115
Réunion Annuelle Bourgogne / Franche-Comté « Les bonnes pratiques de prise en charge des Sarcomes des Tissus Mous » JY Blay

Réunion Annuelle Bourgogne / Franche-Comté « Les bonnes ... · navig_home_logo. logo. Histological discordances. Healthcare system should ensure that accurate pathology is critical

Embed Size (px)

Citation preview

Réunion Annuelle Bourgogne / Franche-Comté« Les bonnes pratiques de prise en charge des Sarcomes

des Tissus Mous »

JY Blay

Avec tous les laboratoires pharmaceutiques et autres fabricant des médicaments destinés aux sarcomes (et aux autres cancers).

Recherche, honoraires, voyages professionnels

Diagnostic

0 0.2 0.4 0.6 0.8 1 1.2 1.4

GISTLiposarcomasSarcoma NOS

Leiomyos non ut.Kaposi

DermatofibroSUterine LMS

MFH

MyxofibrosarcomasRhabdomyosarcomas

Synovialosarcomas

Ewing sarcomasAngiosarcomas

LG Fibromyxoid Sarcomas

MPNST

FibrosarcomasSolitary fibrous tumors

OsteoS

Incidence / 100,000 / yr

Others

Over 80 histotypes of sarcomas…

Besancon 18 1 18

GIST

Sarcomas and

aggressive connective

tissue tumors

Mutations

kinases

G5

Ntl

Group

Translocations

DFSP

SyS

Ewing

Mutations

APC/bCat

Desmoids

WD/DDLPSAmplification

12q13-15

MDM2/CDK4

MPNST

PEComas

TSG loss

NF1, TSC1/2

Genomique

complexe

LMS, UPS

• Specific translocations generating fusion genes (Myx LPS) 15%

• Kinase mutations (KIT…) ?

• Gene inactivation (NF1…) ?

• Amplifications chromosome 12 (MDM2+CDK4) 75-80%

• Complex genetic alterations (Pleo LPS, ...) 5-10%

Ducimetière F et al. PLoS One 2011; 6: e20294.

For 35 % the diagnosis is not totally correct

Rate of concordance by patient sub-group

188

Second opinion requested

53 (28%)

Total

discordance

53 (28%)

Partial

concordance

82 (44%)

Total

concordance

178

Second opinion not requested

17 (10%)

Total

discordance

44 (25%)

Partial

concordance

117 (65%)

Total

concordance

For 56 % the diagnosis is not totally correct

Accuracy of histological diagnosis

Ducimetière F, et al. PLoS One. 2011;6(8):e20294.

Cassier PA, etal. Br J Cancer. 2010 Jul 13;103(2):165-70.

Lurkin A, et al. BMC Cancer. 2010 Apr 19;10:150.

Histological discordances

Healthcare system should ensure that accurate pathology is critical to good care. Histopathologists should be members of a quality assurance scheme which allows second opinions to be routine practice.

Histological reviews registered in 2010: 14% of major discordances (341 cases)

Cost of the treatments assessed for the initial diagnosis: 2 186 816 € vs final diagnosis: 1 060 174 €

Histological reviews/molecular biology result in a cost saving of more than €1,000,000

Lionel Perrier, ISPOR 19th, Canada, June 2014

Prise en charge en phase localisée

• Biopsy first

– Assessment by an experienced team

• En bloc surgical resection

– Planning R0

– If R1, consider re resection

• Post operative radiotherapy

– (G2-3 and/or deep seated, and/or >5cm)

• Preoperative radiotherapy

SMAC 1997, Lancet 350:1647-54

1568 patients in 14 randomized trials comparing chemotherapy vs 0

Ove

rall

su

rviv

al Chemo

No chemo

Meta-analysis of the clinical trials evaluating

adjuvant chemotherapy vs no treatment in STS

ISG adjuvant trial

DFS OS

Frustaci et al. JCO 2001; 19:1238Frustaci 2001

EORTC 62931

(years)

0 2 4 6 8 10 12

0

10

20

30

40

50

60

70

80

90

100

O N Number of patients at risk : TRT1

86 176 108 78 54 27 5

82 175 115 69 38 19 4

Observation

Adjuvant

Overall Logrank test: p=0.497

5 year RFS

Observation 53% [95% CI 46, 61]

Chemotherapy 51% [95% CI 43, 59]

No difference in local

or distant RFS

Meta-analysis of the 2 randomized EORTC adjuvant trialsN=819 patients

• Which are the predictive factors for benefit of adjuvant treatment (OS/PFS)?

– Young (<40) age : NO

– Synovials : NO

– Uterine LMS: NO

– R1: YES

• Patients benefiting for adjuvant treatment:

– PFS: >40, males, R1

– OS: R1

Gortzak E, et al. A randomised phase II study on neo-adjuvant chemotherapy for 'high-risk' adult soft-tissue sarcoma.

Eur J Cancer. 2001 Jun;37(9):1096-103.

• 134 evaluable patients

• Randomisation

– 3 courses of CT + surgery

– Surgery alone

• At a median follow-up of 7.3 years:

– 5 year disease-free survival is 52% for the no chemotherapy and 56% for the chemotherapy arm (P=0.3548).

– 5 year overall survival for both arms is64 and 65%, respectively (standard error 7%) (P=0.2204).

Regional Hyperthermia (RHT) Technology

Courtesy of BSD Medical Corporation model BSD 2000 3D

ESHO quality assurance guidelines for regional hyperthermia Lagendijk et al. Int J Hyperthermia 1998

R

A

D

I

A

T

I

O

N

Arm A

Arm B

EIA

EIA

RHT

R

R

A

D

I

A

T

I

O

N

Arm A

Arm B

EIA

EIA

RHT

R

EORTC 62961

Risk

groups

Stratification: Center, Risk Group, Extremity, Non-Extremity

S

U

R

G

E

R

Y

S

U

R

G

E

R

Y

S1 = primary tumor ≥5 cm, GII/GIII

S2 = local recurrence of S1 tumor

S3 = inadequate surgery of S1 or S2 tumor

Cox hazard ratio=0.65

CI95=0.49-0.87, p=0.003

Impact of clinical practice guidelines (CPG’s) on survival for

sarcoma patients in first line treatment in Rhone-Alpes region

Olfa DERBEL1, Pierre Etienne HEUDEL1, Claire CROPET1, Gualter VAZ3, Pierre MEEUS1, Olivier COLLARD2, Guy DE LAROCHE2, Philippe THIESSE1, Anne-Valérie DECOUVELAERE1, Dominique

