Skip to main content
Top
Published in: International Journal of Hematology 2/2019

01-08-2019 | Primary Myelofibrosis | Progress in Hematology

Deregulated Polycomb functions in myeloproliferative neoplasms

Authors: Goro Sashida, Motohiko Oshima, Atsushi Iwama

Published in: International Journal of Hematology | Issue 2/2019

Login to get access

Abstract

Polycomb proteins function in the maintenance of gene silencing via post-translational modifications of histones and chromatin compaction. Genetic and biochemical studies have revealed that the repressive function of Polycomb repressive complexes (PRCs) in transcription is counteracted by the activating function of Trithorax-group complexes; this balance fine-tunes the expression of genes critical for development and tissue homeostasis. The function of PRCs is frequently dysregulated in various cancer cells due to altered expression or recurrent somatic mutations in PRC genes. The tumor suppressive functions of EZH2-containing PRC2 and a PRC2-related protein ASXL1 have been investigated extensively in the pathogenesis of hematological malignancies, including myeloproliferative neoplasms (MPN). BCOR, a component of non-canonical PRC1, suppresses various hematological malignancies including MPN. In this review, we focus on recent findings on the role of PRCs in the pathogenesis of MPN and the therapeutic impact of targeting the pathological functions of PRCs in MPN.
Literature
1.
2.
go back to reference Blackledge NP, Rose NR, Klose RJ. Targeting Polycomb systems to regulate gene expression: modifications to a complex story. Nat Rev Mol Cell Biol. 2015;16(11):643–9.PubMedPubMedCentralCrossRef Blackledge NP, Rose NR, Klose RJ. Targeting Polycomb systems to regulate gene expression: modifications to a complex story. Nat Rev Mol Cell Biol. 2015;16(11):643–9.PubMedPubMedCentralCrossRef
4.
go back to reference Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12(9):599–612.PubMedCrossRef Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012;12(9):599–612.PubMedCrossRef
5.
go back to reference Comet I, Riising EM, Leblanc B, Helin K. Maintaining cell identity: PRC2-mediated regulation of transcription and cancer. Nat Rev Cancer. 2016;16(12):803–10.PubMedCrossRef Comet I, Riising EM, Leblanc B, Helin K. Maintaining cell identity: PRC2-mediated regulation of transcription and cancer. Nat Rev Cancer. 2016;16(12):803–10.PubMedCrossRef
6.
go back to reference Sashida G, Iwama A. Multifaceted role of the Polycomb-group gene EZH2 in hematological malignancies. Int J Hematol. 2017;105(1):23–30.PubMedCrossRef Sashida G, Iwama A. Multifaceted role of the Polycomb-group gene EZH2 in hematological malignancies. Int J Hematol. 2017;105(1):23–30.PubMedCrossRef
7.
go back to reference Isshiki Y, Iwama A. Emerging role of non-canonical Polycomb repressive complexes in normal and malignant hematopoiesis. Exp Hematol. 2018 (Published online: October 26, 2018). Isshiki Y, Iwama A. Emerging role of non-canonical Polycomb repressive complexes in normal and malignant hematopoiesis. Exp Hematol. 2018 (Published online: October 26, 2018).
8.
go back to reference Simon J, Kingston RE. Mechanisms of Polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol. 2009;10(10):697–708.PubMedCrossRef Simon J, Kingston RE. Mechanisms of Polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol. 2009;10(10):697–708.PubMedCrossRef
9.
go back to reference Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature. 2004;431(7010):873–8.PubMedCrossRef Wang H, Wang L, Erdjument-Bromage H, Vidal M, Tempst P, Jones RS, et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature. 2004;431(7010):873–8.PubMedCrossRef
10.
go back to reference Gao Z, Zhang J, Bonasio R, Strino F, Sawai A, Parisi F, et al. PCGF Homologs, CBX Proteins, and RYBP define functionally distinct PRC1 family complexes. Mol Cell. 2012;45(3):344–56.PubMedPubMedCentralCrossRef Gao Z, Zhang J, Bonasio R, Strino F, Sawai A, Parisi F, et al. PCGF Homologs, CBX Proteins, and RYBP define functionally distinct PRC1 family complexes. Mol Cell. 2012;45(3):344–56.PubMedPubMedCentralCrossRef
11.
go back to reference Radulovic V, de Haan G, Klauke K. Polycomb-group proteins in hematopoietic stem cell regulation and hematopoietic neoplasms. Leukemia. 2013;27(3):523–33.PubMedCrossRef Radulovic V, de Haan G, Klauke K. Polycomb-group proteins in hematopoietic stem cell regulation and hematopoietic neoplasms. Leukemia. 2013;27(3):523–33.PubMedCrossRef
12.
go back to reference Xie H, Xu J, Hsu JH, Nguyen M, Fujiwara Y, Peng C, et al. Polycomb repressive complex 2 regulates normal hematopoietic stem cell function in a developmental-stage-specific manner. Cell Stem Cell. 2014;14(1):68–80.PubMedCrossRef Xie H, Xu J, Hsu JH, Nguyen M, Fujiwara Y, Peng C, et al. Polycomb repressive complex 2 regulates normal hematopoietic stem cell function in a developmental-stage-specific manner. Cell Stem Cell. 2014;14(1):68–80.PubMedCrossRef
13.
go back to reference Hidalgo I, Herrera-Merchan A, Ligos JM, Carramolino L, Nunez J, Martinez F, et al. Ezh1 is required for hematopoietic stem cell maintenance and prevents senescence-like cell cycle arrest. Cell Stem Cell. 2012;11(5):649–62.PubMedCrossRef Hidalgo I, Herrera-Merchan A, Ligos JM, Carramolino L, Nunez J, Martinez F, et al. Ezh1 is required for hematopoietic stem cell maintenance and prevents senescence-like cell cycle arrest. Cell Stem Cell. 2012;11(5):649–62.PubMedCrossRef
14.
go back to reference Mochizuki-Kashio M, Aoyama K, Sashida G, Oshima M, Tomioka T, Muto T, et al. Ezh2 loss in hematopoietic stem cells predisposes mice to develop heterogeneous malignancies in an Ezh1-dependent manner. Blood. 2015;126(10):1172–83.PubMedCrossRef Mochizuki-Kashio M, Aoyama K, Sashida G, Oshima M, Tomioka T, Muto T, et al. Ezh2 loss in hematopoietic stem cells predisposes mice to develop heterogeneous malignancies in an Ezh1-dependent manner. Blood. 2015;126(10):1172–83.PubMedCrossRef
15.
go back to reference Aoyama K, Oshima M, Koide S, Suzuki E, Mochizuki-Kashio M, Kato Y, et al. Ezh1 targets bivalent genes to maintain self-renewing stem cells in Ezh2-insufficient myelodysplastic syndrome. iScience. 2018;9:161–74.PubMedPubMedCentralCrossRef Aoyama K, Oshima M, Koide S, Suzuki E, Mochizuki-Kashio M, Kato Y, et al. Ezh1 targets bivalent genes to maintain self-renewing stem cells in Ezh2-insufficient myelodysplastic syndrome. iScience. 2018;9:161–74.PubMedPubMedCentralCrossRef
16.
go back to reference Su I, Dobenecker M-W, Dickinson E, Oser M, Basavaraj A, Marqueron R, et al. Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005;121(3):425–36.PubMedCrossRef Su I, Dobenecker M-W, Dickinson E, Oser M, Basavaraj A, Marqueron R, et al. Polycomb group protein ezh2 controls actin polymerization and cell signaling. Cell. 2005;121(3):425–36.PubMedCrossRef
17.
go back to reference Park I, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423(6937):302–5.PubMedCrossRef Park I, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423(6937):302–5.PubMedCrossRef
18.
go back to reference Oguro H, Iwama A, Morita Y, Kamijo T, van Lohuizen M, Nakauchi H. Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice. J Exp Med. 2006;203(10):2247–53.PubMedPubMedCentralCrossRef Oguro H, Iwama A, Morita Y, Kamijo T, van Lohuizen M, Nakauchi H. Differential impact of Ink4a and Arf on hematopoietic stem cells and their bone marrow microenvironment in Bmi1-deficient mice. J Exp Med. 2006;203(10):2247–53.PubMedPubMedCentralCrossRef
19.
go back to reference Iwama A, Oguro H, Negishi M, Kato Y, Morita Y, Tsukui H, et al. Enhanced self-renewal of hematopoietic stem cells mediated by the Polycomb gene product Bmi-1. Immunity. 2004;21(6):843–51.PubMedCrossRef Iwama A, Oguro H, Negishi M, Kato Y, Morita Y, Tsukui H, et al. Enhanced self-renewal of hematopoietic stem cells mediated by the Polycomb gene product Bmi-1. Immunity. 2004;21(6):843–51.PubMedCrossRef
20.
go back to reference Oguro H, Yuan J, Ichikawa H, Ikawa T, Yamazaki S, Kawamoto H, et al. Poised lineage specification in multipotential hematopoietic stem and progenitor cells by the Polycomb protein Bmi1. Cell Stem Cell. 2010;6(3):279–86.PubMedCrossRef Oguro H, Yuan J, Ichikawa H, Ikawa T, Yamazaki S, Kawamoto H, et al. Poised lineage specification in multipotential hematopoietic stem and progenitor cells by the Polycomb protein Bmi1. Cell Stem Cell. 2010;6(3):279–86.PubMedCrossRef
21.
go back to reference Ross K, Sedello AK, Todd GP, Paszkowski-rogacz M, Bird AW, Grinenko T, et al. Polycomb group ring finger 1 cooperates with Runx1 in regulating differentiation and self-renewal of hematopoietic cells. Blood. 2012;119(18):4152–62.PubMedCrossRef Ross K, Sedello AK, Todd GP, Paszkowski-rogacz M, Bird AW, Grinenko T, et al. Polycomb group ring finger 1 cooperates with Runx1 in regulating differentiation and self-renewal of hematopoietic cells. Blood. 2012;119(18):4152–62.PubMedCrossRef
22.
go back to reference Cao Q, Gearhart MD, Gery S, Shojaee S, Yang H, Sun H, et al. BCOR regulates myeloid cell proliferation and differentiation. Leukemia. 2016;30(5):1155–65.PubMedPubMedCentralCrossRef Cao Q, Gearhart MD, Gery S, Shojaee S, Yang H, Sun H, et al. BCOR regulates myeloid cell proliferation and differentiation. Leukemia. 2016;30(5):1155–65.PubMedPubMedCentralCrossRef
23.
go back to reference Tara S, Isshiki Y, Nakajima-Takagi Y, Oshima M, Aoyama K, Tanaka T, et al. Bcor insufficiency promotes initiation and progression of myelodysplastic syndrome. Blood. 2018;132(23):2470–83.PubMedCrossRefPubMedCentral Tara S, Isshiki Y, Nakajima-Takagi Y, Oshima M, Aoyama K, Tanaka T, et al. Bcor insufficiency promotes initiation and progression of myelodysplastic syndrome. Blood. 2018;132(23):2470–83.PubMedCrossRefPubMedCentral
24.
go back to reference Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472–8.PubMedPubMedCentralCrossRef Xie M, Lu C, Wang J, McLellan MD, Johnson KJ, Wendl MC, et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med. 2014;20(12):1472–8.PubMedPubMedCentralCrossRef
25.
go back to reference Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose S, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.PubMedPubMedCentralCrossRef Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose S, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.PubMedPubMedCentralCrossRef
26.
go back to reference Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012;337(6101):1541–6.PubMedPubMedCentralCrossRef Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012;337(6101):1541–6.PubMedPubMedCentralCrossRef
27.
go back to reference Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210(12):2641–59.PubMedPubMedCentralCrossRef Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013;210(12):2641–59.PubMedPubMedCentralCrossRef
28.
go back to reference Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, et al. ASXL1 mutations promote myeloid transformation through Loss of PRC2-mediated gene repression. Cancer Cell. 2012;22(2):180–93.PubMedPubMedCentralCrossRef Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, et al. ASXL1 mutations promote myeloid transformation through Loss of PRC2-mediated gene repression. Cancer Cell. 2012;22(2):180–93.PubMedPubMedCentralCrossRef
29.
30.
go back to reference Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017;129(6):667–79.PubMedCrossRef Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017;129(6):667–79.PubMedCrossRef
31.
go back to reference Iwama A. Polycomb repressive complexes in hematological malignancies. Blood. 2018;130(1):23–30.CrossRef Iwama A. Polycomb repressive complexes in hematological malignancies. Blood. 2018;130(1):23–30.CrossRef
32.
go back to reference Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011;364(26):2496–506.PubMedPubMedCentralCrossRef Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med. 2011;364(26):2496–506.PubMedPubMedCentralCrossRef
33.
go back to reference Bejar R, Stevenson KE, Caughey BA, Abdel-Wahab O, Steensma DP, Galili N, et al. Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes. J Clin Oncol. 2012;30(27):3376–82.PubMedPubMedCentralCrossRef Bejar R, Stevenson KE, Caughey BA, Abdel-Wahab O, Steensma DP, Galili N, et al. Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes. J Clin Oncol. 2012;30(27):3376–82.PubMedPubMedCentralCrossRef
34.
go back to reference Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood. 2011;118(19):5227–34.PubMedCrossRef Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood. 2011;118(19):5227–34.PubMedCrossRef
35.
go back to reference Kotini AG, Chang C, Boussaad I, Delrow JJ, Dolezal EK, Nagulapally AB, et al. Functional analysis of a chromosomal deletion associated with myelodysplastic syndromes using isogenic human induced pluripotent stem cells. Nat Biotechnol. 2015;33(6):646–55.PubMedPubMedCentralCrossRef Kotini AG, Chang C, Boussaad I, Delrow JJ, Dolezal EK, Nagulapally AB, et al. Functional analysis of a chromosomal deletion associated with myelodysplastic syndromes using isogenic human induced pluripotent stem cells. Nat Biotechnol. 2015;33(6):646–55.PubMedPubMedCentralCrossRef
36.
go back to reference Makishima H, Jankowska AM, Tiu RV, Szpurka H, Sugimoto Y, Hu Z, et al. Novel homo- and hemizygous mutations in EZH2 in myeloid malignancies. Leukemia. 2010;24(10):1799–804.PubMedCrossRef Makishima H, Jankowska AM, Tiu RV, Szpurka H, Sugimoto Y, Hu Z, et al. Novel homo- and hemizygous mutations in EZH2 in myeloid malignancies. Leukemia. 2010;24(10):1799–804.PubMedCrossRef
37.
go back to reference Khan SN, Jankowska AM, Mahfouz R, Dunbar AJ, Sugimoto Y, Hosono N, et al. Multiple mechanisms deregulate EZH2 and histone H3 lysine 27 epigenetic changes in myeloid malignancies. Leukemia. 2013;27(6):1301–9.PubMedCrossRef Khan SN, Jankowska AM, Mahfouz R, Dunbar AJ, Sugimoto Y, Hosono N, et al. Multiple mechanisms deregulate EZH2 and histone H3 lysine 27 epigenetic changes in myeloid malignancies. Leukemia. 2013;27(6):1301–9.PubMedCrossRef
38.
go back to reference Sashida G, Harada H, Matsui H, Oshima M, Yui M, Harada Y, et al. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat Commun. 2014;5:4177.PubMedCrossRef Sashida G, Harada H, Matsui H, Oshima M, Yui M, Harada Y, et al. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat Commun. 2014;5:4177.PubMedCrossRef
39.
go back to reference Tanaka S, Miyagi S, Sashida G, Chiba T, Yuan J, Mochizuki-Kashio M, et al. Ezh2 augments leukemogenicity by reinforcing differentiation blockage in acute myeloid leukemia. Blood. 2012;120(5):1107–17.PubMedCrossRef Tanaka S, Miyagi S, Sashida G, Chiba T, Yuan J, Mochizuki-Kashio M, et al. Ezh2 augments leukemogenicity by reinforcing differentiation blockage in acute myeloid leukemia. Blood. 2012;120(5):1107–17.PubMedCrossRef
40.
go back to reference Neff T, Sinha AU, Kluk MJ, Zhu N, Khattab MH, Stein L, et al. Polycomb repressive complex 2 is required for MLL-AF9 leukemia. Proc Natl Acad Sci. 2012;109(13):5028–33.PubMedCrossRefPubMedCentral Neff T, Sinha AU, Kluk MJ, Zhu N, Khattab MH, Stein L, et al. Polycomb repressive complex 2 is required for MLL-AF9 leukemia. Proc Natl Acad Sci. 2012;109(13):5028–33.PubMedCrossRefPubMedCentral
41.
go back to reference Muto T, Sashida G, Oshima M, Wendt GR, Mochizuki-Kashio M, Nagata Y, et al. Concurrent loss of Ezh2 and Tet2 cooperates in the pathogenesis of myelodysplastic disorders. J Exp Med. 2013;210(12):2627–39.PubMedPubMedCentralCrossRef Muto T, Sashida G, Oshima M, Wendt GR, Mochizuki-Kashio M, Nagata Y, et al. Concurrent loss of Ezh2 and Tet2 cooperates in the pathogenesis of myelodysplastic disorders. J Exp Med. 2013;210(12):2627–39.PubMedPubMedCentralCrossRef
42.
go back to reference Sashida G, Wang C, Tomioka T, Oshima M, Aoyama K, Kanai A, et al. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J Exp Med. 2016;213(8):1459–77.PubMedPubMedCentralCrossRef Sashida G, Wang C, Tomioka T, Oshima M, Aoyama K, Kanai A, et al. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J Exp Med. 2016;213(8):1459–77.PubMedPubMedCentralCrossRef
43.
go back to reference Shimizu T, Kubovcakova L, Nienhold R, Zmajkovic J, Meyer SC, Shen HH, et al. Loss of Ezh2 synergizes with JAK2 -V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis. J Exp Med. 2016;213(8):1479–96.PubMedPubMedCentralCrossRef Shimizu T, Kubovcakova L, Nienhold R, Zmajkovic J, Meyer SC, Shen HH, et al. Loss of Ezh2 synergizes with JAK2 -V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis. J Exp Med. 2016;213(8):1479–96.PubMedPubMedCentralCrossRef
44.
go back to reference Yang Y, Akada H, Nath D, Hutchison RE, Mohi G. Loss of Ezh2 cooperates with Jak2V617F in the development of myelofibrosis in a mouse model of myeloproliferative neoplasm. Blood. 2016;127(26):3410–24.PubMedPubMedCentralCrossRef Yang Y, Akada H, Nath D, Hutchison RE, Mohi G. Loss of Ezh2 cooperates with Jak2V617F in the development of myelofibrosis in a mouse model of myeloproliferative neoplasm. Blood. 2016;127(26):3410–24.PubMedPubMedCentralCrossRef
45.
46.
go back to reference Hasegawa N, Oshima M, Sashida G, Matsui H, Koide S, Saraya A, et al. Impact of combinatorial dysfunctions of Tet2 and Ezh2 on the epigenome in the pathogenesis of myelodysplastic syndrome. Leukemia. 2017;31:861–71.PubMedCrossRef Hasegawa N, Oshima M, Sashida G, Matsui H, Koide S, Saraya A, et al. Impact of combinatorial dysfunctions of Tet2 and Ezh2 on the epigenome in the pathogenesis of myelodysplastic syndrome. Leukemia. 2017;31:861–71.PubMedCrossRef
47.
go back to reference Xu B, On DM, Ma A, Parton T, Konze KD, Pattenden SG, et al. Selective inhibition of EZH2 and EZH1 enzymatic activity by a small molecule suppresses MLL- rearranged leukemia. Blood. 2015;125(2):346–57.PubMedPubMedCentralCrossRef Xu B, On DM, Ma A, Parton T, Konze KD, Pattenden SG, et al. Selective inhibition of EZH2 and EZH1 enzymatic activity by a small molecule suppresses MLL- rearranged leukemia. Blood. 2015;125(2):346–57.PubMedPubMedCentralCrossRef
48.
go back to reference Fujita S, Honma D, Adachi N, Araki K, Takamatsu E, Katsumoto T, et al. Dual inhibition of EZH1/2 breaks the quiescence of leukemia stem cells in acute myeloid leukemia. Leukemia. 2018;32(4):855–64.PubMedCrossRef Fujita S, Honma D, Adachi N, Araki K, Takamatsu E, Katsumoto T, et al. Dual inhibition of EZH1/2 breaks the quiescence of leukemia stem cells in acute myeloid leukemia. Leukemia. 2018;32(4):855–64.PubMedCrossRef
49.
go back to reference Italiano A, Soria JC, Toulmonde M, Michot JM, Lucchesi C, Varga A, et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol. 2018;19(5):649–59.PubMedCrossRef Italiano A, Soria JC, Toulmonde M, Michot JM, Lucchesi C, Varga A, et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol. 2018;19(5):649–59.PubMedCrossRef
50.
go back to reference Guglielmelli P, Zini R, Bogani C, Salati S, Pancrazzi A, Bianchi E, et al. Molecular profiling of CD34 + cells in idiopathic myelofibrosis identifies a set of disease-associated genes and reveals the clinical significance of Wilms’ tumor gene 1 (WT1). Stem Cells. 2007;25(1):165–73.PubMedCrossRef Guglielmelli P, Zini R, Bogani C, Salati S, Pancrazzi A, Bianchi E, et al. Molecular profiling of CD34 + cells in idiopathic myelofibrosis identifies a set of disease-associated genes and reveals the clinical significance of Wilms’ tumor gene 1 (WT1). Stem Cells. 2007;25(1):165–73.PubMedCrossRef
51.
go back to reference Dutta A, Hutchison RE, Mohi G. Hmga2 promotes the development of myelofibrosis in Jak2(V617F) knockin mice by enhancing TGF-beta1 and Cxcl12 pathways. Blood. 2017;130(7):920–32.PubMedPubMedCentralCrossRef Dutta A, Hutchison RE, Mohi G. Hmga2 promotes the development of myelofibrosis in Jak2(V617F) knockin mice by enhancing TGF-beta1 and Cxcl12 pathways. Blood. 2017;130(7):920–32.PubMedPubMedCentralCrossRef
52.
go back to reference Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell. 2013;153(2):320–34.PubMedPubMedCentralCrossRef Lovén J, Hoke HA, Lin CY, Lau A, Orlando DA, Vakoc CR, et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell. 2013;153(2):320–34.PubMedPubMedCentralCrossRef
53.
go back to reference Kleppe M, Koche R, Zou L, van Galen P, Hill CE, Dong L, et al. Erratum: dual targeting of oncogenic activation and inflammatory signaling increases therapeutic efficacy in myeloproliferative neoplasms. Cancer Cell. 2018;33(4):785–7.PubMedPubMedCentralCrossRef Kleppe M, Koche R, Zou L, van Galen P, Hill CE, Dong L, et al. Erratum: dual targeting of oncogenic activation and inflammatory signaling increases therapeutic efficacy in myeloproliferative neoplasms. Cancer Cell. 2018;33(4):785–7.PubMedPubMedCentralCrossRef
54.
go back to reference De Raedt T, Beert E, Pasmant E, Luscan A, Brems H, Ortonne N, et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature. 2014;514(7521):247–51.PubMedCrossRef De Raedt T, Beert E, Pasmant E, Luscan A, Brems H, Ortonne N, et al. PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies. Nature. 2014;514(7521):247–51.PubMedCrossRef
55.
go back to reference Huang X, Yan J, Zhang M, Wang Y, Chen Y, Fu X, et al. Targeting epigenetic crosstalk as a therapeutic strategy for EZH2-aberrant solid tumors. Cell. 2018;186–99. Huang X, Yan J, Zhang M, Wang Y, Chen Y, Fu X, et al. Targeting epigenetic crosstalk as a therapeutic strategy for EZH2-aberrant solid tumors. Cell. 2018;186–99.
56.
go back to reference Figueroa ME, Skrabanek L, Li Y, Jiemjit A, Fandy TE, Paietta E, et al. MDS and secondary AML display unique patterns and abundance of aberrant DNA methylation. Blood. 2009;114(16):3448–58.PubMedPubMedCentralCrossRef Figueroa ME, Skrabanek L, Li Y, Jiemjit A, Fandy TE, Paietta E, et al. MDS and secondary AML display unique patterns and abundance of aberrant DNA methylation. Blood. 2009;114(16):3448–58.PubMedPubMedCentralCrossRef
57.
go back to reference Bejar R, Lord A, Stevenson K, Bar-Natan M, Pérez-Ladaga A, Zaneveld J, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–12.PubMedPubMedCentralCrossRef Bejar R, Lord A, Stevenson K, Bar-Natan M, Pérez-Ladaga A, Zaneveld J, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705–12.PubMedPubMedCentralCrossRef
58.
go back to reference Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, et al. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. J Clin Invest. 2015;125(5):1857–72.PubMedPubMedCentralCrossRef Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, et al. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. J Clin Invest. 2015;125(5):1857–72.PubMedPubMedCentralCrossRef
59.
go back to reference Welch JS, Petti AA, Miller CA, Fronick CC, O’Laughlin M, Fulton RS, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 2016;375(21):2023–36.PubMedPubMedCentralCrossRef Welch JS, Petti AA, Miller CA, Fronick CC, O’Laughlin M, Fulton RS, et al. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med. 2016;375(21):2023–36.PubMedPubMedCentralCrossRef
60.
go back to reference Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39(2):237–42.PubMedPubMedCentralCrossRef Ohm JE, McGarvey KM, Yu X, Cheng L, Schuebel KE, Cope L, et al. A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet. 2007;39(2):237–42.PubMedPubMedCentralCrossRef
61.
go back to reference Gebhard C, Glatz D, Schwarzfischer L, Wimmer J, Stasik S, Nuetzel M, et al. Profiling of aberrant DNA methylation in acute myeloid leukemia reveals subclasses of CG-rich regions with epigenetic or genetic association. Leukemia. 2018;1–11. Gebhard C, Glatz D, Schwarzfischer L, Wimmer J, Stasik S, Nuetzel M, et al. Profiling of aberrant DNA methylation in acute myeloid leukemia reveals subclasses of CG-rich regions with epigenetic or genetic association. Leukemia. 2018;1–11.
62.
go back to reference Wang C, Oshima M, Sato D, Matsui H, Kubota S, Aoyama K, et al. Ezh2 loss propagates hypermethylation at T cell differentiation—regulating genes to promote leukemic transformation. J Clin Invest. 2018;128(9):3872–86.PubMedPubMedCentralCrossRef Wang C, Oshima M, Sato D, Matsui H, Kubota S, Aoyama K, et al. Ezh2 loss propagates hypermethylation at T cell differentiation—regulating genes to promote leukemic transformation. J Clin Invest. 2018;128(9):3872–86.PubMedPubMedCentralCrossRef
63.
go back to reference Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, et al. Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol. 2011;29(18):2499–506.PubMedCrossRef Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, et al. Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol. 2011;29(18):2499–506.PubMedCrossRef
64.
go back to reference Inoue D, Kitaura J, Togami K, Nishimura K, Enomoto Y, Uchida T, et al. Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations. J Clin Invest. 2013;123(11):4627–40.PubMedPubMedCentralCrossRef Inoue D, Kitaura J, Togami K, Nishimura K, Enomoto Y, Uchida T, et al. Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations. J Clin Invest. 2013;123(11):4627–40.PubMedPubMedCentralCrossRef
65.
go back to reference Daou S, Barbour H, Ahmed O, Masclef L, Baril C, Sen Nkwe N, et al. Monoubiquitination of ASXLs controls the deubiquitinase activity of the tumor suppressor BAP1. Nat Commun. 2018;9(1):4385.PubMedPubMedCentralCrossRef Daou S, Barbour H, Ahmed O, Masclef L, Baril C, Sen Nkwe N, et al. Monoubiquitination of ASXLs controls the deubiquitinase activity of the tumor suppressor BAP1. Nat Commun. 2018;9(1):4385.PubMedPubMedCentralCrossRef
66.
go back to reference Balasubramani A, Larjo A, Bassein JA, Chang X, Hastie RB, Togher SM, et al. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex. Nat Commun. 2015;6:1–15.CrossRef Balasubramani A, Larjo A, Bassein JA, Chang X, Hastie RB, Togher SM, et al. Cancer-associated ASXL1 mutations may act as gain-of-function mutations of the ASXL1-BAP1 complex. Nat Commun. 2015;6:1–15.CrossRef
67.
go back to reference Asada S, Goyama S, Inoue D, Shikata S, Takeda R, Fukushima T, et al. Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis. Nat Commun. 2018;9(1):1–18.CrossRef Asada S, Goyama S, Inoue D, Shikata S, Takeda R, Fukushima T, et al. Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis. Nat Commun. 2018;9(1):1–18.CrossRef
68.
go back to reference Nagase R, Inoue D, Pastore A, Fujino T, Hou H-A, Yamasaki N, et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J Exp Med. 2018;215(6):1729–47.PubMedPubMedCentralCrossRef Nagase R, Inoue D, Pastore A, Fujino T, Hou H-A, Yamasaki N, et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J Exp Med. 2018;215(6):1729–47.PubMedPubMedCentralCrossRef
69.
go back to reference Yang H, Kurtenbach S, Guo Y, Lohse I, Durante MA, Li J, et al. Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies. Blood. 2018;131(3):328–41.PubMedPubMedCentralCrossRef Yang H, Kurtenbach S, Guo Y, Lohse I, Durante MA, Li J, et al. Gain of function of ASXL1 truncating protein in the pathogenesis of myeloid malignancies. Blood. 2018;131(3):328–41.PubMedPubMedCentralCrossRef
70.
go back to reference Grossmann V, Tiacci E, Holmes AB, Kohlmann A, Martelli MP, Kern W, et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood. 2011;118(23):6153–63.PubMedCrossRef Grossmann V, Tiacci E, Holmes AB, Kohlmann A, Martelli MP, Kern W, et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood. 2011;118(23):6153–63.PubMedCrossRef
71.
go back to reference Damm F, Chesnais V, Nagata Y, Yoshida K, Scourzic L, Okuno Y, et al. BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders. Blood. 2013;122(18):3169–77.PubMedCrossRef Damm F, Chesnais V, Nagata Y, Yoshida K, Scourzic L, Okuno Y, et al. BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders. Blood. 2013;122(18):3169–77.PubMedCrossRef
72.
go back to reference Lasho TL, Mudireddy M, Finke CM, Hanson CA, Ketterling RP, Szuber N, et al. Targeted next-generation sequencing in blast phase myeloproliferative neoplasms. Blood Adv. 2018;2(4):370–80.PubMedPubMedCentralCrossRef Lasho TL, Mudireddy M, Finke CM, Hanson CA, Ketterling RP, Szuber N, et al. Targeted next-generation sequencing in blast phase myeloproliferative neoplasms. Blood Adv. 2018;2(4):370–80.PubMedPubMedCentralCrossRef
73.
go back to reference Tanaka T, Nakajima-Takagi Y, Aoyama K, Tara S, Oshima M, Saraya A, et al. Internal deletion of BCOR reveals a tumor suppressor function for BCOR in T lymphocyte malignancies. J Exp Med. 2017;214(10):2901–13.PubMedPubMedCentralCrossRef Tanaka T, Nakajima-Takagi Y, Aoyama K, Tara S, Oshima M, Saraya A, et al. Internal deletion of BCOR reveals a tumor suppressor function for BCOR in T lymphocyte malignancies. J Exp Med. 2017;214(10):2901–13.PubMedPubMedCentralCrossRef
74.
go back to reference Andricovich J, Kai Y, Peng W, Foudi A, Tzatsos A. Histone demethylase KDM2B regulates lineage commitment in normal and malignant hematopoiesis. J Clin Invest. 2016;126(3):905–20.PubMedPubMedCentralCrossRef Andricovich J, Kai Y, Peng W, Foudi A, Tzatsos A. Histone demethylase KDM2B regulates lineage commitment in normal and malignant hematopoiesis. J Clin Invest. 2016;126(3):905–20.PubMedPubMedCentralCrossRef
75.
go back to reference He J, Nguyen AT, Zhang Y. KDM2b/JHDM1b, an H3K36me2-specific demethylase, is required for initiation and maintenance of acute myeloid leukemia. Blood. 2011;117(14):3869–80.PubMedPubMedCentralCrossRef He J, Nguyen AT, Zhang Y. KDM2b/JHDM1b, an H3K36me2-specific demethylase, is required for initiation and maintenance of acute myeloid leukemia. Blood. 2011;117(14):3869–80.PubMedPubMedCentralCrossRef
76.
go back to reference van den Boom V, Maat H, Geugien M, Rodríguez López A, Sotoca AM, Jaques J, et al. Non-canonical PRC1.1 targets active genes independent of H3K27me3 and is essential for leukemogenesis. Cell Rep. 2016;14(2):332–46.PubMedCrossRef van den Boom V, Maat H, Geugien M, Rodríguez López A, Sotoca AM, Jaques J, et al. Non-canonical PRC1.1 targets active genes independent of H3K27me3 and is essential for leukemogenesis. Cell Rep. 2016;14(2):332–46.PubMedCrossRef
Metadata
Title
Deregulated Polycomb functions in myeloproliferative neoplasms
Authors
Goro Sashida
Motohiko Oshima
Atsushi Iwama
Publication date
01-08-2019
Publisher
Springer Japan
Published in
International Journal of Hematology / Issue 2/2019
Print ISSN: 0925-5710
Electronic ISSN: 1865-3774
DOI
https://doi.org/10.1007/s12185-019-02600-6

Other articles of this Issue 2/2019

International Journal of Hematology 2/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine