throbber
pharmaceuticals
`
`
`
`Review
`
`Hypomethylating Chemotherapeutic Agents as Therapy
`for Myelodysplastic Syndromes and Prevention of Acute
`Myeloid Leukemia
`
`Vincent G. Sorrentino +, Srijan Thota !+®, Edward A. Gonzalez !, Pranela Rameshwar 2©,Victor T. Chang 23
`and Jean-Pierre Etchegaray '*
`
`1 Departmentof Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA;
`vincent.sorrentino@rutgers.edu (V.G.S.); st981@scarletmail.rutgers.edu (S.T.);
`egonzalez1796@gmail.com (E.A.G.)
`Departmentof Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School,
`Newark, NJ 07103, USA; rameshwa@njms.rutgers.edu
`3 Veteran Affairs New Jersey Health Care System, East Orange, NJ 07018, USA; Victor.Chang@va.gov
`Correspondence: jeanpierre.etchegaray@rutgers.edu
`+ These authors contributed equally.
`
`Abstract: Myelodysplastic Syndromes (MDSs) affect the elderly and can progress to Acute Myeloid
`Leukemia (AML). Epigenetic alterations including DNA methylation and chromatin modification
`may contribute to the initiation and progression of these malignancies. DNA hypomethylating agents
`such as decitabine and azacitidine are used as therapeutic treatments and have shown to promote
`expression of genes involved in tumor suppression, apoptosis, and immune response. Another
`anti-cancer drug, the proteasome inhibitor bortezomib, is used as a chemotherapeutic treatment
`for multiple myeloma (MM). PhaseIII clinical trials of decitabine and azacitidine used alone and
`in combination with other chemotherapeutics demonstrated their capacity to treat hematological
`malignancies and prolong the survival of MDS and AMLpatients. Although phaseII] clinical trials
`examining bortezomib’s role in MDS and AMLpatients are limited, its underlying mechanisms in
`MM highlightits potential as a chemotherapeutic for such malignancies. Further research is needed
`to better understand how the epigenetic mechanisms mediated by these chemotherapeutic agents
`andtheir targeted gene networksare associated with the development and progression of MDSinto
`AML.This review discusses the mechanisms by which decitabine, azacitidine, and bortezomib alter
`epigenetic programsandtheir results from phaseIII clinicaltrials.
`
`Keywords: epigenetic; myelodysplastic syndrome; acute myeloid leukemia; DNA methylation;
`decitabine; azacytidine; bortezomib; cancer; hypomethylating agent; myeloma
`
`
`1. Introduction
`
`Myelodysplastic syndrome (MDS) is a hematological malignancy characterized by
`aberrant hematopoiesis and most commonly affects the elderly [1]. From 2007 to 2011,
`the reported incidence of MDS per 100,000 individuals in the United States was 4.9 per
`year; today, the numberis likely higher due to increased awareness of the condition [2].
`MDSpatients display cytopenia, leading to a susceptibility for infection and bleeding[3].
`Principal diagnostic criteria for MDS include the presence of dysplasias in the peripheral
`blood and bone marrow;in bothtissues, more than onecell lineage can be affected [3]. MDS
`subtypesare defined by cytopenia type andcell lineage-specific dysplasias. For example,
`RARSor MDS-RSis defined by refractory anemia, dysplasia associated with erythropoiesis,
`the presence of ringed sideroblasts (RSBs), and erythroid precursorcells with abnormal
`iron accumulation in the mitochondria aroundthe nucleus [4]. Differential blood tests are
`helpful in prognostic classification of dysplasias and hematopoietic insufficiencies; higher
`blast counts in the bone marrow (5-10%) and low peripheral blood cell counts are often
`
`check for
`updates
`
`Citation: Sorrentino, V.G.; Thota,S.;
`Gonzalez, E.A.; Rameshwar,P.;
`Chang,V.T.; Etchegaray,J.-P.
`Hypomethylating Chemotherapeutic
`Agents as Therapy for
`Myelodysplastic Syndromes and
`Prevention of Acute Myeloid
`Leukemia. Pharmaceuticals 2021, 14,
`641. https://doi.org/10.3390/
`ph14070641
`
`Academic Editor: Marcin Ratajewski
`
`Received: 9 June 2021
`Accepted: 30 June 2021
`Published: 4 July 2021
`
`Publisher’s Note: MDPIstays neutral
`with regard to jurisdictional claims in
`published mapsandinstitutional affil-
`iations.
`
`Copyright: © 2021 by the authors.
`Licensee MDPI, Basel, Switzerland.
`This article is an open accessarticle
`distributed under
`the terms and
`conditions of the Creative Commons
`
`Attribution (CC BY)license (https://
`creativecommons.org/licenses/by/
`4.0/).
`
`CELGENE 2118
`APOTEX v. CELGENE
`IPR2023-00512
`
`Pharmaceuticals 2021, 14, 641. https: //doi.org/10.3390/ph14070641
`
`https: / /www.mdpi.com/journal/pharmaceuticals
`
`

`

`
`
`Pharmaceuticals 2021, 14, 641 2 of 36
`
`associated with more severe form of the disease [3]. The 2016 World Health Organization
`classification of myelodysplastic syndromes and neoplasmsrecognizes categories of MDS
`with single lineage dysplasia, MDS with multilineage dysplasia, MDS with ringed siderob-
`lasts, MDS with excess blasts, and MDS Unclassifiable [5]. The IPSS-Rstratifies patients at
`diagnosis into risk groups based upon cytogenetics, marrow blast proportion, hemoglobin,
`absolute neutrophil count, and platelet count [6]. High risk MDSpatients can progress
`to acute myeloid leukemia (AML)[1]. AML is a hematological cancer characterized by
`immature myeloid cell proliferation with blasts >20% and often accompanied by bone mar-
`row failure [7]. Cytological features of the bone marrow such as Auerbodies,crystalline
`rod-like structures found in the cytoplasm of leukemic myeloid cells, are common in AML
`and can be helpful in diagnosis of the disease [8].
`The last decade of research has characterized various epigenetic and genetic abnor-
`malities linked to both MDS and AML,resulting in impaired genetic machinery and
`hematopoietic stem cell function, all of which are more commonin the aged popula-
`tion [1,7]. More than 30 driver mutations have been identified in MDSpatients; these
`mutations affect DNA methylation pathways, RNA splicing, chromatin modifications,
`transcription, and signal transduction [9]. Non-random chromosomal mutations have been
`identified as genetic events that promote disease initiation and progression and are com-
`monin approximately 52% of AML patients [10]. Somatic mutations of genes that encode
`epigenetic, transcription factors and signaling proteins in AMLpatients [11], include DNA
`methyltransferase 3A (DNMT3<A), Ten-Eleven Translocation 2 (TET2), CCAAT Enhancer
`Binding Protein « (CEPBA), p53 and tyrosine kinase receptors [7]. This review selects three
`specific epigenetic drugs capable of inhibiting DNA methylation: decitabine, azacitidine,
`and bortezomib.
`Decitabine wasfirst synthesized in 1964 [12] as a cytostatic compound and then
`identified as a suitable treatment for leukemia in murinestrains with distinct MHC-II class
`in 1968 [13]. Decitabine (5-aza-2'-deoxycytidine) has been the focus of numerousstudies
`to better understand the epigenetic basis for cancer treatments. Early experimentation
`pointed to decitabine’s anti-leukemic properties in mouse models. A 1978, leukemic mice
`injected with decitabine with an 8-h infusion showedanincreasein lifespan with little to
`no toxicity [14]. Additionally, in 1983, treatment of leukemic mice with longer infusion of
`decitabine resulted in long-term survivors [15]. These early studies formed the basis for
`the use of decitabine as a chemotherapy for leukemia.
`Studies on decitabine at lower doses uncoveredits specific inhibitory effects on DNA
`methylation, and thereby identified as a hypomethylating agent (HMA)[16]. One of the
`initial studies found decitabine and azacytidine, another cytidine-analog, to induced gene
`expression via DNA demethylation (Figure 1A-C) [16]. Subsequent research confirmed
`decitabine as a DNA methyltransferase inhibitor (DNMTIi) that targets 14,000 regulatory
`regionsof genesin different cancercell types derived from lung[17], prostate [18], colon [19]
`andblood cancers [20]. Animportant study reported the use of decitabine at three low doses
`in patients diagnosed with high-risk MDS and chronic myelomonocytic leukemia (CML).
`The study foundthe best results with a dose of 20 mg/m?intravenouslyfor 5 days [21]. In
`2006, the U.S. Food and Drug Administration (FDA) approveddecitabine as a treatmentfor
`MDSand CML [22].
`
`

`

`3 of 36
`Pharmaceuticals 2021, 14, 641
`
`A Azacitidine
`
`NH
`
`2
`
`B Decitabine
`
`NH
`
`2
`
`C
`
`Cytidine
`
`NH
`
`2
`
`opt ek
`
`Ck
`

`OH
`
`HO
`
`Hot
`
`HO
`
`OH
`
`D
`Azacitidine
`
`Decitabine
`
`Vv
` yoe
` NMPKul
`
`
`
`
`“|
`yarn”
`
`
`
`
`5-aza-dCMP
`J
`{NMPK
`RNR
`5-aza-CDP >~C5-aza-dCDP
`{NDPK
`grerk
`
`
`
`RNA
`DNA
`
`
`Figure 1. Incorporating mechanismsof azacytidine and decitabine into RNA and DNA.Chemical
`structures of (A) azacytidine, and (B) decitabine, which are analogs of (C) cytidine.
`(D) Both
`azacitidine and Decitabine have similar mechanismsof integration into RNA and DNA,respectively.
`These nucleoside analogs are transportedinto the cell via human nucleoside transport (hNT) channel.
`Azacitidine (5-azacytidine) becomes phosphorylated via the uridine cytidine kinase (UCK) to form
`5-aza-CMP analog, which becomessuccessively phosphorylated via nucleoside monophosphate
`kinases (NMPK) and nucleoside diphosphatase kinase (NDPK) to create 5-aza-CDP and 5-aza-
`CTP, respectively. Decitabine (5-aza-2'-deoxycytodine) becomes phosphorylated by deoxycytidine
`kinase (DCK) to make 5-aza-dCMP analog, which becomessuccessively phosphorylated via NMUPK
`and NDPKto form 5-aza-dCDP and 5-aza-dCTP, respectively. Once these ribonucleoside and
`deoxyribonucleoside analogs are in their active states (5-aza-CTP, 5-aza-dCTP), they can replace
`cytosines in RNA and DNA,respectively. Note that the monophosphate analogs, 5-aza-CMP and
`5-aza-dCMP, can be deaminated into 5-aza-U and 5-aza-dU uridine analogs respectively, by cytidine
`deaminase (CDA), which helps maintain the surplus of pyrimidines. Additionally, the diphosphate
`analog 5-aza-CDP, can be converted into 5-aza-dCDPvia ribonucleotide reductase (RNR) by reducing
`the ribonucleoside into a deoxyribonucleoside.
`
`Azacitidine (5-azacytidine), an analog of cytidine with similar structure to decitabine,
`acts as a DNMTinhibitor by modifying the 5th carbon of the pyrimidine ring (Figure 1A).
`Instead of a carbon atom bondedto a hydrogen atom,azacitidine consists of a nitrogen atom
`
`

`

`4 of 36
`Pharmaceuticals 2021, 14, 641
`
`in the sameposition [23]. The analog wasfirst synthesized in 1963 by Piskala and Sorm
`at the Institute of Organic Chemistry and Biochemistry in Prague [24,25]. Soon after, the
`azanucleoside wasisolated from the gram-positive bacteria Strepotoverticillium ladakanum,
`andits functionality with respect to cytotoxicity and anti-proliferation, were shown[26].
`After initial discovery and synthesis, azacitidine was used for its anti-metabolite properties
`in disturbing normal metabolic processes, leading to its use as a chemotherapeutic for
`leukemia [23]. In 1971, the nucleoside analog was used for its cytostatic properties in
`chemotherapy, as it provedefficacious in the treatment of childhood leukemia. In the 1980s,
`the demethylating properties of the drug were identified. This led to further studies and
`clinical trials to investigate the drug as an epigenetic modulator [25]. With the linkage
`between DNA hypermethylation and the development of cancer, azacitidine, as a hy-
`pomethylating agent, was quickly soughtafter for its antineoplastic properties, specifically
`throughreactivation of previously silenced genes, including tumor-suppressors[23,27].
`In 2004, nearly 40 yearsafter its discovery, Azacitidine (Vidaza; Celgene), an injectable
`suspension, was approved by the FDA, followed by approvalin 2008 by the European
`Medicines Agency (EMA)[23,27]. Vidaza, a bioavailable formulation of 100 mgof azaciti-
`dine and 100 mg mannitol, has been used for subcutaneous administration in MDS, AML
`and CMML patients [23]. Currently, the hypomethylating agents azacitidine and decitabine,
`are the recommendedtherapeutic treatment for MDSpatients classified by the International
`Prognostic Scoring System (IPSS) at intermediate-2/high risk [28]. Subcutaneousinjections
`of azacitidine, however, require the need for in-person treatments and potentially cause
`injection site infections; this can be detrimental for AML patients with severe alterations
`in bone marrowblasts. Recently, azacitidine has becomeavailable as an oral formulation
`CC-486 (ONUREG;Celgene), whichis being clinically tested in patients with MDS, AML,
`and CMML[29,30]. This version was approved by the FDA in 2020 as a maintenance
`treatment for AML patients whofailed intensive induction chemotherapy and achieved
`a first complete remission due to the successful trials in the QUAZAR AML-001 study
`(ClinicalTrials.gov Identifier: NCT01757535) on 1 September 2020 [31].
`In addition to hypermethylation, aberrant proteolysis has been associated with the
`developmentof particular malignancies; as such, targeting protein degradation processes
`has been the focus of many researchers interested in developing anticancer drugs [32].
`Oneof the crucial pathwaysinvolved in protein degradationis the ubiquitin-proteasome
`pathway (UPP), which is responsible for targeting degradation of approximately 80% of
`cellular proteins [33]. This pathway is executed by the proteasome: a multiprotein complex
`that is responsible for the recognition and degradation of ubiquitin-markedproteins[33].
`Malignantcells tend to accumulate defective proteins due to their increased synthesis
`capability; this increases their reliance on the proteasome’s disposal mechanism [34,35].
`Therefore, any compound that interferes with proteasomal disposal of proteins could
`result in the accumulation of defective proteins, increased cellular stress, and apoptosis
`of malignantcells [35]. Since the proteasome degrades and processes mediators of the
`cell-cycle and apoptosis such as cyclins, caspases and nuclear factor of kB (NF-kB) [35],
`this molecule also drives the cell cycle via regulation of protein levels that activate and/or
`inhibit phase transitions in cell growth and replication [36]. Moreover, several studies have
`demonstrated proteasomeinhibition results in cell death [33] due to impaired degradation
`of p53 [37] and p21 [38].
`The chemotherapeutic effect of bortezomibrelies on its ability to regulate protein
`turnover. Bortezomib is a proteasome inhibitor knownto induce apoptosis in malignant
`hematopoietic cells [35,39]. First synthesized by Myogenics in 1995, bortezomib showed
`early in vitro and in vivo results in clinical studies [32] and wasthe first proteasome in-
`hibitor to be usedclinically in the treatment of malignancies, namely refractory/relapsed
`myelomas[40]. After extensiveclinicaltrials, it was approved by the FDA in 2003 for the
`treatmentof relapsed multiple myeloma[32,41]. Current research investigates bortezomib-
`mediated mechanismsassociated with epigenetic pathways implicated in cancer develop-
`ment and progression [42].
`
`

