An Efficacy and Safety Study of Azacitidine Subcutaneous in Combination With Durvalumab (MEDI4736) in Previously Untreated Adults With Higher-Risk Myelodysplastic Syndromes (MDS) or in Elderly Patients With Acute Myeloid Leukemia (AML)

February 1, 2023 updated by: Celgene

A Randomized, Multicenter, Open-label, Phase 2 Study Evaluating the Efficacy and Safety of Azacitidine Subcutaneous in Combination With Durvalumab (MEDI4736) in Previously Untreated Subjects With Higher-Risk Myelodysplastic Syndromes (MDS) or in Elderly (>= 65 Years) Acute Myeloid Leukemia (AML) Subjects Not Eligible for Hematopoietic Stem Cell Transplantation (HSCT)

The primary objective of this study is to evaluate the efficacy of subcutaneous azacitidine in combination with durvalumab as compared with subcutaneous azacitidine alone in adults with previously untreated, higher risk MDS who are not eligible for HSCT or in adults ≥ 65 years old with previously untreated AML who are not eligible for HSCT, with intermediate or poor cytogenetic risk.

Study Overview

Study Type

Interventional

Enrollment (Actual)

213

Phase

  • Phase 2

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Graz, Austria, 8036
        • Medizinische Universität Graz
      • Innsbruck, Austria, 6020
        • Medizinische Universität Innsbruck
      • Linz, Austria, 4020
        • Elisabethinen Hospital Linz
      • Salzburg, Austria, 5020
        • Salzburger Landkliniken St. Johanns-Spital
      • Wein, Austria, 1090
        • Akh Wien
      • Wien, Austria, 1140
        • Hanusch Krankenhaus der Stadt Wien
      • Wien, Austria, 1140
        • Local Institution - 653
      • Brussels, Belgium, 1200
        • Cliniques Universitaires St-Luc
      • Charleroi, Belgium, 6000
        • Grand Hopital de Charleroi
      • Gent, Belgium, 9000
        • Local Institution - 202
      • Gent, Belgium, 9000
        • UH Gent
      • Leuven, Belgium, 3000
        • UH Gasthuisberg
      • Yvoir, Belgium, 5530
        • Cliniques Universitaires UCL de Mont-Godine
    • Alberta
      • Edmonton, Alberta, Canada, T6G2B7
        • University of Alberta
    • Manitoba
      • Winnipeg, Manitoba, Canada, R3E 0V9
        • CancerCare Manitoba
    • New Brunswick
      • Saint John, New Brunswick, Canada, E2L 4L2
        • Saint John Regional Hospital
    • Ontario
      • Ottawa, Ontario, Canada, K1H 8L6
        • Ottawa General Hospital
      • Toronto, Ontario, Canada, M5G 2M9
        • Princess Margaret Cancer Centre
    • Quebec
      • Montreal, Quebec, Canada, H2L 4M1
        • CHUM - Notre Dame
      • Angers, France, 67091
        • Centre Hospitalier Universitaire d' Angers
      • Bobigny Cedex, France, 93009
        • Hôpital Avicenne
      • Creteil, France, 94010
        • Hôpital Henri Mondor
      • La Tronche, France, 38700
        • Centre Hospitalier Universitaire de Grenoble Hopital Albert Michallon
      • Lyon, France, 69008
        • Centre Leon Berard
      • Lyon, France, 69008
        • Local Institution - 261
      • Nantes, France, 44093
        • CHRU de Nantes - Hotel Dieu
      • Paris, France, 75010
        • Hôpital Saint louis
      • Paris, France, 75010
        • Local Institution - 251
      • Pessac, France, 33604
        • CHU Bordeaux
      • Pessac, France, 33604
        • Local Institution - 254
      • Pierre-Bénite Cedex, France, 69495
        • Centre Hospitalier Lyon Sud
      • Toulouse, France, 31059 Cedex 9
        • IUCT Oncopole
      • Dresden, Germany, 01307
        • Universitätsklinikum Carl Gustav Carus
      • Dresden, Germany, 01307
        • Local Institution - 604
      • Dusseldorf, Germany, 40479
        • Marien Hospital
      • Dusseldorf, Germany, 40479
        • Local Institution - 603
      • Essen, Germany, 45122
        • Universitätsklinikum Essen
      • Frankfurt am Main, Germany, 60590
        • Klinikum der Johann Wolfgang Goethe Universitat
      • Freiburg, Germany, 79106
        • Universitatsklinikum Freiburg
      • Hannover, Germany, 30625
        • Medizinische Hochschule HannoverZentrum Innere Medizin
      • Leipzig, Germany, 04103
        • Universitatsklinikum Leipzig
      • Munchen, Germany, 1307
        • Klinikum der LMU Campus Grosshadern
      • Munchen, Germany, 81675
        • Klinikum rechts der Isar der TU Munchen
      • Munchen, Germany, 1307
        • Local Institution - 605
      • Ulm, Germany, 89081
        • Universitatsklinikum Ulm
      • Brecia, Italy, 25123
        • AO Spedali Civili di Brescia
      • Firenze, Italy, 50134
        • Azienda Ospedaliero-Universitaria Careggi
      • Milano, Italy, 20162
        • Ospedale Niguarda Milano
      • Palermo, Italy, 33100
        • Azienda Ospedaliera Ospedali Riuniti Villa Sofia Cervello
      • Pavia, Italy, 27100
