cml: current therapeutic challenges · 2019. 2. 13. · all, acute lymphocytic leukaemia; emr,...

41
Timothy Hughes, SAHMRI, Adelaide, Australia CML: Current Therapeutic Challenges

Upload: others

Post on 27-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Timothy Hughes, SAHMRI, Adelaide, Australia

    CML: Current Therapeutic Challenges

  • Disclosures

    Company Research

    Support

    Honoraria,

    Advisory Board

    Novartis YES YES

    BMS YES YES

    Ariad/Takeda YES YES

    Celgene YES

  • Heterogeneity of response to TKI

    therapy in CML

    6

    Years of TKI therapy

    1 2 3 4 80

    BaselineB

    CR

    -AB

    L%

    IS

    0.01

    0.1

    1.0

    10

    0.001

    10% primary resistance

    10% secondary resistance

    30% plateau response

    STOP

    25%

    25%50% deep mol response

    5-10% CML-related death

  • Heterogeneity of response to TKI

    therapy in CML

    6

    Years of TKI therapy

    1 2 3 4 80

    BaselineB

    CR

    -AB

    L%

    IS

    0.01

    0.1

    1.0

    10

    0.001

  • Heterogeneity of response to TKI

    therapy in CML

    6

    Years of TKI therapy

    1 2 3 4 80

    BaselineB

    CR

    -AB

    L%

    IS

    0.01

    0.1

    1.0

    10

    0.001

    1.Kinase inhibition

    2. Intrinsic biology

    3.Host immunity

  • Hughes TP, et al. Blood. 2014;123:1353-60.

    Lipton JH, et al. Lancet Oncol. 2016;17:612-21.

    TKIImatinib

    (%)

    Nilotinib

    (%)

    Ponatinib

    (%)

    ALL CML patients 34 9 6

    Kinase inhibition intensity

    Probability of EMR failure (>10% BCR-ABL at 3 months) is

    dependent on intensity of kinase inhibition

    ALL, acute lymphocytic leukaemia; EMR, early molecular response.

  • Hughes TP, et al. Blood. 2014;123:1353-60.

    Lipton JH, et al. Lancet Oncol. 2016;17:612-21.

    TKIImatinib

    (%)

    Nilotinib

    (%)

    Ponatinib

    (%)

    ALL CML patients 34 9 6

    Divided by

    SOKAL score:

    High risk 56 14 15

    Intermediate risk 30 8 5

    Low risk 21 7 2

    Kinase inhibition intensity

    ALL, acute lymphocytic leukaemia; EMR, early molecular response.

    Probability of EMR failure (>10% BCR-ABL at 3 months) is

    dependent on intensity of kinase inhibition and risk score

    Hughes TP, et al. Blood. 2014;123:1353-60.

    Lipton JH, et al. Lancet Oncol. 2016;17:612-21.

    TKIImatinib

    (%)Nilotinib

    (%)Ponatinib

    (%)

    ALL CML patients 34 9 6

    Divided by

    SOKAL score:

    High risk 56 14 15

    Intermediate risk 30 8 5

    Low risk 21 7 2

    Kinase inhibition intensity

    ALL, acute lymphocytic leukaemia; EMR, early molecular response.

    Probability of EMR failure (>10% BCR-ABL at 3 months) is

    dependent on intensity of kinase inhibition and risk score

  • For each generation of TKI drugs improved efficacyhas been counterbalanced by increased toxicity

    The result – no overall improvement in survival

  • DASISION: OS and PFS

    Cortes JE, et al. Blood. 2014;124:abstract 152.

    Dasatinib100 mg OD

    (n = 259)

    Imatinib400 mg OD

    (n = 260)

    Hazard ratio

    (95% CI)

    Total number of deaths, n 26 26 –

    CML-related deaths 9 17

    Non-CML-related deaths 17 9

    Estimated 5-year OS, % (95% CI)

    91(87–94)

    90(85–93)

    1.01 (0.58–1.73)

    Estimated 5-year PFS, % (95% CI)

    85(80–89)

    86(80–89)

    1.06 (0.68–1.66)

  • ENESTnd: Survival and CML-Related Deaths

    Nilotinib

    300 mg BID

    (n = 282)

    Imatinib

    400 mg QD

    (n = 283)

    Total deaths, n 18 22

    KM-estimated 5-year OS, % 93.7 91.7

    Hazard ratio vs imatinib (95% CI)0.80 (0.43-1.50) —

    P value vs imatinib .49 —

    CML related deaths6 16

    Non-CML related deaths12 6

    Hazard ratio vs imatinib (95% CI) 0.37 (0.14-0.94) —

    P value vs imatinib .03 —

    1

    0

  • Parameter

    ATE per 100-person yearsP-

    ValueTotal Imatinib Nilotinib Dasatinib Ponatinib

    N 584 281 132 120 51

    Overall 1.1 0.6 1.7 1.8 5.1

  • Possible responses

    • Use more potent (and toxic) TKI where the risk and potential benefit is greatest – a risk-adapted approach

  • Precision medicine approach in CML – bioassay-directed management

    Bioassay-

    based

    risk

    assessment

    Imatinib

    Experimental

    Potent TKIAt

    diagnosis

  • Ph associated fusion

    ABL1 kinase domain

    RUNX1

    BCORL1

    CBFB-MYH11

    GATA2

    IKZF1

    TP53 (R248Q-germline)

    SETD1B

    ASXL1

    IDH1 (R132H/C), IDH2 (R140Q)

    U2AF1 (Q157R)

    KMT2D

    UBE2A

    SETD2

    XPO1 (E571K)

    MECOM fusion

    BCOR

    Novel fusion (not Ph associated)

    PHF6

    MLL fusion

    BCR-ABL1 amplification

    HBS1L-MYB intergenic locus del

    Chr 17p deletion, 17q amplification

    PAX5/CDKN2A whole gene del

    IGH locus deletion

    TCRA/D loci deletion

    IGK locus deletion

    TCRB locus deletion

    TCRG locus deletion or inversion

    Myeloid blast crisis (19) Lymphoid blast crisis (20)

    Clinically relevant variants at transformation

    Mutation Type

    Inversion associated fusion

    Fusion

    Amplification

    Splice site

    Frameshift/Nonsense

    Missense

    Exon deletion

    Whole gene or locus deletion

    Patient number

    Chr 19 amplification

    PAX5-ZCCHC7

    Chr 8 amplification

    57 39 38 60 3 17 23 19 20 18 1 64756

    ● Variant also present at diagnosis

    Variant present at accelerated phase

    but not BC

    **** * *** *** *** **

    * Patients with WES/CNV analysis

    Patients with RNA-Seq analysis+

    + + + + + + + + ++ + + + + + + + ++ + + + + + + +58 54 15 2

    **+ + + +*21* + + +

    44 48 41 52 16 4 10 9 534555 5 42 7 13 1459 22 1211

  • Poor outcome: 27 patients Good outcome: 19 patients

    BCORL1

    CBFB-MYH11 fusion

    RUNX1

    IKZF1

    TP53

    SETD1B

    ASXL1

    IDH1

    EZH2

    KMT2D

    Patient number 14 2210 11 139 23 2416 17752 20 251912 153 4 26 27186 8 211 32 33 34 35 36 37 38 39 40 41 42 43 443128 29 30 4645

    Mutated genes Mutation Type

    Fusion

    Frameshift/Nonsense

    Missense

    Exon deletion

    Whole gene deletion

    Mutated genes at diagnosis of CML. Shown are mutated cancer genes and SETD1B, which was

    a novel recurrently mutated lysine methyltransferase.

  • Possible responses

    • Use more potent (and toxic) TKI where the risk and potential benefit is greatest – a risk-adapted approach

    • Develop better targeted inhibitors to break the linkage between potency and toxicity

  • Asciminib is a potent, specific inhibitor of BCR-ABL1

    with a distinct allosteric mechanism of action

    ATP, adenosine triphosphate. Hughes TP, et al. Blood. 2016;128:abstract 625.

    • Potent BCR-ABL1 inhibition,

    maintained against BCR-ABL1

    mutations that confer resistance

    to TKIs

    • High specificity providing the

    potency without toxicity

    • Potential for combination

    therapy

    TBCR-ABL1

    protein

    Nilotinib

    (ATP site)

    Asciminib

    (myristoyl site)

  • SH2SH2

    Kinase

    SH2

    INACTIVE ACTIVE

    SH3

    Kinase

    SH3

    Myristoylated

    N-terminus

    Regulation of ABL1 enzymatic activity

    Wylie A, et al. Blood. 2014:[abstract 398].

  • SH2SH2

    Kinas

    e

    SH2

    INACTIVE ACTIVE

    SH3

    Kinas

    e

    SH3

    BCR

    t(9;22)

    Regulation of ABL1 enzymatic activity

    Wylie A, et al. Blood. 2014:[abstract

    398].

  • t(9;22)BCR

    SH2SH2

    Kinas

    e

    SH2

    INACTIVE ACTIVE

    SH3

    Kinas

    e

    SH3

    BCRASCIMINIB

    Regulation of ABL1 enzymatic activity

    Wylie A, et al. Blood. 2014:[abstract

    398].

  • Asciminib is quite selective against

    BCR-ABL+

    cells

    IC50, half maximal inhibitory concentration. Wylie AA, et al. Nature. 2017;543:733-7.

    0.0001 0.0004 0.01 0.04 0.1 0.4 1 4 10 40

    Cell proliferation IC50 (µM)

    Asciminib

    GNF-2

    Imatinib

    Bosutinib

    Nilotinib

    Dasatinib

    Ponatinib

    Myristoyl-site

    inhibitors

    ATP-site

    inhibitors

    BCR-ABL1+

    BCR-ABL1−

    Cancer cell line panel

  • Acquired resistance develops to ATP-site and

    allosteric inhibitors

    WT 1 1

    A337V 2 10640

    P465S 1 13620

    V468F 1 10053

    WT 1 1

    T315I >30000 55

    E255K 1267 4

    E255V 268 2

    Y253H 132 5

    F359V 64 167

    Q252H 30 44

    G250H 7 2

    E459K 4 7

    Fold reduction in cell potency (Ba/F3)

    due to presence of BCR-ABL mutations

    Nilotinib ABL001

    Myristoyl-

    site

    ATP-site

    Wylie A, et al. Blood. 2014:[abstract 398].

  • Acquired resistance develops to ATP-site and

    allosteric inhibitors

    WT 1 1

    A337V 2 10640

    P465S 1 13620

    V468F 1 10053

    WT 1 1

    T315I >30000 55

    E255K 1267 4

    E255V 268 2

    Y253H 132 5

    F359V 64 167

    Q252H 30 44

    G250H 7 2

    E459K 4 7

    Fold reduction in cell potency (Ba/F3)

    due to presence of BCR-ABL mutations

    Nilotinib ABL001

    Myristoyl-

    site

    ATP-site

    Wylie A, et al. Blood. 2014:[abstract

    398].

  • Dose Escalation

    Bayesian Logistic Regression

    CML—completed

    ABL001, po, BID

    Dose Expansion

    CML (20 mg, 40 mg)–completed

    T315I mutation (150 mg)–ongoing

    Dose Escalation

    Ph+ ALL/CML-BP

    Combo Dose Escalation

    CML

    ABL001+nilotinib

    MT

    DR

    DE

    Expansion

    Dose Expansion

    Ph+ ALL/CML-BP

    Combo Dose Escalation

    CML

    ABL001+imatinib

    Expansion

    Combo Dose Escalation

    CML

    ABL001+dasatinib

    Expansion

    Dose Escalation

    CML

    ABL001, po, QD

    Dose Expansion

    CMLMT

    DR

    DE

    MT

    DR

    DE

    MT

    DR

    DE

    MT

    DR

    DE

    MT

    DR

    DE

    ABL001X2101: Study DesignA multicenter, phase 1, first-in-human study

    • Primary outcome: estimation of MTD/RDE

    • Secondary outcomes: safety, tolerability, preliminary anti-CML

    activity, pharmacodynamics,

    pharmacokinetic profile

    ALL, acute lymphocytic leukemia; BID, twice daily; BP, blast phase; CML, chronic myeloid leukemia; MTD, maximum tolerated dose;

    Ph+, Philadelphia chromosome–positive; po, peroral; QD, once daily; RDE, recommended dose for expansion.

