Introduction
Epigenetics is defined as heritable changes in genetic function that occur without
alteration of the underlying genetic code. Since its original description, it is now
clear that epigenetic mechanisms are vital for cellular growth, development, and establishment
of a distinct cellular identity.[1] The term “epigenome” describes the epigenetic regulatory mechanisms across the entire
genome; and unlike the genome, it varies across different cell types. Abnormalities
in epigenetic control can lead to a wide variety of cellular dysfunction, and have
been specifically found to contribute to carcinogenesis.[2] Epigenetic alterations are early events in the pathogenesis of certain cancers and
contribute to disease progression, analogous to clonal cytogenetic evolution.[3] Epigenetic modifications are of interest in several malignancies, and have been
utilized most notably for the treatment of acute myeloid leukemia in the form of hypomethylating
agents. Significant molecular data are also emerging on the role of epigenetics for
several other malignancies, and is of specific importance in multiple myeloma (MM).
MM is a common hematologic malignancy with a universal risk of relapse and has undergone
advances in survival over the past two decades.[4] However, it still carries a nearly 100% risk of relapse, necessitating newer agents
to be initiated at each instance of progression.[5] As a result, no functional cure is defined, and therapy must continue indefinitely.
Although several newer novel agents have been introduced in the past 5 years, costs
continue to be prohibitive, especially in resource constrained settings.[6] Thus, there is an ongoing need to identify newer cellular pathways and targetable
genetic lesions to ensure the availability of treatment options at each relapse.
Epigenetic mechanisms have recently emerged as critical mediators of disease pathogenesis
and progression in myeloma. Several mechanisms are linked to drug responsiveness and
known to modify survival. As seen in the case of acute myeloid leukemia (AML), several
components of epigenetic mechanisms are eminently targetable and will likely offer
an opportunity for a new approach to drug development. An added impetus for studying
epigenetic pathways would be definition of differences in disease biology based on
geographic variation. For instance, significant differences in age of incidence of
myeloma have been noted in Indian patients compared with Western data, with higher
likelihood of end organ damage at diagnosis.[7]
We provide a succinct snapshot of the current role of epigenetic mechanisms in disease
pathogenesis in MM, and highlight potential therapeutic applications. We also provide
a snapshot of potential drug targets and current ongoing trials targeting epigenetic
pathways in MM.
Summary of Basic Mechanisms of Epigenetic Control
Epigenetic control is facilitated by several biochemical processes, of which the best
understood mechanisms are (a) DNA methylation, (b) histone modification, and (c) microRNA
(miRNA) expression. These three processes are most widely described and are targets
of several investigational approaches to treatment.
DNA Methylation
DNA methylation consists of addition of a methyl group to the 5′ carbon of cytosine
on DNA. The cytosine bases that are involved in methylation are usually located in
regulatory (enhancer or repressor) sequences, and can recruit other proteins to functionally
silence downstream genes.[8] DNA methylation of promotor genes, therefore, leads to silencing of the downstream
genes regulated with the same. DNA methylation is performed by enzymes called DNA
methyl transferases (DNMT) and typically occurs at sites of CpG (cytosine-phosphate-guanine)
dinucleotides, which are concentrated in sites known as CpG islands, mostly at 5′
ends of DNA.[8] There are ~28 million CpGs in the human genome, and ~60% to 80% are methylated.
Histone Modification
Intracellular DNA is tightly wound around proteins called histones, which are octamers
consisting of eight subunits. This process is best summarized by its original definition
given in the 1960s, “dynamic and reversible mechanism for activation as well as repression
of RNA synthesis.”[9] It is known that modification of histone proteins by biochemical changes enables
regulation of DNA expression by increasing or decreasing chromatin accessibility.
The first mechanism to be elucidated was histone acetylation and deacetylation, following
which several other mechanisms have been described over the last decade.[10] As genetic regulation needs to be reversible and dynamic, most of the processes
are reversible and involve reciprocal “effector” and “eraser” enzymes. These include
acetylation, methylation, and phosphorylation, and the use of so-called “erasers,”
or enzymes with reciprocal activity to the ones above. [Fig. 1] summarizes the basic process of histone modification.
Fig. 1 Histone modifications in DNA. Histone proteins (dotted blue) are tightly wound to
DNA (green), preventing access of transcriptional machinery. Reversible binding of
covalent modifiers can occur either on the histone protein core or on one of the histone
tails (red). Binding or a modifier, for example, an acetyl group (blue), leads to
a conformational change, opening the chromatin structure and enabling access to transcriptional
machinery.
Noncoding RNA Expression
Noncoding (ncRNAs) are RNAs that are not coded into proteins. They are classified
by their size into small and large ncRNAs. The small ncRNAs include miRNA, piwi interacting
RNA, transfer RNA, and small nucleolar RNA.[11] They are involved in regulation of DNA expression by several mechanisms, out of
which epigenetic alteration is one of them. [Fig. 2] summarizes mechanisms of miRNA control with potential therapeutic interest.
Fig. 2 Summary of micro-RNA (miRNA)-mediated control of genetic expression. Starting from
lower left, RNA polymerase II encodes primary miRNA (pri-miRNA) that is converted
to pre-miRNA and exported out of the nucleus by Exportin. After cleavage by the Dicer
(helicase with RNase motif) complex, the miRNA forms an RNA-induced silencing complex
(RISC) that binds to complementary miRNA, silencing its expression. This repressor
function of miRNA against has demonstrated antimyeloma activity in vitro.
Histone Modification in Myeloma Is a Complex Multistep Process
Histone proteins are subject to several covalent posttranslational modifications,
of which over 15 have been described in detail.[10] Histone modification is reversible, and the processes involved have effector and
eraser functions with a set of reciprocally active enzymes. Acetylation, phosphorylation,
and methylation are the most well described, with the first two being clear markers
of active transcription[29] Methylation, on the other hand, is more complex, as the final effects of methylation
depend on the amino acids modified and the degree of methylation (mono, di, or tri).[30] Histone methylation is seen to occur at basic amino acids, namely arginine, histidine,
and lysine, and may have a variable effect on transcription depending on context (i.e.,
type and site of amino acid that is methylated).[31] A common nomenclature is used to define histone modifications, comprising the name
of histone protein (e.g., H2), abbreviation and position of amino acid (K or M), chemical
group added (e.g., methyl = Me or Acetyl = Ac), and copies of the modifier added (1,
2, or 3). Therefore, addition of a dimethyl group to a lysine residue in position
36 on histone protein H3 is abbreviated as H3K36Me2. This process is summarized in
[Fig. 3].
Fig. 3 Patterns of histone methylation. Methylation occurs on specific amino acids in the
histone core protein and is mediated by histone methyl transferases. Depending on
the number of methyl groups added, it can either occur as mono-, di-, or trimethylation.
Histone Dimethylation
Histone dimethylation is catalyzed by enzymes containing the SET domain with methyltransferase
activity. In this well-characterized group, mutations in MMSET, NSD1, and NSD2 are
found in several patients at diagnosis and relapse. MMSET is one of the most well
defined, and catalyzes the addition of H3K36me2, associated with active transcription.[32] It is seen to be associated with the t(4;14) translocation, leading to transcriptional
activation and is associated with a poorer outcome in patients with MM.[33] Over the past decade, there has been an increasing recognition of the biological
contribution of MMSET to pathogenesis of myeloma. MMSET is shown to activate the MAF
gene through the MAP kinase pathway, indicating indirect activation of an oncogene.
MMSET is also implicated in activation of several downstream mitogenic pathways, including
c-myc and IRF4.[34]
Histone Trimethylation
Histone trimethylation is brought about by enzymatic units of enhancer of Zeste homologue
2 (EZH1/2) complexes, which deposit H3K27me3 on target genes. EZH2 is part of the
polycomb repressor complex (PRC), which silences several downstream genes. The role
of EZH2 varies in different malignancies, and in MM it is shown to be upregulated
and correlates with a poor outcome.[35] Similar to MMSET, EZH2 overexpression induces disease progression through a variety
of mechanisms. EZH2 is shown to induce proliferation of MM cells independent of interleukin
6 (IL-6). It is also shown to control the expression of tumor suppressor miRNAs that
target c-myc, BLIMP-1, and IRF4.[36]
Histone Monomethylation
Histone monomethylation at lysine sites deposits H3K9me and is essential for gene
repression at promoter regions. H3K9 transferring enzymes are elevated in MM patients.
KDM3A leads to IRF4 supported cell growth in MM.[37]
Histone Acetylation
Histone acetylation is seen to occur at lysine residues, which opens up chromatin
and makes it transcriptionally active. This process is reversibly regulated by lysine
transferases (KATs) and deacetylases (histone deacetylase inhibitors [HDACs]).[38] Mutated KAT genes are found in several patients with MM, especially at relapse,
possibly loss of function mutations. HDACs are overexpressed in myeloma and are associated
with poor prognosis.[39]
Role of Epigenetic Mechanisms in Disease Progression and Drug Resistance
Epigenetic processes have been found to play a significant role in two important processes
mediating disease progression, that is, PC plasticity and drug resistance.
Analysis of malignant PCs in myeloma reveals a heterogeneous population comprising
PCs (CD138+/CD19-), preplasma cells (pre-PCs: CD19-/CD138-), and plasmablasts (CD19+).
A vast majority of the cells are CD19-/CD138+ and predominant mediators of disease
phenotype, including immunoglobulin secretion and organ damage. A clone of pre-PCs
with a CD19-/CD138- represents a small minority, representing a quiescent population.
This clone confers resistance to therapy and is in dynamic equilibrium with CD138+
PCs.[40]
Several characteristics of this process indicate a significant role played by epigenetic
processes. Pre-PC populations demonstrate high concentrations of several epigenetic
regulators, including PRC, MLL transcriptional activating complex, demethylases, HDACS
and KDM5C/D, indicating epigenetic control. Additionally, the absence of irreversible
genetic mutations or change in CD138 miRNA levels makes genetic level (transcriptional/
translational) control unlikely.[41] These cells are intrinsically resistant to proteasome inhibitors, and may indicate
a reservoir that can proliferate at relapse.
Additionally, epigenetic mechanisms are observed to mediate many aspects of drug resistance
in patients with myeloma. Epigenetic inactivation of RASD1 is essential in mediating
glucocorticoid resistance. Several miRNAs have been found to mediate drug resistance
through diverse mechanisms, including modulation of target genes, upregulation of
oncogenes, and modification of drug efflux mechanisms. A review by Rastgoo et al provides
a succinct overview of drug resistance mediated by miRNA.[42] The control of cell adhesion-mediated drug resistance by altering microenvironment
interactions and modification of antiapoptotic signals also have significant epigenetic
control. The description of all allied mechanisms is out of scope of this review and
is summarized elegantly by Furukawa et al.[43]
Therapeutic Applications of Epigenetic Modifications in Myeloma
All the mechanisms described above provide potential therapeutic targets. The following
discussion summarizes salient features of some of the commonly described mechanisms.
Many mechanisms have been translated to clinical applications, and a summary of recent
or ongoing trials utilizing the same is provided in [Table 1].
Table 1
A snapshot of currently ongoing clinical trials utilizing epigenetic pathways for
treatment of myeloma, predominantly in the relapsed/refractory setting
|
Mechanism targeted
|
Drug/Preclinical molecule
|
Trial phase and design
|
Salient findings
|
|
DNMT
|
Oral azacytidine[86]
|
Oral azacytidine in combination with Len/Dex for RRMM
|
ORR was 37.5%; clinical benefit rate was 50%. Median OS was 10.3 mo; median PFS was
2.6 mo
|
|
Injectable azacytidine[87]
|
Phase 1b, twice a week S/C azacytidine with Len/Dex for RRMM
|
The median PFS was 3.1 mo (95%CI: 2.1–5.1 mo), median TTP 2.7 mo (95% CI: 2.1–7.5
mo), and median OS 18.6 mo (95% CI: 12.9–33.0 mo)
|
|
HDAC
|
Panobinostat[53]
|
Phase 3, panobinostat + Vd vs. Vd alone for RRMM
|
PFS advantage, 11·99 mo (95% CI: 10·33–12·94) vs 8·08 mo (7.56–9.23); HR 0·63, 95%
CI 0.52–0.76; p < 0·0001. OS difference not yet clear
|
|
Tefinostat[88]
|
Phase 1, dose escalation trial of oral drug
|
Safety and maximal tolerated dose defined. Further clinical studies planned
|
|
Romidepsin[89]
|
Phase 2 trial in thirteen patients
|
Initial safety and clinical improvement noted, Phase 3 trials planned
|
|
|
Phase 1/2 trial in combination with Len for refractory disease
|
Active, completed recruitment December 2020. Updates available at NCT01755975
|
|
Vorinostat[90]
|
Bortezomib alone or with Vorinostat in patients with RRMM
|
Median PFS was 7·63 mo (95% CI: 6·87–8·40) in the Vorinostat group and 6·83 mo (5·67–7·73)
in the placebo group (HR 0·77, 95% CI: 0·64–0·94; p = 0·0100. OS difference not yet clear
|
|
Ricolinostat[91]
|
Phase 1/2 trial of ricolinostat + bortezomib/DeX
|
ORR 37%, responses in bortezomib refractory 14%
|
|
BET
|
CPI203[92]
|
In vitro
|
Increased myeloma cell kill in vitro when added to Len/Dex combination
|
|
Molibresib[93]
|
Phase 1
|
Trial including patients with multiple hematological and solid organ cancers. Safety
and tolerability established
|
|
CPI-0160
|
Phase 1
|
Recruitment completed; results awaited. Details at NCT02157636
|
|
RO6870810[94]
|
Phase 1
|
Safety established Partial responses in 16%, including daratumumab resistant patients.
|
|
EZH
|
EPZ 6438[95]
|
In vitro
|
EPZ6438 in combination with Len represses myc and activates tumor suppressor genes
|
|
Tazemetostat
|
Phase 1
|
Currently recruiting for solid tumors and lymphomas
|
Abbreviations: CI, confidence interval; Dex, dexamethasone; DNMT, DNA methyl transferases;
EZH, enhancer of Zeste homologue; HDAC, histone deacetylase inhibitor; HR, hazard
ratio; Len, lenalidomide; ORR, overall response rate; OS, overall survival; PFS, progression-free
survival; RRMM, relapsed/refractory multiple myeloma; Vd, bortezomib-dexamethasone.
Promotor Region Hypermethylation Is a Potential Therapeutic Target
Promoter region hypermethylation of several tumor suppressor genes has been documented
in myeloma (summarized in [Table 2]). The use of hypomethylating agents, azacytidine (Aza) and decitabine, thus has a potential role in
reversing this phenomenon.[44] The in vitro efficacy of Aza against MM cell lines was demonstrated in 2008, where
it was shown that Aza led to demethylation of p16, theoretically restoring its tumor
suppressor function. It also inhibited IL-6 production and expression of IL-6 receptor,
leading to apoptosis of MM cell lines.[45] Further, Aza has been shown to have synergistic activity with several other chemotherapeutic
agents in MM. Bortezomib and doxorubicin have been shown to sensitize PCs in MM to
the effects of Aza by synergistic induction of double strand DNA breaks.[46] Thus, Aza, possibly in combination with other commonly used chemotherapeutic agents,
may have a potential therapeutic role in MM.
Table 2
Summary of pathologic genes hypermethylated in multiple myeloma
|
Name
|
Normal function
|
Frequency
|
Comments
|
|
P16
|
Inhibits CDK 4 and 6
|
19–53%
|
Associated with poor outcomes
Involved in progression from MGUS to myeloma[82]
|
|
SHP and SOCS[83]
|
Inhibition of JAK/STAT function
|
20–79%
|
Overactivity of IL-6 stimulated JAK/STAT
|
|
E-Cadherin
|
Cell adhesion inhibits cellular mobility
|
27–56%
|
Disease progression marker for high-risk disease, immature morphology
|
|
DAPK
|
Proapoptotic kinase
|
12.5–67%
|
Associated with high-risk disease, poorer response to therapy
|
|
DNA damage repair genes
|
Multiple genes[84]
|
|
More information needed for exact function
|
Abbreviation: IL-6, interleukin 6; MGUS, monoclonal gammopathy of undetermined significance.
Source: Adapted from Sharma et al.[84]
Preclinical Efficacy of Targeting miRNA Mechanisms
Several miRNAs have been shown to have therapeutic efficacy against PCs from MM cells
in preclinical studies. MiR-29b is shown to have anti-MM activity through multiple
mechanisms, including inhibition of IL-6 and JAK STAT signaling.[47] It has been shown to inhibit MM cell growth in vitro and potentiates the antitumor
efficacy of bortezomib.[48] Many miRNAs have been found to have tumor suppressor functions. For instance, miR-192,
-194, and -215 upregulate P53 expression, and their downregulation is involved in
disease progression.[49] Similarly, miR-34 is also shown to mediate similar activity by controlling cell
proliferation and differentiation. Synthetic miR-34 constructs were found to have
significant anti-MM activity, and found to potentiate the same in combination with
other anti-MM agents.[50] Anti-MM cell activity is also noted with miR-15a and 16a through multiple mechanisms,
including inhibition of BCL2, IL-17, and angiogenesis (via vascular endothelial growth
factor). Downregulation of antitumor activity is observed to correlate with advanced
disease stages.[51]
Histone Modification Has Potential Therapeutic Applications
As there are multiple pathways leading to histone modification in MM, several potential
therapeutic approaches exist to exploit these pathways for antimyeloma efficacy. Out
of the various drugs studied, HDACs are some of the most well characterized with a
proven clinical efficacy.[52] Panobinostat is a nonselective HDAC inhibitor that has been approved for the treatment
of relapsed/refractory myeloma based on phase 3 data.[53] Two important features noted with the use of HDAC inhibitors. First, a high rate
of cardiac adverse events and cytopenia is noted, indicating several off target epigenetic
effects that are still to be elucidated. The use of more selective HDAC inhibitors
is expected to reduce these serious adverse events. Second, the efficacy of these
drugs, even the newer selective HDAC inhibitors, is only modest as single agents.
The best results are seen in combination with other anti-MM agents.[54] Several clinical scores based on gene expression profile to predict response to
HDAC inhibitors have been described.[55] Histone acetylation is always activating, but histone methylation may be activating
or deactivating. Several small molecule modifiers of histone methylation are still
in preclinical or phase 1 trials (NCT02082977).[56]
Mediators of Histone Methylation Are Forthcoming Therapeutic Targets
Histone proteins are found in the nucleus of all eukaryotic cells and exist as octamers,
around which double-stranded DNA is wrapped. Each cell has two copies of each of the
histone proteins H2A, H2B, H3, and H4 that form nucleosomes around which DNA is compactly
packed. Release of DNA from histone proteins leads to chromatin modification and activation
of transcription, which is otherwise suppressed.[57] Histone modifications act by influencing the recruitment of nonhistone proteins
and the levels of chromatin compaction, altering the accessibility of transcription
factors to DNA. Posttranslational modification of histone proteins is an essential
biologic process, required for normal development and cellular functioning.[58] The most important and clinically relevant mechanisms of histone modification consist
of acetylation and methylation. We will focus on histone methylation for this discussion,
which is amenable to therapeutic intervention in myeloma.
Histone Methylation
Histone methylation occurs on basic amino acids, that is, arginine, lysine, and histidine,
and varies according to the amino acid in question. Lysine can be mono-, di-, or trimethylated,
arginine mono- or demethylated, and histidine monomethylated. Histone methylation
is context dependent, and effects on genetic expression may vary based on the location
of the target residue and degree of methylation.[58] An extensive review by Greer and Shi summarizes histone modification in detail,
and a relevant summary of these changes is provided below.[58]
Enzymes mediating histone changes are thought to be attracted to DNA through specific
sequences, the most prominent being the polycomb repressor group. The methylation
status of histones is read by proteins with methyl binding domains. Many mechanisms
are thought to be in play, but most elegantly, the positive charge created by methyl
residues is thought to increase the binding of hydrophobic proteins.[59] Histone methylation is mediated by a group of enzymes called histone methyltransferases,
of which arginine and lysine methyltransferases are the most relevant.
Arginine methyltransferases comprise nine members in all, as a part of the protein
arginine methyltransferase family (PRMT), which mediate methylation of arginine.[60] These enzymes catalyze two types of dimethylation and one monomethylation. Monomethylation
is regarded as an intermediate metabolite in the formation of di-ch3-arginine. PRMTs
are constitutively active and involved in cell growth and development, and multiple
mechanisms exist for regulation of activity of PRMTs. PTM, including methylation and
phosphorylation, inhibit PRMT function. In addition, PTM already on the substrate
prevents further addition of methyl groups. For example, phosphorylation blocks methylation
and acetylation stimulates methylation.[61] Expression of PRMTs has been altered in several malignancies.
Lysine methyltransferases are part of a more extensive family and are characterized
by the presence of a catalytic SET domain, which is conserved across all members of
the family. The SET domain, named after drosophila proteins from which it was isolated,
executes the final catalytic activity of these enzymes.[62] SET containing proteins can be further subclassified based on sequence homology
around the catalytic domain. Six families, namely, SUV39, EZH, SET2, PRDM, SMYD5,
and KMT4 (an exception with no SET domain), have been identified. Interestingly, SET
containing proteins also have the property of being able to read PTMs, indicating
a linkage of reader and writer functions in mediating overall control of histone modification.[63]
Salient details about the clinically important members from both of the above two
groups are listed below, which also form the focus of our study. [Table 3] summarizes important enzymes involved in histone methylation.
Table 3
A summary of enzymes involved in histone methylation and their therapeutic correlates
|
Methylation modifier
|
Prooncogenic effect
|
Others
|
Drug resistance
|
Adverse effect on survival
|
Preclinical small molecule inhibitor
|
|
MMSET
|
↓ P53 function
|
↑ IRF-4, MAP, c-MYC
|
By resistance to DS-DNA breaks
|
Yes
|
No
|
|
EZH2
|
↓ TS miRNA, ↓TS Genes (CDKN1C, RBPMS, LTB)
|
↑ Oncogenes, c-Myc, JUNB, BLIMP1
|
By cell adhesion mediated drug resistance
|
Yes
|
Yes, E7438, UNC1999 and GSK 126 and shRNA
|
|
PRMT5
|
↑ NF-kB pathway
|
↓ IKKβ
|
No
|
Yes
|
Yes, EPZ015666
|
|
KDM1A
|
↓ p53
|
↑c-MYC
|
No
|
Yes
|
Tranylcypromine, GSK-LSD1, and ORY-1001
|
|
KDM3A
|
↑ KLF2, IRF4, MALAT1
|
↓ Apoptosis
|
No
|
Yes
|
No
|
|
KDM6A
|
↓ Tumor suppressor function (self)
|
Coordinates with EZH2
|
No
|
Yes
|
Sensitizes to EZH2 inhibition
|
|
KDM6B
|
↑ ELK, FOS, MAP-k
|
–
|
No
|
Yes
|
–
|
Source: Adapted Adapted from Sharma et al[84] and Anderson et al[85]
MMSET: MMSET catalyzes the addition of H3K36me2, which is associated with active chromatin.
The significance of MMSET activation is seen in myeloma with t(4;14) translocation,
which shows universal activation of this gene.[64] MMSET expression has been found to promote myeloma tumor growth in vitro, with MMSET
knockdown leading to growth arrest.[65] MMSET also indirectly activates the activation of several oncogenic proteins that
act as transcription activators and play a role in carcinogenesis, including IRF4,
MAF, and c-MYC. Recently, it has also been shown to induce degradation of p53, thus
increasing cellular proliferation.[66] MMSET also enhances the ability of malignant PCs to repair DNA damage, leading to
resistance to alkylating agent-based chemotherapy.[67]
EZH2: EZH2 catalyzes the addition of a trimethyl mark H3K27me3, which is associated with
repression of gene expression. EZH2 is part of a PRC, which contains EZH2, ASXL1,
EED, and other accessory proteins.[68] EZH2 overexpression has been found to be associated with dysregulation of cell cycle
control and overall inferior outcomes. The effects of EZH2 overexpression are context
dependent, and lead to activation of transcription in DLBCL/Follicular lymphoma and
silencing in myeloma.[69] Multiple downstream effects of EZH2 include suppression of tumor suppressor miRNAs
and upregulation of antiapoptotic and pro-oncogenes.[70] EZH2 also leads to poorer outcomes by repression of p21 and p15 via H3K27me3, leading
to uncontrolled cyclin D overexpression and cellular proliferation.[35]
PRMT5: PRMT5 is an arginine methyltransferase and catalyzes dimethylation of arginine.
PRMT5 is overexpressed in myeloma and is associated with inferior outcomes. It activates
multiple oncogenic pathways, most notably the nuclear factor-kappa B pathway.[71] Pharmacologic inhibition of PRMT5 is a potential therapeutic target.
Demethylases: Histone demethylases consist of a large group of enzymes, of which important
members are listed below.
KDM1A: KDM1A (also known as LSD1), demethylates H3K4. It coordinates with several other
cellular proteins, including MMSET and HDAC. KDM1A has several nonhistone targets,
notably p53, whose function it inhibits, and has been associated with a poorer prognosis
for multiple malignancies.[72] In myeloma, inhibition of KDM1A has been found to inhibit interaction with epithelium
and osteoclastogenesis. It has also been associated with activation of multiple pathways,
including the c-myc pathway that contributes to pathogenesis and inferior overall
survival.[73]
KDM3 family: It consists of KDM3A, KDM3B, and JMJD1C. It has been found to be associated
with MM cell survival as part of the KDM3A-KLF2-IRF4 axis, and levels are seen to
be increased in MM patients compared with controls.[37] It has been shown that hypoxia inducible KDM3A knocks out KDM3A induced apoptosis
and leads to an antiapoptotic phenotype in malignant MM cells.[74]
KDM6A: Also known as UTX. It removes H3K27me2 and me3, methyl marks correlated with genomic
silencing. KDM6A mutations have been implicated in several malignancies, including
ALL/CMML and bladder cancers.[75] KDM6A mutations have been noted in over 10% of patients with MM and have been associated
with a poorer prognosis. UTX loss is associated with increased proliferation and clonogenicity
of MM cells.[76] Closely related KDM6B is also noted to play a role in MM cell survival and leads
to activation of MAP-K pathway genes.[77]
Clinical Analysis of Epigenetic Modifications
Changes in epigenetic modifications, including methylation and acetylation status,
can be quantitatively measured and have been shown to have several prognostic implications.
A basic understanding of clinical evaluation of epigenetic modifications is essential
to appraise studies describing epigenetic pathways. DNA methylation has been traditionally
tested with bisulfite sequencing, in which bisulfite is used to convert unmethylated
cytosine to uracil, differentiating it from methylated cytosine. Recently, CpG island
microarray has been utilized to provide a high throughput method for methylation status.[78] Histone modification is traditionally measured by CHiP sequencing, which can detect
any protein as long as a specific antibody is available.[79] This has been upscaled with combination with chip microarray.[80] There is no single standard method of choice for miRNA analysis, and a comparative
analysis can be found in this excellent review.[81] An overview of studies indicating prognostic impact of epigenetic changes at diagnosis
is summarized in [Table 4].
Table 4
A summary of studies describing the prognostic impact of epigenetic modifications
in myeloma
|
Study
|
Parameters
|
Specific genes
|
Salient findings
|
|
Kaiser et al[96]
|
Global methylation status of tumor suppressor genes in myeloma
|
GPX3
|
Median OS high vs. low methylation status, 16 vs. 46 mo, p = 0.0001
|
|
|
RBP1
|
23.9 vs. 47.7 mo, p < 0.0001
|
|
|
SPARC
|
19.4 vs. 47.7 mo, p < 0.0001
|
|
|
TGFB1
|
25.7 vs. 50.9 mo, p < 0.0001
|
|
Barwick et al[97]
|
Global DNA methylome
|
|
Low vs. high methylation status, median OS: 2 y vs. not reached, p = 8.7e-8
|
|
Mithraprabhu et al[39]
|
Histone deacetylase expression
|
Class I HDACs, HDAC 1,2,3, and 8 and Class II HDACs, HDAC5 and 10
|
High vs. low expression shorter PFS (p = 0.07) and OS (p = 0.003)
|
|
|
KDM6A mutation
|
Mutated vs. wild-type PFS: 16.8 vs 26.6 mo; proportion alive at 2 y mutated vs. wild
type: 51 vs. 80%
|
|
Pawlyn et al[98]
|
|
DNA methylation modifier
|
Mutated vs. wild-type PFS: NR vs. 26.6 mo; percent alive at 2 y mutated vs. wild-type:
58 vs. 80%
|
Abbreviations: HDAC, histone deacetylase inhibitor; OS, overall survival; PFS, progression-free
survival.