40
Myelodysplastic Syndromes diagnostics prognostics emerging treatment strategies in lower risk disease Arjan A. van de Loosdrecht Department of Hematology VU University Medical Center VU-Institute of Cancer and Immunology (V-ICI) Cancer Center Amsterdam (CCA) Amsterdam, The Netherlands Nederlands Hematologie Congres 2016 Papendal Vrijdag 22-1-2016

Myelodysplastic Syndromes · Add: MDS(del5q): isolated or with one add chrom abnormality, excl monosomy 7 Delete: non-erythroid blast cell count to distinguish pure erythroleukemia

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • Myelodysplastic Syndromes

    diagnostics

    prognostics

    emerging treatment strategies in lower risk disease

    Arjan A. van de Loosdrecht Department of Hematology

    VU University Medical Center

    VU-Institute of Cancer and Immunology (V-ICI)

    Cancer Center Amsterdam (CCA)

    Amsterdam, The Netherlands

    Nederlands Hematologie Congres 2016

    Papendal

    Vrijdag 22-1-2016

  • Steensma DP, et al., Blood 2015;126:9-16; Malcovati L, et al., ASH 2015: pp 299-307 (educational)

    Cytopenia + mutation – dysplasia = WHAT?

  • Incidence of MDS in the Netherlands A

    SR

    / 1

    00,0

    00

    Year of diagnosis

    0,0

    1,0

    2,0

    3,0

    0,3 2,0 7,2

    20,5

    32,1

    0,0

    10,0

    20,0

    30,0

    40,0

  • Relative survival of MDS in the Netherlands

    RSR: relative survival rate

    0%

    20%

    40%

    60%

    80%

    100%

    RS

    R

    Year Of Diagnosis

    1-year RSR

    3-year RSR

    5-year RSR

    0%

    20%

    40%

    60%

    80%

    100%

    0 1 2 3 4 5 6 7 8 9 10

    RS

    R

    Time From Diagnosis (years)

    2001-2005

    2006-2010

    A B

    0%

    20%

    40%

    60%

    80%

    100%

    RS

    R

    Year Of Diagnosis

    1-year RSR

    3-year RSR

    5-year RSR

    0%

    20%

    40%

    60%

    80%

    100%

    0 1 2 3 4 5 6 7 8 9 10

    RS

    R

    Time From Diagnosis (years)

    2001-2005

    2006-2010

    A B

    Relative survival is the observed patient survival corrected for the

    expected survival of comparable group from the general population

    Dinmohamed AG et al., Eur J Cancer 2014;50:1004-12

  • Diagnostic tool Diagnostic value Priority

    Peripheral blood

    smear

    • Evaluation of dysplasia in one or more cell lines

    • Enumeration of blasts Mandatory

    Bone marrow

    aspirate

    • Evaluation of dysplasia in one or more

    myeloid cell lines

    • Enumeration of blasts

    • Enumeration of ring sideroblasts

    Mandatory

    Bone marrow biopsy • Assessment of cellularity, CD34+ cells, and fibrosis Mandatory

    Cytogenetic

    analysis

    • Detection of acquired clonal chromosomal

    abnormalities that can allow a conclusive diagnosis

    and also prognostic assessment

    Mandatory

    FISH

    • Detection of targeted chromosomal abnormalities

    in interphase nuclei following failure of standard G-

    banding

    Recommended

    Flow cytometry

    immunophenotype

    • Detection of abnormalities in erythroid,

    immature myeloid, maturing granulocytes,

    monocytes, immature lymphoid compartments

    Recommended*

    If according to

    ELN guidelines

    SNP-array

    • Detection of chromosomal defects at a high

    resolution in combination with metaphase

    cytogenetics

    Suggested (likely to

    become a

    diagnostic tool in

    the near future)

    Mutation analysis of

    candidate genes

    • Detection of somatic mutations that can allow

    a conclusive diagnosis and also reliable

    prognostic evaluation

    Suggested (likely

    to become a

    diagnostic tool in

    the near future)

    Diagnostic approach to MDS 2016 Dutch/EU guidelines

    Malcovati L, et al., ELN guidelines. Blood 2013;122:2943-64; Greenberg P et al., J Nat Compr Netw

    Canc 2013;11:838-74; *Westers TM, et al., Leukemia 2012;26:1730-41

  • WHO2016:

    reclassifying Myelodysplastic Syndromes

    MDS with single lineage dysplasia (MDS-SLD)

    MDS with multilineage dysplasia (MDS-MLD)

    MDS with single lineage dyplasia and RS (MDS-RS-SLD)

    MDS with multilineage dysplasia and RS (MDS-RS-MLD)

    MDS with excess blasts-1 (MDS-EB1)

    MDS with excess blasts-2 (MDS-EB2)

    Arber DA and Hasserjian RP. Hematology 2015;294-298; educational session

  • WHO2016:

    reclassifying MDS: comments

    Morphology:

    no changes

    – dysplasia cut-off levels remains 10% in all lineages

    – blast cell counts by cytology [no FCM]

    – due to IPSS-R push towards counts of

  • WHO2016:

    reclassifying MDS: comments

    Additional new concepts:

    Add: SF3B1 mutation and RS 50% erythroid precursors

    Add: Familial myeloid neoplasms with germ line mutations

    (TERT, GATA2)

    Arber DA and Hasserjian RP. Hematology 2015;294-298; educational session

  • WHO-2016 role of FCM: iMDSflow recommendations Discussion slides regarding clinical needs

    • Diagnostics:

    – Non-conclusive cases by morphology/cytogenetics

    – Unilineage versus multilineage dysplasia (vs WHO criteria)

    – IDUS/ICUS/CHIP/CCUS

    – MDS-U vs RA vs RARS

    – CMML and monocytic leukemias

    – PB blast count/aberrancies in MDS +/- sF

    – Diagnostic scores i.e. Ogata score vs complex score vs ABC score

    – Integrated diagnostics

    Cremers EMP, et al., Eur J Cancer 2015;54:49-56

    Westers TM, et al., Leukemia 2012;26:1730-41; Porwit A, et al., Leukemia 2014:28:1793-98

  • Validation of the integrated flowcytometric (iFC)

    diagnostic algorithm in a prospective clinical trial (HOVON89)

    Diagnostic score

  • Specificity: 95%

    Sensitivity: 80%

    By FCM dysplasia

    in myeloid / erythroid lineage

    Cremers EMP et al., 2015 (submitted);

    Loosdrecht AA van de, Westers TM. J Nat Compr Cancer Netw 2013;11:892-902;

    Westers TM, et al., Leukemia 2012;26:1730-41; Porwit A, et al., Leukemia 2014:28:1793-98

    Validation of FCM acc. to integrated Flow Score within

    a prospective clinial trial: HOVON 89 (n=174)

  • Integrated Flow Score:

    inconclusive by cytomorphology/Cytogenetics

    Cremers EMP et al., Eur J Cancer 2015;54:49-56

    iFS VUmc

    sensitivity MDS-CM: 61%

    specificity (PaCo): 95%

    MDS in follow-up by CM

    2.6% (3/114) (based on DysM by CM)

    25% (2/8) (reactive/fe-def/drug-induced)

    0%

    10%

    20%

    30%

    40%

    50%

    60%

    70%

    80%

    90%

    100%

    ??

    CM? CG-FC+

    CM ? CG-FC-/+

    CM? CG-FC-

    n= 379 total: cytopenia

    n = 218 diagnosis MDS/non-MDS)

    n = 161 CM/CG inconclusive

    7%

    t=0 FC-analyse

    Ogata score

    + myelomonocytoid

    FU 122/161

    C

    B

    A

  • Prognostic

    variable

    0 0.5 1 1.5 2 3 4

    Cytogenetics Very Good Good Int Poor Very Poor

    BM Blast % ≤2% >2-10

    %

    Hemoglobin

    g/dl; mmol/l

    ≥10

    ≥6.2

    8-

  • Revised International Prognostic Scoring System (IPSS-Revised)

    for MDS: clinical heterogeneity

    Greenberg P, et al., Blood 2012;120:2454-65; Van

    Spronsen M.F., et al., Eur J Cancer 2014;50:3198-3205

  • very good

    n=81

    (2.8%)

    single:

    del(11q)

