throbber
Hematologic Malignancies
`
`Oral Azacitidine (CC-486) for the Treatment of Myelodysplastic
`Syndromes and Acute Myeloid Leukemia
`CHRISTOPHER R. COGLE,a BART L. SCOTT,b THOMAS BOYD,c GUILLERMO GARCIA-MANEROd
`aDivision of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA;
`bFred Hutchinson Cancer Research Center, Seattle, Washington, USA; cNorth Star Lodge Cancer Center, Yakima, Washington, USA;
`dDepartment of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
`Disclosures of potential conflicts of interest may be found at the end of this article.
`
`Key Words. Leukemia x Myeloid x Acute x Myelodysplastic syndromes x Azacitidine
`
`ABSTRACT
`
`The myelodysplastic syndromes (MDS) comprise a heteroge-
`neous group of clonal myeloid malignancies characterized by
`multilineage cytopenias, recurrent cytogenetic abnormali-
`ties, and risk of progression to acute myeloid leukemia (AML).
`AML, which can occur de novo as well as secondary to MDS, is
`characterized by malignant clones of myeloid lineage in the
`bone marrow and peripheral blood, with dissemination into
`tissues. The cytidine nucleoside analog and epigenetic
`modifier azacitidine is approved in the U.S. for the treatment of
`all French-American-British subtypes of MDS and in many
`countries for the treatment of AML with 20%–30% blasts and
`multilineage dysplasia according to the World Health Organi-
`zation classification. Benefits of azacitidine treatment of
`patients with AML with .30% blasts have also been shown
`in a recent phase III trial. Oral administration of azacitidine may
`
`enhance patient convenience, eliminate injection-site reac-
`tions, allow for alternative dosing and scheduling, and enable
`long-term treatment. Phase I studies with oral azacitidine
`(CC-486) have shown biological activity, clinical responses,
`and tolerability in patients with MDS and AML. Extended
`dosing schedules of oral azacitidine (for 14 or 21 days of
`28-day cycles) are currently under investigation as frontline
`therapy in patients with lower risk MDS, as maintenance
`therapy for patients with AML not eligible for stem cell
`transplant, and as maintenance therapy for patients with
`MDS or AML following stem cell transplant. This review
`presents clinical data supporting the use of injectable
`azacitidine in MDS and AML and examines the rationale for
`and results of the clinical development of oral azacitidine.
`The Oncologist 2015;20:1404–1412
`
`Implications for Practice: Injectable azacitidine can prolong survival, reduce transfusions, and improve quality of life compared
`with conventional care regimens in patients with higher-risk myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML).
`An oral formulation improves convenience and eliminates injection-site reactions but also enables testing of novel, longer term,
`low-dose schedules that may enhance therapeutic activity of azacitidine by increasing exposure to cycling malignant cells. In early
`phase trials, oral azacitidine (CC-486) in extended dosing regimens was biologically and clinically active in patients with MDS and
`AML. Oral azacitidine is being further evaluated in an ongoing phase III program.
`
`INTRODUCTION
`
`The myelodysplastic syndromes (MDS) are a heterogeneous
`group of clonal myeloid malignancies arising from a stem cell
`source and characterized by multiple genetic abnormalities and
`subclonal architectures [1–4]. There is considerable heteroge-
`neity in genetic mutations among patients with MDS that may
`explain the diversity of clinical presentations that differ in the
`numbers and depths of cytopenias, risks toward progression
`to acute myeloid leukemia (AML), responses to treatment, and
`survival times [5–8]. Gene mutations affecting epigenetic
`chemical modifications, such as mutations in TET2 and DNMT3A,
`are among the most common in MDS [9–11].
`
`MDS is primarily a disease of older adults, and advanced
`age at diagnosis is associated with decreased overall survival
`(OS) [12, 13]. The reported incidence rate of MDS overall is
`5.70 per 100,000 persons in the U.S. [14] and 1.82 per 100,000
`persons in Europe [15]. In patients aged $65 years, the
`incidence rate is higher, at 12.97 per 100,000 persons or higher
`in the U.S. [14] and 5 per 100,000 persons or higher in Europe
`[15]; however, these are likely underestimates of true in-
`cidence. Large numbers of MDS cases go unreported by state
`cancer registries due to difficulties in disease diagnosis, under-
`appreciation of MDS as a malignancy, under-reporting by
`
`Correspondence: Christopher R. Cogle, M.D., University of Florida, Medicine/Hematology and Oncology, 1600 SW Archer Road, Box 100278,
`Gainesville, Florida 32610, USA. Telephone: 352-273-7493; E-Mail: christopher.cogle@medicine.ufl.edu Received April 22, 2015; accepted for
`publication August 21, 2015; published Online First on October 13, 2015. ©AlphaMed Press 1083-7159/2015/$20.00/0 http://dx.doi.org/
`10.1634/theoncologist.2015-0165
`CELGENE 2073
`APOTEX v. CELGENE
`IPR2023-00512
`©AlphaMed Press 2015
`
`The Oncologist 2015;20:1404–1412 www.TheOncologist.com
`
`

`

`Cogle, Scott, Boyd et al.
`
`1405
`
`outpatient clinics, changing guidelines for defining MDS,
`and lack of resources [15–18]. When accounting for the
`uncaptured cases, MDS is one of the most common he-
`matologic malignancies.
`The current MDS treatment paradigm is based on prog-
`nostic risk assessment [8, 13, 19, 20]. Patients at lower risk for
`early death or evolution to AML are treated with agents
`to primarily reduce or eliminate blood transfusions and to
`improve quality of life (QOL), whereas patients at higher risk for
`early death or AML progression are generally treated with
`more intensive therapies aiming to induce disease remission
`and lengthen survival [8, 21, 22].
`AML is a multigenetic malignancy characterized by ma-
`lignant clones and subclones of myeloid lineage in the bone
`marrow and peripheral blood, with dissemination into tissues
`[23–25]. It is the most common acute leukemia in adults [23],
`with an estimated incidence rate of 4.06 per 100,000 persons in
`the U.S. [14] and 3.62 per 100,000 persons in Europe [15].
`Similar to MDS, AML incidence increases with advancing age,
`with incidence rates in the U.S. and Europe of 10 per 100,000
`persons or higher for patients aged $65 years [14, 15]. AML
`has a poor prognosis, particularly in older patients and those
`with adverse disease characteristics (e.g., secondary AML,
`complex cytogenetic abnormalities, or FLT3 mutation) [26–30].
`Three-quarters of patients with AML die within 5 years of
`diagnosis, and survival decreases with increasing age [26].
`Disease characteristics and performance status are closely
`considered when evaluating patient eligibility for treatment
`with intensive therapies (e.g., induction followed by consol-
`idation chemotherapy or stem cell transplant [SCT]) versus
`low-intensity options such as injectable azacitidine or de-
`citabine [27–29, 31–33].
`
`AZACITIDINE FOR INJECTION
`Azacitidine is a cytidine nucleoside analog with a substitution
`of the carbon at position 5 with nitrogen that prevents
`methylation by covalently binding DNA methyltransferases
`[32, 34] (Fig. 1). The epigenetic modifier azacitidine is in-
`corporated into DNA and RNA [34, 35]. Antileukemic effects of
`azacitidine are thought to include direct cytotoxicity from
`inhibition of protein synthesis and DNA damage and re-
`expression of aberrantly silenced tumor suppressor genes due
`to DNA hypomethylation [32, 34–37]. Azacitidine is approved
`in the U.S. for the treatment of all French-American-British
`(FAB) subtypes of MDS [32] and is approved in many countries
`(e.g., European Union, Australia, Republic of Korea, Taiwan)
`for patients not eligible for SCT with intermediate-2 (Int-2)
`and high-risk MDS according to the International Prognostic
`Scoring System (IPSS) and patients with AML with 20%–30%
`blasts and multilineage dysplasia according to World Health
`Organization (WHO) classification [38]. Azacitidine is also
`approved by the U.S. Food and Drug Administration for chronic
`myelomonocytic leukemia (CMML) and by the European
`Medicines Agency for “CMML with 10%–29% marrow blasts
`without myeloproliferative disorder.”
`The approved dosing of azacitidine is 75 mg/m2 per day on
`days 1–7 of 28-day cycles [32, 38]. Alternative dosing schedules
`have been explored, including 5-day and 5-2-2-day dosing
`regimens to avoid weekend administration [39–41], but are
`not approved. Although hematologic improvement (HI) with
`
`Figure 1. Structure of azacitidine. Substitution of carbon at
`position 5 with nitrogen prevents DNA methylation [34].
`
`5-day dosing in patients with MDS is compelling, clinicians
`should not extrapolate data for injectable azacitidine based on
`the approved 7-day dosing regimen, especially in patients with
`higher risk MDS. The 5-day and 5-2-2-day alternate regimens
`require more rigorous testing.
