Network


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

Hotspot


Dive into the research topics where Douglas Ferguson is active.

Publication


Featured researches published by Douglas Ferguson.


Xenobiotica | 2012

Practical use of the regression offset approach for the prediction of in vivo intrinsic clearance from hepatocytes

Anna-Karin Sohlenius-Sternbeck; Christopher R. Jones; Douglas Ferguson; Brian Middleton; Denis Projean; E. Floby; Johan Bylund; Lovisa Afzelius

Systematic under-prediction of clearance is frequently associated with in vitro kinetic data when extrapolated using physiological scaling factors, appropriate binding parameters and the well-stirred model. The present study describes a method of removing this systematic bias through application of empirical correction factors derived from regression analyses applied to the in vitro and in vivo data for a defined set of reference compounds. Linear regression lines were established with in vivo intrinsic clearance (CLint), derived from in vivo clearance data and scaled in vitro intrinsic clearance from isolated hepatocyte incubations. The scaled CLint was empirically corrected to a predicted in vivo CLint using the slope and intercept from a uniform weighted linear regression applied to the in vitro to in vivo extrapolation. Cross validation of human data demonstrated that 66% of the reference compounds had a predicted in vivo CLint within two-fold of the observed value. The average absolute fold error (AAFE) for the in vivo CLint predictions was 1.90. For rat, 54% of the compounds had a predicted value within two-fold of the observed and the AAFE was 1.98. Three AstraZeneca projects are used to exemplify how a two-sided prediction interval, applied to the rat regression corrected reference data, can form the basis for assessing the likelihood that, for a given chemical series, the in vitro kinetic data is predictive of in vivo clearance and is therefore appropriate to guide optimisation of compound metabolic stability.


Toxicological Sciences | 2014

Early Prediction of Polymyxin-Induced Nephrotoxicity With Next-Generation Urinary Kidney Injury Biomarkers

Natalie Keirstead; Matthew Wagoner; Patricia Bentley; Marie Blais; Crystal Brown; Letitia Cheatham; Paul Ciaccio; Yvonne P. Dragan; Douglas Ferguson; Jim Fikes; Melanie Galvin; Anshul Gupta; Michael R. Hale; Nakpangi Johnson; Wenli Luo; Frank McGrath; Mark Pietras; Sally A. Price; Abhishek Sathe; Jennifer C. Sasaki; Debra Snow; Robert L. Walsky; Gunther Kern

Despite six decades of clinical experience with the polymyxin class of antibiotics, their dose-limiting nephrotoxicity remains difficult to predict due to a paucity of sensitive biomarkers. Here, we evaluate the performance of standard of care and next-generation biomarkers of renal injury in the detection and monitoring of polymyxin-induced acute kidney injury in male Han Wistar rats using colistin (polymyxin E) and a polymyxin B (PMB) derivative with reduced nephrotoxicity, PMB nonapeptide (PMBN). This study provides the first histopathological and biomarker analysis of PMBN, an important test of the hypothesis that fatty acid modifications and charge reductions in polymyxins can reduce their nephrotoxicity. The results indicate that alterations in a panel of urinary kidney injury biomarkers can be used to monitor histopathological injury, with Kim-1 and α-GST emerging as the most sensitive biomarkers outperforming clinical standards of care, serum or plasma creatinine and blood urea nitrogen. To enable the prediction of polymyxin-induced nephrotoxicity, an in vitro cytotoxicity assay was employed using human proximal tubule epithelial cells (HK-2). Cytotoxicity data in these HK-2 cells correlated with the renal toxicity detected via safety biomarker data and histopathological evaluation, suggesting that in vitro and in vivo methods can be incorporated within a screening cascade to prioritize polymyxin class analogs with more favorable renal toxicity profiles.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of a potent respiratory syncytial virus RNA polymerase inhibitor.

Hui Xiong; Melinda A. Foulk; Lisa Aschenbrenner; Jun Fan; Choi-Lai Tiong-Yip; Kenneth D. Johnson; Demetri T. Moustakas; Paul R. Fleming; Dean G. Brown; Minli Zhang; Douglas Ferguson; Dedong Wu; Qin Yu

Targeting viral polymerases has been a proven and attractive strategy for antiviral drug discovery. Herein we describe our effort in improving the antiviral activity and physical properties of a series of benzothienoazepine compounds as respiratory syncytial virus (RSV) RNA polymerase inhibitors. The antiviral activity and spectrum of this class was significantly improved by exploring the amino substitution of the pyridine ring, resulting in the discovery of the most potent RSV A polymerase inhibitors reported to date.


International Journal of Cancer | 2013

Intratracheal and oral administration of SM-276001: A selective TLR7 agonist, leads to antitumor efficacy in primary and metastatic models of cancer

Erina Koga-Yamakawa; Simon J. Dovedi; Masashi Murata; Hiroyuki Matsui; Andrew J. Leishman; John Bell; Douglas Ferguson; Simon P. Heaton; Toshihiro Oki; Hideyuki Tomizawa; Ash Bahl; Haruo Takaku; Robert W. Wilkinson; Hideyuki Harada

Topical TLR7 agonists such as imiquimod are highly effective for the treatment of dermatological malignancies; however, their efficacy in the treatment of nondermatological tumors has been less successful. We report that oral administration of the novel TLR7‐selective small molecule agonist; SM‐276001, leads to the induction of an inflammatory cytokine and chemokine milieu and to the activation of a diverse population of immune effector cells including T and B lymphocytes, NK and NKT cells. Oral administration of SM‐276001 leads to the induction of IFNα, TNFα and IL‐12p40 and a reduction in tumor burden in the Balb/c syngeneic Renca and CT26 models. Using the OV2944‐HM‐1 model of ovarian cancer which spontaneously metastasizes to the lungs following subcutaneous implantation, we evaluated the efficacy of intratracheal and oral administration of SM‐276001 in an adjuvant setting following surgical resection of the primary tumor. We show that both oral and intratracheal TLR7 therapy can reduce the frequency of pulmonary metastasis, and metastasis to the axillary lymph nodes. These results demonstrate that SM‐276001 is a potent selective TLR7 agonist that can induce antitumor immune responses when dosed either intratracheally or orally.


Cancer Research | 2013

Abstract 919: AZD3463, a novel ALK/IGF1R inhibitor, overcomes multiple mechanisms of acquired resistance to crizotinib.

Lisa Drew; Jane Cheng; Jeffrey A. Engelman; Douglas Ferguson; Ryohei Katayama; Brenda McDermott; Jamal C. Saeh; Alice T. Shaw; Minhui Shen; Dan Widzowski; Allan Wu; Graeme Smith

Genomic rearrangement of Anaplastic Lymphoma Kinase (ALK) has been observed in several tumor types including 60-80% anaplastic large cell lymphoma (ALCL) and 3-6% of non small cell lung cancer (NSCLC). Although the ALK inhibitor crizotinib has clinical efficacy in selected ALK positive NSCLC patients, the majority of patients who show initial responses eventually relapse. Various mechanisms leading to resistance have been proposed and include ALK amplification and resistance mutations, as well as alternative pathway drivers including EGFR, cKIT and, more recently, IGF1R. We have discovered a novel and potent inhibitor of ALK, AZD3463 with a Ki value of 0.75nM which also inhibits additional receptor tyrosine kinases including insulin growth factor receptor (IGF1R) with equivalent potency. AZD3463 inhibits ALK in cells as demonstrated by its ability to decrease ALK autophosphorylation in tumor cell lines containing ALK fusions including DEL (ALCL NPM-ALK), H3122 (NSCLC EML4-ALK) and H2228 (NSCLC EML4-ALK). Inhibition of ALK is associated with perturbations in downstream signaling including ERK, AKT and STAT3 pathways leading to preferential inhibition of proliferation in the ALK fusion containing cell lines in vitro. AZD3463 also demonstrates the ability to dose dependently inhibit pALK in xenograft tumors in vivo resulting in stasis (H3122) or regression (DEL, H2228). AZD3463 retains good activity against a number of clinically relevant crizotinib resistant mutations including the gatekeeper mutant L1196M where equivalent potency to wild type ALK is observed in vitro and in vivo in EML4-ALK containing BAF3 cell lines. To further assess the potential ability of AZD3463 to overcome additional resistance mechanisms, antiproliferative activity was assessed in multiple crizotinib resistant cell lines independently derived in vitro from H3122 cells as well as a patient derived crizotinib relapsed model. These resistant cell lines contain multiple resistance mechanisms including the L1196M gatekeeper and T115Ins mutations, ALK amplification and/or secondary drivers including EGFR and IGF1R. AZD3463 retains antiproliferative potency within 4 fold of parental H3122 cells for 10 out of 12 of these acquired resistance models in vitro. In summary, AZD3463 is a potent ALK inhibitor which inhibits additional kinases including IGF1R and has activity in a number of crizotinib resistant models driven by multiple resistance mechanisms. Citation Format: Lisa Drew, Jane Cheng, Jeffrey Engelman, Douglas Ferguson, Ryohei Katayama, Brenda McDermott, Jamal Saeh, Alice Shaw, Minhui Shen, Dan Widzowski, Allan Wu, Graeme Smith. AZD3463, a novel ALK/IGF1R inhibitor, overcomes multiple mechanisms of acquired resistance to crizotinib. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 919. doi:10.1158/1538-7445.AM2013-919


Cancer Research | 2016

Abstract 3572: AZ5576, a novel potent and selective CDK9 inhibitor, induces rapid cell death and achieves efficacy in multiple preclinical hematological models

Justin Cidado; Minhui Shen; Michael Grondine; Scott Boiko; Haiyun Wang; Alexandra Borodovsky; Anne Marie Mazzola; Alan Wu; Deborah Lawson; Douglas Ferguson; Beirong Gao; Andy Cui; Celina M. D’Cruz; Lisa Drew

Cyclin-dependent kinase 9 (Cdk9) is a serine/threonine kinase that regulates elongation of transcription through phosphorylation of RNA polymerase II at serine 2 (pSer2-RNAPII). Mcl1, an anti-apoptotic protein that has been linked to increased survival and chemotherapy resistance in various cancers, can be indirectly modulated through transient inhibition of Cdk9 due to it having a short-lived transcript and being a labile protein. Transient inhibition of Cdk9, therefore, represents a potential therapeutic opportunity in tumors dependent on Mcl1 for survival, including various hematological malignancies. AZ5576 is a potent, highly selective, and orally bioavailable inhibitor of Cdk9 that inhibits Cdk9 enzyme activity with an IC50 Citation Format: Justin Cidado, Minhui Shen, Michael Grondine, Scott Boiko, Haiyun Wang, Alexandra Borodovsky, Anne Marie Mazzola, Alan Wu, Deborah Lawson, Douglas Ferguson, Beirong Gao, Andy Cui, Celina D’Cruz, Lisa Drew. AZ5576, a novel potent and selective CDK9 inhibitor, induces rapid cell death and achieves efficacy in multiple preclinical hematological models. [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 3572.


Cancer Research | 2013

Abstract 3362: Miniaturized PBPK model improves pharmacodynamic characterization and physiological interpretability for compounds with profound hysteresis in tumor.

Francis D. Gibbons; Dan Widzowski; Minhui Shen; Jane Cheng; Lisa Drew; Jamal C. Saeh; Douglas Ferguson

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DCnnBackground:nnSignificant hysteresis between plasma concentration and target inhibition at the effect site (e.g., tumor) is a frequent observation, commonly described mathematically by connecting the central (i.e., plasma) compartment to an ‘effect compartment’ by a ‘link’ which causes the concentration in the latter to be delayed relative to the plasma. The result is a direct response between effect-compartment concentration and target inhibition. A significant drawback is that the effect compartment cannot be observed (making it impossible to validate) and has no physiological interpretation (rendering communication with other disciplines difficult). We develop a novel approach that is more physiologically meaningful, provides more-precise model parameter estimates, and gives insight into the physico-chemical factors limiting distribution into the tumor.nnMethod:nnWe orally administered single doses of several compounds (including Crizotinib, AZD3463, and others) targeting ALK to mice bearing tumors derived from the DEL and H3122 non-small-cell lung cancer line, at several dose levels. At 6, 24, and 48 hours post-dose, we measured the plasma and tumor concentrations of each compound and associated target inhibition (phosphorylated ALK, pALK) in the tumor. pALK inhibition shows a direct response not to plasma, but to tumor concentration, indicating that the delay is distributional in nature. We constructed a miniature physiologically-based pharmacokinetic (mPBPK) model consisting of a central compartment and a tumor of fixed physiological volume. pALK inhibition was modeled as a direct Emax response to tumor concentration. For each compound, we simultaneously fitted the mPBPK model to the naive-pooled plasma and tumor concentrations, as well as pALK, using all available dose levels. Beyond the standard PK and PD parameters (Emax, E, IC50) we also fitted the tumor partition constant Kp, and tumor blood flow rate Qt. For comparison, we fitted a standard effect-compartment (‘link’) model to the plasma concentrations and pALK levels to the same data.nnResults:nnFor each compound, we computed unbound EC50 for both effect-compartment and mPBPK models. We found that while the point estimates largely agree, the mPBPK model delivers more-precise estimates (typically 50% lower CV%). We attribute this to its use of additional data (tumor concentration) to constrain the model, which more than compensates for the additional parameters in the mPBPK model. We find that there is broad consistency in estimates of tumor flow rate Qt across the compounds studied, indicating that distribution from plasma to site of action is limited by blood flow, rather than by permeability. Additionally, we found that the greater physiological interpretability of the mPBPK model enhances cross-functional communication within project teams.nnCitation Format: Francis D. Gibbons, Dan Widzowski, Minhui Shen, Jane Cheng, Lisa Drew, Jamal C. Saeh, Douglas Ferguson. Miniaturized PBPK model improves pharmacodynamic characterization and physiological interpretability for compounds with profound hysteresis in tumor. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3362. doi:10.1158/1538-7445.AM2013-3362


Cancer Research | 2013

Abstract 2461: Design and synthesis of AZD3463, a novel orally bioavailable dual ALK and IGF1R inhibitor, inhibits growth of crizotinib resistance cell lines with multiple mechanisms of acquired resistance.

Jamal C. Saeh; Bin Yang; Tim Pontz; Kumar Thakur; Bo Peng; Lisa Drew; Caroline Rivard; Dan Widzowski; Jane Cheng; Douglas Ferguson; Brenda McDermott; Minhui Shen; John McNulty; Ryohei Katayama; Jeffrey Engleman; Alice T. Shaw; Daniel John Russell; Graeme Smith

Although the ALK inhibitor crizotinib has clinical efficacy in selected ALK positive NSCLC patients, the majority of patients who initially respond eventually relapse. Several mechanisms of resistance have been proposed including amplification, resistance mutations, as well as alternative pathway drivers including EGFR, cKIT and IGF1R. We report here the discovery of a novel scaffold of ALK inhibitors and optimization effort that led to the discovery of AZD3463 a novel dual in vivo active ALK and IGF1R inhibitor. An in house subset screening of kinase inhibitors and de novo studies identified 4-(1H-indol-3-yl)pyrimidin-2-amine analogs as potent ALK inhibitors. Modeling studies were utilized to guide the SAR strategy around the aminopyrimidine group which afforded several lead compounds. Early SAR efforts quickly determined that smaller substituents, chloro and methyl, were optimal in the C5 position of the pyrimidine, and that aniline is preferred over several other amino heterocycles investigated. Parallel medicinal chemistry strategies were executed for the optimization of the aniline and indole. These studies suggested that a 2,4-substituted aniline provided optimal potency and selectivity in conjunction with a variety of heterocycles in C4 position of the pyrimdine. Cyclic amines in the C4 position of the aniline led to simultaneous improvement of potency and metabolic stability. The inhibition of IGF1R in vitro was maintained and modulation of glucose levels in vivo was observed with the optimized compounds and AZD3463. AZD3463 demonstrated superior potency to crizotinib in vivo (H3122 PD unbound EC50=0.16 nM). In summary, detailed SAR studies were executed on the 4-(1H-indol-3-yl)pyrimidin-2-amine template that produced potent inhibitors of ALK with improved physical chemical and ADME properties, and identified AZD3463, a novel equipotent ALK and IGF1R inhibitor, potent in ALK-driven preclinical models and in a variety of crizotinib-resistant models. We present herein the design and synthesis of AZD3463 as well as its overall properties. Citation Format: Jamal C. Saeh, Bin Yang, Tim Pontz, Kumar Thakur, Bo Peng, Lisa Drew, Caroline Rivard, Dan Widzowski, Jane Cheng, Douglas Ferguson, Brenda McDermott, Minhui Shen, John McNulty, Ryohei Katayama, Jeffrey Engleman, Alice Shaw, Daniel Russell, Graeme Smith. Design and synthesis of AZD3463, a novel orally bioavailable dual ALK and IGF1R inhibitor, inhibits growth of crizotinib resistance cell lines with multiple mechanisms of acquired resistance. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2461. doi:10.1158/1538-7445.AM2013-2461


Cancer Research | 2018

Abstract 1650: Discovery of AZD4573, a potent and selective inhibitor of CDK9 that enables transient target engagement for the treatment of hematologic malignancies

Bernard Christophe Barlaam; Chris De Savi; Lisa Drew; Andrew D. Ferguson; Douglas Ferguson; Chungang Gu; Janet Hawkins; Alexander Hird; Michelle L. Lamb; Nichole O'Connell; Kurt Gordon Pike; Theresa Proia; Maryann San Martin; Melissa Vasbinder; Jeff Varnes; Jianyan Wang; Wenlin Shao


Cancer Research | 2018

Abstract 297: AZD4573: Mechanistic PKPD model linking CDK9 inhibition to Mcl1 depletion and induction of apoptosis in preclinical AML model

Douglas Ferguson; Theresa Proia; Justin Cidado; Scott Boiko; Maryann San Martin; Steven Criscione; Wenlin Shao; Lisa Drew

Collaboration


Dive into the Douglas Ferguson'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge