Safety and Efficacy Study of CC-486 in Subjects With Myelodysplastic Syndromes

Sponsor
Celgene (Industry)
Overall Status
Active, not recruiting
CT.gov ID
NCT02281084
Collaborator
(none)
65
66
2
89.5
1
0

Study Details

Study Description

Brief Summary

Evaluate the safety and efficacy of oral azacitidne (CC-486) twice daily (BID) in subjects with myelodysplastic syndromes who failed to achieve an objective response post injectable hypomethylating agent (iHMA) treatment

Reason for removing the combination arm: Due to difficulties with dose-finding, the durvalumab plus CC-486 combination arm was closed to enrollment.

Extension:

An Extension Phase (EP) has been added to allow subjects who are currently receiving oral azacitidine BID and who are demonstrating clinical benefit as assessed by the Investigator, to continue receiving oral azacitidine until the subject meets the criteria for study discontinuation.

Condition or Disease Intervention/Treatment Phase
Phase 2

Study Design

Study Type:
Interventional
Actual Enrollment :
65 participants
Allocation:
Randomized
Intervention Model:
Parallel Assignment
Masking:
None (Open Label)
Primary Purpose:
Treatment
Official Title:
A Phase 2, International, Multicenter, Randomized, Open-label, Parallel Group to Evaluate the Efficacy and Safety of Cc-486 (Oral Azacitidine) Alone in Combination With Durvalumab (MEDI4736) in Subjects With Myelodysplastic Syndromes Who Fail to Achieve an Objective Response to Treatment With Azacitidine for Injection or Decitabine
Actual Study Start Date :
Jul 6, 2015
Actual Primary Completion Date :
Jun 19, 2019
Anticipated Study Completion Date :
Dec 19, 2022

Arms and Interventions

Arm Intervention/Treatment
Experimental: Monotherapy: Oral Azacitidine

Oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.

Drug: Oral Azacitidine
Oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
Other Names:
  • CC-486
  • Experimental: Combination Therapy: Oral Azacitidine and Durvalumab

    Oral Azacitidine 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous (IV) infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.

    Drug: Oral Azacitidine
    Oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Other Names:
  • CC-486
  • Drug: Durvalumab
    Durvalumab 1500 mg by IV infusion on Day 1 of each 28 day treatment cycle.
    Other Names:
  • Imfinzi
  • MEDI4736
  • Outcome Measures

    Primary Outcome Measures

    1. Overall Response Rate Based on the Modified International Working Group (IWG) 2006 Response Criteria for Myelodysplastic Syndrome (MDS) [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      The overall response rate (ORR) was defined as the percentage of participants who achieved an objective response including: hematologic improvement (HI), partial remission (PR), complete remission (CR), or marrow complete remission (mCR). Hematologic response was defined as: CR: ≤ 5% myeloblasts with normal maturation of all cell lines; peripheral blood (PB) shows: hemoglobin ≥11 g/dL, neutrophils ≥1.0x10^9/L, platelets ≥100x10^9/dL, blasts (0%) PR: same as CR bone marrow (BM) shows blasts decreased by ≥ 50% over pre-treatment but still > 5%; cellularity and morphology not relevant mCR: BM: ≤ 5% myeloblasts and decrease by ≥ 50% over pre-treatment PB, PB: if HI responses, noted in addition to mCR HI: HI erythroid response (HI-E); HI neutrophil response (HI-N) ; HI platelet response (HI-P)

    Secondary Outcome Measures

    1. Kaplan-Meier Estimate of Overall Survival [From day 1 of study drug to the data cut off date of 19 June 2019; median follow-up for OS = 15.96 and 8.35 months respectively in the SD and PD oral AZA alone arms and 13.48 and 13.33 months in the SD and PD combination arms]

      Overall survival (OS) was defined as the time from randomization to death from any cause, and was calculated using date of first dose and date of death, or date of last follow-up for censored subjects. All participants were followed until drop out (withdrawal of consent from further data collection or lost to follow-up), death, or study termination. Participants who dropped out or were alive at study termination (or at the time of the interim analysis) had their OS times censored at the time of last contact, as appropriate.

    2. Kaplan Meier Estimate of Time to Onset of First and Best Response [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      Time to onset of first response was defined as the time between the date of first investigational product (IP) dose and the earliest date any response (CR, PR, mCR, or HI) was first observed. Participants who did not achieve any defined response during the treatment period were censored at the date of treatment discontinuation, disease progression, or death, whichever occurred first. Best response is the best recorded response or treatment outcome from the start of the study treatment until the end of the study treatment taking into account the requirements for confirmation of response.

    3. Kaplan Meier Estimate of Duration of First Response [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      Duration of hematologic response and/or improvement was defined as the time from the date response or improvement was first observed to the date of documented relapse or disease progression as defined by the modified IWG 2006 criteria. Particpants who maintained hematologic response and/or improvement through the end of the treatment period were censored as the date of treatment discontinuation or death, whichever occurred first.

    4. Kaplan Meier Estimate of Duration of Best Response [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      Duration of hematologic response and/or improvement was defined as the time from the date response or improvement was first observed to the date of documented relapse or disease progression as defined by the modified IWG 2006 criteria. Particpants who maintained hematologic response and/or improvement through the end of the treatment period were censored as the date of treatment discontinuation or death, whichever occurred first.

    5. Kaplan-Meier Estimate of Progression Free Survival (PFS) [From day 1 of study drug to the data cut off date of 19 June 2019; median follow-up for OS = 15.96 and 8.35 months respectively in the SD/ PD oral AZA arms and 13.48 and 13.33 months in the SD/PD oral AZA and Durva arms]

      Progression-free survival is defined as the time from randomization to the first documented progressive disease (PD), relapse, or death due to any cause during or after the treatment period, whichever occurred first, according to IWG 2006 response criteria for MDS. Participants who were still alive and progression-free were censored at the date of their last response assessment. Progressive disease is defined as follows: - an increase in BM blasts relative to nadir: •If nadir less than 5% blasts: ≥ 50% increase in blasts to > 5% blasts •If nadir 5% - 10% blasts: ≥ 50% increase in blasts to > 10% blasts •If nadir 10% - 20% blasts: ≥ 50% increase in blasts to > 20% blasts •If nadir 20% - 30% blasts: ≥ 50% increase in blasts to > 30% blasts And any of the following: •At least 50% decrement from maximum remission/response levels in granulocytes or platelets •Reduction in Hgb concentration by ≥ 2 g/dL •Transfusion dependence

    6. Percentage of Participants With Progressive Disease at Baseline Who Achieved Stable Disease [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      A participant was considered as having a stable disease if the disease neither responded nor progressed during or after study treatment.

    7. Kaplan-Meier Estimate of Onset to Achieve Stable Disease [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      A participant was considered as having a stable disease if the disease neither responded nor progressed during or after study treatment.

    8. Kaplan-Meier Estimate of Duration of Stable Disease [Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms]

      The duration of stable disease was defined as the time between any two observations of objective disease progression (modified IWG criteria), starting from the first day of dosing with IP. Participants who maintained stable disease through the end of the treatment period were censored at the date of study termination.

    9. Percentage of Participants Who Progressed to Acute Myelogenous Leukemia (AML) [From Day 1 of study drug up to the data cut off date of 19 June 2019; median follow up time for AML progression = 11.59 and 5.65 months respectively in the SD and PD arms for oral AZA and 6.21 months for SD/PD in the combination arm.]

