The Role of Epigenomics in PM
The Role of Epigenomics in PM
Author manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Author Manuscript
Virginia, USA
cDepartment of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy,
Richmond, Virginia, USA
Abstract
Introduction—Epigenetics is the study of reversible modifications to chromatin and their
extensive and profound effects on gene regulation. To date, the role of epigenetics in personalized
medicine has been under-explored. Therefore, this review aims to highlight the vast potential that
epigenetics holds.
Areas covered—We first review the cell-specific nature of epigenetic states and how these can
vary with developmental stage and in response to environmental factors. We then summarize
Author Manuscript
Keywords
Chromatin remodeling; DNA methylation; DNMT inhibitors; Epigenetic biomarkers; Epigenetic
drugs; HDAC inhibitors; Histone modifications; Oncology; Neuroscience; Pharmacokinetics
*
Correspondence to: Joseph L. McClay, Ph.D., Robert Blackwell Smith Building, 410 North 12th Street, Medical College of Virginia
Campus, Virginia Commonwealth University, Richmond, VA 23298-0533. Tel: +1 804-828 3428, [email protected].
Declaration of Interest
JL McClay received honoraria from the American College of Clinical Pharmacy to present on the topic of pharmacoepigenomics. The
authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or
financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Kronfol et al. Page 2
1. Introduction
Author Manuscript
Personalized medicine is founded upon the concept that individual differences in therapeutic
success are the norm among patients that require pharmacological treatment. This concept is
not new. Hippocrates writing in the 5th century BCE is known to have commented, “give
different ones [drugs] to different patients, for the sweet ones do not benefit everyone, nor do
the astringent ones, nor are all the patients able to drink the same things.” (see [1]). Thus, the
concept of variable response to drugs has been discussed for at least two and a half
millennia. However, being able to predict who will respond to a given drug has proven an
enduring challenge. With the advent of modern genomic technologies, which enable us to
read each patient’s genetic make-up, the idea of personalized medicine is becoming a reality.
Pharmacogenetics, the core discipline of personalized medicine, has already delivered some
profound and meaningful successes. The effectiveness of Cytochrome P450 (CYP450)
Author Manuscript
In the last decade, pharmacogenetics has harnessed the power of genome-wide association
studies (GWAS). This has enabled the field to move beyond the study of candidate genes to
scanning hundreds of thousands of genetic markers for each subject. Several promising new
leads have been discovered. Arguably, however, the success of GWAS in pharmacogenomics
Author Manuscript
has not mirrored that of complex disease studies. Primarily this may be an issue of statistical
power, whereby the clinical trials necessary to measure drug response are costly and so
sample sizes currently tend to be small. As studies grow in size and number and meta-
analyses are conducted across samples, we can expect GWAS to yield additional insight over
time [4]. However, GWAS will not yield all the answers for any given drug response
phenotype. Beyond the limitation where GWAS focuses on common polymorphisms, even if
all the relevant variants for response to a given drug were mapped, we would still be unable
to explain all the phenotypic variation in drug response [5]. Drug response is complex and,
like other complex traits, it likely arises from the interplay of multiple genetic and
environmental factors over the life course [6]. DNA sequence is just one component of this
complexity.
Most genotype associations in complex traits such as drug response are probabilistic
Author Manuscript
indicators of phenotype, which typically say little of certainty about the state of the organism
at the time of sampling. When treating an individual patient with a specific drug, substantial
supporting information in addition to genotype information may be required before making
a clinical decision. Even phenotypes that are strongly influenced by genetics, such as the
CYP450 drug metabolism phenotypes, will be modified by the effects of concurrent
medications or alcohol and tobacco use that may inhibit or interfere with CYP enzyme
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 3
activity [7,8]. This further illustrates the need to consider information beyond genotype
Author Manuscript
alone.
There are two broad complexities to living organisms that are not addressed by genotype
information. These are 1) spatial and 2) temporal variation in biological function or
phenotypic expression within the same organism. Consider that humans are composed of
multiple cell types with a diverse array of functions (spatial, or cell-specific variation) and
that we take on very different macroscopic forms in early versus later life (temporal, or
developmental variation). Yet essentially the same genome is present in all nucleated cells at
all time points. In this review, we will show how the processes that lead to cellular diversity
and organismal development, i.e. epigenetics, can be harnessed to provide more nuanced
DNA-based biomarkers and novel treatment strategies [9]. Indeed, epigenetics may also
yield an environmental exposure record of the patient that we are just beginning to
comprehend [10]. Epigenetic biomarkers are therefore fundamentally different to studies of
Author Manuscript
2. Epigenetics Overview
The term “epigenetics” was first described by the British developmental biologist Conrad
Waddington in the 1940s as “the branch of biology which studies the causal interactions
between genes and their products, which bring the phenotype into being” [12,13].
Waddington’s definition therefore predates the discovery of DNA and so the term
Author Manuscript
Today, backed by knowledge of the genome and some core molecular processes, epigenetics
can be defined as the study of mitotically stable changes in genetic regulation that do not
involve changes to nucleotide sequence [14]. Mitotic stability, in this sense, means that the
epigenetic state of the parent cell is written to the daughter cell after mitosis, thereby
continuing the developmental trajectory of the parent. This regulation is enacted via
epigenetic marks, which are reversible regulatory modifications to chromatin.
Author Manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 4
5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) [17,18]. The functions of these exotic
marks are still being elucidated, but 5hmC may play an important role in the central nervous
system, where it is prevalent, and in the regulation of pluripotency in stem cells [19,20].
reported [23]. Standard nomenclature abbreviates the histone, the modified residue and the
type of modification, such that histone 3 lysine 27 acetylation is written as “H3K27Ac”.
These modifications are written and erased by specific enzyme families, such as histone
acetyltransferases (HATs) and histone deacetylases (HDACs) in the case of acetylation
marks, or histone methyltransferases (HMTs) and demethylases (HDMs) in the case of
methylation marks [23].
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 5
epigenetic signature [29], although not all histone modifications are coupled with DNA
Author Manuscript
Massive studies such as ENCODE [31] and Roadmap Epigenomics [32] have significantly
advanced our understanding of genetic and epigenetic regulation. The ENCODE project
aims to identify all functional elements in the genome, while RoadMap Epigenomics aims to
elucidate epigenetic processes that contribute to human biology and disease. Both projects
make extensive use of next-generation sequencing (NGS) to profile reference epigenomes
and genome-wide protein-DNA binding patterns, including binding patterns for specific
modified histones. The most recent culmination of these efforts was the publication of 111
reference epigenomes by RoadMap Epigenomics [32]. This study revealed epigenetic
Author Manuscript
provides concrete biological pathways that are involved in the persistence of environmental
effects [36].
Epigenetic states can also vary between individuals because of genetic differences. In the
case of methylation, one of the simplest examples involves polymorphic CpG sites [37]. If a
nucleotide substitution ablates a CpG in some individuals, those individuals cannot be
methylated at that locus. There are several examples of disease-associated polymorphic
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 6
CpGs, suggesting that this is a significant contributor to individual differences in disease risk
Author Manuscript
[38,39]. In addition to polymorphic CpGs, DNA sequence variation may also affect the
binding of chromatin-interacting proteins and thus influence epigenetic states [40]. Thus,
individual differences in epigenetic states, whether arising via genotype or maladaptive
responses to environmental factors, can lead to disease.
approved tests that rely exclusively on epigenetic biomarkers. However, one commercially
available test with an epigenetic component has received full FDA approval. This is
ColoGuard®, a screening test for colorectal cancer in adults over 50. The test uses DNA
methylation levels at BMP3 and NDRG4, in combination of mutated KRAS and an
immunochemical assay for hemoglobin (Table 1). This test was reported to have superior
sensitivity but slightly lower specificity for colorectal cancer compared to the traditional
screening method, fecal immunochemical testing (FIT) [47]. However, more recent results
suggest FIT may be more effective and less costly than ColoGuard®, the latter necessitating
either very high patient uptake or a 60% reduction in cost per test to become the preferred
testing method [48]. This illustrates the economic barriers that diagnostic tests must
overcome, beyond the demonstration of efficacy and reproducibility, in order to become
widespread.
Author Manuscript
Two other epigenetic tests are currently available in the US, classified as Laboratory
Developed Tests (LDTs) and regulated under the Clinical Laboratory Improvement
Amendments (CLIA) program. This means that the test may only be conducted “in house”
in the laboratory where it was developed, once the lab meets CLIA performance standards.
The two tests are ConfirmMDx and AssureMDx, for prostate cancer and bladder cancer
respectively. Hypermethylation of the glutathione S-transferase gene (GSTP1) promoter in
prostrate cancer was first shown in the 1990s [49]. This marker, plus APC and RASSF1, are
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 7
now components of the ConfirmMDx test (Table 1), which is used to address false-negative
Author Manuscript
prostate biopsy concerns [50]. The AssureMDx test for bladder cancer involves the analysis
of DNA methylation levels of three genes (TWIST1, ONECUT2 and OTX1) in combination
with mutation analysis of three others [51].
In lung cancer, the DNA methylation of the SHOX2 gene was reported to be an accurate
marker for identifying lung cancer based on analysis of bronchial aspirates [52]. In Europe,
this biomarker is now commercially available as the Epi proLungVR BL Reflex Assay [53].
However, this test has not yet received regulatory approval for use in the USA.
In breast and ovarian cancer, hypermethylation of the BRCA1 promoter region has been
observed repeatedly [54,55]. BRCA1 is also thought to epigenetically repress expression of
the oncogenic microRNA miR-155 via a mechanism involving histone deacetylase 2
(HDAC2) [56]. A recent study by Anjum et al. (2014) identified a blood cell DNA
Author Manuscript
methylation signature at BRCA1 that was able to predict breast cancer risk several years
prior to diagnosis [57]. However, this biomarker is not yet available in a commercial kit or
test.
The biomarker potential of circulating cell-free DNA (cfDNA) has been investigated in
breast cancer and other cancers. Circulating cfDNA is extracted from plasma or serum and is
derived from dying tumor cells that release their DNA into the bloodstream. Kloten et al
(2013) used a panel of three genes (ITIH5, DKK3 and RASSF1A) that showed
hypermethylation in serum cfDNA from breast cancer patients and found these could
discriminate between patients and controls with a sensitivity of 67% and specificity of 69%
[58]. Fackler et al. (2014) followed this with a panel of 10 genes and cancer-specific DNA
was detected in sera with a sensitivity of 91% and specificity of 96% in the test samples
Author Manuscript
[59]. The researchers of the latter study are reportedly working with the diagnostics
company Cepheid to bring this test to market [60].
While epigenetic studies of cancer are arguably the most advanced relative to other areas,
several diseases have shown promising findings, particularly with respect to DNA
methylation. These include neurological disorders such as Alzheimer’s Disease [61,62] and
Parkinson’s Disease [63], autoimmune disorders such as systemic lupus erythematosus [64],
and psychiatric disorders such as schizophrenia [65,66] and autism [67]. Despite these
advances, there are no currently available diagnostic kits for these diseases that employ
epigenetic markers. Nevertheless, it is hoped that the clinical value of epigenomics already
seen in oncology will be replicated in these areas [68].
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 8
inhibitors. Currently there are two classes of epigenetic drugs that have been approved by
FDA for clinical use in the United States: DNMTi and HDACi (see Table 2).
The first approved epidrug in the US was azacitidine (Vidaza, Azadine), a DNMTi indicated
to treat chronic myelomonocytic leukemia and myelodysplastic syndrome. Azacitidine was
approved in 2004 and quickly followed by decitabine (Dacogen) with same indication two
years later. Both drugs cause broad hypomethylation that leads to cellular dysregulation that
most seriously affects rapidly dividing cells. It is important to note that these drugs are not
highly locus-specific and these agents can cause hypomethylation at many genomic sites.
Even though current drugs are designed to favorably induce genes that have been silenced in
cancer [70], they may also activate the expression of prometastatic genes as well as
oncogenes [71]. There remains a need to develop more selective DNMTi to improve the
efficacy and reduce side effects for this class of drug.
Author Manuscript
The potential application of DNMTi to other diseases is also under investigation and
examples include multiple sclerosis [72], HIV [73], pain [74] and memory [75]. For
example, DNMT activity was observed in HIV-1 infection of CD4(+) T-cells in vitro and
induced hypermethylation of distinct cellular promoters [73]. Studies from Rajasethupathy
et al. suggested that DNA methylation is necessary for serotonin-dependent long-term
facilitation in memory formation [76]. For a curative therapy of AIDS patients, a
combination of antiretroviral drugs and epidrugs has been suggested for the reactivation of
latent HIV-1 genomes. These epidrugs include DNMTi, HDACi, histone methyltransferase
inhibitors (HMTi) and histone demethylase inhibitors [73].
The HDAC inhibitors suberoylanilide hydroxamic acid (SAHA, vorinostat, in 2006) and
Author Manuscript
the DNMTi drugs, current HDACi have broad effects across the genome and lack locus-
specificity. These drugs can have serious side effects [43] and use of currently approved
HDACi in cancer is often indicated only after other treatments have failed, or as combination
therapies (Table 2).
Besides these two approved epidrug classes, HMTi and bromodomain inhibitors are other
emerging epidrug classes under development. Pinometostat is a small molecule inhibitor of
the histone methyltransferase DOT1L for the treatment of MLL-r leukemia [81].
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 9
Tazemetostat is an orally administered, first-in-class small molecule HMTi that targets the
Author Manuscript
indications outside oncology. We restrict our listing of early phase cancer indications
because these are too numerous to list concisely.
Finally, several HATi are in preclinical studies at time of writing. Aberrant function of
HATs, also called lysine acetyltransferases (KATs), is correlated with cancer and other
diseases [84]. HATi are great candidates with potential therapeutic utility, but current HATi
only have moderate potency and specificity and none are in clinical trial at time of writing.
Nevertheless, some HATi have shown efficacy in preclinical studies. Compound C646 is a
pyrazolone-containing small molecule inhibitor of the p300/CBP HAT subfamily [85]. It has
been shown to cause growth arrest in melanoma cell lines and inhibit cancer cell growth in
prostate and lung cancer cell lines. PU139 is a pyridoisothiazolone that inhibits several HAT
subfamilies and was shown to block neuroblastoma xenograft growth in mice [86]. These
agents and others in development are indicative that HATi are still in infancy relative to other
Author Manuscript
epigenetic drugs, but they show enormous promise and need further investment to reach
their potential as therapeutic compounds.
O-6 positions of guanine residues and the resulting DNA damage triggers tumor cell death.
Hypomethylation of MGMT leads to expression of O6-alkylguanine DNA alkyltransferase,
which can repair the DNA damage, whereas hypermethylation leads to silencing of the gene
and thus greater susceptibility to the drug. In addition to glioma, a role for MGMT in
predicting response to metastatic colorectal cancer (mCRC) treatment has also been
suggested [89].
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 10
Other published epigenetic biomarker examples include GSTP1 and BRCA1. Methylation of
Author Manuscript
the promoter of GSTP1 is correlated with survival in breast cancer patients and may be
predictive of treatment efficacy with doxorubicin [90] or DNA methyltransferase (DNMT)
inhibitors [91]. The BRCA1 gene plays a role in DNA damage response and
hypermethylation of the BRCA1 promoter region may be predictive of enhanced sensitivity
to platinum-derived drugs in cancer cell lines and xenografted tumors; it also may be
predictive of increased time to relapse and survival in ovarian cancer patients under cisplatin
treatment [92].
The impact of epigenetics in drug response has been investigated beyond oncology. For
example, methylation of the P2 promoter of the IGF1 gene affects transcriptional response to
growth hormone (GH) [93]. GH is mainly used to treat children with short stature due to
growth hormone deficiency. Ouni et al. [93] measured P2-driven and total IGF1 transcripts
before and 12 h after the GH injection and found an increase in P2-driven transcripts with a
Author Manuscript
very strong inverse correlation with CG-137 methylation. This correlation accounted for ~
25% of the variability in the response to GH.
Unfortunately, research to date has not yet directly addressed this question, but individual
variation in epigenetic states of ADME genes has been correlated with a range of outcomes.
For example, Parkinson’s disease has been associated with hypomethylation of the CYP2E1
gene promoter in the brain [95]. Methylation levels at CYP1B1 [96] and CYP1A1 [97] have
been associated with prostate cancer, and CYP2W1 with colon cancer [98]. Methylation
levels at the drug transporter genes OCT1 [99] and OCT2 [100], responsible for the renal
excretion of drugs, have been associated with renal carcinoma. These findings demonstrate
the existence of inter-patient variability in ADME gene epigenetic states, some of which
have functional effects on gene expression. However, the extent of normal epigenetic
variation at these loci in the population and the extent to which it will affect pharmacokinetic
phenotypes remains to be determined.
Author Manuscript
3.5 Conclusion
Epigenomic medicine is already here, with numerous epigenetic disease associations
reported, six epidrugs and a handful of epigenetic biomarker tests available the US, plus a
small number of other products available worldwide. The largest number of findings and
applications to date is in the field of oncology. However, the field of epigenetics is only a
few decades old and epigenomic medicine is a very recent arrival, so we are still in early
days. The perceived benefits that epigenomics will bring to healthcare are emphatically
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 11
illustrated by the large number of epidrugs currently in development and the large sums of
Author Manuscript
4. Expert Commentary
Several factors are currently driving innovation in epigenomic medicine. First is the general
level of interest in the field, which is high. Second is the ongoing characterization of
reference epigenomes to enrich and accelerate research efforts. Third is the availability of
powerful methods such as next-generation sequencing (NGS) to characterize epigenomes.
Discussion of technical methods is largely outside the scope of this article and reviews have
been published elsewhere [101,102]. However, with NGS approaches already in use to
Author Manuscript
Considering epigenomic biomarker research, among the most significant difficulties are data
complexity and the clean interpretation of findings [103]. Unlike studies of genotype,
epigenomics has a direction of causality problem. While epigenetic biomarkers may be
predictive of disease state or drug response, epigenetic changes are also inducible by
pharmacological treatments [11,104]. As a result, there is the risk that epigenetic differences
between cases and controls in an epigenome-wide association study could be the result of
drug treatment in cases, rather than causal variation. Furthermore, evidence from genome-
wide studies suggests that not all epigenetic changes are functional or cause identifiable
changes to gene expression [105]. Targeting specific populations, such as drug-naïve
Author Manuscript
patients, may go some way to solving issues related to the direction of causality, but it is
certain that experimental model systems will be needed to adequately disentangle causality
and establish functionality of epigenetic changes.
Another complexity is that epigenetic modifications are cell-specific. While this is in many
ways an advantage, and can give precise insight into the workings of the cell of origin, it
also leads to some challenges in sample collection, particularly with respect to clinical
studies. Blood DNA is the most readily-accessible source from humans, but the extent to
which blood DNA methylation is reflective of methylation changes in other tissues is
debated and it seems there may not be a hard and fast rule with respect to which changes are
reflected in blood as compared to which are not. Aging epigenetic signatures, also known as
the “epigenetic clock”, appear to transcend tissue barriers [106], but the extent to which a
blood DNA methylation mark is informative about a disease of, for example, the lung or
Author Manuscript
heart remains an open question. Circulating cfDNA is an exception, since it is sourced from
the diseased tissue of interest and is merely liberated into the bloodstream.
While these considerations apply to the discovery of novel epigenetic biomarkers, a separate
set of considerations apply to novel epigenetic drugs. Paramount among priorities for future
epidrug development is improving target specificity. This can be viewed in two ways. First,
as mentioned above, current drugs lack genomic locus specificity and affect DNA
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 12
correction of aberrant changes, some sort of nucleic acid targeting adjunct is likely to be
required. While antisense RNA (MG98) has already been used to modulate DNMT activity
with some success [107], it is difficult to speculate how this could be used to target
epigenetic modifications at specific target loci. On the other hand, it may be possible to
capitalize on the locus targeting abilities of CRISPR/Cas9 systems to deliver epigenetic
modifying agents to specific loci. Indeed, epigenome editing has already been demonstrated
using this broad approach [108]. A second consideration involves the specificity of epidrugs
to specific members of families of chromatin modifying enzymes. For example, there are
numerous human DNMTs and HDAC enzymes with somewhat different functions and
substrate specificities but currently available DNMTi and HDACi are non-selective and
inhibit many isozymes. However, some drugs currently in clinical trials appear to be more
selective, e.g. mocetinostat that inhibits only HDAC 1 and 4 (see Table 3). Thus, the problem
Author Manuscript
of specificity does not appear to be insurmountable. To conclude, we mention two areas, one
technological and one clinical, that we consider to be of significant interest going forward.
The advent of chromatin conformation capture (3C) sequencing technology marked the
beginning of a new era in precision medicine and our understanding of epigenomic
regulation. Its evolution into Hi-C technology allows insight into the well-organized 3D
structure of the human genome within the nucleus [109]. Numerous studies demonstrated
highly conserved topologically associated domains (TADs) - spatially close units of
chromatin bringing together enhancers, promoters of genes, and other regulatory elements.
These TADs have well-defined boundaries marked by strongly interacting chromatin regions
(chromatin loops) [110]. TADs harbor multiple active RNA polymerases anchored to a
nuclear substructure, with genes within TADs showing co-expression patterns [111]. TADs
are increasingly recognized as regulatory units orchestrating expression of thousands of
Author Manuscript
In the clinical arena, aging is an area where epigenomic medicine may make an impact.
Older adults are at increased risk for adverse drug events and this may be partly because
aging is associated with changes in physiology that can affect drug pharmacokinetics and
pharmacodynamics [116]. The clinical challenge is to identify those patients who are more
likely to experience an adverse drug event or altered drug response among the older adult
Author Manuscript
population when weighing the risk versus the benefit of a drug therapy. Chronologic age
alone is insufficient as an indicator that dosage adjustment or avoidance of a particular
therapeutic agent is warranted. Pharmacogenetic information alone is also insufficient, as
altered drug response and risk of adverse drug events changes across the lifespan while
genotype remains constant [117,118]. Epigenetic alterations may be a better indicator than
chronological age for personalizing drug therapy for the older population. For example, it
has been proposed that epigenetic regulation of cytochrome P450 (CYP) enzymes
responsible for drug metabolism through DNA methylation may result in altered drug
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 13
exposure in geriatric patients [119]. More research is needed to elucidate the relationships
Author Manuscript
between epigenetics and drug exposure and response during senescence, but is a promising
alternative to chronologic age for adjusting pharmacotherapy in older adults.
Five-year view
We expect to see the introduction of several new epidrugs in the next five years, given that
many are currently in later stage clinical trials. Perhaps the most significant change in this
area will be the introduction of epidrugs with indications outside cancer. For example,
Apabetalone (RVX-208) from Resverlogix is in Phase III trials for high risk Type 2 diabetes
mellitus patients with coronary artery disease (Table 3). Resverlogix also reports the
planning of Phase II trials for Alzheimer’s disease (www.resverlogix.com/programs/
rvx-208-clinical-development/). Successful approval for either of these indications would be
a first step for epidrugs outside oncology. We also expect to see new classes of epidrug enter
Author Manuscript
clinical trials. As mentioned above, several HATi are in preclinical development and show
therapeutic promise. There is growing recognition of the importance of histone acetylation
in many diseases, including cancer, so we expect the first HATi to enter clinical trials in
three to five years.
Given the high level of current research interest, we expect a number of epigenetic
biomarker tests to become commercially available on a five-year horizon. There are
numerous clinical trials currently ongoing to evaluate epigenetic biomarkers. In addition to
the developments cited in Sections 3.1 and 3.3 above, clinical trials of DNA methylation
biomarkers for disease diagnosis, prediction of treatment efficacy and even quantifying toxin
exposure (see for example clinical trial NCT01815385) are ongoing or planned. One area
that should see significant growth is the use of epigenetic markers on cfDNA. As mentioned
in Section 3.1, cfDNA is sourced from dead cells that have released their contents into the
Author Manuscript
As researchers continue to seek out new epigenetic biomarkers, we will see more large-scale
epigenome-wide studies of complex diseases. These studies will primarily focus on DNA
methylation but we expect the current, prevailing use of high density microarrays to largely
give way to NGS as costs continue to fall. Whole genome bisulfite shotgun sequencing
Author Manuscript
(WGBS) is poised to become the dominant method of choice, because it is the only way to
assay DNA methylation at single base resolution. Correlation between neighboring
methylated sites is low beyond very short distances [40], so this level of resolution is
necessary for a comprehensive genome-wide DNA methylation analysis. While cost is the
primary factor limiting large-scale implementation of WGBS, improved analysis methods
will be needed to extract maximum information from these complex data. Research into
whole genome sequence analysis methods will intensify over the next five years with efforts
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 14
In sum, the next five years should see intensifying research efforts in personalized epigenetic
Author Manuscript
medicine, the introduction of several new epidrugs, initiation of clinical trials for new drug
classes, and the clinical introduction of novel DNA methylation biomarker tests. Overall it
should prove an exciting time for this nascent area.
Acknowledgments
Funding
JL McClay was partially supported by grant R21MH099419 from the US National Institutes of Health. MG
Dozmorov was supported in part by the Virginia Commonwealth University Presidential Research Quest award. R
Huang was supported by an American Cancer Society Institutional Research Grant. Sponsors had no role in the
preparation of this manuscript.
References
Author Manuscript
1. Sykiotis GP, Kalliolias GD, Papavassiliou AG. Pharmacogenetic Principles in the Hippocratic
Writings. J Clin Pharmacol. 2005; 45:1218–1220. [PubMed: 16239353]
2. Ingelman-Sundberg M. Pharmacogenetics of cytochrome P450 and its applications in drug therapy:
the past, present and future. Trends Pharmacol Sci. 2004; 25:193–200. [PubMed: 15063083]
3. McCarthy JJ, McLeod HL, Ginsburg GS. Genomic medicine: a decade of successes, challenges, and
opportunities. Sci Transl Med. 2013; 5:189sr4. [PubMed: 23761042]
4. Ritchie MD. The success of pharmacogenomics in moving genetic association studies from bench to
bedside: study design and implementation of precision medicine in the post-GWAS era. Hum Genet.
2012; 131:1615–1626. [PubMed: 22923055]
5. Manolio TA, Collins FS, Cox NJ, et al. Finding the missing heritability of complex diseases. Nature.
2009; 461:747–753. [PubMed: 19812666]
6. Cardon LR, Harris T. Precision medicine, genomics and drug discovery. Hum Mol Genet. 2016;
25:R166–R172. [PubMed: 27538422]
7. Tracy TS, Chaudhry AS, Prasad B, et al. Interindividual Variability in Cytochrome P450-Mediated
Author Manuscript
Drug Metabolism. Drug Metab Dispos Biol Fate Chem. 2016; 44:343–351. [PubMed: 26681736]
8. He Z-X, Chen X-W, Zhou Z-W, et al. Impact of physiological, pathological and environmental
factors on the expression and activity of human cytochrome P450 2D6 and implications in precision
medicine. Drug Metab Rev. 2015; 47:470–519. [PubMed: 26574146]
9. Bock C. Epigenetic biomarker development. Epigenomics. 2009; 1:99–110. [PubMed: 22122639]
10. Ladd-Acosta C. Epigenetic Signatures as Biomarkers of Exposure. Curr Environ Health Rep. 2015;
2:117–125. [PubMed: 26231361]
11. Feinberg AP. Phenotypic plasticity and the epigenetics of human disease. Nature. 2007; 447:433–
440. [PubMed: 17522677]
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 15
12. Goldberg AD, Allis CD, Bernstein E. Epigenetics: A Landscape Takes Shape. Cell. 2007;
128:635–638. [PubMed: 17320500]
Author Manuscript
13. Noble D. Conrad Waddington and the origin of epigenetics. J Exp Biol. 2015; 218:816–818.
[PubMed: 25788723]
14. Riggs, A., Russo, V., Martiensen, R. Epigenetic Mechanisms of Gene Regulation. Plainview, NY:
Cold Spring Harbor Laboratory Press; 1996.
15. Irizarry RA, Ladd-Acosta C, Wen B, et al. The human colon cancer methylome shows similar
hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;
41:178–186. [PubMed: 19151715]
16. Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science. 2001;
293:1089–1093. [PubMed: 11498579]
17. Ito S, Shen L, Dai Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-
carboxylcytosine. Science. 2011; 333:1300–1303. [PubMed: 21778364]
18. Bachman M, Uribe-Lewis S, Yang X, et al. 5-Formylcytosine can be a stable DNA modification in
mammals. Nat Chem Biol. 2015; 11:555–557. [PubMed: 26098680]
19. Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje
Author Manuscript
33. Ernst J, Kheradpour P, Mikkelsen TS, et al. Mapping and analysis of chromatin state dynamics in
nine human cell types. Nature. 2011; 473:43–49. [PubMed: 21441907]
34. Fraga MF, Ballestar E, Paz MF, et al. Epigenetic differences arise during the lifetime of
monozygotic twins. Proc Natl Acad Sci U S A. 2005; 102:10604–10609. [PubMed: 16009939]
35. Feil R, Fraga MF. Epigenetics and the environment: emerging patterns and implications. Nat Rev
Genet. 2011; 13:97–109.
36. Rutter M. Why is the topic of the biological embedding of experiences important for translation?
Dev. Psychopathol. 2016; 28:1245–1258.
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 16
37. Zhi D, Aslibekyan S, Irvin MR, et al. SNPs located at CpG sites modulate genome-epigenome
interaction. Epigenetics. 2013; 8:802–806. [PubMed: 23811543]
Author Manuscript
38. Zhou D, Li Z, Yu D, et al. Polymorphisms involving gain or loss of CpG sites are significantly
enriched in trait-associated SNPs. Oncotarget. 2015; 6:39995–40004. [PubMed: 26503467]
39. Cazaly E, Charlesworth J, Dickinson JL, et al. Genetic Determinants of Epigenetic Patterns:
Providing Insight into Disease. Mol Med. 2015; 21:400–409. [PubMed: 25822796]
40. McClay JL, Shabalin AA, Dozmorov MG, et al. High density methylation QTL analysis in human
blood via next-generation sequencing of the methylated genomic DNA fraction. Genome Biol.
2015; 16:291. [PubMed: 26699738]
41. García-Giménez, JL. Epigenetic Biomarkers and Diagnostics. Academic Press; 2015.
42. Szyf M. Epigenetics, DNA methylation, and chromatin modifying drugs. Annu Rev Pharmacol
Toxicol. 2009; 49:243–263. [PubMed: 18851683]
43. Hunter P. The second coming of epigenetic drugs: a more strategic and broader research framework
could boost the development of new drugs to modify epigenetic factors and gene expression.
EMBO Rep. 2015; 16:276–279. [PubMed: 25662153]
44. Suvà ML, Riggi N, Bernstein BE. Epigenetic reprogramming in cancer. Science. 2013; 339:1567–
Author Manuscript
56. Chang S, Wang R-H, Akagi K, et al. Tumor suppressor BRCA1 epigenetically controls oncogenic
microRNA-155. Nat Med. 2011; 17:1275–1282. [PubMed: 21946536]
57. Anjum S, Fourkala E-O, Zikan M, et al. A BRCA1-mutation associated DNA methylation
signature in blood cells predicts sporadic breast cancer incidence and survival. Genome Med.
2014; 6:47. [PubMed: 25067956]
58. Kloten V, Becker B, Winner K, et al. Promoter hypermethylation of the tumor-suppressor genes
ITIH5, DKK3, and RASSF1A as novel biomarkers for blood-based breast cancer screening. Breast
Cancer Res BCR. 2013; 15:R4. [PubMed: 23320751]
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 17
59*. Fackler MJ, Lopez Bujanda Z, Umbricht C, et al. Novel methylated biomarkers and a robust assay
to detect circulating tumor DNA in metastatic breast cancer. Cancer Res. 2014; 74:2160–2170.
Author Manuscript
bladder cancer cells exposed to the methylation inhibitor 5-aza-2′-deoxycytidine. Cancer Res.
2002; 62:961–966. [PubMed: 11861364]
71. Cheishvili D, Boureau L, Szyf M. DNA demethylation and invasive cancer: implications for
therapeutics. Br J Pharmacol. 2015; 172:2705–2715. [PubMed: 25134627]
72. Peedicayil J. Epigenetic Drugs for Multiple Sclerosis. Curr Neuropharmacol. 2016; 14:3–9.
[PubMed: 26813117]
73. Abdel-Hameed EA, Ji H, Shata MT. HIV-Induced Epigenetic Alterations in Host Cells. Adv Exp
Med Biol. 2016; 879:27–38. [PubMed: 26659262]
74. Sun Y, Sahbaie P, Liang D, et al. DNA Methylation Modulates Nociceptive Sensitization after
Incision. PloS One. 2015; 10:e0142046. [PubMed: 26535894]
75. Singh P, Konar A, Kumar A, et al. Hippocampal chromatin-modifying enzymes are pivotal for
scopolamine-induced synaptic plasticity gene expression changes and memory impairment. J
Neurochem. 2015; 134:642–651. [PubMed: 25982413]
76. Rajasethupathy P, Antonov I, Sheridan R, et al. A role for neuronal piRNAs in the epigenetic
control of memory-related synaptic plasticity. Cell. 2012; 149:693–707. [PubMed: 22541438]
Author Manuscript
77. Lane AA, Chabner BA. Histone Deacetylase Inhibitors in Cancer Therapy. J Clin Oncol. 2009;
27:5459–5468. [PubMed: 19826124]
78. Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological
diseases and immune disorders. Nat Rev Drug Discov. 2014; 13:673–691. [PubMed: 25131830]
79. Kurita M, Holloway T, García-Bea A, et al. HDAC2 regulates atypical antipsychotic responses
through the modulation of mGlu2 promoter activity. Nat Neurosci. 2012; 15:1245–1254.
[PubMed: 22864611]
80. Sharma S, Taliyan R. Histone deacetylase inhibitors: Future therapeutics for insulin resistance and
type 2 diabetes. Pharmacol Res. 2016; 113:320–326. [PubMed: 27620069]
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 18
81*. Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of MLL-
fusion leukemia. Blood. 2013; 122:1017–1025. An interesting paper demonstrating the
Author Manuscript
landmark study that was one of the first to show association between epigenetic factors and
therapeutic drug response. [PubMed: 11070098]
88. Hegi ME, Diserens A-C, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in
glioblastoma. N Engl J Med. 2005; 352:997–1003. [PubMed: 15758010]
89. Fornaro L, Vivaldi C, Caparello C, et al. Pharmacoepigenetics in gastrointestinal tumors: MGMT
methylation and beyond. Front Biosci Elite Ed. 2016; 8:170–180. [PubMed: 26709653]
90. Chiam, K., Centenera, MM., Butler, LM., et al. GSTP1 DNA Methylation and Expression Status Is
Indicative of 5-aza-2′-Deoxycytidine Efficacy in Human Prostate Cancer Cells. In: Agoulnik, I.,
editor. PLoS ONE. Vol. 6. 2011. p. e25634
91. Dejeux E, Rønneberg J, Solvang H, et al. DNA methylation profiling in doxorubicin treated
primary locally advanced breast tumours identifies novel genes associated with survival and
treatment response. Mol Cancer. 2010; 9:68. [PubMed: 20338046]
92. Stefansson OA, Villanueva A, Vidal A, et al. BRCA1 epigenetic inactivation predicts sensitivity to
platinum-based chemotherapy in breast and ovarian cancer. Epigenetics. 2012; 7:1225–1229.
Author Manuscript
[PubMed: 23069641]
93. Ouni M, Belot MP, Castell AL, et al. The P2 promoter of the IGF1 gene is a major epigenetic locus
for GH responsiveness. Pharmacogenomics J. 2016; 16:102–106. [PubMed: 25869012]
94. Ivanov M, Kacevska M, Ingelman-Sundberg M. Epigenomics and interindividual differences in
drug response. Clin Pharmacol Ther. 2012; 92:727–736. [PubMed: 23093317]
95. Kaut O, Schmitt I, Wüllner U. Genome-scale methylation analysis of Parkinson’s disease patients’
brains reveals DNA hypomethylation and increased mRNA expression of cytochrome P450 2E1.
Neurogenetics. 2012; 13:87–91. [PubMed: 22238121]
96. Tokizane T, Shiina H, Igawa M, et al. Cytochrome P450 1B1 is overexpressed and regulated by
hypomethylation in prostate cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2005; 11:5793–
5801.
97. Okino ST, Pookot D, Li L-C, et al. Epigenetic inactivation of the dioxin-responsive cytochrome
P4501A1 gene in human prostate cancer. Cancer Res. 2006; 66:7420–7428. [PubMed: 16885337]
98. Gomez A, Karlgren M, Edler D, et al. Expression of CYP2W1 in colon tumors: regulation by gene
methylation. Pharmacogenomics. 2007; 8:1315–1325. [PubMed: 17979506]
Author Manuscript
99. Schaeffeler E, Hellerbrand C, Nies AT, et al. DNA methylation is associated with downregulation
of the organic cation transporter OCT1 (SLC22A1) in human hepatocellular carcinoma. Genome
Med. 2011; 3:82. [PubMed: 22196450]
100. Liu Y, Zheng X, Yu Q, et al. Epigenetic activation of the drug transporter OCT2 sensitizes renal
cell carcinoma to oxaliplatin. Sci Transl Med. 2016; 8:348ra97.
101. Laird PW. Principles and challenges of genomewide DNA methylation analysis. Nat Rev Genet.
2010; 11:191–203. [PubMed: 20125086]
102. Krueger F, Kreck B, Franke A, et al. DNA methylome analysis using short bisulfite sequencing
data. Nat Methods. 2012; 9:145–151. [PubMed: 22290186]
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 19
103. Ledford H. Epigenetics: The genome unwrapped. Nature. 2015; 528:S12–S13. [PubMed:
26630592]
Author Manuscript
104. Wang Y, Krishnan HR, Ghezzi A, et al. Drug-induced epigenetic changes produce drug tolerance.
PLoS Biol. 2007; 5:e265. [PubMed: 17941717]
105*. Stricker SH, Köferle A, Beck S. From profiles to function in epigenomics. Nat Rev Genet. 2016
advance online publication. An outstanding and up-to-date review of complex epigenomic
regulation.
106. Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013; 14:R115.
[PubMed: 24138928]
107. Reid GK, Besterman JM, MacLeod AR. Selective inhibition of DNA methyltransferase enzymes
as a novel strategy for cancer treatment. Curr Opin Mol Ther. 2002; 4:130–137. [PubMed:
12044034]
108. Thakore PI, Black JB, Hilton IB, et al. Editing the epigenome: technologies for programmable
transcription and epigenetic modulation. Nat Methods. 2016; 13:127–137. [PubMed: 26820547]
109*. Lieberman-Aiden E, van Berkum NL, Williams L, et al. Comprehensive mapping of long-range
interactions reveals folding principles of the human genome. Science. 2009; 326:289–293. A
Author Manuscript
116. Slattum, P., Peron, E., Ogbonna, K. The Pharmacology of Aging. In: Fill, HM.Rockwood,
K.Woodhouse, K., Young, JB., editors. Brocklehursts Textb Geriatr Med Gerontol. 8. Philadephia
PA: Saunders/Elsevier; 2016. p. Chapter 26
117. Pal S, Tyler JK. Epigenetics and aging. Sci Adv. 2016; 2:e1600584. [PubMed: 27482540]
118. Brunet A, Berger SL. Epigenetics of aging and aging-related disease. J Gerontol A Biol Sci Med
Sci. 2014; 69(Suppl 1):S17–20. [PubMed: 24833581]
119. Seripa D, Panza F, Daragjati J, et al. Measuring pharmacogenetics in special groups: geriatrics.
Expert Opin Drug Metab Toxicol. 2015; 11:1073–1088. [PubMed: 25990744]
120. Thakur BK, Zhang H, Becker A, et al. Double-stranded DNA in exosomes: a novel biomarker in
cancer detection. Cell Res. 2014; 24:766–769. [PubMed: 24710597]
121*. Sun W, Poschmann J, Cruz-Herrera Del Rosario R, et al. Histone Acetylome-wide Association
Study of Autism Spectrum Disorder. Cell. 2016; 167:1385–1397. e11. The first study, to our
knowledge, to systematically compare epigenome-wide histone modification profiles in disease
cases versus controls. [PubMed: 27863250]
Author Manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 20
Key issues
Author Manuscript
• Epigenetic marks are reversible, therefore much effort has been expended in
Author Manuscript
• Future directions may include the “druggable nucleosome” concept and life
stage-specific biomarkers of drug response.
Author Manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 21
Author Manuscript
Figure 1.
Waddington represented the developmental process as a series of “decisions” made by
differentiating cells that could be represented as forks in the valleys of the “developmental
landscape”. Panels A and B represent the alternate fates of the cell, or ball by analogy. As
the pluripotent stem cell of the egg (ball at the top), begins to specialize, the differentiation
Author Manuscript
“decisions” made are irreversible. Its pattern of epigenetic regulation is established by the
point of terminal differentiation at the bottom of the landscape. With epigenetic drugs and
therapies, the aim is to artificially reverse maladaptive epigenetic states and essentially “push
the ball back up the hill”. Figure from Noble (2015) [13] reproduced with permission.
Author Manuscript
Author Manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Kronfol et al. Page 22
Author Manuscript
Author Manuscript
Author Manuscript
Figure 2.
Epigenetic regulation of gene expression via chromatin remodeling. The diagram shows two
generic chromatin activity states. At the top, active chromatin is open and accessible to
transcription factors and polymerases, with nucleosomes spread apart, DNA typically in an
unmethylated state and acetylation marks on histones. HAT is histone acetyltransferase,
SWI/SNF is a nucleosome remodeling complex, RNA Pol II is RNA polymerase II. The
Author Manuscript
lower panel shows the opposite inactive chromatin scenario, where the nucleosomes are
tightly packed, the DNA is methylated and inaccessible to transcription factors, while
histones have their acetylation marks removed. HDAC is histone deacetylase, HMT is
histone methyltransferase. Figure is adapted from Luong, P. Basic Principles of Genetics,
Connexions Web site. [http://cnx.org/content/m26565/1.1/] (2009) under a Creative
Commons Attribution License ([http://creativecommons.org/licenses/by/3.0/CC-BY 3.0]).
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Table 1
Cologuard Exact sciences/2014 Colorectal cancer Stool DNA methylation of NDRG4 and BMP3 (plus other genetic markers) FDA
ConfirmMDx MDxHealth/2012 Prostate cancer Tissue DNA methylation of GSTP1, RASSF1 and APC. LDT/CLIA
AssureMDx MDxHealth/2016 Bladder cancer Urine DNA methylation of TWIST, ONECUT2 and OTX1 (plus other genetic markers) LDT/CLIA
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Page 23
Kronfol et al. Page 24
Table 2
Mechanism of Action Active Ingredient (Trade name®, Proprietor) Date of Approval Indication(s)
DNA N-Methyltransferase Azacitidine (Vidaza®, Celgene) May 19, 2004 Chronic Myelomonocytic
Inhibitor (DNMTi) Leukemia. Myelodysplastic
Syndrome i.
Decitabine(Dacogen®, Otsuka) May 2, 2006 Chronic Myelomonocytic
Leukemia. Myelodysplastic
Syndromes ii.
i
Subtypes: refractory anemia or refractory anemia with ringed sideroblasts (if accompanied by neutropenia or thrombocytopenia or requiring
transfusions), refractory anemia with excess blasts, refractory anemia with excess blasts in transformation.
ii
Including 90 previously treated and untreated, de novo and secondary MDS of all French-American-British subtypes 91 (refractory anemia,
refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, 92 refractory anemia with excess blasts in transformation, and 93
intermediate-1, intermediate-2, and high-risk International Prognostic Scoring System groups).
iii
In patients who have received at least one prior systemic therapy.
Author Manuscript
iv
In combination with bortezomib and dexamethasone.
Author Manuscript
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Table 3
Classification of epigenetic drug classes in active clinical trials registered in the US (clinicaltrials.gov) according to mechanism of action.
Mechanism of action Active ingredient (Proprietor) Indication Clinical Trial Phase Trial Ref ID
Kronfol et al.
BET Bromodomain Inhibitors Apabetalone/RVX-208 (Resverlogix) High-risk type 2 diabetes mellitus with Phase III NCT02586155
coronary artery disease
CPI-0610 (Constellation Pharmaceuticals) Malignant Peripheral Nerve Sheath Tumor Phase II NCT02986919
DNA N-Methyltransferase Inhibitor Guadecitabine (Astex) Acute Myeloid Leukemia Phase III NCT02920008
(DNMTi)
TdCyd (NCI) Advanced Solid Tumors Phase I NCT02423057
Histone Non-Selective Entinostat (Syndax) Advanced Hormone Receptor positive (HR Phase III NCT02115282
Deacetylase +) Breast Cancer ii
inhibitors
(HDACi)
Givinostat/ITF2357 (Italfarmaco) Chronic Myeloproliferative Neoplasms Phase II NCT01761968
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Resminostat (4SC AG) Advanced Stage Mycosis Fungoides or Phase II NCT02953301
Sézary Syndrome
Quisinostat/JNJ-26481585 (Janssen) Ovarian cancer iii Phase II NCT02948075
Pracinostat/SB939 (NCIC Clinical Trials Acute Myeloid Leukemia iv, Myelofibrosis Phase II NCT01912274iv NCT02267278v
Group) v
Mechanism of action Active ingredient (Proprietor) Indication Clinical Trial Phase Trial Ref ID
AR-42 (Celgene) Relapsed multiple myeloma vi Phase I NCT02569320
HDAC 6 Selective ACY 241 (Acetylon) Non-Small Cell Lung Cancer vii Phase I NCT02635061
Histone Lysine LSD1 inhibitors GSK2879552 (GlaxoSmithKline) Myelodysplastic Syndrome Phase II NCT02929498
De-methylases
(KDM/HDM)
Tranylcypromine (Martin Luther Universität) Relapsed/Refractory Acute Myeloid Phase I/II NCT02261779
Leukemia viii
INCB059872 (Incyte) Advanced Malignancies Phase I/II NCT02712905
Histone Lysine DOT1L inhibitor Pinometostat/EPZ-5676 (Epizyme) Relapsed/Refractory Leukemias ix Phase I NCT01684150
Methyl
Transferase
(KMT/HMT) EZH1/2 inhibitor DS-3201b (Daiichi Sankyo) Lymphomas Phase I NCT02732275
EZH2 inhibitor Tazemetostat/EPZ-6438 (Epizyme) Advanced Solid Tumors or B-cell Phase I/II NCT01897571
lymphomas
MAK683 (Novartis) Diffuse Large B-cell Lymphoma Phase I/II NCT02900651
PRMT5 inhibitor GSK3326595 (GlaxoSmithKline) Solid Tumors and Non-Hodgkin’s Phase I NCT02783300
Lymphoma
i
In combination with Enzalutamide.
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
ii
In combination with Aromasin (Exemestene).
iii
In combination with Paclitaxel and Carboplatin chemotherapy.
iv
In combination with Azacitidine.
v
In combination with Ruxolitinib.
vi
In combination with Pomalidomide.
Page 26
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
vii
In Combination with Nivolumab.
viii
In combination with all-trans-retinoic acid (ATRA) chemotherapy.
ix
Only patients with rearrangements involving the MLL gene.
LSD1: Lysine (K)-specific demethylase 1A, DOT1L: Disruptor of telomeric silencing 1-like, EZH1 or EZH2: Enhancer Of Zeste Homolog 1 or 2 Polycomb Respressive Complex 2 Subunit, PRMT5:
Protein Arginine Methyl Transferase 5.
Kronfol et al.
Expert Rev Precis Med Drug Dev. Author manuscript; available in PMC 2018 January 31.
Page 27