Next Article in Journal
CaMKII Inhibition Attenuates Distinct Gain-of-Function Effects Produced by Mutant Nav1.6 Channels and Reduces Neuronal Excitability
Next Article in Special Issue
Impact of Molecular Biology in Diagnosis, Prognosis, and Therapeutic Management of BCR::ABL1-Negative Myeloproliferative Neoplasm
Previous Article in Journal
Selective Ablation of BCL11A in Epidermal Keratinocytes Alters Skin Homeostasis and Accelerates Excisional Wound Healing In Vivo
 
 
Correction published on 19 December 2022, see Cells 2022, 11(24), 4126.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis

Department of Medicine, LKS Faculty of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
Cells 2022, 11(13), 2107; https://doi.org/10.3390/cells11132107
Submission received: 11 June 2022 / Revised: 28 June 2022 / Accepted: 2 July 2022 / Published: 3 July 2022 / Corrected: 19 December 2022
(This article belongs to the Special Issue The Myeloid Diseases from the Biology to the Clinic)

Abstract

:
Myelofibrosis (MF) is the most symptomatic form of myeloproliferative neoplasm and carries the worst outcome. Allogeneic hematopoietic stem cell transplantation is the only therapy with potential for cure at present, but is limited by significant mortality and morbidity. JAK inhibition is the mainstay of treatment for intermediate- and high-risk MF. Ruxolitinib is the most widely used JAK1/2 inhibitor and provides durable effects in controlling symptom burden and spleen volumes. Nevertheless, ruxolitinib may not adequately address the underlying disease biology. Its effects on mutant allele burden, bone marrow fibrosis, and the prevention of leukemic transformation are minimal. Multiple small molecules are being tested in multiple phase 2 and 3 studies as either monotherapy or in combination with JAK2 inhibitors. In this review, the role of LSD1/KDM1A inhibition as a potential disease-modification strategy in patients with myelofibrosis is described and discussed.

Graphical Abstract

1. Introduction

Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in hematopoiesis [1]. They are characterized by stem cell-derived clonal myeloproliferation. The classical Philadelphia (Ph) chromosome-negative MPNs comprise polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), and are associated with the driver genes JAK2, CALR, and MPL. PMF is characterized by reticulin fibrosis, abnormal cytokine-mediated systemic symptoms, anemia, hepatosplenomegaly, and a propensity for progression to AML. Patients with PV and ET may progress to post-PV (PPV) and post-ET (PET)—myelofibrosis (MF), respectively. The incidence of PV, ET, and PMF is approximately 0.4 to 2.8, 0.38 to 1.7, and 0.1 to 1.0 per 100,000 persons per year, respectively [2]. MPNs generally affect the middle-aged, with the median age at presentation of PV, ET, and PMF being 65, 68, and 70 years, respectively [3]. Patients with MF carry the worst prognosis [4,5]. Median survival for PV, ET, and PMF is 14, 20, and 5.7 years respectively [6,7,8]. Allogeneic hematopoietic stem cell transplantation is the only therapy with potential for cure at present, but is limited by significant mortality and morbidity [9]. Thus, JAK inhibition is the cornerstone of treatment for intermediate- and high-risk MF. Ruxolitinib is the most widely used JAK1/2 inhibitor, provides durable effects in controlling patient symptoms and spleen volumes, and may prolong survival [10,11,12,13,14,15,16]. Nevertheless, the effect of ruxolitinib on mutant allele burden, bone marrow fibrosis, and the prevention of leukemic transformation has been little observed. Even the three other JAK2 inhibitors—fedratinib, pacritinib, and momelotinib—may not address all the unmet needs in patients with MF, especially in the second-line setting and the setting of disease modification [17,18,19,20]. Multiple “non-JAK inhibitor” molecules are being tested in phase 2 and 3 studies, either as monotherapy or in combination with JAK2 inhibitors [20,21]. In this review, the role of LSD1/KDM1A inhibition as a potential disease-modification strategy in patients with myelofibrosis is appraised.

2. Mutations in Epigenetic Regulators in Myelofibrosis

With the advent of next-generation sequencing and whole-genome analyses in myeloid malignancies, mutations in DNA methylation genes (TET2, DNMT3A, IDH1/2), histone modification genes (EZH2, ASXL1), RNA splicing factors (SRSF2, SF3B1, U2AF1, ZRSR2), and transcription factors (TP53, CUX1, IKZF1, ETV6, RUNX1) have been described in MPNs [22,23,24]. Nevertheless, these mutations are not restricted to MPNs, and are also seen in myelodysplastic syndrome (MDS), AML, and other myeloid malignancies. These mutations are involved in the phenotypic and disease evolution of MPNs. TET2 (10–20% of MPNs) and DNMT3A (5–10% of MPNs) have been found to precede JAK2V617F mutations, and have a central role in self-renewal and disease initiation in MPN hematopoietic stem cells (HSCs) [25,26,27,28,29,30,31,32]. The role of DNMT3A and TET2 in the progression to MF or secondary AML is yet to be elucidated. EZH2 mutations are seen in 5% to 10% of PMF and portend a poor prognosis [33]. In murine MPN models, Ezh2 loss modifies phenotype and is associated with disease progression [34,35,36]. Mutations in ASXL are seen in 25% of PMF and are associated with worse outcomes [37,38,39]. Loss-of-function mutations in ASXL1 are associated with a higher risk of leukemic transformation. In murine models, Asxl1 loss is associated with dysplasia, cytopenia, and defective HSC self-renewal [40,41]. SRSF2 mutations are mostly restricted to ET and PMF and associated with poor outcomes [42]. They are especially enriched in patients with secondary AML from preceding MF. TP53 mutations are uncommon in PMF or other MPNs in the chronic phase. However, they are found in up to 50% of patients with secondary AML and are often associated with ASXL1, SRSF2, IDH1/2, CBL, and LNK mutations [43,44]. They are especially common in secondary AML from preceding post-PV or post-ET MF and are associated with DNMT3A mutations. Other mutations associated with late events in the clonal progression of MPNs and secondary AML include RUNX1, FLT3-ITD, NRAS, NF1, IKZF1 and CUX1 [45,46]. Mutations in epigenetic regulators and transcription factors are often associated with advanced MF and increased risk of progression [27,28,47,48,49]. Manipulation of these genetic alterations may offer an additional therapeutic option in patients with an otherwise dismal outcome, and targeting epigenetic regulators with novel agents may potentially alter disease biology in MF (Figure 1).