    -Y

    OS

    60.8 months

    HR

    0.5 (0.3-0.7)

    good

    n=1809

    (65.7%)

    single:

    normal

    del(5q)

    del(12p)

    del(20q)

    two:

    del(5q)

    and others

    OS

    48.6 months

    HR

    1.0 (0.9-1.1)

    intermediate

    n=529

    (19.2%)

    single:

    del(7q)

    +8

    i(17q)

    +19

    other unrel

    clones

    two:

    w/o

    del(5q)/

    del(7q)

    OS

    26 months

    HR

    1.6 (1.4-1.8)

    poor

    n=148

    (5.4%)

    single:

    inv(3)/t(3q)/

    del(3q)

    -7

    two:

    del(7q)

    and others

    complex:

    3

    OS

    15.8 months

    HR

    2.6 (2.1-3.2)

    very poor

    n=187

    (6.8%)

    complex:

    >3

    OS

    5.9 months

    HR

    4.2 (3.4-5.2)

    Cytogenetic Score within the IPSS-R

    Schanz J, et al., J Clin Oncol. 2012 Mar 10;30(8):820-9. Greenberg PL, et al., Blood 2012:120;2454-2465

  • Prognostic classification of rare abnormailties

    into the IPSS-R: n = 7245

    del(3p)

    -13/13q-

    very good

    -X

    -21

    good

    any balanced translocation

    +1/+1q/dup1q intermediate

    der(1;7)

    -9/9q-/9p-

    -18/18q- poor

    +21

    very poor

    Haase D and Schanz J, ASH2015;

    presented with permission for Dutch

    Educational program 2015-16

  • 78

    22

    74

    26

    52

    48

    36

    64

    83

    17

    65

    35

    59

    41

    42

    58

    0

    20

    40

    60

    80

    100

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    Age at diagnosis Age at diagnosis

    MDS CMML

    No cytogenetics performed Cytogenetics performed

    % o

    f p

    atie

    nts

    78

    22

    74

    26

    52

    48

    36

    64

    83

    17

    65

    35

    59

    41

    42

    58

    0

    20

    40

    60

    80

    100

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    Age at diagnosis Age at diagnosis

    MDS CMML

    No cytogenetics performed Cytogenetics performed

    78

    22

    74

    26

    52

    48

    36

    64

    83

    17

    65

    35

    59

    41

    42

    58

    0

    20

    40

    60

    80

    100

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    18-5

    9 ye

    ars

    60-6

    9 ye

    ars

    70-7

    9 ye

    ars

    80 yea

    rs

    Age at diagnosis Age at diagnosis

    MDS CMML

    No cytogenetics performed Cytogenetics performed

    0% 25% 50% 75% 100%

    Very low

    Low

    Intermediate

    High

    Very high

    Percent of patients

    Low Int-1 Int-2 High

    IPSS

    IPS

    S-R

    Prognostication of MDS in daily practice Results from the Dutch PHAROS MDS registry

    Dinmohamed AG, et al., 2016 (submitted)

    Dinmohamed AG et al. Leukemia. 29: 2449-2451 (2015)

  • Alhan C, et al., Leukemia 2015; Oct 27. doi: 10.1038/leu.2015.295. [Epub ahead of print]

    Haferlach T, et al., Leukemia 2014;28:241-7

    Emerging prognostic models in MDS:

    FCM and molecular abnormalities

    IPSS-R low

  • Outcomes After An MDS Diagnosis: kindly provided by DP Steensma (Leuk Lymphoma 2016:57;17-22)

    If the ~32,000/yr MDS patients diagnosed in the U.S. were represented as 100 people…

  • 6 will undergo allogeneic transplant: 2 will be cured, 3 will relapse and die, 1 will die of complications

