54
¿Cómo tratamos al paciente EGFRm?, la importancia de buscar el mecanismo de resistencia Antonio Calles Servicio de Oncología Médica Hospital General Universitario Gregorio Marañón Instituto Investigación Sanitaria Gregorio Marañón Madrid, España

¿Cómo tratamos al paciente EGFRm?, la importancia …hitosoncologicos.com/wp-content/uploads/2017/11/03_M2_Dr.Antonio... · ¿Cómo tratamos al paciente EGFRm?, la importancia de

Embed Size (px)

Citation preview

¿Cómo tratamos al paciente EGFRm?, la importancia de buscar el mecanismo de resistencia

Antonio Calles

Servicio de Oncología Médica

Hospital General Universitario Gregorio Marañón

Instituto Investigación Sanitaria Gregorio Marañón

Madrid, España

NSCLC: From Histology to Genomics

Adenocarcinoma

“Druggable” genomic alterations

2004

Osimertinib Rociletinib Olmutinib

2014

3rd Generation EGFR TKIs presented at ASCO

Corral et al ELCC 2017

2017

Resistance to targeted therapy may have different underlying

mechanisms

Camidge et al, Nat Rev Clin Oncol 2014

Baseline: Start erlotinib 3m: Response 14m: PD

30m: Re-biopsy

37m: Offered trial 39m: First dyspnea

18m 24m

35m

Radiographic progression does not always result in

clinical or symptomatic progression

Courtesy Dr. Jänne

Aquired EGFR-TKI resistance: different resistance mechanisms

PD: rebiopsy Initial biopsy

Erlotinib, Gefitinib,

Afatinib (8-12m)

EGFR

del 19

L858R

Yu et al., Clin Cancer Res 2013

On target

Off target

How does resistance develop during targeted therapy ?

Treatment pressure – bottleneck phenomenon

Adapted from Gerlinger M & Swanton C, Br J Cancer 2010;103:1139‒1143.

Kobayashi et al., NEJM 2005

T790M

secondary EGFR exon 20

gatekeeper-mutation

•Methionine bulk replaces drug from ATP binding-pocket

The EGFR T790M mutation confers resistance to 1st and 2nd gen. EGFR-TKIs

Chmielecki Sci Transl Med 2011 Zhou Nature 2009

EGFR T790M confers resistance to quinazoline inhibitors but sensitivity to

pyrimidine-based inhibitors

Osimertinib tras progresión a Gefitinib:

Inhibidor EGFR de 3ª generación

Inicio de tratamiento Tras 8 semanas

3rd gen. EGFR-TKI show high efficacy in T790M + acquired resistance

Erlotinib, Gefitinib,

Afatinib (8-12m)

PD: rebiopsy

T790M Osimertinib (8-10m) EGFR

del 19

L858R

Initial biopsy

also: liquid biopsy

Phase I (Aura)

mPFS 9.6 m

ORR: 61%

DCR: 95%

Phase III (Aura 3)

(Osimertinib vs. CT)

mPFS 10.1 vs. 4.4 m

Jänne et al., NEJM 2015 Mok et al., NEJM 2016

AURA3 study design

Key eligibility criteria

• ≥18 years (≥20 years in Japan)

• Locally advanced or metastatic NSCLC

• Evidence of disease progression following first-

line EGFR-TKI therapy

• Documented EGFRm and central confirmation

of tumour EGFR T790M mutation from a tissue

biopsy taken after disease progression on first-

line EGFR-TKI treatment

• WHO performance status of 0 or 1

• No more than one prior line of treatment for

advanced NSCLC

• No prior neo-adjuvant or adjuvant

chemotherapy treatment within 6 months prior

to starting first EGFR-TKI treatment

• Stable* asymptomatic CNS metastases allowed

R

2:1

Osimertinib (n=279)

80 mg orally

QD

Platinum-pemetrexed (n=140)

Pemetrexed 500 mg/m2 +

carboplatin AUC5 or

cisplatin 75 mg/m2

Q3W for up to 6 cycles

+ optional maintenance

pemetrexed# Optional crossover

Protocol amendment allowed patients on

chemotherapy to begin post-BICR confirmed

progression open-label osimertinib treatment

Endpoints

Primary:

• PFS by investigator assessment (RECISTv1.1)

Secondary and exploratory:

• Overall survival

• Objective response rate

• Duration of response

• Disease control rate

• Tumour shrinkage

• BICR-assessed PFS

• Patient reported outcomes

• Safety and tolerability

• Patients were stratified at randomisation based on ethnicity (Asian/Non-Asian)

