Network


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

Hotspot


Dive into the research topics where Amanda L. Christie is active.

Publication


Featured researches published by Amanda L. Christie.


Nature | 2010

Selective inhibition of BET bromodomains

Panagis Filippakopoulos; Jun Qi; Sarah Picaud; Yao Shen; William B. Smith; Oleg Fedorov; Elizabeth Morse; Tracey Keates; Tyler Hickman; I. Felletar; Martin Philpott; Shonagh Munro; Michael R. McKeown; Yuchuan Wang; Amanda L. Christie; Nathan West; Michael J. Cameron; Brian S. Schwartz; Tom D. Heightman; Nicholas B. La Thangue; Christopher A. French; Olaf Wiest; Andrew L. Kung; Stefan Knapp; James E. Bradner

Epigenetic proteins are intently pursued targets in ligand discovery. So far, successful efforts have been limited to chromatin modifying enzymes, or so-called epigenetic ‘writers’ and ‘erasers’. Potent inhibitors of histone binding modules have not yet been described. Here we report a cell-permeable small molecule (JQ1) that binds competitively to acetyl-lysine recognition motifs, or bromodomains. High potency and specificity towards a subset of human bromodomains is explained by co-crystal structures with bromodomain and extra-terminal (BET) family member BRD4, revealing excellent shape complementarity with the acetyl-lysine binding cavity. Recurrent translocation of BRD4 is observed in a genetically-defined, incurable subtype of human squamous carcinoma. Competitive binding by JQ1 displaces the BRD4 fusion oncoprotein from chromatin, prompting squamous differentiation and specific antiproliferative effects in BRD4-dependent cell lines and patient-derived xenograft models. These data establish proof-of-concept for targeting protein–protein interactions of epigenetic ‘readers’, and provide a versatile chemical scaffold for the development of chemical probes more broadly throughout the bromodomain family.


Nature | 2011

SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction.

Hiroyuki Inuzuka; Shavali Shaik; Ichiro Onoyama; Darning Gao; Alan Tseng; Richard S. Maser; Bo Zhai; Lixin Wan; Alejandro Gutierrez; Alan W. Lau; Yonghong Xiao; Amanda L. Christie; Jeffrey Settleman; Steven P. Gygi; Andrew L. Kung; Thomas Look; Keiichi I. Nakayama; Ronald A. DePinho; Wenyi Wei

The effective use of targeted therapy is highly dependent on the identification of responder patient populations. Loss of FBW7, which encodes a tumour-suppressor protein, is frequently found in various types of human cancer, including breast cancer, colon cancer and T-cell acute lymphoblastic leukaemia (T-ALL). In line with these genomic data, engineered deletion of Fbw7 in mouse T cells results in T-ALL, validating FBW7 as a T-ALL tumour suppressor. Determining the precise molecular mechanisms by which FBW7 exerts antitumour activity is an area of intensive investigation. These mechanisms are thought to relate in part to FBW7-mediated destruction of key proteins relevant to cancer, including Jun, Myc, cyclin E and notch 1 (ref. 9), all of which have oncoprotein activity and are overexpressed in various human cancers, including leukaemia. In addition to accelerating cell growth, overexpression of Jun, Myc or notch 1 can also induce programmed cell death. Thus, considerable uncertainty surrounds how FBW7-deficient cells evade cell death in the setting of upregulated Jun, Myc and/or notch 1. Here we show that the E3 ubiquitin ligase SCFFBW7 (a SKP1–cullin-1–F-box complex that contains FBW7 as the F-box protein) governs cellular apoptosis by targeting MCL1, a pro-survival BCL2 family member, for ubiquitylation and destruction in a manner that depends on phosphorylation by glycogen synthase kinase 3. Human T-ALL cell lines showed a close relationship between FBW7 loss and MCL1 overexpression. Correspondingly, T-ALL cell lines with defective FBW7 are particularly sensitive to the multi-kinase inhibitor sorafenib but resistant to the BCL2 antagonist ABT-737. On the genetic level, FBW7 reconstitution or MCL1 depletion restores sensitivity to ABT-737, establishing MCL1 as a therapeutically relevant bypass survival mechanism that enables FBW7-deficient cells to evade apoptosis. Therefore, our work provides insight into the molecular mechanism of direct tumour suppression by FBW7 and has implications for the targeted treatment of patients with FBW7-deficient T-ALL.


Journal of Experimental Medicine | 2012

Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition

Oliver Weigert; Andrew A. Lane; Liat Bird; Nadja Kopp; Bjoern Chapuy; Diederik van Bodegom; Angela V. Toms; Sachie Marubayashi; Amanda L. Christie; Michael R. McKeown; Ronald M. Paranal; James E. Bradner; Akinori Yoda; Christoph Gaul; Eric Vangrevelinghe; Vincent Romanet; Masato Murakami; Ralph Tiedt; Nicolas Ebel; Emeline Evrot; Alain De Pover; Catherine H. Regnier; Dirk Erdmann; Francesco Hofmann; Michael J. Eck; Stephen E. Sallan; Ross L. Levine; Andrew L. Kung; Fabienne Baffert; Thomas Radimerski

Hsp90 inhibition in B cell acute lymphoblastic leukemia overcomes resistance to JAK2 inhibitors.


Nature Medicine | 2012

Autocrine activation of the MET receptor tyrosine kinase in acute myeloid leukemia

Alex Kentsis; Casie Reed; Kim L. Rice; Takaomi Sanda; Scott J. Rodig; Eleni Tholouli; Amanda L. Christie; Ruud Delwel; Vu N. Ngo; Jeffery L. Kutok; Suzanne E. Dahlberg; Lisa A. Moreau; Richard Byers; James G. Christensen; George F. Vande Woude; Jonathan D. Licht; Andrew L. Kung; Louis M. Staudt; A. Thomas Look

Although the treatment of acute myeloid leukemia (AML) has improved substantially in the past three decades, more than half of all patients develop disease that is refractory to intensive chemotherapy. Functional genomics approaches offer a means to discover specific molecules mediating the aberrant growth and survival of cancer cells. Thus, using a loss-of-function RNA interference genomic screen, we identified the aberrant expression of hepatocyte growth factor (HGF) as a crucial element in AML pathogenesis. We found HGF expression leading to autocrine activation of its receptor tyrosine kinase, MET, in nearly half of the AML cell lines and clinical samples we studied. Genetic depletion of HGF or MET potently inhibited the growth and survival of HGF-expressing AML cells. However, leukemic cells treated with the specific MET kinase inhibitor crizotinib developed resistance resulting from compensatory upregulation of HGF expression, leading to the restoration of MET signaling. In cases of AML where MET is coactivated with other tyrosine kinases, such as fibroblast growth factor receptor 1 (FGFR1), concomitant inhibition of FGFR1 and MET blocked this compensatory HGF upregulation, resulting in sustained logarithmic cell killing both in vitro and in xenograft models in vivo. Our results show a widespread dependence of AML cells on autocrine activation of MET, as well as the key role of compensatory upregulation of HGF expression in maintaining leukemogenic signaling by this receptor. We anticipate that these findings will lead to the design of additional strategies to block adaptive cellular responses that drive compensatory ligand expression as an essential component of the targeted inhibition of oncogenic receptors in human cancers.


Leukemia | 2013

Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells

Julia Etchin; Qi Sun; Alex Kentsis; Alicia Farmer; Zi Chao Zhang; Takaomi Sanda; Marc R. Mansour; C Barcelo; Dilara McCauley; Michael Kauffman; Sharon Shacham; Amanda L. Christie; Andrew L. Kung; Scott J. Rodig; Yuh Min Chook; A T Look

Drugs that target the chief mediator of nuclear export, chromosome region maintenance 1 protein (CRM1) have potential as therapeutics for leukemia, but existing CRM1 inhibitors show variable potencies and a broad range of cytotoxic effects. Here, we report the structural analysis and antileukemic activity of a new generation of small-molecule inhibitors of CRM1. Designated selective inhibitors of nuclear export (SINE), these compounds were developed using molecular modeling to screen a small virtual library of compounds against the nuclear export signal (NES) groove of CRM1. The 2.2-Å crystal structure of the CRM1-Ran-RanBP1 complex bound to KPT-251, a representative molecule of this class of inhibitors, shows that the drug occupies part of the groove in CRM1 that is usually occupied by the NES, but penetrates much deeper into the groove and blocks CRM1-directed protein export. SINE inhibitors exhibit potent antileukemic activity, inducing apoptosis at nanomolar concentrations in a panel of 14 human acute myeloid leukemia (AML) cell lines representing different molecular subtypes of the disease. When administered orally to immunodeficient mice engrafted with human AML cells, KPT-251 had potent antileukemic activity with negligible toxicity to normal hematopoietic cells. Thus, KPT-SINE CRM1 antagonists represent a novel class of drugs that warrant further testing in AML patients.


Cancer Research | 2011

Differentiation of NUT midline carcinoma by epigenomic reprogramming.

Brian E. Schwartz; Matthias D. Hofer; Madeleine E. Lemieux; Daniel E. Bauer; Michael J. Cameron; Nathan West; Elin S. Agoston; Nicolas Reynoird; Saadi Khochbin; Tan A. Ince; Amanda L. Christie; Katherine A. Janeway; Sara O. Vargas; Antonio R. Perez-Atayde; Stephen E. Sallan; Andrew L. Kung; James E. Bradner; Christopher A. French

NUT midline carcinoma (NMC) is a lethal pediatric tumor defined by the presence of BRD-NUT fusion proteins that arrest differentiation. Here we explore the mechanisms underlying the ability of BRD4-NUT to prevent squamous differentiation. In both gain-of and loss-of-expression assays, we find that expression of BRD4-NUT is associated with globally decreased histone acetylation and transcriptional repression. Bulk chromatin acetylation can be restored by treatment of NMC cells with histone deacetylase inhibitors (HDACi), engaging a program of squamous differentiation and arrested growth in vitro that closely mimics the effects of siRNA-mediated attenuation of BRD4-NUT expression. The potential therapeutic utility of HDACi differentiation therapy was established in three different NMC xenograft models, where it produced significant growth inhibition and a survival benefit. Based on these results and translational studies performed with patient-derived primary tumor cells, a child with NMC was treated with the FDA-approved HDAC inhibitor, vorinostat. An objective response was obtained after five weeks of therapy, as determined by positron emission tomography. These findings provide preclinical support for trials of HDACi in patients with NMC.


British Journal of Haematology | 2013

KPT-330 inhibitor of CRM1 (XPO1)-mediated nuclear export has selective anti-leukaemic activity in preclinical models of T-cell acute lymphoblastic leukaemia and acute myeloid leukaemia

Julia Etchin; Takaomi Sanda; Marc R. Mansour; Alex Kentsis; Joan Montero; Bonnie Thi Le; Amanda L. Christie; Dilara McCauley; Scott J. Rodig; Michael Kauffman; Sharon Shacham; Richard Stone; Anthony Letai; Andrew L. Kung; A. Thomas Look

This study explored the anti‐leukaemic efficacy of novel irreversible inhibitors of the major nuclear export receptor, chromosome region maintenance 1 (CRM1, also termed XPO1). We found that these novel CRM1 antagonists, termed SINE (Selective Inhibitors of Nuclear Export), induced rapid apoptosis at low nanomolar concentrations in a panel of 14 human T‐cell acute lymphoblastic leukaemia (T‐ALL) cell lines representing different molecular subtypes of the disease. To assess in vivo anti‐leukaemia cell activity, we engrafted immunodeficient mice intravenously with the human T‐ALL MOLT‐4 cells, which harbour activating mutations of NOTCH1 and NRAS as well as loss of function of the CDKN2A, PTEN and TP53 tumour suppressors and express a high level of oncogenic transcription factor TAL1. Importantly, we examined the in vivo anti‐leukaemic efficacy of the clinical SINE compound KPT‐330 against T‐ALL and acute myeloid leukaemia (AML) cells. These studies demonstrated striking in vivo activity of KPT‐330 against T‐ALL and AML cells, with little toxicity to normal murine haematopoietic cells. Taken together, our results show that SINE CRM1 antagonists represent promising ‘first‐in‐class’ drugs with a novel mechanism of action and wide therapeutic index, and imply that drugs of this class show promise for the targeted therapy of T‐ALL and AML.


Blood | 2014

Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma

Yu-Tzu Tai; Patrick Mayes; Chirag Acharya; Mike Y. Zhong; Michele Cea; Antonia Cagnetta; Jenny L. Craigen; John Yates; Louise Gliddon; William Fieles; Bao Hoang; James Tunstead; Amanda L. Christie; Andrew L. Kung; Paul G. Richardson; Nikhil C. Munshi; Kenneth C. Anderson

B-cell maturation antigen (BCMA), highly expressed on malignant plasma cells in human multiple myeloma (MM), has not been effectively targeted with therapeutic monoclonal antibodies. We here show that BCMA is universally expressed on the MM cell surface and determine specific anti-MM activity of J6M0-mcMMAF (GSK2857916), a novel humanized and afucosylated antagonistic anti-BCMA antibody-drug conjugate via a noncleavable linker. J6M0-mcMMAF specifically blocks cell growth via G2/M arrest and induces caspase 3-dependent apoptosis in MM cells, alone and in coculture with bone marrow stromal cells or various effector cells. It strongly inhibits colony formation by MM cells while sparing surrounding BCMA-negative normal cells. J6M0-mcMMAF significantly induces effector cell-mediated lysis against allogeneic or autologous patient MM cells, with increased potency and efficacy compared with the wild-type J6M0 without Fc enhancement. The antibody-dependent cell-mediated cytotoxicity and apoptotic activity of J6M0-mcMMAF is further enhanced by lenalidomide. Importantly, J6M0-mcMMAF rapidly eliminates myeloma cells in subcutaneous and disseminated mouse models, and mice remain tumor-free up to 3.5 months. Furthermore, J6M0-mcMMAF recruits macrophages and mediates antibody-dependent cellular phagocytosis of MM cells. Together, these results demonstrate that GSK2857916 has potent and selective anti-MM activities via multiple cytotoxic mechanisms, providing a promising next-generation immunotherapeutic in this cancer.


Nature | 2015

Mediator kinase inhibition further activates super-enhancer-associated genes in AML.

Henry E. Pelish; Brian B. Liau; Ioana I. Nitulescu; Anupong Tangpeerachaikul; Zachary C. Poss; Diogo H. Da Silva; Brittany T. Caruso; Alexander Arefolov; Olugbeminiyi Fadeyi; Amanda L. Christie; Karrie Du; Deepti Banka; Elisabeth V. Schneider; Anja Jestel; Ge Zou; Chong Si; Christopher C. Ebmeier; Roderick T. Bronson; Andrei V. Krivtsov; Andrew G. Myers; Nancy E. Kohl; Andrew L. Kung; Scott A. Armstrong; Madeleine E. Lemieux; Dylan J. Taatjes; Matthew D. Shair

Super-enhancers (SEs), which are composed of large clusters of enhancers densely loaded with the Mediator complex, transcription factors and chromatin regulators, drive high expression of genes implicated in cell identity and disease, such as lineage-controlling transcription factors and oncogenes. BRD4 and CDK7 are positive regulators of SE-mediated transcription. By contrast, negative regulators of SE-associated genes have not been well described. Here we show that the Mediator-associated kinases cyclin-dependent kinase 8 (CDK8) and CDK19 restrain increased activation of key SE-associated genes in acute myeloid leukaemia (AML) cells. We report that the natural product cortistatin A (CA) selectively inhibits Mediator kinases, has anti-leukaemic activity in vitro and in vivo, and disproportionately induces upregulation of SE-associated genes in CA-sensitive AML cell lines but not in CA-insensitive cell lines. In AML cells, CA upregulated SE-associated genes with tumour suppressor and lineage-controlling functions, including the transcription factors CEBPA, IRF8, IRF1 and ETV6 (refs 6, 7, 8). The BRD4 inhibitor I-BET151 downregulated these SE-associated genes, yet also has anti-leukaemic activity. Individually increasing or decreasing the expression of these transcription factors suppressed AML cell growth, providing evidence that leukaemia cells are sensitive to the dosage of SE-associated genes. Our results demonstrate that Mediator kinases can negatively regulate SE-associated gene expression in specific cell types, and can be pharmacologically targeted as a therapeutic approach to AML.


Leukemia | 2012

Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib

Ellen Weisberg; Abdel Kareem Azab; Paul W. Manley; Andrew L. Kung; Amanda L. Christie; Rod Bronson; Irene M. Ghobrial; James D. Griffin

Drug resistance is a growing area of concern. It has been shown that a small, residual pool of leukemic CD34+ progenitor cells can survive in the marrow microenvironment of chronic myeloid leukemia (CML) patients after years of kinase inhibitor treatment. Bone marrow (BM) stroma has been implicated in the long-term survival of leukemic cells, and contributes to the expansion and proliferation of both transformed and normal hematopoietic cells. Mechanistically, we found that CML cells expressed CXCR4, and that plerixafor diminished BCR–ABL-positive cell migration and reduced adhesion of these cells to extra cellular-matrix components and to BM stromal cells in vitro. Moreover, plerixafor decreased the drug resistance of CML cells induced by co-culture with BM stromal cells in vitro. Using a functional mouse model of progressive and residual disease, we demonstrated the ability of the CXCR4 inhibitor, plerixafor, to mobilize leukemic cells in vivo, such that a plerixafor–nilotinib combination reduced the leukemia burden in mice significantly below the baseline level suppression exhibited by a moderate-to-high dose of nilotinib as single agent. These results support the idea of using CXCR4 inhibition in conjunction with targeted tyrosine kinase inhibition to override drug resistance in CML and suppress or eradicate residual disease.

Collaboration


Dive into the Amanda L. Christie's collaboration.

Top Co-Authors

Avatar

Andrew L. Kung

Memorial Sloan Kettering Cancer Center

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

Scott J. Rodig

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge