Bernard Escudier Gustave Roussy, Villejuif FRANCE · 2020. 12. 23. · [TFE3] Transloc. (6;11)...

54
Non Clear Cell RCC Bernard Escudier Gustave Roussy, Villejuif FRANCE

Transcript of Bernard Escudier Gustave Roussy, Villejuif FRANCE · 2020. 12. 23. · [TFE3] Transloc. (6;11)...

  • Non Clear Cell RCC

    Bernard Escudier

    Gustave Roussy, Villejuif

    FRANCE

  • From morphology to molecular profile of RKCs

    RKC, rare kidney cancer.

    Albiges L, et al. J Clin Oncol 2018:JCO2018792531 [Epub ahead of print].

    Papillary Chromophobe TranslocationCollecting

    DuctMedullary Sarcomatoid

    Cytogenetic Alterations

    Type 1

    Gain Chr. 7,17

    Type 2

    Del Chr. 9p

    Del Chr. 1, 2, 6, 10, 13, 17

    Transloc. Xp11.2 [TFE3]Transloc. (6;11) [TFEB]

    Del Chr. 8p, 16p, 1p, 9pGain Chr. 13q

    Del. 22q –

    Molecular Alterations

    Type 1

    METTERTCDKN2A/BEGFR

    Type 2

    SETD2CDKN2A/BNF2FHTERT

    TP53PTENTERT fusionMTOR, TSC1/2MT-ND5

    TFE3 fusionTFEB fusion

    NF2SETD2SMARCB1CDKN2A

    SMARCB1 rearrangements

    TP53CDKN2ANF2RELNBAP1ARID1A

    Pathway Deregulartions

    ActivationCell cycleMAP

    kinases

    DeregulationChromatin

    remodeling

    ActivationCell cycleHippoNRF2-ARE

    DeregulationChromatin

    remodelingMetabolismMethylation

    ActivationMTORAPOBEC

    DeregulationMetabolism

    ActivationTNFTGF-βMOTOR

    DownregulationHIF/VEGF

    DeregulationChromatin remodeling

    ActivationImmune responseCell cycle

    DeregulationMetabolism

    – ActivationCell cycleTGF-β

    DeregulationChromatin

    remodeling

  • Outcome of papillary vs. clear cell RCC

    CSS, cancer-specific survival; cRCC, clear cell RCC; pRCC, papillary RCC; RCC, renal cell carcinoma.

    Steffens S et al. Eur J Cancer. 2012;48:2347–52.

    pRCC ccRCC P

    value

    Lymph

    node mets

    9.0% 7.3% 0.17

    Visceral mets 9.6% 15.2%

  • Outcome of chromophobe vs. clear cell RCC

    • Outcomes between metastatic chrRCC and ccRCC are similar when treated withconventional targeted therapies

    ccRCC, clear-cell renal cell carcinoma; chrRCC, chromophobe renal cell carcinoma; mRCC, metastatic renal cell carcinoma; OS, overall survival; TTF, time-to-treatment failure.

    Yip SM et al. Kidney Cancer. 2017;1:41–7.

    0

    Months

    20 40 8060

    0.25

    0.00

    0.50

    0.75

    1.00

    100 120 140 0

    Months

    20 40 8060

    0.25

    0.00

    0.50

    0.75

    1.00

    100 120

    Overall Survival: Kaplan-Meier Curve (N=4970)

    Metastatic chrRCC OS 23.8 mo (95% CI 16.7–28.1) (N=109)

    Clear cell mRCC OS 22.4 mo (95% CI, 21.4–23.4) (N=4861)

    P=0.0908

    Time–to-Treatment Failure

    Metastatic chrRCC TTF 6.9 mo (95% CI 4.1–8.5) (N=109)

    Clear cell mRCC TTF 7.6 mo (95% CI, 7.2–8.0) (N=4861)

    P=0.53

  • Non clear cell RCC(first line)

    Papillary Chromophobe CDC/medullary

    Standard: Sunitinib [II, B]

    Pazopanib [V, C]

    Option: Everolimus [II, C]

    Cabozantinib[IV, C]

    Option: Sunitinib [II, C]

    Pazopanib, [IV, C]Everolimus [II, C]

    Option: Cisplatin-based regiment [IV, C]Sunitinib [V, C]

    Pazopanib, [V, C]

    Sarcomatoid(predominant)

    Option: Nivolumab +

    Ipilimumab[IV, A]Sunitinib [II, B]

    Pazopanib, [V, C]

  • What is the first-line standard of care?

    Identifier Population Pts (n) Non-clear cell

    histology

    Line Comparator

    Armstrong (2015) ASPEN

    NCT01108445

    Non-clear cell 108 Papillary 76/108 First Everolimus vs

    sunitinib

    Tannir (2014) ESPN

    NCT01185366

    Non-clear cell 68 Papillary 27/68

    Chromophobe 12/68

    Translocational

    Unclassified

    Sarcomatoid

    First

    Second

    Everolimus vs

    sunitinib

    Motzer (2014) RECORD3

    NCT

    00903175

    Clear +

    non-clear cell

    66 Papillary 50/66

    Chromophobe 12/66

    Unclassified

    First

    Second

    Everolimus vs

    sunitinib

    Twardowski

    (2015)

    SWOG 1107

    NCT01688973

    Non-clear cell 50 Papillary 50/50 First

    Second

    Tivantinib vs

    tivantinib +

    erlotinib

    Dutcher (2009) ARCC

    NCT00065468

    Clear +

    non-clear cell

    73 Papillary 55/73

    Unclassified

    First IFN-α vs

    temsirolimus

  • Evidence for of RKCs: The ASPEN trial

    Armstrong AJ et al. Lancet Oncol 2016; 17: 378–88.

    Sunitinib

    (n=51)

    Everolimus

    (n=57)

    Histological subtype

    - Papillary histology overall 33 (65%) 37 (65%)

    - Papillary histology type 1 4 (8%) 2 (4%)

    - Chromophobe 10 (20%) 6 (10%)

    - Unclassified 8 (16%) 14 (25%)

    - Translocation carcinoma 6 (12%) 2 (4%)

    - Minor clear cell

    component

    5 (10%) 8 (14%)

    - Sarcomatoid

    differentiation

    5 (11%) 11 (27%)

    Prior nephrectomy 41 (80%) 45 (79%)

    Elevated lactate

    dehydrogenase

    14 (27%) 13 (25%)

    Liver metastases 16 (31%) 15 (26%)

    Lung metastases 30 (59%) 25 (44%)

    Bone metastases 12 (24%) 15 (26%)

    MSKCC risk group

    0 15 (29%) 14 (25%)

    1-2 32 (63%) 32 (56%)

    ≥3 4 (8%) 11 (19%)

    Events/patients

    Median progression-free survival (months, 80% CI)

    Hazard ratio(80% CI)

    Sunitinib Everolimus

    MSKCC risk group

    Good 21/29 14 (11.5–19.7) 5.7 (5.6–8.4) 2.9 (1.5–5.7)

    Intermediate 51/64 6.5 (5.7–11.0) 4.9 (3.0–5.6) 1.4 (0.9–2.0)

    Poor 15/15 4.0 (0.9–5.8) 6.1 (3.1–7.3) 0.3 (0.1–0.7)

    Histology

    Papillary 60/70 8.1 (5.8–11.1) 5.5 (4.4–5.6) 1.6 (1.1–2.3)

    Chromophobe 12/16 5.5 (3.2–19.7)11.4 (5.7–19.4)

    0.7 (0.3–1.7)

    Unclassified 15/2211.5 (5.3–Not reached)

    5.7 (2.8–7.2) 1.9 (0.8–4.9)

    Baseline LDH

    ≤ULN 62/75 8.4 (5.8–13.0) 5.6 (5.5–6.1) 1.5 (1.1–2.2)

    >ULN 19/26 5.8 (3.2–15.4) 4.1 (2.6–6.0) 1.1 (0.6–2.3)

    Total 87/108 8.3 (5.8–11.4) 5.6 (5.5–6.0) 1.4 (1.0–1.9)-3.0-2.5 2.52.01.51.00.50-2.0-1.5-1.0-0.5

    Favours everolimusFavours sunitinibLog (HR)

    0Time since randomisation (months)

    6 12 18 24 30

    20

    0

    40

    60

    80

    100

    Pro

    gre

    ss

    -fre

    e s

    urv

    ival (%

    )

    Sunitinib

    Everolimus

    HR 1.41 (80% CI, 1.03–1.92)

    P=0.16

    36

    5157

    2621

    178

    104

    83

    42

    11

    No. at riskSunitinib

    Everolimus

    0Time since randomisation (months)

    6 12 18 24 30

    20

    0

    40

    60

    80

    100

    Ov

    era

    ll s

    urv

    ival (%

    )

    Sunitinib

    Everolimus

    HR 1.12 (95% CI, 0.70–2.1)

    P=0.60

    36

    5157

    4044

    3427

    1915

    149

    106

    42

    No. at riskSunitinib

    Everolimus

  • Molecular profile of rare kidney tumours

    pRCC, papillary renal cell carcinoma. Linehan WM, et al. N Engl J Med 2016;374:135–45.

    Methylation and

    metabolic profile

    of pRCC

    Somatic

    mutational

    profile of

    pRCC

  • Mutations within the MET

    receptor

    MET in RKCs

    RKC, rare kidney cancer.

    Adapted from Sánchez-Gastaldo A et al. Cancer Treat Rev 2017;60:77–89;. 2. Linehan WM, et al. N Engl J Med 2016;374:135–45.

    Transcriptional activation of

    HIF target genes: expression

    of VEGF, PDGF, TGFα

    Energy and

    nutrient sensing

    RENAL CELL

    CANCER

    MET

    RECEPTOR

    Cell survival

    AMPK

    FNIP1 FNIP2

    FLCN

    LKB1

    HGF

    FORETINIB

    SAVOLITINIB

    CABOZANTINIB

    AMG1

    02

    PI3K-AKT-

    mTOR

    PATHWAY

    HIFα

    GAB1GAB2

    PP

    PP

    1. 2.

  • Screened ptsPRCC1 per local pathology

    N=41

    MET+ n=4 MET- n=16 MET? n=3

    Eligible and evaluableN=4

    Eligible and evaluableN=16

    Eligible and evaluableN=3

    Centrally confirmed PRCC1and enrolled

    N=23

    MET+ analysis set

    PRCC1 analysis set

    (incl. 1 MET amplified) (incl. 1 MET amplified)

    MET?: Technical failure, mutational analysis could not be done

    CREATE phase II (EORTC study)

    MET- analysis set

    Schoffski et al, AACR Annual Meeting, April 17, 2016, New Orleans

  • Treatment duration (considering METamplification)

    MET+ (mutation in exons 16-19) and/or MET amplification

    MET- (no mutation in exons 16-19), no METamplification

    RECIST PR first documented

    Treatment ongoing

    Treatment stopped due to

    Adverse event

    Progression

    Patient’s decision or

    other reason

    MET amplification (exploratory)

    MET? (technical failure)

  • Maximum shrinkage of target lesions

    MET+ MET- MET?

    MET amplification

  • MET/VEGFR inhibitors in papillary RCC

    • Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma

    • 65 patients with locally-advanced, bilateral multifocal, or metastatic sporadic PRCC or known HPRC

    • Foretinib bisphosphate (GSK1363089A), formerly XL880, is an oral, multikinase inhibitor targeting MET, RON, AXL, TIE-2, and VEGF receptors

    HPRC, hereditary papillary RCC; PR, partial response; PRCC, papillary renal cell carcinoma; RCC, renal cell carcinoma; VEGF, vascular endothelial growth factor.

    Choueiri TK, et al. J Clin Oncol. 2013;31:181–6.

    0

    Time to progressive disease or death (months)

    2 1416 200

    0.6

    1.4

    Pro

    bab

    ilit

    y o

    f p

    ati

    en

    ts s

    urv

    ivin

    g

    wit

    ho

    ut

    dis

    ease p

    rog

    ressio

    n

    Median PFS

    All patients, 9.3 months

    Intermittent dosing, 11.6 months

    Daily dosing, 9.1 months

    304 24

    0.10.20.30.40.5

    0.70.80.91.01.11.21.3

    282622186 8 1012

    100

    Best

    perc

    en

    tag

    e c

    han

    ge

    fro

    m b

    aselin

    e

    Germline mutations

    MET aberration (MET amplification, gain of chromosome 7 or somatic mutation;

    exluding germline mutations)

    No germline mutation; testing for MET aberration profile incomplete

    * Confirmed PR

    80

    60

    40

    20

    0

    -20

    -40

    -60

    -80

  • MET independent

    pRCC

    MET/VEGFR inhibitors in papillary RCC

    Biomarker-Based Phase II Trial of Savolitinib in Patients with Advanced Papillary Renal Cell Cancer

    • 109 Patients with metastatic pRCC

    • Savolitinib (AZD6094, HMPL-504, volitinib) is a highly selective MET tyrosine kinase inhibitor.

    • MET-driven pRCC was defined as any of the following: chromosome 7 copy gain, focal MET or HGF gene amplification, or MET kinase domain mutations

    NE, not evaluable; PD, progressive disease; PR, partial response; pRCC, papillary RCC; RECIST, Response Evaluation Criteria in Solid Tumours; RCC, renal cell carcinoma; SD, stable disease, VEGF, vascular endothelial growth factor.Choueiri TK, et al. J Clin Oncol 2017,35:2993–3001.

    RECIST Response

    No. (%)

    MET Driven(n=44)

    MET Independen

    t (n=46)

    MET Unknown

    (n=19)

    Total(N=109)

    PR 8 (18) 0 (0) 0 (0) 8 (7)

    SD 22 (50) 11 (24) 5 (26) 38 (35)

    PD 11 (25) 28 (61) 9 (47) 48 (44)

    NE 3 (7) 7 (15) 5 (26) 15 (14)

    Tumour

    responses in

    overall

    treatment

    population and

    by MET status

    0

    Time (months)

    8 12 14 160.0

    0.2

    0.4

    0.6

    1.0

    PF

    S p

    rob

    ab

    ilit

    y

    20

    0.8

    10 18

    MET independentMET drivenMET unknown

    642

    Best

    ch

    an

    ge f

    rom

    baselin

    e

    in t

    arg

    et

    lesio

    n s

    ize (

    %) 100

    8060

    4020

    0-20-40-60-80

    -100

    MET driven pRCC

    Best

    ch

    an

    ge f

    rom

    baselin

    e

    in t

    arg

    et

    lesio

    n s

    ize (

    %) 100

    8060

    4020

    -40-60-80

    -100

    0-20

  • Fumarate

    hydratase

    (FH)

    Fumarate

    Fumarate

    Fumarate

    PHD 2

    Fumarate

    HIF-α

    HIF

    -αHIF

    HIF

    HIF

    HIF

    GLUT1

    TGFα/

    EGFR

    VEGF

    HIF prolyl-hydroxylase. Ubiquitin-

    proteasome

    degradation

    pathway

    Erlotinib

    Bevacizumab

    Fumarate hydratase pathway in RKC

    EGFR, epidermal growth factor receptor; GLUT 1, glucose transporter 1; HIF, hypoxia-inducible factor; PHD, HIF prolyl hydroxylase; RKC, rare kidney cancer; TCA cycle , tricarboxylic acid (Krebs) cycle; TGFα, transforming growth factor alpha; VEGF, vascular endothelial growth factor.

    Adapted from Srinivasan R, et al. Clin Cancer Res 2015; 21(1):10–17.

  • 41 patients

    with pRCC

    Fumarate hydratase pathway in RKC

    HLRCC, hereditary leiomyomatosis and renal cell carcinoma; NR, not reached; ORR, overall response rate; PFS, progression-free survival; pRCC, papillary RCC; RECIST, Response Evaluation Criteria in Solid Tumours; RCC, renal cell carcinoma; RKC, rare kidney cancer.

    Modified from Srinivasan R. Presented at Genitourinary Cancers Symposium 2016, San Francisco, USA, 7−9 January 2016.

    Best response

    by RECIST

    HLRCC Sporadic

    pRCC

    Total

    ORR 65% 29% 46%

    Stable disease 35% 62% 95%

    Disease control

    rate

    100% 91% 95%

    Bevacizumab 10 mg/kg

    Q15,

    Erlotinib 150mg/day

    Cohort 1 (n=20)

    HLRCC

    Cohort 2 (n=21)

    sporadic pRCC

    0

    Time since study entry (months)

    5 15 20 250.0

    0.2

    0.4

    0.6

    1.0

    Pro

    po

    rtio

    n f

    ree f

    rom

    pro

    gre

    ssio

    n

    Median PFS 12.8 (95% CI 7.47–26.3)

    35

    0.8

    10 30 0

    Time since study entry (months)

    5 15 20 250.0

    0.2

    0.4

    0.6

    1.0

    Pro

    po

    rtio

    n f

    ree f

    rom

    pro

    gre

    ssio

    n

    35

    0.8

    10 30

    40

    Be

    st

    pe

    rce

    nta

    ge

    ch

    an

    ge

    fro

    m b

    as

    elin

    e

    20

    0

    -20

    -40

    -60

    -80

    -100

    Non-HLRCC HLRCC

    Entire study: 12.8 (95% CI 7.47–26.3)HLRCC: 24.2 mo (95% CI 12.8–NR)Non-HLRCC: 7.4 mo (95% CI 3.73–10.2)

  • PD-L1 expression in rare kidney tumours

    Ch-RCC, chromophobe RCC; PD-L1, programmed death ligand-1; p-RCC, papillary RCC; RCC, renal cell carcinoma; TIMC, tumour-infiltrating mononuclear cells.

    Choueiri TK et al. Ann Oncol 2014;25:2178–84.

    N=101 %

    Histology

    Chromophobe 36 36

    Papillary 50 49

    Translocation 10 10

    Collecting duct

    carcinomas

    5 5

    Metastatic

    disease

    No 78 77.2

    Yes 23 22.8

  • PD-L1 expression in RKCs

    PD-L1, programmed death ligand-1; RKC, rare kidney cancer.

    Choueiri TK et al. Ann Oncol 2014; 25: 2178–2184.

    0Years from diagnosis

    1 2 3 4

    0

    0.2

    0.4

    0.6

    1.0

    Ov

    era

    ll s

    urv

    ival p

    rob

    ab

    ilit

    y

  • KEYNOTE-427: (NCT02853344)

    aPD-L1 positive defined as combined positive score [CPS] ≥1.

    Pembrolizumab

    200 mg Q3W

    Cohort A

    clear cell

    RCC

    (N = 110)

    Cohort B

    nccRCC*

    (N = 165)

    Response

    assessed at

    week 12

    and Q6W until

    week 54, and

    Q12W

    thereafter

    • Endpoints

    • Primary: ORR per RECIST v1.1 (blinded

    independent central review)

    • Secondary: DOR, DCR, PFS, OS,

    safety, and tolerability

    • Exploratory: ORR by histology (blinded

    independent central review) and PD-L1

    expression;a tissue-based biomarkers

    (eg, IHC, RNA sequencing)

    Screen for

    eligibility

    Patients

    • Recurrent or advanced/metastatic disease

    • Measurableper RECIST v1.1

    • No prior systemic therapy

    • Karnofsky performance status ≥70%

    *nccRCC diagnosis

    confirmed by central

    pathology

  • Baseline Characteristics

    Database cutoff: September 7, 2018.

    Characteristic, n

    (%)N = 165

    Age, median (range), 62 (22-86)

    Men 109 (66)

    Karnofsky

    performance scale

    90-100 124 (75)

    70-80 41 (25)

    Characteristic, n (%) N = 165

    Confirmed RCC histology

    Papillary 118 (71)

    Chromophobe 21 (13)

    Unclassified 26 (16)

    IMDC risk category

    Favorable 53 (32)

    Intermediate/poor 112 (68)

    PD-L1 status

    CPS ≥1 102 (62)

    CPS

  • Confirmed ORR by Blinded Independent Central Review

    aIncludes patients who discontinued or died before first postbaseline scan. bIncludes patients with insufficient data for response assessment.

    Database cutoff: September 7, 2018.

    N = 165

    n % 95% CI

    ORR 41 24.8 18.5-32.2

    DCR (CR + PR + SD ≥6 months) 67 40.6 33.0-48.5

    Best overall response

    CR 8 4.8

    PR 33 20.0

    SD 53 32.1

    PD 61 37.0

    No assessmenta 8 4.8

    Not evaluableb 2 1.2

  • Maximum Change From Baseline in Target Lesions by Central Review

    Includes patients who received ≥1 dose of pembrolizumab, had a baseline scan with measurable disease per RECIST v1.1, and had a postbaseline assessment (n = 155).

    *Patient had an increase in target lesions above 100%.

    Database cutoff: September 7, 2018.

    • 91/165 (55.2%) experienced a reduction in tumor burden

    • 20/165 (12.1%) experienced a tumor burden reduction ≥80%

    • 7/165 (4.2%) experienced 100% tumor burden reduction

    +20%

    –30%

    –80%

    Pe

    rce

    nta

    ge

    Ch

    an

    ge

    Fro

    m B

    as

    eli

    ne

    Papillary

    Chromophobe

    Unclassified

    –100

    –80

    –60

    –40

    –20

    0

    20

    40

    60

    80

    100 **

  • Time to Response and Response Duration

    Data are presented for responders.

    Database cutoff: September 7, 2018.

    Time to last scan

    CR

    PR

    PD

    Time to last dose

    Ongoing response*

    Continued response after last dose

    Months

    0 5 10 15 20

  • Time to Response and Response Duration

    Database cutoff: September 7, 2018.

    Time to Response,

    median (range), months

    DOR,median (range),

    months

    Response≥6

    Months, %

    2.8 (0.1-8.3) NR (2.8-15.2+) 81.5

    Months

    Re

    ma

    inin

    g in

    Re

    spo

    nse

    , %

    41 37 22 12 6 3 0

    No. at risk

    0 3 6 9 12 15 18

    0

    20

    40

    60

    80

    100

  • ORR by Confirmed RCC Histology per Blinded Independent Central Review

    aDCR = CR + PR + SD ≥6 months. bIncludes patients who discontinued or died before first postbaseline scan. cIncludes patients with insufficient data for response assessment.

    Database cutoff: September 7, 2018.

    Papillary

    n = 118

    Chromophobe

    n = 21

    Unclassified

    n = 26

    Confirmed ORR, %

    (95%CI)25.4 (17.9-34.3) 9.5 (1.2-30.4) 34.6 (17.2-55.7)

    DCR, % (95%CI)a 43.2 (34.1-52.7) 33.3 (14.6-57.0) 34.6 (17.2-55.7)

    Confirmed BOR, %

    CR

    PR

    SD

    PD

    No assessmentb

    4.2

    21.2

    34.7

    33.9

    5.1

    4.8

    4.8

    47.6

    42.9

    0.0

    7.7

    26.9

    7.7

    46.2

    7.7

    Not evaluablec 0.8 0.0 3.8

  • ORR by PD-L1 Expression

    CPS ≥1

    n = 102

    CPS

  • Progression-Free Survival and Overall Survival in the Total Population

    Database cutoff: September 7, 2018.

    Median OSNR (95% CI, NR)

    OS,

    %

    165 151 140 108 75 42 14 1

    No. At Risk

    0

    Months

    0 3 6 9 12 15 18 21 24

    0

    20

    40

    60

    80

    100

    72%

    Median PFS4.1 months (95% CI, 2.8-5.6 months)

    165 91 62 38 20 9 3 1 0

    No. at risk

    Months

    PFS

    , %

    0 3 6 9 12 15 18 21 24

    0

    20

    40

    60

    80

    100

    23%

  • Nivolumab

    Ipilimumab

    200 pts

    Cohort 1:

    ccRCC

    Cohort 2:

    RKCs

    Phase: IIIb/IV

    Primary endpoint: Safety

    Lenvatinib

    Everolimus

    31 pts with

    RKCs

    Phase: II

    Primary endpoint: ORR

    Bevacizumab

    Atezolizumab

    60 pts with

    RKCs

    Phase: II

    Primary endpoint: ORR

    Nivolumab

    Cabozantinib

    37 pts with

    RKCs

    Phase: II

    Primary endpoint: ORR

    Cabozantinib48 pts with

    RKCs post-

    CPIs

    (or CPI

    unsuitable)Phase: II

    Primary endpoint: ORR

    Nivolumab

    Sunitinib

    306 pts

    with RKCs

    Phase: II

    Primary endpoint: 12-mos

    OS

    Ipilimumab

    Current trials for RKCs

    ccRCC, clear cell RCC; CPI, checkpoint inhibitor; ORR, overall response rate; OS, overall survival; RCC, renal cell carcinoma; RKC, rare kidney cancer1. https://clinicaltrials.gov/ct2/show/NCT02982954; 2. https://clinicaltrials.gov/ct2/show/NCT02915783; 3. https://clinicaltrials.gov/ct2/show/NCT02724878; 4. https://clinicaltrials.gov/ct2/show/NCT03635892; 5. https://clinicaltrials.gov/ct2/show/NCT03685448; 6. https://clinicaltrials.gov/ct2/show/NCT03075423. (All accessed February 2019).

    +

    +

    + +

    +

    https://clinicaltrials.gov/ct2/show/NCT02982954https://clinicaltrials.gov/ct2/show/NCT02915783https://clinicaltrials.gov/ct2/show/NCT03635892https://clinicaltrials.gov/ct2/show/NCT03685448https://clinicaltrials.gov/ct2/show/NCT03075423

  • Exploratory Analysis in Sarcomatoid Patients

    • Minimum follow-up 30 months as of August 6, 2018 database lock

    • Endpoints: OS, PFS (per investigator), ORR (per investigator), best overall response, and safety

    • No central pathology review leading to unknown tumor sample origin (nephrectomy vs biopsy) and percent sarcomatoid dedifferentiation

    IMDC, International Metastatic Renal Cell Carcinoma Database Consortium

    All randomized CheckMate 214 patients (n = 1096)

    Excluded; no available pathology report (n = 254)

    Available local pathology report accompanying tumor sample (n = 842)

    Excluded; “sarcomatoid” not present (n = 592)

    Had “sarcomatoid” keyword (n = 250)

    • Identified as sarcomatoid positive (n = 115)

    ‒ IMDC intermediate/poor risk (n = 111)

    ‒ IMDC favorable risk (n = 4)

    Intermediate/poor risk NIVO+IPI (n = 60)

    • Treated (n = 59) / not treated (n = 1)Intermediate/poor risk SUN (n = 52)

    • Treated (n = 52)

    CheckMate 214

    Excluded; sarcomatoid negative (n = 135)

  • Baseline Characteristics

    Sarcomatoid

    intermediate/poor riskAll intermediate/poor risk1

    NIVO + IPI

    N = 60

    SUN

    N = 52

    NIVO + IPI

    N = 425

    SUN

    N = 422

    Median age, years 58 60 62 61

    Male, % 70 75 74 71

    IMDC prognostic score, %

    Intermediate (1–2)

    Poor (3–6)

    75

    25

    71

    29

    79

    21

    79

    21

    Previous nephrectomy, % 88 81 80 76

    No. of sites with ≥1 lesion, %a

    1

    ≥2

    17

    83

    25

    75

    21

    79

    20

    80

    Sites of metastasis, %a

    Lung

    Lymph node

    Bone

    Liver

    76

    51

    20

    15

    81

    54

    19

    13

    69

    45

    22

    21

    70

    51

    23

    21

    Quantifiable tumor PD-L1 expression, %b

  • Sarcomatoid

    intermediate/poor riskAll intermediate/poor risk1

    NIVO + IPI

    N = 60

    SUN

    N = 52

    NIVO + IPI

    N = 425

    SUN

    N = 422

    Confirmed ORR (95%

    CI), %56.7

    (43.2–69.4)

    19.2 (9.6–32.5)

    41.9 (37.1–46.7)

    29.4 (25.1–34.0)

    P value

  • Best Tumor Reduction Investigator:

    Intermediate/Poor-Risk Sarcomatoid Patients

    * = respondersPatients with target lesion at baseline and at least 1 on-treatment tumor assessment are included. Best reduction is maximum reduction in sum of diameters of target lesions. A negative value

    means true reduction, positive value means increase only observed over time. Dashed reference line indicates the 30% reduction consistent with a RECIST v1.1 response

    Be

    st

    red

    ucti

    on

    fro

    m b

    as

    eli

    ne

    in

    ta

    rge

    t le

    sio

    n (

    %)

    NIVO+IPI SUN

    50

    100

    25

    75

    0

    –25*

    ******

    ****

    **

    ****

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

    –50

    –75

    –100* ** **

    *****

    *

    CheckMate 214

  • PFS per Investigator:

    Intermediate/Poor-Risk Sarcomatoid PatientsP

    rog

    res

    sio

    n-f

    ree

    su

    rviv

    al

    (pro

    ba

    bil

    ity)

    Months

    0.8

    1.0

    0.7

    0.9

    0.6

    0.5

    0.4

    0.3

    0.2

    0.1

    0.0

    0 63 9 12 15 18 21 24 27 30 33 36 39 42

    60 41 35 28 23 23 21 19 19 17 16 10 4 2 0

    52 32 20 11 8 7 6 6 5 4 4 2 1 0 0

    No. at risk

    NIVO+IPI

    SUN

    CheckMate 214

    NIVO + IPI

    N = 60

    SUN

    N = 52

    Events, n (%) 37 (62) 40 (77)

    Median PFS, (95% CI), mo 8.4 (5.2–24.0) 4.9 (4.0–7.0) Hazard ratio (95% CI)

    P value0.61 (0.38‒0.97)

    0.0329

  • OS: Intermediate/Poor-Risk Sarcomatoid

    Patients

    NE, not estimable

    OS probability,

    % (95% CI)

    NIVO + IPI

    N = 60

    SUN

    N = 52

    12 month 80 (67‒88) 56 (41‒68)

    24 month 58 (45‒70) 35 (22‒47)

    30 month 53 (39‒65) 29 (17‒41)

    Ove

    rall

    su

    rviv

    al

    (pro

    ba

    bil

    ity)

    Months

    0 63 9 12 15 18 21 24 27 30 33 36 39 42 45

    60 56 52 49 47 45 43 37 32 30 29 22 10 5 1 0

    52 48 36 32 29 23 22 19 18 17 15 15 9 3 1 0

    No. at risk

    NIVO+IPI

    SUN

    CheckMate 214

    NIVO + IPI

    N = 60

    SUN

    N = 52

    Events, n (%) 31 (52) 39 (75)

    Median OS, (95% CI),

    mo31.2 (23.0‒NE) 13.6 (7.7‒20.9)

    Hazard ratio (95% CI)

    P value0.55 (0.33‒0.90)

    0.0155

    0.8

    1.0

    0.7

    0.9

    0.6

    0.5

    0.4

    0.3

    0.2

    0.1

    0.0

  • OS: PD-L1–Evaluable Sarcomatoid Patients

  • Translocation RCC

    Malouf GG et al. Clinical Cancer Research, 2014

    Activation of CTLA4 and PDL 1

  • Young patient

    • June 2014: right RN + LND for translocation RCC (TFEB) with lung netastases and lymph nodes

    • Sunitinib July 2014 (stopped for liver toxicity)

    • Pazopanib July 2014-Sept 2014

    • Everolimus Sept 2014-Jan 2015

  • 17q gain is common in translocation RCC

    Malouf GG et al. Clinical Cancer Research, 2013

  • Activation of CTLA4 in tumors with 17q gain

    Malouf GG et al. Clinical Cancer Research, 2013

  • Ipilimumab initiated in 4th line

  • CR in abdominal lesion

  • Initial tumor left kidney 01/2015

    Collecting duct tumor

  • Treatment schedule

    1/15

    Nephrectomy Oophorectomy

    4/15

    CT scanBrain MRI

    CT scan

    Cisplatin Gemcitabine

    Bevacizumab

  • Ovarian metastasis: origin of the PDX

    MRI 04/2015

    CT 04/2015

  • Genomic Alterations (oophorectomy)

    BRAF G466A mutation (Foundation Medicine)

    FBXW7 mutation

    SMARCB1 mutation

    High-level MET amplification (Oncoplex)

    IHC: PDL1 5% tumor cells

    Molecular biology

  • Treatment schedule

    1/15

    Nephrectomy Oophorectomy

    4/15 8/15 1/16 2/16

    CT scanBrain MRI

    PET-CTCT scan

    Cisplatin Gemcitabine

    PET-CT

    Bevacizumab

    Trametinib

    Gene

    sequencing

  • PET-CT 01/2016 PET-CT 02/2016

    M1 MEKi

  • PET-CT 01/2016 PET-CT 02/2016

    M1 MEKi

  • PET-CT 01/2016 PET-CT 02/2016

    M1 MEKi

  • PDX treatment with

    trametinib alone

    PDX treatment with

    trametinib+INC280

    0

    100

    200

    300

    400

    500

    600

    4 9 13 16 20 23 27 30 34 37

    TumorVolume(%)

    Days

    Vehicle

    Trametinib

    - 100

    0

    100

    200

    300

    400

    500

    600

    7 11 14 16 21 24 28 32 35

    TumorVolume(%)

    Days

    Vehicle

    INC280

    Trametinib + INC280

  • Treatment schedule

    1/15

    Nephrectomy Oophorectomy

    4/15 8/15 1/16 2/16

    CT scanBrain MRI

    PET-CTCT scan

    Cisplatin Gemcitabine

    PET-CT

    Bevacizumab

    Trametinib

    6/16

    Trametinib +Crizotinib

    10/16

    Gene

    sequencing

    Gene

    sequencing

  • Rare kidney tumors

    • Young lady (25 years)

    • Hematuria

  • What is your diagnosis?

    • Radical nephrectomy: choriocarcinoma

    • HCG 10 times normal

    • Chemotherapy with BEP

    • Relapse one year later: lung surgery and TIP

    • Currently in CR…..

  • Conclusions

    • RKCs are heterogeneous and represents 25% of RCC

    • The outcome of patients with RKC depends on the histotype and stage at diagnosis

    • Sunitinib remains a standard of care

    • The discovery of MET- and FH-driven disease suggests interesting possibilities for tailored therapy in specific patient groups and paved the way for subtype-driven studies

    • IO revealed promising clinical activity in RKCs

    • Some rare tumors might have different treatments