715 Optimization of an -free Virus Therapy Regimen Containing the HCV NS3/4A Protease Inhibitor , IDWeek 2013 the Non-nucleoside NS5B Inhibitor Deleobuvir, and for Genotyp e-1b-infected Patients Parvez Mantry 1, Marcus Schuchmann 2, Ansgar W. Lohse 3, Keikawus Arasteh 4, Michael Manns 5, Thomas Berg 6, Stefan Mauss 7, Michael Geissler 8, Federico Mensa 9, Wulf O. Böcher 10 , Stefan Zeuzem 11 1The Liver Institute at Methodist Dallas Medical Center, Dallas, TX; 2University Hospital Mainz, Mainz, Germany; 3University Hospital Hamburg-Eppendorf, Hamburg, Germany; 4EPIMED - GmbH Berlin, Germany; 5Hannover Medical School, Hannover, Germany; 6University Hospital Leipzig, Leipzig, Germany; 7Center for HIV and Hepatogastroenterology, Düsseldorf, Germany; 8Esslingen Hospital, Esslingen, Germany; 9Boehringer Ingelheim, Ridgefield, CT, USA; 10 , Biberach, Germany; 11 J. W. Goethe University Hospital, Frankfurt am Main, Germany

INTRODUCTION • SOUND-C2 included a loading dose of deleobuvir 1200 mg on Day 1 leading to high plasma RESULTS Figure 6. Individual HCV RNA over time concentrations that were associated with an increased incidence of gastrointestinal (GI) • New direct acting antivirals (DAAs) currently in development for treatment of chronic adverse events (AEs) (Figure 3) 3 • The interferon-free combination of faldaprevir, deleobuvir and RBV for 16 weeks was highly 10 8 (HCV) are being tested in conjunction with and without both pegylated efficacious in patients infected with HCV GT-1b with SVR12 rates of 95% (Figure 5) HCV GT-1b EOT Figure 3. Delobuvir plasma levels correlate with GI AEs in SOUND-C2 interferon-alpha and ribavirin (PegIFN/RBV) 10 7 – Faldaprevir is an NS3/4A HCV protease inhibitor currently in phase 3 trials, with a Figure 5. Primary endpoint SVR 12 (ITT) pharmacokinetic profile that supports once-daily (QD) dosing 1.0 10 6

100 100 GT-1a, IL28B CC GT-1b ) L – Deleobuvir is an HCV NS5B polymerase inhibitor with a pharmacokinetic profile that 8 0.9 100 95 ) m y 5 /

L 10 supports twice-daily (BID) dosing a U

D Odds ratio (per log 10 increase of deleobuvir) I m (

0.8 / o 1.771 (95% CI, 1.042–3.010), n= 339 U t A 4

I 80

( 10 N RATIONALE p

u 0.7 A R N g V 3 n R 58

C Cirrhotic patients i 60 10 • The aim of SOUND-C3 was to optimize IFN-free treatment with faldaprevir, deleobuvir, and t 0.6 Estimated probability i V H C RBV following the SOUND-C2 study: m o 0.5 Upper 95% confidence limit H 2

v 10 e l – SOUND-C2 investigated the safety, efficacy, and treatment duration of faldaprevir 120 mg QD, f Lower 95% confidence limit 40 33 b o

0.4 a y deleobuvir 600 mg BID or TID with or without RBV for 16, 28, or 40 weeks in 362 HCV t 1 t

i 10 Event, 1= Yes, 0=No c BLD l i GT1-infected patients e 17 t b 0.3 20 e a

d 0 – Cirrhotic patients were included and accounted for 9% of the total population b 10 n o

r 7/12 20/20 4/12 20/20 2/12 19/20 0.2 U 0 5 10 15 20 25 30 – In SOUND-C2, patients infected with HCV GT-1b who received faldaprevir 120 mg QD, p 0 d EoT SVR4 SVR12 Time (weeks) deleobuvir 600 mg BID, and RBV achieved an SVR12 rate of 85% e

t 0.1

a EoT, end of treatment; ITT, Intention-to-treat analysis.

– Patients infected with HCV GT-1a and carrying the IL28B CC genotype achieved a similar m i

t 0.0 SVR12 to those with GT-1b (Figure 1) s 8 HCV GT-1a, IL28b CC E • One GT-1b-infected patient discontinued treatment at Day 69 due to AEs, did not achieve 10 EOT – In SOUND-C2 the deleobuvir BID regimen had a 28-week duration; the BID regimen was SVR12, and subsequently relapsed 1 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0 tested for 16 weeks in SOUND-C3 10 7 Log 10 median deleobuvir plasma concentration Day 4–8 • Another GT-1b infected patient discontinued treatment early at Day 41 due to AEs but went on to achieve SVR12 Figure 1. SOUND-C2: SVR12 by HCV GT-1 subtype and IL28B genotype 10 6

• Eight GT-1a treatment-naïve patients experienced rebound or relapse, two patients were ) L

Faldaprevir 120 mg QD, deleobuvir 600 mg BID and RBV for 28 weeks below the limit of detection (BLD) (~10 IU/mL) at SVR12 follow-up, and one patient m 5 100 / 10 U

91 METHODS I

discontinued while BLD and did not complete follow-up (Figure 6) (

84 A 10 4 • Adverse events and worst on-treatment changes in laboratory values are shown in Tables 2 & 3 N 80 75 • The Phase 2b SOUND-C3 study (N=32) tested the BID regimen from SOUND-C2 for a R

period of only 16 weeks (Figure 4); the primary endpoint was SVR12 and patients with V 3 C ) Table 2. Adverse event summary 10 H

% 60 compensated liver cirrhosis were eligible and accounted for 13% of the population (

2 AE, n (%) N=32 10 2 1 • Treatment-naïve GT-1a patients (n=12) carrying the IL28b CC genotype (rs12979860, R Any AE 30 (94) V 40 GT-1a-CC) and GT-1b patients (n=20) carrying any IL28b genotype were treated with

S 32 1 faldaprevir 120 mg QD + deleobuvir 600 mg BID Serious AEs 1 (3) 10 BLD a 20 Dehydration 1 (3) • No loading dose of deleobuvir was administered on Day 1 in SOUND-C3 10 0 AEs leading to discontinuation 3 (9) 0 5 10 15 20 25 30 6/8 7/22 10/ 11 31/37 • Baseline characteristics are shown in Table 1 0 Dehydration 1 (3) a Time (weeks) 1a CC 1a non-CC 1b CC 1b non-CC Vomiting 1 (3) BID, twice-daily; QD, once-daily Figure 4. SOUND-C3 trial design EOT, end of treatment. Graph truncated at first measurement >1000 IU/mL after nadir. Last observation carried forward (LOCF) for missing time points. Pruritus 1 (3) Lower panel: One patient discontinued at BLD and could not complete follow-up visits due to personal circumstances. LOCF for missing time points. • Pharmacokinetic and pharmacodynamic analysis in SOUND-C2 indicated an interaction between Any AE Grade ≥2 15 (47) faldaprevir and deleobuvir that increased plasma concentrations of each drug (Figure 2) 2 Faldaprevir 120 mg QD AEs of interest of highest frequency Anemia 5 (16) CONCLUSIONS Figure 2. Pharmacokinetics of faldaprevir and deleobuvir in SOUND-C2 N=32 + deleobuvir 600 mg BID Follow-up Fatigue 3 (9) + RBV Vomiting 3 (9) 6000 Faldaprevir 20 Deleobuvir • The interferon-free combination of faldaprevir, deleobuvir, and RBV for 16 weeks ) ) Nausea 3 (9) L L was highly efficacious in patients infected with HCV GT-1b with SVR12 rates m m

/ Life threatening AEs 0 / 15 g Combination therapy g Day 1 Week 16 Week 28 a n 4000 Same patient of 95% µ ( ( Combination therapy r r i i v

v 10

e • All four GT-1b-infected patients with cirrhosis achieved SVR12 u r Table 3. Grade 3 and 4 changes in laboratory values b p

2000 o a

e Table 1. Baseline characteristics d l 5 • The combination of faldaprevir, deleobuvir, and RBV was well tolerated with few

l Grade 3 and 4 adverse events, n (%) N=32 e a D F Monotherapy Monotherapy GT-1a (n=12) GT-1b (n=20) Hemoglobin ≤8.9 g/dL 4 (13) discontinuations due to AEs 0 0 Mean age, years (SD) 47 (5) 50 (15) 3 02468 02468 White blood cell count ≤1499/mm 0 • Virological response in GT-1a-infected patients was significantly lower Time (weeks) Time (weeks) Male, n (%) 7 (58) 5 (25) Lymphocytes ≤499/mm 3 0 Caucasian, n (%) 12 (100) 20 (100) Neutrophils ≤749/mm 3 1 (3) a References: 1. Zeuzem et al. J Hepatol. 2012;56(Suppl2):S45 (Abstract 101). 2. Sabo et al. Hepatology. 2012;56:568A (Abstract 777). 3. SOUND-C2, data on file. UGT1A1 OATP P- gp MRP-2 BCR P Acknowledgments: This poster was originally presented at the The German Society for Digestive and Metabolic Diseases, September 11-14, 2013, Nürnberg, Germany. This work was supported by Boehringer Ingelheim Pharmaceuticals, Inc. 2 3 (BIPI). The authors met criteria for authorship as recommended by the International Committee of Medical Journal Editors (ICMJE) and were fully responsible for all content and editorial decisions, and were involved at all stages of poster Mean BMI, kg/m (SD) 26 (4) 26 (4) Platelets ≤49,999/mm 0 develop ment. The authors received no compensation related to the development of the poster, but are consultants for BIPI and did receive research grant support for this study. Medical writing assistance, supported financially by Boehringer Ingelheim, Substrate Substrate Substrate was provided by Katharine Howe of Adelphi Communications Ltd. Editorial support and formatting assistance for this poster was provided by Adelphi Communications, which was contracted and compensated by BIPI for these services. Faldaprevir IL28B CC, n (%) 12 (100) 3 (15) Disclosures: P Mantry has served as a speaker and received grant and research support from Abbott, Boehringer Ingelheim, Genentech, Merck, Salix, and Vertex. M Schuchmann has received medical writing assistance from Inhibitor Inhibitor Inhibitor ALT ≥5.1 x ULN 0 Boehringer Ingelheim; served as a consultant for Bristol-Myers Squibb, Boehringer Ingelheim, Gilead, Norgine, and Roche; has received grants/grants pending from DFG; and has served on speakers’ bureaus for Bristol-Myers Squibb, Liver cirrhosis, n (%) 0 4 (20) Boehringer Ingelheim, Falk, Gilead, Merck, MSD, and Roche. A Lohse has received grant/research support from Roche, MSD, Boehringer Ingelheim, Bristol-Myers Squibb, and Gilead and has served as a speaker/teacher for Roche, Substrate Substrate Substrate Bilirubin (total) ≥2.6 x ULN 14 (44) MSD, and Falk. K Arasteh has received grant/research support, medical writing assistance, and served as a board member, consultant, and on speakers’ bureaus for Boehringer Ingelheim. M Manns has served as a consultant for Deleobuvir Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, GlaxoSmithKline, Idenix, Janssen, Merck, Novartis, and Roche; has received grants/grants pending from Boehringer Ingelheim, Bristol-Myers Squibb, Gilead, Janssen, Merck, Novar tis, Mean baseline HCV RNA, log 10 IU/mL (SD) 6.5 (0.7) 6.3 (0.7) DAIDS Grade 3 and 4 laboratory changes shown. Three patients had Grade 4 bilirubin changes (>5 x ULN); all other events were Grade 3. aOne patient had severe and Roche; and has served on speakers’ bureaus for Bristol-Myers Squibb, Gilead, Janssen, Merck, and Roche. T Berg has served as a consultant for and has grants/grants pending from AbbVie, Boehringer Ingelheim, Bristol-Myers Inhibitor Inhibitor Inhibitor Squibb, Gilead, MSD, Novartis, Roche, and Vertex; and has served as a board member and on speakers’ bureaus for AbbVie, Bristol-Myers Squibb, Gilead, MSD, Novartis, Roche, and Vertex. S Mauss has served as a speaker for neutopenia and had received metamizole (dipyrone) for from Day 14 to Day 22. Neutropenia was seen at visits on Day 30, 44, and 58. Neutrophil levels returned to Abbott, Bristol-Myers Squibb, Tibotec, and Roche; has served on advisory boards for Abbot, GlaxoSmithKline, Gilead, and Tibotec; and has received research funding from Abbott and Roche. M Geissler has nothing to disclose. Baseline HCV RNA ≥800,000 IU/mL, n (%) 10 (83) 17 (85) normal at the next visit (Day 90). ALT, alanine transaminase; DAIDS, division of AIDS. Federic Mensa is an employee of Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT. WO Böcher is an employee of Boehringer Ingelheim Pharma, Biberach, Germany. S Zeuzem has served as a consultant for Abbot, Achillion, AstraZeneca, BMS, Boehringer Ingelheim, Gilead, Idenix, Janssen, Merck, Novartis, Presidio, Roche, Santaris, and Vertex; and has served on speakers’ bureaus for Bristol-Myers Squibb, Boehringer Ingelheim, Gilead, MSD, Roche and Janssen.

IDWeek™ 2013, October 2–6, 2013, San Francisco, CA