keyword
https://read.qxmd.com/read/32343799/fedratinib-in-myelofibrosis
#21
JOURNAL ARTICLE
Ann Mullally, John Hood, Claire Harrison, Ruben Mesa
Following the discovery of the JAK2V617F mutation in myeloproliferative neoplasms in 2005, fedratinib was developed as a small molecular inhibitor of JAK2. It was optimized to yield low-nanomolar activity against JAK2 (50% inhibitory concentration = 3 nM) and was identified to be selective for JAK2 relative to other JAK family members (eg, JAK1, JAK3, and TYK2). It quickly moved into clinical development with a phase 1 clinical trial opening in 2008, where a favorable impact on spleen and myelofibrosis (MF) symptom responses was reported...
April 28, 2020: Blood Advances
https://read.qxmd.com/read/31917838/both-sides-now-losses-and-gains-of-mutant-calr
#22
COMMENT
Ann Mullally
No abstract text is available yet for this article.
January 9, 2020: Blood
https://read.qxmd.com/read/31732720/distinct-effects-of-ruxolitinib-and-interferon-alpha-on-murine-jak2v617f-myeloproliferative-neoplasm-hematopoietic-stem-cell-populations
#23
JOURNAL ARTICLE
Rebecca J Austin, Jasmin Straube, Claudia Bruedigam, Gabor Pali, Sebastien Jacquelin, Therese Vu, Joanne Green, Julius Gräsel, Lianne Lansink, Leanne Cooper, Shin-Jye Lee, Nien-Tsu Chen, Chung-Wei Lee, Ashraful Haque, Florian H Heidel, Richard D'Andrea, Geoff R Hill, Ann Mullally, Michael D Milsom, Megan Bywater, Steven W Lane
JAK2V617F is the most common mutation in patients with BCR-ABL negative myeloproliferative neoplasms (MPNs). The eradication of JAK2V617F hematopoietic stem cells (HSCs) is critical for achieving molecular remissions and cure. We investigate the distinct effects of two therapies, ruxolitinib (JAK1/2 inhibitor) and interferon-alpha (IFN-α), on the disease-initiating HSC population. Whereas ruxolitinib inhibits Stat5 activation in erythroid progenitor populations, it fails to inhibit this same pathway in HSCs...
April 2020: Leukemia
https://read.qxmd.com/read/31562135/mutant-calreticulin-in-myeloproliferative-neoplasms
#24
JOURNAL ARTICLE
Joan How, Gabriela S Hobbs, Ann Mullally
Recurrent mutations in calreticulin are present in ∼20% of patients with myeloproliferative neoplasms (MPNs). Since its discovery in 2013, we now have a more precise understanding of how mutant CALR, an endoplasmic reticulum chaperone protein, activates the JAK/STAT signaling pathway via a pathogenic binding interaction with the thrombopoietin receptor MPL to induce MPNs. In this Spotlight article, we review the current understanding of the biology underpinning mutant CALR-driven MPNs, discuss clinical implications, and highlight future therapeutic approaches...
December 19, 2019: Blood
https://read.qxmd.com/read/31548225/the-molecular-genetics-of-myeloproliferative-neoplasms
#25
REVIEW
Anna E Marneth, Ann Mullally
Activated JAK-STAT signaling is central to the pathogenesis of BCR-ABL -negative myeloproliferative neoplasms (MPNs) and occurs as a result of MPN phenotypic driver mutations in JAK2 , CALR , or MPL The spectrum of concomitant somatic mutations in other genes has now largely been defined in MPNs. With the integration of targeted next-generation sequencing (NGS) panels into clinical practice, the clinical significance of concomitant mutations in MPNs has become clearer. In this review, we describe the consequences of concomitant mutations in the most frequently mutated classes of genes in MPNs: (1) DNA methylation pathways, (2) chromatin modification, (3) RNA splicing, (4) signaling pathways, (5) transcription factors, and (6) DNA damage response/stress signaling...
February 3, 2020: Cold Spring Harbor Perspectives in Medicine
https://read.qxmd.com/read/30504299/jak2-and-other-genes-be-nimble-with-mpn-diagnosis-prognosis-and-therapy
#26
REVIEW
Michele Ciboddo, Ann Mullally
Now that the spectrum of somatic mutations that initiate, propagate, and drive the progression of myeloproliferative neoplasms (MPNs) has largely been defined, recent efforts have focused on integrating this information into clinical decision making. In this regard, the greatest progress has been made in myelofibrosis, in which high-molecular-risk mutations have been identified and incorporated into prognostic models to help guide treatment decisions. In this chapter, we focus on advances in 4 main areas: (1) What are the MPN phenotypic driver mutations? (2) What constitutes high molecular risk in MPN (focusing on ASXL1 )? (3) How do we risk-stratify patients with MPN? And (4) What is the significance of molecular genetics for MPN treatment? Although substantial progress has been made, we still have an incomplete understanding of the molecular basis for phenotypic diversity in MPN, and few rationally designed therapeutic approaches to target high-risk mutations are available...
November 30, 2018: Hematology—the Education Program of the American Society of Hematology
https://read.qxmd.com/read/30504235/the-kinases-ikbke-and-tbk1-regulate-myc-dependent-survival-pathways-through-yb-1-in-aml-and-are-targets-for-therapy
#27
JOURNAL ARTICLE
Suhu Liu, Anna E Marneth, Gabriela Alexe, Sarah R Walker, Helen I Gandler, Darwin Q Ye, Katherine Labella, Radhika Mathur, Patricia A Toniolo, Michelle Tillgren, Prafulla C Gokhale, David Barbie, Ann Mullally, Kimberly Stegmaier, David A Frank
To identify novel therapeutic targets in acute myeloid leukemia (AML), we examined kinase expression patterns in primary AML samples. We found that the serine/threonine kinase IKBKE, a noncanonical IkB kinase, is expressed at higher levels in myeloid leukemia cells compared with normal hematopoietic cells. Inhibiting IKBKE, or its close homolog TANK-binding kinase 1 (TBK1), by either short hairpin RNA knockdown or pharmacological compounds, induces apoptosis and reduces the viability of AML cells. Using gene expression profiling and gene set enrichment analysis, we found that IKBKE/TBK1-sensitive AML cells typically possess an MYC oncogenic signature...
December 11, 2018: Blood Advances
https://read.qxmd.com/read/30075132/gli1-mesenchymal-stromal-cells-are-a-key-driver-of-bone-marrow-fibrosis-and-an-important-cellular-therapeutic-target
#28
Rebekka K Schneider, Ann Mullally, Aurelien Dugourd, Fabian Peisker, Remco Hoogenboezem, Paulina M H Van Strien, Eric M Bindels, Dirk Heckl, Guntram Büsche, David Fleck, Gerhard Müller-Newen, Janewit Wongboonsin, Monica Ventura Ferreira, Victor G Puelles, Julio Saez-Rodriguez, Benjamin L Ebert, Benjamin D Humphreys, Rafael Kramann
No abstract text is available yet for this article.
August 2, 2018: Cell Stem Cell
https://read.qxmd.com/read/29643232/increased-neutrophil-extracellular-trap-formation-promotes-thrombosis-in-myeloproliferative-neoplasms
#29
JOURNAL ARTICLE
Ofir Wolach, Rob S Sellar, Kimberly Martinod, Deya Cherpokova, Marie McConkey, Ryan J Chappell, Alexander J Silver, Dylan Adams, Cecilia A Castellano, Rebekka K Schneider, Robert F Padera, Daniel J DeAngelo, Martha Wadleigh, David P Steensma, Ilene Galinsky, Richard M Stone, Giulio Genovese, Steven A McCarroll, Bozenna Iliadou, Christina Hultman, Donna Neuberg, Ann Mullally, Denisa D Wagner, Benjamin L Ebert
Thrombosis is a major cause of morbidity and mortality in Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), clonal disorders of hematopoiesis characterized by activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. Neutrophil extracellular trap (NET) formation, a component of innate immunity, has been linked to thrombosis. We demonstrate that neutrophils from patients with MPNs are primed for NET formation, an effect blunted by pharmacological inhibition of JAK signaling...
April 11, 2018: Science Translational Medicine
https://read.qxmd.com/read/29364275/using-crispr-cas9-gene-editing-to-investigate-the-oncogenic-activity-of-mutant-calreticulin-in-cytokine-dependent-hematopoietic-cells
#30
JOURNAL ARTICLE
Nouran S Abdelfattah, Ann Mullally
Clustered regularly interspaced short palindromic repeats (CRISPR) is an adaptive immunity system in prokaryotes that has been repurposed by scientists to generate RNA-guided nucleases, such as CRISPR-associated (Cas) 9 for site-specific eukaryotic genome editing. Genome engineering by Cas9 is used to efficiently, easily and robustly modify endogenous genes in many biomedically-relevant mammalian cell lines and organisms. Here we show an example of how to utilize the CRISPR/Cas9 methodology to understand the biological function of specific genetic mutations...
January 5, 2018: Journal of Visualized Experiments: JoVE
https://read.qxmd.com/read/29288169/defining-the-requirements-for-the-pathogenic-interaction-between-mutant-calreticulin-and-mpl-in-mpn
#31
JOURNAL ARTICLE
Shannon Elf, Nouran S Abdelfattah, April J Baral, Danielle Beeson, Jeanne F Rivera, Amy Ko, Natalie Florescu, Gabriel Birrane, Edwin Chen, Ann Mullally
Mutations in calreticulin ( CALR ) are phenotypic drivers in the pathogenesis of myeloproliferative neoplasms. Mechanistic studies have demonstrated that mutant CALR binds to the thrombopoietin receptor MPL, and that the positive electrostatic charge of the mutant CALR C terminus is required for mutant CALR-mediated activation of JAK-STAT signaling. Here we demonstrate that although binding between mutant CALR and MPL is required for mutant CALR to transform hematopoietic cells; binding alone is insufficient for cytokine independent growth...
February 15, 2018: Blood
https://read.qxmd.com/read/28673398/kinase-inhibitors-in-the-treatment-of-myeloid-malignancies
#32
EDITORIAL
Ann Mullally
No abstract text is available yet for this article.
August 2017: Hematology/oncology Clinics of North America
https://read.qxmd.com/read/28673391/the-development-and-use-of-janus-kinase-2-inhibitors-for-the-treatment-of-myeloproliferative-neoplasms
#33
REVIEW
Gabriela S Hobbs, Sarah Rozelle, Ann Mullally
Following the discovery of the JAK2V617F mutation, Janus kinase (JAK) 2 inhibitors were developed as rationally designed therapy in myeloproliferative neoplasms (MPNs). Although JAK2 inhibitors have clinical efficacy in MPN, they are not clonally selective for the JAK2V617F-mutant cells. Because activated JAK-signal transducer and activator of transcription (STAT) signaling is a common feature of MPN, JAK2 inhibitors are efficacious regardless of the specific MPN phenotypic driver mutation. The Food and Drug Administration approved the JAK1/JAK2 inhibitor, ruxolitinib, for the treatment of myelofibrosis and polycythemia vera...
August 2017: Hematology/oncology Clinics of North America
https://read.qxmd.com/read/28457748/gli1-mesenchymal-stromal-cells-are-a-key-driver-of-bone-marrow-fibrosis-and-an-important-cellular-therapeutic-target
#34
JOURNAL ARTICLE
Rebekka K Schneider, Ann Mullally, Aurelien Dugourd, Fabian Peisker, Remco Hoogenboezem, Paulina M H Van Strien, Eric M Bindels, Dirk Heckl, Guntram Büsche, David Fleck, Gerhard Müller-Newen, Janewit Wongboonsin, Monica Ventura Ferreira, Victor G Puelles, Julio Saez-Rodriguez, Benjamin L Ebert, Benjamin D Humphreys, Rafael Kramann
Bone marrow fibrosis (BMF) develops in various hematological and non-hematological conditions and is a central pathological feature of myelofibrosis. Effective cell-targeted therapeutics are needed, but the cellular origin of BMF remains elusive. Here, we show using genetic fate tracing in two murine models of BMF that Gli1+ mesenchymal stromal cells (MSCs) are recruited from the endosteal and perivascular niche to become fibrosis-driving myofibroblasts in the bone marrow. Genetic ablation of Gli1+ cells abolished BMF and rescued bone marrow failure...
June 1, 2017: Cell Stem Cell
https://read.qxmd.com/read/28159736/myeloproliferative-neoplasm-stem-cells
#35
REVIEW
Adam J Mead, Ann Mullally
Myeloproliferative neoplasms (MPNs) arise in the hematopoietic stem cell (HSC) compartment as a result of the acquisition of somatic mutations in a single HSC that provides a selective advantage to mutant HSC over normal HSC and promotes myeloid differentiation to engender a myeloproliferative phenotype. This population of somatically mutated HSC, which initiates and sustains MPNs, is termed MPN stem cells. In >95% of cases, mutations that drive the development of an MPN phenotype occur in a mutually exclusive manner in 1 of 3 genes: JAK2 , CALR , or MPL The thrombopoietin receptor, MPL, is the key cytokine receptor in MPN development, and these mutations all activate MPL-JAK-STAT signaling in MPN stem cells...
March 23, 2017: Blood
https://read.qxmd.com/read/27712165/editorial
#36
JOURNAL ARTICLE
(no author information available yet)
SO WEare to have new chief nursing officers (CNOs) for England and Scotland. Sarah Mullally's surprise resignation last month paves the way for a new leader in England, while Paul Martin has been appointed Anne Jarvie's successor in Scotland. Maybe a new era for nurse leadership is in the offing?
July 1, 2004: Nursing Management (Harrow)
https://read.qxmd.com/read/27622333/physiologic-expression-of-sf3b1-k700e-causes-impaired-erythropoiesis-aberrant-splicing-and-sensitivity-to-therapeutic-spliceosome-modulation
#37
JOURNAL ARTICLE
Esther A Obeng, Ryan J Chappell, Michael Seiler, Michelle C Chen, Dean R Campagna, Paul J Schmidt, Rebekka K Schneider, Allegra M Lord, Lili Wang, Rutendo G Gambe, Marie E McConkey, Abdullah M Ali, Azra Raza, Lihua Yu, Silvia Buonamici, Peter G Smith, Ann Mullally, Catherine J Wu, Mark D Fleming, Benjamin L Ebert
More than 80% of patients with the refractory anemia with ring sideroblasts subtype of myelodysplastic syndrome (MDS) have mutations in Splicing Factor 3B, Subunit 1 (SF3B1). We generated a conditional knockin mouse model of the most common SF3B1 mutation, Sf3b1(K700E). Sf3b1(K700E) mice develop macrocytic anemia due to a terminal erythroid maturation defect, erythroid dysplasia, and long-term hematopoietic stem cell (LT-HSC) expansion. Sf3b1(K700E) myeloid progenitors and SF3B1-mutant MDS patient samples demonstrate aberrant 3' splice-site selection associated with increased nonsense-mediated decay...
September 12, 2016: Cancer Cell
https://read.qxmd.com/read/26951227/mutant-calreticulin-requires-both-its-mutant-c-terminus-and-the-thrombopoietin-receptor-for-oncogenic-transformation
#38
JOURNAL ARTICLE
Shannon Elf, Nouran S Abdelfattah, Edwin Chen, Javier Perales-Patón, Emily A Rosen, Amy Ko, Fabian Peisker, Natalie Florescu, Silvia Giannini, Ofir Wolach, Elizabeth A Morgan, Zuzana Tothova, Julie-Aurore Losman, Rebekka K Schneider, Fatima Al-Shahrour, Ann Mullally
UNLABELLED: Somatic mutations in calreticulin (CALR) are present in approximately 40% of patients with myeloproliferative neoplasms (MPN), but the mechanism by which mutant CALR is oncogenic remains unclear. Here, we demonstrate that expression of mutant CALR alone is sufficient to engender MPN in mice and recapitulates the disease phenotype of patients with CALR-mutant MPN. We further show that the thrombopoietin receptor MPL is required for mutant CALR-driven transformation through JAK-STAT pathway activation, thus rendering mutant CALR-transformed hematopoietic cells sensitive to JAK2 inhibition...
April 2016: Cancer Discovery
https://read.qxmd.com/read/26686625/recql5-suppresses-oncogenic-jak2-induced-replication-stress-and-genomic-instability
#39
JOURNAL ARTICLE
Edwin Chen, Jong Sook Ahn, David B Sykes, Lawrence J Breyfogle, Anna L Godfrey, Jyoti Nangalia, Amy Ko, Daniel J DeAngelo, Anthony R Green, Ann Mullally
JAK2V617F is the most common oncogenic lesion in patients with myeloproliferative neoplasms (MPNs). Despite the ability of JAK2V617F to instigate DNA damage in vitro, MPNs are nevertheless characterized by genomic stability. In this study, we address this paradox by identifying the DNA helicase RECQL5 as a suppressor of genomic instability in MPNs. We report increased RECQL5 expression in JAK2V617F-expressing cells and demonstrate that RECQL5 is required to counteract JAK2V617F-induced replication stress. Moreover, RECQL5 depletion sensitizes JAK2V617F mutant cells to hydroxyurea (HU), a pharmacological inducer of replication stress and the most common treatment for MPNs...
December 22, 2015: Cell Reports
https://read.qxmd.com/read/26569382/targeting-megakaryocytic-induced-fibrosis-in-myeloproliferative-neoplasms-by-aurka-inhibition
#40
JOURNAL ARTICLE
Qiang Jeremy Wen, Qiong Yang, Benjamin Goldenson, Sébastien Malinge, Terra Lasho, Rebekka K Schneider, Lawrence J Breyfogle, Rachael Schultz, Laure Gilles, Priya Koppikar, Omar Abdel-Wahab, Animesh Pardanani, Brady Stein, Sandeep Gurbuxani, Ann Mullally, Ross L Levine, Ayalew Tefferi, John D Crispino
Primary myelofibrosis (PMF) is characterized by bone marrow fibrosis, myeloproliferation, extramedullary hematopoiesis, splenomegaly and leukemic progression. Moreover, the bone marrow and spleens of individuals with PMF contain large numbers of atypical megakaryocytes that are postulated to contribute to fibrosis through the release of cytokines, including transforming growth factor (TGF)-β. Although the Janus kinase inhibitor ruxolitinib provides symptomatic relief, it does not reduce the mutant allele burden or substantially reverse fibrosis...
December 2015: Nature Medicine
keyword
keyword
165587
2
3
Fetch more papers »
Fetching more papers... Fetching...
Remove bar
Read by QxMD icon Read
×

Save your favorite articles in one place with a free QxMD account.

×

Search Tips

Use Boolean operators: AND/OR

diabetic AND foot
diabetes OR diabetic

Exclude a word using the 'minus' sign

Virchow -triad

Use Parentheses

water AND (cup OR glass)

Add an asterisk (*) at end of a word to include word stems

Neuro* will search for Neurology, Neuroscientist, Neurological, and so on

Use quotes to search for an exact phrase

"primary prevention of cancer"
(heart or cardiac or cardio*) AND arrest -"American Heart Association"

We want to hear from doctors like you!

Take a second to answer a survey question.