`Initial approval was based primarily on results from two
`phase III studies. CALGB-9221 was a phase III multicenter
`randomized controlled trial of subcutaneous (s.c.) azacitidine
`(75 mg/m2 per day on days 1–7 in 28-day cycles; n 5 99) versus
`best supportive care (BSC; n 5 92) in patients with all FAB
`MDS subtypes [42]. In this trial, the overall response rate (ORR)
`with azacitidine was significantly improved versus BSC (60%
`vs. 5%, p , .0001) [42]. Response rates to azacitidine were
`comparable in patients with lower and higher risk MDS [42].
`Myelosuppression was the most common toxicity, and adverse
`events (AEs) were generally transient, resolving before the
`next treatment cycle [42].
`AZA-001 was a phase III multicenter randomized con-
`trolled trial of s.c. azacitidine (75 mg/m2 per day on days 1–7
`in 28-day cycles) versus conventional care regimens (CCR:
`BSC, low-dose cytarabine [LDAC], or intensive chemotherapy
`[IC]) in patients with IPSS-defined higher risk (Int-2 or high-
`risk) MDS (N 5 358) [43]. Azacitidine treatment resulted
`in significantly longer OS (median: 24.5 vs. 15 months;
`p 5 .0001), higher rates of hematologic response (p 5 .0001)
`and HI (p , .0001) assessed by International Working Group
`(IWG) 2000 criteria [21], and longer durations of response
`(median: 13.6 vs. 5.2 months; p 5 .0002) versus CCR [43].
`Azacitidine prolonged OS compared with CCR regardless of
`IPSS cytogenetic risk group [43].
`A multivariate analysis of AZA-001 showed that achieve-
`ment of hematologic response or HI (IWG 2000 criteria [21])
`was associated with improved OS with azacitidine treatment
`[44], and patients who achieved a hematologic response to
`azacitidine had significantly prolonged OS and reduced risk of
`death versus patients who achieved a response to CCR [44]. In
`addition, stable disease or achievement of complete re-
`sponse (CR), marrow CR (mCR), partial response (PR) or HI
`(IWG 2003 [45] and 2006 criteria [22]) with alternative dosing
`schedules of azacitidine has been shown to significantly
`reduce the risk of death versus disease progression (p , .001)
`[39].
`In the AZA-001 study, the median time to first response
`with azacitidine was 2 cycles (range: 1–16), with 91% of
`responding patients achieving first response within 6 cycles
`and all but 1 achieving first response by cycle 12 [46].
`Continued treatment improved response quality in 48% of
`patients [46], with a median of 3 cycles from first to best
`
`www.TheOncologist.com
`
`©AlphaMed Press 2015
`
`

`

`1406
`
`Oral Azacitidine (CC-486) for MDS and AML
`
`response. By cycle 12, 92% of responding patients achieved
`their best response. Based on this experience, some cli-
`nicians have advised administering at least six cycles of
`azacitidine [47], and the National Comprehensive Cancer
`Network guidelines for MDS recommend at least four to six
`cycles before assessing for treatment failure [8]. Assess-
`ments for treatment failure include evaluation of peripheral
`blood, bone marrow aspiration or biopsy, cytogenetic testing,
`and genetic studies. If response is achieved, azacitidine is
`typically continued until disease progression, unacceptable
`toxicity, or definitive therapy with SCT. If no response is
`achieved after six cycles of azacitidine, then prognosis is
`generally poor [48, 49]. Small case series have shown that
`decitabine treatment after azacitidine failure yields modest
`responses (ORR: 0%–28%) that are generally short lived [48,
`50–53]; however, this is not currently standard treatment
`practice. The current standard of care for patients with MDS
`who have not responded after at least six cycles of azacitidine
`treatment is to consider clinical trials [48, 49] or proceed
`with SCT [51].
`The most common grade 3/4 AEs with azacitidine treatment
`in patients with MDS in the AZA-001 trial were cytopenias [43],
`and most were transient and resolved during therapy [54].
`The highest rates of AEs reported with azacitidine in AZA-001
`occurred during cycles 1–2, with decreased frequency with
`continued treatment. Median duration of hematologic AEs
`was 14–16 days.
`Azacitidine has also been investigated for the treatment of
`patients with AML. In a subanalysis of AZA-001 of low-blast-
`count AML (20%–30% blasts; n 5 113), the 2-year OS rate for
`patients treated with azacitidine was 50%, and the median OS
`was 24.5 months compared with 16.0 months for patients
`treated with CCR (p 5 .005) and 16.4 months for patients not
`preselected to receive IC (BSC or LDAC; p 5 .004) [31]. Survival
`benefits with azacitidine may not require CR because CR rates
`were similar for azacitidine versus CCR (18% vs. 16%; p 5 .8),
`whereas rates of red blood cell transfusion independence
`(RBC-TI) were significantly higher with azacitidine (41% vs.
`18%; p 5 .04) [31].
`Recently, the global phase III randomized open-label AZA-
`AML-001 study of azacitidine (75 mg/m2 per day on days 1–7 in
`28-day cycles) versus CCR extended these findings in patients
`aged $65 years with newly diagnosed AML with .30% blasts
`(N 5 488) [55]. Azacitidine treatment demonstrated a clinically
`meaningful improvement in median OS of 10.4 months versus
`6.5 months with CCR (p 5 .1009). Azacitidine significantly
`improved 1-year survival: 46.5% versus 34.2% with CCR,
`a 12.3% difference (95% confidence interval: 3.5%–21.0%). In
`patients who did not achieve CR (IWG 2003 criteria [45]),
`median OS was prolonged from 6.9 months with azacitidine
`versus 4.2 months with CCR (p 5 .0170).
`The safety profile of azacitidine for the treatment of AML
`was consistent with previous observations [31, 43, 55]. The
`most common grade 3/4 hematologic AEs were cytopenias,
`which occurred more frequently during earlier treatment
`cycles [31, 55].
`
`RATIONALE FOR ORAL ADMINISTRATION OF AZACITIDINE
`Oral administration of azacitidine avoids injection-site re-
`actions and may enhance patient convenience compared
`
`©AlphaMed Press 2015
`
`with an injectable formulation [56, 57]. It allows for the
`evaluation of alternative doses and schedules,
`including
`extended dosing schedules. Early trials with injectable
`azacitidine showed decreased toxicity and increased efficacy
`at lower doses over several days versus a single higher dose
`[58–61]. In addition, in patients with MDS, continued treat-
`ment with s.c. azacitidine was shown to improve response
`quality [46].
`The benefits of extended azacitidine dosing and long-
`term treatment are likely related to the impact on hypo-
`methylation. Hypomethylating effects are cell cycle dependent
`[62], and serial cycles of DNA replication are needed to in-
`duce hypomethylation [63, 64]. Extensive demethylation
`requires prolonged drug exposure [63]. Due to the short
`plasma half-life of azacitidine [34, 38] and cell cycle-restricted
`DNA incorporation [62], extended dosing schedules enabled
`by oral administration have the potential to enhance clinical
`activity of azacitidine by increasing exposure to cycling ma-
`lignant cells.
`
`CLINICAL INVESTIGATIONS WITH ORAL AZACITIDINE
`(CC-486)
`
`Pilot Study of Oral Azacitidine
`The bioavailability and safety of oral azacitidine was initially
`studied in an open-label, pharmacokinetic (PK), and feasibility
`pilot study of patients with MDS, leukemia, or solid tumors
`[56]. Four patients received 60- or 80-mg single doses of oral
`azacitidine. All four patients had measurable plasma concen-
`trations, allowing for comparison with historical s.c. azacitidine
`PK data. The 80-mg oral azacitidine dose had mean bio-
`availability of 17% of that of s.c. azacitidine [56, 65]. No severe
`drug-related toxicities were observed, and results from this
`pilot study led to the development of a phase I study of oral
`azacitidine.
`
`Dose-Finding Study of Oral Azacitidine
`Because of the demonstrated safety and efficacy of inject-
`able azacitidine in patients with MDS and AML [32, 38], the
`oral azacitidine phase I program initially focused on these
`patient populations. AZA PH US 2007 CL 005 was a phase I
`open-label dose-escalation study that evaluated the safety,
`PK, and pharmacodynamics (PD) of oral azacitidine in
`patients with MDS, CMML, or AML [66]. This trial had two
`parts (Fig. 2) [57, 66]: In part 1, 41 patients (71% MDS, 10%
`CMML, 20% AML) received 7-day dosing of s.c. azacitidine
`for a single 28-day cycle, followed by oral azacitidine for
`7 days of 28-day cycles (cycles 2 and beyond; Table 1). Dur-
`ing cycles 1 and 2, PK (days 1 and 7) and PD (days 1, 3, 8, 15,
`22, and 28) profiles were assessed.
`The maximum tolerated dose (MTD) of oral azacitidine
`was 480 mg once daily (QD) for 7 days [66]. Dose-limiting
`toxicities (DLTs) were reported in 2 of 3 patients treated
`with oral azacitidine 600 mg QD for 7 days (1 grade 3
`diarrhea and 1 grade 4 diarrhea despite medical interven-
`tion) with no other DLTs observed. Gastrointestinal AEs
`were the most common nonhematologic AEs and were
`primarily grade 1/2 and manageable [66] with gastric
`acid-reducing agents, antiemetics, and antidiarrheal
`
`TheOncologist®
`
`

`

`Cogle, Scott, Boyd et al.
`
`1407
`
`Figure 2. Trial schema for AZA PH US 2007 CL 005, the phase I trial of oral azacitidine (CC-486) in patients with myelodysplastic syndromes,
`acute myeloid leukemia, and chronic myelomonocytic leukemia [67].
`Abbreviations: BID, twice daily; QD, once daily; s.c., subcutaneous.
`
`Table 1. Summary of clinical results from the expanded phase I study of oral azacitidine (CC-486) in patients with MDS and AML [66,
`69, 71]
`
`Variable
`
`Dosing
`
`Patient
`population,
`n (%)
`
`Key safety
`data
`
`Key efficacy
`data
`
`Dose-finding study in MDS,
`CMML, and AML [66]
`Cycle 1: s.c. AZA 75 mg/m2
`QD 3 7 days
`Cycles $2: CC-486 120–600 mg
`QD 3 7 days
`MDS: 29 (71)
`CMML: 4 (10)
`AML: 8 (20)
`
`MTD: 480 mg QD 3 7 days
`DLTs: grade 3 or 4 diarrhea
`Most common grade 3/4 AEs:
`febrile neutropenia (20%),
`diarrhea (12%)
`First-line patients: 73% ORR,
`56% any HI, 33% mCR
`Previously-treated patients:
`35% ORR, 38% any HI, 67% mCR
`
`Extended-dosing in patients
`with lower risk MDS [69]
`CC-486 300 mg QD 3 14 or 21
`days
`
`IPSS low-risk MDS: 15 (28)
`IPSS Int-1 risk MDS: 38 (72)
`RBC-TD: 30 (57)
`Platelet-TD: 4 (8)
`Most common AEs (any grade)
`were gastrointestinal in origin
`Most common grade 3/4 AEs:
`neutropenia (13%)
`8 patients D/C due to AEs
`300 mg QD 3 14 days: 42%
`ORR, 27% any HI, 20% RBC-TI
`sustained 84 days
`300 mg QD 3 21 days: 37%
`ORR, 30% any HI, 33% RBC-TI
`sustained 84 days
`
`CC-486 300 mg QD or 200 mg
`BID 3 14 or 21 days
`
`Subset analysis: 7-day and extended dosing in patients
`with AML [71]
`Cycle 1: s.c. AZA 75 mg/m2
`QD 3 7 days
`Cycles $2: CC-486
`120–600 mg QD 3 7 days
`De novo AML: 13 (57)
`Secondary AML: 10 (43)
`RBC-TD: 14 (61)
`Platelet-TD: 8 (35)
`Most common AEs (any grade) were gastrointestinal in
`origin
`Most common grade 3/4 AEs: febrile neutropenia (35%),
`pneumonia (17%), syncope (17%), nausea (13%)
`6 patients D/C due to AEs
`38% ORR, 13% HI, 25%
`mCR, 13% mPR, 25%
`RBC-TIa
`
`47% ORR, 27% HI, 40% RBC-
`TI, 17% platelet-TI, 33%
`mPRb
`
`aEight evaluable patients for each, except four evaluable patients for RBC-TI.
`bFifteen evaluable patients for ORR and HI, 10 evaluable patients for RBC-TI, 6 evaluable patients for platelet-TI, and 9 evaluable patients for mPR.
`Abbreviations: AE, adverse event; AML, acute myeloid leukemia; AZA, azacitidine; BID, twice daily; CMML, chronic myelomonocytic leukemia; D/C,
`discontinued; DLT, dose-limiting toxicity; HI, hematologic improvement; Int-1, intermediate-1; IPSS, International Prognostic Scoring System; mCR,
`marrow complete response; mPR, marrow partial response; MDS, myelodysplastic syndromes; MTD, maximum tolerated dose; ORR, overall response
`rate; QD, once daily; RBC, red blood cell; s.c., subcutaneous; TD, transfusion dependence; TI, transfusion independence.
`
`medications [67]. The most common ($10%) grade 3/4 AEs
`were febrile neutropenia (20%), diarrhea (12%), and fatigue
`(10%). Notably, 4 of 8 patients with grade 3/4 febrile
`neutropenia had an absolute neutrophil count #500/mL at
`baseline [66]. Grade 3/4 nausea and vomiting were each
`reported in 7% of patients. Three patients discontinued due
`to an AE.The investigators of part 1 of the trial concluded that
`7-day oral azacitidine dosing was clinically active (Tables 1,
`2). Responses in patients treated with oral azacitidine as
`first-line therapy included a 73% ORR, 56% with any HI, and
`33% with mCR. Notably, responses were also achieved in
`previously treated patients: 35% ORR, 38% any HI, and 67%
`mCR [66].
`
`Extended Dosing of Oral Azacitidine in Patients With
`Lower Risk MDS
`Approximately two-thirds of newly diagnosed patients with
`MDS present with lower risk disease (IPSS low or Int-1 risk) [19].
`Lower risk patients are generally viewed as having favorable
`prognoses, with a median OS of 5.7 years for low-risk and 3.5
`years for Int-1 risk disease [19]. However, a subgroup within the
`lower risk MDS population actually has a worse prognosis, with
`shortened survival time and clonal evolution to AML [68].
`Approximately one-third of patients with lower risk MDS were
`shown to have poor prognostic features, with a median OS of
`only 1.2 years [68], the same as that reported for patients with
`Int-2 risk disease [19]. Severe anemia and thrombocytopenia
`
`www.TheOncologist.com
`
`©AlphaMed Press 2015
`
`

`

`1408
`
`Oral Azacitidine (CC-486) for MDS and AML
`
`Table 2. Hematologic responses in patients with MDS, CMML,
`or AML treated with 1 cycle of s.c. azacitidine followed by oral
`azacitidine (CC-486) in 7-day dosing schedules [66]
`
`Table 3. Hematologic responses in patients with IPSS
`lower-risk MDS treated with oral azacitidine (CC-486) in
`extended dosing regimens [69]
`
`First-line-treated
`patients
`
`Previously
`treated
`patientsa
`
`Total
`
`Response
`
`300 mg QD 3
`14 days
`
`300 mg QD 3
`21 days
`
`Total
`
`EP, n R, n (%)
`
`EP, n R, n (%)
`
`EP, n R, n (%)
`
`Response
`
`Overall
`responseb
`CRc
`Any HI
`HI-E
`HI-P
`HI-N
`mCRd
`TI
`
`RBC
`Platelet
`
`EP, n
`
`R, n (%)
`
`EP, n R, n (%) EP, n R, n (%)
`
`15
`
`15
`9
`4
`6
`7
`6
`3
`3
`0
`
`11 (73)
`
`6 (40)
`5 (56)
`2 (50)
`2 (33)
`2 (29)
`2 (33)
`1 (33)
`1 (33)
`0
`
`17
`
`17
`16
`10
`14
`10
`9
`5
`3
`4
`
`6 (35)
`
`0
`6 (38)
`3 (30)
`5 (36)
`0
`6 (67)
`0
`0
`0
`
`32
`
`32
`25
`14
`20
`17
`15
`8
`6
`4
`
`17 (53)
`
`6 (19)
`11 (44)
`5 (36)
`7 (35)
`2 (12)
`8 (53)e
`1 (13)
`1 (17)
`0
`
`aIncludes erythropoiesis-stimulating agents, chemotherapy,
`hypomethylating agents, and investigational and/or other agents.
`bDoes not include patients with mCR only.
`cPatients achieving CR were not included in other categories.
`dTwo patients with mCR in the first-line group also had HI (HI-P [n 5 1]
`and HI-E and HI-N [n 5 1]) and 1 patient with mCR in the previously
`treated group also had HI (both HI-E and HI-P); these patients were
`included in the mCR and HI categories.
`eIn the 8 patients with mCR, the response began in cycle 1 of s.c. dosing
`(n 5 4) or very early in cycle 2 of CC-486 dosing (n 5 4); therefore, a single
`cycle of azacitidine s.c. likely contributed to the response.
`Abbreviations: AML, acute myeloid leukemia; CMML, chronic
`myelomonocytic leukemia; CR, complete response; E, erythroid; EP,
`evaluable patients; HI, hematologic improvement; mCR, marrow
`complete response; MDS, myelodysplastic syndromes; N, neutrophil; P,
`platelet; R, responders; RBC, red blood cell; s.c., subcutaneous; TI,
`transfusion independence.
`
`are common poor prognostic features in patients with lower
`risk MDS [19, 68].
`Although part 2 of AZA PH US 2007 CL 005 was initially
`designed to investigate extended dosing schedules of oral
`azacitidine (300 mg QD or 200 mg twice daily [BID] for 14 or
`21 days per 28-day cycle) in patients with MDS, CMML,
`or AML (Fig. 2) [57], because of the new prognostic in-
`formation about lower risk MDS, the trial was amended to
`focus on patients with lower risk MDS with poor prognostic
`features (low platelet count and/or low hemoglobin and/or
`RBC transfusion dependence [TD] and/or platelet TD). Part
`2 included 53 patients with IPSS low-risk (28%) or Int-1 risk
`(72%) MDS; most patients were RBC-TD [69] (Table 1).
`Extended dosing with oral azacitidine (300 mg QD for 14 or
`21 days of repeated 28-day cycles) resulted in response (IWG
`2006 criteria [22]) in more than one-third of patients with
`lower risk MDS (Tables 1, 3), and the rate of RBC-TI increased
`from baseline with increasing cycles of treatment. The most
`common nonhematologic AEs were gastrointestinal, and
`there were no unexpected AEs based on the known safety
`profile of injectable azacitidine [32, 38, 69]. Eight patients
`(four in each arm) discontinued due to an AE. The most
`common grade 3/4 AEs ($10%) were neutropenia (8% and
`19%, respectively), pneumonia (15% and 4%), anemia (12%
`
`©AlphaMed Press 2015
`
`26
`
`26
`23
`17
`10
`
`15
`
`15
`
`11 (42)
`
`27
`
`10 (37)
`
`53
`
`21 (40)
`
`7 (27)
`4 (17)
`4 (24)
`3 (30)
`
`27
`25
`15
`6
`
`8 (30)
`6 (24)
`3 (20)
`0
`
`53
`48
`32
`16
`
`15 (28)
`10 (21)
`7 (22)
`3 (19)
`
`8 (54)
`
`15
`
`6 (40)
`
`30
`
`14 (47)
`
`3 (20)
`
`15
`
`5 (33)
`
`30
`
`8 (27)
`
`Overall
`responsea
`Any HI
`HI-E
`HI-P
`HI-N
`RBC-TI
`Sustained
`56 days
`Sustained
`84 days
`aComplete response, partial response, any HI, and TI by International
`Working Group 2006 criteria.
`Abbreviations: E, erythroid; EP, evaluable patients; HI, hematologic
`improvement; IPSS, International Prognostic Scoring System; MDS,
`myelodysplastic syndromes; N, neutrophil; P, platelet; QD, once daily; R,
`responders; RBC, red blood cell; TI, transfusion independence.
`
`and 7%), thrombocytopenia (12% and 4%), diarrhea (8% and
`11%), and febrile neutropenia (4% and 11%) [69].
`
`Preliminary data in patients with MDS, CMML, and
`AML suggest that oral azacitidine in extended dosing
`regimens may be associated with significant DNA
`hypomethylation through cycle end; however, this
`correlation must be confirmed in a larger patient
`population.
`
`Oral Azacitidine in Patients With AML
`IC is not appropriate for all patients with AML, and eligibility is
`influenced by age, performance status, comorbidities, and
`preexisting MDS [27–29, 70]. There is an unmet need for
`effective treatment options for patients who are ineligible or
`unwilling to receive IC [29]. Data from patients with AML in
`parts 1 and 2 of AZA PH US 2007 CL 005 were pooled (n 5 23;
`13 patients with de novo disease and 10 secondary to MDS)
`to assess response (IWG 2003 [45] and 2006 criteria [22]) to
`oral azacitidine [71] (Table 1). At baseline, 52% and 35% of
`patients had intermediate and unfavorable cytogenetics,
`respectively; 61% and 35% of patients were RBC-TD and
`platelet-TD, respectively; and 57% of patients were relapsed
`or refractory to prior treatment [71]. Of 8 patients treated
`with oral azacitidine at 120–600 mg QD for 7 days, 3 achieved
`a response (38%), including 1 with HI, 1 with RBC-TI, 2 with
`mCR, and 1 with marrow PR. Of 15 patients treated with oral
`azacitidine in extended dosing schedules (300 mg QD or 200
`mg BID for 14 or 21 days), 7 achieved a response (47%),
`including 4 with HI, 4 with RBC-TI, 1 with platelet-TI, and 3
`
`TheOncologist®
`
`

`

`Cogle, Scott, Boyd et al.
`
`1409
`
`with marrow PR [71]. No patients with AML achieved CR or
`PR. The most common AEs were gastrointestinal, and 6
`patients discontinued due to an AE.The most common grade
`3/4 AEs (7-day and extended dosing regimens combined,
`$10%) were febrile neutropenia (35%), pneumonia (17%),
`syncope (17%), and nausea (13%) [71].
`
`PK and PD of Oral Azacitidine
`For patients with MDS, CMML, or AML in the AZA PH US 2007
`CL 005 study, the maximum concentration (Cmax) of oral
`azacitidine occurred at 1.0 hour (range: 0.3–3.6 hours) after
`dosing, with a mean half-life of 0.6 hour [66]. In patients with
`lower risk MDS, oral azacitidine 300 mg QD for 14 or 21 days
`had a mean exposure per cycle of 38% and 56%, respectively, of
`that of s.c. azacitidine 75 mg/m2 QD for 7 days (the approved
`dosing regimen) [72]. Exposure generally increased with dose,
`and no drug accumulation was observed following multiple
`administrations.
`In patients evaluated for DNA methylation, 7-day dosing
`with s.c. azacitidine or oral azacitidine resulted in maximal
`decreases in highly methylated DNA loci at day 15 that
`returned to near-baseline levels by cycle end [66, 72].
`Preliminary data in patients with MDS, CMML, and AML
`suggest that oral azacitidine in extended dosing regimens
`may be associated with significant DNA hypomethylation
`through cycle end [72]; however, this correlation must be
`confirmed in a larger patient population. The use of hypo-
`methylation to predict response to hypomethylating agents
`remains controversial [73]. Other potential biomarkers for
`azacitidine response actively being investigated include
`mutations in TET2 and IDH1/2, which are involved in DNA
`methylation [74–76].
`Two phase I studies in patients with MDS, CMML, or AML
`evaluated the PK of immediate-release, enteric-coated, and
`capsule formulations of oral azacitidine and the effect of
`food and gastric pH alterations with concomitant admin-
`istration of proton-pump inhibitors (PPIs) on PK parameters
`(n 5 47) [67]. An immediate-release formulation was
`chosen for upcoming studies because it had the greatest
`mean oral bioavailability relative to s.c. azacitidine. PK
`parameters were similar under fasting and postprandial
`conditions; Cmax was delayed by ∼0.5 hour, and time to
`reach Cmax was significantly delayed from 1.0 hour to 2.0
`hours (p , .05) under postprandial conditions. Coadmin-
`istration of the PPI omeprazole did not significantly affect
`PK parameters [67]. The lack of food effect or need for dose
`adjustments with PPIs allows for convenient administration
`of oral azacitidine.
`
`KEY ONGOING OR PLANNED TRIALS OF ORAL AZACITIDINE
`(CC-486) IN MDS AND AML
`Promising results of the expanded phase I program for oral
`azacitidine in MDS and AML led to the initiation of several
`additional studies, including two ongoing phase III trials.
`
`QUAZAR Lower Risk MDS (AZA-MDS-003) Study
`No currently approved therapies have demonstrated signif-
`icantly prolonged survival in patients with lower risk MDS
`with thrombocytopenia and TD anemia. An alternative-
`dosing study of injectable azacitidine in patients with MDS
`
`(most with lower-risk disease) suggested that prolonged
`exposure to a lower dose of azacitidine may benefit patients
`with multiple cytopenias [40]. The phase III QUAZAR Lower
`Risk MDS (AZA-MDS-003) study (ClinicalTrials.gov identifier
`NCT01566695) has a planned enrollment of 386 patients
`with RBC-TD anemia (average of $2 RBC units per 28 days,
`confirmed for $84 days immediately preceding randomiza-
`tion) and thrombocytopenia (2 platelet counts #75 3 109/L
`$21 days apart) due to IPSS lower risk MDS [77]. This study
`will assess the efficacy of oral azacitidine (300 mg QD for 21
`days of 28-day cycles) plus BSC versus placebo plus BSC in
`these patients. Enrolled patients must have MDS (WHO 2008
`classification; excludes secondary MDS unless patients
`received their last dose of prior antineoplastic therapy $24
`weeks prior to randomization), have Eastern Cooperative
`Oncology Group (ECOG) performance status 0–2, have no prior
`treatment with hypomethylating agents, have no prior
`treatment with lenalidomide within 24 weeks prior to
`randomization, and be SCT ineligible with no prior transplant.
`The primary outcome of interest is RBC-TI, with key secondary
`outcomes of survival, progression to AML, platelet-TI, he-
`matologic response, platelet HI, erythroid HI, safety, health-
`related QOL (HRQOL), and health care resource utilization
`[77].
`
`An alternative-dosing study of injectable azacitidine in
`patients with MDS (most with lower risk disease)
`suggested that prolonged exposure to a lower dose
`of azacitidine may benefit patients with multiple
`cytopenias.
`
`QUAZAR AML Maintenance (CC-486-AML-001) Study
`Most patients with AML currently will relapse following
`achievement of remission with IC, and it has been shown that
`duration of first remission is predictive of response to salvage
`therapies and long-term outcomes [78]. To improve out-
`comes, there is a need to prolong remissions, particularly
`in patients unable or unwilling to receive SCT. The phase
`III

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