        • I.R.C.C.S. Policlinico San Matteo - Universita di Pavia
      • Pavia, Italy, 27100
        • Local Institution - 302
      • Roma, Italy, 133
        • Azienda Ospedaliera Bianchi-Melacrino-Morelli
      • Roma, Italy, 133
        • Local Institution - 303
      • Roma, Italy, 89100
        • Policlinico Agostino Gemelli - Istituto di Ematologia
      • Rome, Italy, 133
        • Azienda Ospedaliera Universitaria Policlinico Tor Vergata
      • Rome, Italy, 133
        • Local Institution - 304
      • Udine, Italy, 30174
        • Azienda Ospedaliero-Universitaria Santa Maria della Misericordia die Udine
      • Varese, Italy, 21100
        • Universita degli Studi dell'Insubria - Ospedale di Circolo e Fondazione Macchi - Varese
      • Amsterdam, Netherlands, 1081 HV
        • VU University Medical Center
      • Brzozow, Poland, 36-200
        • Oddzial Hematologii Onkologicznej Szpital Specjalistyczny w Brzozowie Podkarpacki Osrodek Onkologicz
      • Gdansk, Poland, 80-211
        • Katedra i Klinika Hematologii i Transplantologii Uniwersyteckie Centrum Kliniczne
      • Lubin, Poland, 20-081
        • Samodzielny Publiczny Szpital Kliniczny nr 1 Klinika Hematoonkologii i Transplantacji Szpiku
      • Olsztyn, Poland, 10-228
        • Oddzial Hematologii Samodzielny Publiczny Zaklad Opieki Zdrowotnej MSW
      • Wroclaw, Poland, 50-367
        • Klinika Hematologii Nowotworów Krwi i Transplantacji Szpiku
      • Coimbra, Portugal, 3000-076
        • Hospitais da universidade de Coimbra
      • Lisbon, Portugal, 1099-023
        • Instituto Portugues de Oncologia de Lisboa Francisco Gentil EPE
      • Porto, Portugal, 4200
        • Hospital de Sao Joao
      • Porto, Portugal, 4200-072
        • Ipo Instituto Portugues De Oncologia Porto
      • Porto, Portugal, 4200-072
        • Local Institution - 502
      • Barcelona, Spain, 8036
        • Hospital Clinic i Provincial de Barcelona
      • Barcelona, Spain, 28040
        • Hospital Universitario Vall d Hebron
      • Caceres, Spain, 10003
        • Complejo Hospitalario San Pedro de Alcantara
      • Madrid, Spain, 28006
        • Hospital Universitario La Princesa
      • Madrid, Spain, 37007
        • Hospital Gregorio Marañón
      • Madrid, Spain, 37007
        • Local Institution - 555
      • Malaga, Spain, 29010
        • Hospital Universitario Virgen de la Victoria
      • Palma de Mallorca, Spain, 7198
        • Hospital Son Llatzer
      • Salamanca, Spain, 37007
        • Hospital Universitario de Salamanca
      • Sevilla, Spain, 41013
        • Hospital Virgen del Rocío
      • Valencia, Spain, 46010
        • Hospital Clínico Universitario de Valencia
      • Valencia, Spain, 46009
        • Hospital Universitario La Fe
      • Valencia, Spain, 46009
        • Local Institution - 559
      • Birmingham, United Kingdom, B15 2TH
        • University Hospital Birmingham
      • Leeds, United Kingdom, LS1 3EX
        • St James University Hospital
      • London, United Kingdom, SE5 9RS
        • Kings College Hospital
      • London, United Kingdom, EC1 7BE
        • St Bartholomews Hospital
      • London Bloomsbury, United Kingdom, WC1E 6AU
        • University College London Hospital
      • Manchester, United Kingdom, M20 4BX
        • The Christie NHS Foundation Trust
      • Oxford, United Kingdom, OX3 9DU
        • John Radcliffe Hospital
      • Oxford, United Kingdom, OX3 9DU
        • Local Institution - 453
      • Sutton, United Kingdom, SM2 5PT
        • The Royal Marsden NHS Foundation Trust
    • Connecticut
      • New Haven, Connecticut, United States, 06520
        • Yale Cancer Center
    • District of Columbia
      • Washington, District of Columbia, United States, 20007
        • Georgetown University Hospital
    • Florida
      • Gainesville, Florida, United States, 32610
        • University of Florida
      • Tampa, Florida, United States, 33612
        • Moffitt Cancer Center
    • Illinois
      • Chicago, Illinois, United States, 60637
        • University of Chicago
    • New Jersey
      • Hackensack, New Jersey, United States, 07601
        • Hackensack University Medical Center
    • New York
      • Buffalo, New York, United States, 14263
        • Roswell Park Cancer Institute
      • New York, New York, United States, 10029
        • Icahn School of Medicine at Mount Sinai
    • North Carolina
      • Durham, North Carolina, United States, 27705
        • Duke University Medical Center
    • South Dakota
      • Sioux Falls, South Dakota, United States, 57105
        • Avera Cancer Institute
    • Tennessee
      • Nashville, Tennessee, United States, 37203
        • Vanderbilt University Medical Center
    • Texas
      • Houston, Texas, United States, 77230
        • University of Texas- MD Anderson

