Network


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

Hotspot


Dive into the research topics where Casie Reed is active.

Publication


Featured researches published by Casie Reed.


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.


Journal of Clinical Investigation | 2014

Phenothiazines induce PP2A-mediated apoptosis in T cell acute lymphoblastic leukemia

Alejandro Gutierrez; Li Pan; Richard W.J. Groen; Frederic Baleydier; Alex Kentsis; Jason J. Marineau; Ruta Grebliunaite; Elena Kozakewich; Casie Reed; Françoise Pflumio; Sandrine Poglio; Benjamin Uzan; Paul A. Clemons; Lynn VerPlank; Frank An; Jason Burbank; Stephanie Norton; Nicola Tolliday; Hanno Steen; Andrew P. Weng; H. Yuan; James E. Bradner; Constantine S. Mitsiades; A. Thomas Look

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer that is frequently associated with activating mutations in NOTCH1 and dysregulation of MYC. Here, we performed 2 complementary screens to identify FDA-approved drugs and drug-like small molecules with activity against T-ALL. We developed a zebrafish system to screen small molecules for toxic activity toward MYC-overexpressing thymocytes and used a human T-ALL cell line to screen for small molecules that synergize with Notch inhibitors. We identified the antipsychotic drug perphenazine in both screens due to its ability to induce apoptosis in fish, mouse, and human T-ALL cells. Using ligand-affinity chromatography coupled with mass spectrometry, we identified protein phosphatase 2A (PP2A) as a perphenazine target. T-ALL cell lines treated with perphenazine exhibited rapid dephosphorylation of multiple PP2A substrates and subsequent apoptosis. Moreover, shRNA knockdown of specific PP2A subunits attenuated perphenazine activity, indicating that PP2A mediates the drugs antileukemic activity. Finally, human T-ALLs treated with perphenazine exhibited suppressed cell growth and dephosphorylation of PP2A targets in vitro and in vivo. Our findings provide a mechanistic explanation for the recurring identification of phenothiazines as a class of drugs with anticancer effects. Furthermore, these data suggest that pharmacologic PP2A activation in T-ALL and other cancers driven by hyperphosphorylated PP2A substrates has therapeutic potential.


Leukemia | 2016

Activity of a selective inhibitor of nuclear export, selinexor (KPT-330), against AML-initiating cells engrafted into immunosuppressed NSG mice.

Julia Etchin; Joan Montero; Alla Berezovskaya; Bonnie Thi Le; Alex Kentsis; Amanda L. Christie; Amy Saur Conway; Wen Chen; Casie Reed; Marc R. Mansour; Christopher Ng; Sophia Adamia; Scott J. Rodig; Ilene Galinsky; Richard Stone; B Klebanov; Yosef Landesman; Michael Kauffman; Sharon Shacham; Andrew L. Kung; Jean C.Y. Wang; Anthony Letai; A T Look

Currently available combination chemotherapy for acute myeloid leukemia (AML) often fails to result in long-term remissions, emphasizing the need for novel therapeutic strategies. We reasoned that targeted inhibition of a prominent nuclear exporter, XPO1/CRM1, could eradicate self-renewing leukemia-initiating cells (LICs) whose survival depends on timely XPO1-mediated transport of specific protein and RNA cargoes. Using an immunosuppressed mouse model bearing primary patient-derived AML cells, we demonstrate that selinexor (KPT-330), an oral antagonist of XPO1 that is currently in clinical trials, has strong activity against primary AML cells while sparing normal stem and progenitor cells. Importantly, limiting dilution transplantation assays showed that this cytotoxic activity is not limited to the rapidly proliferating bulk population of leukemic cells but extends to the LICs, whose inherent drug resistance and unrestricted self-renewal capacity has been implicated in the difficulty of curing AML patients with conventional chemotherapy alone.


Nature Genetics | 2017

PGBD5 promotes site-specific oncogenic mutations in human tumors

Anton Henssen; Richard Koche; Jiali Zhuang; Eileen Jiang; Casie Reed; Amy Eisenberg; Eric Still; Ian Macarthur; Elias Rodríguez-Fos; Santiago Gonzalez; Montserrat Puiggròs; Andrew N. Blackford; Christopher E. Mason; Elisa de Stanchina; Mithat Gonen; Anne Katrin Emde; Minita Shah; Kanika Arora; Catherine Reeves; Nicholas D. Socci; Elizabeth J. Perlman; Cristina R. Antonescu; Charles W. M. Roberts; Hanno Steen; Elizabeth Mullen; David Torrents; Zhiping Weng; Scott A. Armstrong; Alex Kentsis

Genomic rearrangements are a hallmark of human cancers. Here, we identify the piggyBac transposable element derived 5 (PGBD5) gene as encoding an active DNA transposase expressed in the majority of childhood solid tumors, including lethal rhabdoid tumors. Using assembly-based whole-genome DNA sequencing, we found previously undefined genomic rearrangements in human rhabdoid tumors. These rearrangements involved PGBD5-specific signal (PSS) sequences at their breakpoints and recurrently inactivated tumor-suppressor genes. PGBD5 was physically associated with genomic PSS sequences that were also sufficient to mediate PGBD5-induced DNA rearrangements in rhabdoid tumor cells. Ectopic expression of PGBD5 in primary immortalized human cells was sufficient to promote cell transformation in vivo. This activity required specific catalytic residues in the PGBD5 transposase domain as well as end-joining DNA repair and induced structural rearrangements with PSS breakpoints. These results define PGBD5 as an oncogenic mutator and provide a plausible mechanism for site-specific DNA rearrangements in childhood and adult solid tumors.


British Journal of Haematology | 2015

Targeting oncogenic interleukin-7 receptor signalling with N-acetylcysteine in T cell acute lymphoblastic leukaemia.

Marc R. Mansour; Casie Reed; Amy Eisenberg; Jen Chieh Tseng; Jean-Claude Twizere; Sarah Daakour; Akinori Yoda; Scott J. Rodig; Noa Tal; Chen Shochat; Alla Berezovskaya; Daniel J. DeAngelo; Stephen E. Sallan; David M. Weinstock; Shai Izraeli; Andrew L. Kung; Alex Kentsis; A. Thomas Look

Activating mutations of the interleukin‐7 receptor (IL7R) occur in approximately 10% of patients with T cell acute lymphoblastic leukaemia (T‐ALL). Most mutations generate a cysteine at the transmembrane domain leading to receptor homodimerization through disulfide bond formation and ligand‐independent activation of STAT5. We hypothesized that the reducing agent N‐acetylcysteine (NAC), a well‐tolerated drug used widely in clinical practice to treat acetaminophen overdose, would reduce disulfide bond formation, and inhibit mutant IL7R‐mediated oncogenic signalling. We found that treatment with NAC disrupted IL7R homodimerization in IL7R‐mutant DND‐41 cells as assessed by non‐reducing Western blot, as well as in a luciferase complementation assay. NAC led to STAT5 dephosphorylation and cell apoptosis at clinically achievable concentrations in DND‐41 cells, and Ba/F3 cells transformed by an IL7R‐mutant construct containing a cysteine insertion. The apoptotic effects of NAC could be rescued in part by a constitutively active allele of STAT5. Despite using doses lower than those tolerated in humans, NAC treatment significantly inhibited the progression of human DND‐41 cells engrafted in immunodeficient mice. Thus, targeting leukaemogenic IL7R homodimerization with NAC offers a potentially effective and feasible therapeutic strategy that warrants testing in patients with T‐ALL.


Journal of Proteome Research | 2015

A High-Efficiency Cellular Extraction System for Biological Proteomics.

Avantika Dhabaria; Paolo Cifani; Casie Reed; Hanno Steen; Alex Kentsis

Recent developments in quantitative high-resolution mass spectrometry have led to significant improvements in the sensitivity and specificity of the biochemical analyses of cellular reactions, protein-protein interactions, and small-molecule-drug discovery. These approaches depend on cellular proteome extraction that preserves native protein activities. Here, we systematically analyzed mechanical methods of cell lysis and physical protein extraction to identify those that maximize the extraction of cellular proteins while minimizing their denaturation. Cells were mechanically disrupted using Potter-Elvehjem homogenization, probe- or adaptive-focused acoustic sonication, and were in the presence of various detergents, including polyoxyethylene ethers and esters, glycosides, and zwitterions. Using fluorescence spectroscopy, biochemical assays, and mass spectrometry proteomics, we identified the combination of adaptive focused acoustic (AFA) sonication in the presence of a binary poloxamer-based mixture of octyl-β-glucoside and Pluronic F-127 to maximize the depth and yield of the proteome extraction while maintaining native protein activity. This binary poloxamer extraction system allowed for native proteome extraction comparable in coverage to the proteomes extracted using denaturing SDS or guanidine-containing buffers, including the efficient extraction of all major cellular organelles. This high-efficiency cellular extraction system should prove useful for a variety of cell biochemical studies, including structural and functional proteomics.


Cancer Discovery | 2018

MEF2C phosphorylation is required for chemotherapy resistance in acute myeloid leukemia

Fiona Brown; Eric Still; Richard Koche; Christina Y. Yim; Sumiko Takao; Paolo Cifani; Casie Reed; Shehana Gunasekera; Scott B. Ficarro; Peter Romanienko; Willie Mark; Craig R. McCarthy; Elisa de Stanchina; Mithat Gonen; Venkatraman E. Seshan; Patrick Bhola; Conor O'Donnell; Barbara Spitzer; Crystal Stutzke; Vincent-Philippe Lavallée; Josée Hébert; Andrei V. Krivstov; Ari Melnick; Elisabeth Paietta; Martin S. Tallman; Anthony Letai; Guy Sauvageau; Gayle Pouliot; Ross L. Levine; Jarrod A. Marto

In acute myeloid leukemia (AML), chemotherapy resistance remains prevalent and poorly understood. Using functional proteomics of patient AML specimens, we identified MEF2C S222 phosphorylation as a specific marker of primary chemoresistance. We found that Mef2cS222A/S222A knock-in mutant mice engineered to block MEF2C phosphorylation exhibited normal hematopoiesis, but were resistant to leukemogenesis induced by MLL-AF9 MEF2C phosphorylation was required for leukemia stem cell maintenance and induced by MARK kinases in cells. Treatment with the selective MARK/SIK inhibitor MRT199665 caused apoptosis and conferred chemosensitivity in MEF2C-activated human AML cell lines and primary patient specimens, but not those lacking MEF2C phosphorylation. These findings identify kinase-dependent dysregulation of transcription factor control as a determinant of therapy response in AML, with immediate potential for improved diagnosis and therapy for this disease.Significance: Functional proteomics identifies phosphorylation of MEF2C in the majority of primary chemotherapy-resistant AML. Kinase-dependent dysregulation of this transcription factor confers susceptibility to MARK/SIK kinase inhibition in preclinical models, substantiating its clinical investigation for improved diagnosis and therapy of AML. Cancer Discov; 8(4); 478-97. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 371.


Science Translational Medicine | 2017

Therapeutic targeting of PGBD5-induced DNA repair dependency in pediatric solid tumors

Anton Henssen; Casie Reed; Eileen Jiang; Heathcliff Dorado García; Jennifer von Stebut; Ian Macarthur; Patrick Hundsdoerfer; Jun Hyun Kim; Elisa de Stanchina; Yasumichi Kuwahara; Hajime Hosoi; Neil J. Ganem; Filemon Dela Cruz; Andrew L. Kung; Johannes H. Schulte; John H.J. Petrini; Alex Kentsis

PGBD5 DNA transposase confers therapeutically actionable dependency in solid tumors. Synthetic lethality, pediatric edition Although a variety of therapeutic regimens are available for pediatric solid tumors, they are often ineffective and typically nonspecific. Henssen et al. determined that expression of a DNA transposase called PGBD5 is common in these tumors and presents a therapeutic vulnerability. The authors demonstrated that cells expressing PGBD5 are dependent on DNA repair through nonhomologous end joining, then identified a drug that inhibits this DNA repair pathway and is therefore active against many pediatric tumor types, particularly when combined with chemotherapy, while sparing surrounding nontumor tissues. Despite intense efforts, the cure rates of childhood and adult solid tumors are not satisfactory. Resistance to intensive chemotherapy is common, and targets for molecular therapies are largely undefined. We have found that the majority of childhood solid tumors, including rhabdoid tumors, neuroblastoma, medulloblastoma, and Ewing sarcoma, express an active DNA transposase, PGBD5, that can promote site-specific genomic rearrangements in human cells. Using functional genetic approaches, we discovered that mouse and human cells deficient in nonhomologous end joining (NHEJ) DNA repair cannot tolerate the expression of PGBD5. In a chemical screen of DNA damage signaling inhibitors, we identified AZD6738 as a specific sensitizer of PGBD5-dependent DNA damage and apoptosis. We found that expression of PGBD5, but not its nuclease activity–deficient mutant, was sufficient to induce sensitivity to AZD6738. Depletion of endogenous PGBD5 conferred resistance to AZD6738 in human tumor cells. PGBD5-expressing tumor cells accumulated unrepaired DNA damage in response to AZD6738 treatment and underwent apoptosis in both dividing and G1-phase cells in the absence of immediate DNA replication stress. Accordingly, AZD6738 exhibited nanomolar potency against most neuroblastoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumor cells tested while sparing nontransformed human and mouse embryonic fibroblasts in vitro. Finally, treatment with AZD6738 induced apoptosis and regression of human neuroblastoma and medulloblastoma tumors engrafted in immunodeficient mice in vivo. This effect was potentiated by combined treatment with cisplatin, including substantial antitumor activity against patient-derived primary neuroblastoma xenografts. These findings delineate a therapeutically actionable synthetic dependency induced in PGBD5-expressing solid tumors.


Nature Genetics | 2017

Erratum: PGBD5 promotes site-specific oncogenic mutations in human tumors

Anton Henssen; Richard Koche; Jiali Zhuang; Eileen Jiang; Casie Reed; Amy Eisenberg; Eric Still; Ian Macarthur; Elias Rodríguez-Fos; Santiago Gonzalez; Montserrat Puiggròs; Andrew N. Blackford; Christopher E. Mason; Elisa de Stanchina; Mithat Gonen; Anne-Katrin Emde; Minita Shah; Kanika Arora; Catherine Reeves; Nicholas D. Socci; Elizabeth J. Perlman; Cristina R. Antonescu; Charles W. M. Roberts; Hanno Steen; Elizabeth Mullen; David Torrents; Zhiping Weng; Scott A. Armstrong; Alex Kentsis

Nat. Genet.; doi:10.1038/ng.3866; corrected online 24 May 2017 In the version of this article initially published online, the affiliations for Jiali Zhuang listed an incorrect present address instead of an equal contribution. The error has been corrected in the print, PDF and HTML versions of this article.


Nature Medicine | 2018

DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia

Darren C. Johnson; Cornelius Y. Taabazuing; Marian C. Okondo; Ashley J. Chui; Sahana D. Rao; Fiona Brown; Casie Reed; Elizabeth Peguero; Elisa de Stanchina; Alex Kentsis; Daniel A. Bachovchin

Small-molecule inhibitors of the serine dipeptidases DPP8 and DPP9 (DPP8/9) induce a lytic form of cell death called pyroptosis in mouse and human monocytes and macrophages1,2. In mouse myeloid cells, Dpp8/9 inhibition activates the inflammasome sensor Nlrp1b, which in turn activates pro-caspase-1 to mediate cell death3, but the mechanism of DPP8/9 inhibitor-induced pyroptosis in human myeloid cells is not yet known. Here we show that the CARD-containing protein CARD8 mediates DPP8/9 inhibitor-induced pro-caspase-1-dependent pyroptosis in human myeloid cells. We further show that DPP8/9 inhibitors induce pyroptosis in the majority of human acute myeloid leukemia (AML) cell lines and primary AML samples, but not in cells from many other lineages, and that these inhibitors inhibit human AML progression in mouse models. Overall, this work identifies an activator of CARD8 in human cells and indicates that its activation by small-molecule DPP8/9 inhibitors represents a new potential therapeutic strategy for AML.Small-molecule inhibitors of the serine dipeptidases DPP8 and DPP9 block AML progression by promoting CARD8-dependent pyroptosis of leukemic myeloid cells.

Collaboration


Dive into the Casie Reed's collaboration.

Top Co-Authors

Avatar

Alex Kentsis

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Elisa de Stanchina

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hanno Steen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew L. Kung

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anton Henssen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eileen Jiang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Eric Still

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Richard Koche

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amy Eisenberg

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge