Network


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

Hotspot


Dive into the research topics where Margaret Weidner is active.

Publication


Featured researches published by Margaret Weidner.


Cancer Research | 2015

SLC46A3 Is Required to Transport Catabolites of Noncleavable Antibody Maytansine Conjugates from the Lysosome to the Cytoplasm

Kevin J. Hamblett; Allison P. Jacob; Jesse L. Gurgel; Mark E. Tometsko; Brooke M. Rock; Sonal Patel; Robert R. Milburn; Sophia Siu; Seamus Ragan; Dan A. Rock; Christopher J. Borths; Jason W. O'Neill; Wesley S. Chang; Margaret Weidner; Matthew M. Bio; William C. Fanslow

Antibody-drug conjugates (ADC) target cytotoxic drugs to antigen-positive cells for treating cancer. After internalization, ADCs with noncleavable linkers are catabolized to amino acid-linker-warheads within the lysosome, which then enter the cytoplasm by an unknown mechanism. We hypothesized that a lysosomal transporter was responsible for delivering noncleavable ADC catabolites into the cytoplasm. To identify candidate transporters, we performed a phenotypic shRNA screen with an anti-CD70 maytansine-based ADC. This screen revealed the lysosomal membrane protein SLC46A3, the genetic attenuation of which inhibited the potency of multiple noncleavable antibody-maytansine ADCs, including ado-trastuzumab emtansine. In contrast, the potencies of noncleavable ADCs carrying the structurally distinct monomethyl auristatin F were unaffected by SLC46A3 attenuation. Structure-activity experiments suggested that maytansine is a substrate for SLC46A3. Notably, SLC46A3 silencing led to relative increases in catabolite concentrations in the lysosome. Taken together, our results establish SLC46A3 as a direct transporter of maytansine-based catabolites from the lysosome to the cytoplasm, prompting further investigation of SLC46A3 as a predictive response marker in breast cancer specimens.


Molecular Cancer Therapeutics | 2014

Preclinical Evaluation of AMG 925, a FLT3/CDK4 Dual Kinase Inhibitor for Treating Acute Myeloid Leukemia

Kathleen Keegan; Cong Li; Zhihong Li; Ji Ma; Mark L. Ragains; Suzanne Coberly; David Hollenback; John Eksterowicz; Lingming Liang; Margaret Weidner; Justin Huard; Xianghong Wang; Grace Alba; Jessica Orf; Mei-Chu Lo; Sharon Zhao; Rachel Ngo; Ada Chen; Lily Liu; Timothy J. Carlson; Christophe Quéva; Lawrence R. McGee; Julio C. Medina; Alexander Kamb; Dineli Wickramasinghe; Kang Dai

Acute myeloid leukemia (AML) remains a serious unmet medical need. Despite high remission rates with chemotherapy standard-of-care treatment, the disease eventually relapses in a major proportion of patients. Activating Fms-like tyrosine kinase 3 (FLT3) mutations are found in approximately 30% of patients with AML. Targeting FLT3 receptor tyrosine kinase has shown encouraging results in treating FLT3-mutated AML. Responses, however, are not sustained and acquired resistance has been a clinical challenge. Treatment options to overcome resistance are currently the focus of research. We report here the preclinical evaluation of AMG 925, a potent, selective, and bioavailable FLT3/cyclin-dependent kinase 4 (CDK4) dual kinase inhibitor. AMG 925 inhibited AML xenograft tumor growth by 96% to 99% without significant body weight loss. The antitumor activity of AMG 925 correlated with the inhibition of STAT5 and RB phosphorylation, the pharmacodynamic markers for inhibition of FLT3 and CDK4, respectively. In addition, AMG 925 was also found to inhibit FLT3 mutants (e.g., D835Y) that are resistant to the current FLT3 inhibitors (e.g., AC220 and sorafenib). CDK4 is a cyclin D–dependent kinase that plays an essential central role in regulating cell proliferation in response to external growth signals. A critical role of the CDK4–RB pathway in cancer development has been well established. CDK4-specific inhibitors are being developed for treating RB-positive cancer. AMG 925, which combines inhibition of two kinases essential for proliferation and survival of FLT3-mutated AML cells, may improve and prolong clinical responses. Mol Cancer Ther; 13(4); 880–9. ©2014 AACR.


European Journal of Medicinal Chemistry | 2014

Synthesis and structure–activity relationship of trisubstituted thiazoles as Cdc7 kinase inhibitors

Andreas Reichelt; Julie M. Bailis; Michael D. Bartberger; Guomin Yao; Hong Shu; Matthew R. Kaller; John G. Allen; Margaret Weidner; Kathleen Keegan; Jennifer Dao

The Cell division cycle 7 (Cdc7) protein kinase is essential for DNA replication and maintenance of genome stability. We systematically explored thiazole-based compounds as inhibitors of Cdc7 kinase activity in cancer cells. Our studies resulted in the identification of a potent, selective Cdc7 inhibitor that decreased phosphorylation of the direct substrate MCM2 in vitro and in vivo, and inhibited DNA synthesis and cell viability in vitro.


Journal of Pharmacokinetics and Pharmacodynamics | 2014

FLT3 and CDK4/6 inhibitors: Signaling mechanisms and tumor burden in subcutaneous and orthotopic mouse models of acute myeloid leukemia

Yaping Zhang; Cheng-Pang Hsu; Jian-Feng Lu; Mita Kuchimanchi; Yu-Nien Sun; Ji Ma; Guifen Xu; Yilong Zhang; Yang Xu; Margaret Weidner; Justin Huard; David Z. D’Argenio

FLT3ITD subtype acute myeloid leukemia (AML) has a poor prognosis with currently available therapies. A number of small molecule inhibitors of FLT3 and/or CDK4/6 are currently under development. A more complete and quantitative understanding of the mechanisms of action of FLT3 and CDK4/6 inhibitors may better inform the development of current and future compounds that act on one or both of the molecular targets, and thus may lead to improved treatments for AML. In this study, we investigated in both subcutaneous and orthotopic AML mouse models, the mechanisms of action of three FLT3 and/or CDK4/6 inhibitors: AMG925 (Amgen), sorafenib (Bayer and Onyx), and quizartinib (Ambit Biosciences). A composite model was developed to integrate the plasma pharmacokinetics of these three compounds on their respective molecular targets, the coupling between the target pathways, as well as the resulting effects on tumor burden reduction in the subcutaneous xenograft model. A sequential modeling approach was used, wherein model structures and estimated parameters from upstream processes (e.g. PK, cellular signaling) were fixed for modeling subsequent downstream processes (cellular signaling, tumor burden). Pooled data analysis was employed for the plasma PK and cellular signaling modeling, while population modeling was applied to the tumor burden modeling. The resulting model allows the decomposition of the relative contributions of FLT3ITD and CDK4/6 inhibition on downstream signaling and tumor burden. In addition, the action of AMG925 on cellular signaling and tumor burden was further studied in an orthotopic tumor mouse model more closely representing the physiologically relevant environment for AML.


Cancer Research | 2013

Abstract 2351: CDK4/FLT3 dual inhibitors as potential therapeutics for acute myeloid leukemia.

Zhihong Li; Kang Dai; Kathleen Keegan; Ji Ma; Mark L. Ragains; Jacob Kaizerman; Dustin L. McMinn; Jiasheng Fu; Benjamin Fisher; Michael W. Gribble; Lawrence R. McGee; John Eksterowicz; Cong Li; Lingming Liang; Margaret Weidner; Justin Huard; Robert Cho; Timothy J. Carlson; Grace Alba; David Hollenback; John S. Hill; Darrin M. Beaupre; Alexander Kamb; Dineli Wickramasinghe; Julio C. Medina

CDK4 is a cyclin D dependent kinase that promotes cell cycle progression in a broad range of tumor types by phosphorylating the tumor suppressor retinoblastoma protein (Rb) and releasing transcription factor E2F. Critical involvement of the cyclin D-CDK4-Rb pathway in carcinogenesis is strongly supported by a large amount of genetic evidence. In addition, promoter methylation with consequent silencing of expression of the CDK4 inhibitor, p15, has been reported in 44-60% of acute myeloid leukemia (AML) patients. It is also well established that constitutive activation of the tyrosine kinase FLT3 via mutation contributes to the development of AML, with 30% of AML carrying such activating mutations. FLT3 tyrosine kinase inhibitors used as single agents reduce peripheral blood and bone marrow blasts in only a minority of AML patients, and the effect tends to be transient. This may be due to insufficient FLT3 inhibition, the selection of drug-resistant clones, or the independence of the cell on FLT3 signaling for proliferation and survival. In preclinical models, a synergistic effect of CDK4 inhibition and FLT3 inhibition resulting in increased apoptosis of AML cell lines was reported (Wang et al., Blood, 2007). From a HTS hit through SAR optimization led to AM-5992, a potent and orally bioavailable dual inhibitor of CDK4 and FLT3 including all FLT3 mutants reported to date. AM-5992 inhibits the proliferation of a panel of human tumor cell lines including MDA-MB-435(Rb+), colo-205(Rb+), U937(FLT3WT) and induced cell death in MOLM13(FLT3ITD), MV4-11(FLT3ITD), and even in MOLM13(FLT3ITD, D835Y) which exhibits resistance to a number of FLT3 inhibitors currently under clinical development. In mouse models of leukemia using cells with the FLT3ITD mutation, AM-5992 treatment at 150 mpk qd on days 6-16 after leukemia cell injection significantly reduced the leukemia burden and prolonged survival 11 days over that of vehicle controls. Collectively, these data support the hypothesis that simultaneously inhibition of CDK4 and FLT3 may improve the durability of clinical response in AML; and consequently that this hypothesis should be tested in the clinic. Citation Format: Zhihong Li, Kang Dai, Kathleen Keegan, Ji Ma, Mark Ragains, Jacob Kaizerman, Dustin McMinn, Jiasheng Fu, Benjamin Fisher, Michael Gribble, Lawrence R. McGee, John Eksterowicz, Cong Li, Lingming Liang, Margaret Weidner, Justin Huard, Robert Cho, Timothy Carlson, Grace M. Alba, David Hollenback, John Hill, Darrin Beaupre, Alexander Kamb, Dineli Wickramasinghe, Julio C. Medina. CDK4/FLT3 dual inhibitors as potential therapeutics for acute myeloid leukemia. [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 2351. doi:10.1158/1538-7445.AM2013-2351


Archive | 2012

Fused tricyclic dual inhibitors of CDK 4/6 and FLT3

Xiaoqi Chen; Kang Dai; Jason Duquette; Michael W. Gribble; Justin Huard; Kathleen Keegan; Zhihong Li; Sarah E. Lively; Lawrence R. McGee; Mark L. Ragains; Xianghong Wang; Margaret Weidner; Jian Zhang


Archive | 2012

Doubles inhibiteurs tricycliques fusionnés de cdk 4/6 et de flt3

Xiaoqi Chen; Kang Dai; Jason Duquette; Michael W. Gribble; Justin Huard; Kathleen S. Keegan; Zhihong Li; Sarah E. Lively; Lawrence R. McGee; Mark L. Ragains; Xianghong Wang; Margaret Weidner; Jian Zhang


Archive | 2012

DEHYDRATED tricyclic DUALINHIBITORER OF CDK 4/6 AND FLT3

Xiaoqi Chen; Kang Dai; Michael W. Gribble; Zhihong Li; Sarah E. Lively; Lawrence R. McGee; Xianghong Wang; Margaret Weidner; Jian Zhang; Kathleen S. Keegan; Jason Duquette; Justin Huard; Mark L. Ragains


Archive | 2012

Fusionierte tricyclische duale inhibitoren von cdk 4/6 und flt3

Xiaoqi Chen; Kang Dai; Jason Duquette; Michael W. Gribble; Justin Huard; Kathleen S. Keegan; Zhihong Li; Sarah E. Lively; Lawrence R. McGee; Mark L. Ragains; Xianghong Wang; Margaret Weidner; Jian Zhang


Archive | 2004

Utilisation d'inhibiteurs de la chk1 pour lutter contre la proliferation cellulaire

Darcey Clark; Kathleen S. Keegan; Scott Peterson; Margaret Weidner

Collaboration


Dive into the Margaret Weidner's collaboration.

Researchain Logo
Decentralizing Knowledge