• RECISTv1.1 assessments performed every 6 weeks until objective disease progression; patients could receive study treatment beyond RECISTv1.1 defined

progression as long as they experienced clinical benefit

• With 221 events of progression or death, the study would have 80% power to reject the null hypothesis of no significant difference in duration of PFS between the

two treatment groups, assuming a treatment effect HR of 0.67 at 5% two-sided significance

*Defined as not requiring corticosteroids for 4 weeks prior to study treatment; #For patients whose disease had not progressed after 4 cycles of platinum-pemetrexed

HR, hazard ratio; Q3W, every 3 weeks; R, randomisation; RECIST, Response Evaluation Criteria In Solid Tumors; WHO, World Health Organization

• Analysis of PFS by BICR was consistent with the investigator-based analysis: HR 0.28 (95% CI 0.20, 0.38), p<0.001;

median PFS 11.0 vs 4.2 months.

Population: intent-to-treat

Progression-free survival defined as time from randomisation until date of objective disease progression or death. Progression included deaths in the absence of RECIST progression.

Tick marks indicate censored data; CI, confidence interval

AURA3 primary endpoint:

PFS by investigator assessment 1.0

0.8

0.6

0.4

0.2

0 0 3 6 9 12 15 18

Pro

ba

bilit

y o

f

pro

gre

ss

ion

-fre

e s

urv

iva

l

No. at risk Osimertinib

Platinum-pemetrexed

Months 279 140

240

93

162

44

88 17

50

7

13

1

0 0

Median PFS, months (95% CI)

HR (95% CI)

10.1 (8.3, 12.3) 0.30 (0.23, 0.41) p<0.001 4.4 (4.2, 5.6)

Osimertinib

Platinum-pemetrexed

PFS benefit in AURA3 patients with

CNS metastases at baseline

With CNS metastases Without CNS metastases

Population: intent-to-treat

Progression-free survival defined as time from randomisation until date of objective disease progression or death. Progression included deaths in the absence of RECIST progression.

Tick marks indicate censored data. CNS metastases determined programmatically from baseline data of CNS lesion site, medical history, and/or surgery, and/or radiotherapy.

Pro

ba

bil

ity o

f

pro

gre

ss

ion

-fre

e s

urv

iva

l

1.0

0.8

0.6

0.4

0.2

0

No. at risk

Osimertinib

Platinum-

pemetrexed

0 3 6 9 12 15 18

93

51

80

32

46

9

27

4

14

2

4

0

0

0

Months

Osimertinib (n=93)

Platinum-pemetrexed (n=51)

Median PFS, months (95% CI)

8.5 (6.8, 12.3)

4.2 (4.1, 5.4)

HR 0.32

(95% CI 0.21, 0.49)

Pro

ba

bil

ity o

f

pro

gre

ss

ion

-fre

e s

urv

iva

l

1.0

0.8

0.6

0.4

0.2

0

0 3 6 9 12 15 18

186

89

160

61

116

35

61

13

36

5

9

1

0

0

Months

Osimertinib (n=186)

Platinum-pemetrexed (n=89)

Median PFS, months (95% CI)

10.8 (8.3, 12.5)

5.6 (4.2, 6.8)

HR 0.40

(95% CI 0.29, 0.55)

All causality adverse events* in AURA3

N (%)

15% cut-off

Osimertinib (N=279) Platinum-pemetrexed (N=136)

Any grade Grade ≥3 Any grade Grade ≥3

Any AE 273 (98) 63 (23) 135 (99) 64 (47)

Diarrhoea 113 (41) 3 (1) 15 (11) 2 (1)

Rash# 94 (34) 2 (1) 8 (6) 0

Dry skin# 65 (23) 0 6 (4) 0

Paronychia# 61 (22) 0 2 (1) 0

Decreased appetite 50 (18) 3 (1) 49 (36) 4 (3)

Cough 46 (16) 0 19 (14) 0

Nausea 45 (16) 2 (1) 67 (49) 5 (4)

Fatigue 44 (16) 3 (1) 38 (28) 1 (1)

Stomatitis 41 (15) 0 21 (15) 2 (1)

Constipation 39 (14) 0 47 (35) 0

Vomiting 31 (11) 1 (<1) 27 (20) 3 (2)

Thrombocytopenia# 28 (10) 1 (<1) 27 (20) 10 (7)

Neutropenia# 22 (8) 4 (1) 31 (23) 16 (12)

Leukopenia# 22 (8) 0 20 (15) 5 (4)

Anaemia# 21 (8) 2 (1) 41 (30) 16 (12)

Asthenia 20 (7) 3 (1) 20 (15) 6 (4)

Select adverse events

Interstitial lung disease# 10 (4) 1 (<1) 1 (1) 1 (1)

QT prolongation 10 (4) 1 (<1) 1 (1) 0

Population: safety analysis set (all patients who received at least one dose of study drug and for whom post-dose data were available)

*Includes AEs with an onset date on or after the date of first dose and up to and including 28 days following discontinuation of randomised treatment or the day before first administration of cross-over

treatment. Some patients had more than one AE. #Grouped term: if a patient has multiple preferred term level events within a specific grouped term AE, then the maximum CTCAE grade across those events is

counted.

• DNA fragments of 120-200bp with half life of ~2 hours

• Real-time, non-invasive, multi-lesions

• Cheaper because it does not require a biopsy

• Often very low amount of ctDNA

in the sea of wild type DNA - ”Needle in a farm”

• Specific to tumor

Diaz J Clin Oncol 2016

Liquid Biopsy

Plasma EGFR T790M • Plasma from AURA trial sent for BEAMing

– Paired tumor and plasma available for 216 patients

• Plasma T790M testing not as predictive as tumor T790M testing

Oxnard et al, JCO, 2016

T790M+

in tumor:

62% RR

T790M-

in tumor

26% RR

• Plasma from AURA trial sent for BEAMing

– Paired tumor and plasma available for 216 patients

18 T790M+

in plasma,

not tumor 111 T790M+

in tumor and

plasma

47

T790M+ in

tumor, not

plasma

40 patients T790M- tumor and

plasma

Plasma EGFR T790M

Oxnard et al, JCO, 2016

• Plasma from AURA trial sent for BEAMing

– Paired tumor and plasma available for 216 patients

18 T790M+

in plasma,

not tumor 111 T790M+

in tumor and

plasma

47

T790M+ in

tumor, not

plasma

40 patients T790M- tumor and

plasma

T790M+ in tumor:

62% RR, 10m PFS

T790M+ in plasma:

63% RR, 10m PFS

Plasma EGFR T790M

Oxnard et al, JCO, 2016

• Plasma from AURA trial sent for BEAMing

– Paired tumor and plasma available for 216 patients

18 T790M+

in plasma,

not tumor 111 T790M+

in tumor and

plasma

47

T790M+ in

tumor, not

plasma

40 patients T790M- tumor and

plasma

T790M+ in tumor:

62% RR, 10m PFS

T790M+ in plasma:

63% RR, 10m PFS

16.5m

PFS

4.3m

PFS

Plasma EGFR T790M

Oxnard et al, JCO, 2016

• Plasma from AURA trial sent for BEAMing

– Paired tumor and plasma available for 216 patients

• Tumor T790M-/plasma T790M+ has lower relative levels of T790M

Plasma EGFR T790M

Oxnard et al, JCO, 2016

WCLC 2017

Plasma EGFR T790M

• Proposed algorithm where plasma genotyping is

a screening test prior to biopsy:

– If you have tumor, test the tumor

– If you don’t have tumor, plasma is a convenient

first pass test

– If plasma doesn’t reveal an actionable finding,

tumor genotyping is needed

– Tumor result trumps plasma result

Oxnard et al, JCO, 2016

Challenge 1: about 40% of AR cases are T790M negative

PD: rebiopsy Initial biopsy

Erlotinib, Gefitinib,

Afatinib (8-12m)

EGFRd

el 19

L858R

Yu et al., Clin Cancer Res 2013

On target

Off target

EGFR By-Pass Pathways: MET amplification

Zhang et al, Biomedicines 2015

Engelman Science 2007

MET/CEP7 probe set with specimens HCC827 (A) and HCC827 GR6 (B).

Scheffler et al, J Thorac Oncol 2015

Rebiopsy left arm: EGFR L858R, high-level METamp, TP53 C277*, TP53 R110C

Start crizotinib, continue erlotinib After 8 days of crizotinib + erlotinib

Molecular analysis pleural effusion: EGFR L858R T790M, low-level METamp Stop crizotinib and erlotinib, start AZD9291

Patient with activating EGFR mutation and good response for about 2 years:

now presenting with acquired resistance and massive progression

Waterfall plot based on evaluable patients (n = 64): all patients dosed and with on-treatment assessment or discontinuation prior to first tumour assessment

Data cut-off 31 Aug 2017

*17 patients did not have central FISH confirmation of MET-positive status (n = 6 MET-negative; n = 11 unknown by central lab); †Confirmed by a later scan performed at

least 4 weeks after initial response observed

TATTON Phase Ib expansion cohort: osimertinib plus savolitinib for patients with EGFR-

mutant MET-amplified NSCLC after progression on prior EGFR-TKI

Preliminary anti-tumour activity in all MET-positive patients*, n = 64

100

75

50

25

0

-25

-50

-75

-100

Best %

change fro

m b

aselin

e

in tum

our

lesio

n s

ize

Prior 3rd Gen T790M directed EGFR-TKI

No prior 3rd gen EGFR-TKI, T790M+

No prior 3rd gen EGFR-TKI, T790M-

TATTON Part B

NCT02143466

Objective

response rate,

n (%)

Prior 3rd Gen

T790M

directed

EGFR-TKI

(n = 30)

No prior 3rd Gen T790M

directed EGFR-TKI

Total

(n = 64)

T790M+

(n = 11)

T790M-

(n = 23)

ORR† 10 (33) 6 (55) 14 (61) 30 (47)

Ercan Cancer Discovery 2009

FISH amplification in MAPK1

Curative effect of W+T treatment in vivo.

Erin M. Tricker et al. Cancer Discovery 2015;5:960-971

EGFR Downstream Activation: ERK Signaling Resistance to EGFR TKI

Targeted and non-targeted treatment options in T790M neg. AR

PD: rebiopsy Initial biopsy

Erlotinib, Gefitinib,

Afatinib T79M

hl

MET

ampl.

HER2

ampl.

SCLC

???

Osimertinib (10m) EGFR

del 19

L858R

EGFR-TKI + METinh.

EGFR-TKI + HER2inh.

Chemo, I-O, anti-DLL3

Chemo

clinical

trials

PD: rebiopsy Initial biopsy

Erlotinib, Gefitinib,

Afatinib (10m) T790M Osimertinib (10m)

EGFR

del 19

L858R

also: liquid biopsy

Osimertinib (20m)

PD: rebiopsy

Challenge 2: acquired resistance to 3rd gen. EGFR inhibitors

?

Yu et al. JAMA Oncology 2015

Erlotinib

AZD9291

EGFR Mutations and Resistance to Irreversible Pyrimidine-Based EGFR Inhibitors

EGFR C797S mutation mediates resistance to 3rd generation EGFR TKI

Zhou et al, Nature 2009; Thress et al, Nature Medicine 2015

abolishes covalent binding

of 3rd gen. EGFR inhibitor

EGFR C797S in cis- or trans- position

Novel dominant EGFR solvent-front mutations in AR to osimertinib

Detection by plasma testing of a 69y old

Caucasian female with AR to osimertinib:

•L858R (MAF 17.9%)

•T790M (MAF 18.2%)

•G796S/R (MAF 14.4%)

•C797S/G (MAF 2.26%)

•L792F/H (MAF 0.35%)

•V802F (MAF 0.36%)

disables osimertinib

binding by sterical

interference

Ou et al, Lung Cancer 2017

Osimertinib resistance on tumor NGS

• EGFR C797S was detected only in patients

with maintained T790M

119 T790M+ patients treated with single-agent

osimertinib for acquired resistance

29 remain on therapy without

progression

10 off due to AE without progression

80 patients eligible for resistance analysis

35 without post-progression biopsy

12 without complete tissue testing

33 patients with progression tumor NGS

11

maintained

T790M

22 lost T790M

7 C797S

5 SCLC histologic

transformation*

1 BRAF V600E

1 KRAS Q61K

1 CCDC6-RET fusion

1 FGFR3-TACC

fusion

3 MET amplification

2 PIK3CA mutation*

1 PIK3CA

mutation

*Patient with both

PIK3CA + SCLC

transformation

• Competing resistance mechanisms were

detected in patients with loss of T790M

0 3 6 9

1 0

1 0 0

1 0 0 0

1 0 0 0 0

M o n th s s in c e s ta r t in g o s im e r t in ib

Co

pie

s/m

L o

f p

las

ma

E G F R e x o n 1 9 d e l

E G F R T 7 9 0 M

K R A S Q 6 1 K

B io p s y d a teS ta r t o f o s im e rtin ib

N /D

Oxnard et al, AACR 2017

Loss of T790M new competing resistance mechanisms

Validation cohort:

loss of T790M associated with early resistance

• Those with T790M loss had a shorter TTF compared to those with maintained T790M

0 6 12 18 24 300

25

50

75

100

TTF (months)

Pe

rce

nt

wit

ho

ut

trea

tme

nt

failu

re

T790M lossMedian: 5.5

T790M+/C797S-Median: 12.6

T790M+/C797S+Median: 12.4

N = 110

<=5.5 >5.5 to <=13 >130

10

20

30

40

TTF (months)

Nu

mb

er

of

pati

en

ts

T790Mloss

T790M+/C797S-

N = 38 N = 36 N = 36

T790M+/C797S+

• Plasma available for 157 subjects on the first-in-human AURA trial, 110 with a detectable

driver at time of osimertinib resistance

• Among those with early resistance (TTF <=5.5 months), 68% had loss of T790M

Oxnard et al, AACR 2017

Serial plasma genotyping on osimertinb

Rapid loss of T790M with

continued increase in driver

mutation in a patient with

small cell transformation on

biopsy

Emergence of C797S mutation with the

re-emergence of exon 19 del and T790M

after initial response to osimertinib

0 3 6 9 12

10

100

1000

10000

Time on osimertinib (months)

Co

pie

s/m

L o

f p

las

ma

Biopsy dateStart of osimertinib

EGFR C797S TA

EGFR C797S GC

N/D

EGFR T790M

EGFR exon 19 del

0 1 2

10

100

1000

10000

Time on osimertinib (months)

Co

pie

s/m

L o

f p

las

ma

EGFR exon 19 del

EGFR T790M

Start of osimertinib

Biopsy date

N/D

Acquired EGFR C797S Small cell transformation

• Resistance due to loss of T790M is often seen early on osimertinib

• Resistance due to EGFR C797S is often seen later on osimertinib

Conclusion

• Loss of T790M does not indicate resensitization to first-generation EGFR TKI, but often indicates overgrowth of a competing resistance mutation [NOMBRE

DE LA CATEGORÍA]

T790M+/ C797S-

Unknown

Other rare genotypes

MET amp

SCLC

T790M loss T790M maintained

• Range of rare genetic resistance mechanisms to look out for:

• KRAS mutations

• RET fusions

• FGFR fusions

Osimertinib resistance

• Timing of resistance impacts biology of resistance

• Repeat plasma T790M testing can guide next steps?

Oxnard et al, AACR 2017

Osimertinib

for T790M+

NSCLC

Early

Resistance

Late

Resistance

Suspect outgrowth of competing

resistance mechanism

Suspect resistance with

maintained EGFR addiction

Consider trial of osimertinib combined with

additional EGFR inhibitor (e.g. necitumumab)

Consider trial of osimertinib combined with

alternate pathway inhibitor (e.g. MET inhibitor)

Nazartinib (EGF816) Phase I dose-escalation part

Presented By Daniel Tan at 2017 ASCO Annual Meeting

Gene alterations at progression and nazartinib efficacy

Presented By Daniel Tan at 2017 ASCO Annual Meeting

Tumor heterogeneity:

different clones may drive

different metastatic sites

Govindan, Science 2014

Clonal heterogeneity

over time

Spatial heterogeneity at given timepoint

Evolutionary patterns of resistance differ on time and location

Treatment at clinical progression

Residual disease:

Treatment at time of response

Preventive Treatments

Immunotherapy is not a good option for 2L EGFRm NSCLC

Akbay, Cancer Discovery 2013

Is progression

clinically significant?

Progression on first-line EGFR TKI

Approach to the management of EGFR mutant NSCLC with progression on first-line EGFR TKI

Sacher, Jänne & Oxnard Cancer 2014

Is progression localized?

Is re-biopsy feasible?

Start chemotherapy +/- continued

TKI

There is not one uniform approach to treating resistance

• Paradigma de tratamiento dirigido en tumores sólidos.

– Rápidos avances, aumento de la complejidad, repercusión en supervivencia del paciente.

• No vamos a “curar” a los pacientes con terapias dirigidas anti-EGFR, pero sí tendremos pacientes “crónicos”

• No todos los EGFR TKIs son iguales y sus mecanismos de resistencia, tampoco.

• Los pacientes con adenocarcinoma de pulmón EGFR mutado requieren de un manejo individualizado (y multidisciplinar):

– Cirugía, Radioterapia, Quimioterapia, Terapias Dirigidas, ¿Inmunoterapia?

• Por definir el “mejor tratamiento” de entrada, la “mejor secuencia” de tratamientos, y ¿la mejor combinación? (sinergias on-target + off-target)

• Entender significado de biopsia líquida (falsos negativos) y monitorización

• Rebiopsiar las progresiones y tener fármacos disponibles

Take Home Messages Mecanismos de resistencia EGFRm

Thanks for

your attention!

[email protected]