Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Claire Sadler is active.

Publication


Featured researches published by Claire Sadler.


Toxicologic Pathology | 2011

Induction of Heart Valve Lesions by Small-Molecule ALK5 Inhibitors

Mark J. Anderton; Howard R. Mellor; Alex R. Bell; Claire Sadler; Martin Pass; Steve Powell; Samantha J. Steele; Ruth A. Roberts; Annabelle Heier

Aberrant signaling by transforming growth factor-β (TGF-β) and its type I (ALK5) receptor has been implicated in a number of human diseases and this pathway is considered a potential target for therapeutic intervention. Transforming growth factor-β signaling via ALK5 plays a critical role during heart development, but the role of ALK5 in the adult heart is poorly understood. In the current study, the preclinical toxicology of ALK5 inhibitors from two different chemistry scaffolds was explored. Ten-week-old female Han Wistar rats received test compounds by the oral route for three to seven days. Both compounds induced histopathologic heart valve lesions characterized by hemorrhage, inflammation, degeneration, and proliferation of valvular interstitial cells. The pathology was observed in all animals, at all doses tested, and occurred in all four heart valves. Immunohistochemical analysis of ALK5 in rat hearts revealed expression in the valves, but not in the myocardium. Compared to control animals, protein levels of ALK5 were unchanged in the heart valves of treated animals. We also observed a physeal dysplasia in the femoro-tibial joint of rats treated with ALK5 inhibitors, a finding consistent with a pharmacological effect described previously with ALK5 inhibitors. Overall, these findings suggest that TGF-β signaling via ALK5 plays a critical role in maintaining heart valve integrity.


Cancer Research | 2016

AZD9496: An Oral Estrogen Receptor Inhibitor That Blocks the Growth of ER-Positive and ESR1-Mutant Breast Tumors in Preclinical Models.

Hazel M. Weir; Robert Hugh Bradbury; Mandy Lawson; Alfred A. Rabow; David Buttar; Rowena Callis; Jon Owen Curwen; Camila de Almeida; Peter Ballard; Micheal Hulse; Craig S. Donald; Lyman Feron; Galith Karoutchi; Philip A. MacFaul; Thomas A. Moss; Richard A. Norman; Stuart E. Pearson; Michael Tonge; Gareth Davies; Graeme Walker; Zena Wilson; Rachel Rowlinson; Steve Powell; Claire Sadler; Graham Richmond; Brendon Ladd; Ermira Pazolli; Anne Marie Mazzola; Celina D'Cruz; Chris De Savi

Fulvestrant is an estrogen receptor (ER) antagonist administered to breast cancer patients by monthly intramuscular injection. Given its present limitations of dosing and route of administration, a more flexible orally available compound has been sought to pursue the potential benefits of this drug in patients with advanced metastatic disease. Here we report the identification and characterization of AZD9496, a nonsteroidal small-molecule inhibitor of ERα, which is a potent and selective antagonist and downregulator of ERα in vitro and in vivo in ER-positive models of breast cancer. Significant tumor growth inhibition was observed as low as 0.5 mg/kg dose in the estrogen-dependent MCF-7 xenograft model, where this effect was accompanied by a dose-dependent decrease in PR protein levels, demonstrating potent antagonist activity. Combining AZD9496 with PI3K pathway and CDK4/6 inhibitors led to further growth-inhibitory effects compared with monotherapy alone. Tumor regressions were also seen in a long-term estrogen-deprived breast model, where significant downregulation of ERα protein was observed. AZD9496 bound and downregulated clinically relevant ESR1 mutants in vitro and inhibited tumor growth in an ESR1-mutant patient-derived xenograft model that included a D538G mutation. Collectively, the pharmacologic evidence showed that AZD9496 is an oral, nonsteroidal, selective estrogen receptor antagonist and downregulator in ER(+) breast cells that could provide meaningful benefit to ER(+) breast cancer patients. AZD9496 is currently being evaluated in a phase I clinical trial. Cancer Res; 76(11); 3307-18. ©2016 AACR.


Antimicrobial Agents and Chemotherapy | 2014

1,4-Azaindole, a Potential Drug Candidate for Treatment of Tuberculosis

Monalisa Chatterji; Radha Shandil; M. R. Manjunatha; Suresh Solapure; Naveen Kumar; Ramanatha Saralaya; Jitendar Reddy; K. R. Prabhakar; Sreevalli Sharma; Claire Sadler; Christopher B. Cooper; Khisi Mdluli; Pravin S. Iyer; Shridhar Narayanan; Pravin S. Shirude

ABSTRACT New therapeutic strategies against multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mycobacterium tuberculosis are urgently required to combat the global tuberculosis (TB) threat. Toward this end, we previously reported the identification of 1,4-azaindoles, a promising class of compounds with potent antitubercular activity through noncovalent inhibition of decaprenylphosphoryl-β-d-ribose 2′-epimerase (DprE1). Further, this series was optimized to improve its physicochemical properties and pharmacokinetics in mice. Here, we describe the short-listing of a potential clinical candidate, compound 2, that has potent cellular activity, drug-like properties, efficacy in mouse and rat chronic TB infection models, and minimal in vitro safety risks. We also demonstrate that the compounds, including compound 2, have no antagonistic activity with other anti-TB drugs. Moreover, compound 2 shows synergy with PA824 and TMC207 in vitro, and the synergy effect is translated in vivo with TMC207. The series is predicted to have a low clearance in humans, and the predicted human dose for compound 2 is ≤1 g/day. Altogether, our data suggest that a 1,4-azaindole (compound 2) is a promising candidate for the development of a novel anti-TB drug.


ACS Chemical Biology | 2014

Pyrazolopyrimidines Establish MurC as a Vulnerable Target in Pseudomonas aeruginosa and Escherichia coli

Shahul Hameed P; Praveena Manjrekar; Murugan Chinnapattu; Vaishali Humnabadkar; Gajanan Shanbhag; Chaitanyakumar Kedari; Naina Vinay Mudugal; Anisha Ambady; Boudewijn L. M. de Jonge; Claire Sadler; Beena Paul; Shubha Sriram; Parvinder Kaur; Supreeth Guptha; Anandkumar Raichurkar; Paul R. Fleming; Charles J. Eyermann; David C. McKinney; Vasan K. Sambandamurthy; Manoranjan Panda; Sudha Ravishankar

The bacterial peptidoglycan biosynthesis pathway provides multiple targets for antibacterials, as proven by the clinical success of β-lactam and glycopeptide classes of antibiotics. The Mur ligases play an essential role in the biosynthesis of the peptidoglycan building block, N-acetyl-muramic acid-pentapeptide. MurC, the first of four Mur ligases, ligates l-alanine to UDP-N-acetylmuramic acid, initiating the synthesis of pentapeptide precursor. Therefore, inhibiting the MurC enzyme should result in bacterial cell death. Herein, we report a novel class of pyrazolopyrimidines with subnanomolar potency against both Escherichia coli and Pseudomonas aeruginosa MurC enzymes, which demonstrates a concomitant bactericidal activity against efflux-deficient strains. Radio-labeled precursor incorporation showed these compounds selectively inhibited peptidoglycan biosynthesis, and genetic studies confirmed the target of pyrazolopyrimidines to be MurC. In the presence of permeability enhancers such as colistin, pyrazolopyrimidines exhibited low micromolar MIC against the wild-type bacteria, thereby, indicating permeability and efflux as major challenges for this chemical series. Our studies provide biochemical and genetic evidence to support the essentiality of MurC and serve to validate the attractiveness of target for antibacterial discovery.


ChemMedChem | 2016

Nitroarenes as Antitubercular Agents: Stereoelectronic Modulation to Mitigate Mutagenicity

Sudhir Landge; Anupriya Kumar; João Neres; Kannan Murugan; Claire Sadler; Mick D. Fellows; Vaishali Humnabadkar; Prakash Vachaspati; Anandkumar Raichurkar; Sreevalli Sharma; Sudha Ravishankar; Supreeth Guptha; Vasan K. Sambandamurthy; Tanjore S. Balganesh; Bheemarao G. Ugarkar; V. Balasubramanian; Balachandra Bandodkar; Manoranjan Panda