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion Criteria:

For both cohorts:

  1. Subject must understand and voluntarily sign an informed consent form (ICF) prior to any study-related assessments/procedures being conducted.
  2. Have an Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1, or 2.
  3. Female subjects of childbearing potential may participate, providing they meet the following conditions:

    1. Have 2 negative pregnancy tests as verified by the Investigator prior to starting any investigational product (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 abstinence from heterosexual contact.
    2. Agree to practice true abstinence (which must be reviewed on a monthly basis and source documented) or agree to the use of a highly effective method of contraception use from 28 days prior to starting durvalumab or azacitidine, and must agree to continue using such precautions while taking durvalumab or azacitidine (including dose interruptions) and up to 90 days after the last dose of durvalumab or azacitidine. Cessation of contraception after this point should be discussed with a responsible physician.
    3. Agree to abstain from breastfeeding during study participation and for at least 90 days after the last dose of IP.
    4. Refrain from egg cell donation while taking durvalumab and for at least 90 days after the last dose of durvalumab.
  4. Male subject must:

    1. Either practice true abstinence 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 childbearing 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 durvalumab or azacitidine.
    2. 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 a biomarker-specific component of the informed consent form prior to any study-related procedures conducted.
  6. Willing and able to adhere to the study visit schedule and other protocol requirements.

    MDS Cohort:

  7. Age ≥ 18 years at the time of signing the informed consent form.
  8. Central confirmation of diagnosis of previously untreated primary or secondary myelodysplastic syndromes (MDS) as per World Health Organization (WHO) classification. Results of central pathology review are required prior to receiving the first dose of IP.
  9. Central confirmation of the categorization of the MDS risk classification, as per the Revised - International prognostic scoring system (IPSS-R) Intermediate risk with >10% blasts or poor or very poor cytogenetics, or IPSS-R High or Very High risk (results of central pathology review required prior to receiving the first dose of IP).

    Acute myeloid leukemia (AML) Cohort:

  10. Age ≥ 65 years at the time of signing the informed consent form (ICF).
  11. Central confirmation of diagnosis of one of the following untreated AML as per WHO classification:

    • Newly diagnosed, histologically confirmed de novo AML (bone marrow blasts ≥ 20%), or
    • AML secondary to prior MDS, or
    • AML secondary to exposure to potentially leukemogenic therapies or agents (eg, radiation therapy, alkylating agents, topoisomerase II inhibitors) with the primary malignancy in remission for at least 2 years.
  12. Central confirmation of intermediate or poor risk status, based on Cytogenetics for acute myeloid leukemia.

