SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

4
SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates High SVR Rates in Pa ents With HCV Genotype 2, 4, 5, or 6 Infecon Without Cirrhosis Following an 8-Week Treatment Dura on Tarek Hassanein 1 , David Wyles 2 , Stanley Wang 3 , Paul Y Kwo 4 , Mitchell L Shiman 5 , Ziad Younes 6 , Susan Greenbloom 7 , Catherine A Stedman 8 , Joseph Sasadeusz 9 , Humberto Aguilar 10 , Jeong Heo 11 , Ran Liu 3 , Teresa I Ng 3 , Chih-Wei Lin 3 , Federico Mensa 3 1 Southern California GI and Liver Centers and Southern California Research Center, Coronado, California, United States; 2 Denver Health Medical Center, Denver, Colorado, United States; 3 AbbVie Inc., North Chicago, Illinois, United States; 4 Indiana University School of Medicine, Indianapolis, Indiana, United States; 5 Liver Ins tute of Virginia, Bon Secours Health System, Newport News and Richmond, Virginia, United States; 6 GastroOne, Germantown, Tennessee, United States; 7 Toronto Digesve Disease Associates, Toronto, Ontario, Canada; 8 Christchurch Hospital and University of Otago, Christchurch, New Zealand; 9 Island Health Authority, Secon of Infecous Diseases, Victoria, Brish Columbia, Canada; 10 Louisiana Research Center, Shreveport, Louisiana, United States; 11 College of Medicine, Pusan Naonal University and Medical Research Instute, Pusan Naonal University Hospital, Busan, Republic of Korea CONCLUSIONS 97% (196/203, ITT) of GT2, 4, 5, or 6-infected paents without cirrhosis achieved SVR12 following 8 weeks of G/P In DAA-naïve paents with GT2 infecon, 8-week treatment was non-inferior to the historical 95% SVR12 rate achieved with 12 weeks SOF/RBV There were no virologic failures in paents with GT4–6 infecon Baseline viral load, genotype/subtype, F0–F3 brosis stage, presence of baseline polymorphisms, and prior treatment experience with interferon- or SOF-based regimens did not impact achievement of SVR12 SVR rates were similar to observed rates following 12-week treatment with G/P G/P for 8 weeks was well-tolerated, with no disconnuaons due to AEs, no DAA-related serious AEs, and rare grade 3 or higher lab abnormalies (0.5%) The all-oral, once-daily, IFN- and RBV-free regimen of G/P is highly ecacious for treatment of paents with HCV GT 2, 4, 5, or 6 infecon without cirrhosis following an 8-week treatment duraon INTRODUCTION • Hepas C virus genotypes (HCV GT) 2, 4, 5, and 6 have diverse global distribuons, accounng for approximately 9%, 8%, 1%, and 5% of HCV infecons worldwide, respecvely 1 Global Prevalence of HCV Genotype 2, 4, 5, and 6 Infecon <10% prevalence GT2 GT4 GT5 GT6 >10% prevalence of more than one (GT2, 4-6) GT • A pangenotypic treatment that is safe and highly ecacious, with a treatment duraon as short as possible, could improve paent adherence and access to care Next Generaon Direct-acng Anvirals In vitro: 2,3 High barrier to resistance Potent against common NS3 polymorphisms (eg, posions 80, 155, and 168) and NS5A polymorphisms (eg, posions 28, 30, 31, and 93) Addive/synergisc anviral acvity Clinical PK & metabolism: Oral dosing of 3 pills once-daily Minimal metabolism and primary biliary excreon Negligible renal excreon (<1%) h b ba ar r rr r r r i i i i i e e e r r r t t t t t o o o r r r e e e s s s i i i i i s s s t t t t t a a an n n c c c e e e e n t a g a i n s t c o mm on NS3 po l ymo r p posi ons 8 0 , 1 5 5 , an d 1 6 8) an d NS posi ons 2 8 , 3 0 , 3 1, an d 9 3 ) Glecaprevir (formerly ABT-493) pangenotypic NS3/4A protease inhibitor Pibrentasvir (formerly ABT-530) pangenotypic NS5A inhibitor Collecvely: G/P G/P is co-formulated and dosed once daily as three 100 mg/40 mg pills for a total dose of 300 mg/120 mg. Glecaprevir was idened by AbbVie and Enanta. • In previous phase 2 studies, sustained virologic response at 12 weeks aer treatment (SVR12) rates of 98% and 100% were achieved following treatment with GLE + PIB (G/P) for 8 weeks in GT2-infected paents or 12-weeks in GT 4–6 infected paents, respecvely, with no virologic failures 4 OBJECTIVE • SURVEYOR-II, Part 4 is a phase 2, open-label, mulcenter, single-arm study that evaluated the safety and ecacy of an 8-week G/P regimen in paents with GT4–6 infecon and a larger cohort of GT2-infected paents Presented at the 67th Annual Meeng of the American Associaon for the Study of Liver Diseases, November 11–15, 2016, Boston, Massachuse s LB-15 REFERENCES 1. Messina, et al. Hepatology. 