      For all participants who received at least one dose of study drug, continuous monitoring for progression to AML occurred in the post treatment follow up period.

    10. Kaplan-Meier Estimate of Time to Progression to AML [From Day 1 of study drug up to the data cut off date of 19 June 2019; median follow up time for AML progression = 11.59 and 5.65 months respectively in the SD and PD arms for oral AZA and 6.21 months for SD/PD in the combination arm.]

      Time to AML progression was defined as the time from the date of first dose of IP until the date the participant had documented progression to AML.

    11. Number of Participants With Treatment Emergent Adverse Events (TEAEs) [From first dose of IP until 28 days after final oral AZA dose, 90 days after final durva dose and/or treatment stopped; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively; 1.84 months for AZA and Durva SD/PD arms]

      TEAEs were defined as AEs occurring or worsening on or after the date of the first dose of oral aza or durva and within 28 days after last dose of oral aza or 90 days after last dose of durva A serious adverse event (SAE) is any: Death; Life-threatening event; Any inpatient hospitalization or prolongation of existing hospitalization; Persistent or significant disability or incapacity; Congenital anomaly or birth defect; Any other important medical event The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 4.0) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    18 Years and Older
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No
    Inclusion Criteria:
    1. Male or female, ≥ 18 years of age at the time of signing the informed consent document

    2. Documented diagnosis of MYELODYSPLASTIC SYNDROMES (MDS), classified according to FRENCH-AMERICAN BRITISH (FAB) classification criteria

    3. Adequate course of treatment with an injectable hypomethylating agent (azacitidine for injection or decitabine) as the last therapeutic intervention for MDS prior to beginning screening for this study. Adequate is defined as:

    • having received at least 6 consecutive 4-week treatment cycles with azacitidine for injection, or

    • having received at least 4 consecutive 6-week treatment cycles with decitabine (3-day regimen) or at least 6 consecutive 4-week treatment cycles with decitabine (5-day regimen), or

    • having demonstrated inability to tolerate treatment with an injectable hypomethylating agent because of unacceptable drug-related toxicity after at least 3 months of attempted treatment: Three 28-day cycles of azacitidine for injection or decitabine 5-day regimen; two 42-day cycles of decitabine 3-day regimen.

    1. Documented disease progression or stable disease as best response to treatment (or attempted treatment) with azacitidine for injection or decitabine. Those achieving an objective response to treatment regimen with an injectable hypomethylating agent (HMA) are excluded from participation in this study.

    Definitions of disease progression are modified from INTERNATIONAL WORKING GROUP (IWG) 2006 criteria and include:

    • Pre-injectable hypomethylating agent baseline bone marrow myeloblasts:
    1. Less than 5%: ≥ 100% increase to ≥ 8% blasts

    2. ≥ 5%: ≥ 50% increase to ≥ 10% blasts Note: ≥ 30% blasts is considered acute myeloid leukemia (AML )per FAB classification. Subjects known to have ≥ 30% blasts are not eligible for inclusion in this study.recognizing eastern cooperative oncology group) limitations of blast cell quantification, Protocol will allow subjects with pre-enrollment bone marrow blast counts up to 33% on the screening bone marrow examination to be considered for inclusion. Assessment may be made according to local bone marrow examination to facilitate enrollment of eligible subjects into the treatment phase of the study.

    • Any clinical worsening from pre-injectable hypomethylating agents (HMA) baseline condition, including:
    1. sustained clinically-significant worsening (investigator's assessment) from baseline granulocyte, platelet, or hemoglobin values (≥ 2 values, separated by ≥ 2 weeks) - worsening granulocytes should be ≥ 50% decrease from pre-injectable HMA baseline value - worsening platelets should be ≥ 50% decrease from pre-injectable HMA baseline value (untransfused)
    • worsening hemoglobin should be ≥ 1.5 g/dL decrease from preinjectable HMA baseline value in subjects not receiving RBC transfusions
    1. meaningful worsening in RBC or platelet transfusion requirement
    Definition of stable disease is based on modified IWG 2006 criteria:
    • Failure to achieve any objective response (CR - complete remission, PR - partial remissino, mCR - marrow complete remission, or HI - hematologic improvement), but no evidence of disease progression within the 8 weeks leading to the subject's first dose of investigational product (IP), Cycle 1, Day 1
    1. Have the last dose of the prior treatment regimen injectable HMA - (azacitidine for injection or decitabine) not more than 16 weeks prior to screening for this study (date of informed consent signature).

    2. No less than 3 weeks between the last dose of the prior treatment regimen injectable HMA - (azacitidine for injection or decitabine) and the planned date of first dose of IP (

    3. Have an eastern cooperative oncology group (ECOG) performance status of 0, 1, or 2

    4. Females subjects of childbearing potential (FCBP)1 may participate, providing they meet the following conditions:

    5. Have two negative pregnancy tests as verified by the investigator prior to starting any IP therapy: serum pregnancy test at screening and negative serum or urine pregnancy test (investigator's discretion) within 72 hours prior to starting treatment with IP (Cycle 1, Day 1). They must agree to ongoing pregnancy testing during the course of the study (before beginning each subsequent cycle of treatment), and after the last dose of any IP. This applies even if the subject practices complete abstinence2 from heterosexual contact.

    6. Agree to practice true abstinence2 (which must be reviewed on a monthly basis and source documented) or agree to the use of highly effective methods of contraception from 28 days prior to starting azacitidine, and must agree to continue using such precautions while taking azacitidine (including dose interruptions) and for up to 90 days after the last dose of azacitidine. Cessation of contraception after this point should be discussed with a responsible physician

    7. Agree to abstain from breastfeeding during study participation and for at least 90 days after the last dose of IP.

    Note that the screening serum pregnancy test can also be used as the test prior to starting IP if it is performed within the 72-hour timeframe.

    1. Male subjects must:

    2. Male subjects must:

    3. Either practice true abstinence2 from heterosexual contact (which must be reviewed on a monthly basis) or agree to avoid fathering a child, to use highly effective methods of contraception, male condom plus spermicide during sexual contact with a pregnant female or a female of child bearing potential (even if he has undergone a successful vasectomy) from starting dose of IP (Cycle 1 Day 1), including dose interruptions through 90 days after receipt of the last dose of azacitidine.

    4. Refrain from semen or sperm donation while taking IP and for at least 90 days after the last dose of IP.

    5. Understand and voluntarily sign an informed consent document prior to any study-related assessments or procedures conducted.

    6. Be able to adhere to the study visit schedule and other protocol requirements.

    7. Understand and voluntarily sign a biomarker-specific component of the informed consent document prior to any study-related procedures conducted.

    Extension Phase

    At the Investigator's discretion and following confirmation of eligibility criteria below, subjects can enter the Extension Phase (EP):

    • Subjects who have signed the informed consent for the EP of the study;

    • Subjects receiving oral azacitidine and continuing in the treatment phase demonstrating clinical benefit as assessed by the Investigator are eligible to receive oral azacitidine in the EP;

    • Subjects who do not meet any of the criteria for study discontinuation

    Exclusion Criteria:
    1. Rapidly-progressing MDS defined as:

    2. Known clinically-significant doubling in marrow or per IP peripheral blood blast percentage (to ≥ 20%) in the 8-week period leading to the first dose of IP (Cycle 1, Day 1)

    3. ≥100% increase in WBC count (myeloid cell line and monocyte series) within the 8-week period leading to Cycle 1, Day 1

    4. AML - FAB (FRENCH-AMERICAN-BRITISH) classification: ≥ 30% blasts in bone marrow). Subjects known to have ≥ 30% blasts are not eligible for inclusion in this study. Recognizing limitations of blast cell quantification, this protocol will allow subjects with pre-enrollment (screening/baseline) bone marrow blast counts up to 33% to be considered for inclusion.

    5. Prior allogeneic stem cell transplant

    6. Prior exposure to the investigational oral formulation of decitabine, or other oral azacitidine derivative at any time in the subject's prior history

    7. Prior or ongoing response (IWG 2006: HI, PR, CR, or marrow CR) to treatment with azacitidine for injection or decitabine, at any time in the subject's prior history, which includes relapsed disease

    8. Ongoing medically significant adverse events from previous treatment, regardless of the time period

    9. Use of any of the following within 28 days prior to the first dose of IP:

    10. thrombopoiesis-stimulating agents ([TSAs]; eg, Romiplostim, Eltrombopag, Interleukin-11)

    11. ESAs (Erythropoiesis stimulating agent) and other RBC hematopoietic growth factors (eg, interleukin-3)

    12. hydroxyurea

    13. Concurrent use of corticosteroids unless the subject is on a stable or decreasing dose for ≥ 1 week prior to enrollment for medical conditions other than MDS

    14. History of inflammatory bowel disease (eg, Crohn's disease, ulcerative colitis), celiac disease (ie, sprue), prior gastrectomy or upper bowel removal, or any other gastrointestinal disorder or defect that would interfere with the absorption, distribution, metabolism or excretion of the IP and/or predispose the subject to an increased risk of gastrointestinal toxicity

    15. Prior history of malignancies, other than MDS, unless the subject has been free of the disease for ≥ 3 years. However, subjects with the following history/concurrent conditions are allowed:

    16. Basal or squamous cell carcinoma of the skin

    17. Carcinoma in situ of the cervix

    18. Carcinoma in situ of the breast

    19. Incidental histologic finding of prostate cancer (T1a or T1b using the tumor, nodes, metastasis [TNM] clinical staging system)

    20. Significant active cardiac disease within the previous 6 months, including:

    21. New York Heart Association (NYHA) class IV congestive heart failure;

    22. Unstable angina or angina requiring surgical or medical intervention; and/or

    23. Myocardial infarction

    24. Uncontrolled systemic fungal, bacterial, or viral infection (defined as ongoing signs/symptoms related to the infection without improvement despite appropriate antibiotics, antiviral therapy, and/or other treatment)

    25. Known Human Immunodeficiency Virus (HIV) or Hepatitis C (HCV) infection, or evidence of active Hepatitis B Virus (HBV) infection

    26. Any of the following laboratory abnormalities:

    27. Serum Aspartate transaminase / Serum glutamic oxaloacetic transaminase (AST/SGOT) Alanine aminotransaminase / Serum glutamic pyruvate transaminase (ALT/SGPT) > 2.5 x ULN (upper limit of normal)

    28. Serum total bilirubin > 1.5 x upper limit of normal (ULN). Higher levels are acceptable if these can be attributed to active red blood cell precursor destruction within the bone marrow (ie, ineffective erythropoiesis). Subjects are excluded if there is evidence of autoimmune hemolytic anemia manifested as a corrected reticulocyte count of > 2% with either a positive Coombs' test or over 50% of indirect bilirubin

    29. Serum creatinine > 2.5 x ULN (upper limit of normal)

    30. Absolute WBC (white blood cell) count ≥ 20 x 109/L

    31. Known or suspected hypersensitivity to azacitidine, mannitol, its constituents, or to any other humanized monoclonal antibody

    32. Pregnant, planning to become pregnant starting from 28 days prior to receiving CC-486 throughout your participation in the study, and for at least 90 days following your last dose of study treatment, or breast-feeding females

    33. Any significant medical condition, laboratory abnormality, or psychiatric illness that would prevent the subject from participating in the study

    34. Any condition including the presence of laboratory abnormalities, which places the subject at unacceptable risk if he/she were to participate in the study

    35. Any condition that confounds the ability to interpret data from the study, including known or suspected conditions other than MDS, associated with anemia

    36. Having received any prior MAb (monoclonal antibodies) against CTLA-4 (cytotoxic T lymphocyte-associated antigen), PD-1, or PD-L1 or having received other investigational monoclonalantibodies (MAbs) within 6 months

    37. Clinical evidence of central nervous system (CNS) or pulmonary leukostasis, or CNS leukemia

    38. Active or prior documented autoimmune or inflammatory disorders (including inflammatory bowel disease [eg, colitis, Crohn's disease], diverticulitis with the exception of a prior episode that has resolved or diverticulosis, celiac disease, irritable bowel disease, or other serious gastrointestinal chronic conditions associated with diarrhea; systemic lupus erythematosus; Wegener's syndrome [granulomatosis with polyangiitis]; myasthenia gravis; Graves' disease; rheumatoid arthritis; hypophysitis, uveitis; etc) within the past 3 years prior to the start of treatment. The following are exceptions to this criterion:

    39. Subjects with vitiligo or alopecia;

    40. Subjects with hypothyroidism (eg, following Hashimoto syndrome) stable on hormone replacement for ≥ 3 months; or

    41. Subjects with psoriasis not requiring systemic treatment

    42. History of primary immunodeficiency

    43. Active myeloproliferative neoplasms (MPN) and chronic myelomonocytic leukemia (CMML)

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 Local Institution - 113 New Haven Connecticut United States 06520
    2 Yale University New Haven Connecticut United States 06520
    3 University of Miami Miller School of Medicine Miami Florida United States 33136
    4 H Lee Moffitt Cancer Center and Research Institute Tampa Florida United States 33612
    5 University of Chicago Medicine Chicago Illinois United States 60637
    6 Ingalls Memorial Hospital Harvey Illinois United States 60426-3558
    7 University of Iowa Hospitals and Clinics Iowa City Iowa United States 52242
    8 James Graham Brown Cancer Center Louisville Kentucky United States 40202
    9 John Theurer Cancer Center at Hackensack University Medical Center Hackensack New Jersey United States 07601
    10 Icahn School of Medicine at Mount Sinai New York New York United States 10029
    11 University of Pennsylvania Philadelphia Pennsylvania United States 19104
    12 Hillman Cancer Institute at UPMC Pittsburgh Pennsylvania United States 15232
    13 University of Texas- MD Anderson Houston Texas United States 77030
    14 Cancer Care Centers of South Texas - HOAST San Antonio Texas United States 78229
    15 Medical College of Wisconsin Milwaukee Wisconsin United States 53226
    16 Westmead Hospital Westmead New South Wales Australia 2145
    17 Royal Adelaide Hospital Adelaide South Australia Australia 5000
    18 Monash Medical Centre Clayton Victoria Australia 3168
    19 Cabrini Hospital Malvern Victoria Australia 3144
    20 Local Institution - 405 Malvern Victoria Australia 3144
    21 Royal Brisbane and Women's Hospital Herston Australia 4029
    22 Royal Perth Hospital Perth Australia 6000
    23 Universitair Ziekenhuis Brussel Brussel Belgium 1090
    24 Hopital Erasme Bruxelles Belgium 1070
    25 Centre Hospitalier Universitaire de Liege Liège Belgium 4000
    26 Clinique Saint-Pierre Ottignies Belgium 1340
    27 Institut Paoli Calmettes Marseille Cedex 9 France 13273
    28 Hopital Saint-Louis Paris France 75010
    29 Local Institution - 201 Paris France 75010
    30 CHU Purpan Toulouse France 31059
    31 Local Institution - 500 Dresden Germany D-01307
    32 Universitatsklinikum Carl Gustav Carus an der TU Dresden Dresden Germany D-01307
    33 Local Institution - 502 Dusseldorf Germany 40479
    34 Marien Hospital Dusseldorf Germany 40479
    35 Universitatsklinikum Freiburg Freiburg Germany 79106
    36 Universitatsklinikum Halle Saale Halle Germany 06120
    37 Universitatsklinikum Leipzig Leipzig Germany 04103
    38 Medizinische Klinik III Klinikum der Universität München-Großhadern München Germany 81377
    39 Azienda Ospedaliera Santi Antonio Biagio E Cesare Arrigo Allessandria Italy 15100
    40 Azienda Ospedaliera Universitaria Policlinico Sant Orsola Malpighi Bologna Italy 40138
    41 Azienda Ospedaliero-Universitaria Careggi Firenze Italy 50134
    42 Ospedale San Raffaele S.r.l. Milano Italy 20132
    43 Azienda Ospedaliera Sant Andrea Roma Italy 00189
    44 Local Institution - 603 Roma Italy 00189
    45 Zaklad Opieki Zdrowotnej MSW z Warminsko-Mazurskim Centrum Onkologii Olsztyn Poland 10-228
    46 MTZ Clinical Research Sp. z o.o. Warszawa Poland 02-106
    47 Uniwersytecki Szpital Kliniczny Wroclaw Poland 50-367
    48 Institut Calatà d'Oncologia, L'Hospitalet Barcelona Spain 08907
    49 Local Institution - 306 Barcelona Spain 08907
    50 Hospital Universitari Vall d'Hebron Barcelona Spain 8035
    51 Hospital Virgenes de las Nieves Granada Spain 18014
    52 Hospital General Gregorio Maranon Madrid Spain 28009
    53 Hospital Universitario Virgen De La Victoria Malaga Spain 29010
    54 Hospital Central de Asturias Oviedo Spain 33006
    55 Hospital Universitario de Salamanca Salamanca Spain 37007
    56 Hospital Universitario Virgen Del Rocio Sevilla Spain 41013
    57 CEIC Hospital Universitario La Fe Valencia Spain 46009
    58 United Lincolnshire Hospitals NHS Trust Boston United Kingdom PE21 9QS
    59 Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital Cambridge United Kingdom CB2 0QQ
    60 Broomfield Hospital Chelmsford United Kingdom CM17ET
    61 Saint James University Hospital Leeds United Kingdom LS1 3EX
    62 King's College HospitalSchool of Medicine London United Kingdom SE5 9RS
    63 Hammersmith Hospital London United Kingdom W12 0HS
    64 University College London London United Kingdom WC1E 6BT
    65 Northwick Park Hospital Middlesex United Kingdom HA1 3UJ
    66 Southampton General Hospital Southampton United Kingdom SO16 6YD

    Sponsors and Collaborators

    • Celgene

    Investigators

    • Study Director: Bristol-Myers Squibb, Bristol-Myers Squibb

    Study Documents (Full-Text)

    More Information

    Publications

    None provided.
    Responsible Party:
    Celgene
    ClinicalTrials.gov Identifier:
    NCT02281084
    Other Study ID Numbers:
    • CC-486-MDS-006
    • 2014-002675-29
    First Posted:
    Nov 2, 2014
    Last Update Posted:
    Aug 23, 2022
    Last Verified:
    Aug 1, 2022

    Study Results

    Participant Flow

    Recruitment Details Participants were randomized at 33 sites globally. The sites were located in: Australia (3), Europe (18) and the United States (12). Results are reported as of the data cut-off date of 19 June 2019.
    Pre-assignment Detail Participants were eligible who did not respond to an adequate course of therapy with an injectable hypomethylating agent (iHMA - azacitidine or decitabine) or were unable to tolerate an iHMA following at least 3 months of attempted treatment.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Period Title: Overall Study
    STARTED 32 22 6 5
    COMPLETED 10 4 2 0
    NOT COMPLETED 22 18 4 5

    Baseline Characteristics

    Arm/Group Title Progressive Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Stable Disease (SD) Cohort: Oral Azacitidine Total
    Arm/Group Description Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Total of all reporting groups
    Overall Participants 32 22 6 5 65
    Age (Years) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [Years]
    73.9
    (7.62)
    75.1
    (7.56)
    70.0
    (7.21)
    72.4
    (5.55)
    73.9
    (7.42)
    Sex: Female, Male (Count of Participants)
    Female
    12
    37.5%
    7
    31.8%
    0
    0%
    0
    0%
    19
    29.2%
    Male
    20
    62.5%
    15
    68.2%
    6
    100%
    5
    100%
    46
    70.8%
    Race/Ethnicity, Customized (Count of Participants)
    White
    28
    87.5%
    18
    81.8%
    6
    100%
    5
    100%
    57
    87.7%
    Black
    0
    0%
    1
    4.5%
    0
    0%
    0
    0%
    1
    1.5%
    Asian
    1
    3.1%
    1
    4.5%
    0
    0%
    0
    0%
    2
    3.1%
    Native Hawaiian or Other Pacific Islander
    0
    0%
    1
    4.5%
    0
    0%
    0
    0%
    1
    1.5%
    American Indian or Alaska Native
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    Other
    1
    3.1%
    1
    4.5%
    0
    0%
    0
    0%
    2
    3.1%
    Not Collected or Reported
    2
    6.3%
    0
    0%
    0
    0%
    0
    0%
    2
    3.1%
    Race/Ethnicity, Customized (Count of Participants)
    Hispanic or Latino
    1
    3.1%
    1
    4.5%
    0
    0%
    2
    40%
    4
    6.2%
    Not Hispanic or Latino
    30
    93.8%
    21
    95.5%
    6
    100%
    3
    60%
    60
    92.3%
    Unknown
    1
    3.1%
    0
    0%
    0
    0%
    0
    0%
    1
    1.5%
    Myelodysplastic Syndrome (MDS) World Health Organization Classification 2008 (Count of Participants)
    Refractory Anemia (RA) with Ringed Sideroblasts
    1
    3.1%
    0
    0%
    0
    0%
    0
    0%
    1
    1.5%
    Refractory Cytopenia with Multilineage Dysplasia
    11
    34.4%
    2
    9.1%
    1
    16.7%
    0
    0%
    14
    21.5%
    RA With Excess Blasts-1 (RAEB-1)
    10
    31.3%
    6
    27.3%
    1
    16.7%
    2
    40%
    19
    29.2%
    RA With Excess Blasts-2 (RAEB-2)
    8
    25%
    11
    50%
    4
    66.7%
    3
    60%
    26
    40%
    MDS Unclassified (MDS-U)
    1
    3.1%
    3
    13.6%
    0
    0%
    0
    0%
    4
    6.2%
    MDS Associated with Isolated del (5q)
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    Missing
    1
    3.1%
    0
    0%
    0
    0%
    0
    0%
    1
    1.5%
    International Prognostic Scoring System Risk Classification (Count of Participants)
    Low (0)
    4
    12.5%
    2
    9.1%
    0
    0%
    0
    0%
    6
    9.2%
    Intermediate 1 (0.5-1.0)
    12
    37.5%
    5
    22.7%
    1
    16.7%
    2
    40%
    20
    30.8%
    Intermediate (2) (1.0-2.0)
    7
    21.9%
    7
    31.8%
    4
    66.7%
    2
    40%
    20
    30.8%
    High (2) (≥ 2.5)
    8
    25%
    8
    36.4%
    1
    16.7%
    1
    20%
    18
    27.7%
    Unknown
    1
    3.1%
    0
    0%
    0
    0%
    0
    0%
    1
    1.5%
    French-American-British (FAB) Classification (Count of Participants)
    Refractory Anemia (RA)
    6
    18.8%
    2
    9.1%
    0
    0%
    0
    0%
    8
    12.3%
    Refractory Anemia with Ringed Sideroblasts (RARS)
    6
    18.8%
    1
    4.5%
    1
    16.7%
    0
    0%
    8
    12.3%
    Refractory Anemia with Excess Blasts (RAEB)
    17
    53.1%
    16
    72.7%
    5
    83.3%
    4
    80%
    42
    64.6%
    RAEB in Transformation
    2
    6.3%
    3
    13.6%
    0
    0%
    1
    20%
    6
    9.2%
    Chronic Myelomonocytic Leukemia (CMML)
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    Missing
    1
    3.1%
    0
    0%
    0
    0%
    0
    0%
    1
    1.5%
    Time Since Initial Diagnosis of MDS (months) [Mean (Standard Deviation) ]
    Mean (Standard Deviation) [months]
    26.79
    (29.678)
    26.97
    (33.835)
    40.20
    (61.663)
    23.51
    (30.524)
    27.84
    (34.266)
    Eastern Cooperative Oncology Group (ECOG) Performance Status (PS) (Count of Participants)
    0 (Fully active)
    5
    15.6%
    6
    27.3%
    3
    50%
    2
    40%
    16
    24.6%
    1 (Restricted but Ambulatory)
    22
    68.8%
    13
    59.1%
    3
    50%
    3
    60%
    41
    63.1%
    2 (Ambulatory But Unable to Work)
    5
    15.6%
    3
    13.6%
    0
    0%
    0
    0%
    8
    12.3%
    3 (Limited Self-Care)
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    4 (Completely Disabled)
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    5 (Death)
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    Baseline Platelet Transfusion Status (Count of Participants)
    Dependent
    4
    12.5%
    5
    22.7%
    0
    0%
    1
    20%
    10
    15.4%
    Independent
    25
    78.1%
    13
    59.1%
    6
    100%
    4
    80%
    48
    73.8%
    Other
    3
    9.4%
    4
    18.2%
    0
    0%
    0
    0%
    7
    10.8%
    Baseline Red Blood Cell (RBC) Transfusion Status (Count of Participants)
    Dependent
    8
    25%
    7
    31.8%
    0
    0%
    2
    40%
    17
    26.2%
    Independent
    8
    25%
    6
    27.3%
    3
    50%
    2
    40%
    19
    29.2%
    Other
    16
    50%
    9
    40.9%
    3
    50%
    1
    20%
    29
    44.6%
    Average Red Blood Cell (RBC) Transfusion Requirement (units per 56 days) [Median (Full Range) ]
    Median (Full Range) [units per 56 days]
    4.00
    2.50
    1.00
    4.00
    3.00

    Outcome Measures

    1. Primary Outcome
    Title Overall Response Rate Based on the Modified International Working Group (IWG) 2006 Response Criteria for Myelodysplastic Syndrome (MDS)
    Description The overall response rate (ORR) was defined as the percentage of participants who achieved an objective response including: hematologic improvement (HI), partial remission (PR), complete remission (CR), or marrow complete remission (mCR). Hematologic response was defined as: CR: ≤ 5% myeloblasts with normal maturation of all cell lines; peripheral blood (PB) shows: hemoglobin ≥11 g/dL, neutrophils ≥1.0x10^9/L, platelets ≥100x10^9/dL, blasts (0%) PR: same as CR bone marrow (BM) shows blasts decreased by ≥ 50% over pre-treatment but still > 5%; cellularity and morphology not relevant mCR: BM: ≤ 5% myeloblasts and decrease by ≥ 50% over pre-treatment PB, PB: if HI responses, noted in addition to mCR HI: HI erythroid response (HI-E); HI neutrophil response (HI-N) ; HI platelet response (HI-P)
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The intent-to-treat (ITT) population included all enrolled participants who received at least one dose of investigational product (IP).
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Number (95% Confidence Interval) [Percentage of Participants]
    6.3
    19.7%
    4.5
    20.5%
    16.7
    278.3%
    0
    0%
    2. Secondary Outcome
    Title Kaplan-Meier Estimate of Overall Survival
    Description Overall survival (OS) was defined as the time from randomization to death from any cause, and was calculated using date of first dose and date of death, or date of last follow-up for censored subjects. All participants were followed until drop out (withdrawal of consent from further data collection or lost to follow-up), death, or study termination. Participants who dropped out or were alive at study termination (or at the time of the interim analysis) had their OS times censored at the time of last contact, as appropriate.
    Time Frame From day 1 of study drug to the data cut off date of 19 June 2019; median follow-up for OS = 15.96 and 8.35 months respectively in the SD and PD oral AZA alone arms and 13.48 and 13.33 months in the SD and PD combination arms

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Median (95% Confidence Interval) [Months]
    17.00
    6.28
    14.70
    14.56
    3. Secondary Outcome
    Title Kaplan Meier Estimate of Time to Onset of First and Best Response
    Description Time to onset of first response was defined as the time between the date of first investigational product (IP) dose and the earliest date any response (CR, PR, mCR, or HI) was first observed. Participants who did not achieve any defined response during the treatment period were censored at the date of treatment discontinuation, disease progression, or death, whichever occurred first. Best response is the best recorded response or treatment outcome from the start of the study treatment until the end of the study treatment taking into account the requirements for confirmation of response.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Onset of First Response
    NA
    11.97
    NA
    NA
    Onset of Best Response
    3.68
    4.41
    3.29
    2.17
    4. Secondary Outcome
    Title Kaplan Meier Estimate of Duration of First Response
    Description Duration of hematologic response and/or improvement was defined as the time from the date response or improvement was first observed to the date of documented relapse or disease progression as defined by the modified IWG 2006 criteria. Particpants who maintained hematologic response and/or improvement through the end of the treatment period were censored as the date of treatment discontinuation or death, whichever occurred first.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP; participants who achieved a response.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 3 1 6 5
    Median (95% Confidence Interval) [Months]
    9.24
    NA
    NA
    NA
    5. Secondary Outcome
    Title Kaplan Meier Estimate of Duration of Best Response
    Description Duration of hematologic response and/or improvement was defined as the time from the date response or improvement was first observed to the date of documented relapse or disease progression as defined by the modified IWG 2006 criteria. Particpants who maintained hematologic response and/or improvement through the end of the treatment period were censored as the date of treatment discontinuation or death, whichever occurred first.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP; participants who achieved a best response.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Median (95% Confidence Interval) [Months]
    NA
    NA
    NA
    NA
    6. Secondary Outcome
    Title Kaplan-Meier Estimate of Progression Free Survival (PFS)
    Description Progression-free survival is defined as the time from randomization to the first documented progressive disease (PD), relapse, or death due to any cause during or after the treatment period, whichever occurred first, according to IWG 2006 response criteria for MDS. Participants who were still alive and progression-free were censored at the date of their last response assessment. Progressive disease is defined as follows: - an increase in BM blasts relative to nadir: •If nadir less than 5% blasts: ≥ 50% increase in blasts to > 5% blasts •If nadir 5% - 10% blasts: ≥ 50% increase in blasts to > 10% blasts •If nadir 10% - 20% blasts: ≥ 50% increase in blasts to > 20% blasts •If nadir 20% - 30% blasts: ≥ 50% increase in blasts to > 30% blasts And any of the following: •At least 50% decrement from maximum remission/response levels in granulocytes or platelets •Reduction in Hgb concentration by ≥ 2 g/dL •Transfusion dependence
    Time Frame From day 1 of study drug to the data cut off date of 19 June 2019; median follow-up for OS = 15.96 and 8.35 months respectively in the SD/ PD oral AZA arms and 13.48 and 13.33 months in the SD/PD oral AZA and Durva arms

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Median (95% Confidence Interval) [Months]
    14.86
    6.28
    14.70
    12.10
    7. Secondary Outcome
    Title Percentage of Participants With Progressive Disease at Baseline Who Achieved Stable Disease
    Description A participant was considered as having a stable disease if the disease neither responded nor progressed during or after study treatment.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    Includes participants who had progressive disease at baseline who achieved stable disease.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 0 22 0 5
    Number [Percentage of Participants]
    31.8
    99.4%
    20.0
    90.9%
    8. Secondary Outcome
    Title Kaplan-Meier Estimate of Onset to Achieve Stable Disease
    Description A participant was considered as having a stable disease if the disease neither responded nor progressed during or after study treatment.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    Includes all participants in the ITT population with progressive disease at baseline and achieved stable disease; those who didn't achieve SD or better were censored.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 0 22 0 5
    Median (95% Confidence Interval) [Months]
    NA
    NA
    9. Secondary Outcome
    Title Kaplan-Meier Estimate of Duration of Stable Disease
    Description The duration of stable disease was defined as the time between any two observations of objective disease progression (modified IWG criteria), starting from the first day of dosing with IP. Participants who maintained stable disease through the end of the treatment period were censored at the date of study termination.
    Time Frame Response was assessed every 2 cycles following treatment during the first 6 cycles, then every 3 cycles thereafter; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively, and 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The population includes participants who achieved stable disease as their best response.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 0 5
    Median (95% Confidence Interval) [Months]
    NA
    NA
    NA
    10. Secondary Outcome
    Title Percentage of Participants Who Progressed to Acute Myelogenous Leukemia (AML)
    Description For all participants who received at least one dose of study drug, continuous monitoring for progression to AML occurred in the post treatment follow up period.
    Time Frame From Day 1 of study drug up to the data cut off date of 19 June 2019; median follow up time for AML progression = 11.59 and 5.65 months respectively in the SD and PD arms for oral AZA and 6.21 months for SD/PD in the combination arm.

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of IP.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Number [Percentage of Participants]
    28.1
    87.8%
    18.2
    82.7%
    16.7
    278.3%
    60.0
    1200%
    11. Secondary Outcome
    Title Kaplan-Meier Estimate of Time to Progression to AML
    Description Time to AML progression was defined as the time from the date of first dose of IP until the date the participant had documented progression to AML.
    Time Frame From Day 1 of study drug up to the data cut off date of 19 June 2019; median follow up time for AML progression = 11.59 and 5.65 months respectively in the SD and PD arms for oral AZA and 6.21 months for SD/PD in the combination arm.

    Outcome Measure Data

    Analysis Population Description
    The ITT population included all enrolled participants who received at least one dose of investigational product (IP).
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    Median (95% Confidence Interval) [Months]
    NA
    NA
    NA
    6.21
    12. Secondary Outcome
    Title Number of Participants With Treatment Emergent Adverse Events (TEAEs)
    Description TEAEs were defined as AEs occurring or worsening on or after the date of the first dose of oral aza or durva and within 28 days after last dose of oral aza or 90 days after last dose of durva A serious adverse event (SAE) is any: Death; Life-threatening event; Any inpatient hospitalization or prolongation of existing hospitalization; Persistent or significant disability or incapacity; Congenital anomaly or birth defect; Any other important medical event The severity of an AE was evaluated by the investigator according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) (Version 4.0) where Grade 1 = Mild, Grade 2 = Moderate, Grade 3 = Severe, Grade 4 = Life-threatening and Grade 5 = Death.
    Time Frame From first dose of IP until 28 days after final oral AZA dose, 90 days after final durva dose and/or treatment stopped; median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively; 1.84 months for AZA and Durva SD/PD arms

    Outcome Measure Data

    Analysis Population Description
    The safety population included all enrolled participants who received at least 1 dose of IP and had at least 1 post-dose safety assessment.
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    Measure Participants 32 22 6 5
    ≥ 1 TEAE
    32
    100%
    22
    100%
    6
    100%
    5
    100%
    ≥ 1 TEAE Related to (R/T) Oral Azacitidine (AZA)
    28
    87.5%
    20
    90.9%
    6
    100%
    4
    80%
    ≥ 1 TEAE R/T Durvalumab (Durva)
    0
    0%
    0
    0%
    5
    83.3%
    4
    80%
    ≥ 1 TEAE R/T Oral AZA or Durva
    28
    87.5%
    20
    90.9%
    6
    100%
    4
    80%
    ≥ 1 Serious TEAE
    25
    78.1%
    15
    68.2%
    4
    66.7%
    5
    100%
    ≥ 1 Serious TEAE R/T Oral AZA
    4
    12.5%
    3
    13.6%
    2
    33.3%
    1
    20%
    ≥ 1 Serious TEAE R/T Durva
    0
    0%
    0
    0%
    1
    16.7%
    1
    20%
    ≥ 1 Serious TEAE R/T Oral AZA or Durva
    4
    12.5%
    3
    13.6%
    2
    33.3%
    1
    20%
    ≥ 1 NCI CTC Grade (GR) 3 or 4 TEAE
    32
    100%
    19
    86.4%
    5
    83.3%
    5
    100%
    ≥ 1 NCI CTC GR 3 or 4 TEAE R/T Oral AZA
    18
    56.3%
    10
    45.5%
    4
    66.7%
    3
    60%
    ≥ 1 NCI CTC GR 3 or 4 TEAE R/T Durva
    0
    0%
    0
    0%
    2
    33.3%
    2
    40%
    ≥ 1 NCI CTC GR 3 or 4 TEAE R/T AZA or Durva
    18
    56.3%
    10
    45.5%
    4
    66.7%
    3
    60%
    ≥ 1 TEAE Leading to Death
    4
    12.5%
    4
    18.2%
    2
    33.3%
    0
    0%
    ≥ 1 TEAE Leading to Death R/T Oral AZA
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    ≥ 1 TEAE Leading to Death R/T Durva
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    ≥ 1 TEAE Leading to Death R/T AZA or Durva
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    ≥ 1 TEAE Leading to Dose Reduction of AZA
    9
    28.1%
    8
    36.4%
    1
    16.7%
    0
    0%
    ≥ 1 TEAE Leading to Reduction of Durva
    0
    0%
    0
    0%
    0
    0%
    0
    0%
    ≥ 1 TEAE Leading to Reduction of AZA or Durva
    9
    28.1%
    8
    36.4%
    1
    16.7%
    0
    0%
    ≥1 TEAE Leading to Interruption of AZA
    21
    65.6%
    13
    59.1%
    3
    50%
    4
    80%
    ≥1 TEAE Leading to Interruption of Durva
    0
    0%
    0
    0%
    2
    33.3%
    0
    0%
    ≥1 TEAE Leading to Interruption of AZA or Durva
    21
    65.6%
    12
    54.5%
    3
    50%
    4
    80%
    ≥ 1 TEAE Leading to Discontinuation (D/C) of AZA
    15
    46.9%
    8
    36.4%
    4
    66.7%
    4
    80%
    ≥ 1 TEAE Leading to D/C of Durva
    0
    0%
    0
    0%
    3
    50%
    3
    60%
    ≥ 1 TEAE Leading to D/C of AZA or Durva
    15
    46.9%
    8
    36.4%
    4
    66.7%
    4
    80%

    Adverse Events

    Time Frame From the date of the first dose of study drug until 28 days after the last dose of CC-486, 90 days after the last dose of durvalumab, or until the treatment discontinuation visit, whichever was later and those SAEs known to the investigator at any time thereafter that were suspected of being related to IP.
    Adverse Event Reporting Description Median duration of treatment = 5.26 and 3.81 months for SD/PD for oral AZA arms respectively; 1.84 months for AZA and Durva SD/PD arms
    Arm/Group Title Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Arm/Group Description Participants were given oral azacitidine (AZA) 100 mg, 150 mg, or 200 mg tablets twice daily (BID) on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants were given oral azacitidine 100 mg, 150mg, or 200mg tablets BID on days 1 to 21 of each 28-day treatment cycle. Participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine (AZA) tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab (Durva) 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred. Participants received 100 mg oral azacitidine tablets BID on days 1 to 14 or days 1 to 21 of each 28-day treatment cycle and durvalumab 1500 mg by intravenous infusion on day 1 of each 28-day treatment cycle; participants continued to receive their assigned study treatment unless disease progression, unacceptable toxicity, lost to follow-up or withdrawal by participant occurred.
    All Cause Mortality
    Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 20/32 (62.5%) 16/22 (72.7%) 3/6 (50%) 5/5 (100%)
    Serious Adverse Events
    Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 25/32 (78.1%) 15/22 (68.2%) 4/6 (66.7%) 5/5 (100%)
    Blood and lymphatic system disorders
    Febrile neutropenia 2/32 (6.3%) 1/22 (4.5%) 2/6 (33.3%) 0/5 (0%)
    Hyperleukocytosis 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Leukocytosis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Thrombocytopenia 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Cardiac disorders
    Myocardial infarction 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Gastrointestinal disorders
    Constipation 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Diarrhoea 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Haematemesis 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Intestinal ischaemia 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Nausea 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Rectal haemorrhage 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    General disorders
    Death 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    General physical health deterioration 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Oedema peripheral 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pyrexia 2/32 (6.3%) 0/22 (0%) 1/6 (16.7%) 2/5 (40%)
    Hepatobiliary disorders
    Autoimmune hepatitis 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Hepatitis 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Infections and infestations
    Appendicitis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Arthritis infective 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Bacteraemia 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Bacterial sepsis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Cellulitis 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Cellulitis gangrenous 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Clostridium difficile colitis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Diverticulitis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Fungaemia 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Gastroenteritis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Gastroenteritis norovirus 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Lower respiratory tract infection 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Neutropenic sepsis 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Oral herpes 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pneumonia 8/32 (25%) 2/22 (9.1%) 0/6 (0%) 2/5 (40%)
    Pseudomonal sepsis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Sepsis 2/32 (6.3%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Serratia bacteraemia 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Skin infection 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Upper respiratory tract infection 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Urinary tract infection enterococcal 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Injury, poisoning and procedural complications
    Femur fracture 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Investigations
    Ejection fraction decreased 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    White blood cell count increased 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Metabolism and nutrition disorders
    Failure to thrive 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Tumour lysis syndrome 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Musculoskeletal and connective tissue disorders
    Back pain 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Bone pain 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Joint effusion 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Myositis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Neck pain 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Neoplasms benign, malignant and unspecified (incl cysts and polyps)
    Glioma 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Sarcoma 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Transformation to acute myeloid leukaemia 8/32 (25%) 3/22 (13.6%) 1/6 (16.7%) 3/5 (60%)
    Nervous system disorders
    Ataxia 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Dementia 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Psychiatric disorders
    Confusional state 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Renal and urinary disorders
    Acute kidney injury 1/32 (3.1%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Haematuria 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Urinary retention 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Respiratory, thoracic and mediastinal disorders
    Epistaxis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pneumonia aspiration 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pulmonary oedema 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Respiratory distress 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Respiratory failure 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Skin and subcutaneous tissue disorders
    Acute febrile neutrophilic dermatosis 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Erythema 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pyoderma gangrenosum 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Other (Not Including Serious) Adverse Events
    Stable Disease (SD) Cohort: Oral Azacitidine Progressive Disease (PD) Cohort: Oral Azacitidine Stable Disease Cohort: Oral Azacitidine and Durvalumab Progressive Disease Cohort: Oral Azacitidine and Durvalumab
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 32/32 (100%) 22/22 (100%) 6/6 (100%) 5/5 (100%)
    Blood and lymphatic system disorders
    Anaemia 11/32 (34.4%) 9/22 (40.9%) 1/6 (16.7%) 3/5 (60%)
    Febrile neutropenia 3/32 (9.4%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Neutropenia 12/32 (37.5%) 6/22 (27.3%) 1/6 (16.7%) 2/5 (40%)
    Thrombocytopenia 6/32 (18.8%) 5/22 (22.7%) 1/6 (16.7%) 4/5 (80%)
    Cardiac disorders
    Atrial fibrillation 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 2/5 (40%)
    Pericardial effusion 0/32 (0%) 0/22 (0%) 0/6 (0%) 2/5 (40%)
    Sinus bradycardia 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Sinus tachycardia 0/32 (0%) 0/22 (0%) 0/6 (0%) 2/5 (40%)
    Tachycardia 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Tricuspid valve disease 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Ear and labyrinth disorders
    Ear pain 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Hypoacusis 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Vertigo positional 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Endocrine disorders
    Hypothyroidism 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Eye disorders
    Conjunctival haemorrhage 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Gastrointestinal disorders
    Abdominal distension 3/32 (9.4%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Abdominal pain 3/32 (9.4%) 2/22 (9.1%) 2/6 (33.3%) 1/5 (20%)
    Abdominal pain upper 3/32 (9.4%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Constipation 12/32 (37.5%) 9/22 (40.9%) 1/6 (16.7%) 2/5 (40%)
    Diarrhoea 20/32 (62.5%) 11/22 (50%) 4/6 (66.7%) 2/5 (40%)
    Dry mouth 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 1/5 (20%)
    Duodenogastric reflux 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Dyspepsia 1/32 (3.1%) 3/22 (13.6%) 2/6 (33.3%) 1/5 (20%)
    Dysphagia 2/32 (6.3%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Flatulence 3/32 (9.4%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Gastrointestinal haemorrhage 0/32 (0%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Gastrooesophageal reflux disease 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Gingival bleeding 2/32 (6.3%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Haemorrhoids 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Mouth haemorrhage 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 2/5 (40%)
    Nausea 24/32 (75%) 12/22 (54.5%) 2/6 (33.3%) 2/5 (40%)
    Oral mucosa haematoma 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Perianal erythema 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Periodontal disease 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Stomatitis 4/32 (12.5%) 3/22 (13.6%) 1/6 (16.7%) 1/5 (20%)
    Vomiting 18/32 (56.3%) 11/22 (50%) 2/6 (33.3%) 3/5 (60%)
    General disorders
    Asthenia 9/32 (28.1%) 4/22 (18.2%) 0/6 (0%) 0/5 (0%)
    Catheter site erythema 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Chills 1/32 (3.1%) 0/22 (0%) 1/6 (16.7%) 1/5 (20%)
    Fatigue 10/32 (31.3%) 6/22 (27.3%) 2/6 (33.3%) 4/5 (80%)
    Generalised oedema 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Oedema peripheral 6/32 (18.8%) 2/22 (9.1%) 1/6 (16.7%) 3/5 (60%)
    Pyrexia 7/32 (21.9%) 3/22 (13.6%) 1/6 (16.7%) 1/5 (20%)
    Immune system disorders
    Haemophagocytic lymphohistiocytosis 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Infections and infestations
    Cellulitis 2/32 (6.3%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Diverticulitis 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Folliculitis 0/32 (0%) 3/22 (13.6%) 0/6 (0%) 0/5 (0%)
    Nasopharyngitis 2/32 (6.3%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Oral candidiasis 3/32 (9.4%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Oral herpes 4/32 (12.5%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Pneumonia 3/32 (9.4%) 2/22 (9.1%) 0/6 (0%) 2/5 (40%)
    Rash pustular 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Staphylococcal infection 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Upper respiratory tract infection 6/32 (18.8%) 2/22 (9.1%) 1/6 (16.7%) 0/5 (0%)
    Urinary tract infection 3/32 (9.4%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Injury, poisoning and procedural complications
    Contusion 5/32 (15.6%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Fall 5/32 (15.6%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Skin wound 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Spinal compression fracture 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Transfusion reaction 1/32 (3.1%) 2/22 (9.1%) 0/6 (0%) 1/5 (20%)
    Investigations
    Alanine aminotransferase increased 2/32 (6.3%) 4/22 (18.2%) 0/6 (0%) 1/5 (20%)
    Aspartate aminotransferase increased 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Blood alkaline phosphatase increased 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Blood bilirubin increased 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Blood creatinine increased 3/32 (9.4%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Cardiac murmur 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Electrocardiogram QT prolonged 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Platelet count decreased 0/32 (0%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Urine output decreased 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Weight decreased 10/32 (31.3%) 6/22 (27.3%) 0/6 (0%) 0/5 (0%)
    White blood cell count increased 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Metabolism and nutrition disorders
    Decreased appetite 15/32 (46.9%) 8/22 (36.4%) 1/6 (16.7%) 2/5 (40%)
    Dehydration 2/32 (6.3%) 1/22 (4.5%) 1/6 (16.7%) 0/5 (0%)
    Gout 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Hyperglycaemia 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Hyperkalaemia 1/32 (3.1%) 3/22 (13.6%) 0/6 (0%) 0/5 (0%)
    Hyperphosphataemia 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Hyperuricaemia 0/32 (0%) 2/22 (9.1%) 1/6 (16.7%) 1/5 (20%)
    Hypoalbuminaemia 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Hypocalcaemia 2/32 (6.3%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Hypokalaemia 4/32 (12.5%) 0/22 (0%) 0/6 (0%) 2/5 (40%)
    Hypomagnesaemia 3/32 (9.4%) 2/22 (9.1%) 0/6 (0%) 1/5 (20%)
    Hypophosphataemia 0/32 (0%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Musculoskeletal and connective tissue disorders
    Arthralgia 4/32 (12.5%) 3/22 (13.6%) 0/6 (0%) 0/5 (0%)
    Back pain 7/32 (21.9%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Muscle spasms 1/32 (3.1%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Muscular weakness 4/32 (12.5%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Myalgia 3/32 (9.4%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Polyarthritis 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Nervous system disorders
    Dizziness 6/32 (18.8%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Dysgeusia 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Headache 4/32 (12.5%) 1/22 (4.5%) 1/6 (16.7%) 0/5 (0%)
    Lethargy 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Somnolence 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Psychiatric disorders
    Delirium 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Depression 0/32 (0%) 2/22 (9.1%) 0/6 (0%) 1/5 (20%)
    Insomnia 5/32 (15.6%) 2/22 (9.1%) 0/6 (0%) 1/5 (20%)
    Irritability 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Renal and urinary disorders
    Acute kidney injury 1/32 (3.1%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Dysuria 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pollakiuria 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Renal cyst 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Renal disorder 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Urinary incontinence 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Urinary retention 2/32 (6.3%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Urinary tract obstruction 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Reproductive system and breast disorders
    Oedema genital 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Penile pain 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Respiratory, thoracic and mediastinal disorders
    Cough 7/32 (21.9%) 4/22 (18.2%) 0/6 (0%) 1/5 (20%)
    Dyspnoea 9/32 (28.1%) 4/22 (18.2%) 0/6 (0%) 0/5 (0%)
    Dyspnoea exertional 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Epistaxis 3/32 (9.4%) 1/22 (4.5%) 0/6 (0%) 2/5 (40%)
    Hypoxia 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Nasal congestion 1/32 (3.1%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Oropharyngeal pain 3/32 (9.4%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Pleural effusion 1/32 (3.1%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Productive cough 2/32 (6.3%) 2/22 (9.1%) 0/6 (0%) 0/5 (0%)
    Pulmonary hypertension 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Skin and subcutaneous tissue disorders
    Decubitus ulcer 0/32 (0%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Dermatitis bullous 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Erythema multiforme 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)
    Night sweats 1/32 (3.1%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Petechiae 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pruritus 3/32 (9.4%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Pruritus generalised 2/32 (6.3%) 1/22 (4.5%) 0/6 (0%) 0/5 (0%)
    Rash 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Rash maculo-papular 3/32 (9.4%) 1/22 (4.5%) 1/6 (16.7%) 1/5 (20%)
    Urticaria 1/32 (3.1%) 0/22 (0%) 1/6 (16.7%) 0/5 (0%)
    Vascular disorders
    Haematoma 1/32 (3.1%) 1/22 (4.5%) 0/6 (0%) 1/5 (20%)
    Hypertension 2/32 (6.3%) 0/22 (0%) 0/6 (0%) 0/5 (0%)
    Hypotension 1/32 (3.1%) 1/22 (4.5%) 0/6 (0%) 3/5 (60%)
    Thrombophlebitis superficial 0/32 (0%) 0/22 (0%) 0/6 (0%) 1/5 (20%)

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    Results from a center cannot be submitted for publication before results of multicenter study are published unless it is > 1 year since study completion. Then, Investigator can publish if manuscript is submitted to Celgene 60 days prior to submission. If Celgene decides publication would hinder drug development, Investigator must delay submission for up to 90 additional days. Investigator must delete confidential information before submission and defer publication to permit patent applications.

    Results Point of Contact

    Name/Title Anne McClain, Senior Manager, Clinical Trial Disclosure
    Organization Celgene Corporation
    Phone 908-673-9100
    Email ClinicalTrialDisclosure@Celgene.com
    Responsible Party:
    Celgene
    ClinicalTrials.gov Identifier:
    NCT02281084
    Other Study ID Numbers:
    • CC-486-MDS-006
    • 2014-002675-29
    First Posted:
    Nov 2, 2014
    Last Update Posted:
    Aug 23, 2022
    Last Verified:
    Aug 1, 2022