1 - ATMi increases the rate of mitotic catastrophe in glioma cells when p53 is knocked down. from Orally Bioavailable and Blood–Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Kristoffer Valerie, Stephen T. Durant, Martin Pass, Kan Chen, Yingchun Wang, Tianwei Zhang, Li Zheng, Ian P. Barrett, Aaron Smith, Joanne Wilson, Nicola Colclough, Jason Kettle, Sebastien L. Degorce, Bernard Barlaam, Nitai Mukhopadhyay, Jason M. Beckta, Jenna Kahn, Laura Biddlestone-Thorpe, Nicholas C.K. Valerie, Amrita Sule, Thomas A. Hunt, Kurt G. Pike, Alan Lau, Jennifer Vincent, Bhavika Patel, Andrew G. Thomason, Antonio Garcia-Trinidad, Lucy C. Riches, Elaine B. Cadogan, Paul Farrington, Rajesh Odedra, Victoria Sheridan, Gareth Hughes, Syed F. Ahmad, Jasmine Allen, and Jeremy Karlin
- Abstract
U87/Centrin2-EGFP/H2B-mCherry/puro cells were seeded on dishes with cover slip bottoms and irradiated (5 Gy). Time-lapse videos were recorded intermittently (every 7 min) over 16 hrs. Treatment results in 2.7% aberrant mitoses (see Fig. 3F).
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Orally Bioavailable and Blood–Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Kristoffer Valerie, Stephen T. Durant, Martin Pass, Kan Chen, Yingchun Wang, Tianwei Zhang, Li Zheng, Ian P. Barrett, Aaron Smith, Joanne Wilson, Nicola Colclough, Jason Kettle, Sebastien L. Degorce, Bernard Barlaam, Nitai Mukhopadhyay, Jason M. Beckta, Jenna Kahn, Laura Biddlestone-Thorpe, Nicholas C.K. Valerie, Amrita Sule, Thomas A. Hunt, Kurt G. Pike, Alan Lau, Jennifer Vincent, Bhavika Patel, Andrew G. Thomason, Antonio Garcia-Trinidad, Lucy C. Riches, Elaine B. Cadogan, Paul Farrington, Rajesh Odedra, Victoria Sheridan, Gareth Hughes, Syed F. Ahmad, Jasmine Allen, and Jeremy Karlin
- Abstract
Inhibition of ataxia-telangiectasia mutated (ATM) during radiotherapy of glioblastoma multiforme (GBM) may improve tumor control by short-circuiting the response to radiation-induced DNA damage. A major impediment for clinical implementation is that current inhibitors have limited central nervous system (CNS) bioavailability; thus, the goal was to identify ATM inhibitors (ATMi) with improved CNS penetration. Drug screens and refinement of lead compounds identified AZ31 and AZ32. The compounds were then tested in vivo for efficacy and impact on tumor and healthy brain. Both AZ31 and AZ32 blocked the DNA damage response and radiosensitized GBM cells in vitro. AZ32, with enhanced blood–brain barrier (BBB) penetration, was highly efficient in vivo as radiosensitizer in syngeneic and human, orthotopic mouse glioma model compared with AZ31. Furthermore, human glioma cell lines expressing mutant p53 or having checkpoint-defective mutations were particularly sensitive to ATMi radiosensitization. The mechanism for this p53 effect involves a propensity to undergo mitotic catastrophe relative to cells with wild-type p53. In vivo, apoptosis was >6-fold higher in tumor relative to healthy brain after exposure to AZ32 and low-dose radiation. AZ32 is the first ATMi with oral bioavailability shown to radiosensitize glioma and improve survival in orthotopic mouse models. These findings support the development of a clinical-grade, BBB-penetrating ATMi for the treatment of GBM. Importantly, because many GBMs have defective p53 signaling, the use of an ATMi concurrent with standard radiotherapy is expected to be cancer-specific, increase the therapeutic ratio, and maintain full therapeutic effect at lower radiation doses. Mol Cancer Ther; 17(8); 1637–47. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Preclinical Comparison of the Blood–brain barrier Permeability of Osimertinib with Other EGFR TKIs
- Author
-
Darren A.E. Cross, Zack Cheng, Lars Farde, Paul D. Smith, James W.T. Yates, Richard A. Ward, M. Raymond V. Finlay, Natasha A. Karp, Albert D. Windhorst, Mohammad Mahdi Moein, Ana Vazquez-Romero, Evgeny Revunov, Jonas Malmquist, Mikhail Kondrashov, Ryosuke Arakawa, Akihiro Takano, Aaron Smith, Joanne Wilson, Annika Janefeldt, Lin Zhang, James Atkinson, Peter Barton, Gareth Maglennon, Don X. Nguyen, Minghui Zhao, Sally J. Adua, Katarina Varnäs, Richard Goodwin, Magnus Schou, Gail L. Wrigley, Yumei Yan, Nicole Strittmatter, Peter Johnström, Kan Chen, and Nicola Colclough
- Abstract
Purpose:Osimertinib is a potent and selective EGFR tyrosine kinase inhibitor (EGFR-TKI) of both sensitizing and T790M resistance mutations. To treat metastatic brain disease, blood–brain barrier (BBB) permeability is considered desirable for increasing clinical efficacy.Experimental Design:We examined the level of brain penetration for 16 irreversible and reversible EGFR-TKIs using multiple in vitro and in vivo BBB preclinical models.Results:In vitro osimertinib was the weakest substrate for human BBB efflux transporters (efflux ratio 3.2). In vivo rat free brain to free plasma ratios (Kpuu) show osimertinib has the most BBB penetrance (0.21), compared with the other TKIs (Kpuu ≤ 0.12). PET imaging in Cynomolgus macaques demonstrated osimertinib was the only TKI among those tested to achieve significant brain penetrance (Cmax %ID 1.5, brain/blood Kp 2.6). Desorption electrospray ionization mass spectroscopy images of brains from mouse PC9 macrometastases models showed osimertinib readily distributes across both healthy brain and tumor tissue. Comparison of osimertinib with the poorly BBB penetrant afatinib in a mouse PC9 model of subclinical brain metastases showed only osimertinib has a significant effect on rate of brain tumor growth.Conclusions:These preclinical studies indicate that osimertinib can achieve significant exposure in the brain compared with the other EGFR-TKIs tested and supports the ongoing clinical evaluation of osimertinib for the treatment of EGFR-mutant brain metastasis. This work also demonstrates the link between low in vitro transporter efflux ratios and increased brain penetrance in vivo supporting the use of in vitro transporter assays as an early screen in drug discovery.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S1 from Preclinical Comparison of the Blood–brain barrier Permeability of Osimertinib with Other EGFR TKIs
- Author
-
Darren A.E. Cross, Zack Cheng, Lars Farde, Paul D. Smith, James W.T. Yates, Richard A. Ward, M. Raymond V. Finlay, Natasha A. Karp, Albert D. Windhorst, Mohammad Mahdi Moein, Ana Vazquez-Romero, Evgeny Revunov, Jonas Malmquist, Mikhail Kondrashov, Ryosuke Arakawa, Akihiro Takano, Aaron Smith, Joanne Wilson, Annika Janefeldt, Lin Zhang, James Atkinson, Peter Barton, Gareth Maglennon, Don X. Nguyen, Minghui Zhao, Sally J. Adua, Katarina Varnäs, Richard Goodwin, Magnus Schou, Gail L. Wrigley, Yumei Yan, Nicole Strittmatter, Peter Johnström, Kan Chen, and Nicola Colclough
- Abstract
Supplementary Figure S1 - Structure and position of label for compounds assessed for BBB penetrance in non human primates (NHP). Majority of compounds were labelled with carbon-11 except afatinib which was labelled with fluorine-18. (*) Indicates position for carbon-11 label
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data from Preclinical Comparison of the Blood–brain barrier Permeability of Osimertinib with Other EGFR TKIs
- Author
-
Darren A.E. Cross, Zack Cheng, Lars Farde, Paul D. Smith, James W.T. Yates, Richard A. Ward, M. Raymond V. Finlay, Natasha A. Karp, Albert D. Windhorst, Mohammad Mahdi Moein, Ana Vazquez-Romero, Evgeny Revunov, Jonas Malmquist, Mikhail Kondrashov, Ryosuke Arakawa, Akihiro Takano, Aaron Smith, Joanne Wilson, Annika Janefeldt, Lin Zhang, James Atkinson, Peter Barton, Gareth Maglennon, Don X. Nguyen, Minghui Zhao, Sally J. Adua, Katarina Varnäs, Richard Goodwin, Magnus Schou, Gail L. Wrigley, Yumei Yan, Nicole Strittmatter, Peter Johnström, Kan Chen, and Nicola Colclough
- Abstract
Supplementary Data
- Published
- 2023
- Full Text
- View/download PDF
12. Interpreting physicochemical experimental data sets.
- Author
-
Nicola Colclough and Mark C. Wenlock
- Published
- 2015
- Full Text
- View/download PDF
13. Preclinical Comparison of the Blood–brain barrier Permeability of Osimertinib with Other EGFR TKIs
- Author
-
Aaron Smith, Peter Johnström, Katarina Varnäs, James Atkinson, Minghui Zhao, Nicole Strittmatter, Yumei Yan, Ryosuke Arakawa, Nicola Colclough, Annika Janefeldt, M. Raymond V. Finlay, Albert D. Windhorst, Evgeny Revunov, Natasha A. Karp, Kan Chen, Lin Zhang, Gareth Maglennon, Peter Barton, Richard J. A. Goodwin, Ana Vazquez-Romero, Magnus Schou, Akihiro Takano, Gail L. Wrigley, Darren Cross, Mikhail Kondrashov, Mohammad Mahdi Moein, Zack Cheng, Paul D. Smith, Sally J. Adua, Richard A. Ward, Lars Farde, James W.T. Yates, Joanne Wilson, Jonas Malmquist, Don X. Nguyen, CCA - Imaging and biomarkers, Amsterdam Neuroscience - Brain Imaging, and Radiology and nuclear medicine
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Afatinib ,Brain tumor ,Pharmacology ,Permeability ,Madin Darby Canine Kidney Cells ,Mice ,03 medical and health sciences ,T790M ,Dogs ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Humans ,Tissue Distribution ,Osimertinib ,Protein Kinase Inhibitors ,Acrylamides ,Aniline Compounds ,Brain Neoplasms ,business.industry ,Transporter ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,ErbB Receptors ,Macaca fascicularis ,030104 developmental biology ,Oncology ,Blood-Brain Barrier ,030220 oncology & carcinogenesis ,Efflux ,business ,Brain metastasis ,medicine.drug - Abstract
Purpose: Osimertinib is a potent and selective EGFR tyrosine kinase inhibitor (EGFR-TKI) of both sensitizing and T790M resistance mutations. To treat metastatic brain disease, blood–brain barrier (BBB) permeability is considered desirable for increasing clinical efficacy. Experimental Design: We examined the level of brain penetration for 16 irreversible and reversible EGFR-TKIs using multiple in vitro and in vivo BBB preclinical models. Results: In vitro osimertinib was the weakest substrate for human BBB efflux transporters (efflux ratio 3.2). In vivo rat free brain to free plasma ratios (Kpuu) show osimertinib has the most BBB penetrance (0.21), compared with the other TKIs (Kpuu ≤ 0.12). PET imaging in Cynomolgus macaques demonstrated osimertinib was the only TKI among those tested to achieve significant brain penetrance (Cmax %ID 1.5, brain/blood Kp 2.6). Desorption electrospray ionization mass spectroscopy images of brains from mouse PC9 macrometastases models showed osimertinib readily distributes across both healthy brain and tumor tissue. Comparison of osimertinib with the poorly BBB penetrant afatinib in a mouse PC9 model of subclinical brain metastases showed only osimertinib has a significant effect on rate of brain tumor growth. Conclusions: These preclinical studies indicate that osimertinib can achieve significant exposure in the brain compared with the other EGFR-TKIs tested and supports the ongoing clinical evaluation of osimertinib for the treatment of EGFR-mutant brain metastasis. This work also demonstrates the link between low in vitro transporter efflux ratios and increased brain penetrance in vivo supporting the use of in vitro transporter assays as an early screen in drug discovery.