    7 will die of anemia-related complications (CVA, MI etc)

    27 will progress to AML and die

    26 will die of unrelated causes (e.g., geriatric conditions)

    2 die of iron overload 12 will die of hemorrhage

    20 will die of infection

  • What are the major needs in MDS

    • Optimizing geriatric assessment in (EU/ELN MDS-Right

    HORIZON2020)

    • Optimizing supportive care (infection/bleeding/iron)

    • Development of new targeted therapies in mds

    – ESA refractory lower risk MDS (HOVON/HOVON-associated

    studies)

    • Identification of the poor prognostic lower risk patients (HOVON and

    (EU/ELN MDS-Right HORIZON2020)

    • Understanding mechanisms of resistance to ESA/IMiDs and

    epigenetic therapies

    • Understanding mechanisms of transformation to AML

    • Incorporation of alloTx in MDS

    – Minimizing risk of TRM/relapse post alloTx in MDS

    MDS-F symposium 2015; ASH2015 educational session; oral and poster ASH2015

  • What are the current and emerging new clinical trial

    options in low/high risk MDS?

    Lower risk

    • IMiDs (lenalidomide) (HOVON89)

    • IRAK-1 inhibition (Pacritinib) (HOVON)

    • Toll-like receptor inhibition (OPN-301)

    • Oral Azacitidine (MDS-003) (NL)

    • Luspatercept (NL)

    • Telomerase Inhibition (Imetelstat) (NL)

    Higher risk

    • Genomic annotations: IDH1, IDH2, RAS, FLT3

    • Anti-PD1/PDL1 combinations +/- HMA (NL)

    • Rigosertib (PI3k/PLK1 inhibitor) (under evaluation in NL)

    • (oral) second generation hypomethylating agents

    • Decitabine +/- ibrutinib (HOVON135)

    MDS-F sympoisum 2015; ASH2015 educational session; oral and poster ASH2015

  • Therapeutic options for lower IPSS risk MDS:

    ELN/Dutch guidelines (update 2016 expected)

    Low IPSS risk

    Symptomatic anaemia asymptomatic cytopenia

    Immunosuppressive therapy with ATG

    Watchful-waiting RBC transfusion and ICT

    Age < 60 years, BM blasts

  • KM plot of the time to first post-ESA transfusion for ESA-treated patients that did not have any

    transfusions prior to ESA treatment stratified by whether the patient was defined as a responder or not.

    Log-rank test p=0.0011

    Time to first post-ESA transfusion: Transfusion Naive

    patients; all ESA treated

    EU-MDS registry 2015 (submitted; presented with permission)

  • HOVON89: Epo refr/TD non-del(5q) low risk MDS:

    preliminary conclusions/perspectives

    • HOVON89:

    – closed august 12, 2015

    – Target number of patients n=200 is reached

    – No safety/no efficacy issues in interim analysis (n=100/FU 10

    month)

    – DB closed April 1, 2016:

    • ASH2016 1st clinical data to be presented

    • Add-on studies:

    – Molecular profile follows expected mutations in lower risk MDS

    – Validations of FCM: specificity 95%; sensitivity 85%

    – Association studies between FCM and Mol. Mutations ongoing

    – MDS/Niche studies ongoing

  • A phase II, multicentre, randomised, open-label comparative study evaluating the efficacy

    of LEN with or without EPO in patients with RBC-TD lower-risk MDS without chromosome

    5 abnormalities who are resistant to ESAs (NCT01718379*)/GFM-len-epo-08)

    Key eligibility criteria

    •Patients with de novo MDS or IPSS low- or int-1-risk MDS

    •RBC-TD anaemia

    •Resistance or loss of response to a previous treatment with epoetin alpha/beta

    (60,000 IU/week) or darbepoetin (>250µg/week), administered for ≥12 weeks

    •ECOG performance status ≤2

    N=132

    Randomisation

    Arm A 4 cycles of lenalidomide 10mg p.o.