  • Asciminib, a Specific Allosteric BCR-ABL1 Inhibitor,

    in Patients with Chronic Myeloid Leukemia

    Carrying the T315I Mutation in a Phase 1 Trial

    Delphine Rea, MD, PhD

  • First-in-human phase 1 study design

    Dose Escalation: CML

    asciminib BID completed

    10 – 200 mg BID

    Dose Expansion:

    CML (20 mg and 40 mg BID) completed

    Dose Escalation: CMLasciminib QD

    80 – 200 mg

    Dose Expansion: CML

    Dose Expansion:

    Ph+ ALL/CML-BP

    Dose Escalation: Ph+ ALL/CML-BPasciminib BID

    40 – 280 mg

    MTD

    RDE

    MTD

    RDE

    MTD

    RDE

    MTD

    RDE

    MTD

    RDE

    MTD

    RDE

    Dose Expansion:

    CMLCombo Dose Escalation: CML

    asciminib + NIL 300 mg BID

    20 and 40 mg BID

    Dose Expansion:

    CMLCombo Dose Escalation: CML

    asciminib + IMA 400 mg QD

    40, 60, and 80 mg QD; 40 mg BID

    Dose Expansion:

    CMLCombo Dose Escalation: CML

    asciminib + DAS 100 mg QD

    80 mg QD; 40 mg BID

    Dose Escalation: CML T315I

    asciminib BID and QD completed

    Asciminib BID20 – 200 mg BID

    80 – 200 mg QD

    Dose Expansion:

    T315I mutation (200 mg BID) ongoing MTD

    RDE

    ALL, acute lymphoblastic leukemia; BID, twice daily; BP, blast phase; DAS, dasatinib; IMA, imatinib; MTD, maximum tolerated dose; NIL, nilotinib; QD, once daily; RDE, recommended dose for expansion.

  • Baseline disease characteristicsDemographics and Prior Therapy N = 32Age, median, years (range) 54.0 (29-77)

    Male, n (%) 25 (78.1)

    ECOG performance status, n (%)

    0 26 (81.3)

    1 6 (18.8)

    Number of prior TKIs, n (%)

    1 3 (9.4)

    2 11 (34.4)

    3 11 (34.4)

    ≥ 4 7 (21.9)

    Prior ponatinib treatment, n (%) 19 (59.4)

    Ponatinib resistanta 12 (37.5)

    Ponatinib intolerantb 7 (21.9)

    AP, accelerated phase; CCyR, complete cytogenetic response; CHR, complete hematologic response; MMR, major molecular response (BCR-ABL1 ≤ 0.1% on the International Scale [IS]).a Patients who discontinued due to disease progression, completed prescribed regimen, or showed lack of efficacy at their last ponatinib regimen. b Patients who discontinued due to toxicity or adverse events (AEs) at their last ponatinib regimen. c Insufficient number of metaphases available to evaluate for CCyR. d One patient had atypical BCR-ABL1 transcript.

    Disease Characteristics N = 32Disease phase, n (%)

    CML-CP 30 (93.8)CML-AP 2 (6.3)

    CHR at screening, n (%)No 15 (46.9)Yes 17 (53.1)

    CCyR at screening, n (%)No 22 (68.8)Yes 7 (21.9)Incompletec 3 (9.4)

    MMR at screening, n (%)No 30 (93.8)Yes 1 (3.1)Not assessabled 1 (3.1)

  • Baseline disease characteristicsDemographics and Prior Therapy N = 32Age, median, years (range) 54.0 (29-77)

    Male, n (%) 25 (78.1)

    ECOG performance status, n (%)

    0 26 (81.3)

    1 6 (18.8)

    Number of prior TKIs, n (%)

    1 3 (9.4)

    2 11 (34.4)

    3 11 (34.4)

    ≥ 4 7 (21.9)

    Prior ponatinib treatment, n (%) 19 (59.4)

    Ponatinib resistanta 12 (37.5)

    Ponatinib intolerantb 7 (21.9)

    AP, accelerated phase; CCyR, complete cytogenetic response; CHR, complete hematologic response; MMR, major molecular response (BCR-ABL1 ≤ 0.1% on the International Scale [IS]).a Patients who discontinued due to disease progression, completed prescribed regimen, or showed lack of efficacy at their last ponatinib regimen. b Patients who discontinued due to toxicity or adverse events (AEs) at their last ponatinib regimen. c Insufficient number of metaphases available to evaluate for CCyR. d One patient had atypical BCR-ABL1 transcript.

    Disease Characteristics N = 32Disease phase, n (%)

    CML-CP 30 (93.8)CML-AP 2 (6.3)

    CHR at screening, n (%)No 15 (46.9)Yes 17 (53.1)

    CCyR at screening, n (%)No 22 (68.8)Yes 7 (21.9)Incompletec 3 (9.4)

    MMR at screening, n (%)No 30 (93.8)Yes 1 (3.1)Not assessabled 1 (3.1)

  • Baseline disease characteristicsDemographics and Prior Therapy N = 32Age, median, years (range) 54.0 (29-77)

    Male, n (%) 25 (78.1)

    ECOG performance status, n (%)

    0 26 (81.3)

    1 6 (18.8)

    Number of prior TKIs, n (%)

    1 3 (9.4)

    2 11 (34.4)

    3 11 (34.4)

    ≥ 4 7 (21.9)

    Prior ponatinib treatment, n (%) 19 (59.4)

    Ponatinib resistanta 12 (37.5)

    Ponatinib intolerantb 7 (21.9)

    AP, accelerated phase; CCyR, complete cytogenetic response; CHR, complete hematologic response; MMR, major molecular response (BCR-ABL1 ≤ 0.1% on the International Scale [IS]).a Patients who discontinued due to disease progression, completed prescribed regimen, or showed lack of efficacy at their last ponatinib regimen. b Patients who discontinued due to toxicity or adverse events (AEs) at their last ponatinib regimen. c Insufficient number of metaphases available to evaluate for CCyR. d One patient had atypical BCR-ABL1 transcript.

    Disease Characteristics N = 32Disease phase, n (%)

    CML-CP 30 (93.8)CML-AP 2 (6.3)

    CHR at screening, n (%)No 15 (46.9)Yes 17 (53.1)

    CCyR at screening, n (%)No 22 (68.8)Yes 7 (21.9)Incompletec 3 (9.4)

    MMR at screening, n (%)No 30 (93.8)Yes 1 (3.1)Not assessabled 1 (3.1)

  • Patient disposition

    Patients, n (%) 200 mg BID (N = 32)Treatment ongoing 29 (90.6)

    Treatment ended 3 (9.4)

    Lack of responsea 2 (6.3)

    Adverse event 1 (3.1)

    a One patient had BCR-ABL1IS > 10% and thrombocytosis at screening, and discontinued after continued BCR-ABL1IS > 10% and grade 1 thrombocytosis. The other patient had CML-CP at study entry and persistent thrombocytosis with no progression to AP on study.

    • Analysis is based on a data cutoff date of July 15, 2018• Median duration of exposure: 27.6 weeks (range, 1.0-85.6 weeks)

  • Hematologic and cytogenetic responses

    a Maintenance or achievement of CHR by the data cutoff, based on clinical review of laboratory parameters. b One patient with persistent thrombocytosis was in CHR at screening (platelets 395K) and not in CHR during follow-up due to thrombocytosis (platelets 539K). c Evaluable patients had a cytogenetic evaluation within the specified time window or achieved a CCyR or discontinued treatment before that time window. d Maintenance or achievement of CCyR.

    0

    20

    40

    60

    80

    100

    CHRa CCyR

    CHR at screening(n = 17)

    No CHR at screening(n = 15)

    Pati

    ents

    , % 94.1% (16/17)

    maintained

    CHR

    100%

    (15/15)

    achieved

    CHR

    5.9% (1/17)b

    without CHR

    0

    20

    40

    60

    80

    100

    By 24 weeks By 48 weeksPa

    tien

    ts W

    ith

    Res

    po

    nse

    , %

    75.0%

    (15/20)

    62.5%

    (5/8)

    80.0%

    (16/20)

    66.7%

    (6/9)

    All evaluable patientsc,d

    Evaluable patientsc with > 35% Ph+ at screening

  • Cumulative molecular response rates

    MR4, BCR-ABL1IS ≤ 0.01%; MR4.5, BCR-ABL1IS ≤ 0.0032%. a One patient with atypical BCR-ABL1 transcript and no molecular data was excluded from analyses of cumulative molecular responses; 1 patient with MMR at screening was excluded from analysis of cumulative MMR. b Among patients who achieved MMR.

