Targeting FGFR alterations in cancer with BGJ398, a selective FGFR inhibitor

Targeting FGFR alterations in cancer with BGJ398, a selective FGFR inhibitor Randi Isaacs MD Novartis Institutes for BioMedical Research Translational...
33 downloads 0 Views 3MB Size
Targeting FGFR alterations in cancer with BGJ398, a selective FGFR inhibitor Randi Isaacs MD Novartis Institutes for BioMedical Research Translational Clinical Oncology

2 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Targeting FGFRs - The original hypothesis !  FGF/R Genetic Alterations in Cancer:

4 FGFRs, 22 FGFs FGFR3 mutations FGF in bladder cancer Ig I

! FGFR3 Activating Mutations:

acid box

•  superficial bladder cancer: 70% •  invasive bladder cancer: 10%

Ig II Ig III

!  GOF studies showed mutant FGFR3 transforms NIH3T3 cells

GRB2 SOS FRS2

PI3K

!  LOF studies showed dependence on FGFR3 mutant

FGFR R248C S249C Ras GTP

G372C

TM Ras Y375C GDP

TK-1

SurvivalTK-2

MAPK

K652E

Proliferation

3 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Targeting FGFRs - The extended hypothesis FGF/R Genetic Alterations in Cancer: ! Activating Mutations: •  FGFR3: bladder •  FGFR2: endometrial, lung •  FGFR4: rhabdomyosarcoma

! Gene amplification •  FGFR1: breast, lung •  FGFR2: gastric, breast •  FGF19: liver

! Gene rearrangements → fusions •  FGFR1, FGFR2, FGFR3: cholangio, GBM, bladder, prostate, breast, lung YM Wo et al Cancer Discovery 2013

4 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398 is a potent pan-FGFR inhibitor Cellular FGFR autophosphorylation assay

Biochemical kinase assay

IC50 µM

FGFR1

FGFR2

FGFR3

FGFR4

FGFR1

FGFR2

FGFR3

FGFR4

0.0009

0.0014

0.001

0.06

0.0065

0.0058

0.0058

0.225

BGJ398 has predominant activity against FGFR1, FGFR2 and FGFR3 5 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398 is selective against a panel of kinases Biochemical assay

BGJ398

6 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Kinase

IC50 nM

Kinase

IC50 nM

Kinase

IC50 nM

FGFR1 FGFR2 FGFR3 FGFR4

0.9 1.4 1 60

ABL ALK Aurora AXL BTK CAMK2 CK1 CDK1 CDK2 CDK4 COT1 CSK ERK2 EPHA4 EPHB4 FAK FLT3 FYN GSK3β HCK

2300 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 1900 > 10000 > 10000

HER1 HER2 HER4 IGF1R INS1R IRAK4 JAK1 JAK2 JAK3 JNK2 JNK3 KIT VEGFR2 LCK LYN MER MET MK2 MK5 MNK1 MNK2 MSTIR mTOR P38a P38g

> 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 750 180 2500 300 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 9000 > 10000 > 10000

PAK2 ac. PDGFRα PDK1 PI3Ka PI3Kb PI3Kd PIK4b PIM2 PKA PKBa PKCa PKCθ PKN1 PKN2 PLK1 RET ROCK2 RON SRC S6K SYK TYK2 VPS34 YES ZAP70

> 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 9000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 10000 > 9000 1100 > 10000

The Cancer Cell Line Encyclopedia BGJ398 profiled in 748 cell lines

BGJ398 IC50 µM

>8

7 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

~5%

BGJ398 cellular sensitivity is greatest among the FGFR genetically altered cell lines FGFR genet. alter.

FGFR wt

BGJ398 IC50 uM

2.5

1.5

0.5

48%

3%

8 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398 cellular sensitivity is greatest among the FGFR genetically altered cell lines

BGJ398 IC50 uM

0.25

0.20

0.15

0.1

0.05

FGFR fusion 9 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

FGFR1 amp

FGFR2 amp

FGFR2 mut

FGFR3 mut

BGJ398 inhibits proliferation of bladder cancer cell lines with FGFR3 genomic alterations *

>3000 FGFR3 mutation FGFR3 fusion

BGJ398 IC50 nM

2500

FGFR3 wt 2000

*FGFR3 not expressed

1500

*

1000

10 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

EJ198

HT1376

J82

KU1919

T24

UMUC3

VMCUB1

647-V

SW1710

BFTC805

HTB9

HT1197

TCCSUP

SW780

RT4

MGHU3

RT112

0

RT112/82

500

BGJ398 shows in vivo anti-tumor activity in the RT112 bladder cancer rat model (FGFR3 fusion)

Vehicle

BGJ398

BGJ398

5 mg/kg

10 mg/kg

Vehicle

pFRS2 pMAPK MAPK 11 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398

BGJ398

5 mg/kg

10 mg/kg

pFRS2

3h

pMAPK ß-tubulin

24h

BGJ398 Is Effective in FGFR1amp Lung Cancer Models Pharmacologic Activity of BGJ398 in Lung Cancer Cell Lines (N = 112)

Antitumor Efficacy in H1581 FGFR1amp Lung Cancer Xenografts

H1581 Tumor Volume, mm3

FGFR1 Copy Number

Vehicle qd

> 4 FGFR1 copies

NCI-H1581 DMSO

DMSO

BGJ398

pFRS2 α-actinin

BGJ398

!  BGJ398 is effective in DMS 114 and NCI-H1581 lung cancer models

•  Pharmacologic activity, with IC50 < 0.04 nM •  Reduced FGFR signaling, as determined by a decrease in phosphorylated FRS2 •  Antitumor activity in xenografts

Graus-Porta D, et al. AACR 2012 [abstract 854]; Wolf J, et al. AACR 2012 [abstract LB-122]. 12 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398 45 mg/kg qd

**One-way ANOVA: Dunnett vs vehicle.

BGJ398 IC50 nM DMS 114

BGJ398 10 mg/kg qd BGJ398 20 mg/kg qd

PTX’s with FGFR2 amplification respond to BGJ398 GAM033 (Crownbio)

CHGA010

Vehicle pFGFR2

BGJ398 15mg/kg

Vehicle pErk1/2 Erk1/2

FGFR2

13 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

β-tubulin

BGJ398 15mg/kg

BGJ398 FIM Phase 1 study design Dose expansion Arm 1: ~20-40 FGFR1 amplified squamous NSCLC continuous daily dosing

Dose escalation Oral, once-daily BGJ398, 28-day cycle MTD declaration 28Jun 2012 125mg QD

Dose levels Decision to dose escalate based on review of toxicity (DLT) in Cycle 1 and other clinical, PK, and laboratory data

Solid tumors FGFR1 or 2 amplified or FGFR3 mutated or other FGFR genetic alterations 14 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Arm 2: 21 FGFR ampl / mut solid tumors continuous daily dosing Arm 3: ≥ 10 solid tumors with FGFR genetic alterations 3 weeks on/1 week off dosing Arm 4: Up to 80 bladder cancer with FGFR3 mut/ fusion 3 weeks on/1 week off dosing 14

Baseline patient characteristics N = 119 Median age (range), years Age ≥ 65 years, n (%) Male / female, n (%)

59 (25-86) 31 (26) 52 (44) / 67 (56)

WHO performance status, n (%) 0/1/2

61 (52) / 54 (46) / 3 (3)

Primary tumor type, n (%) Breast

42 (35)

Lung

42 (35)

Bladdera

11 (10)

Other

24 (20)

Median prior lines of therapy, n a

Includes urothelial cell carcinoma of any origin.

15 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

3 Data as of September 9, 2014 cutoff

Patient disposition N = 119 Treatment ongoing, n (%) Treatment discontinued, n (%)

8 (7) 111 (93)

Primary reason for discontinuation, n (%)

• 

Disease progression

78 (66)

Withdrew consent

14 (12)

Adverse event

12 (10)

Death

5 (4)

Other

2 (2)

Most frequent AEs leading to discontinuation were eye disorders (n = 4)

16 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Treatment emergent adverse events (all grades) regardless of study drug relationship 5-60 mg n = 19

All patients =115; n (%) 100 mg 125 mg (c) 150 mg n=6 n = 53 n=6

125 mg (i) n = 31

Hyperphosphatemia

5 (26)

6 (100)

43 (81)

5 (83)

23 (74)

Constipation

1 (5)

3 (50)

28 (53)

0

15 (48)

0

4 (67)

23 (43)

2 (33)

12 (39)

Decreased appetite

3 (16)

3 (50)

24 (45)

3 (50)

13 (42)

Diarrhea

7 (37)

5 (83)

15 (28)

1 (17)

11 (36)

Fatigue

7 (37)

0

16 (30)

1 (17)

11 (36)

Nausea

6 (32)

4 (67)

14 (26)

3 (50)

9 (29)

Asthenia

1 (5)

2 (33)

11 (21)

1 (17)

6 (19)

Blood creatinine increased

1 (5)

2 (33)

12 (23)

1 (17)

7 (23)

ALT increased

2 (10)

1 (17)

9 (17)

1 (17)

3 (10)

AST increased

2 (10)

1 (17)

11 (21)

1 (17)

3 (10)

Stomatitis

17 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

BGJ398 pharmacokinetics accumulation upon multiple dosing at doses ≥ 60 mg

!  Accumulation in the range of 4- to 8-fold on day 15 (≥ 60 mg) likely due to autoinhibition of Cyp3A4 clearance pathways

Data as of September 24, 2013 cutoff. 18 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

125 mg/day

1000

BGJ398 Concentration, ng/mL

!  Consistent

Day 15 100

10

Day 1

1

0

0 1 2 3 4

6

8

Time, h

24

125 mg/day, mean (CV%) Day 1 (n = 40) Day 15 (n = 31) Cmax, ng/mL

102 (73)

253 (51)

AUClast, h⋅ng/mL

890 (101)

3614 (57)

BGJ398 treatment causes soft tissue mineralization in preclinical tox species !  Linked to elevated

systemic levels of Pi, VitD3

!  Preceded the onset of tissue mineralization

!  Inactive analogue of BGJ398 did not alter Pi, VitD3 Alveolar Mineralization 19 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

FGF23: an FGFR ligand that controls phosphate homeostasis and VitD3 metabolism Lumen

Kidney: Proximal tubule VitD3 metabolism

_

Pi re-absorption _

Basolateral membrane

FGFR1

FGF23

Bone: Osteocytes

FGF23

ɑ-KLOTHO

FGF23

Pi ↑ VitD3 ↑ •  FGF23 and ɑ-klotho ko mice develop tissue mineralization and hyperphosphatemia

•  Patients with familial tumoral calcinosis (LOF mutations in FGF23) suffer from hyperphosphatemia and ectopic calcifications 20 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Can we identify a monitorable biomarker that predicts exposures associated with pathway inhibition? Biomarker study:

!  ROC Analysis:

Biomarker

AUC

Pi

0.9

Sensitivity Specificity

measures diagnostic power of a biomarker

1

0.88

!  Reversibility upon treatment discontinuation? Treatment days 1 3

Pi (mg/dL)

11

7

9

15

7 5 vehicle

BGJ398 10mg/kg

BGJ398 20mg/kg

21 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

vehicle

BGJ398 10mg/kg

BGJ398 20mg/kg

Hyperphosphatemia as a pharmacodynamic marker of FGFR pathway inhibition mechanism of action1

!  The FGF23/FGFR pathway normally mediates phosphate secretion from renal tubules

!  Dose- and exposure-related

hyperphosphatemia seen in majority of patients at doses ≥ 100 mg daily !  Not seen with less potent FGFR inhibitors (multi-TKIs)

!  Managed through dietary

restrictions, phosphate-lowering therapy, and drug interruptions

1 Dienstmann R, et al. Ann Oncol. 2013;25:552-563. 22 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Individual Time Plot of Phosphate in 100-mg Dose Cohort 2.0

Change From Baseline of Phosphate, mmol/L

!  Biologic rationale and

Dose delay

1.5 1.0 0.5 0.0

Dose reduction (60 mg/day)

-0.5 -1.0 Baseline

10

20

30

40

50

Study Day of Assessment

•  • 

Serum Pi level in cycle 1 increases after exposure to BGJ398 Dose interruption results in quick decrease of serum Pi level

60

Hyperphosphatemia leads to predictable dose interruptions and empiric selection of alternate dosing schedule 100 mg/day continuous n=6

125 mg/day continuous n = 43

Median time to first dose interruption or reduction, days

23.5

22

Median duration of first dose interruption, days

4.5

7

160

Days to the First Dose Interruption/Reduction

140 120 100 80 60 40 20

Data as of 31JAN14 cutoff

0

23 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

100 mg Continuous Dosing

125 mg Continuous Dosing

Intermittent dosing schedule demonstrates greater tolerability and compliance 125 mg/day Continuous n = 53, n (%)

125 mg/day 3 Weeks On/1 Week Off n = 31, n (%)

Cycle 1 dose interruptions

24 (45)

5 (16)

Cycle 1 dose reductions

13 (24)

2 (6)

24 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Best Percent Change from Baseline

BGJ398 has clinical activity across multiple tumor types with FGFR genetic alterations

25 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

62-year-old male with metastatic cholangiocarcinoma to liver with FGFR2-BICC1 gene fusion Baseline and 8-week follow-up CT scans Patient history and course High throughput NGS demonstrated FGFR2 gene fusion and 27 mutations, including TP53 (MI-ONCOSEQ) Received 6 cycles 5-FU/Gem/CDDP with best response SD BGJ398 125 mg qd administered for 3 cycles, dose reduced to 100 mg for hyperphosphatemia/grade 1 elevated creatinine Disease progression at end of cycle 3

Wu et al Cancer Discovery 2013 26 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Phase 2 study of BGJ398 in cholangiocarcinoma opened in July 2014 N ≈55 Advanced cholangiocarcinoma w/FGFR2 gene fusion/other FGFR genetic alteration Prior CDDP-based chemotherapy or gemcitabinecontaining therapy for pts unable to receive CDDP

BGJ398 125mg qd x 21 days (28d cycles)

Primary endpoint: - ORR Secondary endpoint: -  PFS, DCR, BOR, OS -  Safety -  PK

27 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Continue until disease progression, unacceptable toxicity, withdrawal of consent or death

Conclusions !  The 125-mg once-daily dose was identified as the MTD !  BGJ398 has a tolerable safety profile, with hyperphosphatemia an on-target and manageable AE

!  The 3-weeks-on/1-week-off intermittent schedule

demonstrates greater compliance and tolerability vs continuous dosing

!  Clinical activity has been observed in multiple tumor types, patients with FGFR3-mutated bladder cancer may be especially sensitive to BGJ398 ! Refinement of molecular profile for Identification of sensitive subpopulations of patients with other tumors is ongoing

!  Clinical trials in bladder cancer, cholangiocarcinoma,

glioblastoma, squamous NSCLC, and endometrial cancer are open or planned

28 | Cholangiocarcinoma Foundation | 06Feb15 | RIsaacs |

Suggest Documents