3. The Functional Role of LSD1 in Hematopoiesis

LSD1 is an essential gene, the loss of which leads to early embryonic lethality [50,51]. The protein also regulates the balance between self-renewal and proliferation [52]. Conditional in vivo LSD1 knockdown using a doxycycline-inducible short hairpin LSD1 (shLSD1) established LSD1 as a central regulator of HSPCs [53]. An inducible LSD1 knockdown resulted in profound but reversible thrombocytopenia, neutropenia, and anemia with concurrent monocytosis. LSD1 knockdown for 27 days led to an increase in circulating multipotent progenitors and HSCs with a concomitant downregulation of chemokine (C-X-C motif) receptor 4 (CXCR4) without affecting the size of the quiescent long-term hematopoietic stem cell (HSC) pool [53].
LSD1 is a key regulator of the progression from pluripotency to terminal differentiation and balancing self-renewal and proliferation [52,54]. LSD1 is recruited to promoters and enhancers of genes essential for normal development by the transcription factors octamer-binding transcription factor 4 (OCT4), SRY (sex determining region Y)-box 2 (SOX2), Nanog, and the coactivator mediator. LSD1 maintains the pluripotency program allowing embryonic stem cells (ESCs) to differentiate. LSD1 is also essential for the complete shutdown of ESC gene expression, as cells undergo transition to more differentiated cell states [54]. LSD1 plays a similar role during myelopoiesis, allowing commitment of progenitors to specific myeloid lineages [55]. Enhancers essential for terminal myeloid differentiation in lineage-specific progenitor cells are activated by the placement of H3K4me1 marks. As progenitors commit to differentiation, LSD1 is significantly downregulated, allowing enhancers and promoters to be gradually activated with progressive addition of methyl or acetyl groups to H3K4 and H3K27, respectively [55].

4. LSD1 as an Epigenetic Regulator

Lysine methyltransferases and demethylases are able to catalyze N-methylation and N-demthylation of histone (H) lysines (K) [56,57]. LSD1, or KDM1A, is an enzyme that removes mono- and dimethyl groups from the histone H3 at the critical lysines K4 and K9 [58]. Methylation of histone H3K4 and H3K9 is a posttranslational modification that results in conformational change of chromatins [59,60]. Chromatins are a constellation of nuclear macromolecules and comprise DNA, protein scaffolding, and enzymes that drive RNA transcription and synthesis [61]. The DNA and its protein scaffold of histones form the nucleosome. Each nucleosome comprises two copies of each of the four histone proteins—H2A, H2B, H3, and H4—forming an octamer around which DNA is wrapped. The rates of gene transcription are heavily influenced by the accessibility of transcription factors and RNA polymerase complexes to template DNA at promoters and enhancers [59,60].
Histone and nucleic acid modifications provide binding sites for proteins and components of the transcriptional machinery that affect transcriptional gene silencing or activation. Histone modifications include acetylation (Ac), methylation (Me), phosphorylation (Ph), and ubiquitination (Ub). LSD1 acts as an epigenetic regulator of gene expression by altering the local state of the chromatin. Inhibition of LSD1 results in alteration of gene expression and inhibits the maturation of JAK-STAT activated megakaryocytes and myeloid cells from their progenitors. This also results in the inhibition of self-renewal potential of HSPCs harboring the pathogenic driver mutations [62,63,64].
LSD also regulates nonhistone substrates [65]. LSD1 is localized to specific sites in the genome through various transcription factors that bind DNA [54,66]. Transcription activators, such as V-Myb avian myeloblastosis viral oncogene homolog (MYB) and steroid hormone receptors, as well as repressors, such as growth factor independence 1 transcription repressor (GFI1) and RE-1 silencing transcription factor (REST), recruit LSD1 to specific locations on the genome [67,68]. LSD1 is part of a larger protein complex, containing Co-RE-1 silencing transcription factor (CoREST), nucleosome remodeling and histone deacetylase (NuRD), or other factors that determine cell- and site-specific chromatin remodeling [51,69]. These complexes may also include DNA methyltransferase 1 (DNMT1) and histone deacetylases 1, 2, and 3 (HDAC1, 2, and 3) activities, all of which contribute to maintaining or modifying the epigenetic state at that genomic site [70,71]. Therefore, an important property of LSD1 is its function as a scaffold for other proteins and epigenetic enzymes that are corecruited to genomic sites. LSD1 bound to specific sites precludes the binding of other factors that may influence transcription.

5. The Biological Role of LSD1 and the Effect of LSD1 Inhibitors in Murine Models of MPNs

Overexpression of LSD1 messenger RNA (mRNA) and excess LSD1 protein have been observed in various malignancies, including neuroblastoma, squamous-cell carcinoma, Ewing sarcoma, AML, neuroendocrine tumors, breast cancer, prostate cancer, bladder cancer, small-cell lung cancer, and colorectal cancer [63,64,67,72,73,74]. In MPNs, LSD1 is overexpressed, mainly in the megakaryocytes, erythroid precursors, and to a certain extent in in the early myeloid series [75]. Treatment of various malignant cell types in vitro with LSD1 inhibitors suppresses tumor growth, reduces their invasiveness, reduces clonogenic potential, eliminates cancer stem cells, induces markers of differentiation appropriate to the cell lineage, and induces apoptosis [76,77,78]. In various models of mouse leukemia, treatment with LSD1 inhibitors induced monocytic markers of differentiation, reduced clonogenic potential of leukemia-initiating cells (LICs), and induced apoptosis [78]. LSD1 is expressed in a high proportion of leukemic myeloblasts [79,80]. LSD1 gene expression is among the highest in the malignant myeloid stem and progenitor cell population [77,78]. LSD1 plays a direct role in regulating pathogenic JAK-STAT signaling pathways. The key MPN driver genes JAK2V617F, CALR, and MPL activate JAK-STAT signaling via phosphorylation of STAT3, STAT5, and transcription factors, which activate specific genes with pleiotropic effects [81]. STAT3 activity is modulated by methylation on lysine (K140) and is one of the substrates for LSD1 [82]. Proof-of-concept studies have been performed on well-established, preclinical mouse models of MPNs (JAK2V617F, MplW515L). LSD1 inhibition in MplW515L mice markedly suppressed myeloproliferation, reducing leukocyte and platelet counts. Spleens in animals treated with LSDi showed a dose-proportional decrease in weight. Histologically, a marked reduction in myeloid proliferation was demonstrated in the bone marrow and the spleen alongside a reversal of extramedullary hematopoiesis (EMH). Intriguingly, there was marked reduction in the degree of bone marrow reticulin fibrosis with LSD1 inhibition. LSD1 inhibition also significantly reduced serum inflammatory cytokine concentrations, in particular the plasma concentration of the chemokine (C-X-C motif) ligand 5 (Cxcl5 or IL-8 in humans), a key mediator of the inflammatory state seen in MPNs. In these mouse models, a reduction in mutant allele burden of driver mutations was also demonstrated. The observation of reduction in the degree of marrow fibrosis and the mutant allele burden of driver genes support the proposition that targeting LSD1 may induce disease modification in MPNs.