- Published
- 2021
- Full Text
- View/download PDF
14. Potent and Selective Inhibitors of the Epidermal Growth Factor Receptor to Overcome C797S-Mediated Resistance
- Author
-
Darren Cross, Matthew J. Martin, Carine M. Guérot, Nicolas Floc'h, Clare Gregson, Jonathan P. Orme, Michal Bista, Lin Xue, Xu Li, Amar Rahi, Xiliang Zhao, Richard A. Ward, L. Evans, Gail L. Wrigley, Arash Mosallanejad, Tieguang Yao, Claire McWhirter, David J. Hargreaves, Nicola Colclough, Sue Bickerton, Darren Mckerrecher, Peter Barton, Yang Ye, Yi Liu, Marta Wylot, M. Raymond V. Finlay, Xiaoming Kang, Eva M. Lenz, Daniel O'Neill, Verity Talbot, Olivier Lorthioir, and Paul D. Smith
- Subjects
Mice, Nude ,Antineoplastic Agents ,Mice, SCID ,medicine.disease_cause ,T790M ,Organophosphorus Compounds ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Osimertinib ,Epidermal growth factor receptor ,Lung cancer ,Cell potency ,Protein Kinase Inhibitors ,EGFR inhibitors ,Mutation ,biology ,Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,ErbB Receptors ,Pyrimidines ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Molecular Medicine ,Female - Abstract
The epidermal growth factor receptor (EGFR) harboring activating mutations is a clinically validated target in non-small-cell lung cancer, and a number of inhibitors of the EGFR tyrosine kinase domain, including osimertinib, have been approved for clinical use. Resistance to these therapies has emerged due to a variety of molecular events including the C797S mutation which renders third-generation C797-targeting covalent EGFR inhibitors considerably less potent against the target due to the loss of the key covalent-bond-forming residue. We describe the medicinal chemistry optimization of a biochemically potent but modestly cell-active, reversible EGFR inhibitor starting point with sub-optimal physicochemical properties. These studies culminated in the identification of compound 12 that showed improved cell potency, oral exposure, and in vivo activity in clinically relevant EGFR-mutant-driven disease models, including an Exon19 deletion/T790M/C797S triple-mutant mouse xenograft model.
- Published
- 2021
15. Brain metastatic outgrowth and osimertinib resistance are potentiated by RhoA in EGFR-mutant lung cancer
- Author
-
Sally J. Adua, Anna Arnal-Estapé, Minghui Zhao, Bowen Qi, Zongzhi Z. Liu, Carolyn Kravitz, Heather Hulme, Nicole Strittmatter, Francesc López-Giráldez, Sampada Chande, Alexandra E. Albert, Mary-Ann Melnick, Bomiao Hu, Katerina Politi, Veronica Chiang, Nicola Colclough, Richard J. A. Goodwin, Darren Cross, Paul Smith, and Don X. Nguyen
- Subjects
Multidisciplinary ,Lung Neoplasms ,Aniline Compounds ,General Physics and Astronomy ,Brain ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,ErbB Receptors ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Mutation ,Tumor Microenvironment ,Humans ,Neoplasm Recurrence, Local ,rhoA GTP-Binding Protein ,Protein Kinase Inhibitors - Abstract
The brain is a major sanctuary site for metastatic cancer cells that evade systemic therapies. Through pre-clinical pharmacological, biological, and molecular studies, we characterize the functional link between drug resistance and central nervous system (CNS) relapse in Epidermal Growth Factor Receptor- (EGFR-) mutant non-small cell lung cancer, which can progress in the brain when treated with the CNS-penetrant EGFR inhibitor osimertinib. Despite widespread osimertinib distribution in vivo, the brain microvascular tumor microenvironment (TME) is associated with the persistence of malignant cell sub-populations, which are poised to proliferate in the brain as osimertinib-resistant lesions over time. Cellular and molecular features of this poised state are regulated through a Ras homolog family member A (RhoA) and Serum Responsive Factor (SRF) gene expression program. RhoA potentiates the outgrowth of disseminated tumor cells on osimertinib treatment, preferentially in response to extracellular laminin and in the brain. Thus, we identify pre-existing and adaptive features of metastatic and drug-resistant cancer cells, which are enhanced by RhoA/SRF signaling and the brain TME during the evolution of osimertinib-resistant disease.
- Published
- 2021
16. An Investigation into the Prediction of the Plasma Concentration-Time Profile and Its Interindividual Variability for a Range of Flavin-Containing Monooxygenase Substrates Using a Physiologically Based Pharmacokinetic Modeling Approach
- Author
-
Joanne Wilson, Nicola Colclough, Danxi Li, Venkatesh Pilla Reddy, Abhishek Srivastava, and Barry Jones
- Subjects
Adult ,Male ,Physiologically based pharmacokinetic modelling ,Time Factors ,Adolescent ,Metabolic Clearance Rate ,Pharmacokinetic modeling ,Pharmaceutical Science ,Flavin-containing monooxygenase ,Models, Biological ,030226 pharmacology & pharmacy ,Substrate Specificity ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,In vivo ,Humans ,Aged ,Aged, 80 and over ,Pharmacology ,Chromatography ,Chemistry ,Middle Aged ,Monooxygenase ,030220 oncology & carcinogenesis ,Plasma concentration ,Hepatocytes ,Microsomes, Liver ,Oxygenases ,Microsome ,Female ,Forecasting - Abstract
Our recent paper demonstrated the ability to predict in vivo clearance of flavin-containing monooxygenase (FMO) drug substrates using in vitro human hepatocyte and human liver microsomal intrinsic clearance with standard scaling approaches. In this paper, we apply a physiologically based pharmacokinetic (PBPK) modeling and simulation approach (MS seven of the nine compounds fell within 2-fold when human liver microsomal data were used. rFMO overpredicted the AUC by approximately 2.5-fold for three of the nine compounds. Applying a calculated intersystem extrapolation scalar or tissue-specific scalar for the rFMO data resulted in better prediction of clinical data. The PBPK M&S results from this study demonstrate that human hepatocytes and human liver microsomes can be used along with our standard scaling approaches to predict human in vivo pharmacokinetic parameters for FMO substrates.
- Published
- 2018
- Full Text
- View/download PDF
17. The Identification of Potent, Selective, and Orally Available Inhibitors of Ataxia Telangiectasia Mutated (ATM) Kinase: The Discovery of AZD0156 (8-{6-[3-(Dimethylamino)propoxy]pyridin-3-yl}-3-methyl-1-(tetrahydro-2H-pyran-4-yl)-1,3-dihydro-2H-imidazo[4,5-c]quinolin-2-one)
- Author
-
Camila de-Almeida, Keith R. Mulholland, Kang Zhao, Barlaam Bernard Christophe, Gilles Ouvry, Gareth Hughes, Martin Pass, Elaine Cadogan, Zhenhua Wang, Andrew D. Campbell, Nidal Al-Huniti, Natalie Stratton, Sébastien L. Degorce, Joanne Wilson, Myriam Didelot, Philip A. MacFaul, Stephen T. Durant, Richard Ducray, Baochang Zhai, Kurt Gordon Pike, Lorraine A. Hassall, Jane L. Holmes, Thomas M. McGuire, Nichola L. Davies, Allan Dishington, Yingxue Chen, Nicola Colclough, and Graeme R. Robb
- Subjects
Models, Molecular ,0301 basic medicine ,Protein Conformation ,Pyridines ,Administration, Oral ,Biological Availability ,Ataxia Telangiectasia Mutated Proteins ,Quinolones ,Pharmacology ,Substrate Specificity ,Olaparib ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Potency ,Protein Kinase Inhibitors ,Volume of distribution ,Chemistry ,Atm kinase ,Irinotecan ,030104 developmental biology ,Pyran ,Drug Design ,030220 oncology & carcinogenesis ,Quinolines ,Molecular Medicine ,medicine.drug - Abstract
ATM inhibitors, such as 7, have demonstrated the antitumor potential of ATM inhibition when combined with DNA double-strand break-inducing agents in mouse xenograft models. However, the properties of 7 result in a relatively high predicted clinically efficacious dose. In an attempt to minimize attrition during clinical development, we sought to identify ATM inhibitors with a low predicted clinical dose (50 mg) and focused on strategies to increase both ATM potency and predicted human pharmacokinetic half-life (predominantly through the increase of volume of distribution). These efforts resulted in the discovery of 64 (AZD0156), an exceptionally potent and selective inhibitor of ATM based on an imidazo[4,5- c]quinolin-2-one core. 64 has good preclinical phamacokinetics, a low predicted clinical dose, and a high maximum absorbable dose. 64 has been shown to potentiate the efficacy of the approved drugs irinotecan and olaparib in disease relevant mouse models and is currently undergoing clinical evaluation with these agents.
- Published
- 2018
- Full Text
- View/download PDF
18. Further Considerations Towards an Effective and Efficient Oncology Drug Discovery DMPK Strategy
- Author
-
Barry Jones, Beth Williamson, Nicola Colclough, Rhys Dafydd Owen Jones, Adrian J. Fretland, and Dermot F. McGinnity
- Subjects
Pharmacology ,Drug ,business.industry ,Drug discovery ,media_common.quotation_subject ,In silico ,Clinical Biochemistry ,Drug Evaluation, Preclinical ,Phases of clinical research ,Administration, Oral ,Antineoplastic Agents ,Computational biology ,Metabolic stability ,Intestinal Absorption ,Drug Discovery ,Dose escalation ,Medicine ,Oncology drug ,Animals ,Humans ,Drug Interactions ,business ,ADME ,media_common - Abstract
Background: DMPK data and knowledge are critical in maximising the probability of developing successful drugs via the application of in silico, in vitro and in vivo approaches in drug discovery. Methods: The evaluation, optimisation and prediction of human pharmacokinetics is now a mainstay within drug discovery. These elements are at the heart of the ‘right tissue’ component of AstraZeneca’s ‘5Rs framework’ which, since its adoption, has resulted in increased success of Phase III clinical trials. With the plethora of DMPK related assays and models available, there is a need to continually refine and improve the effectiveness and efficiency of approaches best to facilitate the progression of quality compounds for human clinical testing. Results: This article builds on previously published strategies from our laboratories, highlighting recent discoveries and successes, that brings our AstraZeneca Oncology DMPK strategy up to date. We review the core aspects of DMPK in Oncology drug discovery and highlight data recently generated in our laboratories that have influenced our screening cascade and experimental design. We present data and our experiences of employing cassette animal PK, as well as re-evaluating in vitro assay design for metabolic stability assessments and expanding our use of freshly excised animal and human tissue to best inform first time in human dosing and dose escalation studies. Conclusion: Application of our updated drug-drug interaction and central nervous system drug exposure strategies are exemplified, as is the impact of physiologically based pharmacokinetic and pharmacokinetic-pharmacodynamic modelling for human predictions.
- Published
- 2019
19. Utilization of Structure-Based Design to Identify Novel, Irreversible Inhibitors of EGFR Harboring the T790M Mutation
- Author
-
Darren Cross, Lakshmaiah Gingipalli, Teresa Klinowska, Cath Eberlein, Nicola Colclough, Edward J. Hennessy, Jonathan P. Orme, Li Sha, Claudio Chuaqui, Xiaoyun Wu, Judit E. Debreczeni, and Susan Ashton
- Subjects
0301 basic medicine ,Mutation ,Kinase ,Organic Chemistry ,Autophosphorylation ,Mutant ,Biology ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,T790M ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Drug Discovery ,medicine ,Cancer research ,biology.protein ,Epidermal growth factor receptor ,EGFR inhibitors - Abstract
A novel series of covalent inhibitors of EGFR (epidermal growth factor receptor) kinase was discovered through a combination of subset screening and structure-based design. These compounds preferentially inhibit mutant forms of EGFR (activating mutant and T790M mutant) over wild-type EGFR in cellular assays measuring EGFR autophosphorylation and proliferation, suggesting an improved therapeutic index in non-small cell lung cancer patients would be achievable relative to established EGFR inhibitors. We describe our design approaches, resulting in the identification of the lead compound 5, and our efforts to develop an understanding of the structure-activity relationships within this series. In addition, strategies to overcome challenges around metabolic stability and aqueous solubility are discussed. Despite limitations in its physical properties, 5 is orally bioavailable in mice and demonstrates pronounced antitumor activity in in vivo models of mutant EGFR-driven cancers.
- Published
- 2016
- Full Text
- View/download PDF
20. Building on the success of osimertinib: achieving CNS exposure in oncology drug discovery
- Author
-
Nicola Colclough, Dermot F. McGinnity, Kan Chen, Peter Johnström, and Markus Fridén
- Subjects
0301 basic medicine ,Antineoplastic Agents ,Bioinformatics ,Piperazines ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Medicine ,Animals ,Humans ,Osimertinib ,Pharmaceutical sciences ,Protein Kinase Inhibitors ,Pharmacology ,Acrylamides ,Aniline Compounds ,business.industry ,Drug discovery ,Brain Neoplasms ,Brain ,Limiting ,Preclinical data ,030104 developmental biology ,030220 oncology & carcinogenesis ,Quinazolines ,Oncology drug ,business - Abstract
Due to the blood-brain barrier (BBB) limiting the exposure of therapeutics to the central nervous system (CNS), patients with brain malignancies are challenging to treat, typically have poor prognoses, and represent a significant unmet medical need. Preclinical data report osimertinib to have significant BBB penetration and emerging clinical data demonstrate encouraging activity against CNS malignancies. Here, we discuss the oncology drug candidates AZD3759 and AZD1390 as case examples of discovery projects designing in BBB penetrance. We demonstrate how these innovative kinase inhibitors were recognized as brain penetrant and outline our view of experimental approaches and strategies that can facilitate the discovery of new brain-penetrant therapies for the treatment of primary and secondary CNS malignancies as well as other CNS disorders.
- Published
- 2018
21. Discovery of a Series of 3-Cinnoline Carboxamides as Orally Bioavailable, Highly Potent, and Selective ATM Inhibitors
- Author
-
Allan Dishington, Guohong Xin, Nicola Colclough, Stephen T. Durant, Anil Patel, Stuart E. Pearson, Lorraine A. Hassall, Kurt Gordon Pike, Kristin Goldberg, Thomas M. McGuire, Andrew D. Campbell, Baochang Zhai, Jens Petersen, Gareth Hughes, Elaine Cadogan, Barlaam Bernard Christophe, Natalie Stratton, Graeme R. Robb, Martin Pass, and Philip A. MacFaul
- Subjects
0301 basic medicine ,Kinase ,Organic Chemistry ,Cell ,Pharmacology ,Biochemistry ,Irinotecan ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Pharmacokinetics ,chemistry ,In vivo ,030220 oncology & carcinogenesis ,Drug Discovery ,medicine ,Potency ,IC50 ,Cinnoline ,medicine.drug - Abstract
[Image: see text] We report the discovery of a novel series of 3-cinnoline carboxamides as highly potent and selective ataxia telangiectasia mutated (ATM) kinase inhibitors. Optimization of this series focusing on potency and physicochemical properties (especially permeability) led to the identification of compound 21, a highly potent ATM inhibitor (ATM cell IC(50) 0.0028 μM) with excellent kinase selectivity and favorable physicochemical and pharmacokinetics properties. In vivo, 21 in combination with irinotecan showed tumor regression in the SW620 colorectal tumor xenograft model, superior inhibition to irinotecan alone. Compound 21 was selected for preclinical evaluation alongside AZD0156.
- Published
- 2018
22. The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models
- Author
-
Joanne Wilson, Kristoffer Valerie, Stephen T. Durant, Caroline Roberts, Antonio García Trinidad, Li Zheng, Jacqueline H. L. Fok, Nicola Colclough, Jonathan Stott, Jasmine Allen, Venkatesh Pilla Reddy, Melissa Chapman, Thomas Anthony Hunt, Stephanie Ling, Gemma N Jones, Lucy Riches, Jenna M. Kahn, Tianwei Zhang, Katarina Varnäs, Amrita Sule, Kan Chen, Ruth Illingworth, Ian P. Barrett, Jeremy Karlin, Andrew J. Pierce, Anna Cronin, Andrew Sykes, Peter Johnström, Aaron Smith, Yingchun Wang, Lingli Zhang, Zhenfan Yang, Martin Pass, Annika Janefeldt, Jonathan P. Orme, Akihiro Takano, Martine P. Roudier, and Kurt Gordon Pike
- Subjects
0301 basic medicine ,Radiation-Sensitizing Agents ,Radiosensitizer ,Cell Membrane Permeability ,Cell ,Drug Evaluation, Preclinical ,Brain tumor ,Apoptosis ,Ataxia Telangiectasia Mutated Proteins ,Radiation Tolerance ,Cell cycle phase ,Mice ,03 medical and health sciences ,In vivo ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Multidisciplinary ,Brain Neoplasms ,Chemistry ,Kinase ,X-Rays ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Blood-Brain Barrier ,Cancer research ,Tumor Suppressor Protein p53 ,Signal Transduction - Abstract
Poor survival rates of patients with tumors arising from or disseminating into the brain are attributed to an inability to excise all tumor tissue (if operable), a lack of blood-brain barrier (BBB) penetration of chemotherapies/targeted agents, and an intrinsic tumor radio-/chemo-resistance. Ataxia-telangiectasia mutated (ATM) protein orchestrates the cellular DNA damage response (DDR) to cytotoxic DNA double-strand breaks induced by ionizing radiation (IR). ATM genetic ablation or pharmacological inhibition results in tumor cell hypersensitivity to IR. We report the primary pharmacology of the clinical-grade, exquisitely potent (cell IC50, 0.78 nM), highly selective [>10,000-fold over kinases within the same phosphatidylinositol 3-kinase–related kinase (PIKK) family], orally bioavailable ATM inhibitor AZD1390 specifically optimized for BBB penetration confirmed in cynomolgus monkey brain positron emission tomography (PET) imaging of microdosed 11C-labeled AZD1390 (Kp,uu, 0.33). AZD1390 blocks ATM-dependent DDR pathway activity and combines with radiation to induce G2 cell cycle phase accumulation, micronuclei, and apoptosis. AZD1390 radiosensitizes glioma and lung cancer cell lines, with p53 mutant glioma cells generally being more radiosensitized than wild type. In in vivo syngeneic and patient-derived glioma as well as orthotopic lung-brain metastatic models, AZD1390 dosed in combination with daily fractions of IR (whole-brain or stereotactic radiotherapy) significantly induced tumor regressions and increased animal survival compared to IR treatment alone. We established a pharmacokinetic-pharmacodynamic-efficacy relationship by correlating free brain concentrations, tumor phospho-ATM/phospho-Rad50 inhibition, apoptotic biomarker (cleaved caspase-3) induction, tumor regression, and survival. On the basis of the data presented here, AZD1390 is now in early clinical development for use as a radiosensitizer in central nervous system malignancies.
- Published
- 2018
- Full Text
- View/download PDF
23. Utilizing microphysiological systems and induced pluripotent stem cells for disease modeling: a case study for blood brain barrier research in a pharmaceutical setting
- Author
-
Damian C. Crowther, Ryan Hicks, Lorna Ewart, Nicola Colclough, Kristin M. Fabre, and Louise Delsing
- Subjects
Drug ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Pharmaceutical Science ,02 engineering and technology ,Disease ,Blood–brain barrier ,Models, Biological ,Translational Research, Biomedical ,03 medical and health sciences ,Drug Discovery ,Medicine ,Animals ,Humans ,Induced pluripotent stem cell ,030304 developmental biology ,Pharmaceutical industry ,media_common ,0303 health sciences ,Drug discovery ,business.industry ,021001 nanoscience & nanotechnology ,medicine.anatomical_structure ,Drug development ,Blood-Brain Barrier ,Stem cell ,0210 nano-technology ,business ,Neuroscience - Abstract
Microphysiological systems (MPS) may be able to provide the pharmaceutical industry models that can reflect human physiological responses to improve drug discovery and translational outcomes. With lack of efficacy being the primary cause for drug attrition, developing MPS disease models would help researchers identify novel targets, study mechanisms in more physiologically-relevant depth, screen for novel biomarkers and test/optimize various therapeutics (small molecules, nanoparticles and biologics). Furthermore, with advances in inducible pluripotent stem cell technology (iPSC), pharmaceutical companies can access cells from patients to help recreate specific disease phenotypes in MPS platforms. Combining iPSC and MPS technologies will contribute to our understanding of the complexities of neurodegenerative diseases and of the blood brain barrier (BBB) leading to development of enhanced therapeutics.
- Published
- 2018
24. Using quantitative structure activity relationship models to predict an appropriate solvent system from a common solvent system family for countercurrent chromatography separation
- Author
-
Svetlana Ignatova, Nicola Colclough, Siân Marsden-Jones, Ian Garrard, and Neil Sumner
- Subjects
Quantitative structure–activity relationship ,Chromatography ,Molecular Structure ,Chemistry ,Organic Chemistry ,Analytical chemistry ,Quantitative Structure-Activity Relationship ,General Medicine ,Models, Theoretical ,Biochemistry ,Chemistry Techniques, Analytical ,Analytical Chemistry ,Solvent ,Partition coefficient ,Countercurrent chromatography ,Column chromatography ,Molecular property ,Phase (matter) ,Solvents ,Chromatography column ,Countercurrent Distribution - Abstract
Countercurrent chromatography (CCC) is a form of liquid–liquid chromatography. It works by running one immiscible solvent (mobile phase) over another solvent (stationary phase) being held in a CCC column using centrifugal force. The concentration of compound in each phase is characterised by the partition coefficient ( K d ), which is the concentration in the stationary phase divided by the concentration in the mobile phase. When K d is between approximately 0.2 and 2, it is most likely that optimal separation will be achieved. Having the K d in this range allows the compound enough time in the column to be separated without resulting in a broad peak and long run time. In this paper we report the development of quantitative structure activity relationship (QSAR) models to predict log K d . The QSAR models use only the molecule's 2D structure to predict the molecular property log K d .
- Published
- 2015
- Full Text
- View/download PDF
25. Discovery and Optimization of a Novel Series of Dyrk1B Kinase Inhibitors To Explore a MEK Resistance Hypothesis
- Author
-
Susan E. Critchlow, Judit E. Debreczeni, Louise Goodwin, Shaun M. Fillery, Gary Fairley, David Whittaker, Nicola Colclough, Richard A. Ward, Mark A. Graham, Catherine Bardelle, Jason Grant Kettle, Kevin Hudson, Sylvie Guichard, Peter Ballard, and Mark Cockerill
- Subjects
Models, Molecular ,DYRK1B ,Drug Resistance ,Drug resistance ,Protein Serine-Threonine Kinases ,RNA interference ,Cell Line, Tumor ,Chlorocebus aethiops ,Drug Discovery ,Animals ,Humans ,RNA, Small Interfering ,Rats, Wistar ,Protein Kinase Inhibitors ,COS cells ,Chemistry ,Kinase ,Drug discovery ,Mechanism (biology) ,Protein-Tyrosine Kinases ,MAP Kinase Kinase Kinases ,Biochemistry ,Cell culture ,COS Cells ,Cancer research ,Molecular Medicine ,RNA Interference - Abstract
Potent and selective inhibitors of Dyrk1B kinase were developed to explore the hypothesis, based on siRNA studies, that Dyrk1B may be a resistance mechanism in cells undergoing a stress response.
- Published
- 2015
- Full Text
- View/download PDF
26. Orally Bioavailable and Blood-Brain Barrier-Penetrating ATM Inhibitor (AZ32) Radiosensitizes Intracranial Gliomas in Mice
- Author
-
Ian P. Barrett, Li Zheng, Jason M. Beckta, Jasmine Allen, Antonio Garcia-Trinidad, Aaron Smith, Yingchun Wang, Amrita Sule, Joanne Wilson, Paul Farrington, Andrew G. Thomason, Stephen T. Durant, Victoria Sheridan, Jeremy Karlin, Jenna M. Kahn, Nitai D. Mukhopadhyay, Tianwei Zhang, Jason Grant Kettle, Syed Farhan Ahmad, Nicholas C.K. Valerie, Kan Chen, Barlaam Bernard Christophe, Kurt Gordon Pike, Lucy Riches, Rajesh Odedra, Gareth Hughes, Laura Biddlestone-Thorpe, Kristoffer Valerie, Elaine Cadogan, Martin Pass, Sébastien L. Degorce, Alan Lau, Jennifer L. Vincent, Nicola Colclough, Thomas Anthony Hunt, and Bhavika Patel
- Subjects
0301 basic medicine ,Cancer Research ,Radiosensitizer ,Radiation-Sensitizing Agents ,DNA damage ,Administration, Oral ,Mice, Nude ,Ataxia Telangiectasia Mutated Proteins ,Blood–brain barrier ,Article ,03 medical and health sciences ,Mice ,Therapeutic index ,In vivo ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Mitotic catastrophe ,Protein Kinase Inhibitors ,business.industry ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,Blood-Brain Barrier ,Cancer research ,business - Abstract
Inhibition of ataxia-telangiectasia mutated (ATM) during radiotherapy of glioblastoma multiforme (GBM) may improve tumor control by short-circuiting the response to radiation-induced DNA damage. A major impediment for clinical implementation is that current inhibitors have limited central nervous system (CNS) bioavailability; thus, the goal was to identify ATM inhibitors (ATMi) with improved CNS penetration. Drug screens and refinement of lead compounds identified AZ31 and AZ32. The compounds were then tested in vivo for efficacy and impact on tumor and healthy brain. Both AZ31 and AZ32 blocked the DNA damage response and radiosensitized GBM cells in vitro. AZ32, with enhanced blood–brain barrier (BBB) penetration, was highly efficient in vivo as radiosensitizer in syngeneic and human, orthotopic mouse glioma model compared with AZ31. Furthermore, human glioma cell lines expressing mutant p53 or having checkpoint-defective mutations were particularly sensitive to ATMi radiosensitization. The mechanism for this p53 effect involves a propensity to undergo mitotic catastrophe relative to cells with wild-type p53. In vivo, apoptosis was >6-fold higher in tumor relative to healthy brain after exposure to AZ32 and low-dose radiation. AZ32 is the first ATMi with oral bioavailability shown to radiosensitize glioma and improve survival in orthotopic mouse models. These findings support the development of a clinical-grade, BBB-penetrating ATMi for the treatment of GBM. Importantly, because many GBMs have defective p53 signaling, the use of an ATMi concurrent with standard radiotherapy is expected to be cancer-specific, increase the therapeutic ratio, and maintain full therapeutic effect at lower radiation doses. Mol Cancer Ther; 17(8); 1637–47. ©2018 AACR.
- Published
- 2017
27. An Investigation into the Prediction of in Vivo Clearance for a Range of Flavin-containing Monooxygenase Substrates
- Author
-
Danxi Li, Venkatesh Pilla Reddy, Abhishek Srivastava, Barry Jones, Nicola Colclough, Sara Amberntsson, and Joanne Wilson
- Subjects
Male ,Imipramine ,Dinitrocresols ,Metabolic Clearance Rate ,Pharmaceutical Science ,Flavin-containing monooxygenase ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,Cytochrome P-450 Enzyme System ,In vivo ,Benzyl Compounds ,Humans ,Pharmacology ,chemistry.chemical_classification ,biology ,In vitro toxicology ,Cytochrome P450 ,Substrate (chemistry) ,Monooxygenase ,Kinetics ,Enzyme ,Biochemistry ,chemistry ,Liver ,030220 oncology & carcinogenesis ,Benzamides ,biology.protein ,Microsome ,Hepatocytes ,Microsomes, Liver ,Female ,Aryl Hydrocarbon Hydroxylases ,Oxidation-Reduction - Abstract
Flavin-containing monooxygenases (FMO) are metabolic enzymes mediating the oxygenation of nucleophilic atoms such as nitrogen, sulfur, phosphorus, and selenium. These enzymes share similar properties to the cytochrome P450 system but can be differentiated through heat inactivation and selective substrate inhibition by methimazole. This study investigated 10 compounds with varying degrees of FMO involvement to determine the nature of the correlation between human in vitro and in vivo unbound intrinsic clearance. To confirm and quantify the extent of FMO involvement six of the compounds were investigated in human liver microsomal (HLM) in vitro assays using heat inactivation and methimazole substrate inhibition. Under these conditions FMO contribution varied from 21% (imipramine) to 96% (itopride). Human hepatocyte and HLM intrinsic clearance (CLint) data were scaled using standard methods to determine the predicted unbound intrinsic clearance (predicted CLint u) for each compound. This was compared with observed unbound intrinsic clearance (observed CLint u) values back calculated from human pharmacokinetic studies. A good correlation was observed between the predicted and observed CLint u using hepatocytes (R2 = 0.69), with 8 of the 10 compounds investigated within or close to a factor of 2. For HLM the in vitro-in vivo correlation was maintained (R2 = 0.84) but the accuracy was reduced with only 3 out of 10 compounds falling within, or close to, twofold. This study demonstrates that human hepatocytes and HLM can be used with standard scaling approaches to predict the human in vivo clearance for FMO substrates.
- Published
- 2017
28. P58 - Utilising a dual human transporter MDCK_MDR1_BCRP cell line to assess efflux at the blood brain barrier
- Author
-
Ravindra Alluri, Nicola Colclough, Yumei Yan, Danxi Li, Kan Chen, and Dermot F. McGinnity
- Subjects
Pharmacology ,medicine.anatomical_structure ,Cell culture ,Chemistry ,medicine ,Pharmaceutical Science ,Pharmacology (medical) ,Transporter ,Efflux ,Blood–brain barrier ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
29. Abstract 4813: Comparative activity profiling of tyrosine kinase inhibitors (TKIs) against exon 20 insertions and the wild-type form of epidermal growth factor receptor (EGFR)
- Author
-
Martina Fitzek, Jelena Urosevic, Emanuela M. Cuomo, Darren Mckerrecher, Ambra Bianco, Nicolas Floc'h, Sladjana Gagrica, M. Raymond V. Finlay, Matthew J. Martin, Beverley Hammond, Richard A. Ward, Jonathan P. Orme, Darren Cross, Paul D. Smith, James W.T. Yates, Nicky Whalley, Nicola Colclough, Daniel O'Neill, and Anna Staniszewska
- Subjects
0301 basic medicine ,Cancer Research ,Cetuximab ,biology ,business.industry ,Afatinib ,Poziotinib ,Cancer ,medicine.disease ,03 medical and health sciences ,Exon ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,Osimertinib ,Epidermal growth factor receptor ,business ,Tyrosine kinase ,medicine.drug - Abstract
Exon 20 insertions (Ex20Ins) have been identified in approximately 5% of epidermal growth factor receptor (EGFR)-mutated lung tumours in patients presenting with non-small cell lung cancer (NSCLC). Several small molecule tyrosine kinase inhibitors (TKIs) have been reported to have pre-clinical activity against such insertions including afatinib, poziotinib, osimertinib, nazartinib, AP32788/TAK-788 and TAS6417. However, there remains a lack of approved treatments for patients with Ex20Ins with early approved EGFR agents appearing to be ineffective in this setting. Poziotinib, osimertinib and AP32788/TAK-788 are undergoing clinical evaluation in patients whose tumours carry Ex20Ins and in some cases clinical responses have been reported giving hope that such insertions can be targeted by small molecules. There is however a need for comparable data across such compounds that would enable understanding of the relative activity of these compounds between Ex20Ins and the wild-type form of EGFR. As many of the Exon 20 insertions are not part of the ATP binding pocket achieving selectivity over wild type EGFR is highly challenging and may limit the clinical utility of agents due to dose limiting EGFR wild-type driven toxicity. A selection of TKIs were profiled for Ex20Ins and wild-type EGFR activity using biochemical, in vitro cellular phosphorylation and proliferation assays. This has enabled us to differentiate the Ex20Ins versus wild-type EGFR selectivity profiles of a range of pre-clinical, clinical and proprietary compounds. As part of this evaluation we utilized a CRISPR CAS9 approach in H2073 EGFR wild-type NSCLC cell line, where we have established cellular disease models against the most prevalent insertions including D770-N771insSVD (22%). Finally, we will show anti-tumour efficacy data for a selection of these inhibitors along with a potential combination approach of osimertinib and cetuximab. EGFR D770-N771InsSVD cell phospho IC50 (µM)EGFR WT (H2073) cell phospho IC50 (µM)Fold-EGFR WT margin (cell)afatinib0.0060.00330.5osimertinib0.0940.414.4poziotinib0.00340.00351TAS64170.0230.0672.9AZ62810.0972.223 Citation Format: Richard A. Ward, Ambra Bianco, Nicola Colclough, Darren Cross, Emanuela M. Cuomo, M. Raymond V. Finlay, Martina Fitzek, Nicolas Floc’h, Sladjana Gagrica, Beverley Hammond, Matthew J. Martin, Darren McKerrecher, Daniel J. O’Neill, Jonathan P. Orme, Paul D. Smith, Anna D. Staniszewska, Jelena Urosevic, Nicky Whalley, James W. Yates. Comparative activity profiling of tyrosine kinase inhibitors (TKIs) against exon 20 insertions and the wild-type form of epidermal growth factor receptor (EGFR) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4813.