    Median time to MMRa,b:

    12.2 weeks (range, 4.1-60.0 weeks)

    MMR % (N = 30): 0.0 26.7 33.3 33.3 33.3 36.7 36.7 36.7MR4 % (N = 31): 0.0 6.5 16.1 19.4 19.4 19.4 19.4 19.4

    MR4.5 % (N = 31): 0.0 0.0 12.9 12.9 16.1 16.1 16.1 16.1

    200 mg BID (N = 32)a

    Time (28-day cycles)

    Pati

    ents

    Wit

    h R

    esp

    on

    se (

    %)

    MMR: 36.7%

    MR4: 19.4%

    MR4.5: 16.1%

    MMRMR4MR4.5

    100

    90

    80

    70

    60

    50

    40

    30

    20

    10

    0

    0 3 6 9 12 15 18 21

  • Summary and conclusions

    • Asciminib at 200 mg BID shows promising efficacy and a favorable safety profile in patients with T315I-mutated CML-CP or -AP

    • Clinical benefit is observed in ponatinib-naive and ponatinib-resistant/intolerant patients

    • Asciminib is a promising treatment option in these patients with high unmet medical need

  • Current asciminib trials

    Frontline add-on therapy (phase 2; NCT03578367)1

    • Enrolling patients without DMR on long-term frontline imatinib

    • Patients randomized to:

    o Asciminib 40 mg QD + imatinib 400 mg QD

    o Asciminib 60 mg QD + imatinib 400 mg QD

    o Continue imatinib 400 mg QD

    o Switch to nilotinib 300 mg BID

    • Primary endpoint: MR4.5 rate at 48 weeks

    Third- or later-line therapy (phase 3; NCT03106779)2

    • Enrolling patients previously treated with ≥ 2 ATP binding–site TKIs

    • Patients randomized to asciminib 40 mg BID or bosutinib 500 mg QD

    • Primary endpoint: MMR rate at 24 weeks

  • 65%

    57%

    39%

    15 YRS:

    Adelaide Cohort: Cumulative Incidence of TFR

  • Multivariate Analysis of Factors

    Predicting TFR

    Forest plot of multivariate analysis of factors predicting TFR success at 12 months

    Variable p-value

    Hazard ratio

    [LCL,UCL]Favouring TFR

  • Bioassay-Based Risk Assessment

    AND

    Molecular Dynamics

    ATDiagnosis

    STABLEDMR

    IMATINIB

    Experimental

    CONTINUE

    POTENT TKI

    Intensify/immunomodify

    STOP

    Future of CML Management: Precision Medicine ApproachBioassay Directed Management

  • Current therapeutic challenges in

    CML

    • Rational selection of frontline therapy– based on risk profile, comorbidities and TFR priority

    – Future prospect of risk-adapted therapy based on biomarker algorithm

    • Break the linkage between potency of kinase inhibition and toxicity– Asciminib shows great potential

    • Majority of CML patients eventually eligible for TFR attempt

  • Bordeaux, FranceSEPTEMBER 12-15, 2019

    21st Annual John Goldman Conference on CHRONIC MYELOID LEUKEMIA: BIOLOGY AND THERAPY

    Chairs: J. Cortes, T. P. Hughes, D. S. KrauseOrganizers: R. Bhatia, T. Brümmendorf, M. Copland, M. Deininger, O. Hantschel, F.X. Mahon, D. Perrotti, J. Radich, D. RéaAdvisory Committee: J. Apperley, S. Branford, C. Gambacorti-Passerini, F. Guilhot, R. Hehlmann, P. Laneuville, G. Saglio,

    C. Schiffer, S. Soverini, P. Valent, R. Van Etten

    DEADLINE FOR ABSTRACTS: MAY 8th, 2019

    For further information: www.esh.orgor contact [email protected]

  • Mount Kilimanjaro can be climbed

    CML can be cured