Exclusion Criteria:

For both cohorts:

  1. Prior hematopoietic stem cell transplant.
  2. Considered eligible for hematopoietic stem cell transplant (allogeneic or autologous) at the time of signing the ICF.
  3. Prior exposure to azacitidine, decitabine or prior exposure to the investigational oral formulation of decitabine, or other oral azacitidine derivative.
  4. Inaspirable bone marrow.
  5. Use of any of the following within 28 days prior to the first dose of IP:

    • Thrombopoiesis-stimulating agents (eg, romiplostim, eltrombopag, Interleukin-11)
    • Any hematopoietic growth factors (erythropoietin-stimulating agents [ESAs], granulocyte colony-stimulating factor (G-CSF) and other red blood cell (RBC) hematopoietic growth factors (eg, Interleukin-3)
    • Any investigational agents within 28 days or 5 half-lives (whichever is longer) of initiating study treatment
  6. Prior history of malignancies (except MDS for AML subjects), unless the subject has been free of the disease for ≥ 2 years. However, subjects with the following history/concurrent conditions are allowed:

    • Basal or squamous cell carcinoma of the skin
    • Carcinoma in situ of the cervix
    • Carcinoma in situ of the breast
    • Incidental histologic finding of prostate cancer (T1a or T1b using the tumor, nodes, metastasis [tumor, node, metastases (TNM)] clinical staging system).
  7. Pregnant or breast-feeding females or females who intend to become pregnant during study participation.
  8. Subject has 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 [exclude only if active within the last 6 months prior to signing the ICF], 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:

    • Subjects with vitiligo or alopecia;
    • Subjects with hypothyroidism (eg, following Hashimoto syndrome) stable on hormone replacement for ≥ 3 months prior to signing the ICF; or
    • Subjects with psoriasis not requiring systemic treatment
  9. Significant active cardiac disease within the previous 6 months prior to signing the ICF, including:

    • New York Heart Association (NYHA) Class III or IV congestive heart failure;
    • Unstable angina or angina requiring surgical or medical intervention; and/or
    • Significant cardiac arrhythmia
    • Myocardial infarction
  10. Uncontrolled intercurrent illness including, but not limited to, ongoing or active systemic fungal, bacterial, or viral infection (defined as ongoing signs/symptoms related to the infection without improvement despite appropriate antibiotics or other treatment), uncontrolled hypertension, cardiac arrhythmia, pneumonitis, interstitial lung disease, active peptic ulcer disease or gastritis that would limit compliance with study requirement.
  11. Known human immunodeficiency virus (HIV) or hepatitis C (HCV) infection, or evidence of active hepatitis B virus (HBV) infection.
  12. Known or suspected hypersensitivity to azacitidine, mannitol, or durvalumab, its constituents, or to any other humanized monoclonal antibody.
  13. Any significant medical condition, laboratory abnormality, or psychiatric illness that would prevent the subject from participating in the study.
  14. Any condition including the presence of laboratory abnormalities, which places the subject at unacceptable risk if he/she were to participate in the study.
  15. Prior anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death-1 (PD-1), or programmed death ligand-1 (PD-L1) or other immune checkpoint mAb exposure.
  16. Other investigational monoclonal antibodies (mAbs) within 6 months prior to first dose of IP.
  17. Current or prior use of immunosuppressive medication within 14 days prior to the first dose of IP. The following are exceptions to this criterion:

    • Intranasal, inhaled, topical, or local steroid injections (eg, intra-articular injection)
    • Systemic corticosteroids at physiologic doses not to exceed 10 mg/day of prednisone or equivalent
    • Steroids as premedication for hypersensitivity reactions (eg, computed tomography [CT] scan premedication)
  18. History of primary immunodeficiency.
  19. Receipt of live, attenuated vaccine within 30 days prior to the first dose of IP (NOTE: Subjects, if enrolled, should not receive live vaccine during the study and for 30 days after the last dose of durvalumab).
  20. Unwilling or unable to complete subject reported outcome assessments without assistance or with minimal assistance from trained site personnel and/or caregiver.
  21. Subjects who have had clinical evidence of central nervous system (CNS) or pulmonary leukostasis, disseminated intravascular coagulation, or CNS leukemia.
  22. Presence of advanced malignant hepatic tumors.
  23. Any of the following laboratory abnormalities:

    • Serum aspartate aminotransferase (AST/SGOT) or alanine aminotransferase (ALT/SGPT) > 2.5 × upper limit of normal (ULN)
    • Serum total bilirubin > 1.5 × 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
    • Serum creatinine > 2.5 × ULN.