- Published
- 2019
- Full Text
- View/download PDF
30. Abstract 4868: A preclinical PK/PD model based on a mouse glioblastoma survival model for AZD1390, a novel, brain-penetrant ATM kinase inhibitor, to predict the inhibition of DNA damage response induced by radiation and the human efficacious dose
- Author
-
Stephen T. Durant, Lenka Oplustil O'Connor, Andy Sykes, Matthias Hoch, Serena De Vita, Nicola Colclough, Nuria Buil Bruna, Martin Pass, Melinda S. Merchant, and Venkatesh Pilla Reddy
- Subjects
0301 basic medicine ,Cancer Research ,Kinase ,business.industry ,medicine.medical_treatment ,Brain tumor ,Cancer ,Cell cycle ,medicine.disease ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,business ,PK/PD models - Abstract
AZD1390 is a novel, highly selective, brain-penetrant, potent inhibitor of the Ataxia Telangiectasia Mutated (ATM) kinase. ATM is activated in response to double-strand breaks (DSBs) and coordinates cellular responses to ionising radiation and other insults. Radiation is the mainstay of treatment for patients with brain tumors, and glioma cells are exquisitely sensitive to ATM inhibition. Therefore, AZD1390 is in clinical development in combination with radiation therapy for the treatment of patients with Glioblastoma Multiforme (GBM) and brain metastases from solid tumours (NCT03423628). [1] The aim of this work was to develop a translational PK/PD-efficacy model for AZD1390 that would enable the project team to assess the extent/duration of inhibition of target engagement required and help to predict the optimal dose and regimen of AZD1390 to treat patients with brain malignancies in combination with radiation. Time-course data of pharmacokinetics (PK), pharmacodynamics (PD) and tumor growth inhibition from mouse NCI-H2228 brain tumor orthotopic model studies were used to build a PK-PD/Efficacy or survival model. ATM activation is induced by IR treatment, becoming transiently phosphorylated within minutes of IR exposure and dissipates over a 24-hour period. This model quantitatively and dynamically integrates AZD1390 brain PK to the rate and extent of inhibition of phosphorylation of ATM, a proximal PD marker of ATM kinase activation caused by radiation induced DNA damage in the tumor and rate of induction of cell death (determined by bioluminescence signalling as a measure of tumor growth). In parallel, we also modelled cell cycle profiles in GBM cell lines to complement the in-vivo modelling work. The pATM time-course relative to IR radiation (2 Gy) was modelled using AZD1390 concentrations in the brain tumor and the corresponding pATM inhibition, which was then linked to efficacy by estimating tumor cell kill rate via exponential tumor growth model. The free brain concentration of AZD1390 that resulted in half-maximal inhibition (EC50) of pATM after radiation was estimated to be 0.8 nM (range 0.4 to 1.6 nM). Significant tumor regression in orthotopic tumor model was observed at doses >5 mg/kg. An average pATM inhibition over 24h in the mouse GBM survival model was in the range of 44% (5 mg/kg QD) to 88% (20 mg/kg BD). The % overall survival at 5 mg/kg QD and 20 mg/kg BD dose were 56% and 100%, respectively. Conclusion: The translation of mouse survival data to clinical schedules is unknown, thus efficacious dose was anchored to pATM inhibition required to deliver significant tumor regression. References: 1. Durant ST et al., The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. Sci. Adv. 2018;4. Citation Format: Venkatesh Pilla Reddy, Andy Sykes, Nicola Colclough, Stephen T. Durant, Lenka Oplustil O'Connor, Matthias Hoch, Nuria Buil Bruna, Serena De Vita, Melinda Merchant, Martin Pass. A preclinical PK/PD model based on a mouse glioblastoma survival model for AZD1390, a novel, brain-penetrant ATM kinase inhibitor, to predict the inhibition of DNA damage response induced by radiation and the human efficacious dose [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4868.
- Published
- 2019
- Full Text
- View/download PDF
31. Abstract 4451: Evaluation of the therapeutic potential of phosphine oxide pyrazole inhibitors in tumors harboring EGFR C797S mutation
- Author
-
Nicolas Floch, M. Raymond V. Finlay, Ambra Bianco, Sue Bickerton, Nicola Colclough, Darren A. Cross, Emanuela M. Cuomo, Carine M. Guerot, David Hargreaves, Matthew J. Martin, Darren McKerrecher, Daniel J. O’Neill, Jonathan P. Orme, Amar Rahi, Paul D. Smith, and Richard A. Ward
- Subjects
Cancer Research ,Oncology - Abstract
Osimertinib is a next-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) with activity against both the activating and the ‘gatekeeper’ T790M EGFR mutations. An acquired EGFR C797S mutation has been reported to mediate osimertinib resistance in approximately 15% and 7% of patients in second-line and first-line treatment respectively. This percentage in the first-line setting will likely evolve as the first line data mature. The C797S mutation leads to the loss of covalent binding of osimertinib to mutant EGFR. The high affinity of the EGFR triple mutant for ATP presents a challenge for reversible inhibitor design, particularly as the loss of the cysteine at position 797 precludes the previously exploited covalent approaches. We have explored various approaches to address this challenge, including an effort to maximise reversible affinity to target the C797S mutation without requiring a covalent bond. We describe herein the therapeutic potential of reversible phosphine oxide pyrazole inhibitors in tumors harboring C797S. Using structure-based design, we were able to design a series of phosphine oxide pyrazole inhibitors that displayed exceptionally high biochemical potency against EGFR C797S mutation, which translated into good activity in cell-based assays. Using CRISPR-Cas 9 genome editing technology, we engineered cellular disease-relevant models to express the C797S mutation to evaluate potency in vitro and in vivo. By modulating the physicochemical properties of our in vitro leads, we were able to achieve good oral exposure of cellularly active EGFR C797S inhibitors such as AZ’7608. We showed that AZ’7608 inhibits signalling pathways and cellular growth of C797S EGFR cell lines in vitro and demonstrated an improved WT EGFR margin. This translated into 52% (p The work presented herein shows a proof of concept for reversible phosphine oxide pyrazole inhibitors to target tumors harboring C797S. The emergence of the C797S EGFR mutation remains a key area of unmet need and warrants further efforts in drug discovery. Citation Format: Nicolas Floch, M. Raymond V. Finlay, Ambra Bianco, Sue Bickerton, Nicola Colclough, Darren A. Cross, Emanuela M. Cuomo, Carine M. Guerot, David Hargreaves, Matthew J. Martin, Darren McKerrecher, Daniel J. O’Neill, Jonathan P. Orme, Amar Rahi, Paul D. Smith, Richard A. Ward. Evaluation of the therapeutic potential of phosphine oxide pyrazole inhibitors in tumors harboring EGFR C797S mutation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4451.
- Published
- 2019
- Full Text
- View/download PDF
32. Species differences in drug plasma protein binding
- Author
-
J. Matthew Wood, Philip A. MacFaul, Nicola Colclough, and Linette Ruston
- Subjects
Pharmacology ,Drug ,Drug discovery ,media_common.quotation_subject ,Organic Chemistry ,Pharmaceutical Science ,Plasma protein binding ,Biology ,Biochemistry ,Blood proteins ,Human plasma ,Drug Discovery ,Molecular Medicine ,media_common - Abstract
Comparison of the human plasma protein binding data for a variety of drug discovery compounds indicates that compounds tend to be slightly more bound to human plasma proteins, than compared to plasma proteins from rats, dogs or mice. However, the majority of measurements from the pre-clinical species fall within 5-fold of the human plasma value, although there are some compounds that do show significantly different interspecies plasma protein binding.
- Published
- 2014
- Full Text
- View/download PDF
33. Structure- and Reactivity-Based Development of Covalent Inhibitors of the Activating and Gatekeeper Mutant Forms of the Epidermal Growth Factor Receptor (EGFR)
- Author
-
Clare Lane, Nicola Colclough, Les A. Dakin, M. Raymond V. Finlay, Claudio Chuaqui, Mark J. Anderton, Christopher G. Chorley, Matthew Grist, Darren Cross, Richard A. Ward, Michael J. Waring, Teresa Klinowska, Jonathon P. Orme, Cath Eberlein, Judit E. Debreczeni, Scott W. Martin, Matthew R. Box, Peter D. Smith, Susan Ashton, Paul A. Bethel, Fengjiang Wang, Sam Butterworth, and George B. Hill
- Subjects
Models, Molecular ,biology ,Chemistry ,Mutant ,ErbB Receptors ,Structure-Activity Relationship ,Gefitinib ,Biochemistry ,Mutation ,Drug Discovery ,medicine ,biology.protein ,Molecular Medicine ,Structure–activity relationship ,Erlotinib ,Epidermal growth factor receptor ,Binding site ,Tyrosine kinase ,medicine.drug ,Cysteine - Abstract
A novel series of small-molecule inhibitors has been developed to target the double mutant form of the epidermal growth factor receptor (EGFR) tyrosine kinase, which is resistant to treatment with gefitinib and erlotinib. Our reported compounds also show selectivity over wild-type EGFR. Guided by molecular modeling, this series was evolved to target a cysteine residue in the ATP binding site via covalent bond formation and demonstrates high levels of activity in cellular models of the double mutant form of EGFR. In addition, these compounds show significant activity against the activating mutations, which gefitinib and erlotinib target and inhibition of which gives rise to their observed clinical efficacy. A glutathione (GSH)-based assay was used to measure thiol reactivity toward the electrophilic functionality of the inhibitor series, enabling both the identification of a suitable reactivity window for their potency and the development of a reactivity quantitative structure-property relationship (QSPR) to support design.
- Published
- 2013
- Full Text
- View/download PDF
34. Small-molecule androgen receptor downregulators as an approach to treatment of advanced prostate cancer
- Author
-
Nicola Colclough, David G. Acton, Alan Girdwood, Andrew A. Mortlock, David M. Andrews, Peter Ballard, Neil James Hales, Alfred A. Rabow, Graeme Walker, David A. Jude, Owen Jones, Nigel Brooks, Robert Hugh Bradbury, Urs Hancox, and Sarah A. Loddick
- Subjects
Male ,Models, Molecular ,Clinical Biochemistry ,Down-Regulation ,Pharmaceutical Science ,Pharmacology ,Ligands ,Biochemistry ,Structure-Activity Relationship ,Prostate cancer ,Downregulation and upregulation ,Oral administration ,Drug Discovery ,medicine ,Humans ,Structure–activity relationship ,Potency ,Receptor ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Organic Chemistry ,Prostatic Neoplasms ,Stereoisomerism ,medicine.disease ,Small molecule ,Molecular Weight ,Pyridazines ,Androgen receptor ,Receptors, Androgen ,Molecular Medicine - Abstract
Chemical starting points were investigated for downregulation of the androgen receptor as an approach to treatment of advanced prostate cancer. Although prototypic steroidal downregulators such as 6a designed for intramuscular administration showed insufficient cellular potency, a medicinal chemistry program derived from a novel androgen receptor ligand 8a led to 6-[4-(4-cyanobenzyl)piperazin-1-yl]-3-(trifluoromethyl)[1,2,4]triazolo[4,3-b]pyridazine (10b), for which high plasma levels following oral administration in a preclinical model compensate for moderate cellular potency.
- Published
- 2011
- Full Text
- View/download PDF
35. The discovery of benzanilides as c-Met receptor tyrosine kinase inhibitors by a directed screening approach
- Author
-
Christine Marie Paul Lambert, Alan Girdwood, Andrew G. Leach, Catherine Bardelle, Andrew P. Garner, Helen Plant, J. S. Major, Brian Law, Dave Buttar, Kevin Blades, Louise Chapman, Nicola Colclough, Joanne V. Allen, Anthony M. Slater, and Alexander G. Dossetter
- Subjects
Models, Molecular ,C-Met ,Molecular model ,Clinical Biochemistry ,Pharmaceutical Science ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Anilides ,Secondary metabolism ,Protein Kinase Inhibitors ,Molecular Biology ,Ligand efficiency ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Kinase ,Organic Chemistry ,Binding potential ,Stereoisomerism ,Proto-Oncogene Proteins c-met ,C-Met Receptor Tyrosine Kinase ,In vitro ,Molecular Medicine - Abstract
A directed screen of a relatively small number of compounds, selected for kinase ATP pocket binding potential, yielded a novel series of hit compounds (1). Hit explosion on two binding residues identified compounds 27 and 43 as the best leads for an optimization program having reduced secondary metabolism, as measured by in vitro rat hepatocytes incubation, leading to oral bio-availability. Structure–activity relationships and molecular modeling have suggested a binding mode for the most potent inhibitor 12.
- Published
- 2011
- Full Text
- View/download PDF
36. Gradient elution in counter-current chromatography: A new layout for an old path
- Author
-
Nicola Colclough, Neil Sumner, Svetlana Ignatova, and Ian Sutherland
- Subjects
Solvent system ,Chromatography ,Series (mathematics) ,Elution ,Chemistry ,Organic Chemistry ,General Medicine ,Biochemistry ,Method development ,Analytical Chemistry ,Countercurrent chromatography ,Pharmaceutical Preparations ,Stationary phase ,Path (graph theory) ,Gradient elution ,Countercurrent Distribution - Abstract
Gradient elution in CCC is a powerful tool, which needs further systematic development to become robust and easy to use. The first attempt to build a correlation between gradient elution profile and distribution ratio (KD) values for model mixtures containing typical representatives of pharmaceutical compounds is presented in this paper. The three step estimation of the solvent system composition of a heptane-ethyl acetate-methanol-water (HEMWat) series is described. The estimation is based on simple measurements of initial and final stationary phase retention for gradient elution run, calculating gradient distribution ratio and correlating it with static KD against HEMWat number. © 2011 Elsevier B.V. All rights reserved.
- Published
- 2011
- Full Text
- View/download PDF
37. A combined spectroscopic and crystallographic approach to probing drug–human serum albumin interactions
- Author
-
Stefan Gerhardt, Stefan Steinbacher, Paul Robert Owen Whittamore, Kin Yip Tam, Holger Steuber, Alleyn T. Plowright, Philip A. MacFaul, Klaus Maskos, Nicola Colclough, David Buttar, and Scott D. Phillips
- Subjects
Molecular model ,Stereochemistry ,Clinical Biochemistry ,Serum albumin ,Fluorescence spectrometry ,Pharmaceutical Science ,Plasma protein binding ,Crystallography, X-Ray ,Biochemistry ,Fluorescence spectroscopy ,Drug Discovery ,medicine ,Humans ,Drug Interactions ,Binding site ,Molecular Biology ,Serum Albumin ,Binding Sites ,biology ,Chemistry ,Organic Chemistry ,Human serum albumin ,Protein Structure, Tertiary ,Crystallography ,Spectrometry, Fluorescence ,Pharmaceutical Preparations ,Lipophilicity ,biology.protein ,Molecular Medicine ,Protein Binding ,medicine.drug - Abstract
The displacement of probes that bind selectively to subdomains IIA or IIIA on human serum albumin (HSA) by competing compounds has been followed using fluorescence spectroscopy, and has therefore been used to assign a primary binding site for these compounds in the presence and absence of fatty acids. The crystal structures have also been solved for three compounds: a matched pair of carboxylic acids whose binding strength to HSA unexpectedly decreased as the lipophilicity increased; and a highly bound sulphonamide that appeared not to displace the probes in the displacement assay. The crystallography results support the findings from the fluorescence displacement assay. The results indicate that drug binding to subdomain IB might also be important location for certain compounds.
- Published
- 2010
- Full Text
- View/download PDF
38. Aqueous Solubility in Drug Discovery Chemistry, DMPK, and Biological Assays
- Author
-
Linette Ruston, Kin Yip Tam, and Nicola Colclough
- Subjects
Chemistry ,Drug discovery ,Aqueous solubility ,Bioassay ,Organic chemistry ,Drug metabolism - Published
- 2008
- Full Text
- View/download PDF
39. Structure-Guided Design of Highly Selective and Potent Covalent Inhibitors of ERK1/2
- Author
-
Philip Hopcroft, Lyman Feron, Tina Howard, Kay Yeung, Mairi Challinor, Judit E. Debreczeni, Michael Tonge, Vikki Flemington, Lindsay Snow, Clifford David Jones, Ryan Greenwood, Michael James, Nicola Colclough, Mark A. Graham, Kay Eckersley, Gary Fairley, Christopher R. Jones, Karen Roberts, Richard A. Ward, and Jonathan Renshaw
- Subjects
Drug ,Models, Molecular ,media_common.quotation_subject ,Immunoblotting ,Molecular Sequence Data ,Crystallography, X-Ray ,Structure-Activity Relationship ,In vivo ,Drug Discovery ,medicine ,Structure–activity relationship ,Transferase ,Humans ,Amino Acid Sequence ,Protein Kinase Inhibitors ,Cells, Cultured ,media_common ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Molecular Structure ,Chemistry ,Small molecule ,In vitro ,Mechanism of action ,Biochemistry ,Covalent bond ,Drug Design ,Molecular Medicine ,medicine.symptom - Abstract
The RAS/RAF/MEK/ERK signaling pathway has been targeted with a number of small molecule inhibitors in oncology clinical development across multiple disease indications. Importantly, cell lines with acquired resistance to B-RAF and MEK inhibitors have been shown to maintain sensitivity to ERK1/2 inhibition by small molecule inhibitors. There are a number of selective, noncovalent ERK1/2 inhibitors reported along with the promiscuous hypothemycin (and related analogues) that act via a covalent mechanism of action. This article reports the identification of multiple series of highly selective covalent ERK1/2 inhibitors informed by structure-based drug design (SBDD). As a starting point for these covalent inhibitors, reported ERK1/2 inhibitors and a chemical series identified via high-throughput screening were exploited. These approaches resulted in the identification of selective covalent tool compounds for potential in vitro and in vivo studies to assess the risks and or benefits of targeting this pathway through such a mechanism of action.
- Published
- 2015
40. Matched Molecular Pairs as a Guide in the Optimization of Pharmaceutical Properties; a Study of Aqueous Solubility, Plasma Protein Binding and Oral Exposure
- Author
-
Philip A. MacFaul, David A. Cosgrove, J. Matthew Wood, Andrew G. Leach, H. Jones, Brian Law, Nicola Colclough, Peter W. Kenny, and Linette Ruston
- Subjects
Models, Molecular ,Property (philosophy) ,Databases, Factual ,Stereochemistry ,Heteroatom ,Administration, Oral ,Methylation ,Piperazines ,Structure-Activity Relationship ,Piperidines ,Computational chemistry ,Drug Discovery ,Animals ,Molecule ,Pharmacokinetics ,skin and connective tissue diseases ,Aqueous solution ,Molecular Structure ,Chemistry ,Water ,Hydrogen Bonding ,Aromaticity ,Blood Proteins ,Rats ,Pharmaceutical Preparations ,Solubility ,Structural change ,Drug Design ,Lipophilicity ,Molecular Medicine ,sense organs ,Matched molecular pair analysis ,Software ,Protein Binding - Abstract
By identifying every pair of molecules that differ only by a particular, well-defined, structural transformation in a database of measured properties and computing the corresponding change in property, we obtain an overview of the effect that structural change has upon the property and set an expectation for what will happen when that transformation is applied elsewhere. The mean change indicates the expected magnitude of the change in the property and the number of cases in which the property increases give the probability that the structural transformation will cause the property to increase. Outliers indicate potential ways of avoiding the general trend. Comparing to changes in lipophilicity highlights structural transformations that have unusual effects, some of which can be explained by conformational changes. In this paper, we focus upon the effects on aqueous solubility, plasma protein binding and oral exposure of adding substituents to aromatic rings and methylating heteroatoms.
- Published
- 2006
- Full Text
- View/download PDF
41. Abstract A104: AZD1390, a potent and selective orally bioavailable blood-brain barrier-penetrant ATM inhibitor, radiosensitizes and improves survival of orthotopic glioma and metastatic brain tumor models
- Author
-
Jonathan Stott, Antonio Garcia-Trinidad, Nicola Colclough, Stephanie Ling, Steve T. Durant, Kurt Gordon Pike, Martin Pass, Yingchun Wang, Venkatesh Pilla Reddy, Peter Johnström, Ruth Illingworth, Andrew J. Pierce, Ian P. Barrett, Gemma N Jones, Andrew Sykes, Lucy Riches, Jeremy Karlin, Tianwei Zhang, Kan Chen, Jenna M. Kahn, Li Zheng, Jasmine Allen, Thomas Anthony Hunt, and Kristoffer Valerie
- Subjects
0301 basic medicine ,Cancer Research ,Radiosensitizer ,Temozolomide ,business.industry ,Brain tumor ,medicine.disease ,Blood–brain barrier ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,Stem cell ,Clonogenic assay ,business ,medicine.drug - Abstract
ATM plays a central role in the detection, signalling, and repair of DNA double-strand breaks (DSB), the most cytotoxic lesion induced by ionizing radiation (IR) and certain chemotherapies. ATM is also activated by reactive oxygen species (ROS) induced by cellular exposures to IR. Genetic ablation and pharmacologic inhibition of ATM is associated with extreme hypersensitivity of glioblastoma multiforme (GBM) tumor cells to IR, especially those with checkpoint defects such as p53 abrogations. GBM is the most common and lethal form of brain tumor. Median survival of patients is 12-15 months, despite surgery, fractionated radiotherapy, and temozolomide standard of care. Poor survival is attributed to an inability to excise all tumor tissue (if operable), dissemination of disease into regions with an intact blood-brain barrier (BBB), and an intrinsic radio- and chemo-resistance. With ATM activity robustly upregulated in GBM stem cells, ATM represents an attractive radiosensitization target. Here, we describe the activity of AZD1390, a potent, selective, and orally bioavailable ATM inhibitor optimized for BBB penetration in preclinical model species. AZD1390 demonstrates exquisitely potent cellular inhibition of ATM activity (IC50 0.78 nM) with >1000-fold activity over closely related (PIKKs) and distant kinases. We confirm target and pathway engagement by Western blot and imaging pan-nuclear and discreet pATM foci staining (IC50 0.6-3nM). Radiosensitization of a panel of GBM cell lines and NCI-H2228 lung cells was confirmed in antiproliferation and clonogenic assays (IC50 3 nM). DEF37 of 2.7 was seen in p53 mutant GBM cells dosed at 3nM and p53 mutant GBM cell lines were more radiosensitized than wild type cells. Radiosensitization was confirmed in vivo in mouse orthotopic NCI-H2228 lung tumor models implanted directly into brain or via carotid artery injection showing dose-dependent tumor growth inhibition and remarkable increases in survival of mice when dosing AZD1390 PO an hour before four daily fractions of 2.5 Gy IR to the whole head. 20 mg/kg QD or BID gave the best survival benefit that correlated with bioluminescent tumor growth inhibition. Doses lower than 2 mg/kg were not efficacious, suggesting free brain PK cover over ATM IC50 of 3 hours or more are required for efficacy. Efficacy was also achieved in a dose-dependent manner in orthotopic GL261 murine GBM syngeneic models dosed in combination with either whole head radiotherapy or stereotactic beam radiotherapy. A PK PD efficacy relationship was establish by correlating AZD1390 free brain PK levels, phospho-ATM/Rad50 detection in tumor by IHC, and tumor growth inhibition and survival. Significant brain exposure was observed in a nonhuman primate PET study utilizing 11C-labelled AZD1390, further supporting the ability of the compound to efficiently cross the BBB. With confirmation that AZD1390 is not a substrate for human efflux transporters and having favorable pharmacokinetic and pharmacodynamic properties, AZD1390 is now in early clinical development for use as a radiosensitizer in central nervous system malignancies. Citation Format: Steve T. Durant, Kurt G. Pike, Nicola Colclough, Lucy Riches, Antonio Garcia-Trinidad, Thomas Hunt, Stephanie Ling, Jonathan Stott, Ian Barrett, Li Zheng, Yingchun Wang, Kan Chen, Tianwei Zhang, Venkatesh Pilla Reddy, Andrew Sykes, Peter Johnstrom, Gemma Jones, Andrew Pierce, Jeremy Karlin, Jenna Kahn, Jasmine Allen, Kristoffer Valerie, Ruth Illingworth, Martin Pass. AZD1390, a potent and selective orally bioavailable blood-brain barrier-penetrant ATM inhibitor, radiosensitizes and improves survival of orthotopic glioma and metastatic brain tumor models [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr A104.
- Published
- 2018
- Full Text
- View/download PDF
42. Abstract 2079: Osimertinib, an irreversible mutant selective EGFR tyrosine kinase inhibitor, exerts anti tumor activity in NSCLC harbouring exon 20 insertion mutant-EGFR
- Author
-
Susan Ashton, Ludovic Ménard, Matthew J. Martin, M. Raymond V. Finlay, Daniel O'Neill, Jonathan P. Orme, Maria Emanuela Cuomo, Darren Cross, Anna Staniszewska, Ambra Bianco, James W.T. Yates, Nicola Colclough, Darren Mckerrecher, Nicolas Floc'h, and Richard A. Ward
- Subjects
Genetics ,Cancer Research ,Mutation ,Cell growth ,Mutant ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,T790M ,Exon ,Oncology ,Cell culture ,medicine ,Cancer research ,Osimertinib - Abstract
EGFR exon 20 insertion (Ex20Ins) mutations represent a combination of in-frame insertions and/or duplications that account for 4-10% of all EGFR mutants in non-small cell lung cancer (NSCLC). To date, more than one hundred different Ex20Ins mutations have been identified. With the notable exception of the rare A763_Y764insFQEA insertion ( Using CRISPR-Cas 9 genome editing technology, we engineered cellular disease relevant models carrying the most prevalent Ex20Ins mutations, namely Ex20Ins D770_N771InsSVD (22%) or Ex20Ins V769_D770InsASV (17%). Using these models, we show that osimertinib and AZ5104 inhibit signalling pathways and cellular growth of Ex20Ins CRISPR-Cas9 engineered cell lines in vitro. This translates into sustained tumor growth inhibition in vivo in both the Ex20Ins SVD (65%, p EGFR signal. Two patients from the AURA Phase 2 osimertinib trials with plasma positive Ex20Ins (concurrent with Ex19del and T790M) had durable PFS responses of 6.4 and 13.9 months, supporting the premise that osimertinib has the potential to be clinically beneficial in tumors harboring Ex20Ins. The work presented herein demonstrates that osimertinib has the potential to improve upon the current treatment options for NSCLC patients whose tumors harbour an Ex20Ins mutation, and warrants its further clinical investigation. Citation Format: Nicolas Floc'h, Susan Ashton, Ambra Bianco, Nicola Colclough, Darren AE Cross, Maria Emanuela Cuomo, M. Raymond V. Finlay, Matthew J Martin, Ludovic Menard, Darren McKerrecher, Daniel J O'Neill, Jonathan P Orme, Anna D Staniszewska, Richard A Ward, James W T Yates. Osimertinib, an irreversible mutant selective EGFR tyrosine kinase inhibitor, exerts anti tumor activity in NSCLC harbouring exon 20 insertion mutant-EGFR [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2079. doi:10.1158/1538-7445.AM2017-2079
- Published
- 2017
- Full Text
- View/download PDF
43. Discovery of a potent and selective EGFR inhibitor (AZD9291) of both sensitizing and T790M resistance mutations that spares the wild type form of the receptor
- Author
-
Peter Ballard, Michael James, Daniel S. James, Peter D. Smith, Simon J. Brown, M. Raymond V. Finlay, Michael J. Waring, Paula Perkins, Andrew D. Campbell, Heather L. McFarland, David Perkins, Gordon S. Currie, Gillian M. Lamont, Graham Richmond, Matthew R. Box, David Whittaker, Mark J. Anderton, Richard A. Ward, Teresa Klinowska, Scott G. Lamont, Nicola Colclough, Stuart L. Wells, Darren Cross, Gail L. Wrigley, George B. Hill, Nathaniel G. Martin, Paul D. Kemmitt, Susan Ashton, Lorraine A. Hassall, Paul A. Bethel, Robert Hugh Bradbury, Sam Butterworth, Christopher G. Chorley, Martine J. Mellor, Matthew Grist, and Jonathon P. Orme
- Subjects
Male ,Lung Neoplasms ,Antineoplastic Agents ,Drug resistance ,Chemistry Techniques, Synthetic ,Pharmacology ,medicine.disease_cause ,T790M ,Inhibitory Concentration 50 ,Mice ,Carcinoma, Non-Small-Cell Lung ,Drug Discovery ,medicine ,Animals ,Humans ,Rociletinib ,Epidermal growth factor receptor ,Lung cancer ,Protein Kinase Inhibitors ,EGFR inhibitors ,Mutation ,Acrylamides ,Aniline Compounds ,biology ,Wild type ,Rats, Inbred Strains ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,ErbB Receptors ,Drug Resistance, Neoplasm ,biology.protein ,Molecular Medicine ,Female - Abstract
Epidermal growth factor receptor (EGFR) inhibitors have been used clinically in the treatment of non-small-cell lung cancer (NSCLC) patients harboring sensitizing (or activating) mutations for a number of years. Despite encouraging clinical efficacy with these agents, in many patients resistance develops leading to disease progression. In most cases, this resistance is in the form of the T790M mutation. In addition, EGFR wild type receptor inhibition inherent with these agents can lead to dose limiting toxicities of rash and diarrhea. We describe herein the evolution of an early, mutant selective lead to the clinical candidate AZD9291, an irreversible inhibitor of both EGFR sensitizing (EGFRm+) and T790M resistance mutations with selectivity over the wild type form of the receptor. Following observations of significant tumor inhibition in preclinical models, the clinical candidate was administered clinically to patients with T790M positive EGFR-TKI resistant NSCLC and early efficacy has been observed, accompanied by an encouraging safety profile.
- Published
- 2014
44. Biophysical methods in drug discovery from small molecule to pharmaceutical
- Author
-
Geoffrey, Holdgate, Stefan, Geschwindner, Alex, Breeze, Gareth, Davies, Nicola, Colclough, David, Temesi, and Lara, Ward
- Subjects
Binding Sites ,Magnetic Resonance Spectroscopy ,Protein Conformation ,Proteins ,Stereoisomerism ,Calorimetry ,Surface Plasmon Resonance ,High-Throughput Screening Assays ,Small Molecule Libraries ,Kinetics ,Nucleic Acids ,Drug Discovery ,Humans ,Thermodynamics ,Protein Binding - Abstract
Biophysical methods have become established in many areas of drug discovery. Application of these methods was once restricted to a relatively small number of scientists using specialized, low throughput technologies and methods. Now, automated high-throughput instruments are to be found in a growing number of laboratories. Many biophysical methods are capable of measuring the equilibrium binding constants between pairs of molecules crucial for molecular recognition processes, encompassing protein-protein, protein-small molecule, and protein-nucleic acid interactions, and several can be used to measure the kinetic or thermodynamic components controlling these biological processes. For a full characterization of a binding process, determinations of stoichiometry, binding mode, and any conformational changes associated with such interactions are also required. The suite of biophysical methods that are now available represents a powerful toolbox of techniques which can effectively deliver this full characterization.The aim of this chapter is to provide the reader with an overview of the drug discovery process and how biophysical methods, such as surface plasmon resonance (SPR), isothermal titration calorimetry (ITC), nuclear magnetic resonance, mass spectrometry (MS), and thermal unfolding methods can answer specific questions in order to influence project progression and outcomes. The selection of these examples is based upon the experiences of the authors at AstraZeneca, and relevant approaches are highlighted where they have utility in a particular drug discovery scenario.
- Published
- 2013
45. Biophysical Methods in Drug Discovery from Small Molecule to Pharmaceutical
- Author
-
Gareth M. Davies, Geoffrey A. Holdgate, David G. Temesi, Lara Ward, Nicola Colclough, Stefan Geschwindner, and Alexander L. Breeze
- Subjects
Computer science ,Drug discovery ,Stereoisomerism ,Isothermal titration calorimetry ,Computational biology ,Pharmacology ,Mass spectrometry ,Small molecule ,Characterization (materials science) ,Molecular recognition ,Protein structure ,High-Throughput Screening Assays ,Molecule ,Binding site ,Surface plasmon resonance - Abstract
Biophysical methods have become established in many areas of drug discovery. Application of these methods was once restricted to a relatively small number of scientists using specialized, low throughput technologies and methods. Now, automated high-throughput instruments are to be found in a growing number of laboratories. Many biophysical methods are capable of measuring the equilibrium binding constants between pairs of molecules crucial for molecular recognition processes, encompassing protein-protein, protein-small molecule, and protein-nucleic acid interactions, and several can be used to measure the kinetic or thermodynamic components controlling these biological processes. For a full characterization of a binding process, determinations of stoichiometry, binding mode, and any conformational changes associated with such interactions are also required. The suite of biophysical methods that are now available represents a powerful toolbox of techniques which can effectively deliver this full characterization.The aim of this chapter is to provide the reader with an overview of the drug discovery process and how biophysical methods, such as surface plasmon resonance (SPR), isothermal titration calorimetry (ITC), nuclear magnetic resonance, mass spectrometry (MS), and thermal unfolding methods can answer specific questions in order to influence project progression and outcomes. The selection of these examples is based upon the experiences of the authors at AstraZeneca, and relevant approaches are highlighted where they have utility in a particular drug discovery scenario.
- Published
- 2013
- Full Text
- View/download PDF
46. Abstract 3041: Blood-brain barrier penetrating ATM inhibitor (AZ32) radiosensitises intracranial gliomas in mice
- Author
-
Catherine Bardelle, Gareth Hughes, Kristoffer Valerie, Elaine Cadogan, Nitai D. Mukhopadhyay, Jeremy Karlin, Mary E. Tokarz, Nicola Colclough, Alan Lau, Andrew G. Thomason, Kurt Gordon Pike, Bhavika Patel, Syed Farhan Ahmad, Martin Pass, Ian P. Barrett, Steve T. Durant, and J M. Bekta
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Cell cycle checkpoint ,business.industry ,DNA repair ,DNA damage ,Cancer ,medicine.disease ,Oncology ,Apoptosis ,Radioresistance ,Glioma ,Cancer research ,Medicine ,Stem cell ,business - Abstract
Ataxia-telangiectasia mutated (ATM) kinase is a central DNA damage response (DDR) component signalling the presence of DNA double strand breaks (DSBs) to DNA repair, checkpoint and survival pathways. Clinical doses of fractionated radiotherapy directed at tumours kill cells by inducing single strand breaks and DSBs, the latter being particularly lethal to all cells if not repaired. Poor survival rates of glioblastoma multiforme (GBM) patients is attributed to an inability to excise all invasive tumor tissue (if operable) and an intrinsic tumour chemo/radioresistance, which has been linked to elevated ATM activity in glioma stem cells. ATM inhibition (ATMi) radiosensitises cancer cell lines in vitro and in vivo. ATMi also acutely radiosensitises patient-derived glioma stem cells more effectively than by inhibiting other DDR or cell cycle checkpoint components (PARP, ATR, Chk1). Checkpoint-defective glioblastoma multiforme (GBM) cell lines seem to be particularly radio-sensitised by ATMi. In addition, several studies suggest that normal brain is radioprotected when ATM activity is deficient, suggesting ATM is required for apoptosis in neurons. ATMi in brain may therefore provide a wide therapeutic margin. Furthermore, ATM's role in signalling DNA damage independent redox stress has been linked to promoting neoangiogenesis in tumour vasculature. Taken together, these studies suggest ATM is an extremely attractive target to inhibit during and potentially following radiotherapy. However, one impediment to preclinical and clinical studies is that current ATMi's have limited CNS bioavailability. We report AZ32 as the first known selective, orally bioavailable blood-brain barrier (BBB) penetrating ATMi probe. AZ32 inhibits the DDR and radiosensitizes p53/checkpoint-defective GBM cells in vitro. Oral daily dosing providing sufficient mouse free brain pharmacokinetic exposures over AZ32's ATM IC50, during just four daily doses of whole brain irradiation results in significant improvement in median overall survival of syngeneic orthotopic mouse glioma models (log-rank AZ32/radiation vs. radiation p = 0.0194). Tumor eradication was confirmed by tumor imaging. This result was recapitulated with a human orthotopic model. These findings support the development of clinical grade BBB-penetrating ATMi as a potential treatment for GBM and potentially other intracranial tumours dosed in combination with fractionated radiotherapy used in standard clinical practice. Citation Format: Steve T. Durant, Jeremy Karlin, Kurt Pike, Nicola Colclough, N Mukhopadhyay, S F. Ahmad, J M. Bekta, M Tokarz, Catherine Bardelle, Gareth Hughes, Bhavika Patel, Andrew Thomason, Elaine Cadogan, Ian Barrett, Alan Lau, Martin Pass, Kristoffer Valerie. Blood-brain barrier penetrating ATM inhibitor (AZ32) radiosensitises intracranial gliomas in mice. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3041.
- Published
- 2016
- Full Text
- View/download PDF
47. High throughput solubility determination with application to selection of compounds for fragment screening
- Author
-
Alison Hunter, Rod S. Kittlety, Mark A. Timms, Nicola Colclough, Lynsey Lobedan, Kin Yip Tam, and Peter W. Kenny
- Subjects
Chromatography ,Molecular Structure ,Chemistry ,High-throughput screening ,Organic Chemistry ,Clinical Biochemistry ,Fragment-based lead discovery ,Drug Evaluation, Preclinical ,Pharmaceutical Science ,Biochemistry ,Fully automated ,Pharmaceutical Preparations ,Solubility ,X-Ray Diffraction ,Drug Discovery ,Lipophilicity ,Aqueous solubility ,Molecular Medicine ,Molecular Biology ,Throughput (business) - Abstract
The development and application of a high throughput aqueous solubility assay is reported. Measurements for up to 637 compounds can be made in a fully automated experiment. Results from this assay were used to quantify risk of unacceptable solubility as a function of lipophilicity for neutral fragment-like compounds. Assessment of risk of unacceptable solubility was combined with experimental solubility measurement to select compounds for inclusion in a fragment-screening library.
- Published
- 2008
48. Abstract 4744: Structure-based development of covalent inhibitors of the activating and T790M gatekeeper mutant forms of the epidermal growth factor receptor (EGFR) leading to the discovery of AZD9291
- Author
-
Nicola Colclough, Mark J. Anderton, Martine J. Mellor, Rob H. Bradbury, Heather L. McFarland, Richard A. Ward, Sam Butterworth, M Ray V. Finlay, Darren Cross, Pete G. Ballard, Michael J. Waring, and Susan Ashton
- Subjects
Cancer Research ,biology ,business.industry ,Wild type ,Cancer ,medicine.disease ,T790M ,Gefitinib ,Oncology ,medicine ,Cancer research ,biology.protein ,Epidermal growth factor receptor ,Erlotinib ,business ,Tyrosine kinase ,medicine.drug ,EGFR inhibitors - Abstract
Small molecule inhibitors of the Epidermal Growth Factor Receptor (EGFR) tyrosine kinase such as gefitinib and erlotinib have been employed successfully in the treatment of non-small cell lung cancer (NSCLC) patients harboring an activating mutation (EGFRm+). However, resistance to these inhibitors in the form of additional mutations such as T790M, (mutation of the gatekeeper residue), is recognized as a clinical issue. This presentation will describe the discovery and evolution of one of our novel chemical series, leading ultimately to the identification of AZD9291, an orally bioavailable, covalent EGFR inhibitor of both the resistance (NCI-H1975, cell phosphorylation IC50 0.5 uM). Wild type EGFR inhibition is believed to drive the observed dose limiting toxicities (such as skin rash and diarrhea) for these first generation therapies in the clinic. New data will be presented for the first time including a broader description of the medicinal chemistry program that led to the identification of AZD9291. We shall also present previously undisclosed work on the identification of additional distinct chemical series and an update of recent data from ongoing AZD9291 Phase I clinical studies in NSCLC patients. L858R/T790M Double Mutant (resistance) cell IC50 uMEGFRm+ Single Mutant (activating) cell IC50 uMEGFR Wild Type cell IC50 uMAqueous solubility, (salt) (pH=6.8, ug/mL)L858R/T790M Double Mutant efficacy (% TGI at 5mpk PO QD for 14 days)0.5>490119 Citation Format: Richard A. Ward, Susan Ashton, Mark Anderton, Pete G. Ballard, Rob H. Bradbury, Sam Butterworth, Nicola Colclough, Darren A E Cross, M Ray V. Finlay, Heather L. McFarland, Martine Mellor, Mike J. Waring. Structure-based development of covalent inhibitors of the activating and T790M gatekeeper mutant forms of the epidermal growth factor receptor (EGFR) leading to the discovery of AZD9291. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4744. doi:10.1158/1538-7445.AM2014-4744
- Published
- 2014
- Full Text
- View/download PDF
49. The Development and Evaluation of a Microfluidic Device for the Determination of Organic-Aqueous Partition Coefficients to Support Agrochemical and Drug Discovery
- Author
-
Julie Deacon, Chris Turner, Eric Daniel Clarke, Brian Law, Nicola Colclough, Bina Mackenzie, and Dave Temesi
- Subjects
Partition coefficient ,Chromatography ,Aqueous solution ,Agrochemical ,business.industry ,Chemistry ,Drug discovery ,Microfluidics ,business - Abstract
A prototype multi-channel microfluidic device has been developed and tested for the determination of organic-aqueous partition coefficients.
- Published
- 2001
- Full Text
- View/download PDF
50. Abstract B94: Discovery of and first disclosure of the clinical candidate AZD9291, a potent and selective third-generation EGFR inhibitor of both activating and T790M resistant mutations that spares the wild type form of the receptor
- Author
-
Heather L. McFarland, Peter Ballard, Claudio Chuaqui, Susan Ashton, M. Raymond V. Finlay, Sam Butterworth, Martine J. Mellor, Darren Cross, Nicola Colclough, Rob H. Bradbury, Richard A. Ward, Mark J. Anderton, and Michael J. Waring
- Subjects
Cancer Research ,biology ,business.industry ,Wild type ,Cancer ,Pharmacology ,medicine.disease ,T790M ,Gefitinib ,Oncology ,medicine ,biology.protein ,Erlotinib ,Epidermal growth factor receptor ,business ,Tyrosine kinase ,EGFR inhibitors ,medicine.drug - Abstract
Small molecule inhibitors of the epidermal growth factor receptor (EGFR) tyrosine kinase such as gefitinib and erlotinib have been employed successfully in the treatment of non-small cell lung cancer (NSCLC) patients harboring an activating mutation (EGFRm+). However, resistance to these inhibitors in the form of additional mutations in the kinase domain such as T790M is emerging as a growing clinical issue. This presentation will describe the discovery of AZD9291, an orally bioavailable, irreversible EGFR inhibitor of both the resistance (NCI-H1975, cell phosphorylation IC50 0.5 μM). Wild type EGFR inhibition is believed to drive the observed dose limiting toxicities (such as skin rash and diarrhea) for these first generation therapies in the clinic. New data will be discussed for the first time including the medicinal chemistry program that led to the identification of AZD9291, details of significant in vivo oral activity in pre-clinical xenograft models (including tumor regression in the L858R/T790M double mutant setting at a dose of 5 mpk) and the first disclosure of the candidate drug structure. The pre-clinical findings from this work strongly supported selection of AZD9291 as a clinical candidate, and first dose in man was achieved with AZD9291 in March 2013. L858R/T790M Double Mutant (resistance) cell IC50 (μM)EGFRm+ Single Mutant (activating) cell IC50 (μM)EGFR Wild Type cell IC50 (μM)Aqueous solubility, (salt) (pH=6.8, μg/mL)L858R/T790M Double Mutant efficacy (% TGI at 5mpk PO QD for 14 days)AZD92910.5>490119 Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B94. Citation Format: M. Raymond V. Finlay, Mark Anderton, Susan Ashton, Peter G. Ballard, Rob H. Bradbury, Sam Butterworth, Nicola Colclough, Darren A. E. Cross, Heather L. McFarland, Martine J. Mellor, Richard A. Ward, Mike J. Waring. Discovery of and first disclosure of the clinical candidate AZD9291, a potent and selective third-generation EGFR inhibitor of both activating and T790M resistant mutations that spares the wild type form of the receptor. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B94.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.