Accepted Manuscript New therapeutic approaches in myelodysplastic syndromes: hypomethylating agents and lenalidomide Clémence Loiseau, Ashfaq Ali, Raphael Itzykson PII:
S0301-472X(15)00197-6
DOI:
10.1016/j.exphem.2015.05.014
Reference:
EXPHEM 3272
To appear in:
Experimental Hematology
Received Date: 15 May 2015 Revised Date:
28 May 2015
Accepted Date: 29 May 2015
Please cite this article as: Loiseau C, Ali A, Itzykson R, New therapeutic approaches in myelodysplastic syndromes: hypomethylating agents and lenalidomide, Experimental Hematology (2015), doi: 10.1016/ j.exphem.2015.05.014. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT New therapeutic approaches in myelodysplastic syndromes: hypomethylating agents and lenalidomide
Clémence Loiseau1, Ashfaq Ali2, Raphael Itzykson1,2 1
Department of Hematology, Saint-Louis Hospital, Assistance Publique – Hopitaux de Paris, Paris
Diderot University, Paris, France 2
INSERM U944, Saint-Louis Institute, Paris France
Corresponding Author: Raphael Itzykson, MD PhD Adult Hematology Unit Hopital Saint-Louis 1 avenue Claude Vellefaux 75010 Paris, France Tel: +33142385127 Fax : +33142385128 Email :
[email protected]
Abstract: 102 words Text: 5095 words References: 150 Figures: 3 Keywords: myelodysplastic syndromes; hypomethylating agents; azacitidine; decitabine; lenalidomide
ACCEPTED MANUSCRIPT Abstract Recent advances in the treatment of myelodysplastic syndromes have come from the use of the hypomethylating agents decitabine and azacitidine, and the immunomodulatory drug lenalidomide. Their clinical benefit has been demonstrated by randomized phase III clinical trials, mostly in higher risk and del(5q) myelodyslastic syndromes, respectively. However, neither drug appears to eradicate myelodysplastic stem cells, and thus do not currently represent curative options. Here we review data from both clinical and translational research on those drugs, in order to identify their molecular and cellular mechanisms of action, and to delineate paths for improved treatment allocation and further therapeutic advances in myelodysplastic syndromes.
Introduction MDS are clonal disorders arising in aging HSC as a result of cell-intrinsic genetic (and potentially epigenetic) insults, and micro-environmental alterations1. These alterations result in chronic cytopenias, with transfusion-dependent anemia being present in 80% of patients, and increased risk of progression to AML. Prognosis of MDS depends on the number and severity of cytopenias, percentage of bone marrow blasts, and cytogenetics2. MDS is often broadly categorized in lower-risk disease, where therapy mainly aims at coping with anemia, and higher-risk disease, where therapeutic interventions seek to delay progression to AML and improve survival. Hematopoietic stem cell transplantation (HSCT) remains the only curative option in MDS patients, though relapse remains frequent after HSCT. In recent years, two major advances in the treatment of myelodysplastic syndromes have occurred. The DNA methyltransferase (DNMT) inhibitors azacitidine and decitabine, also called ‘hypomethylating agents’ (HMA) have been shown to alter the natural course of higher-risk MDS, significantly prolonging survival of patients compared to other treatment options (aside from transplantation), by delaying progression to AML and improving cytopenias. The immunomodulatory agent lenalidomide durably, though not indefinitely, restores erythropoiesis and induces clonal suppression in lower-risk MDS patients with del(5q), and also improves anemia in a subset of patients without del(5q). Here we review the lessons learnt from clinical trials and translational studies performed with these drugs in MDS.
Hypomethylating agents Epigenetics of MDS The methylome of cancer cells is characterized by global DNA hypomethylation, specifically in heterochromatin, contrasting with focal aberrant hypermethylation of promoter CpG islands that
ACCEPTED MANUSCRIPT have been linked to repression of tumor suppressor gene expression and tumor progression. In euchromatin, the regulatory roles of DNA methylation extend outside of CpG islands. Intragenic DNA methylation is involved in RNA transcription processivity and splicing 3. DNA methylation at distal enhancers can also play an important role in transcriptional regulation 4. In MDS, aberrant promoter hypermethylation of the CDKN2B promoter is a prototypic driver ‘epimutation’ associated with disease progression 5, and HMA were thought to erase these epimutations and thereby delay MDS progression to AML. Comprehensive analyses of MDS methylomes are emerging, and have only recently extended beyond CpG islands, with the advent of whole genome bisulfite sequencing (WGBS)
6
or reduced
representation bisulfite sequencing (RRBS) 7. Early methylome studies focusing on CpG islands identified a signature of promoter hypermethylation in MDS compared to normal hematopoietic stem/progenitors cells, with dynamic accumulation of aberrant hypermethylation marks during disease progression from low- to high- risk and AML 8,9. Intergenic methylation maps remain poorly characterized in MDS, with recent data emerging in the CMML subset 7. Detailed cell sorting experiments have confirmed that aberrant methylation marks arise in MDS ‘stem cells’
10
. Mutations in the DNMT3A gene that encodes one of the three DNA
methyltransferases involved in de novo (as well as maintenance) methylation are present in MDS, though at lower frequencies than in AML. Mutations in the TET2 gene, and to a lesser extent in the IDH1 and IDH2 genes whose many functions include regulation of TET2 enzymatic activity, are prevalent in MDS. TET2 catalyzes hydroxymethylation of methylated cytosines, leading to demethylation. There is limited data regarding the inter-relationship between aberrant methylomes and gene mutations in MDS. In CMML, aberrant methylation seems to be only partly dictated by mutations in epigenetic regulators such as TET2, DNMT3A or ASXL1 7,11. Mutations in splicing factors, which are specific of MDS or secondary AML compared to other subtypes of myeloid neoplasms, also have a distinct methylation signature, presumably because these mutations induce mis-splicing of epigenetic regulators 7. An alternative mechanism of acquisition and selection of aberrant methylation is aging of HSC itself, which leads to accumulation of DNA hypermethylation in polycomb-regulated genes
12
. TET2 mutations provide pre-leukemic HSC with a competitive
advantage over normal HSC in murine models
13,14
, and probably in healthy aging subjects
15,16
. The
HSC ‘self-renewal’ pathways involved in this clonal advantage remain elusive, although methylome studies have pointed to HOX transcription factors 17 and Wnt signaling pathways as candidates 8.
Lessons learnt from clinical trials with HMA
ACCEPTED MANUSCRIPT Azacitidine and decitabine have been compared to conventional care, mostly supportive care or lowdose cytarabine, in several large randomized clinical trials of higher-risk MDS patients: both drugs were shown to induce significant overall response rate, mostly correction of cytopenias (‘hematological improvement’) contrasting with a limited 15-20% complete remission rate, and to delay progression of MDS to AML higher-risk MDS
19
18-21
. Azacitidine was also shown to prolong overall survival in
and AML with lower blast count
22
. No survival benefit was demonstrated for
decitabine, most likely because of difference in study designs 20,21, though it remains possible that the two drugs have different biological activities in MDS. Several lessons have been taken from detailed analyses of these trials that shed light on the possible mechanism of action of these drugs. First, several (4-6) months of exposure to HMA are required to achieve a response, and this response can improve over time
23
. Second, continued exposure to HMA is required to maintain response, and
relapse occurs within months of HMA withdrawal in most cases
24
. Achievement of complete
remission is not a prerequisite for prolonged survival with HMA, as a survival benefit is also seen in patients with hematological improvement despite persistence of blast excess
25,26
. Cytogenetic
response is not a better surrogate for survival benefit 27. Response is difficult to predict with routine clinical variables, though these can identify subsets of patients with different survival with HMA 26, stressing the need for biomarkers of HMA. Finally, outcome of patients after HMA failure is dismal 28.
Metabolism of hypomethylating agents The hypomethylating agents (HMA) azanucleotides 5-azacytidine (azacitdine, AZA) and 5azadeoxycytidine (decitabine, DAC) are cytosine analogs with unmodified sugar moiety, therefore with reduced cytotoxicity upon incorporation into DNA compared to cytarabine. At high doses, these compounds nonetheless display significant cytotoxic activity in AML cells. At lower doses, incorporation of azanucleotides leads to cell-cycle dependent global demethylation, in part because the aza- moiety inhibits maintenance DNA methyl- group transfers during replication, but mostly because azanucleotides covalently bind DNMTs and induce their proteosomal degradation 29. After several rounds of cycling, the DNMT pool is depleted, leading to a profound and global hypomethylation, which is rapidly reversible upon HMA withdrawal. DNA incorporation-independent DNMT depletion has also been described in vitro at high doses of DAC 30. The relevance of such cellcycle independent- DNA hypomethylation at clinically relevant DAC concentrations has not yet been determined. Increasing evidence suggest that inter-individual differences in intra-cellular uptake and metabolism of HMA are responsible for the variable, and hardly predictable clinical activity of these compounds in patients. In vitro experiments
31
and in vivo correlative studies
7,32,33
suggest that sensitivity to
ACCEPTED MANUSCRIPT azanucleotides mostly depends on the level of its incorporation in DNA, which can be modulated by differences in intracellular metabolism or rate of DNA replication. These phenomena as summarized on Figure 1. Similar to other cytosine analogs, intracellular uptake of azanucleotides depends on equilibrative nucleoside transporters hENT1 and hENT2 34. The first step of decitabine intracellular metabolism is conversion to 5-aza-dCMP, which is catalyzed by deoxycytidine Kinase (dCK), whereas uridinecytidine kinase (UCK) is responsible for initial activation of AZA to 5-aza-CMP. Conversely, cytidine deaminase (CDA) catalyzes inactivation of 5-aza-dCTP to 5-aza-dUTP, which can no longer be incorporated into DNA. In vitro resistance to DAC in leukemia cell lines has been assigned to insufficient DNA incorporation, through low expression of DCK and hENT1, and high expression of CDA 31. In different studies, patients who fail to respond to DAC have been reported to have lower hENT1 expression 35, or a higher ratio of CDA/DCK expression than responders 32. One study found lower DCK expression at relapse after achieving response to DAC
35
, but this result has not been
confirmed 32. Lower expression of UCK at baseline predicts resistance to AZA, whereas SNP in CDA have not been reported to impact the outcome of MDS patients treated with AZA 33. Finally, slow DNA replication also increases the rate of DAC incorporation 36, and high expression of genes involved in DNA replication and cell cycle predict clinical response to DAC 7. Hyperexpression of the chemokines CXCL4 and CXCL7 have been associated to resistance to DAC, and this is thought to result from induction of MDS stem cell quiescence 7. Only 20-30% of intracellular 5-azacytidine is incorporated into DNA, and this requires an additional enzymatic step of reduction of 5-aza-CDP to 5-aza-dCDP catalyzed by ribonucleotide reductase (RNR). The majority of AZA is converted to 5-aza-CTP and is incorporated into RNA, with potential consequences on protein synthesis and cell viability that have been incompletely explored in MDS 37,38
. One of the key proteins downregulated through this pathway could be the RNR enzyme itself,
leading to depletion of nucleotide pools and to further cellular stress 39. Recent data have implicated AZA in the reversion of nonsense mediated RNA decay (NMD) in a MYC-dependent fashion 40. The NMD pathway targets transcripts that contain premature stop codons for degradation. Mis-spliced transcripts frequently accumulate in MDS as a result of mutations in components of the spliceosome machinery
41
. Further efforts are required to explore the role of AZA on reverting the post-
transcriptional consequences of splice mutations in MDS, as technological advances now render feasible the monitoring of RNA incorporation of AZA 42.
Epigenetic modulation induced by HMA
ACCEPTED MANUSCRIPT Incorporation of HMA into DNA has been reported to trigger the DNA damage response pathway, which in turn can lead to apoptosis 43. DNA damage resulting from HMA incorporation is repaired by the BER machinery 44, which can be blocked by PARP inhibitors, suggesting possible synergy between both drug classes. Most of the DNA damaging properties of HMA have been reported in in vitro experiments performed in AML cells lines at micromolar concentrations
43-45
, whereas
pharmacokinetic studies indicate that peak plasmatic concentrations of AZA are in the range of 50 – 500 nM 46. The relevance of the genotoxic activity of HMA on the current clinical activity in MDS or AML is thus dubious, although DNMT1 depletion per se has been reported to induce mismatch repair defects in human embryonic cells 47. At clinically active doses, HMA exposure leads to genome-wide uniform demethylation, as assessed by WGBS 6,48. Several studies have also reported genome-wide modifications in gene expression upon AZA or DAC exposure
37,49
. Of note, the patterns induced by the two drugs only partly overlap,
presumably because AZA incorporation into RNA can affect the stability of certain transcripts. However, genome-wide studies focusing on promoter CpG islands failed to identify a clear correlation between promoter demethylation patterns and gene expression modifications upon HMA exposure, whether in vitro 48,50 or in vivo 51. Several non-mutually exclusive hypotheses may account for this puzzling observation. First, overall promoter methylation may not be the best surrogate for methylation ‘locking’ of gene expression. Application of a sophisticated algorithm to WGBS data for instance identified a subset of upregulated genes with a shared hypomethylation signature at a very precise location, near the TSS, suggesting that demethylation of these specific TSS-flanking CpGs by HMA was directly responsible for gene expression upregulation 6. Another explanation is that demethylation outside of promoter CpG islands, notably in distant enhancers, can account for the gene expression modifications induced by HMA 7. In particular, demthylation of the PU.1 upstream regulatory element (URE) may account for the differentiating effect of AZA 52. Finally, demethylation of CpG promoters may also not be sufficient to induce gene expression without concomitant modification of repressive histone marks
53
. This key observation has set the ground for the
combination of HMA with other classes of ‘epidrugs’, in particular HDAC inhibitors.
Cellular consequences of demethylation The cellular consequences of clinically-relevant HMA exposure in MDS or AML remain elusive, notably because of the lack of a suitable in vivo murine model for this study. Most hypotheses are thus based on in vitro experiments or on correlative studies investigating potential biomarkers of HMA activity. They are summarized in Figure 2.
ACCEPTED MANUSCRIPT Many groups have focused on investigating cell death induced by HMA, mostly in AML cell lines. Autophagic cell death has been reported in a MDS cell line upon AZA exposure, and hyperexpression of the anti-apoptotic protein BCL2L10 correlates with resistance to AZA in patients
54,55
. Studies
investigating the induction of apoptosis (or autophagic cell death) in the recently identified ‘MDS stem cells’
10,56
in vivo are eagerly awaited. It is however unlikely that HMA induce significant MDS
stem cell eradication in the clinical setting: not only is the complete remission rate low with HMA, but response typically requires several months, compared to the few weeks required to achieve remission after intensive chemotherapy. Finally, cytogenetic response is infrequent in patients treated with HMA, and poorly correlates with long term survival 27, suggesting that clonal eradication at the stem cell level is not the main cause of improved outcome in MDS treated by HMA. Other studies, also relying on cell lines, have suggested that HMA exposure, though not inducing MDS/AML stem cell death, may reduce their self-renewal advantage 57,58, in keeping with studies in mice where reduced Dnmt1 expression impaired MLL-AF9 leukemic stem cell self-renewal
59
.
Interestingly, other studies performed in vivo suggest that HMA have an opposite effect on healthy HSC 60,61, although this property is not recapitulated in a Dnmt1+/- haplo-insuficient mouse model 62. These observations raise the intriguing possibility that HMA, rather than eradicating MDS stem cells, reverse their competitive advantage over normal HSC in contributing to hematopoiesis. This model could be addressed more directly by in vivo exposure to HMA in competitive transplant experiments in Tet2 knockout mice, because TET2-mutated MDS have a higher response rate to AZA because Tet2 invalidation provides a competitive advantage to murine HSC
63-65
, and
13,14
. A model of HMA-
induced MDS stem cell quiescence fits better with the transient reduction in leukemic stem cell frequency observed in patients by flow cytometry 66, the delay in progression to AML, and relatively rapid relapse after withdrawal of HMA 24. A third, non-mutually exclusive, direct mechanism of action of HMA could be to enhance the differentiation of MDS progenitor cells. This mechanism could in particular account for the frequent correction of cytopenias noted in clinical trials, which seem to convey part of the survival benefit of HMA
25,26
, where the restored hematopoiesis is often clonal
67,68
. HMA exposure can in particular
modulate the expression level of several key myeloid transcription factors, including PU.1, through its upstream regulatory element
52
, and the CEBP family
69
. In vitro data suggest synergism of these
differentiation properties with growth factors such as G-CSF 52, while limited clinical data indicate that addition of recombinant EPO can synergize with HMA in improving anemia, even in higher-risk MDS patients who have very limited sensitivity to EPO alone 70.
ACCEPTED MANUSCRIPT Whether the response to HMA is TP53-dependent is a matter of controversy. In vitro data suggest that HMA-induced cell death may be P53-independent 71, and in vivo experiments support the notion that HMA-induced differentiation is also P53-independent 58. The latter can be reconciled with the finding that response rates to HMA appear independent of TP53 status or expression 72,73. However, the survival of TP53 patients remains poor under HMA
72,73
, and there is no direct evidence from
randomized trials that the poor prognosis of TP53 is even partly alleviated by HMA. In the AZA-001 trial, the outcome of patients with complex karyotype, half of whom harbor TP53 mutations 74, was dismal regardless of treatment 19,75.
HMA as immunomodulatory drugs Non cell-autonomous mechanisms of action of HMA are also an area of investigation. The bone marrow niche has been involved in the pathogenesis of MDS 76,77, but there is so far limited data on the activity of HMA on the stromal environment of MDS stem and progenitor cells (Götze et al., 13th International Symposium on Myelodysplastic Syndromes, Washington DC, May 2015). Conversely, several authors have studied the consequences of HMA on the immune system of patients with MDS or AML, notably in the context of ASCT. Clinical evidence supports an immunomodulatory role of HMA in MDS: treatment with HMA alleviates auto-immune MDS-related manifestations
78
, and
patients treated with a combination of HMA and donor lymphocyte infusions (DLI) after HSCT seem to be less frequently subject to aggressive GVHD models
68,79
. These findings, as well as data from murine
80,81
, support the contention that HMA have immunomodulatory properties against GVHD yet
preserve allogeneic graft-versus-leukemia effects. This model will be addressed by ongoing trials of HMA maintenance after HSCT. The molecular underpinnings of such dissociated immunomodulatory role remain unclear, but could involve enhanced generation of regulatory T cells (Tregs) through demethylation of the FOXP3 receptor
80,81
. A similar induction of Tregs has been noted in patients
receiving HMA after HSCT 82,83, though the functionality of these expanded Tregs is debated 84,85. HMA may also induce anti-tumoral immune responses outside of the transplant setting, notably by restoring the expression of tumor antigens at the surface of MDS cells. HMA have for instance been shown to induce a CD8+ T-cell response to cancer testis antigens in MDS/AML patients
86,87
.
Prospective studies investigating immune parameters as biomarkers of HMA in higher-risk MDS are thus eagerly awaited, as HMA can be double-edged swords in anti-tumoral immune responses. For instance, recent data suggest that azacitidine demethylates the promoter of PD-1 of on T cells, restoring expression of this auto-inhibitory receptor, potentially leading to exhaustion of antitumoral T-cell mediated responses 88,89.
ACCEPTED MANUSCRIPT
Predicting the response to HMA There have been numerous attempts at validating biomarkers of response or survival benefit in MDS or AML patients treated with HMA. Baseline global methylation levels do not predict response to HMA 7,51,90,91, and, in studies where global methylation has been found to impact overall survival, this impact was also found in cohorts not receiving HMA 91. Conversely, demethylation under HMA has repeatedly been found to predict outcome
90,91
, again stressing the role of variable DNA
incorporation of HMA as a key parameter. Gene expression profiles may predict sensitivity to HMA, and most profiles appear directly or indirectly related to cell cycle or to global regulation of methylation 7, such as mir-29b, which regulates DNMT expression
92
, or MLL5, which has been
proposed to interact with TET2 and thus with active demethylation 93. Mutations in TET2 have been shown by several groups to predict superior response rates to HMA 6365
, whereas evidence regarding mutations in other epigenetic regulators such as DNMT3A or ASXL1 is
more conflictual 7,64,65,94,95. The molecular underpinning of the higher response rate in TET2-mutated patients is currently unknown, and, as previously discussed, is unlikely to reflect differences in global methylation levels. However, the superior response rate of TET2-mutated patients does not translate in a significant survival benefit. TET2 mutations do not have a strong effect on MDS prognosis outside of HMA treatment 96, thus ruling out a potential ‘Simpson’s paradox’ as explanation to this lack of survival benefit, and stressing the need for a better understanding of HMA mode of action to identify relevant biomarkers predicting survival rather than response with HMA.
Improving the therapeutic benefit of HMA Current clinical research aiming at improving the outcome of MDS patients with HMA follow two different but non-exclusive routes: achieving more continuous hypomethylation without undue toxicity, and combining HMA with other drug classes. Current administration schemes of both AZA and DAC are discontinuous (7 and 5 days per monthly cycles in the most frequently used schedules, respectively), and thus only induce transient hypomethylation, with complete genomic remethylation before the start of the next HMA cycle 90. More continuous administration of lower, non-genotoxic doses of DAC seem to yield similar clinical efficacy in MDS 97. Oral azacitidine, though it has variable inter-individual pharmacokinetics, can also potentially achieve more continuous hypomethylation than parenteral administration 46. SGI-110, a dinucleotide of decitabine and deoxyguanosine, is a new HMA that increases in vivo exposure of
ACCEPTED MANUSCRIPT decitabine by preventing deamination of DAC by CDA. Because of its prolonged half-like, SGI-110 could allow more stable hypomethylation, with a favorable toxicity profile due to its lower peak concentration compared to DAC 98. Another approach to improve the efficacy of HMA is to combine it with another drug class. HDAC inhibitors have been thoroughly investigated, because of the abundant in vitro data showing synergism in restoring silenced gene expression. HDAC inhibition can also restore the expression of mir-29b, which in turn downregulates DNMT expression. The levels of expression of mir-29b have indeed been shown to impact response to HMA 92. This observation advocates for the use of HDACi as priming before HMA administration 99. However, this strong pre-clinical rationale has been faced by disappointing clinical data. Randomized trials have yet to demonstrate superiority of a HMA / HDACi combination 100. Several reasons can explain this lack of clinical efficacy. First, detailed in vitro studies have shown that the timing of combination is of critical importance 101. Second, HDACi are in fact most often deacetylase inhibitors, with incomplete specificity for histones. Off-target effects may thwart their activity in leukemic cells, and probably accounts for the limiting GI toxicity of this drug class. Combination of HMA with other classes of ‘epidrugs’ are also being investigated to restore gene expression. These drugs either inhibit the histone demethylase LSD1 involved in erasing activating histone marks such as H3K4me3 102, or instead inhibit the histone methyltransferase EZH2 that writes the inactivating mark H3K27m3
103
. Like HDACs, LSD1 also has non-histone substrates, including
DNMT1. Interestingly, methylation of DNMT1 by LSD1 stabilizes DNMT1, and combination of HMA with LSD1 inhibitors could synergize in inducing global hypomethylation
102
. Combination of HMA
with myeloid differentiating agents such as vitamin D or vitamin A derivatives including RAR and RXR agonists has also been investigated
104-106
, again without clinical evidence for strong synergism.
Finally, the most promising combination of HMA may be with the lenalidomide, though strong biological rationale for such combination is currently lacking. These empirically designed combinations take advantage of the non-overlapping extra-hematological toxicity of these drugs, and of the specific activity of LEN in MDS 107.
Lenalidomide Lenalidomide is a thalidomide derivative with immunomodulatory (IMiD) and anti-neoplastic activities in some solid tumors, in myeloma, lymphoma, and in MDS. IMiDs including lenalidomide, regulate the E3 ubiquitin ligase complex CRBN-CRL4 through the DNA damage-binding protein-1 (DDB1) and cereblon (CRBN) subunits, both blocking the degradation of endogeneous substrates of
ACCEPTED MANUSCRIPT this ubiquitin ligase, and recruiting novel targets for degradation (Figure 3) 108-110. In myeloma and T cells, lenalidomide interaction with the CRBN-CRL4 complex leads to selective ubiquitination and degradation of the lymphoid transcription factors IKZF1 and IKZF3 109.
LEN in 5q- syndromes In early trials of LEN in the treatment of MDS, its clinical activity appeared prominent in patients with lower-risk disease (ie. with predominant anemia and no blast excess) and del(5q) 111,112. In later trials of LEN focusing on del(5q) patients, LEN was found to achieve prolonged RBC transfusion independence in 50-60% of patients, potentially improving survival 113,114. Cytogenetic responses are also noted in half of cases but do not seem to delay transformation to AML or prolong overall survival 115. In MDS, del(5q) induces haploinsufficiency of a commonly deleted region (CDR) that encompasses 40 genes and several miRNAs. A functional screen has identified that haploinsufficiency of the RPS14 gene involved in ribosomal biogenesis plays a major role in the macrocytic anemia of 5q- syndromes 116
. Ribosomal stress resulting from reduced RPS14 gene dosage is thought to result in degradation of
MDM2, in turn activating p53 117. This activation of the P53 pathway, which is also evident in GEP of del(5q) patients
118
, is predominant in the erythroid compartment and it is not certain that it is
involved in the stem cell advantage of del(5q)
117
. Haploinsufficiency of two microRNA contained in
the CDR, mir-145 and mir-146, accounts for the thrombocytosis found in these patients, in part through non cell-autonomous mechanisms, and may as well contribute to the clonal dominance of del(5q)
119
. However, the exact mechanisms leading to clonal advantage of del(5q) MDS stem cells
remains to be fully determined. CSNK1A1, which encodes casein kinase 1A1 and SPARC, a gene involved in cellular adhesion to extracellular matrix, are potential haploinsufficient tumor suppressor genes located in the CDR that may contribute to clonal dominance 120,121. There is conflictual evidence as to whether LEN restores expression of RPS14 122, mir-145 123 or SPARC 120
in del(5q) cells. In particular, in vivo pre/post-treatment comparisons may be influenced by the
reduction in del(5q) cells and concomitant expansion of non-clonal progenitors. In vitro, one group found a statistical link between induction of mir-145 expression by LEN in CD34+ del(5q) stem/progenitor cells and clinical response to LEN 123. A first model of LEN activity in del(5q) MDS cells involves stabilization of MDM2 through inhibition of of two haploinsufficient phosphatases, PP2A and CDC25C. Stabilization of MDM2 in turn leads to P53 degradation, cell cycle reentry, and G2/M arrest of del(5q) cells
124,125
. In keeping with this model,
resistance to LEN has been shown to be associated with restoration of P53 accumulation, and
ACCEPTED MANUSCRIPT inhibition of TP53 expression by an antisense oligonucleotide modestly enhances del(5q) erythropoiesis
126
. Finally, sub-clonal TP53 mutations are associated to lower rates of cytogenetic
responses to LEN, though they do not influence the restoration of erythropoiesis 127. A second, not mutually exclusive mechanism has been reported, so far only in abstract form
128
. In
this study, high throughput proteomic screening identified CSNK1A1 as targeted for degradation by ubiquitination by the cereblon complex upon LEN treatment. The cellular consequences of CSNK1A1 degradation remain to be determined but preliminary data suggest that it induces a p21- and p53mediated response, in keeping with previous observations on the role of TP53 in resistance to LEN 128
. Because these mechanisms require happloinsufficiency of PP2A, CDC25C, or CSNK1A1, they can
account for the selective kill of del(5q) MDS progenitors compared to non-del(5q) progenitors induced by LEN, in keeping with the important rate of cytogenetic response noted in these patients, and with the finding that LEN-induced hematological ‘toxicity’ is correlated with achievement of response. LEN hematological ‘toxicity’ likely reflects suppression of clonal hematopoiesis
129
.
However, LEN does not seem to eradicate MDS stem cells, as demonstrated in elegant studies combining detailed immunophenotyping and FISH
130
. This finding can be reconciled with the
observation that CSNK1A1 haploinsufficiency drives clonal dominance of del(5q) cells, because the same study found that complete abrogation of Csnk1a1 in mice, mimicking the potential action of LEN on del(5q) HSC, conversely impairs HSC self-renewal
121
.
There has been initial controversy as to whether LEN treatment accelerates progression of 5qsyndromes to higher-risk MDS or AML, reports that may have been obscured by the greater propensity of progression in the heavily transfused del(5q) MDS enrolled in the LEN trials
131,132
.
Achievement of erythroid response with LEN prolongs survival, and may even delay progression to AML
114
. Detailed analyses on a limited number of patients suggest that LEN does not prevent the
accumulation of driver mutations in del(5q) stem cells 56. It will be necessary to better understand the selection pressure that LEN exerts on the subclonal TP53 mutations found in ~20% of lower-risk del(5q) MDS patients that often pre-date LEN exposure and confer a poor prognosis 133. High dose LEN induces some CR in higher-risk MDS and AML with del(5q)
134,135
. In higher-risk MDS
and AML, del(5q) is larger, frequently associated to a complex karyotype, and detailed genetic studies have identified an additional CDR in those patients
136
. Whether PP2A, CDC25C or CSKN1A1
are involved in these responses remains to be determined. Pretreatment of AML cells with LEN has been shown to increase their chemosensitivity through upregulation of mir-181a 137, in keeping with preliminary clinical reports of LEN-chemotherapy combinations 138.
ACCEPTED MANUSCRIPT Non-cell autonomous mechanisms of LEN therapy on MDS clones are also under investigation. Preliminary reports suggest that LEN has a modest effect on MDS MSC
139
. Thalidomide derivatives
including LEN have important immunomodulatory properties (thus their denomination as IMiDs), inducing activation of effector T cells and NK cells, whereas inhibiting Tregs. Such properties may also be involved in LEN activity in MDS. Indeed, one study found that LEN can overcome anergy in MDS T cells 140.
LEN in non-del(5q) anemia LEN also induces 20-25% erythroid responses in lower-risk MDS without del(5q), including in patients resistant or unlikely to respond to EPO
141,142
. In those patients, the mechanism of action of LEN is
likely different, as no cytogenetic response has been noted in informative cases and hematological toxicity is milder than in del(5q) MDS. Only a subset of non-del(5q) MDS have reduced expression of RPS14 and there is to date limited evidence that this confers sensitivity to LEN in non-del(5q) lowerrisk MDS 143. A gene expression signature indicative of impaired erythroid differentiation was initially reported to predict LEN sensitivity in non-del(5q) patients later trials
144
, but has not received independent validation in
142
. In vitro, LEN promotes erythropoiesis and induces fetal hemoglobin expression 145. LEN
acts cell-autonomously at the CFU-E stage of differentiation, even in the absence of ribosomal stress 146
. LEN can also have non-cell autonomous effects on differentiation, notably by modulating
macrophages from the erythroblastic island
146
. EPO receptor (EpoR) signaling requires specialized
membrane microdomains called ‘lipid rafts’ as signaling scaffold, which are partly deficient in MDS erythroid progenitors. Formation of these rafts requires the activity of Rho and Rac GTPases, which can be activated by IMiDs 147. Indeed, treatment with LEN improves raft assembly, enhancing EpoR signaling and erythropoiesis in vitro 148. This model is in keeping with the finding that LEN synergizes with EPO in the treatment of non-del(5q) lower-risk MDS patients who have failed EPO alone 149. LEN activity in non del(5q) MDS is likely to be CRBN-dependent, as a polymorphism located at tyrosine 244 has been shown to predict response, independent of CRBN expression 150.
Conclusion The discovery of HMA and LEN activity in MDS owes more to serendipity and to the dedication of clinician researchers than to rational drug design. In spite of the significant advances made over the recent years, a lot remains to be done to bring together evidence from clinical and biological studies performed with these drugs and shed light on their mechanism of action, in order to allocate them
ACCEPTED MANUSCRIPT more adequately and to improve their activity. This will require relevant murine or xenotransplant models. As none of these drug classes leads to clonal eradication, which probably remains the only road to a cure in MDS, renewed efforts in understanding MDS biology, and ceaseless empirical clinical research will be required to design novel therapeutic approaches for our patients, for whom approval of HMA and LEN only represents the “end of the beginning” in the therapeutic management of MDS.
Figure Legends Figure 1: Metabolism of hypomethylating agents. hENT: equilibrative nucleoside transporters, CDA: cytidine deaminase, RNR: ribonucleotide reductase, UCK: uridine-cytidine kinase, dCK : deoxycytidine kinase. Dotted arrows indicate molecular consequences of HMA that occur at higher doses of HMA.
Figure 2: Potential Mechanisms of action of hypomethylating agents. In vitro or in vivo properties of HMA are italicized. Clinical benefits of HMA are underlined.
Figure 3: Potential Mechanisms of action of lenalidomide. Top panels summarize the presumed targets of LEN in MDS with del(5q), including induction of degradation of CSNK1A1, and inhibition of the PP2A and CDC25C phosphatases. Bottom right panel indicates the synergism of LEN with EPOR in non-del(5q) MDS, through modulation of lipid rafts, and direct modulation of erythropoiesis genes. Bottom left panel recapitulates the cellular consequences of these mechanisms on del(5q) hematopoiesis (inhibition of MDS stem cell self-renewal, eradication of clonal progenitors), and on non-del(5q) MDS progenitors (promotion of erythroid differentiation).
ACCEPTED MANUSCRIPT References 1. Ades L, Itzykson R, Fenaux P. Myelodysplastic syndromes. Lancet. 2014 Jun 28;383(9936):2239-52. 2. Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Sole F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012 Sep 20;120(12):2454-65. 3. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012 Jul;13(7):484-92. 4. Sanyal A, Lajoie BR, Jain G, Dekker J. The long-range interaction landscape of gene promoters. Nature. 2012 Sep 6;489(7414):109-13. 5. Quesnel B, Guillerm G, Vereecque R, Wattel E, Preudhomme C, Bauters F, et al. Methylation of the p15(INK4b) gene in myelodysplastic syndromes is frequent and acquired during disease progression. Blood. 1998 Apr 15;91(8):2985-90. 6. Lund K, Cole JJ, VanderKraats ND, McBryan T, Pchelintsev NA, Clark W, et al. DNMT inhibitors reverse a specific signature of aberrant promoter DNA methylation and associated gene silencing in AML. Genome Biol. 2014;15(8):406. 7. Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, et al. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. The Journal of clinical investigation. 2015 Mar 30. 8. 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 Aug 3. 9. Jiang Y, Dunbar A, Gondek LP, Mohan S, Rataul M, O'Keefe C, et al. Aberrant DNA methylation is a dominant mechanism in MDS progression to AML. Blood. 2009 Feb 5;113(6):131525. 10. Will B, Zhou L, Vogler TO, Ben-Neriah S, Schinke C, Tamari R, et al. Stem and progenitor cells in myelodysplastic syndromes show aberrant stage-specific expansion and harbor genetic and epigenetic alterations. Blood. 2012 Sep 6;120(10):2076-86. 11. Perez C, Martinez-Calle N, Martin-Subero JI, Segura V, Delabesse E, Fernandez-Mercado M, et al. TET2 mutations are associated with specific 5-methylcytosine and 5-hydroxymethylcytosine profiles in patients with chronic myelomonocytic leukemia. PLoS One. 2012;7(2):e31605. 12. Beerman I, Bock C, Garrison BS, Smith ZD, Gu H, Meissner A, et al. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell. 2013 Apr 4;12(4):413-25. 13. Quivoron C, Couronne L, Della Valle V, Lopez CK, Plo I, Wagner-Ballon O, et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell. 2011 Jul 12;20(1):25-38. 14. Moran-Crusio K, Reavie L, Shih A, Abdel-Wahab O, Ndiaye-Lobry D, Lobry C, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011 Jul 12;20(1):11-24. 15. Genovese G, Kahler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014 Dec 25;371(26):2477-87. 16. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 2014 Dec 25;371(26):2488-98. 17. Bocker MT, Tuorto F, Raddatz G, Musch T, Yang FC, Xu M, et al. Hydroxylation of 5methylcytosine by TET2 maintains the active state of the mammalian HOXA cluster. Nat Commun. 2012;3:818. 18. Silverman LR, McKenzie DR, Peterson BL, Holland JF, Backstrom JT, Beach CL, et al. Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B. J Clin Oncol. 2006 Aug 20;24(24):3895-903.
ACCEPTED MANUSCRIPT 19. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009 Mar;10(3):223-32. 20. Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M, DiPersio J, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006 Apr 15;106(8):1794-803. 21. Lubbert M, Suciu S, Baila L, Ruter BH, Platzbecker U, Giagounidis A, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J Clin Oncol. 2011 May 20;29(15):1987-96. 22. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Gattermann N, Germing U, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. J Clin Oncol. 2010;28(4):562-9. 23. Silverman LR, Fenaux P, Mufti GJ, Santini V, Hellstrom-Lindberg E, Gattermann N, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011 Jun 15;117(12):2697-702. 24. Cabrero M, Jabbour E, Ravandi F, Bohannan Z, Pierce S, Kantarjian HM, et al. Discontinuation of hypomethylating agent therapy in patients with myelodysplastic syndromes or acute myelogenous leukemia in complete remission or partial response: Retrospective analysis of survival after long-term follow-up. Leuk Res. 2015 May;39(5):520-4. 25. Gore SD, Fenaux P, Santini V, Bennett JM, Silverman LR, Seymour JF, et al. A multivariate analysis of the relationship between response and survival among patients with higher-risk myelodysplastic syndromes treated within azacitidine or conventional care regimens in the randomized AZA-001 trial. Haematologica. 2013 Jul;98(7):1067-72. 26. Itzykson R, Thepot S, Quesnel B, Dreyfus F, Beyne-Rauzy O, Turlure P, et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood. 2011 Jan 13;117(2):403-11. 27. Sebert M, Komrokji RS, Sekeres MA, Prebet T, Cluzeau T, Santini V, et al. Impact Of Cytogenetics and Cytogenetic Response On Outcome In Myelodysplastic Syndromes (MDS) treated With Azacitidine (AZA). A Collaborative Study In 878 Patients. ASH Annual Meeting Abstracts. 2013;submitted. 28. Prebet T, Gore SD, Esterni B, Gardin C, Itzykson R, Thepot S, et al. Outcome of high-risk myelodysplastic syndrome after azacitidine treatment failure. J Clin Oncol. 2011 Aug 20;29(24):33227. 29. Ghoshal K, Datta J, Majumder S, Bai S, Kutay H, Motiwala T, et al. 5-Aza-deoxycytidine induces selective degradation of DNA methyltransferase 1 by a proteasomal pathway that requires the KEN box, bromo-adjacent homology domain, and nuclear localization signal. Mol Cell Biol. 2005 Jun;25(11):4727-41. 30. Datta J, Ghoshal K, Motiwala T, Jacob ST. Novel Insights into the Molecular Mechanism of Action of DNA Hypomethylating Agents: Role of Protein Kinase C delta in Decitabine-Induced Degradation of DNA Methyltransferase 1. Genes Cancer. 2012 Jan;3(1):71-81. 31. Qin T, Jelinek J, Si J, Shu J, Issa JP. Mechanisms of resistance to 5-aza-2'-deoxycytidine in human cancer cell lines. Blood. 2008 Oct 17. 32. Qin T, Castoro R, El Ahdab S, Jelinek J, Wang X, Si J, et al. Mechanisms of resistance to decitabine in the myelodysplastic syndrome. PLoS One. 2011;6(8):e23372. 33. Valencia A, Masala E, Rossi A, Martino A, Sanna A, Buchi F, et al. Expression of nucleosidemetabolizing enzymes in myelodysplastic syndromes and modulation of response to azacitidine. Leukemia. 2014 Mar;28(3):621-8.
ACCEPTED MANUSCRIPT 34. Damaraju VL, Mowles D, Yao S, Ng A, Young JD, Cass CE, et al. Role of human nucleoside transporters in the uptake and cytotoxicity of azacitidine and decitabine. Nucleosides Nucleotides Nucleic Acids. 2012;31(3):236-55. 35. Wu P, Geng S, Weng J, Deng C, Lu Z, Luo C, et al. The hENT1 and DCK genes underlie the decitabine response in patients with myelodysplastic syndrome. Leuk Res. 2015 Feb;39(2):216-20. 36. Desjobert C, El Mai M, Gerard-Hirne T, Guianvarc'h D, Carrier A, Pottier C, et al. Combined analysis of DNA methylation and cell cycle in cancer cells. Epigenetics. 2015;10(1):82-91. 37. Hollenbach PW, Nguyen AN, Brady H, Williams M, Ning Y, Richard N, et al. A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS One. 2010;5(2):e9001. 38. Schaefer M, Hagemann S, Hanna K, Lyko F. Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Cancer Res. 2009 Oct 15;69(20):8127-32. 39. Aimiuwu J, Wang H, Chen P, Xie Z, Wang J, Liu S, et al. RNA-dependent inhibition of ribonucleotide reductase is a major pathway for 5-azacytidine activity in acute myeloid leukemia. Blood. 2012 May 31;119(22):5229-38. 40. Bhuvanagiri M, Lewis J, Putzker K, Becker JP, Leicht S, Krijgsveld J, et al. 5-azacytidine inhibits nonsense-mediated decay in a MYC-dependent fashion. EMBO Mol Med. 2014 Dec;6(12):1593-609. 41. Chesnais V, Kosmider O, Damm F, Itzykson R, Bernard OA, Solary E, et al. Spliceosome mutations in myelodysplastic syndromes and chronic myelomonocytic leukemia. Oncotarget. 2012 Nov;3(11):1284-93. 42. Khoddami V, Cairns BR. Identification of direct targets and modified bases of RNA cytosine methyltransferases. Nat Biotechnol. 2013 May;31(5):458-64. 43. Thepot S, Lainey E, Cluzeau T, Sebert M, Leroy C, Ades L, et al. Hypomethylating agents reactivate FOXO3A in acute myeloid leukemia. Cell Cycle. 2011 Jul 15;10(14):2323-30. 44. Orta ML, Hoglund A, Calderon-Montano JM, Dominguez I, Burgos-Moron E, Visnes T, et al. The PARP inhibitor Olaparib disrupts base excision repair of 5-aza-2'-deoxycytidine lesions. Nucleic Acids Res. 2014 Aug;42(14):9108-20. 45. Zhang X, Li B, de Jonge N, Bjorkholm M, Xu D. The DNA methylation inhibitor induces telomere dysfunction and apoptosis of leukemia cells that is attenuated by telomerase overexpression. Oncotarget. 2015 Mar 10;6(7):4888-900. 46. Garcia-Manero G, Gore SD, Cogle C, Ward R, Shi T, Macbeth KJ, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol. 2011 Jun 20;29(18):2521-7. 47. Loughery JE, Dunne PD, O'Neill KM, Meehan RR, McDaid JR, Walsh CP. DNMT1 deficiency triggers mismatch repair defects in human cells through depletion of repair protein levels in a process involving the DNA damage response. Hum Mol Genet. 2011 Aug 15;20(16):3241-55. 48. Klco JM, Spencer DH, Lamprecht TL, Sarkaria SM, Wylie T, Magrini V, et al. Genomic impact of transient low-dose decitabine treatment on primary AML cells. Blood. 2013 Feb 28;121(9):1633-43. 49. Flotho C, Claus R, Batz C, Schneider M, Sandrock I, Ihde S, et al. The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells. Leukemia. 2009 Jun;23(6):1019-28. 50. Wong YF, Jakt LM, Nishikawa S. Prolonged Treatment with DNMT Inhibitors Induces Distinct Effects in Promoters and Gene-Bodies. PLoS One. 2013;8(8):e71099. 51. Fandy TE, Herman JG, Kerns P, Jiemjit A, Sugar EA, Choi SH, et al. Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood. 2009;114(13):2764-73. 52. Curik N, Burda P, Vargova K, Pospisil V, Belickova M, Vlckova P, et al. 5-azacitidine in aggressive myelodysplastic syndromes regulates chromatin structure at PU.1 gene and cell differentiation capacity. Leukemia. 2012 Aug;26(8):1804-11. 53. Pandiyan K, You JS, Yang X, Dai C, Zhou XJ, Baylin SB, et al. Functional DNA demethylation is accompanied by chromatin accessibility. Nucleic Acids Res. 2013 Apr;41(7):3973-85.
ACCEPTED MANUSCRIPT 54. Cluzeau T, Robert G, Mounier N, Karsenti JM, Dufies M, Puissant A, et al. BCL2L10 is a predictive factor for resistance to azacitidine in MDS and AML patients. Oncotarget. 2012 Apr;3(4):490-501. 55. Cluzeau T, Robert G, Puissant A, Jean-Michel K, Cassuto JP, Raynaud S, et al. Azacitidineresistant SKM1 myeloid cells are defective for AZA-induced mitochondrial apoptosis and autophagy. Cell Cycle. 2011 Jul 15;10(14):2339-43. 56. Woll PS, Kjallquist U, Chowdhury O, Doolittle H, Wedge DC, Thongjuea S, et al. Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo. Cancer Cell. 2014 Jun 16;25(6):794-808. 57. Tsai HC, Li H, Van Neste L, Cai Y, Robert C, Rassool FV, et al. Transient Low Doses of DNADemethylating Agents Exert Durable Antitumor Effects on Hematological and Epithelial Tumor Cells. Cancer Cell. 2012 Mar 20;21(3):430-46. 58. Ng KP, Ebrahem Q, Negrotto S, Mahfouz RZ, Link KA, Hu Z, et al. p53 independent epigeneticdifferentiation treatment in xenotransplant models of acute myeloid leukemia. Leukemia. 2011 Nov;25(11):1739-50. 59. Trowbridge JJ, Sinha AU, Zhu N, Li M, Armstrong SA, Orkin SH. Haploinsufficiency of Dnmt1 impairs leukemia stem cell function through derepression of bivalent chromatin domains. Genes Dev. 2012 Feb 15;26(4):344-9. 60. Araki H, Yoshinaga K, Boccuni P, Zhao Y, Hoffman R, Mahmud N. Chromatin-modifying agents permit human hematopoietic stem cells to undergo multiple cell divisions while retaining their repopulating potential. Blood. 2007 Apr 15;109(8):3570-8. 61. Hu Z, Negrotto S, Gu X, Mahfouz R, Ng KP, Ebrahem Q, et al. Decitabine maintains hematopoietic precursor self-renewal by preventing repression of stem cell genes by a differentiation-inducing stimulus. Mol Cancer Ther. 2010 Jun;9(6):1536-43. 62. Trowbridge JJ, Snow JW, Kim J, Orkin SH. DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell. 2009 Oct 2;5(4):442-9. 63. Itzykson R, Kosmider O, Cluzeau T, Mansat-De Mas V, Dreyfus F, Beyne-Rauzy O, et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias. Leukemia. 2011 Jul;25(7):1147-52. 64. Traina F, Visconte V, Elson P, Tabarroki A, Jankowska AM, Hasrouni E, et al. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia. 2013 Sep 18. 65. Bejar R, Lord A, Stevenson K, Bar-Natan M, Perez-Ladaga A, Zaneveld J, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014 Oct 23;124(17):2705-12. 66. Craddock C, Quek L, Goardon N, Freeman S, Siddique S, Raghavan M, et al. Azacitidine fails to eradicate leukemic stem/progenitor cell populations in patients with acute myeloid leukemia and myelodysplasia. Leukemia. 2012 Nov 6. 67. Lubbert M, Daskalakis M, Kunzmann R, Engelhardt M, Guo Y, Wijermans P. Nonclonal neutrophil responses after successful treatment of myelodysplasia with low-dose 5-aza-2'deoxycytidine (decitabine). Leuk Res. 2004 Dec;28(12):1267-71. 68. Steinmann J, Bertz H, Wasch R, Marks R, Zeiser R, Bogatyreva L, et al. 5-Azacytidine and DLI can induce long-term remissions in AML patients relapsed after allograft. Bone Marrow Transplant. 2015 May;50(5):690-5. 69. Negrotto S, Ng KP, Jankowska AM, Bodo J, Gopalan B, Guinta K, et al. CpG methylation patterns and decitabine treatment response in acute myeloid leukemia cells and normal hematopoietic precursors. Leukemia. 2012 Feb;26(2):244-54. 70. Itzykson R, Thepot S, Beyne-Rauzy O, Ame S, Isnard F, Dreyfus F, et al. Does addition of erythropoiesis stimulating agents improve the outcome of higher-risk myelodysplastic syndromes treated with azacitidine? Leuk Res. 2012 Apr;36(4):397-400.
ACCEPTED MANUSCRIPT 71. Schmelz K, Wagner M, Dorken B, Tamm I. 5-Aza-2'-deoxycytidine induces p21WAF expression by demethylation of p73 leading to p53-independent apoptosis in myeloid leukemia. Int J Cancer. 2005 May 1;114(5):683-95. 72. Bally C, Ades L, Renneville A, Sebert M, Eclache V, Preudhomme C, et al. Prognostic value of TP53 gene mutations in myelodysplastic syndromes and acute myeloid leukemia treated with azacitidine. Leuk Res. 2014 Jul;38(7):751-5. 73. Muller-Thomas C, Rudelius M, Rondak IC, Haferlach T, Schanz J, Huberle C, et al. Response to azacitidine is independent of p53 expression in higher-risk myelodysplastic syndromes and secondary acute myeloid leukemia. Haematologica. 2014 Oct;99(10):e179-81. 74. 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 Jun 30;364(26):2496506. 75. Mufti GJ, Gore SD, Santini V, Fenaux P, Silverman LR, Hagemeijer A, et al. Influence of Karyotype On Overall Survival in Patients with Higher-Risk Myelodysplastic Syndrome Treated with Azacitidine or a Conventional Care Regimen. 2009. p. 1755-. 76. Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 2010 Apr 8;464(7290):852-7. 77. Medyouf H, Mossner M, Jann JC, Nolte F, Raffel S, Herrmann C, et al. Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit. Cell Stem Cell. 2014 Jun 5;14(6):824-37. 78. Pilorge S, Doleris LM, Dreyfus F, Park S. The autoimmune manifestations associated with myelodysplastic syndrome respond to 5-azacytidine: a report on three cases. Br J Haematol. 2011 Jun;153(5):664-5. 79. Schroeder T, Czibere A, Platzbecker U, Bug G, Uharek L, Luft T, et al. Azacitidine and donor lymphocyte infusions as first salvage therapy for relapse of AML or MDS after allogeneic stem cell transplantation. Leukemia. 2013 Jun;27(6):1229-35. 80. Choi J, Ritchey J, Prior JL, Holt M, Shannon WD, Deych E, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood. 2010 Jul 8;116(1):129-39. 81. Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, Caballero-Velazquez T, Blanco B, Herrero-Sanchez C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010 Jan 7;115(1):107-21. 82. Schroeder T, Frobel J, Cadeddu RP, Czibere A, Dienst A, Platzbecker U, et al. Salvage therapy with azacitidine increases regulatory T cells in peripheral blood of patients with AML or MDS and early relapse after allogeneic blood stem cell transplantation. Leukemia. 2013 Sep;27(9):1910-3. 83. Goodyear OC, Dennis M, Jilani NY, Loke J, Siddique S, Ryan G, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012 Apr 5;119(14):3361-9. 84. Costantini B, Kordasti S, Kulasekararaj AG, Jiang J, Seidl T, Perez Abellan P, et al. The effects of 5-azacytidine on the function and number of regulatory T-cells and T-effectors in myelodysplastic syndrome. Haematologica. 2012 Dec 14. 85. Hu Y, Cui Q, Gu Y, Sheng L, Wu K, Shi J, et al. Decitabine facilitates the generation and immunosuppressive function of regulatory gammadeltaT cells derived from human peripheral blood mononuclear cells. Leukemia. 2013 Jul;27(7):1580-5. 86. Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G, et al. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010;116(11):1908-18. 87. Srivastava P, Paluch BE, Matsuzaki J, James SR, Collamat-Lai G, Karbach J, et al. Immunomodulatory action of SGI-110, a hypomethylating agent, in acute myeloid leukemia cells and xenografts. Leuk Res. 2014 Nov;38(11):1332-41.
ACCEPTED MANUSCRIPT 88. Orskov AD, Treppendahl MB, Skovbo A, Holm MS, Friis LS, Hokland M, et al. Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation. Oncotarget. 2015 Apr 20;6(11):9612-26. 89. Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014 Jun;28(6):1280-8. 90. Kantarjian H, Oki Y, Garcia-Manero G, Huang X, O'Brien S, Cortes J, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007 Jan 1;109(1):52-7. 91. Shen L, Kantarjian H, Guo Y, Lin E, Shan J, Huang X, et al. DNA methylation predicts survival and response to therapy in patients with myelodysplastic syndromes. J Clin Oncol. 2010;28(4):60513. 92. Blum W, Garzon R, Klisovic RB, Schwind S, Walker A, Geyer S, et al. Clinical response and miR29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci U S A. 2010;107(16):7473-8. 93. Yun H, Damm F, Yap D, Schwarzer A, Chaturvedi A, Jyotsana N, et al. Impact of MLL5 expression on decitabine efficacy and DNA methylation in acute myeloid leukemia. Haematologica. 2014 Sep;99(9):1456-64. 94. Metzeler KH, Walker A, Geyer S, Garzon R, Klisovic RB, Bloomfield CD, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia. 2012 May;26(5):1106-7. 95. Braun T, Itzykson R, Renneville A, de Renzis B, Dreyfus F, Laribi K, et al. Molecular predictors of response to decitabine in advanced chronic myelomonocytic leukemia: a phase II trial. Blood. 2011 Aug 9;118:3824-31. 96. Kosmider O, Gelsi-Boyer V, Cheok M, Grabar S, Della-Valle V, Picard F, et al. TET2 mutation is an independent favorable prognostic factor in myelodysplastic syndromes (MDS). Blood. 2009 Aug 7. 97. Saunthararajah Y, Sekeres M, Advani A, Mahfouz R, Durkin L, Radivoyevitch T, et al. Evaluation of noncytotoxic DNMT1-depleting therapy in patients with myelodysplastic syndromes. The Journal of clinical investigation. 2015 Mar 2;125(3):1043-55. 98. Foulks JM, Parnell KM, Nix RN, Chau S, Swierczek K, Saunders M, et al. Epigenetic drug discovery: targeting DNA methyltransferases. J Biomol Screen. 2012 Jan;17(1):2-17. 99. Mims A, Walker AR, Huang X, Sun J, Wang H, Santhanam R, et al. Increased anti-leukemic activity of decitabine via AR-42-induced upregulation of miR-29b: a novel epigenetic-targeting approach in acute myeloid leukemia. Leukemia. 2013 Apr;27(4):871-8. 100. Prebet T, Sun Z, Figueroa ME, Ketterling R, Melnick A, Greenberg PL, et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup trial E1905. J Clin Oncol. 2014 Apr 20;32(12):1242-8. 101. Gore SD, Baylin S, Sugar E, Carraway H, Miller CB, Carducci M, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res. 2006 Jun 15;66(12):6361-9. 102. Han H, Yang X, Pandiyan K, Liang G. Synergistic re-activation of epigenetically silenced genes by combinatorial inhibition of DNMTs and LSD1 in cancer cells. PLoS One. 2013;8(9):e75136. 103. Karoopongse E, Yeung C, Byon J, Ramakrishnan A, Holman ZJ, Jiang PY, et al. The KDM2B- let7b -EZH2 axis in myelodysplastic syndromes as a target for combined epigenetic therapy. PLoS One. 2014;9(9):e107817. 104. Soriano AO, Yang H, Faderl S, Estrov Z, Giles F, Ravandi F, et al. Safety and clinical activity of the combination of 5-azacytidine, valproic acid, and all-trans retinoic acid in acute myeloid leukemia and myelodysplastic syndrome. Blood. 2007 Oct 1;110(7):2302-8. 105. Raffoux E, Cras A, Recher C, Boelle PY, de Labarthe A, Turlure P, et al. Phase 2 clinical trial of 5-azacitidine, valproic acid, and all-trans retinoic acid in patients with high-risk acute myeloid leukemia or myelodysplastic syndrome. Oncotarget. 2010;1(1):34-42.
ACCEPTED MANUSCRIPT 106. Thomas S, Schelker R, Klobuch S, Zaiss S, Troppmann M, Rehli M, et al. Biomodulatory therapy induces complete molecular remission in chemorefractory acute myeloid leukemia. Haematologica. 2015 Jan;100(1):e4-6. 107. Platzbecker U, Germing U. Combination of Azacitidine and Lenalidomide in Myelodysplastic Syndromes or acute Myeloid Leukemia-a wise Liaison? Leukemia. 2013 Sep;27(9):1813-9. 108. Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, et al. Identification of a primary target of thalidomide teratogenicity. Science. 2010 Mar 12;327(5971):1345-50. 109. Kronke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 2014 Jan 17;343(6168):301-5. 110. Fischer ES, Bohm K, Lydeard JR, Yang H, Stadler MB, Cavadini S, et al. Structure of the DDB1CRBN E3 ubiquitin ligase in complex with thalidomide. Nature. 2014 Aug 7;512(7512):49-53. 111. List A, Kurtin S, Roe DJ, Buresh A, Mahadevan D, Fuchs D, et al. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med. 2005 Feb 10;352(6):549-57. 112. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med. 2006 Oct 5;355(14):1456-65. 113. Fenaux P, Giagounidis A, Selleslag D, Beyne-Rauzy O, Mufti G, Mittelman M, et al. A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with del5q. Blood. 2011 Oct 6;118(14):3765-76. 114. List AF, Bennett JM, Sekeres MA, Skikne B, Fu T, Shammo JM, et al. Extended survival and reduced risk of AML progression in erythroid-responsive lenalidomide-treated patients with lowerrisk del(5q) MDS. Leukemia. 2014 May;28(5):1033-40. 115. Giagounidis A, Mufti GJ, Mittelman M, Sanz G, Platzbecker U, Muus P, et al. Outcomes in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with isolated deletion 5q treated with lenalidomide: a subset analysis from the MDS-004 study. Eur J Haematol. 2014 Nov;93(5):429-38. 116. Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, et al. Identification of RPS14 as a 5qsyndrome gene by RNA interference screen. Nature. 2008 Jan 17;451(7176):335-9. 117. Barlow JL, Drynan LF, Hewett DR, Holmes LR, Lorenzo-Abalde S, Lane AL, et al. A p53dependent mechanism underlies macrocytic anemia in a mouse model of human 5q- syndrome. Nat Med. 2010;16(1):59-66. 118. Pellagatti A, Marafioti T, Paterson JC, Barlow JL, Drynan LF, Giagounidis A, et al. Induction of p53 and up-regulation of the p53 pathway in the human 5q- syndrome. Blood. 2010 Apr 1;115(13):2721-3. 119. Starczynowski DT, Kuchenbauer F, Argiropoulos B, Sung S, Morin R, Muranyi A, et al. Identification of miR-145 and miR-146a as mediators of the 5q- syndrome phenotype. Nat Med. 2010;16(1):49-58. 120. Pellagatti A, Jadersten M, Forsblom AM, Cattan H, Christensson B, Emanuelsson EK, et al. Lenalidomide inhibits the malignant clone and up-regulates the SPARC gene mapping to the commonly deleted region in 5q- syndrome patients. Proc Natl Acad Sci U S A. 2007;104(27):1140611. 121. Schneider RK, Adema V, Heckl D, Jaras M, Mallo M, Lord AM, et al. Role of casein kinase 1A1 in the biology and targeted therapy of del(5q) MDS. Cancer Cell. 2014 Oct 13;26(4):509-20. 122. Oliva EN, Cuzzola M, Nobile F, Ronco F, D'Errigo MG, Lagana C, et al. Changes in RPS14 expression levels during lenalidomide treatment in Low- and Intermediate-1-risk myelodysplastic syndromes with chromosome 5q deletion. Eur J Haematol. 2010 Sep;85(3):231-5. 123. Venner CP, Woltosz JW, Nevill TJ, Deeg HJ, Caceres G, Platzbecker U, et al. Correlation of clinical response and response duration with miR-145 induction by lenalidomide in CD34(+) cells from patients with del(5q) myelodysplastic syndrome. Haematologica. 2013 Mar;98(3):409-13.
ACCEPTED MANUSCRIPT 124. Wei S, Chen X, Rocha K, Epling-Burnette PK, Djeu JY, Liu Q, et al. A critical role for phosphatase haplodeficiency in the selective suppression of deletion 5q MDS by lenalidomide. Proc Natl Acad Sci U S A. 2009 May 26. 125. Wei S, Chen X, McGraw K, Zhang L, Komrokji R, Clark J, et al. Lenalidomide promotes p53 degradation by inhibiting MDM2 auto-ubiquitination in myelodysplastic syndrome with chromosome 5q deletion. Oncogene. 2013 Feb 28;32(9):1110-20. 126. Caceres G, McGraw K, Yip BH, Pellagatti A, Johnson J, Zhang L, et al. TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomideresistant patients. Proc Natl Acad Sci U S A. 2013 Oct 1;110(40):16127-32. 127. Saft L, Karimi M, Ghaderi M, Matolcsy A, Mufti GJ, Kulasekararaj A, et al. p53 protein expression independently predicts outcome in patients with lower-risk myelodysplastic syndromes with del(5q). Haematologica. 2014 Jun;99(6):1041-9. 128. Fink EC, Krönke J, Hurst SN, Udeshi ND, Svinkina T, Schneider RK, et al. Lenalidomide Induces Ubiquitination and Degradation of CSNK1A1 in MDS with Del(5q)2014. 129. Sekeres MA, Maciejewski JP, Giagounidis AA, Wride K, Knight R, Raza A, et al. Relationship of treatment-related cytopenias and response to lenalidomide in patients with lower-risk myelodysplastic syndromes. J Clin Oncol. 2008;26(36):5943-9. 130. Tehranchi R, Woll PS, Anderson K, Buza-Vidas N, Mizukami T, Mead AJ, et al. Persistent malignant stem cells in del(5q) myelodysplasia in remission. N Engl J Med. 2010 Sep 9;363(11):102537. 131. Ades L, Le Bras F, Sebert M, Kelaidi C, Lamy T, Dreyfus F, et al. Treatment with lenalidomide does not appear to increase the risk of progression in lower risk myelodysplastic syndromes with 5q deletion. A comparative analysis by the Groupe Francophone des Myelodysplasies. Haematologica. 2012 Feb;97(2):213-8. 132. Kuendgen A, Lauseker M, List AF, Fenaux P, Giagounidis AA, Brandenburg NA, et al. Lenalidomide does not increase AML progression risk in RBC transfusion-dependent patients with Low- or Intermediate-1-risk MDS with del(5q): a comparative analysis. Leukemia. 2013 Apr;27(5):1072-9. 133. Jadersten M, Saft L, Smith A, Kulasekararaj A, Pomplun S, Gohring G, et al. TP53 mutations in low-risk myelodysplastic syndromes with del(5q) predict disease progression. J Clin Oncol. 2011 May 20;29(15):1971-9. 134. Sekeres MA, Gundacker H, Lancet J, Advani A, Petersdorf S, Liesveld J, et al. A phase 2 study of lenalidomide monotherapy in patients with deletion 5q acute myeloid leukemia: Southwest Oncology Group Study S0605. Blood. 2011 Jul 21;118(3):523-8. 135. Fehniger TA, Uy GL, Trinkaus K, Nelson AD, Demland J, Abboud CN, et al. A phase 2 study of high-dose lenalidomide as initial therapy for older patients with acute myeloid leukemia. Blood. 2011 Feb 10;117(6):1828-33. 136. Jerez A, Gondek LP, Jankowska AM, Makishima H, Przychodzen B, Tiu RV, et al. Topography, clinical, and genomic correlates of 5q myeloid malignancies revisited. J Clin Oncol. 2012 Apr 20;30(12):1343-9. 137. Hickey CJ, Schwind S, Radomska HS, Dorrance AM, Santhanam R, Mishra A, et al. Lenalidomide-mediated enhanced translation of C/EBPalpha-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia. Blood. 2013 Jan 3;121(1):159-69. 138. Ades L, Prebet T, Stamatoullas A, Recher C, Guieze R, Raffoux E, et al. Lenalidomide (LEN) Combined To Intensive Chemotherapy (IC) In AML and Higher Risk MDS With Del 5q. Results Of a Phase I/II Study Of The Groupe Francophone Des Myelodysplasies (GFM)2013. 139. Ferrer RA, Wobus M, List C, Wehner R, Schonefeldt C, Brocard B, et al. Mesenchymal stromal cells from patients with myelodyplastic syndrome display distinct functional alterations that are modulated by lenalidomide. Haematologica. 2013 Nov;98(11):1677-85. 140. McDaniel JM, Zou JX, Fulp W, Chen DT, List AF, Epling-Burnette PK. Reversal of T-cell tolerance in myelodysplastic syndrome through lenalidomide immune modulation. Leukemia. 2012 Jun;26(6):1425-9.
ACCEPTED MANUSCRIPT 141. Raza A, Reeves JA, Feldman EJ, Dewald GW, Bennett JM, Deeg HJ, et al. Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate-1 risk myelodysplastic syndromes with karyotypes other than deletion 5q. Blood. 2008 Jan 1;111(1):86-93. 142. Santini V, Almeida A, Giagounidis A, Gröpper S, Jonasova A, Vey N, et al. Efficacy and Safety of Lenalidomide (LEN) Versus Placebo (PBO) in RBC-Transfusion Dependent (TD) Patients (Pts) with IPSS Low/Intermediate (Int-1)-Risk Myelodysplastic Syndromes (MDS) without Del(5q) and Unresponsive or Refractory to Erythropoiesis-Stimulating Agents (ESAs): Results from a Randomized Phase 3 Study (CC-5013-MDS-005). ASH Annual Meeting 2014. Abstract #409 143. Wu L, Li X, Xu F, Zhang Z, Chang C, He Q. Low RPS14 expression in MDS without 5q aberration confers higher apoptosis rate of nucleated erythrocytes and predicts prolonged survival and possible response to lenalidomide in lower risk non-5q- patients. Eur J Haematol. 2013 Jun;90(6):486-93. 144. Ebert BL, Galili N, Tamayo P, Bosco J, Mak R, Pretz J, et al. An erythroid differentiation signature predicts response to lenalidomide in myelodysplastic syndrome. PLoS Med. 2008 Feb;5(2):e35. 145. Moutouh-de Parseval LA, Verhelle D, Glezer E, Jensen-Pergakes K, Ferguson GD, Corral LG, et al. Pomalidomide and lenalidomide regulate erythropoiesis and fetal hemoglobin production in human CD34+ cells. The Journal of clinical investigation. 2008 Jan;118(1):248-58. 146. Narla A, Dutt S, McAuley JR, Al-Shahrour F, Hurst S, McConkey M, et al. Dexamethasone and lenalidomide have distinct functional effects on erythropoiesis. Blood. 2011 Aug 25;118(8):2296-304. 147. Xu Y, Li J, Ferguson GD, Mercurio F, Khambatta G, Morrison L, et al. Immunomodulatory drugs reorganize cytoskeleton by modulating Rho GTPases. Blood. 2009 Jul 9;114(2):338-45. 148. McGraw KL, Basiorka AA, Johnson JO, Clark J, Caceres G, Padron E, et al. Lenalidomide induces lipid raft assembly to enhance erythropoietin receptor signaling in myelodysplastic syndrome progenitors. PLoS One. 2014;9(12):e114249. 149. Toma A, Chevret S, Kosmider O, Delaunay J, Stamatoullas A, Rose C, et al. A randomized study of lenalidomide (LEN) with or without EPO in RBC transfusion dependent (TD) IPSS low and int1 (lower risk) myelodysplastic syndromes (MDS) without del 5q resistant to EPO. ASCO Meeting Abstracts. 2013 June 17, 2013;31(15_suppl):7002. 150. Sardnal V, Rouquette A, Kaltenbach S, Bally C, Chesnais V, Leschi C, et al. A G polymorphism in the CRBN gene acts as a biomarker of response to treatment with lenalidomide in low/int-1 risk MDS without del(5q). Leukemia. 2013 Jul;27(7):1610-3.
Figure 1
DAC
hENT
SC
hENT
RI PT
AZA
5-aza-CR UCK 5-aza-CMP
M AN U
5-aza-dCR dCK 5-aza-dCMP
5-aza-CDP
5-aza-dCDP
RNR
5-aza-CTP
D
dNTP pools Depletion?
AC C
EP
Impaired Protein Synthesis
Replication
TE
RNA
5-aza-dCTP
DNMT depletion
5-aza-dUTP DNA
DNA damage?
HSC
M AN U
Enhanced differentiation
MDS
Improvement of cytopenias
AML
AC C
EP
TE
D
Clonal Extinction
Delayed Progression To AML
SC
Healthy Differential Self-renewal
Precursors
RI PT
Progenitors
Enhanced Immunosurveillance
Figure 2
DDB1 CUL4CRBN
RPS14
CRBN
PP2A
CDC25C
SC
MDM2
M AN U
Ub
Ub Ub
2
D
1
EP
AC C
G2/M arrest
EpoR
2
1
HSC
P53 activation
LEN
TE
LEN del(5q)
P53
Ub
CSNK1A1
non del(5q)
LEN
P
LEN
5q32q33
RI PT
Figure 3
Lipid rafts
Precursors
Progenitors
LEN LEN 3
3
STAT HbF…
ACCEPTED MANUSCRIPT
New therapeutic approaches in myelodysplastic syndromes: hypomethylating agents and lenalidomide
Clémence Loiseau, Ashfaq Ali, Raphael Itzykson
Highlights:
•
Hypomethylating agents alter self-renewal and differentiation of MDS stem/progenitors.
•
Metabolism of hypomethylating agents and cell cycling govern the depth of hypomethylation.
•
Azacitidine incorporates into RNA and regulates protein synthesis.
•
Lenalidomide induces del(5q) MDS progenitor eradication, sparing del(5q) stem cells.