22 results on '"Hanne Merritt"'
Search Results
2. The structural mechanism of human glycogen synthesis by the GYS1-GYG1 complex
- Author
-
Nathan M. Fastman, Yuxi Liu, Vyas Ramanan, Hanne Merritt, Eileen Ambing, Anna A. DePaoli-Roach, Peter J. Roach, Thomas D. Hurley, Kevin T. Mellem, Julie C. Ullman, Eric Green, David Morgans, Jr., and Christos Tzitzilonis
- Subjects
CP: Molecular biology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
3. Abstract 4945: Inhibition of wild-type PI3Kα signaling is required for durable efficacy in PI3Kα mutant cancer cells due to robust re-activation of wild-type PI3Kα signaling
- Author
-
John R. MacDougall, Mengqi Zhong, Hanne Merritt, Joselyn S. Del Cid, John Bradley, Raymond Mak, Neil Dhawan, and Wei Chen
- Subjects
Cancer Research ,Oncology - Abstract
PI3Kα is the most mutated oncogene with a high mutation rate in breast, lung, colorectal, gastric, bladder, and other tumor types. However, current inhibitors have had little efficacy in the clinic due to their inability to achieve maximal PI3Kα target engagement in a highly specific and durable manner. Contributing to this is that most PI3Kα mutant cancer cells are prone to robust pathway feedback and re-activation of remaining wild type PI3Kα signaling upon initial inhibition. We have analyzed pathway feedback in a variety of PI3Kα mutant cancer cell lines across multiple diverse indications and found this to be a common trait among PI3Kα mutant cancer cells. Additionally, when the mutant PI3Kα protein alone is targeted in these cells using mutant-specific inhibitors, we observed robust pathway re-activation and tumor cell proliferation. In contrast, TOS-358, a highly selective first-in-class covalent PI3Kα inhibitor, achieved complete inhibition of both PI3Kα mutant and wild type signaling and this correlated with the induction of cell cycle arrest and cell death across a wide variety of PI3Kα mutant tumor types. Furthermore, stimulation of PI3Kα signaling with serum rapidly re-activated the PI3K-AKT pathway in the presence of reversible and mutant specific inhibitors. Covalent molecules (namely TOS-358) achieved complete inhibition of pathway signaling in this format. Overall, this reveals the need to achieve sustained inhibition of both PI3Kα mutant and wild type signaling to achieve durable efficacy in PI3Kα mutant tumors. Citation Format: John R. MacDougall, Mengqi Zhong, Hanne Merritt, Joselyn S. Del Cid, John Bradley, Raymond Mak, Neil Dhawan, Wei Chen. Inhibition of wild-type PI3Kα signaling is required for durable efficacy in PI3Kα mutant cancer cells due to robust re-activation of wild-type PI3Kα signaling. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4945.
- Published
- 2023
- Full Text
- View/download PDF
4. Two Distinct Mechanisms of Inhibition of LpxA Acyltransferase Essential for Lipopolysaccharide Biosynthesis
- Author
-
Ramadevi Prathapam, Alexandra Frommlet, Jacob Shaul, Dita M. Rasper, Xiaoyu Shen, Alexey Ruzin, Sharadha Subramanian, Feng Wang, Micah Steffek, Bret Benton, Jason Vo, Steven Shia, Wooseok Han, Andreas O. Frank, Charles Wartchow, Patrick Lee, Xiaolei Ma, Fergal Casey, Hanne Merritt, Carl J. Balibar, Alun Bermingham, Elizabeth Ornelas, Tsuyoshi Uehara, Andreas Lingel, Chi-Min Ho, Barbara Chie-Leon, William S. Sawyer, Min-Kyu Cho, Sylvia Ma, Katherine R Prosen, Min Li, Christopher M. Rath, and Gianfranco De Pascale
- Subjects
Microbial Sensitivity Tests ,Crystallography, X-Ray ,010402 general chemistry ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,Catalysis ,Colloid and Surface Chemistry ,Escherichia coli ,medicine ,Enzyme Inhibitors ,chemistry.chemical_classification ,biology ,Chemistry ,Drug discovery ,Imidazoles ,General Chemistry ,biology.organism_classification ,In vitro ,Anti-Bacterial Agents ,0104 chemical sciences ,Enzyme ,Acyltransferase ,Pyrazoles ,Antibacterial activity ,Uncompetitive inhibitor ,Acyltransferases ,Bacteria ,Protein Binding - Abstract
The lipopolysaccharide biosynthesis pathway is considered an attractive drug target against the rising threat of multi-drug-resistant Gram-negative bacteria. Here, we report two novel small-molecule inhibitors (compounds 1 and 2) of the acyltransferase LpxA, the first enzyme in the lipopolysaccharide biosynthesis pathway. We show genetically that the antibacterial activities of the compounds against efflux-deficient Escherichia coli are mediated by LpxA inhibition. Consistently, the compounds inhibited the LpxA enzymatic reaction in vitro. Intriguingly, using biochemical, biophysical, and structural characterization, we reveal two distinct mechanisms of LpxA inhibition; compound 1 is a substrate-competitive inhibitor targeting apo LpxA, and compound 2 is an uncompetitive inhibitor targeting the LpxA/product complex. Compound 2 exhibited more favorable biological and physicochemical properties than compound 1 and was optimized using structural information to achieve improved antibacterial activity against wild-type E. coli. These results show that LpxA is a promising antibacterial target and imply the advantages of targeting enzyme/product complexes in drug discovery.
- Published
- 2020
- Full Text
- View/download PDF
5. In-vitro characterization of MZE001, an orally active GYS1 inhibitor to treat Pompe disease
- Author
-
Rebeca Choy, Daniel Reiton, Hanne Merritt, Tarunmeet Gujral, Sandya Ganesh, Aleksandra Denisin, Hannah Powers, Walter Won, Terrence Satterfield, Julie Ullman, Kevin Mellem, Eric Green, David Morgans, and David Beattie
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Genetics ,Molecular Biology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
6. Pharmacology of small molecule inhibitors of GYS1 in a mouse model of Pompe disease
- Author
-
Yannan Xi, Terrence F. Satterfield, Rebeca Choy, Tarunmeet Gujral, Eva Situ, Samnang Tep, Vivian Morton, Cecile Yu, Daniel Reiton, Baiwei Lin, Hanne Merritt, Pam Santiago, Bryan Espanol, Ryan A. Dick, Julie C. Ullman, Kevin T. Mellem, Sarah B. Noonberg, Eric M. Green, David J. Morgans, Sanjay J. Chandriani, and David T. Beattie
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Genetics ,Molecular Biology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
7. Substrate reduction therapy for Pompe disease: Small molecule inhibition of glycogen synthase 1 in preclinical models
- Author
-
Julie C. Ullman, Kevin T. Mellem, Yannan Xi, Terrence F. Satterfield, Tarunmeet Gujral, Rebeca Choy, Julian R. Homburger, Hanne Merritt, Daniel Reiton, Samnang Tep, Baiwei Lin, Cecile Yu, Eva Situ, Walter Won, Hannah Powers, Vyas Ramanan, Maarten Hoek, Robert Graham, Sanjay J. Chandriani, Christopher Sinz, Ryan A. Dick, Sarah B. Noonberg, David T. Beattie, David J. Morgans, null Jr, and Eric M. Green
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Genetics ,Molecular Biology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
8. Imidazo[1,2- a ]pyridin-6-yl-benzamide analogs as potent RAF inhibitors
- Author
-
John Chan, Sandra W. Cowan-Jacob, Takanori Kanazawa, Zheng Chen, Brent A. Appleton, Aaron Smith, Qiong Zhang, Daniel Poon, Clemens Scheufler, Darrin Stuart, Kay Huh, Jeremy Murray, Kevin Shoemaker, Zilin Huang, Hanne Merritt, Susan Kaufman, Laura Tandeske, Cynthia M. Shafer, Zhi-Jie Ni, and Johanna M. Jansen
- Subjects
Models, Molecular ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,MAPK/ERK pathway ,Stereochemistry ,VEGF receptors ,Clinical Biochemistry ,Pharmaceutical Science ,Heterocyclic Compounds, 2-Ring ,Biochemistry ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Humans ,Transferase ,Benzamide ,Protein Kinase Inhibitors ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Kinase ,Organic Chemistry ,Combinatorial chemistry ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Benzamides ,biology.protein ,Molecular Medicine ,Selectivity - Abstract
A series of imidazo[1,2-a]pyridin-6-yl-benzamide analogs was designed as inhibitors of B-RAFV600E. Medicinal chemistry techniques were employed to explore the SAR for this series and improve selectivity versus P38 and VEGFR2.
- Published
- 2017
- Full Text
- View/download PDF
9. Two distinct mechanisms of small molecule inhibition of LpxA acyltransferase essential for lipopolysaccharide biosynthesis
- Author
-
Xiaoyu Shen, William S. Sawyer, Micah Steffek, Wooseok Han, Tsuyoshi Uehara, Andreas O. Frank, Sharadha Subramanian, Alexey Ruzin, Feng Wang, Alexandra Frommlet, C. M. Baxter Rath, Hanne Merritt, Min Li, Alun Bermingham, Elizabeth Ornelas, Jason Vo, Fergal Casey, B. Chie-Leon, Andreas Lingel, Bret Benton, Xiaolei Ma, Steven Shia, Carl J. Balibar, G. de Pascale, Jacob Shaul, Patrick Lee, Dita M. Rasper, Min-Kyu Cho, Charles Wartchow, Sylvia Ma, Chi-Min Ho, Katherine R Prosen, and Ramadevi Prathapam
- Subjects
chemistry.chemical_classification ,Enzyme ,Biochemistry ,Chemistry ,Drug discovery ,Acyltransferase ,medicine ,Wild type ,medicine.disease_cause ,Uncompetitive inhibitor ,Antibacterial activity ,Small molecule ,Escherichia coli - Abstract
The lipopolysaccharide biosynthesis pathway is considered an attractive drug target against the rising threat of multidrug-resistant Gram-negative bacteria. Here, we report two novel small-molecule inhibitors (compounds 1 and 2) of the acyltransferase LpxA, the first enzyme in the lipopolysaccharide biosynthesis pathway. We show genetically that the antibacterial activities of the compounds against efflux-deficient Escherichia coli are mediated by LpxA inhibition. Consistently, the compounds inhibited the LpxA enzymatic reaction in vitro. Intriguingly, using biochemical, biophysical, and structural characterization, we reveal two distinct mechanisms of LpxA inhibition; compound 1 is a substrate-competitive inhibitor targeting apo LpxA and compound 2 is an uncompetitive inhibitor targeting the LpxA-product complex. Compound 2 exhibited more favorable biological and physicochemical properties than compound 1, and was optimized using structural information to achieve improved antibacterial activity against wild type E. coli. These results show that LpxA is a promising antibacterial target and imply the advantages of targeting enzyme-product complexes in drug discovery.
- Published
- 2019
- Full Text
- View/download PDF
10. Discovery of imidazo[1,2- a ]-pyridine inhibitors of pan-PI3 kinases that are efficacious in a mouse xenograft model
- Author
-
Robert Elling, Aaron Smith, Matthew Burger, Kay Huh, Hanne Merritt, Sabina Pecchi, Charles Voliva, Jiong Lan, Wooseok Han, Daniel Menezes, John Chan, Teresa E. Williams, Yongjin Xu, Mark Knapp, Joelle Verhagen, Susan Kaufman, and Zhi-Jie Ni
- Subjects
0301 basic medicine ,Imidazopyridine ,Pyridines ,Stereochemistry ,Clinical Biochemistry ,Drug Evaluation, Preclinical ,Pharmaceutical Science ,Antineoplastic Agents ,Stereoisomerism ,Crystallography, X-Ray ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Protein Kinase Inhibitors ,Molecular Biology ,Phosphoinositide-3 Kinase Inhibitors ,Ovarian Neoplasms ,Binding Sites ,Trifluoromethyl ,Organic Chemistry ,Protein Structure, Tertiary ,Rats ,Enzyme Activation ,Molecular Docking Simulation ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Benzothiazole ,030220 oncology & carcinogenesis ,Alkoxy group ,Heterografts ,Molecular Medicine ,Female ,Enantiomer ,Half-Life - Abstract
Alterations in PI3K/AKT signaling are known to be implicated with tumorigenesis. The PI3 kinases family of lipid kinases has been an attractive therapeutic target for cancer treatment. Imidazopyridine compound 1, a potent, selective, and orally available pan-PI3K inhibitor, identified by scaffold morphing of a benzothiazole hit, was further optimized in order to achieve efficacy in a PTEN-deleted A2780 ovarian cancer mouse xenograft model. With a hypothesis that a planar conformation between the core and the 6-heteroaryl ring will allow for the accommodation of larger 5'-substituents in a hydrophobic area under P-loop, SAR efforts focused on 5'-alkoxy heteroaryl rings at the 6-position of imidazopyridine and imidazopyridazine cores that have the same dihedral angle of zero degrees. 6'-Alkoxy 5'-aminopyrazines in the imidazopyridine series were identified as the most potent compounds in the A2780 cell line. Compound 14 with 1,1,1-trifluoroisopropoxy group at 6'-position demonstrated excellent potency and selectivity, good oral exposure in rats and in vivo efficacy in A2780 tumor-bearing mouse. Also, we disclose the X-ray co-crystal structure of one enantiomer of compound 14 in PI3Kα, confirming that the trifluoromethyl group fits nicely in the hydrophobic hot spot under P-loop.
- Published
- 2016
- Full Text
- View/download PDF
11. Expression, purification and characterization of inactive and active forms of ERK2 from insect expression system
- Author
-
Isabel Zaror, Kenneth Crawford, Mohammad Hekmat-Nejad, Stephania Widger, Janet Sim, Hanne Merritt, Kelly Yan, Gwynn Pardee, and Jan Marie Cheng
- Subjects
Recombinant Fusion Proteins ,MAP Kinase Kinase 1 ,Gene Expression ,Spodoptera ,Biology ,environment and public health ,Serine ,Gene expression ,Escherichia coli ,Sf9 Cells ,Animals ,Humans ,Histidine ,Cloning, Molecular ,Phosphorylation ,Protein kinase A ,Mitogen-Activated Protein Kinase 1 ,Kinase ,Drug discovery ,Assay ,Biochemical Activity ,Small molecule ,Enzyme Activation ,Kinetics ,enzymes and coenzymes (carbohydrates) ,Biochemistry ,biological phenomena, cell phenomena, and immunity ,Genetic Engineering ,Baculoviridae ,Oligopeptides ,hormones, hormone substitutes, and hormone antagonists ,Plasmids ,Biotechnology - Abstract
Extracellular signal-regulated kinase 2 (ERK2) is a serine/threonine protein kinase involved in many cellular programs, such as cell proliferation, differentiation, motility and programed cell-death. It is therefore considered an important target in the treatment of cancer. In an effort to support biochemical screening and small molecule drug discovery, we established a robust system to generate both inactive and active forms of ERK2 using insect expression system. We report here, for the first time, that inactive ERK2 can be expressed and purified with 100% homogeneity in the unphosphorylated form using insect system. This resulted in a significant 20-fold yield improvement compared to that previously reported using bacterial expression system. We also report a newly developed system to generate active ERK2 in insect cells through in vivo co-expression with a constitutively active MEK1 (S218D S222D). Isolated active ERK2 was confirmed to be doubly phosphorylated at the correct sites, T185 and Y187, in the activation loop of ERK2. Both ERK2 forms, inactive and active, were well characterized by biochemical activity assay for their kinase function. Inactive and active ERK2 were the two key reagents that enabled successful high through-put biochemical assay screen and structural drug discovery studies.
- Published
- 2015
- Full Text
- View/download PDF
12. Design and Discovery of N-(2-Methyl-5'-morpholino-6'-((tetrahydro-2H-pyran-4-yl)oxy)-[3,3'-bipyridin]-5-yl)-3-(trifluoromethyl)benzamide (RAF709): A Potent, Selective, and Efficacious RAF Inhibitor Targeting RAS Mutant Cancers
- Author
-
Yan Lou, Sharadha Subramanian, Yingyun Wang, Lifeng Wan, John Tellew, Laura Tandeske, Benjamin R. Taft, Kalyani Gampa, Jacob R. Haling, Gisele Nishiguchi, Lina Setti, Alice Rico, Sylvia Ma, Payman Amiri, Mallika Singh, Huw Tanner, Brent A. Appleton, Robert J. Aversa, Sepideh Vaziri, Shenlin Huang, Johanna M. Jansen, Anne Van Abbema, Jing Yuan, Vesselina G. Cooke, Hanne Merritt, Aaron Smith, Wenlin Shao, Valery Polyakov, Fei Feng, Savithri Ramurthy, Matthew Burger, Mulugeta Mamo, Lesley A. Mathews Griner, Vijay Sethuraman, Victoriano Tamez, Michael Patrick Dillon, Emma Lees, Ina Dix, Paul A. Barsanti, Richard Zang, Darrin Stuart, and Mohammad Hekmat-Nejad
- Subjects
0301 basic medicine ,Proto-Oncogene Proteins B-raf ,Mutant ,Antineoplastic Agents ,medicine.disease_cause ,Crystallography, X-Ray ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,0302 clinical medicine ,2,2'-Dipyridyl ,Dogs ,Drug Stability ,Neoplasms ,Drug Discovery ,medicine ,Structure–activity relationship ,Animals ,Humans ,c-Raf ,Molecular Targeted Therapy ,Benzamide ,Chemistry ,Kinase ,Drug discovery ,Small molecule ,Xenograft Model Antitumor Assays ,Rats ,030104 developmental biology ,Biochemistry ,030220 oncology & carcinogenesis ,Drug Design ,Benzamides ,ras Proteins ,Molecular Medicine ,raf Kinases ,KRAS - Abstract
RAS oncogenes have been implicated in30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.
- Published
- 2017
13. Identification of NVP-BKM120 as a Potent, Selective, Orally Bioavailable Class I PI3 Kinase Inhibitor for Treating Cancer
- Author
-
Susan Kaufman, Zhang Yanchen, Joshua Haznedar, Daniel Menezes, Matthew Burger, Xiaohua Xin, Aaron Smith, Frazier Kelly, Sauveur-Michel Maira, Allan S. Wagman, Zhi-Jie Ni, Keith B. Pfister, Kay Huh, Isabel Zaror, Thomas Hendrickson, Joelle Verhagen, Gordana Atallah, Kevin Shoemaker, Bartulis Sarah, Michael Chin, Simon Ng, Kenneth Crawford, Dirksen E. Bussiere, Ed Iwanowicz, Mark Knapp, Isabelle Lee, Hanne Merritt, Marion Wiesmann, Charles Voliva, and Sabina Pecchi
- Subjects
Phosphoinositide 3-kinase ,biology ,business.industry ,Kinase ,Organic Chemistry ,Buparlisib ,Phases of clinical research ,Cancer ,Pharmacology ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,chemistry ,In vivo ,Drug Discovery ,biology.protein ,Medicine ,Signal transduction ,business ,PI3K/AKT/mTOR pathway - Abstract
Phosphoinositide-3-kinases (PI3Ks) are important oncology targets due to the deregulation of this signaling pathway in a wide variety of human cancers. Herein we describe the structure guided optimization of a series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines where the pharmacokinetic properties were improved by modulating the electronics of the 6-position heterocycle, and the overall druglike properties were fine-tuned further by modification of the 4-position substituent. The resulting 2,4-bismorpholino 6-heterocyclic pyrimidines are potent class I PI3K inhibitors showing mechanism modulation in PI3K dependent cell lines and in vivo efficacy in tumor xenograft models with PI3K pathway deregulation (A2780 ovarian and U87MG glioma). These efforts culminated in the discovery of 15 (NVP-BKM120), currently in Phase II clinical trials for the treatment of cancer.
- Published
- 2011
- Full Text
- View/download PDF
14. Identification and structure–activity relationship of 2-morpholino 6-(3-hydroxyphenyl) pyrimidines, a class of potent and selective PI3 kinase inhibitors
- Author
-
Hendrickson Thomas, Charles Voliva, Kevin Shoemaker, Mark Knapp, Elizabeth Ornelas, Sabina Pecchi, Paul A. Renhowe, Wendy J. Fantl, Clarke Taylor, Hanne Merritt, Marion Wiesmann, and Susan Kaufman
- Subjects
Morpholino ,Morpholines ,Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,Biochemistry ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Humans ,Structure–activity relationship ,Phosphorylation ,Protein Kinase Inhibitors ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,MAPK14 ,biology ,Kinase ,Chemistry ,Organic Chemistry ,Pyrimidines ,Mitogen-activated protein kinase ,biology.protein ,Molecular Medicine ,Signal transduction - Abstract
PI3 Kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration, and differentiation. The PI3 kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations, and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe a novel series of PI3K inhibitors sharing a pyrimidine core and showing significant potency against class I PI3 kinases in the biochemical assay and in cells. The discovery, synthesis and SAR of this chemotype are described.
- Published
- 2010
- Full Text
- View/download PDF
15. Internal AU-rich elements modulate activity of two competing 3' splice sites in plant nuclei
- Author
-
Hanne Merritt, Andrew J. McCullough, and Mary A. Schuler
- Subjects
RNA Splicing ,Molecular Sequence Data ,Guanosine ,Plant Science ,Biology ,medicine.disease_cause ,Zea mays ,Cytosine ,chemistry.chemical_compound ,Genetics ,medicine ,splice ,Dinucleotide Repeats ,Uracil ,AU-rich element ,Mutation ,Splice site mutation ,Base Sequence ,Adenine ,Alcohol Dehydrogenase ,Intron ,RNA ,Cell Biology ,chemistry ,RNA, Plant ,RNA splicing - Abstract
In vivo analyses using an autonomously replicating Agrobacterium/geminivirus vector have enabled identification of AU-rich intronic elements critical for 5' and 3' splice site selection in dicot plant nuclei and development of a model for pre-mRNA intron recognition in plant nuclei. To determine the minimal length, spacing and nucleotide compositions constraining recognition of the 3' boundary of an intron, two or four nucleotide substitutions have been introduced into the two AU-rich elements located between 50 and 66 nucleotides upstream from the 3' splice site of maize Adh1 intron 3. In each case tested, substitutions in the distal left element (-62 to -66) inactivate the downstream 3' splice site at -1 more effectively than substitutions in the proximal right element (-50 to -55). Guanosine or cytosine substitutions in either element reduce recognition of the -1 site significantly; adenosine substitutions have a less severe effect. Mutations in both of these AU elements additively block recognition of the downstream 3' splice site. The strong additive effect of these mutations supports a model in which short sets of AU islands bind interactive factors and cooperatively modulate usage of the downstream splice site. In contrast to the uridine requirements documented for the 3' terminus of plant introns, adenosines are partially interchangeable with uridines within this internal region of the intron.
- Published
- 1997
- Full Text
- View/download PDF
16. Structure guided optimization of a fragment hit to imidazopyridine inhibitors of PI3K
- Author
-
Aaron Smith, Matthew Burger, Charles Voliva, Kay Huh, John Chan, Sabina Pecchi, Johanna Janssen, Jiong Lan, Hanne Merritt, Marion Wiesmann, Wooseok Han, Zhi-Jie Ni, Susan Kaufman, and Mark Knapp
- Subjects
Models, Molecular ,Imidazopyridine ,Pyridines ,Clinical Biochemistry ,Cell ,Pharmaceutical Science ,Imides ,Biochemistry ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Gene duplication ,medicine ,Humans ,Benzothiazoles ,Enzyme Inhibitors ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Phosphoinositide-3 Kinase Inhibitors ,Chemistry ,Kinase ,Organic Chemistry ,Cancer ,medicine.disease ,In vitro ,Enzyme Activation ,medicine.anatomical_structure ,Benzothiazole ,Solubility ,Cancer research ,Molecular Medicine ,Female ,Azo Compounds - Abstract
PI3 kinases are a family of lipid kinases mediating numerous cell processes such as proliferation, migration and differentiation. The PI3 Kinase pathway is often de-regulated in cancer through PI3Kα overexpression, gene amplification, mutations and PTEN phosphatase deletion. PI3K inhibitors represent therefore an attractive therapeutic modality for cancer treatment. Herein we describe how the potency of a benzothiazole fragment hit was quickly improved based on structural information and how this early chemotype was further optimized through scaffold hopping. This effort led to the identification of a series of 2-acetamido-5-heteroaryl imidazopyridines showing potent in vitro activity against all class I PI3Ks and attractive pharmacokinetic properties.
- Published
- 2013
17. Abstract LB-121: Dissecting MAPK pathway in BRAFmut melanoma: Intricacies of ERK1 and ERK2
- Author
-
Michel Faure, Darrin Stuart, Mohammad Hekmat-Nejad, Karen Yu, Ken Crawford, Yumin Dai, Tatiana Zavorotinskaya, Upasana Mehra, Kelly Yan, Xiaolei Ma, Jan Xuan, Charles Voliva, Jan Marie Cheng, and Hanne Merritt
- Subjects
Genetics ,MAPK/ERK pathway ,Cancer Research ,Gene knockdown ,biology ,Cell growth ,Kinase ,Mutant ,medicine.disease_cause ,Receptor tyrosine kinase ,Cell biology ,Oncology ,RNA interference ,biology.protein ,medicine ,Carcinogenesis - Abstract
The MAPK signaling cascade, comprised of the RAS GTPases, the RAF, MEK1/2 and ERK1/2 kinases is frequently deregulated in cancer. ERK1 and ERK2 transmit signals generated by mutant BRAF, Ras or by activated receptor tyrosine kinases to a wide range of nuclear and cytoplasmic substrates, resulting in signal amplification, cell growth, migration and survival. ERK1 and ERK2 have been considered as redundant because of their high homology, large number of overlapping substrates, and ability to substitute for each other in genetically engineered mouse models. Nevertheless, several investigators have identified non-redundant roles for ERK isoforms in oncogenesis; for instance, ERK2, but not ERK1, appears to be responsible for RASmut induced epithelial-to-mesenchymal transformation. Besides, each of the ERK isoforms employs spatially distinct substrate docking domains, DEF (docking site for ERK FXFP) and D (docking domain), to signal to different subsets of substrates and differentially transmit signals downstream. We set out to determine the roles of ERK isoforms as well as DEF- and D-domain dependent signaling in the survival of melanoma tumor cells expressing activating BRAF mutations which are highly sensitive to pharmacological inhibitors of RAF, MEK1/2 and ERK1/2. We designed ERK1 and ERK2 mutants resistant to ATP-competitive ERK1/2 inhibitors and employed auto-activating, MEK-independent, ERK1 and ERK2 mutants to ask if BRAFmut melanoma survival is dependent on either or both ERK isoforms. In addition, we used RNAi and zinc-finger nucleases’ to knockdown or delete each of ERK isoforms. These experimental approaches consistently demonstrated that ERK2, but not ERK1, was the sole driver of cell survival in multiple BRAFmut melanoma cell lines. Moreover, genome-wide gene expression analysis indicated that ERK2, but not ERK1, was largely responsible for transcriptional effects imposed by pharmacological RAF, MEK1/2 or ERK1/2 inhibitors. Thus, in BRAFmut melanoma, functions of ERK1 and 2 are not redundant, and ERK1 cannot substitute for a disabled ERK2. Next, we introduced DEF- and D-substrate docking domain mutations into an ERK inhibitor resistant ERK2 to investigate whether signaling through either domain is sufficient to support melanoma survival. We observed that signaling through D- or DEF- domains of ERK2 had differential effects on gene expression and substrate phosphorylation. Consequently, we have found that a subset of melanoma cell lines was sensitive to elimination of DEF- docking domain interactions, whereas another subset of cell lines tolerated mutations in the DEF-site. Interactions and signaling through ERK D-docking site were dispensable for survival of all melanoma cell lines tested. These data suggest potential novel approaches to target oncogenic MAPK pathway. Citation Format: Tatiana Zavorotinskaya, Upasana Mehra, Yumin Dai, Michel Faure, Ken Crawford, Karen Yu, Jan Marie Cheng, Xiaolei Ma, Jan Xuan, Kelly Yan, Mohammad Hekmat-Nejad, Hanne Merritt, Darrin Stuart, Charles Voliva. Dissecting MAPK pathway in BRAFmut melanoma: Intricacies of ERK1 and ERK2. [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 LB-121. doi:10.1158/1538-7445.AM2014-LB-121
- Published
- 2014
- Full Text
- View/download PDF
18. Abstract LB-54: Identification of a RAF inhibitory auto-phosphorylation site
- Author
-
Zenhai Gao, Darrin Stuart, Hanne Merritt, Julie Lin, Matthew Holderfield, and Frank McCormick
- Subjects
MAPK/ERK pathway ,Cancer Research ,Phosphorylation sites ,business.industry ,Cancer ,Inhibitory postsynaptic potential ,Bioinformatics ,medicine.disease ,Oncology ,Activation loop ,Cancer cell ,Cancer research ,medicine ,Phosphorylation ,business ,Biochemical mechanism - Abstract
Preclinical studies have demonstrated that BRAF wild-type cancer cells are not only refractory but paradoxically activate the MAPK pathway when treated with RAF inhibitors. Potentially through a related mechanism, multiple point mutations in the activation loop and Phosphate binding loop (P. loop) domains of BRAF have been characterized in human cancers, which often render BRAF catalytically impaired, yet stimulate phosphorylation of downstream targets in cells. However, the underlying biochemistry causing these phenomena has yet to be fully understood. In this study we investigate the biochemical mechanism of wild-type RAF activation in response to RAF inhibitor treatment and identify a novel auto-phosphorylation site within the well conserved BRAF/CRAF P. loop. Disruption of the auto-phosphorylation, either through pharmacologic or genetic alterations, results in activation of the MAPK pathway. This work offers new insight into RAF biochemistry by uncovering a previously unrecognized regulatory mechanism of RAF kinases that is bypassed by the BRAF oncoproteins. Citation Format: Matthew Holderfield, Hanne Merritt, Julie Lin, Zenhai Gao, Darrin Stuart, Frank McCormick. Identification of a RAF inhibitory auto-phosphorylation site. [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 LB-54. doi:10.1158/1538-7445.AM2013-LB-54
- Published
- 2013
- Full Text
- View/download PDF
19. Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site
- Author
-
A M McCall, Robert S. Crawford, Michael Yim, Louis M. Weiner, Hanne Merritt, Keith W. Marshall, James D. Marks, Robert Schier, Gregory P. Adams, and Cara Marks
- Subjects
Models, Molecular ,Receptor, ErbB-2 ,Mutant ,Molecular Sequence Data ,Antibody Affinity ,chemical and pharmacologic phenomena ,Complementarity determining region ,Biology ,medicine.disease_cause ,Evolution, Molecular ,Epitopes ,Antigen ,Structural Biology ,In vivo ,medicine ,Tumor Cells, Cultured ,Humans ,Cloning, Molecular ,Molecular Biology ,Immunoglobulin Fragments ,chemistry.chemical_classification ,Mutation ,Alanine ,Base Sequence ,Cell Membrane ,Molecular biology ,Receptor–ligand kinetics ,In vitro ,Amino acid ,Kinetics ,chemistry ,Mutagenesis ,Binding Sites, Antibody - Abstract
We determined the extent to which additional binding energy could be achieved by diversifying the complementarity determining regions (CDRs) located in the center of the antibody combining site of C6.5, a human single-chain Fv (scFv) isolated from a non-immune phage library which binds the tumor antigen c-erbB-2. CDR3 of the light (V(L)) and heavy (V(H)) chain variable region of C6.5 were sequentially mutated, the mutant scFv displayed on phage, and higher affinity mutants selected on antigen. Mutation of V(L) CDR3 yielded a scFv (C6ML3-9) with a 16-fold lower Kd (1.0 x 10(-9) M) than C6.5. Due to its length of 20 amino acids, four V(H) CDR3 libraries of C6ML3-9 were constructed. The greatest increase in affinity from a single library was ninefold (Kd = 1.1 x 10(-10) M). Combination of mutations isolated from separate V(H) CDR3 libraries yielded additional ninefold decreases in Kd, resulting in a scFv with a 1230-fold increase in affinity from wild-type C6.5 (Kd = 1.3 x 10(-11) M). The increase in affinity, and its absolute value, are comparable to the largest values observed for antibody affinity maturation in vivo or in vitro and indicate that mutation of V(L) and V(H) CDR3 may be a particularly efficient means to increase antibody affinity. This result, combined with the location of amino acid conservation and substitution, suggests an overall strategy for in vitro antibody affinity maturation. In addition, the affinities and binding kinetics of the single-chain Fv provide reagents with potential tumor targeting abilities not previously available.
- Published
- 1996
20. Abstract 3790: Preclinical profile of LGX818: A potent and selective RAF kinase inhibitor
- Author
-
Nanxin Li, Richard Zang, Fernando Salangsang, Nancy Turner, Poon Daniel J, Kimberly Aardalen, Allen Li, Swarupa Kulkarni, Nancy Pryer, Majid Ghoddusi, Frank Sun, Shefali Kakar, John Tellew, Edward Lorenzana, Susan Kaufman, Giordano Caponigro, Hanne Merritt, and Darrin Stuart
- Subjects
Cancer Research ,business.industry ,Kinase ,Melanoma ,Phases of clinical research ,Cancer ,Raf Kinase Inhibitor ,Pharmacology ,medicine.disease ,Oncology ,Apoptosis ,Medicine ,Potency ,business ,IC50 - Abstract
Selective RAF inhibitors have significant activity in patients with metastatic melanoma whose tumors express BRAFV600E. However, not all patients respond equally well to treatment and the duration of response is often limited to less than 6 months. LGX818 was developed with the hypothesis that a more potent inhibitor with excellent pharmacological properties would maximize the degree and duration of patient response. LGX818 is a highly potent RAF inhibitor with selective anti-proliferative and apoptotic activity in cells expressing BRAFV600E. In the A375 (BRAFV600E) human melanoma cell line LGX818 suppresses phospho-ERK (EC50 = 3 nM) leading to potent inhibition of proliferation (EC50 = 4 nM). No significant activity was observed against a panel of 100 kinases (IC50 > 900 nM) and LGX818 did not inhibit proliferation of > 400 cell lines expressing wild-type BRAF. Contributing to the high potency of LGX818 is the extremely slow off-rate from BRAFV600E which is not observed with other RAF inhibitors. In biochemical assays the dissociation half-life was >24 hours which translated into sustained target inhibition in cells following drug wash-out. Single dose PK/PD studies in human melanoma xenograft models (BRAFV600E) indicated that LGX818 treatment at oral doses as low as 6 mg/kg resulted in strong (75%) and sustained (>24 hours) decrease in phospho-MEK, even following clearance of drug from circulation. Decreases in phospho-ERK were consistent with phospho-MEK but markers of downstream transcriptional output (DUSP6 and SPRY4) appeared to provide a more sensitive measure of pathway activation. LGX818 induced tumor regression in multiple BRAF mutant human tumor xenograft models grown in immune compromised mice and rats at doses as low as 1 mg/kg. Consistent with the in vitro data, LGX818 was inactive against BRAF wild-type tumors at doses up to 300 mg/kg bid, with good tolerability and linear increase in exposure. Efficacy was also achieved in a more disease-relevant spontaneous metastatic melanoma and a model of melanoma brain metastasis. LGX818 is a potent and selective RAF kinase inhibitor with unique biochemical properties that contribute to an excellent pharmacological profile. A Phase I clinical trial in patients with BRAF mutant tumors is ongoing. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3790. doi:1538-7445.AM2012-3790
- Published
- 2012
- Full Text
- View/download PDF
21. Abstract 20: Raf kinase inhibitors can induce Raf dimerization, downstream signaling, and cell growth
- Author
-
John Chan, Hanne Merritt, Matthew Holderfield, Susan Kaufman, Darrin Stuart, Kevin Shoemaker, Daniel Poon, John Tellew, Brent A. Appleton, Tobi Nagel, Nanxin Li, Marco Wallroth, and Yongjin Xu
- Subjects
MAPK/ERK pathway ,Cancer Research ,Gene knockdown ,Oncology ,Downregulation and upregulation ,Kinase ,Cell growth ,MEK inhibitor ,Phosphorylation ,Biology ,Small molecule ,Cell biology - Abstract
Genetic alterations in the Ras/Raf/MEK/ERK pathway are among the most common in human cancers. Up to 70% of melanomas harbor B-Raf mutations, and roughly 90% of pancreatic tumors have K-Ras mutations. To address these Raf pathway-driven cancers, small molecule Raf kinase inhibitors have been developed and are currently under clinical investigation. In B-RafV600E cells, Raf compounds inhibit signaling through MEK and ERK, resulting in the expected anti-proliferative effects. Paradoxically, in wild-type Raf cells and in mutant Ras cells, these compounds induce downstream signaling and can induce cell growth in some settings in vitro. While the induction of downstream signaling has previously been attributed to published Raf pathway feedback loops, this has not been proven directly. In fact, we show here that induction of pMEK and pERK can occur within minutes of Raf compound treatment, even before reported feedback phosphorylation events are seen on B-Raf and C-Raf. Interestingly, the induction of signaling and cell growth both occur in a biphasic pattern, with low compound concentrations (0.01-0.1 uM) causing maximal induction, and higher compound concentrations (1-10 uM) causing less profound induction. Such a biphasic pattern is also observed in biochemical assays with purified wild-type B-Raf or C-Raf. The biphasic pattern is suggestive of a mechanism involving the interaction of two signaling subunits. In addition, recent literature data (Rajakulendran, Nature, 461:542-6) has demonstrated that Raf dimerization can upregulate pMEK, not through trans-phosphorylation of Raf molecules but presumably by conformational activation of the kinase. Consistent with that model, we show that Raf compound treatment induces B/C-Raf dimer formation in cells. In addition, knockdown of A-, B- or C-Raf with siRNA does not abrogate the Raf compound induction of pMEK and pERK, suggesting that induction might be mediated by Raf homo- as well as hetero-dimerization. Notably, knockdown of K-Ras in K-RasMUT cells also does not abolish the induction, implying that this effect is not mediated by Ras. Taken together, these data suggest a model in which compound binding to one Raf molecule induces dimerization and conformational activation of a partner Raf molecule in the dimer. These observations can explain why wild-type Raf and mutant Ras tumors are insensitive to selective Raf kinase inhibitors and might also have important implications for toxicity, since induction of strong mitogenic signaling could lead to hyperproliferation of normal tissues. Understanding the Raf compound induction mechanism may lead not only to the design of improved inhibitors, but also to methods for overcoming the induction seen with current development compounds. Toward that end, we show that combining a MEK inhibitor with a Raf compound causes inhibition of both pERK and cell growth and may therefore have significant advantages in the clinic. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 20.
- Published
- 2010
- Full Text
- View/download PDF
22. RAF Inhibitors Activate the MAPK Pathway by Relieving Inhibitory Autophosphorylation
- Author
-
Tobi Nagel, Frank McCormick, John Tellew, John Chan, Huili Zhai, Darrin Stuart, Marco Wallroth, Stephen F. Hardy, Matthew Holderfield, Laura Tandeske, Mohammad Hekmat-Nejad, and Hanne Merritt
- Subjects
MAPK/ERK pathway ,Proto-Oncogene Proteins B-raf ,Cancer Research ,endocrine system diseases ,MAP Kinase Signaling System ,medicine.disease_cause ,Adenosine Triphosphate ,Cell Line, Tumor ,medicine ,Humans ,Phosphorylation ,Protein kinase A ,neoplasms ,chemistry.chemical_classification ,Oncogene ,Chemistry ,Autophosphorylation ,Cell Biology ,digestive system diseases ,Proto-Oncogene Proteins c-raf ,Enzyme ,Oncology ,Cell culture ,Cancer research ,raf Kinases ,Carcinogenesis ,V600E - Abstract
ATP competitive inhibitors of the BRAF(V600E) oncogene paradoxically activate downstream signaling in cells bearing wild-type BRAF (BRAF(WT)). In this study, we investigate the biochemical mechanism of wild-type RAF (RAF(WT)) activation by multiple catalytic inhibitors using kinetic analysis of purified BRAF(V600E) and RAF(WT) enzymes. We show that activation of RAF(WT) is ATP dependent and directly linked to RAF kinase activity. These data support a mechanism involving inhibitory autophosphorylation of RAF's phosphate-binding loop that, when disrupted either through pharmacologic or genetic alterations, results in activation of RAF and the mitogen-activated protein kinase (MAPK) pathway. This mechanism accounts not only for compound-mediated activation of the MAPK pathway in BRAF(WT) cells but also offers a biochemical mechanism for BRAF oncogenesis.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.