Network


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

Hotspot


Dive into the research topics where Jacqueline M. Mason is active.

Publication


Featured researches published by Jacqueline M. Mason.


Genes & Development | 2011

Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress

Kathrin Zaugg; Yi Yao; Patrick T. Reilly; Karuppiah Kannan; Reza Kiarash; Jacqueline M. Mason; Ping Huang; Suzanne K. Sawyer; Benjamin J. Fuerth; Brandon Faubert; Tuula Kalliomaki; Andrew Elia; Xunyi Luo; Vincent Nadeem; David Bungard; Sireesha Yalavarthi; Joseph D. Growney; Andrew Wakeham; Yasmin Moolani; Jennifer Silvester; Annick You Ten; Walbert J. Bakker; Katsuya Tsuchihara; Shelley L. Berger; Richard P. Hill; Russell G. Jones; Ming Tsao; Murray Robinson; Craig B. Thompson; Guohua Pan

Tumor cells gain a survival/growth advantage by adapting their metabolism to respond to environmental stress, a process known as metabolic transformation. The best-known aspect of metabolic transformation is the Warburg effect, whereby cancer cells up-regulate glycolysis under aerobic conditions. However, other mechanisms mediating metabolic transformation remain undefined. Here we report that carnitine palmitoyltransferase 1C (CPT1C), a brain-specific metabolic enzyme, may participate in metabolic transformation. CPT1C expression correlates inversely with mammalian target of rapamycin (mTOR) pathway activation, contributes to rapamycin resistance in murine primary tumors, and is frequently up-regulated in human lung tumors. Tumor cells constitutively expressing CPT1C show increased fatty acid (FA) oxidation, ATP production, and resistance to glucose deprivation or hypoxia. Conversely, cancer cells lacking CPT1C produce less ATP and are more sensitive to metabolic stress. CPT1C depletion via siRNA suppresses xenograft tumor growth and metformin responsiveness in vivo. CPT1C can be induced by hypoxia or glucose deprivation and is regulated by AMPKα. Cpt1c-deficient murine embryonic stem (ES) cells show sensitivity to hypoxia and glucose deprivation and altered FA homeostasis. Our results indicate that cells can use a novel mechanism involving CPT1C and FA metabolism to protect against metabolic stress. CPT1C may thus be a new therapeutic target for the treatment of hypoxic tumors.


Molecular Cell | 2015

Targeting Mitosis in Cancer: Emerging Strategies

Carmen Dominguez-Brauer; Kelsie L. Thu; Jacqueline M. Mason; Heiko Blaser; Mark R. Bray; Tak W. Mak

The cell cycle is an evolutionarily conserved process necessary for mammalian cell growth and development. Because cell-cycle aberrations are a hallmark of cancer, this process has been the target of anti-cancer therapeutics for decades. However, despite numerous clinical trials, cell-cycle-targeting agents have generally failed in the clinic. This review briefly examines past cell-cycle-targeted therapeutics and outlines how experience with these agents has provided valuable insight to refine and improve anti-mitotic strategies. An overview of emerging anti-mitotic approaches with promising pre-clinical results is provided, and the concept of exploiting the genomic instability of tumor cells through therapeutic inhibition of mitotic checkpoints is discussed. We believe this strategy has a high likelihood of success given its potential to enhance therapeutic index by targeting tumor-specific vulnerabilities. This reasoning stimulated our development of novel inhibitors targeting the critical regulators of genomic stability and the mitotic checkpoint: AURKA, PLK4, and Mps1/TTK.


Oncogene | 2014

Role of Nek2 on centrosome duplication and aneuploidy in breast cancer cells

Paola Cappello; H. Blaser; Chiara Gorrini; D. C. C. Lin; Andrew J. Elia; Andrew Wakeham; S. Haider; P. C. Boutros; Jacqueline M. Mason; Naomi Miller; Bruce Youngson; Susan J. Done; Tak W. Mak

Breast cancer is the most common solid tumor and the second most common cause of death in women. Despite a large body of literature and progress in breast cancer research, many molecular aspects of this complex disease are still poorly understood, hindering the design of specific and effective therapeutic strategies. To identify the molecules important in breast cancer progression and metastasis, we tested the in vivo effects of inhibiting the functions of various kinases and genes involved in the regulation/modulation of the cytoskeleton by downregulating them in mouse PyMT mammary tumor cells and human breast cancer cell lines. These kinases and cytoskeletal regulators were selected based on their prognostic values for breast cancer patient survival. PyMT tumor cells, in which a selected gene was stably knocked down were injected into the tail veins of mice, and the formation of tumors in the lungs was monitored. One of the several genes found to be important for tumor growth in the lungs was NIMA-related kinases 2 (Nek2), a cell cycle-related protein kinase. Furthermore, Nek2 was also important for tumor growth in the mammary fat pad. In various human breast cancer cell lines, Nek2 knockdown induced aneuploidy and cell cycle arrest that led to cell death. Significantly, the breast cancer cell line most sensitive to Nek2 depletion was of the triple negative breast cancer subtype. Our data indicate that Nek2 has a pivotal role in breast cancer growth at primary and secondary sites, and thus may be an attractive and novel therapeutic target for this disease.


Journal of Medicinal Chemistry | 2013

The Discovery of PLK4 Inhibitors: (E)-3-((1H-Indazol-6-yl)methylene)indolin-2-ones as Novel Antiproliferative Agents

Radoslaw Laufer; Bryan T. Forrest; Sze-Wan Li; Yong Liu; Peter Sampson; Louise Edwards; Yunhui Lang; Donald E. Awrey; Guodong Mao; Olga Plotnikova; Genie Leung; Richard Hodgson; I. P. Beletskaya; Jacqueline M. Mason; Xunyi Luo; Xin Wei; Yi Yao; Miklos Feher; Fuqiang Ban; Reza Kiarash; Erin Green; Tak W. Mak; Guohua Pan; Henry W. Pauls

The family of Polo-like kinases is important in the regulation of mitotic progression; this work keys on one member, namely Polo-like kinase 4 (PLK4). PLK4 has been identified as a candidate anticancer target which prompted a search for potent and selective inhibitors of PLK4. The body of the paper describes lead generation and optimization work which yielded nanomolar PLK4 inhibitors. Lead generation began with directed virtual screening, using a ligand-based focused library and a PLK4 homology model. Validated hits were used as starting points for the design and discovery of PLK4 inhibitors of novel structure, namely (E)-3-((1H-indazol-6-yl)methylene)indolin-2-ones. Computational models, based on a published X-ray structure (PLK4 kinase domain), were used to understand and optimize the in vitro activity of the series; potent antiproliferative activity was obtained. The kinase selectivity profile and cell cycle analysis of selected inhibitors are described. The results of a xenograft study with an optimized compound 50 (designated CFI-400437) support the potential of these novel PLK4 inhibitors for cancer therapy.


Oncogene | 2014

PTPN12 promotes resistance to oxidative stress and supports tumorigenesis by regulating FOXO signaling

Isaac S. Harris; H. Blaser; J Moreno; Aislinn E. Treloar; Chiara Gorrini; Masato Sasaki; Jacqueline M. Mason; Christiane B. Knobbe; Alessandro Rufini; M Hallé; Andrew J. Elia; Andrew Wakeham; Michel L. Tremblay; Gerry Melino; Susan J. Done; Tak W. Mak

It is well known that protein tyrosine phosphatases (PTPs) that become oxidized due to exposure to reactive oxygen species (ROS) undergo a conformational change and are inactivated. However, whether PTPs can actively regulate ROS levels in order to prevent PTP inhibition has yet to be investigated. Here, we demonstrate that PTP non-receptor type 12 (PTPN12) protects cells against aberrant ROS accumulation and death induced by oxidative stress. Murine embryonic fibroblasts (MEFs) deficient in PTPN12 underwent increased ROS-induced apoptosis under conditions of antioxidant depletion. Cells lacking PTPN12 also showed defective activation of FOXO1/3a, transcription factors required for the upregulation of several antioxidant genes. PTPN12-mediated regulation of ROS appeared to be mediated by phosphoinositide-dependent kinase-1 (PDK1), which was hyperstimulated in the absence of PTPN12. As tight regulation of ROS to sustain survival is a key feature of cancer cells, we examined PTPN12 levels in tumors from a cohort of breast cancer patients. Patients whose tumors showed high levels of PTPN12 transcripts had a significantly poorer prognosis. Analysis of tissues from patients with various breast cancer subtypes revealed that more triple-negative breast cancers, the most aggressive breast cancer subtype, showed high PTPN12 expression than any other subtype. Furthermore, both human breast cancer cells and mouse mammary epithelial tumor cells engineered to lack PTPN12 exhibited reduced tumorigenic and metastatic potential in vivo that correlated with their elevated ROS levels. The involvement of PTPN12 in the antioxidant response of breast cancer cells suggests that PTPN12 may represent a novel therapeutic target for this disease.


Bioorganic & Medicinal Chemistry | 2014

Discovery of inhibitors of the mitotic kinase TTK based on N-(3-(3-sulfamoylphenyl)-1H-indazol-5-yl)-acetamides and carboxamides

Radoslaw Laufer; Grace Ng; Yong Liu; Narendra Kumar B. Patel; Louise Edwards; Yunhui Lang; Sze-Wan Li; Miklos Feher; Don E. Awrey; Genie Leung; Irina Beletskaya; Olga Plotnikova; Jacqueline M. Mason; Richard Hodgson; Xin Wei; Guodong Mao; Xunyi Luo; Ping Huang; Erin Green; Reza Kiarash; Dan Chi-Chia Lin; Marees Harris-Brandts; Fuqiang Ban; Vincent Nadeem; Tak W. Mak; Guohua J. Pan; Wei Qiu; Nickolay Y. Chirgadze; Henry W. Pauls

TTK kinase was identified by in-house siRNA screen and pursued as a tractable, novel target for cancer treatment. A screening campaign and systematic optimization, supported by computer modeling led to an indazole core with key sulfamoylphenyl and acetamido moieties at positions 3 and 5, respectively, establishing a novel chemical class culminating in identification of 72 (CFI-400936). This potent inhibitor of TTK (IC50=3.6nM) demonstrated good activity in cell based assay and selectivity against a panel of human kinases. A co-complex TTK X-ray crystal structure and results of a xenograft study with TTK inhibitors from this class are described.


Journal of Medicinal Chemistry | 2015

The Discovery of Orally Bioavailable Tyrosine Threonine Kinase (TTK) Inhibitors: 3-(4-(heterocyclyl)phenyl)-1H-indazole-5-carboxamides as Anticancer Agents

Yong Liu; Yunhui Lang; Narendra Kumar B. Patel; Grace Ng; Radoslaw Laufer; Sze-Wan Li; Louise Edwards; Bryan T. Forrest; Peter Sampson; Miklos Feher; Fuqiang Ban; Donald E. Awrey; I. P. Beletskaya; Guodong Mao; Richard Hodgson; Olga Plotnikova; Wei Qiu; Nickolay Y. Chirgadze; Jacqueline M. Mason; Xin Wei; Dan Chi-Chia Lin; Yi Che; Reza Kiarash; Brian Madeira; Graham C. Fletcher; Tak W. Mak; Mark R. Bray; Henry W. Pauls

The acetamido and carboxamido substituted 3-(1H-indazol-3-yl)benzenesulfonamides are potent TTK inhibitors. However, they display modest ability to attenuate cancer cell growth; their physicochemical properties, and attendant pharmacokinetic parameters, are not drug-like. By eliminating the polar 3-sulfonamide group and grafting a heterocycle at the 4 position of the phenyl ring, potent inhibitors with oral exposure were obtained. An X-ray cocrystal structure and a refined binding model allowed for a structure guided approach. Systematic optimization resulted in novel TTK inhibitors, namely 3-(4-(heterocyclyl)phenyl)-1H-indazole-5-carboxamides. Compounds incorporating the 3-hydroxy-8-azabicyclo[3.2.1]octan-8-yl bicyclic system were potent (TTK IC50 < 10 nM, HCT116 GI50 < 0.1 μM), displayed low off-target activity (>500×), and microsomal stability (T(1/2) > 30 min). A subset was tested in rodent PK and mouse xenograft models of human cancer. Compound 75 (CFI-401870) recapitulated the phenotype of TTK RNAi, demonstrated in vivo tumor growth inhibition upon oral dosing, and was selected for preclinical evaluation.


ACS Medicinal Chemistry Letters | 2016

Discovery of Pyrazolo[1,5-a]pyrimidine TTK Inhibitors: CFI-402257 is a Potent, Selective, Bioavailable Anticancer Agent

Yong Liu; Radoslaw Laufer; Narendra Kumar B. Patel; Grace Ng; Peter Sampson; Sze-Wan Li; Yunhui Lang; Miklos Feher; Richard D. Brokx; I. P. Beletskaya; Richard Hodgson; Olga Plotnikova; Donald E. Awrey; Wei Qiu; Nickolay Y. Chirgadze; Jacqueline M. Mason; Xin Wei; Dan Chi-Chia Lin; Yi Che; Reza Kiarash; Graham C. Fletcher; Tak W. Mak; Mark R. Bray; Henry W. Pauls

This work describes a scaffold hopping exercise that begins with known imidazo[1,2-a]pyrazines, briefly explores pyrazolo[1,5-a][1,3,5]triazines, and ultimately yields pyrazolo[1,5-a]pyrimidines as a novel class of potent TTK inhibitors. An X-ray structure of a representative compound is consistent with 1(1)/2 type inhibition and provides structural insight to aid subsequent optimization of in vitro activity and physicochemical and pharmacokinetic properties. Incorporation of polar moieties in the hydrophobic and solvent accessible regions modulates physicochemical properties while maintaining potency. Compounds with enhanced oral exposure were identified for xenograft studies. The work culminates in the identification of a potent (TTK K i = 0.1 nM), highly selective, orally bioavailable anticancer agent (CFI-402257) for IND enabling studies.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Functional characterization of CFI-402257, a potent and selective Mps1/TTK kinase inhibitor, for the treatment of cancer

Jacqueline M. Mason; Xin Wei; Graham C. Fletcher; Reza Kiarash; Richard D. Brokx; Richard Hodgson; Irina Beletskaya; Mark R. Bray; Tak W. Mak

Significance At present, microtubule-targeting agents are the most important antimitotic drugs used in the clinic. However, there is an urgent need for the discovery of new approaches to more effectively target tumor cells with less toxicity. Emerging strategies for anticancer therapy include exploiting cell-cycle checkpoint vulnerabilities and genomic instability in cancer cells. The spindle assembly checkpoint (SAC) is important for cell survival, and its inactivation generates lethal genomic instability in cancer cells. Inhibition of SAC signaling through targeting of monopolar spindle 1 (Mps1) has provided an indication of the feasibility of such an approach. We report here the cellular and antitumor effects of CFI-402257, a potent and specific small-molecule inhibitor of Mps1. CFI-402257 is currently in a phase I clinical trial (ClinicalTrials.gov ID: NCT02792465). Loss of cell-cycle control is a hallmark of human cancer. Cell-cycle checkpoints are essential for maintaining genome integrity and balanced growth and division. They are specifically deregulated in cancer cells and contain regulators that represent potential therapeutic targets. Monopolar spindle 1 (Mps1; also known as TTK protein kinase) is a core component of the spindle assembly checkpoint (SAC), a genome-surveillance mechanism that is important for cell survival, and has emerged as a candidate target for anticancer therapy. Here, we report the cellular and antitumor effects of CFI-402257, a potent (Mps1 Ki = 0.09 ± 0.02 nM; cellular Mps1 EC50 = 6.5 ± 0.5 nM), highly selective, and orally active small-molecule inhibitor of Mps1 that was identified through a drug-discovery program. Human cancer cells treated with CFI-402257 exhibit effects consistent with Mps1 kinase inhibition, specifically SAC inactivation, leading to chromosome missegregation, aneuploidy, and ultimately cell death. Oral administration of CFI-402257 in monotherapy or in combination with an anti-programmed cell death 1 (PD-1) antibody in mouse models of human cancer results in inhibition of tumor growth at doses that are well-tolerated. Our findings provide a rationale for the clinical evaluation of CFI-402257 in patients with solid tumors.


Cell Death & Differentiation | 2016

The acetyltransferase Tip60 contributes to mammary tumorigenesis by modulating DNA repair

Christian Bassi; Y.-T. Li; K. Khu; F. Mateo; P. S. Baniasadi; Andrew J. Elia; Jacqueline M. Mason; Vuk Stambolic; M. A. Pujana; Tak W. Mak; Chiara Gorrini

The acetyltransferase Tip60/Kat5 acetylates both histone and non-histone proteins, and is involved in a variety of biological processes. By acetylating p53, Tip60 controls p53-dependent transcriptional activity and so is implicated as a tumor suppressor. However, many breast cancers with low Tip60 also show p53 mutation, implying that Tip60 has a tumor suppressor function independent of its acetylation of p53. Here, we show in a p53-null mouse model of sporadic invasive breast adenocarcinoma that heterozygosity for Tip60 deletion promotes mammary tumorigenesis. Low Tip60 reduces DNA repair in normal and tumor mammary epithelial cells, both under resting conditions and following genotoxic stress. We demonstrate that Tip60 controls homologous recombination (HR)-directed DNA repair, and that Tip60 levels correlate inversely with a gene expression signature associated with defective HR-directed DNA repair. In human breast cancer data sets, Tip60 mRNA is downregulated, with low Tip60 levels correlating with p53 mutations in basal-like breast cancers. Our findings indicate that Tip60 is a novel breast tumor suppressor gene whose loss results in genomic instability leading to cancer formation.

Collaboration


Dive into the Jacqueline M. Mason's collaboration.

Top Co-Authors

Avatar

Tak W. Mak

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Reza Kiarash

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Xin Wei

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Mark R. Bray

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Miklos Feher

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Richard Hodgson

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Donald E. Awrey

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Radoslaw Laufer

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Sze-Wan Li

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Yong Liu

University Health Network

View shared research outputs
Researchain Logo
Decentralizing Knowledge