`

`
`
`Pharmaceuticals 2021, 14, 641 5 of 36
`
`Determining the mechanisms and targeted genes by decitabine, azacytidine and
`bortezomib remain anactive area of research in the context of hematological malignancies
`and other cancers. The purposeof this review is to underline relevantfindings with respect
`to each of these drugs in MDS and AML.Theidentification of new epigenetic biomarkers
`linked to these drugs, in the context of MDS and AML,can be used for developing new
`targeted therapies to increase efficacy of existing treatments.
`
`2. Main Body of Review
`2.1. Decitabine (5-Aza-2'-Deoxycytidine)
`2.1.1. Mechanism of Action
`
`Decitabineis a cytidine analog where nitrogen replaces the carbonfive in the pyrimi-
`dine ring (Figure 1B,D). In general, decitabine represents a DNA hypomethylating agent
`with two main mechanismsofaction (Figure 2). At low doses, decitabine reactivatessi-
`lenced genes and promotescellular differentiation. At high doses,it elicits cytotoxic effects,
`leading to cell death [16,22]. In general, azanucleosides like decitabine are chemically
`unstable and considerations must be taken whenidentifying safe and effective methods to
`administer such drugs[43]. Decitabine and its metabolites bind moretightly to enzymes
`that mediate its incorporation into DNA when comparedto the natural substrate; for exam-
`ple, the drug binds 10 times moretightly to cytidine deaminase than deoxycytidine [44].
`Like many drugs, the effectiveness of decitabine treatment is dependent upon the
`targeted cells to transport the drug [43]. Mechanistically, decitabine is incorporated
`intracellularly by nucleoside transport proteins, including the equilibrative uniporters
`(ENTs; SLC29A family) and concentrative uptake transporters (CNTs; SLC28A family),
`which have both been directly linked to the uptake of chemotherapeutic analogues in
`the treatment of leukemias [43]. The drug is then targeted by deoxycytidine kinase,
`which converts decitabine (5-aza-dCR) to 5-aza-2'-deoxycytidine monophosphate (5-aza-
`dCMP)(Figure 1D) [43]. Through a series of phosphorylation reactions by nucleoside
`di-phosphokinase[44,45], 5-aza-dCMPis further converted into the active nucleotide form
`5-aza-2'-deoxycytidine-5’-triphosphate (5-aza-dCTP), which can substitute for cytosine
`during DNAreplication and thereby incorporated into DNA (Figure 1D)[43,46]. DNA
`methyltransferase enzymeswill then recognize 5-aza-dCTP-guanine dinucleotides for DNA
`methylation (Figure 2) [43]. DNMT1 becomesinactivated by forming an irreversible cova-
`lent bond with the 5-aza-dCTP-guanine dinucleotide (Figure 2) [47]. This covalent bond
`results in a rapid loss of DNMTactivity as bound enzymesare unableto carry out further
`downstream methylation activity [48], leading to global hypomethylation (Figure 2) [45].
`This process is known as enzyme DNA adduct formation. Specifically targeting the S-phase
`of the cell cycle [44], decitabine leads to antitumoraction via inhibition of proper DNA
`replication in cancerouscells; however, the drug also interferes with transcription and
`DNArepair processes (Figure 2) [49].
`Experimental evidence suggests that the effectiveness of low-dose decitabine in ma-
`lignancies stems from its ability to demethylate silenced tumor-suppressor genesrelated
`to leukemic malignancies and other myeloid disorders [49] such as p15'NS4>, E-cadherin
`and MYOD1 [50,51]. Moreover, various distinct methylation patterns have been iden-
`tified in AMLpatients, some predictive of clinical outcomes [20]. As mentioned above,
`nucleoside uptake is an important process in the effectiveness of decitabine treatment
`regimens. Reduced expression of uptake transporters like SLC22A4 have been reported
`as strong predictors of poor event-free and overall survival in AMLpatients [52]. DNA
`methylation-based epigenetic repression could be a contributing factor to such poor out-
`comes, as pre-treatment with decitabine restored SLC22A mRNA expression, increased
`cellular uptake of anthracyclines, and wasassociated with increased sensitivity to cytara-
`bine, a chemotherapeutic, in human AML cell lines [52].
`
`

`

`6 of 36
`Pharmaceuticals 2021, 14, 641
`
`
`
`SESSASes Genomic DNA @ Methylated Cytosine
`
`© Unmodified Cytosine
`
`— —> —> 6—__-9+—__A+—_ ©_Decitabine
`DCK
`NMPK NDPK
`NDPK
`RNR
`NMPK UCK
`© 5-aza-dCTP
`|
`W@W Azacitidine
`
`DNAReplication
`
`Maintenance
`
`DNA methylation <p «me DNA methylation
`
`De novo
`
`@ 5-azadCDP
`
`X_
`
`Irreversible bond with DNMTs
`
`"WV Newly synthesized DNA strand
`
`AAA. Parent DNA strand
`
`Inactivated DNMTs
`
` O
`SUSASASASASATAURNRTAN
`
`OPO
`OOO
`Anti-tumoreffect:
`{|
`.
`Inhibition of cell cycle S-phase
`DNA Hypomethylation Cc Activation of tumor suppressors
`Loss of epigenetic memory
`Activation of apoptotic genes

`Impairs DNA repair
`
`Figure 2. Inhibition of DNMTs uponincorporation of decitabine and azacytidine into DNA. The genomic DNA methylation
`landscape consists of methylated and unmethylated cytosines. Upon multiple phosphorylation steps by DCK (deoxycytidine
`kinase), NMPK (nucleoside monophosphate kinases) and NDPK(nucleoside diphosphatase kinase), decitabine becomes
`5-aza-dCTP, which gets incorporated into DNA. Upon phosphorylation steps by UCK(uridine cytidine kinase) and NMPK
`(nucleoside monophosphate kinases), azacytidine is converted into 5-aza-CDP, which is recognized by ribonucleotide
`reductase (RNR) leading to the formation of 5-aza-dCDP. This reactive analog is further phosphorylated to 5-aza-dCTP by
`NDPK(nucleoside diphosphatase kinase), which is then integrated into genomic DNA. Upon DNAreplication, DNMT1
`maintains the methylation status of the genome. However, DNMT1 becomescovalently boundto the cytosine analog, 5-aza-
`dCTP, which prevents DNMT1activity leading to genomic hypomethylation andloss of epigenetic memory. Additionally,
`other DNMT enzymes, DNMT3a and DNMT3b,can also form anirreversible interaction with azanucleosides leading to
`hypomethylation of the genome. Collectively, this drug-mediated DNA hypomethylation causes anti-tumorigenic effects
`includinginhibition of the cell cycle, DNA repair impairments, activation of pro-apoptotic and tumor suppressor genes.
`
`Decitabine has also been shownto induce the expression of tumor-associated anti-
`gens, resulting in induced immunecytotoxiceffect, indicating its indirect role in immune
`therapy [49]. MDSpatients treated with 1.3 uM decitabine at a dose of 15 mg/m?/day
`showed improvedexpression of cancer-testis antigens (CTAs; ie. MAGE-A1, MAGE-A3,
`and SP17) against solid tumors [53]. Decitabine treatments were accompanied by en-
`hanced T-lymphocyte recognition of MDScells, specifically in response to increased CTA
`expression [53]. This study also found decitabine treatments in MDSpatients to increase
`T-lymphocyte function, expression of HLAclass antigen and ICAM-1, a cell adhesion and
`co-stimulatory molecule in adaptive immunity responses [53,54]. This underlines oppor-
`tunities for decitabine treatment regimensto be used in-tandem with immunotherapies
`alreadyin use and highlights the wide variety of genetic targets affected by decitabine’s
`mechanism ofaction.
`
`

`

`
`
`Pharmaceuticals 2021, 14, 641 7 of 36
`
`2.1.2. Decitabine Hematological Malignancies
`Hypomethylating agents are attractive because of their reduced toxicity in elderly
`patients when compared to standard induction chemotherapy [55]. As mentioned above,
`anti-tumoractivity of decitabine in hematological malignancies results from multiple mech-
`anismsincluding, induction tumor-suppressor genes upon hypomethylation, enzyme DNA
`adduct formation, activation of apoptotic pathways and induction of tumor-associated
`antigens [49,53]. In AML cell lines such as OCI-AML2, decitabine was found to induce
`the expression of 81 genes while inhibiting the expression of 96 genes; whereas, normal
`peripheral blood cells displayed significantly fewer changes in gene expression [56]. In-
`terestingly, this study showedthat nearly 50% of decitabine-induced genes are deprived
`of CpG methylation sites in their promoter regions, which suggest a decitabine-mediated
`effect that is independent of DNA methylation status.
`In patients with hematological malignancies like MDS and AML, decitabineis at its
`most effective when used over a prolonged period with fractionated exposures [57,58].
`Moreover, responserates to decitabine tend to be slow, with mostpatients requiring at least
`twoto five monthly cycles to achieve maximum clinical results [59]. DNA demethylation
`in responseto decitabine is a function of the dose, in which lowerdoses (0.1 uM) induced
`higher levels of DNA hypomethylation, whereas very low (0.01 uM) and high (10 uM)
`decitabine doses induced significantly lower DNA hypomethylation with concomitant
`higherrate of cytotoxicity. A 2017 study examinedtheeffect of low dose (20 mg/m?/day x
`5 days) decitabine treatments in lower risk MDSpatients. The results show 70% of patients
`displayed an overall response rate (ORR) to decitabine treatments and 32% of these pa-
`tients became blood transfusion-independent[60], demonstrating an overall hematological
`improvement. Onetrial interested in dose/schedule dependent responses to the drug
`enrolled MDSpatients in one of twodistinct treatment regiments: 3-day treatments (3 h
`IV infusion of 15 mg/m? given every 8 h for 3 consecutive days every 6 weeks) or 5-day
`treatments (1 h IV infusion of 20 mg/m? given once daily on days 1-5, every 4 weeks;[61]).
`Overall Response Rate for the 3-day group was 29.4% and 25.5% for the 5-day group
`with the median AML-free survival time was 23.8 months, and 24-month overall survival
`was 48.9%[61].
`Recent phaseIII clinical trials have demonstrated varied success with decitabinetreat-
`ments. Thesetrials typically use criteria like progression-free survival (PFS) defined as
`duration from the date of treatment to progressive disease or death, event-free survival
`(EFS) defined as the time from study entry until relapse/malignancy/death, and overall
`response rate (ORR) defined as the percentage of participants who achieved stringent
`complete response, very good partial response, or partial response, to quantify the effec-
`tiveness of the drug therapy in question. In a study comparing decitabine (15 mg/m? every
`8 h for 3 days) with best supportive care (BSC)in elderly patients (>60 years) with MDS,
`15%of those receiving decitabine showed complete/partial remission and hematological
`improvement; patients receiving decitabine also had longer PFS than those receiving BSC,
`but similar overall survival (OS) in both groups [62]. Another phase III randomized study
`looking at decitabine in individuals with MDS found an overall improvementin patient
`outcomes whentreated with the drug. Patients received either 15 mg/m? over 3 h every
`8 h decitabine IV repeated every 6 weeks, or BSC; decitabine patients showed an ORR of
`17%, with 9% complete responses, compared to 0% response for BSC group [63]. Decitabine
`responseswereclassified as durable (median, 10.3 months) and prolonged patients’ median
`time to AML progression when comparedto BSC patients [63].
`Additionally, researchers have been searching for improved biomarkers of predictive
`of clinical outcomes in MDS/AML patients undergoing decitabine treatment. A trial in
`patients with MDS whohave unfavorable-risk cytogenetic profiles and TP53 mutations,
`have shown increased response rates to decitabine in MDS treatmentprotocols [19,59];
`therefore, higher-risk individuals may be moresensitive to the drug. Anothertrial looking
`at high-risk MDSpatients with varying cytogenetic profiles found decitabine responses
`specific to patients’ cytogenetic profile, namely regarding autosomal monosomies (MK-—,
`
`

`

`
`
`Pharmaceuticals 2021, 14, 641 8 of 36
`
`MK+, MK1, MK2+) [64]. ORR in cytogenetically normal (CN)patients was 36.1%, 16.7% in
`MK-patients, and 43.6% in MK+ patients; PFS was prolonged in CN and MK2+ groups
`but not MK—/MK+/MkK1 subgroups when compared to BSC patients [64]. One in vivo
`study found elevated fetal hemoglobin (HbF)to beareliable predictor of overall survival
`in MDS/AMLpatients, with decitabine elevating HbF levels in 81% of MDSpatients and
`54% of AML patients [65]. These studies underline the importance of patient karyotype
`whendeciding which treatment regimens may be mosteffective in MDS/AMLtreatment
`and whatphysiological signals clinicians should assess for reliable predictors of patient
`outcome.
`
`2.1.3. Side Effects and Complications
`During decitabine’s initial development phase in the mid-1980s, toxicities associated
`with the drug’s use included myelosuppression, nausea, and mild to moderate fatigue [16].
`Contemporary studies report neutropenia, febrile neutropenia, thrombocytopenia, leukope-
`nia, anemia, fatigue, nausea, diarrhea, and constipation [66]. In general, one of the main
`concerns with the use of decitabine is its cytotoxic effects at high doses [45], especially
`at non-target sites. Despite this concern, decitabine and azacitidine tend to haveless cy-
`totoxic effects compared to other more conventional chemotherapeutics; this is because
`the dosage neededto elicit desirable epigenetic effects is relatively low comparedto the
`dosage that elicits cytotoxic effects [16]. Possible myelosuppressive complications like
`infection/bleeding, or prolonged myelosuppression [defined as hypocellular marrow]
`havealso been reported but tend to be less common than the milder adverse side effects
`mentioned above [21].
`Despite success with decitabine in the treatment of hematological malignancies, some
`MDS/AML patients do not respondeffectively to the drug [67]. Many patients fail to respond
`to initial decitabine treatments (primary resistance); those that do respondinitially often
`relapse and become unresponsive to subsequent exposures (secondary resistance) [68,69].
`Moreover, treatment with decitabineis not curative; different forms of resistance to the drug
`have been identified over the years [70], most notably dueto alterations in the pathwaysthat
`activate and metabolize decitabine, as well as downstream mutations of genes involved in
`DNAmethylation/ demethylation dynamics such as TET2 [71], DNMT3A, and ASXL1[72].
`Thus, a better understanding of these contributing pathways can help researchers and
`clinicians identify which mechanisms should be targeted at different stages of MDS and
`AMLprogression to develop salvage therapies.
`Researchers have begun looking for waysto bolster the effects of decitabine using
`combination therapies with other drugs. Polo-like kinases regulate thecell cycle [73], vari-
`ousportions of mitosis, and contributes to DNA damagerepair and replication stress [74];
`therefore, these proteins make a reasonable target for anticancer therapeutics. Volasertib
`andrigosertib, two polo-like kinase inhibitors currently under phaseIII trial review, are
`currently being used in combination with decitabine in the treatment of MDS/AML pa-
`tients whoare ineligible for intensive remission therapy [75]. 3-Dezazneplanocin (DZNep),
`a histone methyltransferase inhibitor, has also been identified as a drug that could bolster
`decitabine’s efficacy in MDS/AMLpatients [67]. Together, decitabine and DZNep have
`demonstrated synergistic activation of several tumor suppressor genes and synergistic
`activation of apoptosis in human AML cell lines HL-60 and AML-30 [67]. These types of
`studies widen options for developing new combination treatment regimens.
`
`2.2. Azacitidine
`2.2.1. Mechanism of Action
`
`Similar to decitabine, azacitidine elicits two distinct properties, cytotoxicity and DNA
`hypomethylation, depending on dosage schedules. At high dosage, azacitidine promotes
`cytotoxicity due to its incorporation into both RNA and DNA,while low dosage prompts
`DNAhypomethylation effects (Figures 1D and 2D)[76]. The anti-proliferation effect on
`abnormally dividing hematopoietic cells in the S-phase of the cell cycle results from
`
`

`

`
`
`Pharmaceuticals 2021, 14, 641 9 of 36
`
`interference with nucleic acid metabolism [77]. Thereby, further nucleic acid synthesis and
`cellular proliferation are impaired, and apoptotic pathwaysareactivated.
`Azacitidine’s effect is initiated after intake of the oral CC-486 or injection by Vidaza
`via cellular uptake carried out by specific transmembraneproteins; the nucleoside analog
`is transported into a cell via a humanconcentrative nucleoside transport 1 (hCNT1) which
`is part of the SLC28 gene family of three subtypes that transport naturally occurring
`nucleosides and synthetically engineered nucleoside analogs [23]. Upon transport into
`the cell, azacytidine is phosphorylated by uridine cytidine kinase to 5-azacitidine 5’-
`triphosphate, its active conformation [23]. The compoundis then degradedor integrated
`into the nucleic acids. Triphosphate nucleosides are degraded in the cells by cytidine
`deaminase (CDA) and converted to 5-azauridine compounds, rendering them inactive [23].
`Concentration and synthesis of CDA in humanliver and spleen influences the half-life
`of the drug, making it approximately 41 min in vivo. Moreover, reducedlevels of active
`drug by degradation decreases the drug’s efficacy and potency. The various inactive
`metabolites of azacitidine are primarily discarded via urine secretion with minimal (<1%)
`fecal excretion [78].
`Azacitidine is incorporated into the genomeof rapidly proliferating cells during the S-
`phase[23,79] and does not show sufficient affinity for non-proliferating cells [80]. Upon in-
`tegration into RNA,azacitidine interrupts RNA metabolism andtranslation processes. Fur-
`thermore, azacitidine can also incorporate into DNAbyribonucleotide reductase-mediated
`conversion of azacitidine diphosphate into 5-aza-2'-deoxycytidine diphosphate. Phospho-
`rylation of the diphosphate into triphosphate enables azacitidine to be integrated into DNA
`during replication [27]. Azacitidine has an increased affinity to RNA over DNA with a
`ratio of incorporation of 65:35, respectively, in AMLcell lines [76].

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket