Phase I Dose-finding Study of MIW815 (ADU-S100), an ......• Activation of the STING pathway...

1
Phase I Dose-finding Study of MIW815 (ADU -S100), an Intratumoral STING Agonist, in Patients With Advanced Solid Tumors or Lymphomas References 1. Woo SR, et al. Immunity 2014;41:830–842. 2. Glickman LH, et al. Cancer Res 2016;76(14 Suppl):abst 1445. 3. Corrales L, et al. Cell Rep 2015;11:1018–1030. 4. Cheson BD, et al. J Clin Oncol 2014;32:3059–3068. Funda Meric-Bernstam 1 , Theresa L. Werner 2 , F. Stephen Hodi 3 , Wells Messersmith 4 , Nancy Lewis 5 , Craig Talluto 6 , Mirek Dostalek 5 , Aiyang Tao 5 , Sarah M. McWhirter 7 , Damian Trujillo 7 , Jason J. Luke 8 1 Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX; 2 Huntsman Cancer Institute, University of Utah, Salt Lake City, UT; 3 Dana-Farber Cancer Institute, Boston, MA; 4 University of Colorado Cancer Center, Aurora, CO; 5 Novartis Pharmaceuticals Corporation, East Hanover, NJ; 6 Novartis Institutes for BioMedical Research, Cambridge, MA; 7 Aduro Biotech Inc., Berkeley, CA; 8 The University of Chicago Medicine, Chicago, IL This study is sponsored by Novartis Pharmaceuticals Corporation. Introduction The STimulator of INterferon Genes (STING) pathway plays a crucial role in the innate immune response to immunogenic tumors. Activation of the STING pathway increases interferon (IFN)-β production and induces the recruitment and priming of cluster of differentiation 8 (CD8)-positive T cells against tumor antigens. 1 MIW815 (ADU-S100) is a novel synthetic cyclic dinucleotide that activates the STING pathway in vitro ( Figure 1 ). 2 In preclinical tumor mouse models, intratumoral injection of MIW815 (ADU-S100) resulted in tumor regression in both injected and non-injected lesions. 3 This first-in-human Phase I study was designed to evaluate the safety and efficacy of MIW815 (ADU-S100) in patients with advanced solid tumors or lymphomas. In addition, MIW815 (ADU-S100) is currently being investigated in combination with anti-programmed cell death protein 1 (PD-1; NCT03172936) or anti-cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4; NCT02675439) antibodies in Phase I clinical trials in patients with advanced solid tumors and lymphomas. Figure 1. MIW815 (ADU-S100) Mechanism of Action IFN, interferon; STING, stimulator of interferon genes. Methods Study design This is a multicenter, open-label, first-in-human, Phase I study of MIW815 (ADU-S100) in patients with advanced solid tumors or lymphomas currently in dose escalation (Figure 2; NCT02675439; EudraCT 2015-005245-31). Primary objectives Safety and tolerability of MIW815 (ADU-S100). Recommended dose for future studies. Secondary objectives Preliminary antitumor activity of MIW815 (ADU-S100). Pharmacokinetics (PK) and pharmacodynamics (PD) of MIW815 (ADU-S100). Exploratory objective Biomarkers of response to MIW815 (ADU-S100). Patients are treated with weekly intratumoral injections of MIW815 (ADU-S100; 50–3200 μg) on a 3-weeks-on/1-week-off schedule until they experience unacceptable toxicity, disease progression per immune-related response criteria (irRC) for solid tumors, or confirmed progressive disease in lymphoma, and/or patient/investigator decision. Figure 2. MIW815X2101 Study Design Dose Escalation MTD/ RDE Solid tumors and lymphomas MIW815 (ADU-S100), weekly IT injection (3-weeks-on/1-week-off) 100 μg n=6 50 μg n=6 400 μg n=5 800 μg n=6 3200 μg n=6 1600 μg n=7 200 μg n=5 Planned Dose Expansion Group 1: Accessible UV-induced cancers, including melanoma, SCC, and BCC Group 2: Other accessible solid tumors and lymphomas, including CTCL, Merkel cell carcinoma, and HNSCC BCC, basal cell carcinoma; CTCL, cutaneous T-cell lymphoma; HNSCC, head and neck squamous cell carcinoma; IT, intratumoral; MTD, maximum tolerated dose; RDE, recommended dose for expansion; SCC, squamous cell carcinoma; UV, ultraviolet. Patient population Key inclusion criteria Adult patients with advanced/metastatic solid tumors or lymphomas who have progressed despite standard therapy, are intolerant to standard therapy, or for whom standard therapy is not reasonably effective or does not exist. Measurable disease per Response Evaluation Criteria In Solid Tumors (RECIST) v1.1 or Cheson 2014 criteria. 4 At least two distinct lesions are required; both accessible for baseline and on-treatment biopsies, with patient consent. Lesion 1 must measure ≥10 to <100 mm in longest diameter, and be accessible for repeated intratumoral injection. Eastern Cooperative Oncology Group performance status of ≤1. Prior immunotherapy is permitted. Key exclusion criteria Symptomatic central nervous system (CNS) metastases or CNS metastases requiring local CNS-directed therapy. Systemic anticancer therapy within 4 weeks of the first dose of study treatment. Treatment with systemic steroid therapy, other than replacement-dose steroids in the setting of adrenal insufficiency. Systemic treatment with any immunosuppressive medication. History of, or active, autoimmune disease with the exception of vitiligo or resolved childhood asthma/atopy. Assessments Adverse events (AEs) were assessed at every visit according to the Common Terminology Criteria for Adverse Events v4.03. Tumor response was determined locally according to irRC, RECIST v1.1, and Cheson 2014 criteria 4 for lymphoma, at screening and on Day (D) 1 of Cycle (C) 3, every 8 weeks up to C11, and then every 12 weeks until disease progression or patient withdrawal. Blood samples were collected for PK analysis on C1D1, C1D15, and C3D1, and for PD assessment of cytokine levels on C1D1, C1D8, C1D15, and C3D1. Results Patient characteristics and disposition As of August 16, 2018, 41 patients received MIW815 (ADU-S100) and 4 (9.8%) patients were continuing on treatment. Treatment was discontinued in 37 (90.2%) patients due to disease progression (n=29; 70.7%), physician decision (n=4; 9.8%), patient decision (n=3; 7.3%), and death (n=1; 2.4%). Baseline patient demographics are shown in Table 1. The number of patients treated at each MIW815 (ADU-S100) dose level is shown in Figure 2. A summary of the time on MIW815 (ADU-S100) treatment and patient response is shown in Figure 3. Table 1. Baseline Patient Demographics Characteristic All Patients (N=41) Median age, years (range) 62 (26–80) Sex, n (%) Male 18 (43.9) Female 23 (56.1) Race, n (%) Caucasian 27 (65.9) Black 6 (14.6) Asian 2 (4.9) Other/unknown 6 (14.6) ECOG PS, n (%) 0 11 (26.8) 1 30 (73.2) Prior therapy with a checkpoint inhibitor, n (%) Yes 22 (53.7) No 19 (46.3) Number of prior regimens, n (%) 0 3 (7.3) 1 4 (9.8) ≥2 34 (82.9) ECOG PS, Eastern Cooperative Oncology Group performance status. Figure 3. Time on MIW815 (ADU-S100) Treatment and Response Evaluation Ongoing Response PR SD PD Yes Prior checkpoint 50 μg 100 μg 200 μg 400 μg 800 μg 1600 μg Duration of exposure (months) MIW815 (ADU-S100) dose Breast cancer (TNBC) Cutaneous melanoma Ovarian cancer Colorectal cancer ER+/HER2– breast cancer Colorectal cancer Anal cancer Esophageal cancer Merkel cell carcinoma Non-Hodgkin lymphoma 0 2 4 6 8 10 3200 μg Non-Hodgkin lymphoma Cutaneous melanoma Esophageal cancer Breast cancer (TNBC) Melanoma Melanoma Non-Hodgkin lymphoma Melanoma Melanoma Uveal melanoma Hodgkin lymphoma Merkel cell carcinoma Non-Hodgkin lymphoma Uveal melanoma Breast cancer (ER+/HER2+) Melanoma Pancreatic cancer Breast cancer (TNBC) Ovarian cancer Other (adenocystic carcinoma) Angiosarcoma Melanoma Other (leiomyosarcoma) Colorectal cancer Other (sebaceous gland adenocarcinoma) Endometrial cancer Head and neck cancer (parotid gland) Sarcoma Merkel cell carcinoma Renal carcinoma (collecting duct)* Other (epithelioid hemangioepithelioma) Data cut-off: August 16, 2018. *Patient ongoing treatment at 14 months. ER+, estrogen receptor-positive; HER2–, human epidermal growth factor receptor 2-negative; HER2+, HER2-positive; PD, progressive disease; PR, partial response; SD, stable disease; TNBC, triple-negative breast cancer. Safety and tolerability AEs suspected to be related to MIW815 (ADU-S100) treatment were reported in 78.0% of patients; 12.2% of patients experienced Grade 3/4 AEs. The most common suspected related AEs (in ≥10% of all patients) were headache, injection site pain, and pyrexia (in 14.6% of patients each). Elevated lipase was the only Grade 3/4 suspected related AE reported in >1 patient (n=2; 4.9%). Treatment-emergent serious AEs were reported in 1 patient (Grade 3 dyspnea and Grade 4 respiratory failure; dose level: 1600 μg); this was related to underlying disease progression. There were no dose-limiting toxicities (DLTs) observed during the first cycle of treatment and no patients discontinued treatment due to an AE. Pharmacokinetics The mean plasma concentration–time profiles for MIW815 (ADU-S100) on C1D1, C1D15, and C3D1 are shown in Figure 4. MIW815 (ADU-S100) absorption from the intratumoral injection site was rapid, reaching maximal plasma concentration within minutes of dosing. MIW815 (ADU-S100) exposure generally increased in a dose-dependent manner. The apparent clearance of the MIW815 (ADU-S100) from systemic circulation was rapid with an observed terminal half-life of approximately 10–23 minutes. Figure 4. Concentration–Time Profiles for MIW815 (ADU-S100) on C1D1, C1D15, and C3D1 Mean (SD) MIW815 (ADU-S100) plasma concentration (ng/mL) Time (hours) MIW815 dose: C1D15 0 0.5 1 2 4 C3D1 0 0.5 1 2 4 C1D1 0.01 0.3 10 20 50 100 0 0.5 1 2 4 50 μg 100 μg 200 μg 400 μg 800 μg 1600 μg 3200 μg Data cut-off: August 16, 2018. C, Cycle; D, Day; SD, standard deviation. Patient vignette – Case example 1 Patient and disease characteristics 63-year-old male with Stage IV collecting duct carcinoma with metastases to the left supraclavicular lymph node (LN) and left neck. Treatment history: antineoplastic therapy with an investigational mouse double minute 2 homolog (MDM2) inhibitor and prior right radical nephrectomy and retroperitoneal LN dissection. Experience on study Treatment: weekly intratumoral injections of 800 μg MIW815 (ADU-S100; 3-weeks-on/1-week-off). Reported toxicities: Grade 1 headache, nausea, and hyperthyroidism/Graves’ disease. Best overall response (BOR): stable disease (SD); patient remains on study at C14. On-study pharmacodynamics Increases in systemic cytokine levels, including interleukin 6 (IL6), were observed following MIW815 (ADU-S100) injection on C1D1, C1D8, and C3D1 (Figure 5A). Treatment with MIW815 (ADU-S100) resulted in a >2-fold increase in expression of the IFN-γ , programmed death-ligand 1 (PD-L1), and CD8A genes, and the natural killer (NK) cell gene set in the injected lesion (Figure 5B). An increase in CD8-positive tumor-infiltrating lymphocytes (TILs) and PD-L1 stromal staining was observed in the injected lesion following MIW815 (ADU-S100) treatment (Figure 5C). Figure 5. Case Example 1: Pre- and Post-MIW815 (ADU-S100) Therapy Cytokine Levels, RNA, and IHC in a Patient With Collecting Duct Carcinoma A. Fold Change in Systemic Cytokine Levels on C1D1, C1D8, and C3D1 Time of assessment C1D1 Fold change 0 2.5 7.5 5 15 Pre-dose 2 hours 6 hours Pre-dose 2 hours 6 hours Pre-dose 2 hours 6 hours C1D8 C3D1 Cytokine IFN-β1 IL6 IL8 MCP1 MIP1B C, Cycle; D, Day; IFN, interferon; IHC, immunohistochemistry; IL6, interleukin 6; IL8, interleukin 8; MCP1, monocyte chemoattractant protein 1; MIP1B, macrophage inflammatory protein 1 beta. B. Fold Change in Intratumoral RNA Expression in the Injected and Non-injected Lesion From Screening to C3D1 (Pre-dose) Injected Lesion –2 JAK1 STAT1 IFN-γ PD-L1 CD8A NK cell gene set IFN-γ gene set JAK1 STAT1 IFN-γ PD-L1 CD8A NK cell gene set IFN-γ gene set –1 0 1 2 Log2 fold change Gene Non-injected Lesion 0.0 0.5 1.0 1.5 Log2 fold change *NK cell gene set includes: IL12A, IL12B, KIR3DL1, KIR3DL2, KIR3DL3, KLRB1, KLRC1, KLRC2, KLRD1, KLRF1, KLRG1, KLRK1 , and NCR1; IFN-γ gene set includes: TBK1, IRF3, IFNAR1, JAK1, JAK2, TYK2, STAT1, STAT2, STAT3, IFNG, IFNGR1, IDO1, IL15RAm CXCL9, CXCL10, CXCL11 , and CD274. C, Cycle; CD8A, cluster of differentiation 8A; D, Day; IFN, interferon; JAK1, Janus kinase 1; NK, natural killer; PD-L1 , programmed death-ligand 1; STAT1 , signal transducer and activator of transcription 1. C. IHC Micrographs of the MIW815 (ADU-S100) Injected and Non-injected Lesion at Screening and C3D1 (Pre-dose) H&E Screening C3D1 Injected Lesion CD8* CD68* FOXP3* PD-L1 H&E CD8* CD68* FOXP3* PD-L1 Screening C3D1 2.55% 2.55% 0.39% PD-L1-negative stromal staining PD-L1-positive stromal staining PD-L1-negative stromal staining PD-L1 negative stromal staining 9.57% 5.64% 1.44% 16.94% 10.38% 4.05% 17.08% 7.07% 1.26% Non-injected Lesion *Biomarker area % per image analysis; Tumor positive score % per pathologist analysis; PD-L1 measured using the IHC 22C3 pharmDx assay. C, Cycle; CD, cluster of differentiation; D, Day; FOXP3, forkhead box P3; H&E, hematoxylin and eosin; IHC, immunohistochemistry; PD-L1, programmed death-ligand 1. Patient vignette – Case example 2 Patient and disease characteristics 68-year-old male with esophageal cancer and metastases to the mediastinal LN, supraclavicular LN, and adrenal gland. Treatment history: neoadjuvant paclitaxel, carboplatin, and radiation therapy followed by tumor resection, adjuvant 5-fluorouracil and oxaliplatin chemotherapy, and for metastatic disease cisplatin/irinotecan, nivolumab, and investigational anti-matrix metallopeptidase-9 antibody therapy. Experience on study Treatment: weekly intratumoral injections of 800 μg MIW815 (ADU-S100; 3-weeks-on/1-week-off). Reported toxicities: Grade 1 injection site reaction and exertional dyspnea; patient also experienced an incarcerated bowel (unrelated to study drug). BOR: SD for 3 cycles. On-study pharmacodynamics IFN-β1 levels increased following MIW815 (ADU-S100) injection on C1D1, C1D8, C1D15, and C3D1 (Figure 6A). Expression of the T-cell differentiation antigen CD8A gene and the NK cell gene set were increased >2-fold in the injected lesion as a result of MIW815 (ADU-S100) treatment (Figure 6B). Following MIW815 (ADU-S100) injection, an increase in the staining of CD8-positive TILs and PD-L1 stromal staining was observed in the injected lesion (Figure 6C). Figure 6. Case Example 2: Pre- and Post-MIW815 (ADU-S100) Therapy Cytokine Levels, RNA, and IHC in a Patient With Esophageal Cancer A. Fold Change in Systemic Cytokine Levels on C1D1, C1D8, C1D15, and C3D1 C1D1 Fold change 1 2 3 4 Pre-dose 2 hours 6 hours Pre-dose 2 hours 6 hours Pre-dose 2 hours 6 hours Pre-dose 2 hours 6 hours C1D8 C1D15 C3D1 Cytokine IFN-β1 IL6 IL8 MCP1 MIP1B Time of assessment C, Cycle; D, Day; IFN, interferon; IHC, immunohistochemistry; IL6, interleukin 6; IL8, interleukin 8; MCP1, monocyte chemoattractant protein 1; MIP1B, macrophage inflammatory protein 1 beta. B. Fold Change in Intratumoral RNA Expression in the Injected and Non-injected Lesion From Screening to C3D1 (Pre-dose) Gene Injected Lesion JAK1 STAT1 IFN-γ PD-L1 CD8A IFN-γ gene set NK cell gene set JAK1 STAT1 IFN-γ PD-L1 CD8A IFN-γ gene set NK cell gene set –1 0 1 2 Log2 fold change Non-injected Lesion –2 0.0 0.5 1.0 Log2 fold change *NK cell gene set includes: IL12A, IL12B, KIR3DL1, KIR3DL2, KIR3DL3, KLRB1, KLRC1, KLRC2, KLRD1, KLRF1, KLRG1, KLRK1 , and NCR1; IFN-γ gene set includes: TBK1, IRF3, IFNAR1, JAK1, JAK2, TYK2, STAT1, STAT2, STAT3, IFNG, IFNGR1, IDO1, IL15RAm CXCL9, CXCL10, CXCL11 , and CD274. C, Cycle; CD8A, cluster of differentiation 8A; D, Day; IFN, interferon; JAK1 , Janus kinase 1; NK, natural killer; PD-L1 , programmed death-ligand 1; STAT1 , signal transducer and activator of transcription 1. C. IHC Micrographs of the MIW815 (ADU-S100) Injected and Non-injected Lesion at Screening and C3D1 (Pre-dose) Screening C3D1 Injected Lesion Non-injected Lesion H&E Screening C3D1 CD8* CD68* FOXP3* PD-L1 H&E CD8* CD68* FOXP3* PD-L1 3.9% 11.63% 0.66% PD-L1-negative stromal staining PD-L1-positive stromal staining PD-L1-negative stromal staining PD-L1-negative stromal staining 19.97% 12.6% 2.68% 7.59% 25.31% 2.29% 9.85% 14.91% 1.31% *Biomarker area % per image analysis; Tumor positive score % per pathologist analysis; PD-L1 measured using the IHC 22C3 pharmDx assay. C, Cycle; CD, cluster of differentiation; D, Day; FOXP3, forkhead box P3; H&E, hematoxylin and eosin; IHC, immunohistochemistry; PD-L1, programmed death-ligand 1. Conclusions Single-agent MIW815 (ADU-S100) was well tolerated at doses ranging from 50 to 3200 μg in patients with advanced solid tumors and lymphoma with no DLTs noted. Dose escalation is ongoing. Preliminary signs of MIW815 (ADU-S100) biological activity were observed in patients with advanced solid tumors and lymphoma, including those who had previously received treatment with a checkpoint inhibitor. In the highlighted patient cases, the ability of MIW518 (ADU-S100) to activate the antitumor immune response is suggested by observed increases in CD8-positive TILs, genes involved in the antitumor immune response, and systemic cytokines following MIW815 (ADU-S100) injection. Acknowledgments The authors would like to thank the patients who participated in the trial and their families, and the staff at each site who assisted with the study. The authors would also like to thank Sinead Dolan and Marc Pelletier (Novartis Institutes for Biomedical Research, East Hanover, NJ) for additional biomarker contributions. This study was sponsored by Novartis Pharmaceuticals Corporation and Aduro Biotech, Inc. Editorial assistance was provided by Jenny Winstanley PhD of Articulate Science, and was funded by Novartis Pharmaceuticals Corporation. Presented at the Society for Immunotherapy of Cancer (SITC) 33rd Annual Meeting; November 7–11, 2018; Washington, D.C. P10763 Text: Qc2e13 To: 8NOVA (86682) US Only +18324604729 North, Central and South Americas; Caribbean; China +447860024038 UK, Europe & Russia +46737494608 Sweden, Europe Visit the web at: http://novartis.medicalcongressposters.com/Default.aspx?doc=c2e13 Copies of this poster obtained through Quick Response (QR) code are for personal use only and may not be reproduced without written permission from SITC and the authors of this poster Scan this QR code Presenter email address: [email protected]

Transcript of Phase I Dose-finding Study of MIW815 (ADU-S100), an ......• Activation of the STING pathway...

Page 1: Phase I Dose-finding Study of MIW815 (ADU-S100), an ......• Activation of the STING pathway increases interferon (IFN)-β production and induces the recruitment and priming of cluster

Phase I Dose-finding Study of MIW815 (ADU-S100), an Intratumoral STING Agonist, in Patients With Advanced Solid Tumors or Lymphomas

References1. Woo SR, et al. Immunity 2014;41:830–842.

2. Glickman LH, et al. Cancer Res 2016;76(14 Suppl):abst 1445.

3. Corrales L, et al. Cell Rep 2015;11:1018–1030.

4. Cheson BD, et al. J Clin Oncol 2014;32:3059–3068.

Funda Meric-Bernstam1, Theresa L. Werner2, F. Stephen Hodi3, Wells Messersmith4, Nancy Lewis5, Craig Talluto6, Mirek Dostalek5, Aiyang Tao5, Sarah M. McWhirter7, Damian Trujillo7, Jason J. Luke8

1Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX; 2Huntsman Cancer Institute, University of Utah, Salt Lake City, UT; 3Dana-Farber Cancer Institute, Boston, MA; 4University of Colorado Cancer Center, Aurora, CO; 5Novartis Pharmaceuticals Corporation, East Hanover, NJ; 6Novartis Institutes for BioMedical Research, Cambridge, MA; 7Aduro Biotech Inc., Berkeley, CA; 8The University of Chicago Medicine, Chicago, IL

This study is sponsored by Novartis Pharmaceuticals Corporation.

IntroductionThe STimulator of INterferon Genes (STING) pathway plays a crucial role in the innate immune response to immunogenic tumors.

• Activation of the STING pathway increases interferon (IFN)-β production and induces the recruitment and priming of cluster of differentiation 8 (CD8)-positive T cells against tumor antigens.1

MIW815 (ADU-S100) is a novel synthetic cyclic dinucleotide that activates the STING pathway in vitro (Figure 1).2

In preclinical tumor mouse models, intratumoral injection of MIW815 (ADU-S100) resulted in tumor regression in both injected and non-injected lesions.3

This first-in-human Phase I study was designed to evaluate the safety and efficacy of MIW815 (ADU-S100) in patients with advanced solid tumors or lymphomas.

• In addition, MIW815 (ADU-S100) is currently being investigated in combination with anti-programmed cell death protein 1 (PD-1; NCT03172936) or anti-cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4; NCT02675439) antibodies in Phase I clinical trials in patients with advanced solid tumors and lymphomas.

Figure 1. MIW815 (ADU-S100) Mechanism of Action

IFN, interferon; STING, stimulator of interferon genes.

MethodsStudy designThis is a multicenter, open-label, first-in-human, Phase I study of MIW815 (ADU-S100) in patients with advanced solid tumors or lymphomas currently in dose escalation (Figure 2; NCT02675439; EudraCT 2015-005245-31).

Primary objectives

• Safety and tolerability of MIW815 (ADU-S100).

• Recommended dose for future studies.

Secondary objectives

• Preliminary antitumor activity of MIW815 (ADU-S100).

• Pharmacokinetics (PK) and pharmacodynamics (PD) of MIW815 (ADU-S100).

Exploratory objective

• Biomarkers of response to MIW815 (ADU-S100).

Patients are treated with weekly intratumoral injections of MIW815 (ADU-S100; 50–3200 μg) on a 3-weeks-on/1-week-off schedule until they experience unacceptable toxicity, disease progression per immune-related response criteria (irRC) for solid tumors, or confirmed progressive disease in lymphoma, and/or patient/investigator decision.

Figure 2. MIW815X2101 Study Design

Dose Escalation

MTD/RDE

Solid tumors and lymphomasMIW815 (ADU-S100),

weekly IT injection(3-weeks-on/1-week-o�)

100 µg

n=6 50 µgn=6

400 µgn=5800 µgn=6

3200 µgn=6

1600 µg

n=7

200 µgn=5

Planned Dose Expansion

Group 1:Accessible UV-induced cancers,

including melanoma,SCC, and BCC

Group 2:Other accessible solid tumors

and lymphomas, including CTCL,Merkel cell carcinoma,

and HNSCC

BCC, basal cell carcinoma; CTCL, cutaneous T-cell lymphoma; HNSCC, head and neck squamous cell carcinoma;

IT, intratumoral; MTD, maximum tolerated dose; RDE, recommended dose for expansion;

SCC, squamous cell carcinoma; UV, ultraviolet.

Patient populationKey inclusion criteria

Adult patients with advanced/metastatic solid tumors or lymphomas who have progressed despite standard therapy, are intolerant to standard therapy, or for whom standard therapy is not reasonably effective or does not exist.

Measurable disease per Response Evaluation Criteria In Solid Tumors (RECIST) v1.1 or Cheson 2014 criteria.4

• At least two distinct lesions are required; both accessible for baseline and on-treatment biopsies, with patient consent.

• Lesion 1 must measure ≥10 to <100 mm in longest diameter, and be accessible for repeated intratumoral injection.

Eastern Cooperative Oncology Group performance status of ≤1.

Prior immunotherapy is permitted.

Key exclusion criteria

Symptomatic central nervous system (CNS) metastases or CNS metastases requiring local CNS-directed therapy.Systemic anticancer therapy within 4 weeks of the first dose of study treatment.Treatment with systemic steroid therapy, other than replacement-dose steroids in the setting of adrenal insufficiency.Systemic treatment with any immunosuppressive medication. History of, or active, autoimmune disease with the exception of vitiligo or resolved childhood asthma/atopy.

AssessmentsAdverse events (AEs) were assessed at every visit according to the Common Terminology Criteria for Adverse Events v4.03.

Tumor response was determined locally according to irRC, RECIST v1.1, and Cheson 2014 criteria4 for lymphoma, at screening and on Day (D) 1 of Cycle (C) 3, every 8 weeks up to C11, and then every 12 weeks until disease progression or patient withdrawal.

Blood samples were collected for PK analysis on C1D1, C1D15, and C3D1, and for PD assessment of cytokine levels on C1D1, C1D8, C1D15, and C3D1.

ResultsPatient characteristics and dispositionAs of August 16, 2018, 41 patients received MIW815 (ADU-S100) and 4 (9.8%) patients were continuing on treatment. Treatment was discontinued in 37 (90.2%) patients due to disease progression (n=29; 70.7%), physician decision (n=4; 9.8%), patient decision (n=3; 7.3%), and death (n=1; 2.4%).

Baseline patient demographics are shown in Table 1. The number of patients treated at each MIW815 (ADU-S100) dose level is shown in Figure 2.

A summary of the time on MIW815 (ADU-S100) treatment and patient response is shown in Figure 3.

Table 1. Baseline Patient DemographicsCharacteristic

All Patients(N=41)

Median age, years (range) 62 (26–80)Sex, n (%)

Male 18 (43.9)Female 23 (56.1)

Race, n (%)Caucasian 27 (65.9)Black 6 (14.6)Asian 2 (4.9)Other/unknown 6 (14.6)

ECOG PS, n (%)0 11 (26.8)1 30 (73.2)

Prior therapy with a checkpoint inhibitor, n (%)Yes 22 (53.7)No 19 (46.3)

Number of prior regimens, n (%)0 3 (7.3)1 4 (9.8)≥2 34 (82.9)

ECOG PS, Eastern Cooperative Oncology Group performance status.

Figure 3. Time on MIW815 (ADU-S100) Treatment and Response Evaluation

OngoingResponse

PRSDPD

YesPrior checkpoint

50 µ

g10

0 µ

g20

0 µ

g4

00

µg

80

0 µ

g16

00

µg

Duration of exposure (months)

MIW

815

(AD

U-S

100

) dos

e

Breast cancer (TNBC)

Cutaneous melanoma

Ovarian cancerColorectal cancer

ER+/HER2– breast cancer Colorectal cancer

Anal cancerEsophageal cancer

Merkel cell carcinoma

Non-Hodgkin lymphoma

0 2 4 6 8 10

320

0 µ

g

Non-Hodgkin lymphoma

Cutaneous melanomaEsophageal cancer

Breast cancer (TNBC)

Melanoma

Melanoma

Non-Hodgkin lymphoma

MelanomaMelanoma

Uveal melanoma

Hodgkin lymphomaMerkel cell carcinoma

Non-Hodgkin lymphomaUveal melanoma

Breast cancer (ER+/HER2+)Melanoma

Pancreatic cancerBreast cancer (TNBC)

Ovarian cancer

Other (adenocystic carcinoma)

AngiosarcomaMelanomaOther (leiomyosarcoma)

Colorectal cancer

Other (sebaceous gland adenocarcinoma)

Endometrial cancer

Head and neck cancer (parotid gland)

Sarcoma

Merkel cell carcinomaRenal carcinoma (collecting duct)*

Other (epithelioid hemangioepithelioma)

Data cut-off: August 16, 2018.

*Patient ongoing treatment at 14 months.

ER+, estrogen receptor-positive; HER2–, human epidermal growth factor receptor 2-negative; HER2+, HER2-positive;

PD, progressive disease; PR, partial response; SD, stable disease; TNBC, triple-negative breast cancer.

Safety and tolerabilityAEs suspected to be related to MIW815 (ADU-S100) treatment were reported in 78.0% of patients; 12.2% of patients experienced Grade 3/4 AEs.

• The most common suspected related AEs (in ≥10% of all patients) were headache, injection site pain, and pyrexia (in 14.6% of patients each).

• Elevated lipase was the only Grade 3/4 suspected related AE reported in >1 patient (n=2; 4.9%).Treatment-emergent serious AEs were reported in 1 patient (Grade 3 dyspnea and Grade 4 respiratory failure; dose level: 1600 μg); this was related to underlying disease progression.

There were no dose-limiting toxicities (DLTs) observed during the first cycle of treatment and no patients discontinued treatment due to an AE.

PharmacokineticsThe mean plasma concentration–time profiles for MIW815 (ADU-S100) on C1D1, C1D15, and C3D1 are shown in Figure 4.

MIW815 (ADU-S100) absorption from the intratumoral injection site was rapid, reaching maximal plasma concentration within minutes of dosing.

MIW815 (ADU-S100) exposure generally increased in a dose-dependent manner.

The apparent clearance of the MIW815 (ADU-S100) from systemic circulation was rapid with an observed terminal half-life of approximately 10–23 minutes.

Figure 4. Concentration–Time Profiles for MIW815 (ADU-S100) on C1D1, C1D15, and C3D1

Mea

n (S

D) M

IW8

15 (A

DU

-S10

0)

plas

ma

conc

entr

atio

n (n

g/m

L)

Time (hours)

MIW815 dose:

C1D15

0 0.5 1 2 4

C3D1

0 0.5 1 2 4

C1D1

0.01

0.3

10

20

50

100

0 0.5 1 2 4

50 µg 100 µg 200 µg 400 µg 800 µg 1600 µg 3200 µg

Data cut-off: August 16, 2018. C, Cycle; D, Day; SD, standard deviation.

Patient vignette – Case example 1Patient and disease characteristics

63-year-old male with Stage IV collecting duct carcinoma with metastases to the left supraclavicular lymph node (LN) and left neck.

Treatment history: antineoplastic therapy with an investigational mouse double minute 2 homolog (MDM2) inhibitor and prior right radical nephrectomy and retroperitoneal LN dissection.

Experience on study

Treatment: weekly intratumoral injections of 800 μg MIW815 (ADU-S100; 3-weeks-on/1-week-off).

Reported toxicities: Grade 1 headache, nausea, and hyperthyroidism/Graves’ disease.

Best overall response (BOR): stable disease (SD); patient remains on study at C14.

On-study pharmacodynamics

Increases in systemic cytokine levels, including interleukin 6 (IL6), were observed following MIW815 (ADU-S100) injection on C1D1, C1D8, and C3D1 (Figure 5A).

Treatment with MIW815 (ADU-S100) resulted in a >2-fold increase in expression of the IFN-γ, programmed death-ligand 1 (PD-L1), and CD8A genes, and the natural killer (NK) cell gene set in the injected lesion (Figure 5B).

An increase in CD8-positive tumor-infiltrating lymphocytes (TILs) and PD-L1 stromal staining was observed in the injected lesion following MIW815 (ADU-S100) treatment (Figure 5C).

Figure 5. Case Example 1: Pre- and Post-MIW815 (ADU-S100) Therapy Cytokine Levels, RNA, and IHC in a Patient With Collecting Duct Carcinoma

A. Fold Change in Systemic Cytokine Levels on C1D1, C1D8, and C3D1

Time of assessment

C1D1

Fol

d ch

ange

0

2.5

7.5

5

15

Pre

-dos

e

2 ho

urs

6 h

ours

Pre

-dos

e

2 ho

urs

6 h

ours

Pre

-dos

e

2 ho

urs

6 h

ours

C1D8 C3D1

Cytokine

IFN-β1

IL6

IL8

MCP1

MIP1B

C, Cycle; D, Day; IFN, interferon; IHC, immunohistochemistry; IL6, interleukin 6; IL8, interleukin 8;

MCP1, monocyte chemoattractant protein 1; MIP1B, macrophage inflammatory protein 1 beta.

B. Fold Change in Intratumoral RNA Expression in the Injected and Non-injected Lesion From Screening to C3D1 (Pre-dose)

Injected Lesion

–2

JAK1 STAT1 IFN-γ PD-L1 CD8A NK cellgene set

IFN-γgene set

JAK1 STAT1 IFN-γ PD-L1 CD8A NK cellgene set

IFN-γgene set

–1

0

1

2

Log2

fold

cha

nge

Gene

Non-injected Lesion

0.0 0.5 1.0 1.5

Log2 fold change

*NK cell gene set includes: IL12A, IL12B, KIR3DL1, KIR3DL2, KIR3DL3, KLRB1, KLRC1, KLRC2, KLRD1, KLRF1, KLRG1,

KLRK1, and NCR1; †IFN-γ gene set includes: TBK1, IRF3, IFNAR1, JAK1, JAK2, TYK2, STAT1, STAT2, STAT3, IFNG,

IFNGR1, IDO1, IL15RAm CXCL9, CXCL10, CXCL11, and CD274.

C, Cycle; CD8A, cluster of differentiation 8A; D, Day; IFN, interferon; JAK1, Janus kinase 1; NK, natural killer;

PD-L1, programmed death-ligand 1; STAT1, signal transducer and activator of transcription 1.

C. IHC Micrographs of the MIW815 (ADU-S100) Injected and Non-injected Lesion at Screening and C3D1 (Pre-dose)

H&E

Scr

eeni

ngC

3D1

Injected LesionCD8* CD68* FOXP3* PD-L1† H&E CD8* CD68* FOXP3* PD-L1†

Scr

eeni

ngC

3D1

2.55% 2.55% 0.39% PD-L1-negativestromal staining

PD-L1-positive stromal staining

PD-L1-negativestromal staining

PD-L1 negativestromal staining

9.57% 5.64% 1.44%

16.94% 10.38% 4.05%

17.08% 7.07% 1.26%

Non-injected Lesion

*Biomarker area % per image analysis; †Tumor positive score % per pathologist analysis; PD-L1 measured using

the IHC 22C3 pharmDx assay.

C, Cycle; CD, cluster of differentiation; D, Day; FOXP3, forkhead box P3; H&E, hematoxylin and eosin;

IHC, immunohistochemistry; PD-L1, programmed death-ligand 1.

Patient vignette – Case example 2Patient and disease characteristics

68-year-old male with esophageal cancer and metastases to the mediastinal LN, supraclavicular LN, and adrenal gland.

Treatment history: neoadjuvant paclitaxel, carboplatin, and radiation therapy followed by tumor resection, adjuvant 5-fluorouracil and oxaliplatin chemotherapy, and for metastatic disease cisplatin/irinotecan, nivolumab, and investigational anti-matrix metallopeptidase-9 antibody therapy.

Experience on study

Treatment: weekly intratumoral injections of 800 μg MIW815 (ADU-S100; 3-weeks-on/1-week-off).

Reported toxicities: Grade 1 injection site reaction and exertional dyspnea; patient also experienced an incarcerated bowel (unrelated to study drug).

BOR: SD for 3 cycles.

On-study pharmacodynamicsIFN-β1 levels increased following MIW815 (ADU-S100) injection on C1D1, C1D8, C1D15, and C3D1 (Figure 6A).

Expression of the T-cell differentiation antigen CD8A gene and the NK cell gene set were increased >2-fold in the injected lesion as a result of MIW815 (ADU-S100) treatment (Figure 6B).

Following MIW815 (ADU-S100) injection, an increase in the staining of CD8-positive TILs and PD-L1 stromal staining was observed in the injected lesion (Figure 6C).

Figure 6. Case Example 2: Pre- and Post-MIW815 (ADU-S100) Therapy Cytokine Levels, RNA, and IHC in a Patient With Esophageal CancerA. Fold Change in Systemic Cytokine Levels on C1D1, C1D8, C1D15, and C3D1

C1D1

Fol

d ch

ange

1

2

3

4

Pre

-dos

e

2 ho

urs

6 h

ours

Pre

-dos

e

2 ho

urs

6 h

ours

Pre

-dos

e

2 ho

urs

6 h

ours

Pre

-dos

e

2 ho

urs

6 h

ours

C1D8 C1D15 C3D1

Cytokine

IFN-β1

IL6

IL8

MCP1

MIP1B

Time of assessment

C, Cycle; D, Day; IFN, interferon; IHC, immunohistochemistry; IL6, interleukin 6; IL8, interleukin 8;

MCP1, monocyte chemoattractant protein 1; MIP1B, macrophage inflammatory protein 1 beta.

B. Fold Change in Intratumoral RNA Expression in the Injected and Non-injected Lesion From Screening to C3D1 (Pre-dose)

Gene

Injected Lesion

JAK1 STAT1 IFN-γ PD-L1 CD8A IFN-γgene set

NK cellgene set

JAK1 STAT1 IFN-γ PD-L1 CD8A IFN-γgene set

NK cellgene set

–1

0

1

2

Log2

fold

cha

nge

Non-injected Lesion

–2 0.0 0.5 1.0

Log2 fold change

*NK cell gene set includes: IL12A, IL12B, KIR3DL1, KIR3DL2, KIR3DL3, KLRB1, KLRC1, KLRC2, KLRD1, KLRF1, KLRG1,

KLRK1, and NCR1; †IFN-γ gene set includes: TBK1, IRF3, IFNAR1, JAK1, JAK2, TYK2, STAT1, STAT2, STAT3, IFNG,

IFNGR1, IDO1, IL15RAm CXCL9, CXCL10, CXCL11, and CD274.

C, Cycle; CD8A, cluster of differentiation 8A; D, Day; IFN, interferon; JAK1, Janus kinase 1; NK, natural killer;

PD-L1, programmed death-ligand 1; STAT1, signal transducer and activator of transcription 1.

C. IHC Micrographs of the MIW815 (ADU-S100) Injected and Non-injected Lesion at Screening and C3D1 (Pre-dose)

Scr

eeni

ngC

3D1

Injected Lesion Non-injected LesionH&E

Scr

eeni

ngC

3D1

CD8* CD68* FOXP3* PD-L1† H&E CD8* CD68* FOXP3* PD-L1†

3.9% 11.63% 0.66% PD-L1-negativestromal staining

PD-L1-positivestromal staining

PD-L1-negativestromal staining

PD-L1-negativestromal staining

19.97% 12.6% 2.68%

7.59% 25.31% 2.29%

9.85% 14.91% 1.31%

*Biomarker area % per image analysis; †Tumor positive score % per pathologist analysis; PD-L1 measured using the

IHC 22C3 pharmDx assay.

C, Cycle; CD, cluster of differentiation; D, Day; FOXP3, forkhead box P3; H&E, hematoxylin and eosin;

IHC, immunohistochemistry; PD-L1, programmed death-ligand 1.

ConclusionsSingle-agent MIW815 (ADU-S100) was well tolerated at doses ranging from 50 to 3200 μg in patients with advanced solid tumors and lymphoma with no DLTs noted. Dose escalation is ongoing.

Preliminary signs of MIW815 (ADU-S100) biological activity were observed in patients with advanced solid tumors and lymphoma, including those who had previously received treatment with a checkpoint inhibitor.

In the highlighted patient cases, the ability of MIW518 (ADU-S100) to activate the antitumor immune response is suggested by observed increases in CD8-positive TILs, genes involved in the antitumor immune response, and systemic cytokines following MIW815 (ADU-S100) injection.

AcknowledgmentsThe authors would like to thank the patients who participated in the trial and their families, and the staff at each site who assisted with the study.

The authors would also like to thank Sinead Dolan and Marc Pelletier (Novartis Institutes for Biomedical Research, East Hanover, NJ) for additional biomarker contributions.

This study was sponsored by Novartis Pharmaceuticals Corporation and Aduro Biotech, Inc.

Editorial assistance was provided by Jenny Winstanley PhD of Articulate Science, and was funded by Novartis Pharmaceuticals Corporation.

Presented at the Society for Immunotherapy of Cancer (SITC) 33rd Annual Meeting; November 7–11, 2018; Washington, D.C.

P10763

Text: Qc2e13To: 8NOVA (86682) US Only +18324604729 North, Central and South Americas; Caribbean; China +447860024038 UK, Europe & Russia +46737494608 Sweden, Europe

Visit the web at: http://novartis.medicalcongressposters.com/Default.aspx?doc=c2e13

Copies of this poster obtained through Quick Response (QR) code are for personal use only and may not be reproduced without written permission from SITC and the authors of this poster

Scan this QR code

Presenter email address:[email protected]