RANCHERE-VINCE1, Pierre BIRON1, François Noel GILLY7, Peter PAUWELS4, Fadila FARSI5, Dominic CELLIER6, Jean-Yves BLAY1, Isabelle RAY-COQUARD1

1Centre Léon Bérard, Lyon, France 2Institut de Cancérologie de la Loire, Saint-Priest en Jarez, France3Hôpital Edouard Herriot, Lyon, France4Cancéropôle Lyon Auvergne Rhône-Alpes, France5Réseau Espace Santé Cancer, Rhône-Alpes, France6Merck Serono, Lyon, France7Université Claude Bernard, Lyon, France

Adherence to CPG and pt outcome

Surgical conformity and PFS for STS patients

Median PFS

Conformed surgery: NR

Non conformed surgery: 45.2

HR: conformed vs non conformed: 0.44

IC 95%: [0.32, 059]

p ≤0.0001

0,0

0,1

0,2

0,3

0,4

0,5

0,6

0,7

0,8

0,9

1,0

0 6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96 102

Months

Adherent to CPG’s

Not adherent to CPG’s

0,0

0,1

0,2

0,3

0,4

0,5

0,6

0,7

0,8

0,9

1,0

0 6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96102

Pro

bab

ilit

y o

f su

rviv

al

Months

University /conformsurgery

Private +general S-

p=0.006

3 networks of reference for sarcomas

Label by INCa

Review

diagnosis

Mandatory

Review of

discordant

cases

Evaluation :

-activity,

-discrepancies

-delays,

-technics used,

-Frozen/FFPE

Database shared via Internet

Structuration

of MDTB

sarcoma in

France

Organisation

of third level

MDTB

Activity of MDTB :

-new patients / Fup Patients

-Description traitements

local phase

advanced phase

-inclusions trials,

Réseau de Référence en

Pathology des Sarcomes

Tissus mous / viscères

RRePS

Pr Jean-Michel COINDRE

BERGONIE

NetSarc

Pr Jean-Yves BLAY

CENTRE LEON BERARD

Réseau de Référence

Clinical des Sarcomes

Tissus mous / viscères

ResOsPr François GOUIN

CHU Nantes

Pr Gonzague DE PINIEUX

CHU Tours

Réseau de référence

Pathology/clinical

Sarcomes osseux

Besancon 18 1 18

Jean-Yves Blay, Axel Le Cesne, Nicolas Penel, Emmanuelle Bompas, Florence Duffaud, Christine Chevreau, Maria Rios, Pierre Kerbrat, Didier Cupissol, Philippe Anract, Jean-Emmanuel Kurtz, Celeste Lebbe, Nicolas Isambert, Francois Bertucci, Antoine Thyss, Sophie Piperno-Neumann, Pascale Dubray-

Longeras, Francoise Ducimetiere, Jean-Michel Coindre, Antoine Italiano;

Universite Claude Bernard & Centre Léon Bérard, Lyon, France; Gustave Roussy Cancer Campus, Villejuif, France; Centre Oscar Lambret, Lille, France; Department of Medical Oncology, Centre René Gauducheau, Nantes St. Herblain, France; La Timone University Hospital, Marseilles, France; Institut Claudius Regaud, Toulouse, France; Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France; Medical Oncology Eugene Marquis Comprehensive Cancer Center, Rennes,

France; Centre Val d'Aurelle, Montpellier, France; Hopital Cochin Saint Vincent de Paul, Paris, France; Hôpitaux Universitaires de Strasbourg, Strasbourg, France; Dermatology Department, Saint Louis Hospital, Paris, France; Centre Georges François Leclerc, Dijon, France; Institut Paoli Calmettes, Marseille,

France; Centre Antoine-Lacassagne, Nice, France; Institut Curie, Paris, France; Centre Jean Perrin/ERTICa EA 4677, Clermont-Ferrand, France; Centre Léon Bérard, Lyon, France; Institut Bergonié, Department of Pathology, Bordeaux, France; Institut Bergonié, Department of Medical Oncology, Bordeaux, France

The nationwide cohort of 26883 patients with sarcomas & connective tissue tumors treated in NETSARC reference network

between 2010 and 2015 in France: major impact of multidisciplinary board presentation prior to first treatment

The question adressed by this study

• Does presentation of the patient to a NetSARC MDT prior to treatment impact on management and prognosis?

Suspecteddiagnosis Diagnosis

NETSARCMDTB

NETSARCMDTB

NETSARCMDTB

Treatment

NETSARCCenters

NETSARCCenters

NETSARCCenters

Non NETSARCCenters

Non NETSARCCenters

Non NETSARCCenters

ResultsMDT before treatment

• Overall 37% were presented to a Netsarcmultidisciplinary board (NMTB) prior to initial treatment

• Between 2010 and 2015, the proportion of pts reviewed in Netsarc MDT prior to surgery increased from 30,3% to 41,6% .

Results (2)Worse clinical presentation when MDT

before treatment

• Metastases at diagnosis 16% vs 10% p<0.0001

• Larger tumors: median size 104 vs 91mm p<0.0001

• More deep seated: 85,7% vs 76,3% p<0.0001

• Higher grade: G1 15,8% vs 12,0%, p<0.0001

Also: younger age, more male patients, less visceral sarcomas

Results (3)Better management when MDT before

treatment

• A higher number of pts presented in NetsarcMDTB had

– Adequate imaging of the tumor before treatment/ surgery (87,9% vs 67,8%, p<0.0001)

– Biopsy prior the first resection (87,% vs 55,0%, p<0.0001).

Results (4)Better adhesion to CPGs when MDT before treatment

• Primary surgery performed before vs after presentation to a Netsarc MDT:

R0, R1, R2, and R (unk or NE) surgery in:– 53.0%, 26.8%, 9.1%, 11.0% (MDT before) vs

– 34,2%, 32.7%, 17,6%, 15.5% (MDT after)(p<0.0001).

• 1125 (15.3%) pts had secondary resection after primary surgery performed without previous NetSarc MDT vs 99 (5.5%) in NetSARCcenters (p<0.0001).

• Final surgery:R0, R1, R2, and R (unk or NE) surgery in:

– 57.9%, 25.8%, 6.5%, 9.8% (MDT before) vs

– 48,8%, 26,9%, 10,6%, 13.7% (MDT after) (p<0.0001).

Median follow-up 26 months

Logrank, p<0.0001 Logrank, p=0.15

Results (5)Better LRFS when MDT before treatment

LRFS MFS

Besancon 18 1 18

Jean-Yves Blay, Eberhard Stoeckle, Antoine Italiano , R Rochwerger, Florence Duffaud,

Sylvie Bonvalot, Charles Honoré, Guy Decanter, Carlos Maynou, Philippe Anract, Gwenael Ferron,

Francois Guillemin, Francois Gouin, Maria Rios, Antonio Di Marco1, D. Cupissol, Pierre Meeus,

Jean-Michel Coindre, Isabelle Ray-Coquard1, Nicolas Penel, Axel Le Cesne

Universite Claude Bernard & Centre Léon Bérard, Lyon, France; Gustave Roussy Cancer Campus, Villejuif, France; Centre Oscar Lambret, Lille, France; Department of Medical Oncology, Centre René Gauducheau, Nantes St. Herblain, France; La Timone University Hospital, Marseilles, France; Institut Claudius Regaud, Toulouse, France; Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France; Medical Oncology Eugene Marquis Comprehensive Cancer Center, Rennes,

France; Centre Val d'Aurelle, Montpellier, France; Hopital Cochin Saint Vincent de Paul, Paris, France; Hôpitaux Universitaires de Strasbourg, Strasbourg, France; Dermatology Department, Saint Louis Hospital, Paris, France; Centre Georges François Leclerc, Dijon, France; Institut Paoli Calmettes, Marseille,

France; Centre Antoine-Lacassagne, Nice, France; Institut Curie, Paris, France; Centre Jean Perrin/ERTICa EA 4677, Clermont-Ferrand, France; Centre Léon Bérard, Lyon, France; Institut Bergonié, Department of Pathology, Bordeaux, France; Institut Bergonié, Department of Medical Oncology, Bordeaux, France

Better overall and progression free survival after surgery in expert sites for sarcoma patients: a nationwide study of FSG-

GETO/NETSARC

Median follow-up 30 months

Results (5)LRFS but not MFS nor OS is better in Netsarc centers

Logrank, p=0.43

LRFS

Logrank, p<0.0001

OSRFS OS

Logrank, p<0.0001 Logrank, p=0.49

Present question (ESMO 2017)• Does primary surgery the patient within a NetSARC

center impacts survival?

Suspecteddiagnosis

DiagnosisNETSARC

MDTBNETSARC

MDTBNETSARC

MDTBTreatment

NETSARCCenters

NETSARCCenters

NETSARCCenters

Non NETSARCCenters

Non NETSARCCenters

Non NETSARCCenters

Multivariate analysis for LRFS

Parameter HR p value

Grade 3 1,761 ,000

Grade 2 1,330 ,000

Size 1,002 ,000

Surgery in NetSARC center ,669 ,000

Gender ,878 0,01

Depth 0,881 ,07

Multivariate analysis for RFS

Parameter HR p value

Grade 3 2,336 ,000

Grade 2 1,426 ,000

Size 1,002 ,000

Internal trunk 1,125 0,006

Grade 1 0,751 ,000

Surgery in NetSARC center 0,622 ,000

Multivariate analysis for OS

Parameter HR p value

Grade 3 1,768 ,000

Depth 1,605 ,000

Size 1,003 ,000

Surgery in NetSARC center ,622 ,000

Internal trunk 1,246 0,01

Gender ,863 0,06

Grade 1 0,329 ,000

Retroperitoneal sarcomas

Traitements ciblés, traitements systemiques

Systemic treatment of sarcomas 2000-2018

2000

– All sarcomas• Doxorubicin• Ifosfamide• DTIC

– Subtypes• Dactinomycin• CDDP• Vinca-alcaloids• Cyclophosphamide• HDMTX

2018

– Trabectedin

– Other than LPS : Pazopanib

– GIST : Imatinib, sunitinib

– Osteosarcomas : MTPPE

– LPS : Dox, ET743 (MRCL++)

– LMS : Dox, ET743, Gem, GemTax

– EWS : A,I,C,V,Ac, TopoI inh.

– A/E RMS : Topo inh

– ESS : Aromatase inh.

– PEComa : mTOR

– Angio : Paclitaxel, GemTax

– DFSP : Imatinib

– PVNS : Imatinib, MCSFR Ab or TKI

– TD/FA : HT, imatinib, sorafenib

This slide includes the information about the drug that not approved in Japan.

High doses when response or PFS is the most important enpoint (eg presurgery)

Phase Ib/II of doxorubicin ± olaratumab in advanced STS 1st line

Primary endpoint: PFS

Secondary endpoints: ORR, OS

Olaratumab : IgG1 anti-PDGFR-α (> 80 % STS)

synergistic with doxorubicin in vitro

Phase II

STS adults,

PS ≤ 2

(n = 130)

Doxorubicin

75 mg/m2 d1

(n = 67)

Optional :

olaratumab

until progression

or intolerable

Olaratumab

until progression

or intolerable

1:1

Olaratumab

15 mg/kg D1& D8

+ doxorubicin

75 mg/m2 D1

(n = 66)

R8 cycles (J1 = J21)

Phase Ib

Dose

escalation

(n = 15)

After failure of anthracyclins

Ifosfamide

Trabectedine

Pazopanib

Eribuline

Gemcitabine and Docetaxel

Dacarbazine and Gem

HR (95% CI) = 0.55 (0.436,0.696)

p<0.0001

PFS events: 329 (63.5% of 518 patients)

mPFS Trabectedin: 4.2 months

mPFS Dacarbazine: 1.5 months

Unstratified analysis

A Randomized Phase 3 Study of Trabectedin or Dacarbazine for the Treatment of Patients With Advanced Liposarcoma (LPS)

or Leiomyosarcoma (LMS)

CI, confidence interval

• The primary endpoint of OS was met, indicating a 30% improvement (based on HR)

Eribulin 228 197 162 138 120 97 88 64 45 34 25 14 7 1

1 0

Dacarbazine 224 190 158 130 103 81 64 45 32 24 16 8 3 0

0 0

Patients at Risk:

1.0

0.8

0.6

0.4

0.2

Surv

ival P

robabili

ty

0 3 6 9 12 15 21 24 27 30 33 36 39 42 4518

Survival Time (months)

Eribulin

Dacarbazine

Eribulin Dacarbazine

Median(months)

13.5 11.5

HR (95% CI) 0.768 (0.618, 0.954)

Stratified P-value

0.0169

Randomized, open-label, multicenter, phase 3 study of eribulin versus dacarbazine in patients (pts) with

leiomyosarcoma (LMS) and adipocytic sarcoma (ADI)

Secondary endpoint: PFS

Eribulin 228 79 41 27 16 9 5 2 1 0Dacarbazine 224 63 27 14 6 4 2 1 1 0

Patients at Risk:

1.0

0.8

0.6

0.4

0.2

Surv

ival

Pro

bab

ility

0 3 6 9 12 15 21 24 2718

Survival Time (months)

EribulinDacarbazine

0.0

Eribulin Dacarbazine

Median (months) 2.6 2.6

HR (95% CI) 0.877 (0.710, 1.085)

Stratified P-value 0.2287

Preplanned OS subgroups analysis

*Region 1: USA, Canada; Region 2: Western Europe, Australasia, Israel; Region 3: Eastern Europe, Latin America, Asia.

Overall 176/228 181/224 0.768 (0.618, 0.954) 13.5 11.5

Age group (years)

<65

≥65

138/178

38/50

148/178

33/46

0.728 (0.569, 0.931)

0.766 (0.445, 1.319)

13.5

13.5

11.3

13.2

Sex

Female

Male

124/161

52/67

110/142

71/82

0.896 (0.682, 1.175)

0.591 (0.402, 0.868)

13.2

14.7

12.3

9.6

Prior regimens for advanced STS

2

>2

92/121

84/107

92/122

89/102

0.902 (0.671, 1.214)

0.640 (0.466, 0.879)

13.9

13.2

12.3

10.1

Stratification region*

Region 1

Region 2

Region 3

63/87

85/106

28/35

69/86

84/105

28/33

0.669 (0.466, 0.958)

0.890 (0.653, 1.214)

0.667 (0.380, 1.171)

15.3

13.3

11.4

11.5

11.5

9.7

Group/Subgroup Eribulin Dacarbazine

— Events/n —

Eribulin Dacarbazine

Median (months)

HR (95% CI)

0.25 1 4 16

Favors eribulin Favors dacarbazine

Randomized Phase II Study Comparing Gemcitabine Plus Dacarbazine Versus Dacarbazine Alone in Patients With Previously Treated Soft Tissue Sarcoma: A Spanish

Group for Research on Sarcomas Study

• 113 pts with STS (2 previous lines of CT; adria & ifosfamide)

• Gem 1800mg/m² fixed + DTIC 500 mg/m² q2 weeks or DTIC 1200 mg/m² q3 weeks

• Primary endpoint, PFR @ 3 months (40% to 60%)

García-del-Muro et al. J Clin Oncol 29:2528-2533

• Four parallel randomized, double-blind, placebo-controlled, multicenter phase II studies in pts with refractory STS

• Regorafenib (160 mg once daily, 3 weeks on/one week off) plus BSC vs placebo plus BSC

• Stratification: prior exposure to pazopanib and country

• Primary endpoint: Progression-free survival (PFS)

Advanced liposarcomas

N = 50

Advanced other sarcomasN = 50

Advanced synovial sarcomasN = 25

Advanced LMSN = 50

Until unacceptable toxicity or progression. Patients receiving PBO who experience disease progression were offered open-label RE.

R, ratio; RE, regorafenib; PBO, placebo; BSC, best supportive care.Penel N, et al. J Clin Oncol. 2016;34(suppl):abstract 11003; Accessed at https://clinicaltrials.gov/ct2/show/NCT01900743.

Strata A Strata B Strata C Strata D

RE+

BSC

PBO+

BSC

1:1

n=23n=20

R

RE+

BSC

PBO+

BSC

1:1

n=28n=28

R

RE+

BSC

PBO+

BSC

1:1

n=14n=13

R

RE+

BSC

PBO+

BSC

1:1

n=27n=29

R

Activity of cediranib in alveolar soft part sarcoma (ASPS)confirmed by CASPS (cediranib in ASPS), an international,randomised phase II trial

Ian Judson, James Morden, Michael Leahy, Vivek Bhadri, Quentin Campbell-Hewson, Ricardo Cubedo, Adam Dangoor, Ivo Hennig, Warren Joubert, Sarah Kernaghan, Antonio López Pousa, Beatrice Seddon, Claire Snowdon, Martin Tattersall, Christy Toms, Javier Martinez Trufero, Judith Bliss

This presentation is the intellectual property of the author/presenter. Contact them at [email protected] for permission to reprint and/or distribute

in partnership with

C2130/A12118

American Society of Clinical Oncology Annual Meeting, 2–6 June 2017

Progression-free survivalDefined as time from randomisation to first progression or death (any cause)

59

HR (for Cediranib v Placebo) = 0.54 (90% CI 0.30 – 0.97)Log-rank one-sided p = 0.041

Excluding patients with prior cediranib:HR = 0.52 (90% 0.28 – 0.95); Log-rank one-sided p = 0.036

0

25

50

75

100

% s

urv

ivin

g P

FS e

ven

t fr

ee

16 10(6) 5(4) 3(2) 3(0) 1(1) 1(0) 1(0) 1(0) 0(0) 0(0)Placebo: 32 27(4) 17(7) 12(2) 11(1) 8(2) 7(1) 6(1) 5(1) 3(1) 1(1)Cediranib:

N at risk (events)

0 3 6 9 12 15 18 21 24 27 30Time post randomisation (months)

Cediranib

Placebo

Besancon 18 1 18

GIST

Sarcomas and

aggressive connective

tissue tumors

Kinase

mutations

G5

Ntl

Group

Translocations

DFSP

SyS

Ewing

Mutations

APC/bCat

Desmoids

WD/DDLPSAmplification

12q13-15

MDM2/CDK4

MPNST

PEComas

TSG loss

NF1, TSC1/2

Complex genomics

LMS, UPS

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

0 6 12 18 24 30 36 42 48 54 60 66 72 78 84 90 96 102 108 114 120

Months

Su

rviv

al p

rob

ab

ilit

y

BFR14 trial: N = 434 patients

Median OS: 75.9 months (6.4 yrs)

B2222 trial: N =147

Median OS: 58 months

Perol M et al., J Clin Oncol., 2012; 30: 2516-24; Blanke CD et al., J Clin Oncol., 2008; 26:620-25.

Long term OS of GIST patients in the BFR14 trial

Kit and PDGFRα are mutated in GIST

63Heinrich et al. Hum Pathol 2002;33:484; Science 2003, Corless et al. Proc AACR 2003.

Membrane

Cytoplasm

Exon 11 (67.5%)

Exon 9 (11%)

Exon 13,14 (1%)

Exon 17 (0.5%)

Exon 12 (0.9%)

Exon 18 (6.3%)

KIT PDGFRα

• KIT & PDGFRA: 85%

• Other key genes involved:

- NF1, Raf, SDH, IGF1R

Exon 14 (0.3%)

Imatinib sensitive

+ Sunitinib sensitive

JAMA Oncol 2016;2(7):922-8.

64

Immunohistochemical Analysis (IHC) and Genetic Characteristics of Tumors From 95

Patients

With KIT/PDGFRA Wild-Type Gastrointestinal Stromal Tumors

GIST are at least ten diseasesDose Adjuvan

t

KIT

KIT exon 11 Im 400 +

KIT exon 9 Im 800 +

KIT others Im 400 +

PDGFRA

Non-D842V Im 400 +

D842V: ? -

KIT/PDGFR

WT

Im 400 +/?

NF1 ?/Im

400

-

SDH mutated ?/Im

400

-

SDH

epimutated

?/Im

400

-

RAF, Ras ? -

(years)

0 1 2 3 4 5 6

0

10

20

30

40

50

60

70

80

90

100

400mg-ex9 800mg-ex9 400mg-other 800mg-other

Progression free survival

(years)

0 1 2 3 4 5 6

0

10

20

30

40

50

60

70

80

90

100

400mg-ex9 800mg-ex9 400mg-other 800mg-other

Overall survival

BFR14 trial

GIST mutated on codon 557-558

High risk of relapse but good predictive factor

< 556 557/558 > 559

CR 30% 43.1% 23.5%

PR 57.5% 40.3% 45.1%

SD 10% 16.7% 31.4%

JF Emile et al, ASCO 2013

P= 0.02

PDGFRA GIST in advanced phase

Months

Cu

mu

lati

ve s

urv

ival

pro

bab

ility

Months

Pro

bab

ility

of

Pro

gres

sio

n-f

ree

surv

ival

Cassier et al. Clin Cancer Res. 2012; 18: 4458-64.

Secondary GIST mutations in patients progressingon imatinib or sunitinib

69

65

4

67

0

82

9

82

3

82

2

82

2

82

0

82

0

81

6

V T AD YNNDD

Exon 13 Exon 14 Exon 17 Exon 18

A I PH DKYYG

Drug/ATP binding pocket 42.9%

Kinase activation loop 57.1%

-

-Exon 11

Exon 9

Exon 13

544 550 580 589 976937

KIT Kinase Domain

V654 T670K623 810 830

Exon 11 deletions

P-loopActivation

loopCatalytic

loop

DHPLC, denaturing high-pressure liquid chromatography.

Liegl B et al. J Pathol 2008;216(1):64-74; Wilhelm S. 2006; Patent #WO2007059154 A2, C’KIT Cytoplasmic Domain figure.

Primary KIT

Mutation

70

Primary

Mutations

Exon 13: 1%K642E

Exon 9: 12%

Exon 11: 70%

Exon 17: 1%N822H/K, D820Y

Protein

Domain

Ligand binding

JM

ATP binding

Activation Loop

Exon 13 V654A

T670I

D816A/G/H/V

D820A/E/G/Y

N822H/K

Y823D

IM SU

A829P

Secondary

Mutations

Drug

Sensitivity

Exon 14

Exon 17

Exon 18

Sensitive

REG

NR

NR Not reported

Gramza et al, Clinical Cancer Research 15:7510, 2009

Heinrich et al, ASCO 2013 Poster/Abstract 10509

PON

Sunitinib and regorafenib: Second and Third line agents

1473O: Encouraging activity of novel pan-KIT and PDGFRα inhibitor DCC-2618 in

patients (pts) with gastrointestinal stromal tumor (GIST) – Janku F, et al

KEY RESULTS (CONT.)

*66% increase in tumour size; †PR at RP2D; ‡MAF reductions from baseline for Exons 9, 11, 13, 14, 17, and 18 – patients

with detectable plasma cfDNA at baseline and at least one follow-up are included §Patient in first dose cohort; ‖Patient with mixed histology

MAF, mutation allele frequency

• Use of cfDNA as pharmacodynamic biomarker shows pan-KIT activity of DCC-2618 in patients with KIT

mutant and advanced GIST

• DCC-2618 causes MAF reductions in cfDNA in all resistance-associated exons

• Treatment decisions were based on disease control (and not on MAF changes)

≥100 mg/day

+ 100 fold

+10 fold

1

-10

fold

-3

Be

st fo

ld c

ha

ng

e in

mu

tation a

llele

fre

qu

ency, lo

g s

ca

le

Use of cfDNA as pharmacodynamic biomarker

(Best response per RECIST, n=19)‡

-100

fold

§ §

Ex 9 Ex 11 Ex 13Ex 14 Ex 17 Ex 18

40

20

0

-20

-40

Ma

xim

um

re

du

ction

–su

m o

f d

iam

ete

rs

ch

an

ge fro

m b

ase

line

, %

PR

PD

DCC-2618 dose assigned

<100 mg/day

SD† †

*

Waterfall plot of KIT/PDGFRα GIST patients

(Best response per RECIST, n=37)

‖‖

Janku F, et al. Ann Oncol 2017; 28 (suppl 5): Abstr 1473O

Besancon 18 1 18

GIST

Sarcomas and

aggressive connective

tissue tumors

Kinase

mutations

G5

Ntl

Group

Translocations

DFSP

SyS

Ewing

Mutations

APC/bCat

Desmoids

WD/DDLPSAmplification

12q13-15

MDM2/CDK4

MPNST

PEComas

TSG loss

NF1, TSC1/2

Complex

genomics

LMS, UPS

Imatinib mesylate in advanced dermatofibrosarcoma protuberans (DFSP) -pooled analysis of two phase II clinical trials

P. Rutkowski, S. Schuetze, M. M. Van Glabbeke, C. Rankin, W. Ruka, B. P. Rubin, M. Debiec-Rychter, A. Lazar, H. Gelderblom, J-Y Blay, R. Sciot, P. Hohenberger, A. T. van Oosterom; for the EORTC Soft Tissue/Bone Sarcoma Group and South-West Oncology Group

Study

EORTC

(N=16)

SWOG

(N=8)

Total

(N=24)

N (%) N (%) N (%)

Response at 14 weeks

PR 5 (31.3)

SD 6 (37.5)

PD 3 (18.8)

Not evaluable 2 (12.5)

Best overall response

PR (confirmed) 3 (18.8) 4 (50.0) 7 (29.2)

PR (resected) 4 (25.0) 0 (0.0) 4 (16.7)

SD 4 (25.0) 2 (25.0) 6 (25.0)

PD 3 (18.8) 1 (12.5)* 4 (16.7)

Not evaluable 2 (12.5) 1 (12.5) 3 (12.5)

(years)

0 1 2 3 4

0

10

20

30

40

50

60

70

80

90

100

O N Number of patients at risk : all

6 24 20 15 4 1

Overall survival(ITT)

(years)

0 1 2 3 4

0

10

20

30

40

50

60

70

80

90

100

O N Number of patients at risk : Group

8 16 9 6 3

4 8 3 2 0

EORTC

SWOG

Time to progression(ITT)

N=16, CR 7%, PR50%, SD 37%, PD 7%

M-CSFR inhibitors (TKI & Ab) in PVNS with t(1,2), col3A6-CSF1 fusions

• Case report in 2008– (Ann Oncol 2008)

• Retrospective study2011– (Cancer 2011)

• Prospective study2012– (Proc ASCO 2012)

760

1

2

3

4

5

6

7

8

0 0,1 0,2 0,3 0,4 0,5 0,6 0,7 0,8 0,9 1

1st interim analysis

2nd interim analysis

3rd interim analysis

18/09/06 08/11/06 28/02/07

Figure: Response to imatinib in PVNS

The best tumor

shrinkage is

illustrated

according to

RECIST

Response to

imatinib in PVNS

Distribution of the

probability of

efficacy of nilotinib

(Bayesian design)

Besancon 18 1 18

GIST

Sarcomas and

aggressive connective

tissue tumors

Mutations

kinases

G5

Ntl

Group

Translocations

DFSP

SyS

Ewing

Mutations

APC/bCat

Desmoids

WD/DDLPSAmplification

12q13-15

MDM2/CDK4

MPNST

PEComas

TSG loss

NF1, TSC1/2

Genomique

complexe

LMS, UPS

Parameters guiding the treatment

80

• Strong driver mutations?

• Weak driver mutations?

• Sum of weak drivers?

Driver mutations?

• Tumor cell

• Stroma

• Heterogeneity

• Host, patients

• Time

Sum of weak drivers?

81

Besancon 18 1 18

GIST

Sarcomas and

aggressive connective

tissue tumors

Mutations

kinases

G5

Ntl

Group

Translocations

DFSP

SyS

Ewing

Mutations

APC/bCat

Desmoids

WD/DDLPSAmplification

12q13-15

MDM2/CDK4

MPNST

PEComas

TSG loss

NF1, TSC1/2

Genomique

complexe

LMS, UPS

Weak driver mutations?

83

Phamacokinetic and pharmacodynamic data

Weak driver mutations?

84

Genes whose knockdown is anti-proliferative in dedifferentiated liposarcoma, and the consequences of CDK4, MDM2

and YEATS4 knockdown in dedifferentiated liposarcoma

Nat Gen 2010

MDM2/p53 inhibitors

• RG7112 & RO5503781 (Roche)

• CGM097 (Novartis)

• MI-888 & SAR405838 (Sanofi)

• MK-8242 (Merck )

• DS-3032b (Daichii)

• AMG232 (Amgen)

Parameters guiding the treatment

89

• Tumor cell

• The stroma of the tumor

• Heterogeneity

• Host, patients

• Time

Use of PD-1 Targeting, Macrophage Infiltration, and IDO Pathway Activation in Sarcomas: A Phase 2 Clinical Trial, Toulmonde et al JAMA Oncol. 2017 Jun 29. doi: 10.1001/jamaoncol.2017.1617. • 57 patients: n=1 PR.

• The 6-month nonprogression rates were 0%, 0%, 14.3% (95% CI, 1.8%-42.8%) for LMS, UPS, and others, respectively, and 11.1% (95% CI, 2.8%-48.3%) for GIST.

• Strong infiltration by macrophage expressing the inhibitory enzyme indoleamine2,3-dioxygenase 1 (IDO1)

• Significant increase in the kynurenine to tryptophan ratio was observed in patient plasma samples during the study treatment.

KEY RESULTS

11008: Multicenter phase II study of pembrolizumab (P) in

advanced soft tissue (STS) and bone sarcomas (BS): Final

results of SARC028 and biomarker analyses – Burgess MA, et al

Burgess MA, et al. J Clin Oncol 2017; 35 (Suppl): Abstr 11008

Leiomyosarcoma

Pleomorphic undifferentiated sarcoma

Poorly differentiated / dedifferentiated liposarcoma

Synovial sarcoma

Pe

rce

nta

ge

ch

an

ge

300

200

100

0

–100

Change in target lesion size in STS

Sarcoma histologic initial diagnosis

30% reduction

n (%) LMS UPS LPS Synovial Total

CR 0 1 (10) 0 0 1 (2.5)

PR 0 3 (30) 2 (20) 1 (10) 6 (15)

SD 6 3 4 2 15

PD 4 3 4 7 18

Events

/ n

Median,

weeks

12-week

estimate, %

Leiomyosarcoma 10 / 10 16 (8, 20) 60 (30, 90)

Liposarcoma 10 / 10 25 (8, 42) 60 (30, 90)

Pleomorphic 7 / 10 30 (8, 68) 70 (42, 98)

Synovial sarcoma 10 / 10 7 (7, 22) 30 (2, 58)

Deaths

/ n

Median,

weeks

12-week

estimate, %

Leiomyosarcoma 8 / 10 38 (28, 65)100 (100,

100)

Liposarcoma 7 / 10 43 (19, 96) 90 (71, 100)

Pleomorphic 4 / 10 NR 90 (71, 100)

Synovial sarcoma 6 / 10 49 (32, )100 (100,

100)

RECIST best response in STS

STS: median PFS 18 weeks; 12-week PFS 55%

STS: median OS 49 weeks

KEY RESULTS (CONT.)

CONCLUSIONS

• Pembrolizumab is generally well tolerated for patients with advanced sarcomas,

and shows promising activity in UPS and LPS

• PD-L1 correlates with response in UPS

• Pembrolizumab is a potential treatment option in subsets of sarcomas (UPS, LPS)

11008: Multicenter phase II study of pembrolizumab (P) in

advanced soft tissue (STS) and bone sarcomas (BS): Final

results of SARC028 and biomarker analyses – Burgess MA, et al

Burgess MA, et al. J Clin Oncol 2017; 35 (Suppl): Abstr 11008

Pe

rce

nta

ge

ch

an

ge

600

400

200

0

500

300

100

–100

Chondrosarcoma

Ewing

Osteosarcoma

Change in target lesion size in bone sarcoma

Sarcoma histologic initial diagnosis30% reduction

n (%) Chondro Ewing Osteo Total

PR 1 (20) 0 1 (5) 2 (5)

SD 1 2 6 9

PD 3 11 15 29

RECIST best response in bone sarcoma

Events/n

Median,

weeks

12-week estimate,

%

Chondrosarcoma 4 / 5 8 (7, ) 40 (0, 83)

Ewing 13 / 13 7 (7, 8) 15 (0, 35)

Osteosarcoma 22 / 22 8 (7, 18) 32 (12, 51)

Deaths/n

Median,

weeks

12-week estimate,

%

Chondrosarcoma 1 / 5 NR 80 (45, 100)

Ewing 10 / 13 41 (18, 65) 85 (65, 100)

Osteosarcoma 14 / 22 50 (31, 74) 91 (79, 100)

Bone sarcoma: median PFS 8 weeks

Bone sarcoma: median OS 52 weeks

11007: A multi-center phase II study of nivolumab +/- ipilimumab for patients with

metastatic sarcoma (Alliance A091401)

– D’Angelo SP, et al

STUDY OBJECTIVE

• To assess the efficacy and safety of nivolumab with or without ipilimumab in patients with metastatic sarcoma

who were failing prior regimens was assessed

D’Angelo SP, et al. J Clin Oncol 2017; 35 (Suppl): Abstr 11007

Primary endpoint

• Response

Secondary endpoints

• Safety, DoR, clinical benefit rate, TTP,

PFS, OS

Nivolumab 3 mg/kg +

ipilimumab 1 mg/kg

q3w x4

(n=42)

Nivolumab

3 mg/kg q2w

(n=43)

Key patient inclusion criteria

• Advanced or unresectable

sarcoma

• ≥1 prior treatment

• ECOG PS 0–1

(n=85)PD*/

toxicity/

up to

2 years

PD*/

toxicity/

up to

2 years

Nivolumab

3 mg/kg

q2w

Crossover (n=4)

*Treatment beyond PD permitted

in first 12 weeks

KEY RESULTS

11007: A multi-center phase II study of nivolumab +/- ipilimumab

for patients with metastatic sarcoma (Alliance A091401)

– D’Angelo SP, et al

D’Angelo SP, et al. J Clin Oncol 2017; 35 (Suppl): Abstr 11007

Nivolumab + ipilimumab

(n=38)

Nivolumab (n=38)

BOR, n (%)

CR

PR

SD

PD

Death/no assessment

2 (5)

5 (13)

19 (50)

10 (27)

2 (5)

0

3 (8)

15 (39)

20 (53)

0

ORR (CR + PR), n (%) [90%CI] 6 (16) [7, 29] 2 (5) [1, 15]

Clinical benefit rate (CR + PR + SD), %

90%CI) 29 (17, 43) 18 (1, 32)

Nivolumab + ipilimumab (n=42) Nivolumab (n=42)

Total (events) Median

(95%CI)

Total

(events)

Median

(95%CI)

OS 38 (20) 14.3 (9.6, NE) 38 (26) 10.7 (5.5, 15.4)

PFS 38 (30) 4.4 (2.6, 6.3) 38 (34) 2.1 (1.4, 4.4)

Predictive immune markers

Herbst Nature 2014, Tumeh Nature 2014,

Snyder NEJM 2014, Davoli et al, Sciance 2017

Mutation load

PDL1 expression

Immune infiltrates

Aneuploidy

N=19 pediatric patients

Osteosarcoma and Ewing

Detection of CAR T cells in PB

and in the tumor

4 SD, 3 to 14months

Parameters guiding the treatment

98

• Tumor cell

• The stroma of the tumor

• Heterogeneity

• Host

• Time

Host… and genotype

• PS

• Age

• Comorbidities

• Associated treatments

• Observance / Compliance

• …

99

VariableHR

(95% CI)P

KITL541 6.1 (1.8-21) 0.004

Tumor size

(< 50 mm versus ≥ 50

mm)

3.6 (1.1-12) 0.03

Location

(gastric versus non

gastric)

0.5 (0.1-2) 0.06

Mitotic index per 50

HPF (< 5 versus ≥5)6.2 (1.7-23) 0.006

Mutation status

(all other versus KIT

exon 11)

1.5 (0.4-5) 0.6M. Brahmi, L. Alberti, A. Dufresne et al.

100

Rare variant burden analysis for TP53, ATM, BRCA2, ERCC2, and ATRComparisons were made between the Caucasian subset of the sarcoma cases with the combined controls

from the ISKS, LifePool, and Exome Variant Server datasets. Data are odds ratios and 95% Cis for each

comparison. ISKS=International Sarcoma Kindred Study. OR=odds ratio.

Sarcoma are fragmented in a myriad of different histotypesand molecular subtypes

Paradigmatic models for targeted therapy

Strong drivers vs weak drivers: how to distinguish?

Sarcoma stroma and infiltrating cells play an important role , but we do not know how to manipulate in therapy

Constitutional genome

Heterogeneity and complexity increases overtime , and withtherapeutic pressure

The next challenges:

◦ To identify other « strong drivers »

◦ To integrate complexity and multiple sequential treatments

Besancon 18 1 18

JY BlayCentre Leon Berard, Lyon, France

[email protected]

EURACAN

ERN

• The European Commission (EC) is supporting Member States in the development of European Reference Networks (ERNs) to link existing highly specialised healthcare providers across the European Union (EU).

• As expertise in rare or low prevalence complex diseases or conditions is scarce and dispersed, ERNs will facilitate timely access to care, both diagnosis and treatment, by centralising knowledge and experience, medical research and training, and resources for these diseases and conditions.

• The EC has been mandated to define the requirements for ERNs as specified in Article 12 of the Directive 2011/24/EU1 on patient’s rights in cross-border healthcare.

EURACANWhat are the objectives

• Improving the quality of care of patients withRare cancers in EU

• Guidelines

• Communication

• Patient pathways

• Expertise for all

• Cross border

• Research

EURACAN domains and subdomains

EURACAN

G1 Sarcoma

G2Rare female

genital organ / placenta

G3Rare

male genital organ / urinary

tract

G4Rare

neuroendocrine system

G5Rare

Digestive tractG6

Rare Endocrine

organs

G7Rare

Head and neck

G8Rare

thorax

G9Rare skin / eye

melanoma

G10Rare brain /

spinal cords / meninges /

cranial nerves

G1 Soft tissue /

visceralsarcoma

G1 Bone

SarcomaG2

Trophoblastic disease

G2Rare ovarian

G3Testis

G5Peritoneal

G5Biliary tract

G5Anal

G9Skin

G9Eye melanoma

Domains Sub domains

DISTRIBUTION OF MEMBERS BY COUNTRY

COUNTRIES/Towns

BELGIUM (Antwerp, Brussels,

Leuven, Liège)

CZECH REPUBLIC (Brno, Prague)

DENMARK (Aarhus)

GERMANY (Berlin, Essen,

Mannheim, Hamburg-Eppendorf,

Marburg, Würzburg)

FINLAND (Turku)

FRANCE (Lyon, Paris, Villejuif)

HUNGARY (Budapest)

IRELAND (Dublin)

ITALY (Aviano, Bologna, Candiolo,

Firenze, Genoa, Meldola, Milan,

Naples, Rome, Siena, Torino, Treviso)

LITHUANIA (Kaunas)

NETHERLANDS (Amsterdam,

Leiden, Maastricht, Njimegen,

Rotterdam, Gronigen)

NORWAY (Oslo)

POLAND (Warsaw)

Portugal (Coimbra, Lisboa, Porto)

SPAIN (Sevilla, Barcelona)

SWEDEN (Karolinska, Uppsala)

SLOVENIA (Ljubljana)

UNITED KINGDOM (Coventry,

London, Oxford, Sheffield)

GOVERNANCE

EURACAN General Assembly

Board of all HCP members andassociate/affiliate partners

Domains(Clinical action)

Guidelines

Research

Training/Education

Funding/sustainability plan

Communication/

Interaction with PAGs

G1

Sar

com

a

G2

Rar

e G

YN

G3

Rar

e G

U

G4

NET

G5

Rar

e G

I

G6

En

do

crin

e

G7

Rar

e H

&N

G8

Rar

e Th

ora

cic

G9

Rar

e S

kin

G1

0 R

are

Bra

inLeader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.Leader

Secret.

Transversal Task Forces

Steering CommitteeCoordinator

10 Group leaders+ 1 representative/country not already

represented7 task force leaders

Patient Advocacy groups

Scientific Advisory Board6 independent experts

Rare/frequent cancer/diseasesOutside / inside EU

Decisions for key questions

Decisions for daily management

Dissemination

Quality control

Jean-Yves BLAY EURACAN Coordinator Panu JAAKOLA (Turku Finland)

Secretary Miklos KASLER (Budapest Hungaria)

Treasurer Dermot O'TOOLE (Dublin Ireland)

Arimantas Tamasauskas (Kaunas Lithuania) Antonio ARAUJO (Porto Portugal)

Ignace VERGOTE (Leuven Belgium) Henrik FALCONER (Karolinska, Sweden)

Javier Martin BROTO (Seville Spain ) Katerina KOPECKOVA (Prague Czech Republic)

DOMAIN LEADERS TTF LEADERS

Paolo CASALI G1 : SarcomaESMO, Peter HOHENBERGERGuidelines

Michael SECKL G2: Rare gynecological cancersEORTC, Stéphane LEJEUNEResearch

Jourik GIETEMA G3: Rare GU cancersESO,Training

Martyn CAPLIN G4: Neurendocrine tumors NETBassim HASSANFunding

Lucjan WYRWICZ G5: Rare digestive tract tumoursIsabelle Manneh Vangramberen, ECPC - Kathy Oliver, IBTACommunication

Eric BAUDIN G6: Endocrine cancersIsabelle Manneh Vangramberen, ECPC- Kathy Oliver, IBTAdissemination

Lisa LICITRA G7: Rare Head and NeckOECI Belgique Dominique De ValeriolaQuality control

Nicolas GIRARD G8: Rare Thoracic PAG

Dirk SCHADENDORF G9: Rare Skin/Eye melanoma to be determined

Martin VAN DEN BENT J. G10 : Rare Brain tumors

STEERING COMMITTEE

A randomized trial of surgery in metastatic GIST Shi et al Eur J Cancer 2014

Parameters guiding the treatment

111

• Tumor cell

• The stroma of the tumor

• Heterogeneity

• Host, patients

• Time

112

Summary of Truncal and Branched Driver Events across Cancer Types

Parameters guiding the treatment

113

• Tumor cell

• The stroma of the tumor

• Heterogeneity

• Host

• Time

114

This slide includes the information about the drug that not approved in Japan.