Top Banner
Antitumor Activity of Margetuximab plus Pembrolizumab in Patients with Advanced HER2+ (IHC3+) Gastric Carcinoma Daniel V.T. Catenacci 1 , Haeseong Park 2 , Hope Elizabeth Uronis 3 , Yoon-Koo Kang 4 , Matthew C.H. Ng 5 , Philip Jordan Gold 6 , Peter C. Enzinger 7 , Keun Wook Lee 8 , Jill Lacy 9 , Se Hoon Park 10 , Jennifer Yen 11 , Justin Odegaard 12 , Aleksandra Franovic 11 , Sarah Church 15 , Anushka DeCosta 13 , Jan E. Baughman 13 , Aisha Wynter-Horton 13 , Francine Chen 13 , Jeff Nordstrom 13 , Paul A. Moore 13 , Tony Wu 13 , Jan Kenneth Davidson-Moncada 13 , Yung-Jue Bang 14 1 University of Chicago Medical Center and Biological Sciences, Chicago, IL; 2 Washington University School of Medicine, St. Louis, MO; 3 Duke University Medical Center, Durham, NC; 4 Asan Medical Center, Seoul, South Korea; 5 National Cancer Centre Singapore, Singapore; 6 Swedish Cancer Institute, Seattle, WA; 7 Dana-Farber Cancer Institute, Boston, MA; 8 Seoul National University Bundang Hospital, Seongnam, South Korea; 9 Smilow Cancer Hospital, Yale University, New Haven, CT; 10 Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; 11 Guardant Health, Inc., Redwood City, CA; 12 Lifecode, San Francisco, CA; 13 MacroGenics, Inc., Rockville, MD; 14 Seoul National University Hospital, Seoul, South Korea Abstract #65 Presented at The 2019 Gastrointesnal Cancers Symposium, January 17–19, 2019, San Francisco, CA [email protected] Background Trastuzumab + chemotherapy is standard treatment in 1 st line advanced HER2+ gastroesophageal adenocarcinoma (GEA) No HER2-targeted agents have been approved in post-trastuzumab setting in patients with HER2+ GEA Loss of HER2 expression after trastuzumab has been reported in up to 70% of GEA patients with potential consequences for subsequent treatment with HER2-targeted agents 1-5 Margetuximab is a next generation anti-HER2 monoclonal antibody featuring an optimized Fc domain designed to enhance its Fc-dependent functions, including antibody dependent cell cytotoxicity (ADCC) irrespective of the host’s FcγRIIIa (CD16A) genotype Hypothesis: Coordinate engagement of innate and adaptive immunity with margetuximab and anti-PD-1 mediates greater antitumor activity than either single agent alone Coordinate engagement of innate and adaptive immunity with combination of anti-HER2 and anti-PD-1 antibodies achieves greater antitumor activity than either agent alone in murine tumor models 6 Margetuximab and pembrolizumab have both demonstrated monotherapy antitumor activity in patients with GEA; and Checkpoint inhibitors (pembrolizumab, nivolumab) are approved for treatment of 3L PD-L1+ patients with GEA We reported previously that another Fc-optimized antibody (enoblituzumab, anti-B7H3) combined with pembrolizumab achieved greater antitumor activity than historical experience with checkpoint inhibitors alone in checkpoint-naïve patients with SCCHN and NSCLC (PD-L1<1%) (SITC 2018) 7 Preliminary data indicates that Fc-modified antibodies (including margetuximab and enoblituzumab monotherapy) can modulate T-cell repertoire in treated patients 8-9 NK cells may express PD-1, and PD-1/PD-L1 interaction can impair NK cell function, and PD-1/PD-L1 blockade can enhance NK cell function and preclinical antitumor activity 10 Cancer Cells Tumor Destruction Enoblituzumab Anti-PD-1 Antibody NK Cells Tumor Destruction Macrophages T Cells Enhanced Adaptive T-cell-mediated Antitumor Immunity Exhausted T Cells Innate Immunity Adaptive Immunity Enhanced ADCC Sensitize T Cells Counter T-cell Exhaustion Goal is to develop chemotherapy-free approach for treatment of GEA Study Design Dose Escalation Pembro (200 mg) + Margetuximab 10 & 15 mg/kg Gastric IHC 3+ Expansion N = 25 RP2D: Margetuximab (15 mg/kg) + Pembrolizumab (200mg) IV Q3 WK Enrollment completed. 15 patients ongoing Cohort Expansion N = 60 Open label, dose escalation study, conventional 3+3 design Escalating doses of margetuximab (marge; 10 mg/kg & 15 mg/kg) Fixed dose pembrolizumab (pembro; 200 mg) Response assessed by RECIST & irRECIST 92 Patients treated at recommended Phase 2 dose (RP2D) 61 Gastric cancer (GC) 35 Enrolled in cohort 2 (1), and cohort expansion (34) (HER2 IHC2+/3+) 26 Enrolled in HER2 (IHC3+) GC specific cohort 31 Gastroesophageal junction (GEJ) (HER2 IHC2+/3+) Efficacy Results in Gastric Cancer Population by Biomarker Expression n ORR DCR mPFS mOS Total 61 29.5% (18/61) 65.6% (40/61) 4.07 (2.30, 5.45) 14.62 (9.07, NR) IHC3+ 55 32.7% (18/55) 69.1% (38/55) 4.70 (2.66, 7.49) NR (12.48, NR) ERBB2 amp 35 40.0% (14/35) 77.1% (27/35) 4.76 (2.69, 7.59) 14.62 (8.41, NR) PDL1+ 26 46.2% (12/26) 80.8% (21/26) 4.14 (2.60, 7.59) NR (6.74, NR) IHC3+/ PDL1+ 23 52.2% (12/23) 82.6% (12/23) 4.14 (2.60, 15.54) NR (6.74, NR) ORR=Objective Response Rate; DCR=Disease Control Rate=CR/PR/SD; mPFS=Median Progression Free Survival; mOS=Median Overall Survival Clinical Activity in Overall Gastric Cancer Population * * ** * * ** * * * * * -100 -90 -80 -70 -60 -50 -40 -30 -20 -10 0 10 20 30 40 50 60 70 80 90 100 110 120 130 Change from Baseline (%) Best target lesion change from baseline (%) HER-2 IHC 3+: ORR: 32.7% (18/55) DCR: 69.1% (38/55) Median PFS: 4.7 months (95% CI: 2.7, 7.49) Median OS: NR (95% CI: 12.5, NR) HER 2 (IHC 2+) HER2 (IHC 3+) Ongoing * -100 -80 -60 -40 -20 20 40 60 80 100 120 140 160 180 200 Change from Baseline (%) Weeks Since Treatment Initiation 0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 Gastric Cancer GEJ First New Lesion Ongoing 0 Data cut-off January 8, 2019. Patients who received at least one marge and pembro dose in expansion phase, and had baseline measurable disease and at least one post-baseline disease assessment. Duration of Treatment in Overall Gastric Cancer Population Evaluable Patients Weeks 0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 IHC 2+ IHC 3+ Patient remains on therapy *Data cut-off January 8, 2019 Objective Response Observed Irrespective of Fc Receptor Genotype Outcomes for trastuzumab-treated patients who carry lower-affinity CD16A-F allele are worse than those who are homozygous for higher affinity V allele 11 Fc receptor genotyping results available in all GC patients Objective responses and disease stabilization observed in patient subsets irrespective of Fc receptor genotype FcγRIII (CD16A) Genotype Prevalence %(n) PR SD PD NE F/F 42.6% (26) 34.6% (9) 34.6% (9) 26.9% (7) 3.8% (1) V/F 45.9% (28) 25.0% (7) 39.3% (11) 32.1% (9) 3.6% (1) V/V 11.5% (7) 28.6% (2) 14.3% (1) 57.1% 4 NCT02689284 Methods HER2+ (archival), PD-L1-unselected 2 nd line GEA pts post trastuzumab Primary endpoints: Safety, tolerability, overall response rate (ORR) Secondary endpoints: Progression-free survival (PFS) and overall survival (OS); PFS and OS at 6 months Exploratory endpoints: Disease control rate (DCR) = proportion of patients with complete response (CR) + partial response (PR) + stable disease (SD) HER2-expression (post-trastuzumab) was confirmed by NGS of circulating- tumor DNA (ctDNA) for ERBB2 amp (Guardant360 ® ) PD-L1 tested on archival tissue by immunohistochemistry (IHC; Clone 22C3 pharmDx); Combined Positive Score using a provisional CPS 1 cut point Results Demographics Ninety-two patients have been treated at recommended Phase 2 dose (RP2D); 61 gastric adenocarcinoma (GC) and 31 in gastroesophageal junction adenocarcinoma (GEJ) Characteristic GC (n=61) GEJ (n=31) Age Mean ± SD 61.4 ± 13.6 57.9 ± 11.1 Median (Range) 62.0 (19.0, 85.0) 60.0 (35.0, 79.0) Gender [n (%)] Male 48 (78.7) 27 (87.1) Female 13 (21.3) 4 (12.9) Race [n (%)] Asian 48 (78.7) 3 (9.7) White 9 (14.8) 25 (80.6) Other 1 (1.6) 3 (9.7) Black or African American 3 (4.9) 0 ECOG Status [n (%)] 0 20 (32.8) 13 (41.9) 1 41 (67.2) 18 (58.1) *Data cutoff January 8, 2019. Treatment with Combination of Margetuximab and Pembrolizumab is Well-tolerated 64% of patients experienced treatment related AE (TRAE) irrespective of grade 18% of patients with TRAE ≥Grade 3 Most common TRAE: pruritis (16.8%) 8 Treatment-related serious adverse events: autoimmune hepatitis [2], hyponatremia [1], diabetic ketoacidosis [1], and pneumonitis [1], hypotension [1], confusional state [1], dizziness [1] 17 Adverse events of special interest reported: infusion related reaction [11], autoimmune hepatitis [2], pneumonitis [1], endocrinopathy [1], others [1], LVEF dysfunction [1] Adverse Event All Related AE All (N=95)* ≥ Gr 3 TOTAL 61 (64.2) 17 (17.9) Pruritus 16 (16.8) Diarrhea 14 (14.7) Infusion related reaction 13 (13.7) 3 (3.2) Fatigue 12 (12.6) Rash 7 (7.4) Rash maculo-papular 5 (5.3) Anemia 5 (5.3) 2 (2.1) Nausea 4 (4.2) 1 (1.1) Decreased appetite 4 (4.2) Lipase increased 4 (4.2) 1 (1.1) Aspartate aminotransferase increased 4 (4.2) 1 (1.1) Chills 3 (3.2) Alanine aminotransferase increased 3 (3.2) Amylase increased 3 (3.2) 2 (2.1) Hyperthyroidism 3 (3.2) Adrenal insufficiency 3 (3.2) Vomiting 2 (2.1) 1 (1.1) Pyrexia 2 (2.1) Pain 2 (2.1) Ejection fraction decreased 2 (2.1) Blood alkaline phosphatase increased 2 (2.1) 1 (1.1) Pneumonitis 2 (2.1) 1 (1.1) Hypotension 2 (2.1) 1 (1.1) Autoimmune hepatitis 2 (2.1) 2 (2.1) Data cut off January 8, 2019; Events occurring >2% pts; includes all pts treated on study. Enhanced Immune Cell Infiltrate Following Treatment with Margetuximab/Pembrolizumab in a Patient with Subsequent Complete Response 1 3 5 7 9 11 13 15 17 19 -100 -50 0 50 100 Treatment Cycles A B A B 0.00 0.05 0.10 0.15 0.20 2 4 6 8 10 12 14 16 18 20 22 Mean % (positive/total area [μm 2 ]) CD3+ CD16+ CD56+ 0 2 4 6 8 Combined Positive Score Tumor Volume (% change from Baseline) Pre-treatment (C1D1) Post-treatment (C2D1) A B Panel 1 Panel 3 Panel 4 Panel 2 HER2 Panel 5 Panel 6 CD3/CD16/CD56 PD-L1 A. B. A. B. Panel 1: Waterfall plot of percent tumor volume change from baseline through different treatment cycle for patient on study. Red arrow represent timing of tumor biopsy, A (pre-treatment C1D1), B (post-treatment C2D1). Panel 2–4: IHC staining for samples A and B for HER2, CD3 T-cells, CD16 NK cells, CD56 NK cells and PDL1, respectively. Panel 5–6: Quantitation of IHC staining for CD3/16/56 and PDL1, respectively, shows increase in immune cell infiltration and activation in TME upon dosing with margetuximab and pembrolizumab. Conclusions Margetuximab + pembrolizumab is a chemotherapy-free combination, designed to coordinately engage innate and adaptive immunity for treatment of GEA The investigational combination demonstrated an acceptable safety profile that benchmarked favorably to historical experience with SoC with ramucirumab+/-chemotherapy; ≥Grade 3 TRAEs in 18% of treated patients, and events are consistent with margetuximab or pembrolizumab alone Results from the cohort expansion population and ongoing follow-up confirm the anti-tumor activity of the investigational combination of margetuximab and pembrolizumab, and benchmarked favorably to historical experience with single agent checkpoint inhibitors Paired biopsy of tissue from a patient with a complete response demonstrates enhanced infiltration of both innate (CD16/CD56 NK cells) and adaptive (CD3 T-cells) immune cell subsets Prospective patient selection by HER2 (IHC3+) and/or PD-L1 expression may further enrich for patients more likely to respond to treatment with margetuximab plus pembrolizumab Combined administration of Fc-optimized antibodies and checkpoint inhibitors could be an important novel strategy to coordinately engage innate and adaptive immunity, and extend the activity of cancer immunotherapy beyond that achieved with single agent checkpoint inhibition alone References 1. J Clin Oncol 35, 2017 (suppl 4S; abstract 12). 2. J Clin Oncol 34, 2016 (suppl; abstr 4043). 3. J Clin Oncol 34, 2016 (suppl; abstr 11608). 4. J Clin Oncol 35, 2017 (suppl 4S; abstract 27). 5. J Clin Oncol 35, 2017 (suppl 4S; abstract 81). 6. Makiyama et al. Journal of Clinical Oncology 36, no. 15_suppl (May 2018) 4011-4011. 7. Stagg J, et al. Anti–ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti–PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA. 2011 Apr 26; 108(17): 7142–7147. 8. Data presented at SITC 2018, 4030. 9. Hsu, et al., J Clin Invest, 2018. 10. Unpublished. 11. Presented at SITC 2018, P338. 12. Musolino A, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26:1789-96. This study was sponsored by MacroGenics, Inc. Copies of this poster obtained through QR (Quick Response) and/or text key codes are for personal use only and may not be reproduced without written permission of the authors. Clinical Activity in Expansion Cohort Only Population * * ** * * ** * * * * * * * ORR: 22.4% (19/85) DCR: 57.7% (49/85) Median PFS: 2.7 months (95% CI: 21.6, 4.7) Median OS: 13.3 months (95% CI: 9.23, 18.0) Gastric Cancer GEJ Ongoing * -100 -90 -80 -70 -60 -50 -40 -30 -20 -10 0 10 20 30 40 50 60 70 80 90 100 110 120 130 Change from Baseline (%) Best target lesion change from baseline (%) -100 -80 -60 -40 -20 0 20 40 60 80 100 120 140 160 180 200 Change from Baseline (%) Weeks Since Treatment Initiation 0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 Gastric Cancer GEJ First New Lesion Ongoing Data cut-off January 8, 2019. Patients who received at least one marge and pembro dose in expansion phase, and had baseline measurable disease and at least one post-baseline disease assessment. Duration of Treatment in Overall Cohort Expansion Population Evaluable Patients Weeks 0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 Gastric Cancer GEJ Patient remains on therapy *Data cut-off January 8, 2019 Overall Response Rates and Biomarker Incidence (RP2D Cohorts) All Patients* Gastric Cancer GEJ Cancer Objective Response Rate 20/92 (21.7%) 18/61 (29.5%) 2/31 (6.5%) Biomarker Incidence HER2 IHC3+ 71/92 (77.2%) 55/61 (90.2%) 16/31 (51.6%) ERBB2 amp 48/82 (58.5%) 35/56 (62.5%) 13/26 (50.0%) PD-L1+ 33/76 (43.4%) 26/54 (48.1%) 7/22 (31.8%) HER2 IHC3+/PD-L1+ 25/76 (32.9%) 23/54 (42.6%) 2/22 (9.1%) Data cut-off January 8, 2019; *Includes only patients evaluated per assay. Objective responses in 21.7% (20/92) of HER-2+ GEA patients 15 confirmed/5 unconfirmed responses, 15 patients ongoing Responses are higher in GC 29.5% vs GEJ 6.5% Includes initial cohort expansion as well as subsequent IHC-3+ GC cohort expansion Approximately 77% of pts were HER2 IHC3+ at baseline (archival) sample Baseline HER2 IHC3+ is higher in GC (90.2%) vs GEJ (51.6%) patients Approximately 60% of patients tested retained HER2 expression post-trastuzumab ( ERBB2 amp ctDNA), and 43% of patients tested were PD-L1+ by IHC Higher expression of PD-L1 and ERBB2 amp was observed in patients with GC (43%) vs GEJ (9%) Data cut-off January 8, 2019; *Includes only patients evaluated per assay. Biomarker Distribution in GC vs GEJ Populations Total Population Archival HER2 IHC 3+ ERBB2 amp PD-L1+ GC (n=61) GEJ (n=31) 25% 2% 28% 5% 15% 23% 0% 23% 16% 23% 0% 3% 3% 16%
1

Antitumor Activity of Margetuximab plus …...Antitumor Activity of Margetuximab plus Pembrolizumab in Patients with Advanced HER2+ (IHC3+) Gastric Carcinoma Daniel V.T. Catenacci1,

Aug 22, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Antitumor Activity of Margetuximab plus …...Antitumor Activity of Margetuximab plus Pembrolizumab in Patients with Advanced HER2+ (IHC3+) Gastric Carcinoma Daniel V.T. Catenacci1,

Antitumor Activity of Margetuximab plus Pembrolizumab in Patients with Advanced HER2+ (IHC3+) Gastric CarcinomaDaniel V.T. Catenacci1, Haeseong Park2, Hope Elizabeth Uronis3, Yoon-Koo Kang4, Matthew C.H. Ng5, Philip Jordan Gold6, Peter C. Enzinger7, Keun Wook Lee8, Jill Lacy9, Se Hoon Park10, Jennifer Yen11, Justin Odegaard12,

Aleksandra Franovic11, Sarah Church15, Anushka DeCosta13, Jan E. Baughman13, Aisha Wynter-Horton13, Francine Chen13, Jeff Nordstrom13 , Paul A. Moore13, Tony Wu13, Jan Kenneth Davidson-Moncada13, Yung-Jue Bang14

1University of Chicago Medical Center and Biological Sciences, Chicago, IL; 2Washington University School of Medicine, St. Louis, MO; 3Duke University Medical Center, Durham, NC; 4Asan Medical Center, Seoul, South Korea; 5National Cancer Centre Singapore, Singapore; 6Swedish Cancer Institute, Seattle, WA; 7Dana-Farber Cancer Institute, Boston, MA; 8Seoul National University Bundang Hospital, Seongnam, South Korea; 9Smilow Cancer Hospital, Yale University, New Haven, CT; 10Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; 11Guardant Health, Inc., Redwood City, CA;

12Lifecode, San Francisco, CA; 13MacroGenics, Inc., Rockville, MD; 14Seoul National University Hospital, Seoul, South Korea

Abstract #65

Presented at The 2019 Gastrointestinal Cancers Symposium, January 17–19, 2019, San Francisco, CA [email protected]

Background■■ Trastuzumab + chemotherapy is standard treatment in 1st line advanced HER2+ gastroesophageal adenocarcinoma (GEA)

– No HER2-targeted agents have been approved in post-trastuzumab setting in patients with HER2+ GEA

■■ Loss of HER2 expression after trastuzumab has been reported in up to 70% of GEA patients with potential consequences for subsequent treatment with HER2-targeted agents1-5

■■ Margetuximab is a next generation anti-HER2 monoclonal antibody featuring an optimized Fc domain designed to enhance its Fc-dependent functions, including antibody dependent cell cytotoxicity (ADCC) irrespective of the host’s FcγRIIIa (CD16A) genotype

Hypothesis: Coordinate engagement of innate and adaptive immunity with margetuximab and anti-PD-1 mediates greater antitumor activity than either single agent alone■■ Coordinate engagement of innate and adaptive immunity with combination of anti-HER2 and anti-PD-1 antibodies achieves greater antitumor activity than either agent alone in murine tumor models6

■■ Margetuximab and pembrolizumab have both demonstrated monotherapy antitumor activity in patients with GEA; and Checkpoint inhibitors (pembrolizumab, nivolumab) are approved for treatment of 3L PD-L1+ patients with GEA ■■ We reported previously that another Fc-optimized antibody (enoblituzumab, anti-B7H3) combined with pembrolizumab achieved greater antitumor activity than historical experience with checkpoint inhibitors alone in checkpoint-naïve patients with SCCHN and NSCLC (PD-L1<1%) (SITC 2018)7

■■ Preliminary data indicates that Fc-modified antibodies (including margetuximab and enoblituzumab monotherapy) can modulate T-cell repertoire in treated patients8-9

■■ NK cells may express PD-1, and PD-1/PD-L1 interaction can impair NK cell function, and PD-1/PD-L1 blockade can enhance NK cell function and preclinical antitumor activity10

Cancer Cells

TumorDestruction

Enoblituzumab

Anti-PD-1Antibody

NK Cells

Tumor Destruction

Macrophages

T CellsEnhanced Adaptive

T-cell-mediatedAntitumor Immunity

ExhaustedT Cells

Innate Immunity

Adaptive Immunity

EnhancedADCC

SensitizeT Cells

CounterT-cell

Exhaustion

Goal is to develop chemotherapy-free approach for treatment of GEA

Study Design

Dose EscalationPembro (200 mg) +

Margetuximab10 & 15 mg/kg

Gastric IHC 3+ ExpansionN = 25

RP2D: Margetuximab (15 mg/kg) +Pembrolizumab (200mg) IV Q3 WK

Enrollment completed. 15 patients ongoing

Cohort ExpansionN = 60

■■ Open label, dose escalation study, conventional 3+3 design■■ Escalating doses of margetuximab (marge; 10 mg/kg & 15 mg/kg)■■ Fixed dose pembrolizumab (pembro; 200 mg)■■ Response assessed by RECIST & irRECIST■■ 92 Patients treated at recommended Phase 2 dose (RP2D)

– 61 Gastric cancer (GC) – 35 Enrolled in cohort 2 (1), and cohort expansion (34) (HER2 IHC2+/3+) – 26 Enrolled in HER2 (IHC3+) GC specific cohort

– 31 Gastroesophageal junction (GEJ) (HER2 IHC2+/3+)

Efficacy Results in Gastric Cancer Population by Biomarker Expression

n ORR DCR mPFS mOS

Total 61 29.5% (18/61)

65.6% (40/61)

4.07 (2.30, 5.45)

14.62 (9.07, NR)

IHC3+ 55 32.7% (18/55)

69.1% (38/55)

4.70 (2.66, 7.49) NR (12.48, NR)

ERBB2amp 35 40.0% (14/35)

77.1% (27/35)

4.76 (2.69, 7.59)

14.62 (8.41, NR)

PDL1+ 26 46.2% (12/26)

80.8% (21/26)

4.14 (2.60, 7.59)

NR (6.74, NR)

IHC3+/PDL1+ 23 52.2%

(12/23)82.6% (12/23)

4.14 (2.60, 15.54)

NR (6.74, NR)

ORR=Objective Response Rate; DCR=Disease Control Rate=CR/PR/SD; mPFS=Median Progression Free Survival; mOS=Median Overall Survival

Clinical Activity in Overall Gastric Cancer Population

* *

* * * ** *

** *

*

*-100

-90-80-70-60-50-40-30-20-10

0102030405060708090

100110120130

Chan

ge fr

om B

asel

ine

(%)†

Best target lesion change from baseline (%)

HER-2 IHC 3+: ORR: 32.7% (18/55)DCR: 69.1% (38/55) Median PFS: 4.7 months (95% CI: 2.7, 7.49)Median OS: NR (95% CI: 12.5, NR)

HER 2 (IHC 2+)HER2 (IHC 3+)Ongoing*

-100

-80

-60

-40

-20

20

40

60

80

100

120

140

160

180

200

Chan

ge fr

om B

asel

ine

(%)

Weeks Since Treatment Initiation0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120

Gastric CancerGEJ

First New LesionOngoing

0

Data cut-off January 8, 2019. †Patients who received at least one marge and pembro dose in expansion phase, and had baseline measurable disease and at least one post-baseline disease assessment.

Duration of Treatment in Overall Gastric Cancer Population

Eval

uab

le P

atie

nts

Weeks0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100

IHC 2+IHC 3+Patient remains on therapy

*Data cut-off January 8, 2019

Objective Response Observed Irrespective of Fc Receptor Genotype■■ Outcomes for trastuzumab-treated patients who carry lower-affinity CD16A-F allele are worse than those who are homozygous for higher affinity V allele11

■■ Fc receptor genotyping results available in all GC patients ■■ Objective responses and disease stabilization observed in patient subsets irrespective of Fc receptor genotype

FcγRIII (CD16A) Genotype Prevalence %(n) PR SD PD NE

F/F 42.6% (26) 34.6% (9) 34.6% (9) 26.9% (7) 3.8% (1)

V/F 45.9% (28) 25.0% (7) 39.3% (11) 32.1% (9) 3.6% (1)

V/V 11.5% (7) 28.6% (2) 14.3% (1) 57.1% 4

NCT02689284

Methods■■ HER2+ (archival), PD-L1-unselected 2nd line GEA pts post trastuzumab

– Primary endpoints: – Safety, tolerability, overall response rate (ORR)

– Secondary endpoints: – Progression-free survival (PFS) and overall survival (OS); PFS and OS at 6 months

– Exploratory endpoints: – Disease control rate (DCR) = proportion of patients with complete response (CR) + partial response (PR) + stable disease (SD)

■■ HER2-expression (post-trastuzumab) was confirmed by NGS of circulating-tumor DNA (ctDNA) for ERBB2 amp (Guardant360®)■■ PD-L1 tested on archival tissue by immunohistochemistry (IHC; Clone 22C3 pharmDx); Combined Positive Score using a provisional CPS 1 cut point

Results

DemographicsNinety-two patients have been treated at recommended Phase 2 dose (RP2D); 61 gastric adenocarcinoma (GC) and 31 in gastroesophageal junction adenocarcinoma (GEJ)

Characteristic GC (n=61) GEJ (n=31)

AgeMean ± SD 61.4 ± 13.6 57.9 ± 11.1

Median (Range) 62.0 (19.0, 85.0) 60.0 (35.0, 79.0)

Gender [n (%)]Male 48 (78.7) 27 (87.1)

Female 13 (21.3) 4 (12.9)

Race [n (%)]

Asian 48 (78.7) 3 (9.7)

White 9 (14.8) 25 (80.6)

Other 1 (1.6) 3 (9.7)

Black or African American 3 (4.9) 0

ECOG Status [n (%)] 0 20 (32.8) 13 (41.9)

1 41 (67.2) 18 (58.1)*Data cutoff January 8, 2019.

Treatment with Combination of Margetuximab and Pembrolizumab is Well-tolerated■■ 64% of patients experienced treatment related AE (TRAE) irrespective of grade■■ 18% of patients with TRAE ≥Grade 3 ■■ Most common TRAE: pruritis (16.8%)■■ 8 Treatment-related serious adverse events: autoimmune hepatitis [2], hyponatremia [1], diabetic ketoacidosis [1], and pneumonitis [1], hypotension [1], confusional state [1], dizziness [1]■■ 17 Adverse events of special interest reported: infusion related reaction [11], autoimmune hepatitis [2], pneumonitis [1], endocrinopathy [1], others [1], LVEF dysfunction [1]

Adverse EventAll Related AE

All (N=95)* ≥ Gr 3TOTAL 61 (64.2) 17 (17.9)Pruritus 16 (16.8)

Diarrhea 14 (14.7)

Infusion related reaction 13 (13.7) 3 (3.2)

Fatigue 12 (12.6)

Rash 7 (7.4)

Rash maculo-papular 5 (5.3)

Anemia 5 (5.3) 2 (2.1)

Nausea 4 (4.2) 1 (1.1)

Decreased appetite 4 (4.2)

Lipase increased 4 (4.2) 1 (1.1)

Aspartate aminotransferase increased 4 (4.2) 1 (1.1)

Chills 3 (3.2)

Alanine aminotransferase increased 3 (3.2)

Amylase increased 3 (3.2) 2 (2.1)

Hyperthyroidism 3 (3.2)

Adrenal insufficiency 3 (3.2)

Vomiting 2 (2.1) 1 (1.1)

Pyrexia 2 (2.1)

Pain 2 (2.1)

Ejection fraction decreased 2 (2.1)

Blood alkaline phosphatase increased 2 (2.1) 1 (1.1)

Pneumonitis 2 (2.1) 1 (1.1)

Hypotension 2 (2.1) 1 (1.1)

Autoimmune hepatitis 2 (2.1) 2 (2.1)Data cut off January 8, 2019; Events occurring >2% pts; includes all pts treated on study.

Enhanced Immune Cell Infiltrate Following Treatment with Margetuximab/Pembrolizumab in a Patient with Subsequent Complete Response

1 3 5 7 9 11 13 15 17 19

-100

-50

0

50

100

Treatment Cycles

A B A B0.000.050.100.150.20

2468

10121416182022

Mea

n %

(pos

itiv

e/to

tal a

rea

[µm

2 ])

CD3+ CD16+CD56+

0

2

4

6

8

Com

bine

d Po

siti

ve S

core

Tum

or V

olum

e(%

cha

nge

from

Bas

elin

e)Pr

e-tr

eatm

ent

(C1D

1)Po

st-t

reat

men

t(C

2D1)

AB

Panel 1 Panel 3 Panel 4

Panel 2HER2

Panel 5 Panel 6

CD3/CD16/CD56 PD-L1

A.

B.

A.

B.

Panel 1: Waterfall plot of percent tumor volume change from baseline through different treatment cycle for patient on study. Red arrow represent timing of tumor biopsy, A (pre-treatment C1D1), B (post-treatment C2D1).Panel 2–4: IHC staining for samples A and B for HER2, CD3 T-cells, CD16 NK cells, CD56 NK cells and PDL1, respectively.Panel 5–6: Quantitation of IHC staining for CD3/16/56 and PDL1, respectively, shows increase in immune cell infiltration and activation in TME upon dosing with margetuximab and pembrolizumab.

Conclusions■■ Margetuximab + pembrolizumab is a chemotherapy-free combination, designed to coordinately engage innate and adaptive immunity for treatment of GEA ■■ The investigational combination demonstrated an acceptable safety profile that benchmarked favorably to historical experience with SoC with ramucirumab+/-chemotherapy; ≥Grade 3 TRAEs in 18% of treated patients, and events are consistent with margetuximab or pembrolizumab alone ■■ Results from the cohort expansion population and ongoing follow-up confirm the anti-tumor activity of the investigational combination of margetuximab and pembrolizumab, and benchmarked favorably to historical experience with single agent checkpoint inhibitors■■ Paired biopsy of tissue from a patient with a complete response demonstrates enhanced infiltration of both innate (CD16/CD56 NK cells) and adaptive (CD3 T-cells) immune cell subsets ■■ Prospective patient selection by HER2 (IHC3+) and/or PD-L1 expression may further enrich for patients more likely to respond to treatment with margetuximab plus pembrolizumab ■■ Combined administration of Fc-optimized antibodies and checkpoint inhibitors could be an important novel strategy to coordinately engage innate and adaptive immunity, and extend the activity of cancer immunotherapy beyond that achieved with single agent checkpoint inhibition alone

References1. J Clin Oncol 35, 2017 (suppl 4S; abstract 12). 2. J Clin Oncol 34, 2016 (suppl; abstr 4043). 3. J Clin Oncol 34, 2016 (suppl; abstr 11608). 4. J Clin Oncol 35, 2017 (suppl 4S; abstract 27). 5. J Clin Oncol 35, 2017 (suppl 4S; abstract 81). 6. Makiyama et al. Journal of Clinical Oncology 36, no. 15_suppl (May 2018) 4011-4011. 7. Stagg J, et al. Anti–ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti–PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci USA. 2011 Apr 26; 108(17): 7142–7147. 8. Data presented at SITC 2018, 4030. 9. Hsu, et al., J Clin Invest, 2018. 10. Unpublished. 11. Presented at SITC 2018, P338. 12. Musolino A, et al. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26:1789-96.

This study was sponsored by MacroGenics, Inc. Copies of this poster obtained through QR (Quick Response) and/or text key codes are for personal use only and may not be reproduced without written permission of the authors.

Clinical Activity in Expansion Cohort Only Population

* ** * * *

* **

* **

*

**

ORR: 22.4% (19/85)DCR: 57.7% (49/85)Median PFS: 2.7 months (95% CI: 21.6, 4.7)Median OS: 13.3 months (95% CI: 9.23, 18.0)

Gastric CancerGEJOngoing*-100

-90-80-70-60-50-40-30-20-10

0102030405060708090

100110120130

Chan

ge fr

om B

asel

ine

(%)†

Best target lesion change from baseline (%)

-100

-80

-60

-40

-200

20

40

60

80

100

120

140

160

180

200

Chan

ge fr

om B

asel

ine

(%)

Weeks Since Treatment Initiation0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120

Gastric CancerGEJ

First New LesionOngoing

Data cut-off January 8, 2019. †Patients who received at least one marge and pembro dose in expansion phase, and had baseline measurable disease and at least one post-baseline disease assessment.

Duration of Treatment in Overall Cohort Expansion Population

Eval

uab

le P

atie

nts

Weeks0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100

Gastric CancerGEJPatient remains on therapy

*Data cut-off January 8, 2019

Overall Response Rates and Biomarker Incidence (RP2D Cohorts)

All Patients* Gastric Cancer GEJ CancerObjective Response Rate 20/92 (21.7%) 18/61 (29.5%) 2/31 (6.5%)

Biomarker IncidenceHER2 IHC3+ 71/92 (77.2%) 55/61 (90.2%) 16/31 (51.6%)

ERBB2amp 48/82 (58.5%) 35/56 (62.5%) 13/26 (50.0%)

PD-L1+ 33/76 (43.4%) 26/54 (48.1%) 7/22 (31.8%)

HER2 IHC3+/PD-L1+ 25/76 (32.9%) 23/54 (42.6%) 2/22 (9.1%)Data cut-off January 8, 2019; *Includes only patients evaluated per assay.

■■ Objective responses in 21.7% (20/92) of HER-2+ GEA patients – 15 confirmed/5 unconfirmed responses, 15 patients ongoing

■■ Responses are higher in GC 29.5% vs GEJ 6.5% – Includes initial cohort expansion as well as subsequent IHC-3+ GC cohort expansion

■■ Approximately 77% of pts were HER2 IHC3+ at baseline (archival) sample – Baseline HER2 IHC3+ is higher in GC (90.2%) vs GEJ (51.6%) patients

■■ Approximately 60% of patients tested retained HER2 expression post-trastuzumab (ERBB2amp ctDNA), and 43% of patients tested were PD-L1+ by IHC

– Higher expression of PD-L1 and ERBB2amp was observed in patients with GC (43%) vs GEJ (9%)Data cut-off January 8, 2019; *Includes only patients evaluated per assay.

Biomarker Distribution in GC vs GEJ Populations

Total PopulationArchival HER2 IHC 3+ERBB2amp

PD-L1+

GC (n=61) GEJ (n=31)

25%2%

28%

5%

15%

23%

0%

23%

16%23%

0%3%

3%

16%