Nitroarenes are less preferred in drug discovery due to their potential to be mutagenic. However, several nitroarenes were shown to be promising antitubercular agents with specific modes of action, namely, nitroimidazoles and benzothiazinones. The nitro group in these compounds is activated through different mechanisms, both enzymatic and non‐enzymatic, in mycobacteria prior to binding to the target of interest. From a whole‐cell screening program, we identified a novel lead nitrobenzothiazole (BT) series that acts by inhibition of decaprenylphosphoryl‐β‐d‐ribose 2′‐epimerase (DprE1) of Mycobacterium tuberculosis (Mtb). The lead was found to be mutagenic to start with. Our efforts to mitigate mutagenicity resulted in the identification of 6‐methyl‐7‐nitro‐5‐(trifluoromethyl)‐1,3‐benzothiazoles (cBTs), a novel class of antitubercular agents that are non‐mutagenic and exhibit an improved safety profile. The methyl group ortho to the nitro group decreases the electron affinity of the series, and is hence responsible for the non‐mutagenic nature of these compounds. Additionally, the co‐crystal structure of cBT in complex with Mtb DprE1 established the mode of binding. This investigation led to a new non‐mutagenic antitubercular agent and demonstrates that the mutagenic nature of nitroarenes can be solved by modulation of stereoelectronic properties.


Pharmacology & Therapeutics | 2017

Towards better models and mechanistic biomarkers for drug-induced gastrointestinal injury

Daniel F. Carr; Seyoum Ayehunie; Andrea Davies; Carrie A. Duckworth; Stephanie French; Neil Hall; Syed A. Hussain; Howard R. Mellor; Alan Norris; B. Kevin Park; Alexander Penrose; D. Mark Pritchard; Chris Probert; Shashi K. Ramaiah; Claire Sadler; Michael Schmitt; Alison Shaw; James E. Sidaway; Robert G. Vries; Matthew Wagoner; Munir Pirmohamed

ABSTRACT Adverse drug reactions affecting the gastrointestinal (GI) tract are a serious burden on patients, healthcare providers and the pharmaceutical industry. GI toxicity encompasses a range of pathologies in different parts of the GI tract. However, to date no specific mechanistic diagnostic/prognostic biomarkers or translatable pre‐clinical models of GI toxicity exist. This review will cover the current knowledge of GI ADRs, existing biomarkers and models with potential application for toxicity screening/monitoring. We focus on the current gaps in our knowledge, the potential opportunities and recommend that a systematic approach is needed to identify mechanism‐based GI biomarkers with potential for clinical translation.


Molecular Cancer Research | 2017

Abstract B32: CRLX101, an investigational camptothecin-containing nanoparticle-drug conjugate, combined with DDR agents provides a novel approach to increasing therapeutic index

Lenka Oplustil O'Connor; Anderson T. Wang; David Jones; Rajesh Odedra; Michael Spreadborough; Joanne Wilson; Aaron Smith; Peter Cotton; Jaimini Reens; Jen Barnes; Victoria Sheridan; Andres Tellez; Alan Lau; Claire Sadler; Mark J. O'Connor; Scott Eliasof

Topoisomerase I inhibitors are used as standard-of-care chemotherapy in many types of cancer but are associated with significant toxicities. There is potential to improve their efficacy further by combining with inhibitors of the DNA damage response, such as the poly ADP ribose polymerase (PARP) inhibitor olaparib. However, while preclinical data highlight the improved efficacy of this combination, subsequent clinical trials have struggled due to dose limiting myelotoxicity. CRLX101 is an investigational nanoparticle-drug conjugate (NDC) containing the payload camptothecin, a potent topoisomerase I inhibitor. This agent is preferentially targeted to tumours and demonstrated a generally favorable toxicity profile in the clinic. Here, we explored the molecular mechanism and therapeutic potential of combining CRLX101 with either olaparib or the WEE1 G2 checkpoint kinase inhibitor AZD1775, by testing both efficacy and safety in preclinical models. In vitro studies using NCI-H417a small cell lung cancer (SCLC) cells demonstrated that combination with both olaparib and AZD1775 potentiated the efficacy of CRLX101 although by different mechanisms. Cellular analyses revealed that CRLX101 treatment alone predominantly activated ATM-mediated DNA damage response and resulted in late S/G2 cell cycle arrest. Combination with a PARP inhibitor further enhanced the CRLX101-induced DNA damage response and prolonged cell cycle arrest in late S/G2 phase. In contrast, WEE1 inhibition abrogated late S/G2 cell cycle arrest induced by CRLX101, resulting in aberrant mitotic entry and enhanced cell death. Our in vivo studies using wild type Wistar rat model showed that CRLX101, olaparib and AZD1775, are well tolerated as single agents. However, concurrent combination of CRLX101 with either olaparib or AZD1775 resulted in a dose-dependent decrease in hematological parameters. We investigated sequenced schedules and demonstrated that at a 24h delay between the CRLX101 and olaparib mitigates much of the combined bone marrow toxicity, while improving the efficacy above CRLX101 alone in xenograft tumors from NCI-H417a cells. Collectively, these preclinical data demonstrate increased anti-tumor efficacy of CRLX101 when combined with DDR inhibitors. The combination schedule for CRLX101 and olaparib identified in our preclinical models as providing an increased therapeutic index has been used to develop protocols to test this combination in a relapsed (2nd line) SCLC human clinical trial (in collaboration with NCI). Citation Format: Lenka Oplustil O9Connor, Anderson T. Wang, David Jones, Rajesh Odedra, Michael Spreadborough, Joanne Wilson, Aaron Smith, Peter Cotton, Jaimini Reens, Jen Barnes, Victoria Sheridan, Andres Tellez, Alan Lau, Claire Sadler, Mark J. O9Connor, Scott Eliasof. CRLX101, an investigational camptothecin-containing nanoparticle-drug conjugate, combined with DDR agents provides a novel approach to increasing therapeutic index [abstract]. In: Proceedings of the AACR Special Conference on DNA Repair: Tumor Development and Therapeutic Response; 2016 Nov 2-5; Montreal, QC, Canada. Philadelphia (PA): AACR; Mol Cancer Res 2017;15(4_Suppl):Abstract nr B32.


Cancer Research | 2016

Abstract 3721: A camptothecin-containing nanoparticle-drug conjugate combination with DDR agents provides a novel approach to increasing therapeutic index

Lenka Oplustil O’Connor; Anderson T. Wang; David R. Jones; Rajesh Odedra; Michael Spreadborough; Joanne Wilson; Aaron Smith; Peter Cotton; Jaimini Reens; Jen Barnes; Victoria Sheridan; Scott Eliasof; Andres Tellez; Alan Lau; Claire Sadler; Mark J. O’Connor

Topoisomerase I inhibitors are used as standard-of-care chemotherapy in many types of cancer but are associated with significant toxicities. There is potential to improve their efficacy further by combining with inhibitors of the DNA damage response, such as the PARP inhibitor olaparib. However, while preclinical data highlight the improved efficacy of this combination, subsequent clinical trials have struggled due to dose limiting myelotoxicity. CRLX101 is an investigational nanoparticle-drug conjugate (NDC) containing the payload camptothecin (the most potent topoisomerase I inhibitor known). This agent is preferentially targeted to tumours and demonstrated a favourable toxicity profile in the clinic. Here, we explored the molecular mechanism and therapeutic potential of combining CRLX101 with either olaparib or the WEE1 inhibitor AZD1775, by testing both efficacy and safety in preclinical models. In vitro studies using NCI-H417a SCLC cells demonstrated that combination with both olaparib and AZD1775 potentiated the efficacy of CRLX101 although by different mechanisms. Cellular analyses revealed that CRLX101 treatment alone predominantly activated ATM-mediated DNA damage response and resulted in late S/G2 cell cycle arrest. Combination with a PARP inhibitor further enhanced the CRLX101-induced DNA damage response and prolonged cell cycle arrest in late S/G2 phase. In contrast, WEE1 inhibition abrogated late S/G2 cell cycle arrest induced by CRLX101, resulting in aberrant mitotic entry and enhanced cell death. Our in vivo studies using wild type Wistar rat model showed that CRLX101, olaparib and AZD1775, are well tolerated as single agents. However, concurrent combination of CRLX101 with either olaparib or AZD1775 resulted in a dose-dependent decrease in haematological parameters. We investigated sequenced schedules and demonstrated that at a 24h delay between the CRLX101 and olaparib mitigates much of the combined bone marrow toxicity, while improving the efficacy above CRLX101 alone in xenograft tumours from NCI-H417a cells. Collectively, these preclinical data demonstrate increased anti-tumour efficacy of CRLX101 when combined with DDR inhibitors. The combination schedule for CRLX101 and olaparib identified in our preclinical models as providing an increased therapeutic index has been used to develop protocols to test this combination in a relapsed (2nd line) SCLC human clinical trial (in collaboration with NCI). Citation Format: Lenka Oplustil O’Connor, Anderson T. Wang, David R. Jones, Rajesh Odedra, Michael Spreadborough, Joanne Wilson, Aaron Smith, Peter Cotton, Jaimini Reens, Jen Barnes, Victoria Sheridan, Scott Eliasof, Andres Tellez, Alan Lau, Claire Sadler, Mark J. O’Connor. A camptothecin-containing nanoparticle-drug conjugate combination with DDR agents provides a novel approach to increasing therapeutic index. [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 3721.


Molecular Cancer Therapeutics | 2013

Abstract C135: AZ’4425 is a potent, selective, and orally bio-available progesterone receptor antagonist that has shown anti-tumor activity and inhibition of cancer stem cell proliferation.

Christine M. Chresta; Denis Alferez; Georgia Cerillo; Emma Still; Adina Hughes; Graeme Walker; Jane Kendrew; Claire Sadler; Jayne Harris; Iain Simpson; Andrew Peter Thomas; Al Rabow; Robert B. Clarke; Sacha J Howell; Graham Richmond

Progesterone is an important hormone in breast cancer; the paracrine mediator of the progesterone receptor, RANKL plays a critical role in cancer stem cells, metastasis and tumorigenesis. Steroidal inhibitors of PR have shown modest clinical activity but have been limited by toxicity. AZ’4425 is a novel, potent non-steroidal progesterone receptor (PR) antagonist which selectively inhibits PR compared to other steroid hormone receptors (∼ 1000-fold ER, MR, GR, AR). AZ’4425 competitively inhibits progesterone binding, thus preventing progesterone-induced phosphorylation of PR, nuclear translocation and PR-induced transcription with an IC50 of 26nM. Anchorage independent growth of T47D cells is similarly inhibited with an IC50 of 25nM. Using cancer stem cell (CSC) assays (mammosphere formation and ALDH positivity) we have shown broad cellular activity in both ER/PR positive tumor cell lines and early and late stage clinical samples. Furthermore, AZ’4425 prevents the increase in CSCs, and miRNA changes (miR221/2 and miR200c) induced by anti-estrogen therapies, suggesting a potential role in prevention of acquired resistance. In vivo, the PR antagonist inhibits transcription of progesterone-induced genes including RANKL and SGK1. Notably, AZ’4425 results in significant inhibition of new tumor formation and results in stasis and regression of DMBA-induced tumors, both as monotherapy and in combination with letrozole. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C135. Citation Format: Christine M. Chresta, Denis Alferez, Georgia Cerillo, Emma Still, Adina Hughes, Graeme Walker, Jane Kendrew, Claire Sadler, Jayne Harris, Iain Simpson, Andrew P. Thomas, Al Rabow, Robert Clarke, Sacha Howell, Graham Richmond. AZ’4425 is a potent, selective, and orally bio-available progesterone receptor antagonist that has shown anti-tumor activity and inhibition of cancer stem cell proliferation. [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 C135.


Journal of Medicinal Chemistry | 2013

Azaindoles: Noncovalent DprE1 Inhibitors from Scaffold Morphing Efforts, Kill Mycobacterium tuberculosis and Are Efficacious in Vivo

Pravin S. Shirude; Radha Shandil; Claire Sadler; Maruti Naik; Vinayak Hosagrahara; Shahul P. Hameed; Vikas Shinde; Chandramohan Bathula; Vaishali Humnabadkar; Naveen Kumar; Jitendar Reddy; Sreevalli Sharma; Anisha Ambady; Naina Hegde; James Whiteaker; Robert E. McLaughlin; Humphrey Gardner; Prashanti Madhavapeddi; Parvinder Kaur; Ashwini Narayan; Supreeth Guptha; Disha Awasthy; Chandan Narayan; Jyothi Mahadevaswamy; K. G. Vishwas; Vijaykamal Ahuja; Abhishek Srivastava; K. R. Prabhakar; Ramesh R. Kale; Manjunatha Ramaiah

Collaboration


Dive into the Claire Sadler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge