Network


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

Hotspot


Dive into the research topics where Daniel J. Anderson is active.

Publication


Featured researches published by Daniel J. Anderson.


Nature | 2010

RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth

Georgia Hatzivassiliou; Kyung Song; Ivana Yen; Barbara J. Brandhuber; Daniel J. Anderson; Ryan Alvarado; Mary J. C. Ludlam; David Stokoe; Susan L. Gloor; Guy Vigers; Tony Morales; Ignacio Aliagas; Bonnie Liu; Steve Sideris; Klaus P. Hoeflich; Bijay S. Jaiswal; Somasekar Seshagiri; Hartmut Koeppen; Marcia Belvin; Lori S. Friedman; Shiva Malek

Activating mutations in KRAS and BRAF are found in more than 30% of all human tumours and 40% of melanoma, respectively, thus targeting this pathway could have broad therapeutic effects. Small molecule ATP-competitive RAF kinase inhibitors have potent antitumour effects on mutant BRAF(V600E) tumours but, in contrast to mitogen-activated protein kinase kinase (MEK) inhibitors, are not potent against RAS mutant tumour models, despite RAF functioning as a key effector downstream of RAS and upstream of MEK. Here we show that ATP-competitive RAF inhibitors have two opposing mechanisms of action depending on the cellular context. In BRAF(V600E) tumours, RAF inhibitors effectively block the mitogen-activated protein kinase (MAPK) signalling pathway and decrease tumour growth. Notably, in KRAS mutant and RAS/RAF wild-type tumours, RAF inhibitors activate the RAF–MEK–ERK pathway in a RAS-dependent manner, thus enhancing tumour growth in some xenograft models. Inhibitor binding activates wild-type RAF isoforms by inducing dimerization, membrane localization and interaction with RAS–GTP. These events occur independently of kinase inhibition and are, instead, linked to direct conformational effects of inhibitors on the RAF kinase domain. On the basis of these findings, we demonstrate that ATP-competitive kinase inhibitors can have opposing functions as inhibitors or activators of signalling pathways, depending on the cellular context. Furthermore, this work provides new insights into the therapeutic use of ATP-competitive RAF inhibitors.


Nature | 2011

Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7

Ingrid E. Wertz; Saritha Kusam; Cynthia Lam; Toru Okamoto; Wendy Sandoval; Daniel J. Anderson; Elizabeth Helgason; James A. Ernst; Mike Eby; Jinfeng Liu; Lisa D. Belmont; Joshua S. Kaminker; Karen O’Rourke; Kanan Pujara; Pawan Bir Kohli; Adam R. Johnson; Mark L. Chiu; Jennie R. Lill; Peter K. Jackson; Wayne J. Fairbrother; Somasekar Seshagiri; Mary J. C. Ludlam; Kevin G. Leong; Erin C. Dueber; Heather Maecker; David C. S. Huang; Vishva M. Dixit

Microtubules have pivotal roles in fundamental cellular processes and are targets of antitubulin chemotherapeutics. Microtubule-targeted agents such as Taxol and vincristine are prescribed widely for various malignancies, including ovarian and breast adenocarcinomas, non-small-cell lung cancer, leukaemias and lymphomas. These agents arrest cells in mitosis and subsequently induce cell death through poorly defined mechanisms. The strategies that resistant tumour cells use to evade death induced by antitubulin agents are also unclear. Here we show that the pro-survival protein MCL1 (ref. 3) is a crucial regulator of apoptosis triggered by antitubulin chemotherapeutics. During mitotic arrest, MCL1 protein levels decline markedly, through a post-translational mechanism, potentiating cell death. Phosphorylation of MCL1 directs its interaction with the tumour-suppressor protein FBW7, which is the substrate-binding component of a ubiquitin ligase complex. The polyubiquitylation of MCL1 then targets it for proteasomal degradation. The degradation of MCL1 was blocked in patient-derived tumour cells that lacked FBW7 or had loss-of-function mutations in FBW7, conferring resistance to antitubulin agents and promoting chemotherapeutic-induced polyploidy. Additionally, primary tumour samples were enriched for FBW7 inactivation and elevated MCL1 levels, underscoring the prominent roles of these proteins in oncogenesis. Our findings suggest that profiling the FBW7 and MCL1 status of tumours, in terms of protein levels, messenger RNA levels and genetic status, could be useful to predict the response of patients to antitubulin chemotherapeutics.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity

Till Maurer; Lindsay S. Garrenton; Angela Oh; Keith Pitts; Daniel J. Anderson; Nicholas J. Skelton; Benjamin P. Fauber; Borlan Pan; Shiva Malek; David Stokoe; Mary J. C. Ludlam; Krista K. Bowman; Jiansheng Wu; Anthony M. Giannetti; Melissa A. Starovasnik; Ira Mellman; Peter K. Jackson; Joachim Rudolph; Weiru Wang; Guowei Fang

The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.


Nature | 2013

Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers

Georgia Hatzivassiliou; Jacob R. Haling; Huifen Chen; Kyung Song; Steve Price; Robert Heald; Joanne Frances Mary Hewitt; Mark Zak; Ariana Peck; Christine Orr; Mark Merchant; Klaus P. Hoeflich; Jocelyn Chan; Shiuh-Ming Luoh; Daniel J. Anderson; Mary J. C. Ludlam; Christian Wiesmann; Mark Ultsch; Lori Friedman; Shiva Malek; Marcia Belvin

KRAS and BRAF activating mutations drive tumorigenesis through constitutive activation of the MAPK pathway. As these tumours represent an area of high unmet medical need, multiple allosteric MEK inhibitors, which inhibit MAPK signalling in both genotypes, are being tested in clinical trials. Impressive single-agent activity in BRAF-mutant melanoma has been observed; however, efficacy has been far less robust in KRAS-mutant disease. Here we show that, owing to distinct mechanisms regulating MEK activation in KRAS- versus BRAF-driven tumours, different mechanisms of inhibition are required for optimal antitumour activity in each genotype. Structural and functional analysis illustrates that MEK inhibitors with superior efficacy in KRAS-driven tumours (GDC-0623 and G-573, the former currently in phase I clinical trials) form a strong hydrogen-bond interaction with S212 in MEK that is critical for blocking MEK feedback phosphorylation by wild-type RAF. Conversely, potent inhibition of active, phosphorylated MEK is required for strong inhibition of the MAPK pathway in BRAF-mutant tumours, resulting in superior efficacy in this genotype with GDC-0973 (also known as cobimetinib), a MEK inhibitor currently in phase III clinical trials. Our study highlights that differences in the activation state of MEK in KRAS-mutant tumours versus BRAF-mutant tumours can be exploited through the design of inhibitors that uniquely target these distinct activation states of MEK. These inhibitors are currently being evaluated in clinical trials to determine whether improvements in therapeutic index within KRAS versus BRAF preclinical models translate to improved clinical responses in patients.


Nature Chemical Biology | 2013

Covalent and allosteric inhibitors of the ATPase VCP/p97 induce cancer cell death

Paola Magnaghi; Roberto D'alessio; Barbara Valsasina; Nilla Avanzi; Simona Rizzi; Daniela Asa; Fabio Gasparri; Ulisse Cucchi; Christian Orrenius; Paolo Polucci; Dario Ballinari; Claudia Perrera; Antonella Leone; Giovanni Cervi; Elena Casale; Yang Xiao; Chihunt Wong; Daniel J. Anderson; Arturo Galvani; Daniele Donati; Thomas O'Brien; Peter K. Jackson; Antonella Isacchi

VCP (also known as p97 or Cdc48p in yeast) is an AAA(+) ATPase regulating endoplasmic reticulum-associated degradation. After high-throughput screening, we developed compounds that inhibit VCP via different mechanisms, including covalent modification of an active site cysteine and a new allosteric mechanism. Using photoaffinity labeling, structural analysis and mutagenesis, we mapped the binding site of allosteric inhibitors to a region spanning the D1 and D2 domains of adjacent protomers encompassing elements important for nucleotide-state sensing and ATP hydrolysis. These compounds induced an increased affinity for nucleotides. Interference with nucleotide turnover in individual subunits and distortion of interprotomer communication cooperated to impair VCP enzymatic activity. Chemical expansion of this allosteric class identified NMS-873, the most potent and specific VCP inhibitor described to date, which activated the unfolded protein response, interfered with autophagy and induced cancer cell death. The consistent pattern of cancer cell killing by covalent and allosteric inhibitors provided critical validation of VCP as a cancer target.


Blood | 2012

Bcl-2, Bcl-x(L), and Bcl-w are not equivalent targets of ABT-737 and navitoclax (ABT-263) in lymphoid and leukemic cells.

Delphine Mérino; Seong Lin Khaw; Stephan P. Glaser; Daniel J. Anderson; Lisa D. Belmont; Chihunt Wong; Peng Yue; Mikara Robati; Belinda Phipson; W D Fairlie; Erinna F. Lee; Kirsteen J. Campbell; Cassandra J. Vandenberg; Suzanne Cory; Andrew W. Roberts; Mary J. C. Ludlam; David C. S. Huang

The BH3-mimetic ABT-737 and an orally bioavailable compound of the same class, navitoclax (ABT-263), have shown promising antitumor efficacy in preclinical and early clinical studies. Although both drugs avidly bind Bcl-2, Bcl-x(L), and Bcl-w in vitro, we find that Bcl-2 is the critical target in vivo, suggesting that patients with tumors overexpressing Bcl-2 will probably benefit. In human non-Hodgkin lymphomas, high expression of Bcl-2 but not Bcl-x(L) predicted sensitivity to ABT-263. Moreover, we show that increasing Bcl-2 sensitized normal and transformed lymphoid cells to ABT-737 by elevating proapoptotic Bim. In striking contrast, increasing Bcl-x(L) or Bcl-w conferred robust resistance to ABT-737, despite also increasing Bim. Cell-based protein redistribution assays unexpectedly revealed that ABT-737 disrupts Bcl-2/Bim complexes more readily than Bcl-x(L)/Bim or Bcl-w/Bim complexes. These results have profound implications for how BH3-mimetics induce apoptosis and how the use of these compounds can be optimized for treating lymphoid malignancies.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Crystal structure of the proteasomal deubiquitylation module Rpn8-Rpn11

Ganesh Ramnath Pathare; Pawel Sledz; Daniel J. Anderson; Han-Jie Zhou; Els Pardon; Jan Steyaert; Friedrich Förster; Andreas Bracher; Wolfgang Baumeister

Significance The 26S proteasome is a multiprotein complex that degrades proteins marked for destruction by the covalent attachment of polyubiquitin chains. Proteasome activity is essential for the removal of damaged, potentially toxic proteins and for the regulation of numerous cellular processes. Multiple crystal structures of the Rpn8-Rpn11 heterodimer, which is responsible for the removal of polyubiquitin tags before substrate degradation in the proteasome, provide insight into how substrate unfolding and isopeptide bond cleavage might be coupled, and how premature activation of this module is prevented. Its accurate function ensures timely degradation of substrates and, ultimately, the replenishment of the limited cellular pool of free ubiquitin. The ATP-dependent degradation of polyubiquitylated proteins by the 26S proteasome is essential for the maintenance of proteome stability and the regulation of a plethora of cellular processes. Degradation of substrates is preceded by the removal of polyubiquitin moieties through the isopeptidase activity of the subunit Rpn11. Here we describe three crystal structures of the heterodimer of the Mpr1–Pad1–N-terminal domains of Rpn8 and Rpn11, crystallized as a fusion protein in complex with a nanobody. This fusion protein exhibits modest deubiquitylation activity toward a model substrate. Full activation requires incorporation of Rpn11 into the 26S proteasome and is dependent on ATP hydrolysis, suggesting that substrate processing and polyubiquitin removal are coupled. Based on our structures, we propose that premature activation is prevented by the combined effects of low intrinsic ubiquitin affinity, an insertion segment acting as a physical barrier across the substrate access channel, and a conformationally unstable catalytic loop in Rpn11. The docking of the structure into the proteasome EM density revealed contacts of Rpn11 with ATPase subunits, which likely stabilize the active conformation and boost the affinity for the proximal ubiquitin moiety. The narrow space around the Rpn11 active site at the entrance to the ATPase ring pore is likely to prevent erroneous deubiquitylation of folded proteins.


Journal of Medicinal Chemistry | 2015

Discovery of a First-in-Class, Potent, Selective, and Orally Bioavailable Inhibitor of the p97 AAA ATPase (CB-5083)

Han-Jie Zhou; Jinhai Wang; Bing Yao; Steve Wong; Stevan Djakovic; Brajesh Kumar; Julie Rice; Eduardo Valle; Ferdie Soriano; Mary-Kamala Menon; Antonett Madriaga; Szerenke Kiss von Soly; Abhinav Kumar; Francesco Parlati; F. Michael Yakes; Laura Kay Shawver; Ronan Le Moigne; Daniel J. Anderson; Mark Rolfe; David Juergen Wustrow

The AAA-ATPase p97 plays vital roles in mechanisms of protein homeostasis, including ubiquitin-proteasome system (UPS) mediated protein degradation, endoplasmic reticulum-associated degradation (ERAD), and autophagy. Herein we describe our lead optimization efforts focused on in vitro potency, ADME, and pharmaceutical properties that led to the discovery of a potent, ATP-competitive, D2-selective, and orally bioavailable p97 inhibitor 71, CB-5083. Treatment of tumor cells with 71 leads to significant accumulation of markers associated with inhibition of UPS and ERAD functions, which induces irresolvable proteotoxic stress and cell death. In tumor bearing mice, oral administration of 71 causes rapid accumulation of markers of the unfolded protein response (UPR) and subsequently induces apoptosis leading to sustained antitumor activity in in vivo xenograft models of both solid and hematological tumors. 71 has been taken into phase 1 clinical trials in patients with multiple myeloma and solid tumors.


PLOS ONE | 2011

Live-Cell Microscopy Reveals Small Molecule Inhibitor Effects on MAPK Pathway Dynamics

Daniel J. Anderson; Jenni K. Durieux; Kyung Song; Ryan Alvarado; Peter K. Jackson; Georgia Hatzivassiliou; Mary J. C. Ludlam

Oncogenic mutations in the mitogen activated protein kinase (MAPK) pathway are prevalent in human tumors, making this pathway a target of drug development efforts. Recently, ATP-competitive Raf inhibitors were shown to cause MAPK pathway activation via Raf kinase priming in wild-type BRaf cells and tumors, highlighting the need for a thorough understanding of signaling in the context of small molecule kinase inhibitors. Here, we present critical improvements in cell-line engineering and image analysis coupled with automated image acquisition that allow for the simultaneous identification of cellular localization of multiple MAPK pathway components (KRas, CRaf, Mek1 and Erk2). We use these assays in a systematic study of the effect of small molecule inhibitors across the MAPK cascade either as single agents or in combination. Both Raf inhibitor priming as well as the release from negative feedback induced by Mek and Erk inhibitors cause translocation of CRaf to the plasma membrane via mechanisms that are additive in pathway activation. Analysis of Erk activation and sub-cellular localization upon inhibitor treatments reveals differential inhibition and activation with the Raf inhibitors AZD628 and GDC0879 respectively. Since both single agent and combination studies of Raf and Mek inhibitors are currently in the clinic, our assays provide valuable insight into their effects on MAPK signaling in live cells.


Molecular Cancer Therapeutics | 2017

The p97 inhibitor CB-5083 is a unique disrupter of protein homeostasis in models of Multiple Myeloma.

Ronan Le Moigne; Blake T. Aftab; Stevan Djakovic; Eugen Dhimolea; Eduardo Valle; Megan Murnane; Emily M. King; Ferdie Soriano; Mary Kamala Menon; Zhi Yong Wu; Stephen T. C. Wong; Grace J. Lee; Bing Yao; Arun P. Wiita; Christine Lam; Julie Rice; Jinhai Wang; Marta Chesi; P. Leif Bergsagel; Marianne Kraus; Christoph Driessen; Szerenke Kiss von Soly; F. Michael Yakes; David Wustrow; Laura Kay Shawver; Han Jie Zhou; Thomas G. Martin; Jeffrey L. Wolf; Constantine S. Mitsiades; Daniel J. Anderson

Inhibition of the AAA ATPase, p97, was recently shown to be a novel method for targeting the ubiquitin proteasome system, and CB-5083, a first-in-class inhibitor of p97, has demonstrated broad antitumor activity in a range of both hematologic and solid tumor models. Here, we show that CB-5083 has robust activity against multiple myeloma cell lines and a number of in vivo multiple myeloma models. Treatment with CB-5083 is associated with accumulation of ubiquitinated proteins, induction of the unfolded protein response, and apoptosis. CB-5083 decreases viability in multiple myeloma cell lines and patient-derived multiple myeloma cells, including those with background proteasome inhibitor (PI) resistance. CB-5083 has a unique mechanism of action that combines well with PIs, which is likely owing to the p97-dependent retro-translocation of the transcription factor, Nrf1, which transcribes proteasome subunit genes following exposure to a PI. In vivo studies using clinically relevant multiple myeloma models demonstrate that single-agent CB-5083 inhibits tumor growth and combines well with multiple myeloma standard-of-care agents. Our preclinical data demonstrate the efficacy of CB-5083 in several multiple myeloma disease models and provide the rationale for clinical evaluation as monotherapy and in combination in multiple myeloma. Mol Cancer Ther; 16(11); 2375–86. ©2017 AACR.

Collaboration


Dive into the Daniel J. Anderson's collaboration.

Top Co-Authors

Avatar

Mark Rolfe

Millennium Pharmaceuticals

View shared research outputs
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