6. Clinical Data of LSD1 Inhibitors in Myelofibrosis

Given the important roles that LSD1 plays in carcinogenesis, various LSD1 inhibitors have been evaluated in clinical trials. Some of the reported LSD1 inhibitors include tranylcypromine (TCP or PCPA), ORY-1001 (iadademstat), GSK-2879552, IMG-7289 (bomedemstat), INCB059872, CC-90011, and ORY-2001 (vafidemstat). Bomedemstat (IMG-7289, Imago Biosciences, San Francisco, CA) is the most extensively evaluated LSD1 inhibitor in myeloproliferative neoplasms. In JAK2V617F-postive MPN mice, daily dosing improved blood counts, reduced spleen volumes, reduced marrow reticulin fibrosis, and reduced mutant allele frequencies [83].
Bomedemstat is the only LSD1 inhibitor clinically evaluated in patients with advanced myelofibrosis. In an ongoing phase 2 study in 89 patients with advanced MF, the efficacy and safety of bomedemstat was confirmed [84,85]. Eighty-three percent of the patients had a history of treatment failure with ruxolitinib, with 70% also receiving a second treatment with an unfavorable experience. Thirty-seven percent of patients had received at least one red cell transfusion prior to dosing. The most frequently reported adverse event was thrombocytopenia, an expected observation, given that dosing to grade 3 thrombocytopenia was allowed. The commonest nonhematological toxicity was dysgeusia, reported in 33%, with one discontinuation. Other serious adverse events (SAEs) were reported in 49% of the patients. The most common SAEs, regardless of causality, were cellulitis, diverticulitis, and pneumonia. There were no deaths related to the study drug. The efficacy endpoints were reduction in spleen volume and reduction in total symptom scores (TSS) using the MPN symptom assessment form (SAF). At 24 weeks, 64% of patients had a decrease in spleen volume, with 6% of patients having more than 35% reduction. In patients with a high symptom burden (TSS > 20), 65% had a decrease in TSS, and for 19% of patients, the decrease in TSS was greater than 50%. By week 12, 44% of patients had an increase in hemoglobin of 1 g/dL or more and 46% had stable hemoglobin. At the data cutoff point, of 21 patients, three had become transfusion-independent. The improvement in hemoglobin is an intriguing observation and is likely to address the issue of anemia associated with ruxolitinib. A possible explanation for this observation is that the modulation of transcription by LSD1 is lineage-specific or that expression of the γ-globin gene is altered with LSD1 inhibition [86,87]. Eighty-five percent of patients had stable or improved fibrosis score of at least one grade. Patients with an elevated inflammatory chemokine, such as chemokine ligand 5 (CCL5), had a measurable decrease into normal concentrations. The mutant allele frequencies (MAF) of driver and nondriver mutations among the 261 genes were serially sequenced. Fifty-two percent of patients had a decrease in MAF, with ASXL1 being the most sensitive to bomedemstat. No patient progressed to secondary AML in this study.

7. Conclusions and Future Perspectives

As we progress to the era of novel therapies that could alter disease biology, there is a need to utilize end points that inform us how these novel agents could alter the disease trajectory (Table 1). The current data available from clinical studies support the definition of disease modification that comprises clinically meaningful improvement in survival, reduction in the risk of leukemic transformation, restoration of normal hematopoiesis, significant reduction in bone marrow fibrosis, and reduction in the clonal burden of the disease. Achieving disease modification will ultimately lead to beneficial effects in traditional outcome measures, such as symptom improvement and control of splenomegaly. In spite of the limited disease-modifying effect of JAK inhibitors, the JAK-STAT pathway remains pivotal. This is supported by the modest effect of single-agent bomedemstat on spleen volume. Treatment strategies combining JAK inhibitors and novel agents, such as bomedemstat, will provide synergic effects in improving outcomes and altering disease biology. The selection of JAK inhibitors to be used with bomedemstat should take into account the disease characteristics and clinical needs, such as the presence of anemia (where fedratinib or momelotinib could be considered) or thrombocytopenia (where pacritinib could be considered). Based on the effect of LSD1 inhibition on erythropoiesis, it will also be intriguing to observe if bomedemstat can circumvent anemia caused by ruxolitinib. In addition, the impressive responses seen with bomedemstat in patients with essential thrombocythemia [88] suggest that LSD1 inhibitors could potentially be beneficial when used earlier in MF, such as in prefibrotic or early PMF, an area that is yet to be explored.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author would like to acknowledge Hugh Young Rienhoff and his team at Imago Biosciences for their work on the role of LSD1 and the development of IMG-7289 (Bomedemstat), which inspired this manuscript.

Conflicts of Interest

The author declares no relevant conflict of interest.

References

  1. Spivak, J.L. Myeloproliferative Neoplasms. N. Engl. J. Med. 2017, 376, 2168–2181. [Google Scholar] [CrossRef] [PubMed]
  2. Moulard, O.; Mehta, J.; Fryzek, J.; Olivares, R.; Iqbal, U.; Mesa, R.A. Epidemiology of myelofibrosis, essential thrombocythemia, and polycythemia vera in the European Union. Eur. J. Haematol. 2014, 92, 289–297. [Google Scholar] [CrossRef] [PubMed]
  3. Srour, S.A.; Devesa, S.S.; Morton, L.M.; Check, D.P.; Curtis, R.E.; Linet, M.S.; Dores, G.M. Incidence and patient survival of myeloproliferative neoplasms and myelodysplastic/myeloproliferative neoplasms in the United States, 2001-12. Br. J. Haematol. 2016, 174, 382–396. [Google Scholar] [CrossRef] [PubMed]
  4. Hultcrantz, M.; Kristinsson, S.Y.; Andersson, T.M.-L.; Landgren, O.; Eloranta, S.; Derolf, A.R.; Dickman, P.W.; Björkholm, M. Patterns of Survival Among Patients With Myeloproliferative Neoplasms Diagnosed in Sweden From 1973 to 2008: A Population-Based Study. J. Clin. Oncol. 2012, 30, 2995–3001. [Google Scholar] [CrossRef]
  5. Tefferi, A.; Guglielmelli, P.; Larson, D.R.; Finke, C.; Wassie, E.A.; Pieri, L.; Gangat, N.; Fjerza, R.; Belachew, A.A.; Lasho, T.L.; et al. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood 2014, 124, 2507–2513. [Google Scholar] [CrossRef]
  6. Vannucchi, A.M.; Harrison, C.N. Emerging treatments for classical myeloproliferative neoplasms. Blood 2017, 129, 693–703. [Google Scholar] [CrossRef]
  7. Tefferi, A. Primary myelofibrosis: 2019 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2018, 93, 1551–1560. [Google Scholar] [CrossRef]
  8. Tefferi, A.; Barbui, T. Polycythemia vera and essential thrombocythemia: 2019 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2019, 94, 133–143. [Google Scholar] [CrossRef]
  9. England, J.; Gupta, V. Novel therapies vs hematopoietic cell transplantation in myelofibrosis: Who, when, how? Hematol. Am. Soc. Hematol. Educ. Program 2021, 2021, 453–462. [Google Scholar] [CrossRef]
  10. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Levy, R.S.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.; Miller, C.; Silver, R.T.; et al. A Double-Blind, Placebo-Controlled Trial of Ruxolitinib for Myelofibrosis. N. Engl. J. Med. 2012, 366, 799–807. [Google Scholar] [CrossRef]
  11. Harrison, C.; Kiladjian, J.-J.; Al-Ali, H.K.; Gisslinger, H.; Waltzman, R.; Stalbovskaya, V.; McQuitty, M.; Hunter, D.S.; Levy, R.; Knoops, L.; et al. JAK Inhibition with Ruxolitinib versus Best Available Therapy for Myelofibrosis. N. Engl. J. Med. 2012, 366, 787–798. [Google Scholar] [CrossRef] [PubMed]
  12. Mesa, R.A.; Gotlib, J.; Gupta, V.; Catalano, J.V.; Deininger, M.W.; Shields, A.L.; Miller, C.B.; Silver, R.T.; Talpaz, M.; Winton, E.F.; et al. Effect of Ruxolitinib Therapy on Myelofibrosis-Related Symptoms and Other Patient-Reported Outcomes in COMFORT-I: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Clin. Oncol. 2013, 31, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
  13. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Levy, R.S.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.; Miller, C.; Silver, R.T.; et al. The clinical benefit of ruxolitinib across patient subgroups: Analysis of a placebo-controlled, Phase III study in patients with myelofibrosis. Br. J. Haematol. 2013, 161, 508–516. [Google Scholar] [CrossRef] [PubMed]
  14. Verstovsek, S.; Gotlib, J.; Mesa, R.A.; Vannucchi, A.M.; Kiladjian, J.-J.; Cervantes, F.; Harrison, C.N.; Paquette, R.; Sun, W.; Naim, A.; et al. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J. Hematol. Oncol. 2017, 10, 156. [Google Scholar] [CrossRef]
  15. Guglielmelli, P.; Ghirardi, A.; Carobbio, A.; Masciulli, A.; Maccari, C.; Mora, B.; Rumi, E.; Triguero, A.; Finazzi, M.C.; Pettersson, H.; et al. Impact of ruxolitinib on survival of patients with myelofibrosis in the real world: Update of the ERNEST Study. Blood Adv. 2022, 6, 373–375. [Google Scholar] [CrossRef]
  16. Harrison, C.N.; on behalf of the COMFORT-II Investigators; Vannucchi, A.M.; Kiladjian, J.-J.; Al-Ali, H.K.; Gisslinger, H.; Knoops, L.; Cervantes, F.; Jones, M.M.; Sun, K.; et al. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia 2016, 30, 1701–1707. [Google Scholar] [CrossRef]
  17. Harrison, C.N.; Schaap, N.; Vannucchi, A.M.; Kiladjian, J.-J.; Tiu, R.V.; Zachee, P.; Jourdan, E.; Winton, E.; Silver, R.T.; Schouten, H.C.; et al. Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): A single-arm, open-label, non-randomised, phase 2, multicentre study. Lancet Haematol. 2017, 4, e317–e324. [Google Scholar] [CrossRef]
  18. Mascarenhas, J.; Hoffman, R.; Talpaz, M.; Gerds, A.T.; Stein, B.; Gupta, V.; Szoke, A.; Drummond, M.; Pristupa, A.; Granston, T.; et al. Pacritinib vs Best Available Therapy, Including Ruxolitinib, in Patients With Myelofibrosis: A Randomized Clinical Trial. JAMA Oncol. 2018, 4, 652–659. [Google Scholar] [CrossRef]
  19. Chifotides, H.T.; Bose, P.; Verstovsek, S. Momelotinib: An emerging treatment for myelofibrosis patients with anemia. J. Hematol. Oncol. 2022, 15, 1–17. [Google Scholar] [CrossRef]
  20. Vachhani, P.; Verstovsek, S.; Bose, P. Disease Modification in Myelofibrosis: An Elusive Goal? J. Clin. Oncol. 2022, 40, 1147–1154. [Google Scholar] [CrossRef]
  21. Tremblay, D.; Hoffman, R. Emerging drugs for the treatment of myelofibrosis: Phase II & III clinical trials. Expert Opin. Emerg. Drugs 2021, 26, 351–362. [Google Scholar] [CrossRef] [PubMed]
  22. Vainchenker, W.; Kralovics, R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood 2017, 129, 667–679. [Google Scholar] [CrossRef] [PubMed]
  23. Gill, H.; Leung, A.Y.; Kwong, Y.-L. Molecular and Cellular Mechanisms of Myelodysplastic Syndrome: Implications on Targeted Therapy. Int. J. Mol. Sci. 2016, 17, 440. [Google Scholar] [CrossRef] [PubMed]
  24. Gill, H.; Leung, A.Y.; Kwong, Y.-L. Molecularly targeted therapy in acute myeloid leukemia. Futur. Oncol. 2016, 12, 827–838. [Google Scholar] [CrossRef]
  25. Delhommeau, F.; Dupont, S.; Della Valle, V.; James, C.; Trannoy, S.; Massé, A.; Kosmider, O.; Le Couedic, J.-P.; Robert, F.; Alberdi, A.; et al. Mutation inTET2in Myeloid Cancers. N. Engl. J. Med. 2009, 360, 2289–2301. [Google Scholar] [CrossRef]
  26. Tefferi, A.; Pardanani, A.; Lim, K.-H.; Abdel-Wahab, O.; Lasho, T.L.; Patel, J.; Gangat, N.; Finke, C.M.; Schwager, S.; Mullally, A.; et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009, 23, 905–911. [Google Scholar] [CrossRef]
  27. Ortmann, C.A.; Kent, D.G.; Nangalia, J.; Silber, Y.; Wedge, D.C.; Grinfeld, J.; Baxter, E.J.; Massie, C.E.; Papaemmanuil, E.; Menon, S.; et al. Effect of Mutation Order on Myeloproliferative Neoplasms. N. Engl. J. Med. 2015, 372, 601–612. [Google Scholar] [CrossRef]
  28. Kent, D.G.; Ortmann, C.A.; Green, A.R.; Delhommeau, F. Effect of Mutation Order on Myeloproliferative Neoplasms. N. Engl. J. Med. 2015, 372, 1865–1866. [Google Scholar] [CrossRef]
  29. Stegelmann, F.; Bullinger, L.; Schlenk, R.F.; Paschka, P.; Griesshammer, M.; Blersch, C.; Kuhn, S.; Schauer, S.; Dohner, H.; Dohner, K. DNMT3A mutations in myeloproliferative neoplasms. Leukemia 2011, 25, 1217–1219. [Google Scholar] [CrossRef]
  30. Challen, G.A.; Sun, D.; Jeong, M.; Luo, M.; Jelinek, J.; Berg, J.S.; Bock, C.; Vasanthakumar, A.; Gu, H.; Xi, Y.; et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat. Genet. 2011, 44, 23–31. [Google Scholar] [CrossRef]
  31. Jaiswal, S.; Fontanillas, P.; Flannick, J.; Manning, A.; Grauman, P.V.; Mar, B.G.; Lindsley, R.C.; Mermel, C.H.; Burtt, N.; Chavez, A.; et al. Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes. N. Engl. J. Med. 2014, 371, 2488–2498. [Google Scholar] [CrossRef] [PubMed]
  32. Xie, M.; Lu, C.; Wang, J.; McLellan, M.D.; Johnson, K.J.; Wendl, M.C.; McMichael, J.F.; Schmidt, H.K.; Yellapantula, V.; Miller, C.A.; et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 2014, 20, 1472–1478. [Google Scholar] [CrossRef] [PubMed]
  33. Guglielmelli, P.; Biamonte, F.; Score, J.; Hidalgo-Curtis, C.; Cervantes, F.; Maffioli, M.; Fanelli, T.; Ernst, T.; Winkelman, N.; Jones, A.V.; et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood 2011, 118, 5227–5234. [Google Scholar] [CrossRef]
  34. Yang, Y.; Akada, H.; Nath, D.; Hutchison, R.E.; Mohi, G. Loss of Ezh2 cooperates with Jak2V617F in the development of myelofibrosis in a mouse model of myeloproliferative neoplasm. Blood 2016, 127, 3410–3423. [Google Scholar] [CrossRef] [PubMed]
  35. Shimizu, T.; Kubovcakova, L.; Nienhold, R.; Zmajkovic, J.; Meyer, S.C.; Hao-Shen, H.; Geier, F.; Dirnhofer, S.; Guglielmelli, P.; Vannucchi, A.M.; et al. Loss of Ezh2 synergizes with JAK2-V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis. J. Exp. Med. 2016, 213, 1479–1496. [Google Scholar] [CrossRef]
  36. Sashida, G.; Wang, C.; Tomioka, T.; Oshima, M.; Aoyama, K.; Kanai, A.; Mochizuki-Kashio, M.; Harada, H.; Shimoda, K.; Iwama, A. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J. Exp. Med. 2016, 213, 1459–1477. [Google Scholar] [CrossRef]
  37. Carbuccia, N.; Murati, A.; Trouplin, V.; Brecqueville, M.; Adelaide, J.; Rey, J.; Vainchenker, W.; Bernard, O.A.; Chaffanet, M.; Vey, N.; et al. Mutations of ASXL1 gene in myeloproliferative neoplasms. Leukemia 2009, 23, 2183–2186. [Google Scholar] [CrossRef]
  38. Vannucchi, A.M.; Lasho, T.L.; Guglielmelli, P.; Biamonte, F.; Pardanani, A.; Pereira, A.; Finke, C.; Score, J.; Gangat, N.; Mannarelli, C.; et al. Mutations and prognosis in primary myelofibrosis. Leukemia 2013, 27, 1861–1869. [Google Scholar] [CrossRef]
  39. Gelsi-Boyer, V.; Trouplin, V.; Adélaïde, J.; Bonansea, J.; Cervera, N.; Carbuccia, N.; Lagarde, A.; Prebet, T.; Nezri, M.; Sainty, D.; et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br. J. Haematol. 2009, 145, 788–800. [Google Scholar] [CrossRef]
  40. Abdel-Wahab, O.; Gao, J.; Adli, M.; Dey, A.; Trimarchi, T.; Chung, Y.R.; Kuscu, C.; Hricik, T.; Ndiaye-Lobry, D.; LaFave, L.M.; et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J. Exp. Med. 2013, 210, 2641–2659. [Google Scholar] [CrossRef]
  41. Inoue, D.; Kitaura, J.; Togami, K.; Nishimura, K.; Enomoto, Y.; Uchida, T.; Kagiyama, Y.; Kawabata, K.C.; Nakahara, F.; Izawa, K.; et al. Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations. J. Clin. Investig. 2013, 123, 4627–4640. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, S.-J.; Rampal, R.; Manshouri, T.; Patel, J.; Mensah, N.; Kayserian, A.; Hricik, T.; Heguy, A.; Hedvat, C.; Gönen, M.; et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood 2012, 119, 4480–4485. [Google Scholar] [CrossRef] [PubMed]
  43. Harutyunyan, A.; Klampfl, T.; Cazzola, M.; Kralovics, R. p53 Lesions in Leukemic Transformation. N. Engl. J. Med. 2011, 364, 488–490. [Google Scholar] [CrossRef]
  44. Wong, T.N.; Ramsingh, G.; Young, A.L.; Miller, C.A.; Touma, W.; Welch, J.S.; Lamprecht, T.L.; Shen, D.; Hundal, J.; Fulton, R.S.; et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature 2015, 518, 552–555. [Google Scholar] [CrossRef]
  45. Courtier, F.; Carbuccia, N.; Garnier, S.; Guille, A.; Adélaïde, J.; Cervera, N.; Gelsi-Boyer, V.; Mozziconacci, M.-J.; Rey, J.; Vey, N.; et al. Genomic analysis of myeloproliferative neoplasms in chronic and acute phases. Haematologica 2016, 102, e11–e14. [Google Scholar] [CrossRef] [PubMed]
  46. Milosevic, J.D.; Puda, A.; Malcovati, L.; Berg, T.; Hofbauer, M.; Stukalov, A.; Klampfl, T.; Harutyunyan, A.S.; Gisslinger, H.; Gisslinger, B.; et al. Clinical significance of genetic aberrations in secondary acute myeloid leukemia. Am. J. Hematol. 2012, 87, 1010–1016. [Google Scholar] [CrossRef] [PubMed]
  47. Tefferi, A.; Guglielmelli, P.; Lasho, T.L.; Gangat, N.; Ketterling, R.P.; Pardanani, A.; Vannucchi, A.M. MIPSS70+ Version 2.0: Mutation and Karyotype-Enhanced International Prognostic Scoring System for Primary Myelofibrosis. J. Clin. Oncol. 2018, 36, 1769–1770. [Google Scholar] [CrossRef] [PubMed]
  48. Tefferi, A.; Guglielmelli, P.; Nicolosi, M.; Mannelli, F.; Mudireddy, M.; Bartalucci, N.; Finke, C.M.; Lasho, T.L.; Hanson, C.A.; Ketterling, R.P.; et al. GIPSS: Genetically inspired prognostic scoring system for primary myelofibrosis. Leukemia 2018, 32, 1631–1642. [Google Scholar] [CrossRef]
  49. Grinfeld, J.; Nangalia, J.; Baxter, E.J.; Wedge, D.C.; Angelopoulos, N.; Cantrill, R.; Godfrey, A.L.; Papaemmanuil, E.; Gundem, G.; MacLean, C.; et al. Classification and Personalized Prognosis in Myeloproliferative Neoplasms. N. Engl. J. Med. 2018, 379, 1416–1430. [Google Scholar] [CrossRef]
  50. Wang, J.; Hevi, S.; Kurash, J.K.; Lei, H.; Gay, F.; Bajko, J.; Su, H.; Sun, W.; Chang, H.; Xu, G.; et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet. 2009, 41, 125–129. [Google Scholar] [CrossRef]
  51. Foster, C.T.; Dovey, O.M.; Lezina, L.; Luo, J.L.; Gant, T.W.; Barlev, N.; Bradley, A.; Cowley, S.M. Lysine-Specific Demethylase 1 Regulates the Embryonic Transcriptome and CoREST Stability. Mol. Cell. Biol. 2010, 30, 4851–4863. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, J.; Scully, K.; Zhu, X.; Cai, L.; Zhang, J.; Prefontaine, G.G.; Krones, A.; Ohgi, K.A.; Zhu, P.; Garcia-Bassets, I.; et al. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 2007, 446, 882–887. [Google Scholar] [CrossRef]
  53. Sprüssel, A.; Schulte, J.H.; Weber, S.; Necke, M.; Händschke, K.; Thor, T.; Pajtler, K.W.; Schramm, A.; König, K.; Diehl, L.; et al. Lysine-specific demethylase 1 restricts hematopoietic progenitor proliferation and is essential for terminal differentiation. Leukemia 2012, 26, 2039–2051. [Google Scholar] [CrossRef] [PubMed]
  54. Whyte, W.A.; Bilodeau, S.; Orlando, D.A.; Hoke, H.A.; Frampton, G.M.; Foster, C.T.; Cowley, S.M.; Young, R.A. Enhancer decommissioning by LSD1 during embryonic stem cell differentiation. Nature 2012, 482, 221–225. [Google Scholar] [CrossRef] [PubMed]
  55. Lara-Astiaso, D.; Weiner, A.; Lorenzo-Vivas, E.; Zaretsky, I.; Jaitin, D.A.; David, E.; Keren-Shaul, H.; Mildner, A.; Winter, D.; Jung, S.; et al. Chromatin state dynamics during blood formation. Science 2014, 345, 943–949. [Google Scholar] [CrossRef]
  56. Kaniskan, H.U.; Martini, M.L.; Jin, J. Inhibitors of Protein Methyltransferases and Demethylases. Chem. Rev. 2018, 118, 989–1068. [Google Scholar] [CrossRef]
  57. Hoffmann, I.; Roatsch, M.; Schmitt, M.L.; Carlino, L.; Pippel, M.; Sippl, W.; Jung, M. The role of histone demethylases in cancer therapy. Mol. Oncol. 2012, 6, 683–703. [Google Scholar] [CrossRef]
  58. Shi, Y.; Lan, F.; Matson, C.; Mulligan, P.; Whetstine, J.R.; Cole, P.A.; Casero, R.A.; Shi, Y. Histone Demethylation Mediated by the Nuclear Amine Oxidase Homolog LSD1. Cell 2004, 119, 941–953. [Google Scholar] [CrossRef]
  59. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
  60. Beisel, C.; Paro, R. Silencing chromatin: Comparing modes and mechanisms. Nat. Rev. Genet. 2011, 12, 123–135. [Google Scholar] [CrossRef]
  61. Kornberg, R.D. Chromatin Structure: A Repeating Unit of Histones and DNA. Science 1974, 184, 868–871. [Google Scholar] [CrossRef] [PubMed]
  62. Karakaidos, P.; Verigos, J.; Magklara, A. LSD1/KDM1A, a Gate-Keeper of Cancer Stemness and a Promising Therapeutic Target. Cancers 2019, 11, 1821. [Google Scholar] [CrossRef] [PubMed]
  63. Fang, Y.; Liao, G.; Yu, B. LSD1/KDM1A inhibitors in clinical trials: Advances and prospects. J. Hematol. Oncol. 2019, 12, 1–14. [Google Scholar] [CrossRef]
  64. Fang, Y.; Liao, G.; Yu, B. Targeting Histone Lysine Demethylase LSD1/KDM1A as a New Avenue for Cancer Therapy. Curr. Top. Med. Chem. 2019, 19, 889–891. [Google Scholar] [CrossRef] [PubMed]
  65. Zheng, Y.-C.; Ma, J.; Wang, Z.; Li, J.; Bailing, J.; Jiang, B.; Zhou, W.; Shi, X.; Wang, X.; Zhao, W.; et al. A Systematic Review of Histone Lysine-Specific Demethylase 1 and Its Inhibitors. Med. Res. Rev. 2015, 35, 1032–1071. [Google Scholar] [CrossRef] [PubMed]
  66. Whyte, W.A.; Orlando, D.A.; Hnisz, D.; Abraham, B.J.; Lin, C.Y.; Kagey, M.H.; Rahl, P.B.; Lee, T.I.; Young, R.A. Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes. Cell 2013, 153, 307–319. [Google Scholar] [CrossRef] [PubMed]
  67. Metzger, E.; Wissmann, M.; Yin, N.; Müller, J.M.; Schneider, R.; Peters, A.H.; Günther, T.; Buettner, R.; Schüle, R. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 2005, 437, 436–439. [Google Scholar] [CrossRef] [PubMed]
  68. Saleque, S.; Kim, J.; Rooke, H.M.; Orkin, S.H. Epigenetic Regulation of Hematopoietic Differentiation by Gfi-1 and Gfi-1b Is Mediated by the Cofactors CoREST and LSD1. Mol. Cell 2007, 27, 562–572. [Google Scholar] [CrossRef]
  69. Lee, M.G.; Wynder, C.; Cooch, N.; Shiekhattar, R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 2005, 437, 432–435. [Google Scholar] [CrossRef]
  70. Shi, Y.-J.; Matson, C.; Lan, F.; Iwase, S.; Baba, T.; Shi, Y. Regulation of LSD1 Histone Demethylase Activity by Its Associated Factors. Mol. Cell 2005, 19, 857–864. [Google Scholar] [CrossRef]
  71. Orkin, S.H.; Hochedlinger, K. Chromatin Connections to Pluripotency and Cellular Reprogramming. Cell 2011, 145, 835–850. [Google Scholar] [CrossRef] [PubMed]
  72. Kahl, P.; Gullotti, L.; Heukamp, L.C.; Wolf, S.; Friedrichs, N.; Vorreuther, R.; Solleder, G.; Bastian, P.J.; Ellinger, J.; Metzger, E.; et al. Androgen Receptor Coactivators Lysine-Specific Histone Demethylase 1 and Four and a Half LIM Domain Protein 2 Predict Risk of Prostate Cancer Recurrence. Cancer Res. 2006, 66, 11341–11347. [Google Scholar] [CrossRef] [PubMed]
  73. Lim, S.; Janzer, A.; Becker, A.; Zimmer, A.; Schüle, R.; Buettner, R.; Kirfel, J. Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology. Carcinogenesis 2010, 31, 512–520. [Google Scholar] [CrossRef] [PubMed]
  74. Schulte, J.H.; Lim, S.; Schramm, A.; Friedrichs, N.; Koster, J.; Versteeg, R.; Ora, I.; Pajtler, K.; Klein-Hitpass, L.; Kuhfittig-Kulle, S.; et al. Lysine-Specific Demethylase 1 Is Strongly Expressed in Poorly Differentiated Neuroblastoma: Implications for Therapy. Cancer Res. 2009, 69, 2065–2071. [Google Scholar] [CrossRef] [PubMed]
  75. Niebel, D.; Kirfel, J.; Janzen, V.; Höller, T.; Majores, M.; Gütgemann, I. Lysine-specific demethylase 1 (LSD1) in hematopoietic and lymphoid neoplasms. Blood 2014, 124, 151–152. [Google Scholar] [CrossRef] [PubMed]
  76. Somervaille, T.C.; Cleary, M.L. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell 2006, 10, 257–268. [Google Scholar] [CrossRef]
  77. Somervaille, T.C.; Matheny, C.J.; Spencer, G.J.; Iwasaki, M.; Rinn, J.L.; Witten, D.M.; Chang, H.Y.; Shurtleff, S.A.; Downing, J.R.; Cleary, M.L. Hierarchical Maintenance of MLL Myeloid Leukemia Stem Cells Employs a Transcriptional Program Shared with Embryonic Rather Than Adult Stem Cells. Cell Stem Cell 2009, 4, 129–140. [Google Scholar] [CrossRef]
  78. Harris, W.J.; Huang, X.; Lynch, J.T.; Spencer, G.J.; Hitchin, J.R.; Li, Y.; Ciceri, F.; Blaser, J.G.; Greystoke, B.F.; Jordan, A.M.; et al. The Histone Demethylase KDM1A Sustains the Oncogenic Potential of MLL-AF9 Leukemia Stem Cells. Cancer Cell 2012, 21, 473–487. [Google Scholar] [CrossRef]
  79. Lin, X.-M.; Zhong, W.-T.; Wang, C.-L.; Wang, S.-Q. Expression of histone demethylase lysine specific demethylase 1 in acute leukemia and its clinical significance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2011, 19, 1348–1352. [Google Scholar]
  80. Rhodes, D.R.; Kalyana-Sundaram, S.; Mahavisno, V.; Varambally, R.; Yu, J.; Briggs, B.B.; Barrette, T.R.; Anstet, M.J.; Kincead-Beal, C.; Kulkarni, P.; et al. Oncomine 3.0: Genes, Pathways, and Networks in a Collection of 18,000 Cancer Gene Expression Profiles. Neoplasia 2007, 9, 166–180. [Google Scholar] [CrossRef]
  81. Chen, E.; Mullally, A. How does JAK2V617F contribute to the pathogenesis of myeloproliferative neoplasms? Hematol. Am. Soc. Hematol. Educ. Program 2014, 2014, 268–276. [Google Scholar] [CrossRef] [PubMed]
  82. Yang, J.; Huang, J.; Dasgupta, M.; Sears, N.; Miyagi, M.; Wang, B.; Chance, M.R.; Chen, X.; Du, Y.; Wang, Y.; et al. Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proc. Natl. Acad. Sci. USA 2010, 107, 21499–21504. [Google Scholar] [CrossRef] [PubMed]
  83. Jutzi, J.S.; Kleppe, M.; Dias, J.; Staehle, H.F.; Shank, K.; Teruya-Feldstein, J.; Gambheer, S.M.M.; Dierks, C.; Rienhoff, H.Y., Jr.; Levine, R.L.; et al. LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. HemaSphere 2018, 2, e54. [Google Scholar] [CrossRef] [PubMed]
  84. Gill, H.; Yacoub, A.; Pettit, K.M.; Bradley, T.; Gerds, A.T.; Tatarczuch, M.; Shortt, J.; Curtin, N.J.; Rossetti, J.M.; Burbury, K.; et al. A Phase 2 Study of the LSD1 Inhibitor Img-7289 (bomedemstat) for the Treatment of Advanced Myelofibrosis. Blood 2021, 138, 139. [Google Scholar] [CrossRef]
  85. Gill, H.; Yacoub, A.; Pettit, K.; Bradley, T.; Gerds, A.; Tatarczuch, M.; Shortt, J.; Curtin, N.; Rossetti, J.; Burbury, K.; et al. A phase 2 study of IMG-7289 (bomedemstat) in patients with advanced myelofibrosis. HemaSphere 2022, 6, 1809. [Google Scholar] [CrossRef]
  86. Kohrogi, K.; Hino, S.; Sakamoto, A.; Anan, K.; Takase, R.; Araki, H.; Hino, Y.; Araki, K.; Sato, T.; Nakamura, K.; et al. LSD1 defines erythroleukemia metabolism by controlling the lineage-specific transcription factors GATA1 and C/EBPα. Blood Adv. 2021, 5, 2305–2318. [Google Scholar] [CrossRef]
  87. Rivers, A.; Vaitkus, K.; Ibanez, V.; Ruiz, M.A.; Jagadeeswaran, R.; Saunthararajah, Y.; Cui, S.; Engel, J.D.; DeSimone, J.; Lavelle, D. The LSD1 inhibitor RN-1 recapitulates the fetal pattern of hemoglobin synthesis in baboons (P. anubis). Haematologica 2016, 101, 688–697. [Google Scholar] [CrossRef]
  88. Palandri, F.; Vianelli, N.; Ross, D.M.; Cochrane, T.; Lane, S.W.; Larsen, S.R.; Gerds, A.T.; Halpern, A.B.; Shortt, J.; Rossetti, J.M.; et al. A Phase 2 Study of the LSD1 Inhibitor Img-7289 (bomedemstat) for the Treatment of Essential Thrombocythemia (ET). Blood 2021, 138, 386. [Google Scholar] [CrossRef]
  89. Kremyanskaya, M.; Mascarenhas, J.; Palandri, F.; Vannucchi, A.; Verstovsek, S.; Harrison, C.N.; Bose, P.; Schiller, G.J.; Rampal, R.; Drummond, M.W.; et al. Pelabresib (CPI-0610) Monotherapy in Patients with Myelofibrosis—Update of Clinical and Translational Data from the Ongoing Manifest Trial. Blood 2021, 138, 141. [Google Scholar] [CrossRef]
  90. Verstovsek, S.; Salama, M.E.; Mascarenhas, J.; Talpaz, M.; Mesa, R.A.; Vannucchi, A.; Rampal, R.; Oh, S.T.; Olteanu, H.; Chiu, A.; et al. Disease-Modifying Potential of BET Inhibitor Pelabresib (CPI-0610) As Demonstrated By Improvements in Bone Marrow Function and Clinical Activity in Patients with Myelofibrosis-Preliminary Data. Blood 2021, 138, 2568. [Google Scholar] [CrossRef]
  91. Mascarenhas, J.; Komrokji, R.S.; Palandri, F.; Martino, B.; Niederwieser, D.; Reiter, A.; Scott, B.L.; Baer, M.R.; Hoffman, R.; Odenike, O.; et al. Randomized, Single-Blind, Multicenter Phase II Study of Two Doses of Imetelstat in Relapsed or Refractory Myelofibrosis. J. Clin. Oncol. 2021, 39, 2881–2892. [Google Scholar] [CrossRef]
  92. Pemmaraju, N.; Garcia, J.S.; Potluri, J.; Holes, B.L.; Harb, J.; Jung, P.; Hutti, J.E.; Prchal, J.T.; Verstovsek, S.; Harrison, D.C. The Addition of Navitoclax to Ruxolitinib Demonstrates Efficacy within Different High-Risk Populations in Patients with Relapsed/Refractory Myelofibrosis. Blood 2020, 136, 49–50. [Google Scholar] [CrossRef]
  93. Vachani, P.; Lange, A.; Delgado, R.G.; Al-Ali, H.K.; Hernández-Rivas, J.M.; Kiladjian, J.-J.; Vannucchi, A.; Perkins, A.C.; Valmeekam, V.; Krejsa, C.M.; et al. Potential Disease-Modifying Activity of Navtemadlin (KRT-232), a First-in-Class MDM2 Inhibitor, Correlates with Clinical Benefits in Relapsed/Refractory Myelofibrosis (MF). Blood 2021, 138, 3581. [Google Scholar] [CrossRef]
  94. Masarova, L.; Verstovsek, S.; Hidalgo-Lopez, J.E.; Pemmaraju, N.; Bose, P.; Estrov, Z.; Jabbour, E.J.; Ravandi-Kashani, F.; Takahashi, K.; Cortes, J.; et al. A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood 2018, 132, 1664–1674. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mechanism of actions of novel agents targeting epigenetic regulators. (A) Removal of methyl groups by LSD1, inhibiting p53 methylation and abrogating cellular apoptosis. LSD1 inhibitor antagonizes LSD1 to restore tumor-suppressive effects of p53. (B) Anchoring of BET proteins to acetylated lysine residues to activate NF-κB pathway. BET inhibitors block BET proteins and the proinflammatory pathway to reduce synthesis of proinflammatory cytokines. (C) Removal of acetyl groups by HDAC, decreasing tumor-suppressor gene transcription while deacetylating HSP to aggravate JAK/STAT signaling pathway. This effect can be overcome by HDAC inhibitors. (D) Aberrant phosphorylation of PRMT5 by JAK2V617F, leading to impaired methylation activity. Thus, E2F is methylation for cell cycle progression and myeloproliferation. The aberrant activation can be inhibited by an PRMT5 inhibitor. Me: methylation; Ac: acetylation; LSD1: lysine-specific demethylase-1; BET: bromodomain and extraterminal domain; NF-κB: nuclear factor kappa-light-chain enhancer of activated B cells; HDAC: histone deacetylase; TS genes: tumor suppressor genes; HSP: heat shock protein; JAK: Janus kinase; STAT: signal transducer and activator of transcription; PRMT5: protein arginine methyltransferase 5; E2F: E2F transcription factor 1.
Figure 1. Mechanism of actions of novel agents targeting epigenetic regulators. (A) Removal of methyl groups by LSD1, inhibiting p53 methylation and abrogating cellular apoptosis. LSD1 inhibitor antagonizes LSD1 to restore tumor-suppressive effects of p53. (B) Anchoring of BET proteins to acetylated lysine residues to activate NF-κB pathway. BET inhibitors block BET proteins and the proinflammatory pathway to reduce synthesis of proinflammatory cytokines. (C) Removal of acetyl groups by HDAC, decreasing tumor-suppressor gene transcription while deacetylating HSP to aggravate JAK/STAT signaling pathway. This effect can be overcome by HDAC inhibitors. (D) Aberrant phosphorylation of PRMT5 by JAK2V617F, leading to impaired methylation activity. Thus, E2F is methylation for cell cycle progression and myeloproliferation. The aberrant activation can be inhibited by an PRMT5 inhibitor. Me: methylation; Ac: acetylation; LSD1: lysine-specific demethylase-1; BET: bromodomain and extraterminal domain; NF-κB: nuclear factor kappa-light-chain enhancer of activated B cells; HDAC: histone deacetylase; TS genes: tumor suppressor genes; HSP: heat shock protein; JAK: Janus kinase; STAT: signal transducer and activator of transcription; PRMT5: protein arginine methyltransferase 5; E2F: E2F transcription factor 1.
Cells 11 02107 g001
Table 1. Selected novel agents showing improvement in surrogate markers for disease modification in myelofibrosis, either as single agent or in combination with ruxolitinib.
Table 1. Selected novel agents showing improvement in surrogate markers for disease modification in myelofibrosis, either as single agent or in combination with ruxolitinib.
ClassDrugStudy PopulationDesign SVR35 at 24 WeeksTSS50 at 24 WeeksAnemia ResponseVAF ReductionBM Fibrosis Reduction
LSD1 inhibitorBomedemstat [84,85]Ruxolitinib exposed: 83% (74/89)
N = 89
Phase 2 (ongoing)
Single-agent bomedemstat
6% (3/50)19% (5/26)In TD patients: 52% (11/21) had stable or reduced transfusion burden; 14% (3/21) became TIVAF reduction 52% (36/69), most frequently in JAK2V617F and/or ASXL131% (16/52) improved by 1 grade
50% (26/52) stable
BET inhibitorPelabresib
(MANIFEST) [89,90]
Both JAKi exposed and JAKi naïve, N = 271Phase 1/2 (ongoing)
Arm 1: JAKi exposed (pelabresib)
Arm 2: JAKi exposed (pelabrasib + ruxolitinib)
Arm 3: JAKi naïve (pelabresib + ruxolitinib)
Arm 1: 11% (7/64)
Arm 2: 20% (16/81)
Arm 3: 68% (57/84)
Arm 1: 28% (18/64)
Arm 2: 37% (30/81)
Arm 3: 56% (46/82)
Arm 1: In TD patients, 16% (4/25) became TI
Arm 2: In TD patients, 36% (13/36) became TI
Arm 3: In patients with Hb < 10 g/dL; Hb improved by 1 g/dL
Not reportedArm 1: 23% (7/30) improved at 24 weeks
Arm 2: 25% (9/36) improved at 24 weeks
Arm 3: 31% (16/52) improved at 24 weeks
Telomerase inhibitorImetelstat
(IMBark) [91]
JAKi exposed
N = 59
Phase 2 (complete)
Single-agent imetelstat
10.2% (6/59)32.2% (19/59)In TD patients, 25% (3/12) became transfusion-independent42% had ≥25% reduction in VAF41% (15/37) had reduction in BM fibrosis
BH3 mimetic; Bcl-2/Bcl-XL inhibitorNavitoclax
(REFINE) [92]
Ruxolitinib exposed
N = 174
Phase 2 (ongoing)
Navitoclax +/− ruxolitinib
27% (9/34)30% (9/34)In TD patients or patients with Hb < 10 g/dL; TI or ≥ 2 g/dL in 64% (7/11)46% (12/26) had >10% reduction in VAF21% (7/34) had BM fibrosis reduction at 24 weeks
MDM2 inhibitorNavtemadlin
(BOREAS) [93]
JAKi exposed
N = 113
Phase 2 (ongoing)
Single-agent navtemadlin
Not reportedNot reportedNot reported34% had ≥20% reduction in VAF27% ≥ 1 grade reduction in BM fibrosis
Hypomethylating agentAzacitidine [94]JAKi naïve
N = 60
Phase 2
Ruxolitinib + azacitidine
NR54% (25/46)In TD patients, 20% (1/5) became TI81% (13/16) had reduction in JAK2V617F VAF at 24 weeks57% (8/14) had reduction in BM fibrosis at 24 weeks
SVR35: ≥35% reduction in spleen volume from baseline to 24 weeks; TSS50: ≥50% reduction in total symptom score from baseline to 24 weeks; VAF: variant allele frequency; BM: bone marrow; LSD1: lysine-specific demethylase 1; BET: bromodomain and extraterminal; Bcl-2: B-cell lymphoma 2; MDM2: murine double minute 2; TD: transfusion-dependent; TI: transfusion-independent; JAKi: JAK inhibitor; Hb: hemoglobin; NR: not reported.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gill, H. Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis. Cells 2022, 11, 2107. https://doi.org/10.3390/cells11132107

AMA Style

Gill H. Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis. Cells. 2022; 11(13):2107. https://doi.org/10.3390/cells11132107

Chicago/Turabian Style

Gill, Harinder. 2022. "Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis" Cells 11, no. 13: 2107. https://doi.org/10.3390/cells11132107

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop