Barbara Spitzer
Memorial Sloan Kettering Cancer Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Barbara Spitzer.
Nature Medicine | 2015
Lindsay M. LaFave; Wendy Béguelin; Richard Koche; Matt Teater; Barbara Spitzer; Alan Chramiec; Efthymia Papalexi; Matthew Keller; Todd Hricik; Katerina Konstantinoff; Jean Baptiste Micol; Benjamin H. Durham; Sarah K. Knutson; John E. Campbell; Gil Blum; Xinxu Shi; Emma H. Doud; Andrei V. Krivtsov; Young Rock Chung; Inna Khodos; Elisa de Stanchina; Ouathek Ouerfelli; Prasad S. Adusumilli; Paul M. Thomas; Neil L. Kelleher; Minkui Luo; Heike Keilhack; Omar Abdel-Wahab; Ari Melnick; Scott A. Armstrong
The tumor suppressors BAP1 and ASXL1 interact to form a polycomb deubiquitinase complex that removes monoubiquitin from histone H2A lysine 119 (H2AK119Ub). However, BAP1 and ASXL1 are mutated in distinct cancer types, consistent with independent roles in regulating epigenetic state and malignant transformation. Here we demonstrate that Bap1 loss in mice results in increased trimethylated histone H3 lysine 27 (H3K27me3), elevated enhancer of zeste 2 polycomb repressive complex 2 subunit (Ezh2) expression, and enhanced repression of polycomb repressive complex 2 (PRC2) targets. These findings contrast with the reduction in H3K27me3 levels seen with Asxl1 loss. Conditional deletion of Bap1 and Ezh2 in vivo abrogates the myeloid progenitor expansion induced by Bap1 loss alone. Loss of BAP1 results in a marked decrease in H4K20 monomethylation (H4K20me1). Consistent with a role for H4K20me1 in the transcriptional regulation of EZH2, expression of SETD8—the H4K20me1 methyltransferase—reduces EZH2 expression and abrogates the proliferation of BAP1-mutant cells. Furthermore, mesothelioma cells that lack BAP1 are sensitive to EZH2 pharmacologic inhibition, suggesting a novel therapeutic approach for BAP1-mutant malignancies.
Blood | 2014
Swapna Thota; Aaron D. Viny; Hideki Makishima; Barbara Spitzer; Tomas Radivoyevitch; Bartlomiej Przychodzen; Mikkael A. Sekeres; Ross L. Levine; Jaroslaw P. Maciejewski
Somatic cohesin mutations have been reported in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). To account for the morphologic and cytogenetic diversity of these neoplasms, a well-annotated cohort of 1060 patients with myeloid malignancies including MDS (n = 386), myeloproliferative neoplasms (MPNs) (n = 55), MDS/MPNs (n = 169), and AML (n = 450) were analyzed for cohesin gene mutational status, gene expression, and therapeutic and survival outcomes. Somatic cohesin defects were detected in 12% of patients with myeloid malignancies, whereas low expression of these genes was present in an additional 15% of patients. Mutations of cohesin genes were mutually exclusive and mostly resulted in predicted loss of function. Patients with low cohesin gene expression showed similar expression signatures as those with somatic cohesin mutations. Cross-sectional deep-sequencing analysis for clonal hierarchy demonstrated STAG2, SMC3, and RAD21 mutations to be ancestral in 18%, 18%, and 47% of cases, respectively, and each expanded to clonal dominance concordant with disease transformation. Cohesin mutations were significantly associated with RUNX1, Ras-family oncogenes, and BCOR and ASXL1 mutations and were most prevalent in high-risk MDS and secondary AML. Cohesin defects were associated with poor overall survival (27.2 vs 40 months; P = .023), especially in STAG2 mutant MDS patients surviving >12 months (median survival 35 vs 50 months; P = .017).
Nature | 2016
Yu Liu; Chong Chen; Zhengmin Xu; Claudio Scuoppo; Cory D. Rillahan; Jianjiong Gao; Barbara Spitzer; Benedikt Bosbach; Edward R. Kastenhuber; Timour Baslan; Sarah Ackermann; Lihua Cheng; Qingguo Wang; Ting Niu; Nikolaus Schultz; Ross L. Levine; Alea A. Mills; Scott W. Lowe
Mutations disabling the TP53 tumour suppressor gene represent the most frequent events in human cancer and typically occur through a two-hit mechanism involving a missense mutation in one allele and a ‘loss of heterozygosity’ deletion encompassing the other. While TP53 missense mutations can also contribute gain-of-function activities that impact tumour progression, it remains unclear whether the deletion event, which frequently includes many genes, impacts tumorigenesis beyond TP53 loss alone. Here we show that somatic heterozygous deletion of mouse chromosome 11B3, a 4-megabase region syntenic to human 17p13.1, produces a greater effect on lymphoma and leukaemia development than Trp53 deletion. Mechanistically, the effect of 11B3 loss on tumorigenesis involves co-deleted genes such as Eif5a and Alox15b (also known as Alox8), the suppression of which cooperates with Trp53 loss to produce more aggressive disease. Our results imply that the selective advantage produced by human chromosome 17p deletion reflects the combined impact of TP53 loss and the reduced dosage of linked tumour suppressor genes.
Nature Medicine | 2016
Olga A. Guryanova; Kaitlyn Shank; Barbara Spitzer; Luisa Luciani; Richard Koche; Francine E. Garrett-Bakelman; Chezi Ganzel; Benjamin H. Durham; Abhinita Mohanty; Gregor Hoermann; Sharon A. Rivera; Alan Chramiec; Elodie Pronier; Lennart Bastian; Matthew Keller; Daniel Tovbin; Evangelia Loizou; Abby Weinstein; Adriana Rodriguez Gonzalez; Yen K. Lieu; Jacob M. Rowe; Friederike Pastore; Anna Sophia McKenney; Andrei V. Krivtsov; Wolfgang R. Sperr; Justin R. Cross; Christopher E. Mason; Martin S. Tallman; Maria E. Arcila; Omar Abdel-Wahab
Although the majority of patients with acute myeloid leukemia (AML) initially respond to chemotherapy, many of them subsequently relapse, and the mechanistic basis for AML persistence following chemotherapy has not been determined. Recurrent somatic mutations in DNA methyltransferase 3A (DNMT3A), most frequently at arginine 882 (DNMT3AR882), have been observed in AML and in individuals with clonal hematopoiesis in the absence of leukemic transformation. Patients with DNMT3AR882 AML have an inferior outcome when treated with standard-dose daunorubicin-based induction chemotherapy, suggesting that DNMT3AR882 cells persist and drive relapse. We found that Dnmt3a mutations induced hematopoietic stem cell expansion, cooperated with mutations in the FMS-like tyrosine kinase 3 gene (Flt3ITD) and the nucleophosmin gene (Npm1c) to induce AML in vivo, and promoted resistance to anthracycline chemotherapy. In patients with AML, the presence of DNMT3AR882 mutations predicts minimal residual disease, underscoring their role in AML chemoresistance. DNMT3AR882 cells showed impaired nucleosome eviction and chromatin remodeling in response to anthracycline treatment, which resulted from attenuated recruitment of histone chaperone SPT-16 following anthracycline exposure. This defect led to an inability to sense and repair DNA torsional stress, which resulted in increased mutagenesis. Our findings identify a crucial role for DNMT3AR882 mutations in driving AML chemoresistance and highlight the importance of chromatin remodeling in response to cytotoxic chemotherapy.
Leukemia | 2016
Olga A. Guryanova; Yen K. Lieu; Francine E. Garrett-Bakelman; Barbara Spitzer; Jacob L. Glass; Kaitlyn Shank; Ana Belen Valencia Martinez; Sharon A. Rivera; Benjamin H. Durham; Franck Rapaport; Matthew Keller; Suveg Pandey; Lennart Bastian; Daniel Tovbin; Abby Weinstein; Julie Teruya-Feldstein; Omar Abdel-Wahab; Valeria Santini; Christopher E. Mason; Ari Melnick; Siddhartha Mukherjee; Ross L. Levine
DNA methyltransferase 3A (DNMT3A) mutations are observed in myeloid malignancies, including myeloproliferative neoplasms (MPN), myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Transplantation studies have elucidated an important role for Dnmt3a in stem cell self-renewal and in myeloid differentiation. Here, we investigated the impact of conditional hematopoietic Dnmt3a loss on disease phenotype in primary mice. Mx1-Cre-mediated Dnmt3a ablation led to the development of a lethal, fully penetrant MPN with myelodysplasia (MDS/MPN) characterized by peripheral cytopenias and by marked, progressive hepatomegaly. We detected expanded stem/progenitor populations in the liver of Dnmt3a-ablated mice. The MDS/MPN induced by Dnmt3a ablation was transplantable, including the marked hepatomegaly. Homing studies showed that Dnmt3a-deleted bone marrow cells preferentially migrated to the liver. Gene expression and DNA methylation analyses of progenitor cell populations identified differential regulation of hematopoietic regulatory pathways, including fetal liver hematopoiesis transcriptional programs. These data demonstrate that Dnmt3a ablation in the hematopoietic system leads to myeloid transformation in vivo, with cell-autonomous aberrant tissue tropism and marked extramedullary hematopoiesis (EMH) with liver involvement. Hence, in addition to the established role of Dnmt3a in regulating self-renewal, Dnmt3a regulates tissue tropism and limits myeloid progenitor expansion in vivo.
Oncotarget | 2016
Su Fern Tan; Xin Liu; Todd E. Fox; Brian M. Barth; Arati Sharma; Stephen D. Turner; Andy Awwad; Alden Dewey; Kenichiro Doi; Barbara Spitzer; Mithun Vinod Shah; Samy A.F. Morad; Dhimant Desai; Shantu Amin; Junjia Zhu; Jason Liao; Jong K. Yun; Mark Kester; David F. Claxton; Hong-Gang Wang; Myles C. Cabot; Edward H. Schuchman; Ross L. Levine; David J. Feith; Thomas P. Loughran
There is an urgent unmet need for new therapeutics in acute myeloid leukemia (AML) as standard therapy has not changed in the past three decades and outcome remains poor for most patients. Sphingolipid dysregulation through decreased ceramide levels and elevated sphingosine 1-phosphate (S1P) promotes cancer cell growth and survival. Acid ceramidase (AC) catalyzes ceramide breakdown to sphingosine, the precursor for S1P. We report for the first time that AC is required for AML blast survival. Transcriptome analysis and enzymatic assay show that primary AML cells have high levels of AC expression and activity. Treatment of patient samples and cell lines with AC inhibitor LCL204 reduced viability and induced apoptosis. AC overexpression increased the expression of anti-apoptotic Mcl-1, significantly increased S1P and decreased ceramide. Conversely, LCL204 induced ceramide accumulation and decreased Mcl-1 through post-translational mechanisms. LCL204 treatment significantly increased overall survival of C57BL/6 mice engrafted with leukemic C1498 cells and significantly decreased leukemic burden in NSG mice engrafted with primary human AML cells. Collectively, these studies demonstrate that AC plays a critical role in AML survival through regulation of both sphingolipid levels and Mcl-1. We propose that AC warrants further exploration as a novel therapeutic target in AML.
Frontiers in Oncology | 2015
Kavitha Ramaswamy; Barbara Spitzer; Alex Kentsis
Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that is required for normal cell growth and development. PP2A is a potent tumor suppressor, which is inactivated in cancer cells as a result of genetic deletions and mutations. In myeloid leukemias, genes encoding PP2A subunits are generally intact. Instead, PP2A is functionally inhibited by post-translational modifications of its catalytic C subunit, and interactions with negative regulators by its regulatory B and scaffold A subunits. Here, we review the molecular mechanisms of genetic and functional inactivation of PP2A in human cancers, with a particular focus on human acute myeloid leukemias (AML). By analyzing expression of genes encoding PP2A subunits using transcriptome sequencing, we find that PP2A dysregulation in AML is characterized by silencing and overexpression of distinct A scaffold and B regulatory subunits, respectively. We review the mechanisms of functional PP2A activation by drugs such as fingolimod, forskolin, OP449, and perphenazine. This analysis yields two non-mutually exclusive mechanisms for therapeutic PP2A re-activation: (i) allosteric activation of the phosphatase activity, and (ii) stabilization of active holo-enzyme assembly and displacement of negative regulatory factors from A and B subunits. Future studies should allow the development of specific and potent pharmacologic activators of PP2A, and definition of susceptible disease subsets based on specific mechanisms of PP2A dysregulation.
Cancer Discovery | 2018
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.
International Journal of Surgical Pathology | 2013
Olga L. Bohn; Joseph Whitten; Barbara Spitzer; Rachel Kobos; Susan E. Prockop; Farid Boulad; Maria E. Arcila; Lu Wang; Julie Teruya-Feldstein
Dyskeratosis congenita (DC) is a rare inherited disorder characterized by bone marrow failure and cancer predisposition. We present a case of a 28-year-old woman with DC who was admitted for hematopoietic stem cell transplantation (HSCT) for aplastic anemia and who developed acute myeloid leukemia with complex genetic karyotype abnormalities including the MLL (11q23) gene, 1q25, and chromosome 8. After transplantation, a monomorphic Epstein–Barr virus (EBV) negative posttransplant-associated lymphoproliferative disorder (PTLD) diffuse large B-cell lymphoma was discovered involving the liver, omental tissue, and peritoneal fluid samples showing additional MLL (11q23) gene abnormalities by fluorescence in situ hybridization. Despite treatment, the patient died of complications associated with transplantation and invasive fungal infection. This case represents the first bona fide documented case of EBV-negative monomorphic PTLD host derived, with MLL gene abnormalities in a patient with DC, and shows another possible mechanism for the development of a therapy-related lymphoid neoplasm after transplantation.
Pediatric Blood & Cancer | 2015
Barbara Spitzer; Patricia J. Giardina; Richard j. O'Reilly; Farid Boulad
Allo-HSCT is a well-established therapeutic approach for patients with hemoglobinopathies. Post-transplant mixed chimerism (MC) has been described in a subset of thalassemia patients receiving allo-HSCT that appears stable after an initial period of fluctuation. We describe the late complications of two patients with thalassemia who developed MC following allo-HSCT but ultimately lost their grafts, raising concern regarding the long-term stability of post-transplant MC. Two patients received allo-HSCT at our institution for bthalassemia major in the 1980s. These patients received matched sibling BMTs following cytoreduction with TBI 720 cGy and cyclophosphamide 120mg/kg. Patients were 6 years old at the time of transplant; Lucarelli Risk classification was class I for patient 1 and class II for patient 2. Cell doses were 4 10 and 2 10 nucleated cells/kg respectively and GVHDprophylaxis consisted of methotrexate þ/ cyclosporine. At three months post-HSCT, patient 1 had 97% donor cells in unsorted PB. Six years post-HSCT his BM chimerism status was 80% donor. At 16 years post-HSCT, he required resumption of regular transfusions with >90% host PB chimerism (Fig. 1A). One year after restarting transfusions, BM studies revealed host-derived therapy-related MDS with cytogenetic abnormalities and 100% host chimerism. He subsequently received cytoreduction and a secondary graft from his original donor with engraftment and full donor chimerism and remains alive and well. Patient 2 had an early decrease in BM donor chimerism from 97% ondayþ14 to 50%at 2months.Hewas able to sustain his hemoglobin at 9.5 g/dL despite a decrease of donor chimerism to 13% in unsorted PB at 2 years post-HSCT. Subsequently he had progressive loss of donor chimerism, and a gradual decline in his hemoglobin until he required resumption of regular transfusions at 15 years post-HSCT with 0% donor cells and no evidence of MDS (Fig. 1B). Allo-HSCT is the only currently established curative approach for patients with hemoglobinopathies, and despite myeloablative conditioning, mixed donor-host chimerism is a known potential complication. Patients with earlyMCmay develop late graft failure, and are risk-stratified by percentage of residual host cells at two months post-HSCT: high-risk if >25%, low-risk if <10% [1]. At two years post-HSCT, the majority of patients either rejected their grafts or converted to full donor chimerism, while 19–27% had persistent MC. These patients remained transfusion-free from 2 to 11 years post-transplant. Little data are available in patients with persistent MC >11 years post-transplant. One patient was reported with late unstable mixed chimerism 22 years after matched sibling allo-HSCT for thalassemia, despite early full donor chimerism. This patient recovered full donor chimerism after DLI, though suffered both acute and chronic GVHD [2]. Our patients received a low-dose TBI-based regimen with initial stable mixed chimerism, but developed very late complications