    once daily d1–21/28-day cycle

    Arm B 4 cycles of lenalidomide

    10mg p.o. once daily on d1–21/28-day

    cycle

    EPO 60,000 IU/week

    Continue treatment until disease

    progression

    Responders

    (IMG 2006 criteria)

    Responders

    (IMG 2006 criteria)

    Toma A, et al., Leukemia 2015; Oct 26 [epub ahead of print]

    Primary endpoint: erythroid response rate after 4 courses (IMG 2006 criteria)

  • HI-E and RBC-TI in patients who received at least 4 cycles (n= 99)

    LEN + EPO

    N = 50

    LEN

    N = 49

    HI-E 52% 30.6% RR = 1.7, p= 0.03

    RBC-TI 32% 18.4% RR = 1.7, p= 0.12

    HI-E and RBC-TI in the ITT population (n=129)

    LEN + EPO

    N = 65

    LEN

    N = 64

    HI-E 40 % 23.4 % RR1.7; p= 0.043

    RBC-TI 24.6 % 14.1 % RR1.7; p= 0.13

    Toma A, et al., Leukemia 2015; Oct 26 [epub ahead of print]

    A phase II, multicentre, randomised, open-label comparative study evaluating the efficacy

    of LEN with or without EPO in patients with RBC-TD lower-risk MDS without chromosome

    5 abnormalities who are resistant to ESAs (NCT01718379*)/GFM-len-epo-08)

  • MDS-005: phase III trial of LEN vs placebo in patients

    with lower-risk RBC-TD MDS without del(5q)

    *≥2 units packed RBCs/28 days in the 112 days prior to randomisation; †≥40,000 units/week

    rhEPO for 8 weeks, equivalent dose of darbopoetin, or serum EPO >500mU/mL; ‡5mg for

    patients with creatinine clearance 40–60mL/min; §≥5 years from randomisation

    ESA = erythropoietin-stimulating agent; QD = daily; SPM = second primary malignancy

    Santini V, et al. Oral presentation at

    ASH 2014. Abstract 409

    Phase III, randomised, multicentre, placebo-controlled study assessing the efficacy and safety of LEN

    in patients with lower-risk RBC-TD MDS without del(5q) who are unresponsive/refractory to ESAs

    Inclusion criteria

    •IPSS low-/

    int-1-risk

    •Non-del(5q)

    •RBC-TD*

    •Unresponsive/

    refractory to

    ESAs†

    Long-term

    follow-up§

    •OS

    •AML progression

    •Subsequent MDS

    treatment

    •SPMs 2:1

    random

    isation

    Day 1

    68 a

    ssessm

    ent

    Discontinue

    treatment

    Continue treatment

    until erythroid relapse

    or disease

    progression

    LEN 10mg

    orally QD‡

    (n=160)

    Placebo

    (n=79)

    RBC-TI ≥8

    weeks or

    erythroid

    response

    No RBC-TI

    ≥8 weeks or

    erythroid

    response

    • Primary endpoint: RBC-TI ≥8 weeks

    • Secondary endpoints: RBC-TI ≥24 weeks, duration of RBC-TI, erythroid response, time to RBC-TI,

    AML progression, safety

  • MDS-005: phase III trial of LEN vs placebo in patients

    with lower-risk RBC-TD non-del(5q) MDS

    1. Santini V, et al. Poster presentation at MDSF 2015. Abstract 035

    2. Santini V, et al. Oral presentation at ASH 2014. Abstract 409

    3. Ebert BL, et al. PLoS Med 2008;5:e35

    Rate of RBC-TI ≥8 weeks was significantly higher with LEN vs placebo in patients

    with lower-risk MDS non-del(5q), with 90% of patients responding within 4 cycles

    • Median duration of RBC-TI ≥8 weeks with LEN, weeks (95% CI): 32.9 (20.7–71.1)

    • The Ebert signature (a collection of genes whose expression levels have previously appeared

    to correlate with response)3 was not predictive of RBC-TI ≥8 weeks in this population

    Time to RBC-TI ≥8 weeks in patients

    responding to LEN (n=41)1,2

    p

  • New studies in 2016 for low/int-1 risk MDS within

    HOVON/SAKK/Belgium and NMDSG

    • Pacritinib in Epo/Lenalidomide refractory lower risk MDS

    • (phase 2 start q2-2016)/HOVON-associated

    • Pacritinib upfront in non-del5q lower risk MDS

    • (HOVON 1xx; in preparation)

    • MDS with RS/SF3B1: ACE536 [Luspatercept](Pharma)

    • Imetelstat (telomerase inhibitor)(Pharma)

    • Azacitidine oral formulation: MDS-AZA-003

    • risk adapted treatment in low-int-1 risk MDS • (HOVON: concept in development)

    • CMML (Ceplene/IL-2); pilot NMDSG • (HOVON associated)

  • IRAK family signalling network

    Rhyasen GW and Starczynoswski DT. BJC 2015;112:232-237

  • Rhyasen GW and Starczynoswski DT. Br J Cancer 2015;112:232-237;

    Rhyasen GW et al., Cancer Cell 2013;24:90-104

    IRAK family signalling network in disease

  • HOVON-associated study: IRAK1 inhibitor in HOVON89

    relapse/refractory patients

    • Small phase-II study (pilot: n=27)

    • Start q1/2 2016 [MetC approval awaiting]

    • Pacritinib: Epo/Lenalidomide refractory patients/TD:

    – HOVON89: off-study patients

    • Primary Endpoints: HI/TI

    • Secondary Endpoints: Safety/tolerability

    – Time-to-HI/TI

  • Aristoteles Giagounidis1, Uwe Platzbecker2, Ulrich Germing3, Katharina Götze4, Philipp Kiewe5, Karin Tina Mayer6, Oliver Ottmann7, Markus Radsak8, Thomas Wolff9,

    Detlef Haase10, Monty Hankin11, Dawn Wilson11, Xiaosha Zhang11,

    Adberrahmane Laadem12, Matthew L. Sherman11, Kenneth M. Attie11

    1Marien Hospital Düsseldorf, 2Universitätsklinikum Carl Gustav Carus, Dresden, 3Universitätsklinikum Düsseldorf, 4Technical University of Munich, 5Onkologischer Schwerpunkt am Oskar-Helene-Heim, Berlin, 6University Hospital

    Bonn, 7Universitätsklinikum Frankfurt, Goethe Universitaet, Frankfurt/Main, 8Johannes Gutenberg-Universität,

    Mainz, 9OncoResearch Lerchenfeld UG, Hamburg, 10Universitätsmedizin Göttingen, Germany; 11Acceleron

    Pharma, Cambridge, MA, 12Celgene Corporation, Summit, NJ, USA

    Oral presentation:

    Dec 5, 2015

    12:15 PM

    W331, Level 3

    Orange County Convention Center

    Giagoundis A, et al. Luspatercept Treatment Leads to Long Term Increases in Hemoglobin and Reductions in Transfusion Burden in Patients with Low or

    Intermediate-1 Risk MDS: Preliminary Results from the Phase 2 PACE-MDS Extension Study. Oral presented at: Annual Meeting and Exposition of the

    American Society of Hematology 2015; December 5‒8; Orlando, FL. Abstract 92.

    Luspatercept Treatment Leads to Long Term

    Increases in Hemoglobin and Reductions in

    Transfusion Burden in Patients with Low or

    Intermediate-1 Risk MDS: Preliminary Results

    from the Phase 2 PACE-MDS Extension Study

  • Luspatercept in MDS: Results

    • Mean (SE) change in Hb for LTB patients

    – 9 of 13 (69%) LTB patients achieved IWG HI-E response for mean Hb increase

    Giagoundis A, et al. Luspatercept Treatment Leads to Long Term Increases in Hemoglobin and Reductions in Transfusion Burden in Patients with Low or

    Intermediate-1 Risk MDS: Preliminary Results from the Phase 2 PACE-MDS Extension Study. Oral presented at: Annual Meeting and Exposition of the

    American Society of Hematology 2015; December 5‒8; Orlando, FL. Abstract 92.

    • 13 of 19 (68%) HTB patients achieved IWG HI-E response for transfusions

    • 8 of 19 (42%) HTB patients achieved RBC-TI

    – 3 of 3 LTB patients with 2U RBC/8 weeks achieved RBC-TI SE, standard error.

  • Luspatercept: Results

    • Most patients in the extension study were RS+

    • ≥ 50% patients responding to luspatercept had prior ESA

    treatment or baseline EPO of ≤ 500 U/L

    Giagoundis A, et al. Luspatercept Treatment Leads to Long Term Increases in Hemoglobin and Reductions in Transfusion Burden in Patients with Low or

    Intermediate-1 Risk MDS: Preliminary Results from the Phase 2 PACE-MDS Extension Study. Oral presented at: Annual Meeting and Exposition of the

    American Society of Hematology 2015; December 5‒8; Orlando, FL. Abstract 92.

    Baseline feature, n (%) IWG HI-E

    N = 32

    RBC-TI ≥ 8 Weeksa

    N = 22

    All Patients 22 of 32 (69) 11 of 22 (50)

    RS positive 21 of 29 (72) 10 of 19 (53)

    Baseline EPO

    < 200 U/L 16 of 20 (80) 7 of 13 (54)

    200–500 U/L 5 of 7 (70) 2 of 4 (50)

    > 500 U/L 1 of 5 (20) 2 of 5 (40)

    Prior ESA Treatment

    Yes 12 of 19 (63) 7 of 14 (50)

    No 10 of 13 (77) 4 of 8 (50)

    EPO, erythropoetin; ESA, erythropoetin stimulating agent; RS, ring sideroblast.

    a Includes 19 HTB patients and 3 LTB patients evaluable for RBC-TI (≥ 2 units over 8 weeks).

  • CONFIDENTIAL 37

    Rationale for Targeting Telomerase in MDS

    • MDS patients have higher telomerase activity (TA), higher expression level of

    hTERT (Telomerase reverse transcriptase) and shorter telomere length (TL)

    compared to age-match normal control

    • Higher TA & hTERT, shorter TL correlated to IPSS risk score in MDS

    • Shortened telomeres in MDS have been associated with the disease progression

    and conversion to AML

    • High TA & short TL represent poor prognostic features in lower risk MDS

    • MDS pts with high TA have shorter survival

    • Inhibition of TA should hasten apoptosis of malignant clone

    • Preliminary clinical results of imetelstat in int-1 risk MDS showed clinical benefit

    (transfusion independence)

  • A Phase 2/3 Study to Evaluate the Activity of Imetelstat in TD Low

    or Intermediate-1 Risk MDS who have Failed ESA Treatment

    MDS

    Transfusion

    Dependent

    Failed

    ESA

    • IPSS Risk: • Low

    • Intermediate-1

    • RBC 4 units/8w (12w pre-study)

    • Pretransfusion Hb ≤9.0 g/dL

    • Epo 40K qw* x8w without Hb inc ≥1.5 g/dL or RBC dec ≥4 units/8w

    • Relapsed following response

    • Poor candidate: sEPO>500mU/mL

    38

    *or equivalent agent/dose CONFIDENTIAL

  • Imetelstat (n~115) R

    A

    N

    D

    O

    M

    I

    Z

    E

    2:1

    Best Supportive Care (n~55)

    RBC transfusions,

    hematopoietic growth factors

    Stratification factors:

    • IPSS risk (low / intermediate-1)

    • Prior lenalidomide (yes / no)

    39

    Imetelstat

    7.5 mg/kg IV q4w x2 cycles,

    escalate to 9.4 mg/kg IV q4w

    based on tolerability

    Study Design: Phase 2/3, Open Label, N~200

    Phase 2, run-in single arm n up to 30

    Phase 3 randomized 2:1, controlled

    n~170

    CONFIDENTIAL

  • Conclusions: MDS in 2016

    • WHO2016 is coming soon

    – SF3B1 is incorporated in defining MDS-RS if RS