    MDS Cohort:

  24. Any previous cytotoxic, cytostatic, hormonal, biological or immunological treatment for MDS (ESA with or without G-CSF are allowed under certain conditions, see exclusion criterion # 5).
  25. Any investigational therapy within 28 days prior to the first dose of IP.
  26. Use of hydroxyurea within 2 weeks prior to obtaining the screening hematology sample and prior to first dose of IP.
  27. Absolute white blood cell (WBC) count ≥ 15 × 10^9/L.

    AML Cohort:

  28. Previous cytotoxic, cytostatic, hormonal, biological or immunological treatment (ESA with or without G-CSF and iron chelating therapy and hydroxyurea are allowed under certain conditions, see exclusion criterion #5) or biologic treatment for AML.
  29. Any investigational therapy within 28 days prior to the first dose of IP.
  30. Use of hydroxyurea within 2 weeks prior to obtaining the screening hematology sample and prior to first dose of IP.
  31. Prior use of targeted therapy agents (eg, FLT3 inhibitors, other kinase inhibitors).
  32. Suspected or proven acute promyelocytic leukemia (French-American-British (FAB) M3) based on morphology, immunophenotype, molecular assay, or karyotype; AML associated with t(9;22) karyotype, biphenotypic acute leukemia or AML with previous hematologic disorder such as chronic myelogenous leukemia or myeloproliferative neoplasms.
  33. Acute myeloid leukemia associated with inv(16), t(8;21), t(16;16), t(15;17) karyotypes or molecular evidence of such translocations if not associated with a c-Kit mutation.
  34. Absolute WBC count ≥ 15 × 10^⁹/L (NOTE: Hydroxyurea is not allowed to attain a WBC count ≤ 15 x 10⁹/L).
  35. Known history or presence of Sweet Syndrome at screening

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Azacitidine + Durvalumab
Participants received 75 mg/m² subcutaneous azacitidine for 7 days every 4 weeks (Q4W) in combination with 1500 mg intravenous durvalumab on Day 1 of every 4 week cycle for at least 6 cycles. Participants who benefited from treatment may have continued treatment until loss of that benefit, disease progression or other treatment discontinuation criterion were met.
Administered by subcutaneous injection on Days 1 to 7 of each 4-week treatment cycle.
Administered by intravenous infusion on Day 1 of every 4-week treatment cycle.
Other Names:
  • MEDI4736
Active Comparator: Azacitidine Alone
Participants received 75 mg/m² subcutaneous azacitidine for 7 days every 4 weeks for at least 6 cycles. Participants who benefited from treatment may have continued treatment until loss of that benefit, disease progression or other treatment discontinuation criterion were met.
Administered by subcutaneous injection on Days 1 to 7 of each 4-week treatment cycle.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
MDS Cohort: Overall Response Rate
Time Frame: Response was assessed following every 3 treatment cycles until treatment discontinuation; median duration of treatment was 239 days (AZA) and 215 days (DUR) in the AZA + DUR group and 210 days in the AZA alone group.
Overall response rate (ORR) is defined as the percentage of participants achieving a complete remission (CR), partial remission (PR), marrow complete remission (mCR), and/or hematological improvement (HI) based on International Working Group (IWG) 2006 response criteria for MDS and central review. CR: ≤ 5% myeloblasts in bone marrow (BM), and peripheral blood: hemoglobin ≥ 11 g/dL; platelets ≥ 100 × 10⁹/L; neutrophils ≥ 1.0 × 10⁹/L; blasts 0% PR: BM blasts decreased by ≥ 50% but still > 5%; peripheral blood as for CR mCR: BM ≤ 5% myeloblasts and decrease by ≥ 50% HI: Any of the following: •Hemoglobin increase by ≥ 1.5 g/dL or reduction of units of red blood cell (RBC) transfusions of at least 4 RBC transfusions/8 weeks compared with pretreatment •Absolute increase in platelets of ≥ 30 × 10⁹/L if pretreatment value > 20 × 10⁹/L or increase from < 20 × 10⁹/L to > 20 × 10⁹/L and by at least 100% •At least 100% increase in neutrophils and an absolute increase of > 0.5 × 10⁹/L
Response was assessed following every 3 treatment cycles until treatment discontinuation; median duration of treatment was 239 days (AZA) and 215 days (DUR) in the AZA + DUR group and 210 days in the AZA alone group.
AML Cohort: Overall Response Rate
Time Frame: Response was assessed following every 3 treatment cycles until treatment discontinuation; median duration of treatment was 198 days (AZA) and 171 days (DUR) in the AZA + DUR group and 203 days in the AZA alone group.
Overall response rate for AML is defined as the percentage of participants achieving an overall response of morphologic complete remission (CR) or morphologic complete remission with incomplete blood count recovery (CRi) based on modified IWG 2003 response criteria for AML and central review. CR: The following conditions must be met: •Absolute neutrophil count (ANC) ≥ 1.0 x10⁹/L •Platelet count ≥ 100 x10⁹/L •The bone marrow should contain less than 5% blast cells; •Auer rods should not be detectable; •No platelet, or whole blood transfusions for 7days prior to the date of the hematology assessment. CRi: Defined as a morphologic complete remission but the ANC count may be < 1.0 x10⁹/L and/or the platelet count may be < 100 x10⁹/L.
Response was assessed following every 3 treatment cycles until treatment discontinuation; median duration of treatment was 198 days (AZA) and 171 days (DUR) in the AZA + DUR group and 203 days in the AZA alone group.

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Durvalumab Serum Concentration
Time Frame: Cycle 1 Day 1 end of infusion (EOI), Cycle 2 Day 1 pre-infusion, Cycle 4 Day 1 pre-infusion and EOI, and Cycle 6 Day 1 pre-infusion
Cycle 1 Day 1 end of infusion (EOI), Cycle 2 Day 1 pre-infusion, Cycle 4 Day 1 pre-infusion and EOI, and Cycle 6 Day 1 pre-infusion
MDS Cohort: Kaplan Meier Estimate of Time to First Response
Time Frame: From randomization to the earliest date any response (up to approximately 34 months)
Time to first response is defined as the time from randomization to the earliest date any response (complete remission (CR), partial remission (PR), marrow complete remission (mCR), and/or hematological improvement (HI)) based on International Working Group (IWG) 2006 response criteria for MDS and central review. Participants who did not achieve any defined response were censored at the date of last adequate response assessment, disease progression, or death, whichever occurred first. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization to the earliest date any response (up to approximately 34 months)
MDS Cohort: Kaplan Meier Estimate of Relapse-free Survival
Time Frame: From randomization to to the date of disease relapse or death from any cause, whichever occurred first (up to approximately 34 months)
Relapse-free survival is defined as the time from the date of first documented response (complete remission (CR), partial remission (PR)) to the date of disease relapse or death from any cause, whichever occurred first according to the International Working Group (IWG) 2006 response criteria for MDS and central review. Participants who were still alive and progression-free were censored at the date of their last response assessment. Participants who received a subsequent therapy before the date of disease relapse or death were censored at the time of subsequent therapy. Relapse after CR or PR is defined as at least one of the following: •Return to pretreatment bone marrow blast % •Decrement of ≥ 50% from maximum remission/response levels in granulocytes or platelets •Reduction in hemoglobin concentration by ≥ 1.5 g/dL or transfusion dependence. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization to to the date of disease relapse or death from any cause, whichever occurred first (up to approximately 34 months)
MDS Cohort: Percentage of Participants Who Achieved a Cytogenetic Response
Time Frame: From randomization up to approximately 34 months
Cytogenetic response is defined as the percentage of participants who achieved a complete cytogenetic response or partial cytogenetic response according to the International Working Group (IWG) 2006 response criteria and central review. Complete cytogenetic response: Disappearance of the baseline chromosomal abnormality without appearance of new abnormalities. Partial cytogenetic response: At least 50% reduction of the chromosomal abnormality. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization up to approximately 34 months
MDS Cohort: Kaplan-Meier Estimate of Progression-free Survival (PFS)
Time Frame: From randomization to the first documented progressive disease (PD), relapse, or death due to any cause (up to approximately 34 months)
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 the International Working Group (IWG) 2006 response criteria for MDS and central review. 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
From randomization to the first documented progressive disease (PD), relapse, or death due to any cause (up to approximately 34 months)
MDS Cohort: Kaplan-Meier Estimate of Duration of Response
Time Frame: From randomization to the first overall response, or death (up to approximately 34 months)
Duration of response is defined as the time from when the first overall response (complete remission (CR), partial remission (PR), marrow complete remission (mCR), and/or hematological improvement (HI)) was observed until relapse, progressive disease (PD), or death, as defined by the International Working Group (IWG) 2006 response criteria and central review. If no relapse, PD, or death was observed, the duration of response was censored at the last response assessment date that the participant was known to be progression-free. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization to the first overall response, or death (up to approximately 34 months)
MDS Cohort: Kaplan-Meier Estimate of Time to AML Transformation
Time Frame: From randomization to the date the participant had documented transformation to AML (up to approximately 34 months)
Participants were monitored for transformation to acute myeloid leukemia (AML) until death, lost to follow-up, withdrawal of consent for further data collection, or the end of the trial. Time to transformation to AML is defined as the time from the date of randomization until the date the participant had documented transformation to AML (defined as at least 30% of myeloblasts in the bone marrow). Participants with no transformation to AML were censored at the date of their last disease assessment.
From randomization to the date the participant had documented transformation to AML (up to approximately 34 months)
MDS Cohort: Percentage of Participants With Disease Transformation to AML
Time Frame: From randomization until death, lost to follow-up, withdrawal of consent for further data collection, or the end of the trial (up to approximately 34 months)
Disease transformation to acute myeloid leukemia (AML) is defined as at least 30% myeloblasts in the bone marrow. Participants were monitored for transformation to AML until death, lost to follow-up, withdrawal of consent for further data collection, or the end of the trial. Participants with no transformation to AML were censored at the date of their last disease assessment.
From randomization until death, lost to follow-up, withdrawal of consent for further data collection, or the end of the trial (up to approximately 34 months)
AML Cohort: Kaplan Meier Estimate of Time to First Response
Time Frame: From randomization and the earliest date any response (up to approximately 34 months)
Time to first response is defined as the time between the date of randomization and the earliest date any response (CR or CRi) was observed based on the modified International Working Group (IWG) 2003 response criteria for AML and central review. Participants who did not achieve any defined response were censored at the date of last adequate response assessment, disease progression, or death, whichever occurred first. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization and the earliest date any response (up to approximately 34 months)
AML Cohort: Kaplan Meier Estimate of Relapse-free Survival
Time Frame: From randomization to the date of disease relapse or death from any cause, whichever occurred first (up to approximately 34 months)
Relapse-free survival is defined as time from the date of first documented response (morphologic complete remission (CR) or morphologic complete remission with incomplete blood count recovery (CRi)) to the date of disease relapse or death from any cause, whichever occurred first based on the modified International Working Group (IWG) 2003 response criteria for AML and central review. Participants who were still alive and progression-free were censored at the date of their last response assessment. Participants who received a subsequent therapy before the date of disease relapse or death were censored at the time of subsequent therapy.
From randomization to the date of disease relapse or death from any cause, whichever occurred first (up to approximately 34 months)
AML Cohort: Percentage of Participants Who Achieved a Complete Cytogenetic Response
Time Frame: From randomization up to approximately 34 months)
Complete cytogenetic response (CyCR) based on the modified International Working Group (IWG) 2003 response criteria is defined as morphologic complete remission with a reversion to a normal karyotype. The following conditions must be met: • Absolute neutrophil count (ANC) ≥ 1.0 x10⁹/L • Platelet count ≥ 100 x10⁹/L • The bone marrow should contain less than 5% blast cells; • Auer rods should not be detectable; • No platelet, or whole blood transfusions for 7days prior to the date of the hematology assessment. AND • Reversion to normal karyotype at time of CR (based on ≥ 10 metaphases). Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization up to approximately 34 months)
AML Cohort: Percentage of Participants With Hematologic Improvement
Time Frame: From randomization up to approximately 34 months
Hematological improvement was defined as participants with a erythroid response (HI-E), platelet response (HI-P) or neutrophil response (HI-NE) for at least 8 weeks, according to the IWG 2006 response criteria: Hi-E (in participants with pretreatment hemoglobin < 11 g/dL or red blood cell (RBC)-transfusion dependent): Hemoglobin increase of ≥ 1.5 g/dL, or reduction in units of RBC transfusions of at least 4 RBC transfusions/8 weeks compared with the 8 weeks prior to pretreatment. HI-P (in participants with pretreatment platelet count < 100 × 10⁹/L): Absolute increase in platelets of ≥ 30 × 10⁹/L if pretreatment value > 20 × 10⁹/L or increase from < 20 × 10⁹/L to > 20 × 10⁹/L and by at least 100%. HI-N (in participants with pretreatment neutrophils < 1.0 × 10⁹/L): At least 100% increase in neutrophils and an absolute increase of > 0.5 × 10⁹/L. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization up to approximately 34 months
AML Cohort: Kaplan-Meier Estimate of Duration of Response
Time Frame: From randomization until relapse, PD, or death (up to approximately 34 months)
Duration of response is defined as the time from the first response morphologic complete remission (CR) or morphologic complete remission with incomplete blood count recovery (CRi)) was observed until relapse, PD, or death based on the IWG 2003 response criteria and central review. If no relapse, PD, or death was observed, the duration of response was censored at the last response assessment date that the participant was known to be progression-free. Response was assessed following every 3 treatment cycles until treatment discontinuation.
From randomization until relapse, PD, or death (up to approximately 34 months)
Number of Participants With Treatment-emergent Adverse Events (TEAEs)
Time Frame: From first dose to 90 days after last dose of durvalumab or 28 days after last dose of azacitidine proir to the extension study (up to approximately 34 months)
Treatment emergent adverse events are adverse events (AEs) that occurred or worsened on or after the first dose of study drug (durvalumab or azacitidine) and within 90 days after last dose of durvalumab or 28 days after last dose of azacitidine. A treatment-related TEAE is a TEAE where the causal relationship was assessed by the investigator as "Suspected". The intensity of AEs was graded according to the Common Terminology Criteria for Adverse Events (CTCAE) Version 4.03: Grade 1 (Mild): asymptomatic or mild symptoms; clinical or diagnostic observations only; intervention not indicated. Grade 2 (Moderate): minimal, local or noninvasive intervention indicated; limiting age-appropriate activities of daily living. Grade 3: Severe or medically significant but not immediately life-threatening; hospitalization or prolongation of hospitalization indicated; disabling; limiting self care. Grade 4: Life-threatening consequences; urgent intervention indicated. Grade 5: Death due to AE.
From first dose to 90 days after last dose of durvalumab or 28 days after last dose of azacitidine proir to the extension study (up to approximately 34 months)
Kaplan-Meier Estimate of Overall Survival
Time Frame: From randomization to date of death or last known alive date (up to approximately 34 months)
Overall survival is defined as the time between randomization and death/censored date. Participants who were alive at the time of the clinical data cut-off were censored at the last known alive date.
From randomization to date of death or last known alive date (up to approximately 34 months)
One-year Survival
Time Frame: At 12 months after randomization
One-year survival is defined as the probability of survival at 1 year from randomization and is represented by the Kaplan-Meier estimate of the percentage of participants alive after 1 year.
At 12 months after randomization
Change From Baseline in Selected Hematology Parameters I
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Change From Baseline in Selected Hematology Parameters II
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Change From Baseline in Selected Chemistry Parameters I
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Change From Baseline in Selected Chemistry Parameters II
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Change From Baseline in Selected Chemistry Parameters III
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Change From Baseline in Selected Chemistry Parameters IV
Time Frame: Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)
Baseline values are defined as the last assessment of a particular parameter prior to administration of the participants first dose.
Baseline and last lab measurement collected in Cycle 2 (Last measurement could be taken either on Day 1, 8, 15 or 22)

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Sponsor

Investigators

  • Study Director: CL Beach, Pharm D, Celgene Corporation

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

June 3, 2016

Primary Completion (Actual)

December 31, 2018

Study Completion (Actual)

December 27, 2021

Study Registration Dates

First Submitted

May 16, 2016

First Submitted That Met QC Criteria

May 16, 2016

First Posted (Estimate)

May 18, 2016

Study Record Updates

Last Update Posted (Estimate)

February 28, 2023

Last Update Submitted That Met QC Criteria

February 1, 2023

Last Verified

February 1, 2023

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Myelodysplastic Syndromes

Clinical Trials on Azacitidine

3
Subscribe