2015. 2. Ng TI, et al. Abstract 636. CROI, 2014. 3. Ng TI, et al. Abstract 639. CROI, 2014. 4. Kwo, et al. JHep. 2016 (accepted manuscript under revision). DISCLOSURES T Hassanein: Research/Grant Support: AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, Eisai, Gilead, Idenix, Ikaria, Janssen, La Jolla Pharmaceucals, Merck, Mochida, NGM BioPharmaceucals, Obalon, Roche, Ocera, Sundise, Salix, Taigen, Takeda, Tobira, Vertex, Vital Therapies; Speaker: Baxter, Bristol-Myers Squibb, Gilead, Janssen, Salix; Advisor: AbbVie, Bristol-Myers Squibb. DL Wyles: Research Grants: AbbVie, Bristol-Myers Squibb, Gilead, Merck, Tacere (paid to the University of California Regents); Consultant: AbbVie, Bristol-Myers Squibb, Gilead, Janssen, Merck. PY Kwo: Grant Support: AbbVie, Bristol-Myers Squibb, Conatus, Eisai, Gilead, Janssen, Merck; Advisor: Abbo, AbbVie, BMS, Gilead, Janssen, Merck, Quest, Alnylam, Durect, DSMB, Inovio. ML Shiman: Advisor: AbbVie, Achillion, Bristol-Myers Squibb, Gilead, Merck; Grant/Research Support: AbbVie, Achillion, Beckman-Colter, Bristol-Myers Squibb, Gilead, Intercept, Lumena, Novars; Speaker: AbbVie, Bayer, Gilead, Janssen, Merck. Z Younes: Grant/Research Support: AbbVie, Gilead, Bristol-Myers Squibb, Idenix, Vertex, Roche, Merck, Janssen, Tibotec; Speaker: AbbVie, Gilead, Intercept, Vertex; Advisor: AbbVie, Gilead. S Greenbloom: Nothing to disclose. CA Stedman: Grant/Research Support: Gilead; Consultant/Advisor: Janssen, AbbVie, Gilead, MSD. J Sasadeusz: Grants: AbbVie, Gilead, Roche; Advisory Board: AbbVie, BMS, Gilead, Merck; Sponsored Lectures: BMS, Gilead. H Aguilar: Grant/Research Support: AbbVie, Genentech, Gilead. J Heo: Advisory Board: Bristol-Meyers Squibb, Gilead Sciences; Research Support: Bristol-Meyers Squibb, Roche. S Wang, TI Ng, R Liu, CW Lin, and F Mensa are employees of AbbVie Inc. and may hold stock or stock opons. ACKNOWLEDGEMENTS The authors would like to express their gratude to the paents and their families who parcipated in this study. Medical wring support was provided by Zoë Hunter, PhD, of AbbVie. MATERIALS AND METHODS STUDY DESIGN GT 2, 4, 5, 6 N = 203 Day 0 Wk 40 Open-label Treatment Wk 20 Wk 8 G/P 300 mg/120 mg* SVR12 assessment *G/P is co-formulated and dosed once daily as three 100 mg/40 mg pills for a total dose of 300 mg/120 mg. KEY INCLUSION CRITERIA 18 years of age • BMI 18 kg/m 2 • Chronic HCV GT2, 4, 5, or 6 infecon with HCV RNA >1000 IU/mL • Absence of cirrhosis documented by liver biopsy (eg, METAVIR <4 and ISHAK <5), transient elastography (FibroScan® 12.5 kPa) or serum markers (FibroTest® 0.48 + APRI <1) • HCV treatment-naïve or • Treatment-experienced with interferon (IFN) or pegIFN ± ribavirin (RBV), or sofosbuvir (SOF) + RBV ± pegIFN therapy KEY EXCLUSION CRITERIA • Prior HCV treatment experience with any direct-acng anviral (DAA) other than SOF • Co-infecon with hepas B or HIV • ALT >10 × ULN, AST >10 × ULN, direct bilirubin >ULN, albumin <LLN or platelets <90 000 cells/mm 3 ENDPOINTS AND ANALYSES Safety Assessments Primary Efficacy Endpoint Secondary Efficacy Endpoints The percentage of paents with on-treatment virologic failure, post-treatment relapse and SVR4 The percentage of paents with SVR12 (HCV RNA <25 IU/mL 12 weeks aer the last dose of study drug) The percentage of GT2-infected DAA-naïve paents with SVR12 will be non-inferior to the historical 95% SVR12 rate (12 weeks SOF + RBV) if the lower confidence bound of the 2-sided 95% confidence interval is >89% Adverse events (AEs) and laboratory abnormalies were assessed in all paents receiving at least 1 dose of study drug Next generaon sequencing (detecon threshold = 15%) performed to idenfy baseline polymorphisms and, treatment-emergent substuons in NS3 and NS5A Addional Assessments Table 1. Baseline Demographics and Disease Characteris cs Characteristic 8 Week G/P N = 203 Male, n (%) 98 (48) Race, n (%)* White 155 (76) Black 21 (10) Asian 23 (11) Other 4 (2) Age, median (range), years 55 (19-83) BMI, median (range), kg/m 2 26.8 (17.3–65.7) Genotype, n (%) 2 145 (71) Subtype a/b/c/q 21/103/3/1 4 46 (23) Subtype a/d/f/k/m/v/v–q 26/8/1/1/1/3/1 5 2 (1) § 6 10 (5) Subtype a/e/l 3/3/1 HCV Treatment-naïve, n (%) 176 (87) Treatment-experienced, n (%) 27 (13) IFN-based 21 (10) SOF-based 6 (3) HCV RNA, median (range), log 10 IU/mL 6.45 (0.75–7.62) Baseline Fibrosis Stage, n (%) F0–F1 170 (84) F2 12 (6) F3 21 (10) Non-CC IL28B genotype, n (%) 115 (57) PPI use 19 (9) BMI, body mass index; IFN, interferon; SOF, sofosbuvir; HCV, hepas C virus; PPI, proton pump inhibitor. *Race was self-reported. Genotype determined by the Central Laboratory. Subtype conrmed by phylogenec analysis. § One paent with conrmed GT5a subtype. HCV RNA quaned using the Roche COBAS TaqMan RT-PCR assay, v2.0 or higher. Table 2. Baseline Polymorphisms* Sequence, n (%) GT2 N = 123 GT4 N = 41 GT5 N = 1 GT6 N = 6 None 29 (24) 23 (56) 1 (100) 2 (33) NS3 only 0 0 0 0 NS5A only 93 (76) 17 (42) 0 4 (67) NS3 + NS5A 1 (0.8) 1 (2) 0 0 *Baseline polymorphisms detected at 15% next generaon sequencing threshold in samples that had sequences available for both targets (N) at a key subset of amino acid posions (NS3: 155, 156, 168; NS5A: 24, 28, 30, 31, 58, 92, 93). • 42% (53/126) of paents with GT2 infecon had M at NS5A amino acid posion 31 at baseline; 96% (51/53) of these paents achieved SVR12 Table 3. Adverse Events Event, n (%) 8 Week G/P N = 203 Any AE* 128 (63) AEs leading to discontinuation 0 Serious AE 2 (1) AEs occurring in ≥10% total patients Fatigue 37 (18) Headache 28 (14) Nausea 23 (11) *Includes all AEs regardless of relaon to study drug. One paent experienced cholecyss (Day 30 of treatment) and 1 paent experienced urosepsis (post-treatment Day 15), both assessed as not related to study drug. Table 4. Laboratory Abnormalies Event, n (%) 8 Week G/P N = 203 AST Grade 3 (>5–20 × ULN)* 1 (0.5) ALT Grade 3 (>5–20 × ULN) 0 Total Bilirubin Grade 3 (>3–10 × ULN)* 1 (0.5) AST, aspartate aminotransferase; ALT, alanine aminotransferase; ULN, upper limit of normal. *Post-baseline increase in grade. Post-baseline and post-nadir increase in grade. AST elevaon associated with SAE of cholecyss (described in Table 3) in same paent. • The grade 3 bilirubin elevaon occurred on Day 29 of treatment in a paent who had previously experienced grade 2 elevaons All total bilirubin elevaons were predominately indirect There were no associated post-nadir ALT elevaons • There were no grade 4 or higher lab abnormalies Figure 1. E cacy A. SVR12, ITT Populaon Overall GT2 GT4 GT5 GT6 0 20 40 60 80 100 % Patients With SVR12 97 98 93 100 90 142 145 43 46 2 2 9 10 196 203 Breakthrough Relapse Disconnuation 0: 0 0 0 0 2: 2 0 0 0 2: 1* 1 0 0 3: 0 2 0 1 Missing SVR12 data *Paent disconnued on Day 15 due to loss to follow-up. Paent disconnued on Day 36 due to non-compliance. • Of the paents with missing SVR12 data, at the last study visit, 2 had achieved SVR4 and 1 had achieved SVR8 B. SVR12, mITT* Populaon Overall GT2 GT4 GT5 GT6 0 20 40 60 80 100 % Patients With SVR12 99 98 100 100 100 140 142 43 43 2 2 9 9 194 196 *mITT, modied intent-to-treat; excludes non-virologic failures and 2 paents in the GT2 treatment arm that were idened by phylogenec analysis as having GT1 infecon. • 8-week treatment of DAA-naïve GT2-infected paents with G/P yielded a SVR12 rate of 99% (135/137) that was non-inferior to the historical 95% SVR12 rate (12 weeks SOF/RBV) Table 5. Characteris cs of Paents With Virologic Failure Patient A* Patient B Time of Failure Relapse, post-treatment Day 29 Relapse, post-treatment Day 55 Age/Race/Gender 56-year-old white female 55-year-old white male Genotype/Subtype GT2a GT2a IL28B Genotype C/C C/T Fibrosis Stage F0–F1 F3 Baseline Viral Load 2 870 000 IU/mL 11 700 000 IU/mL Prior Treatment Experience Experienced Experienced Treatment compliant Yes Yes Baseline polymorphisms NS3 None None NS5A L31M L31M Treatment-emergent substitutions at time of failure NS3 None None NS5A None None *Paent had a medical history of gastric bypass. Exposure of GLE on Day 1 and Week 4 was >75% lower than the mean in paents in the same treatment arm; exposure of PIB was comparable to the other paents in the cohort. Measured as the percentage of tablets taken relave to the total tablets expected to be taken during the treatment period; compliance achieved if percentage was 80%. RESULTS

Transcript of SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

Page 1: SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates High SVR Rates in Pati ents With HCV Genotype 2, 4, 5, or 6 Infecti on Without Cirrhosis Following an 8-Week Treatment Durati on Tarek Hassanein1, David Wyles2, Stanley Wang3, Paul Y Kwo4, Mitchell L Shiff man5, Ziad Younes6, Susan Greenbloom7, Catherine A Stedman8, Joseph Sasadeusz9, Humberto Aguilar10, Jeong Heo11, Ran Liu3, Teresa I Ng3, Chih-Wei Lin3, Federico Mensa3

1Southern California GI and Liver Centers and Southern California Research Center, Coronado, California, United States; 2Denver Health Medical Center, Denver, Colorado, United States; 3AbbVie Inc., North Chicago, Illinois, United States; 4Indiana University School of Medicine, Indianapolis, Indiana, United States; 5Liver Insti tute of Virginia, Bon Secours Health System, Newport News and Richmond, Virginia, United States; 6GastroOne, Germantown, Tennessee, United States; 7Toronto Digesti ve Disease Associates, Toronto, Ontario, Canada; 8Christchurch Hospital and University of Otago, Christchurch, New Zealand; 9Island Health Authority, Secti on of Infecti ous Diseases, Victoria, Briti sh Columbia, Canada; 10Louisiana Research Center, Shreveport, Louisiana, United States; 11College of Medicine, Pusan Nati onal University and Medical Research Insti tute, Pusan Nati onal University Hospital, Busan, Republic of Korea

CONCLUSIONS 97% (196/203, ITT) of GT2, 4, 5, or 6-infected pati ents without cirrhosis achieved SVR12 following 8 weeks of G/P

– In DAA-naïve pati ents with GT2 infecti on, 8-week treatment was non-inferior to the historical 95% SVR12 rate achieved with 12 weeks SOF/RBV

– There were no virologic failures in pati ents with GT4–6 infecti on

– Baseline viral load, genotype/subtype, F0–F3 fi brosis stage, presence of baseline polymorphisms, and prior treatment experience with interferon- or SOF-based regimens did not impact achievement of SVR12

– SVR rates were similar to observed rates following 12-week treatment with G/P

G/P for 8 weeks was well-tolerated, with no disconti nuati ons due to AEs, no DAA-related serious AEs, and rare grade 3 or higher lab abnormaliti es (0.5%)

The all-oral, once-daily, IFN- and RBV-free regimen of G/P is highly effi cacious for treatment of pati ents with HCV GT 2, 4, 5, or 6 infecti on without cirrhosis following an 8-week treatment durati on

INTRODUCTION• Hepati ti s C virus genotypes (HCV GT) 2, 4, 5, and 6 have diverse

global distributi ons, accounti ng for approximately 9%, 8%, 1%, and 5% of HCV infecti ons worldwide, respecti vely1

Global Prevalence of HCV Genotype 2, 4, 5, and 6 Infecti on

<10% prevalence

GT2

GT4

GT5

GT6

>10% prevalence of more than one (GT2, 4-6) GT

• A pangenotypic treatment that is safe and highly effi cacious, with a treatment durati on as short as possible, could improve pati ent adherence and access to care

Next Generati on Direct-acti ng Anti virals

In vitro:2,3

• High barrier to resistance• Potent against common NS3 polymorphisms

(eg, posi�ons 80, 155, and 168) and NS5A polymorphisms (eg, posi�ons 28, 30, 31, and 93)

• Addi�ve/synergis�c an�viral ac�vity

Clinical PK & metabolism:

• Oral dosing of 3 pills once-daily

• Minimal metabolism and primary biliary excre�on

• Negligible renal excre�on (<1%)

h bbaarrrrrrriiiiieeeerrrr tttttoooo rrrreeeessssiiiiisssstttttaaannnccceeeent against common NS3 polymorpposi�ons 80, 155, and 168) and NSposi�ons 28, 30, 31, and 93)

Glecaprevir(formerly ABT-493)

pangenotypic NS3/4Aprotease inhibitor

Pibrentasvir(formerly ABT-530)

pangenotypic NS5Ainhibitor

Collec�vely: G/P

G/P is co-formulated and dosed once daily as three 100 mg/40 mg pills for a total dose of 300 mg/120 mg.Glecaprevir was identi fi ed by AbbVie and Enanta.

• In previous phase 2 studies, sustained virologic response at 12 weeks aft er treatment (SVR12) rates of 98% and 100% were achieved following treatment with GLE + PIB (G/P) for 8 weeks in GT2-infected pati ents or 12-weeks in GT 4–6 infected pati ents, respecti vely, with no virologic failures4

OBJECTIVE• SURVEYOR-II, Part 4 is a phase 2, open-label, multi center,

single-arm study that evaluated the safety and effi cacy of an 8-week G/P regimen in pati ents with GT4–6 infecti on and a larger cohort of GT2-infected pati ents

Presented at the 67th Annual Meeti ng of the American Associati on for the Study of Liver Diseases, November 11–15, 2016, Boston, Massachusett s

LB-15

REFERENCES1. Messina, et al. Hepatology. 2015. 2. Ng TI, et al. Abstract 636. CROI, 2014.

3. Ng TI, et al. Abstract 639. CROI, 2014.

4. Kwo, et al. JHep. 2016 (accepted manuscript under revision).

DISCLOSUREST Hassanein: Research/Grant Support: AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, Eisai, Gilead, Idenix, Ikaria, Janssen, La Jolla Pharmaceuti cals, Merck, Mochida, NGM BioPharmaceuti cals, Obalon, Roche, Ocera, Sundise, Salix, Taigen, Takeda, Tobira, Vertex, Vital Therapies; Speaker: Baxter, Bristol-Myers Squibb, Gilead, Janssen, Salix; Advisor: AbbVie, Bristol-Myers Squibb. DL Wyles: Research Grants: AbbVie, Bristol-Myers Squibb, Gilead, Merck, Tacere (paid to the University of California Regents); Consultant: AbbVie, Bristol-Myers Squibb, Gilead, Janssen, Merck. PY Kwo: Grant Support: AbbVie, Bristol-Myers Squibb, Conatus, Eisai, Gilead, Janssen, Merck; Advisor: Abbott , AbbVie, BMS, Gilead, Janssen, Merck, Quest, Alnylam, Durect, DSMB, Inovio. ML Shiff man: Advisor: AbbVie, Achillion, Bristol-Myers Squibb, Gilead, Merck; Grant/Research Support: AbbVie, Achillion, Beckman-Colter, Bristol-Myers Squibb, Gilead, Intercept, Lumena, Novarti s; Speaker: AbbVie, Bayer, Gilead, Janssen, Merck. Z Younes: Grant/Research Support: AbbVie, Gilead, Bristol-Myers Squibb, Idenix, Vertex, Roche, Merck, Janssen, Tibotec; Speaker: AbbVie, Gilead, Intercept, Vertex; Advisor: AbbVie, Gilead. S Greenbloom: Nothing to disclose. CA Stedman: Grant/Research Support: Gilead; Consultant/Advisor: Janssen, AbbVie, Gilead, MSD. J Sasadeusz: Grants: AbbVie, Gilead, Roche; Advisory Board: AbbVie, BMS, Gilead, Merck; Sponsored Lectures: BMS, Gilead. H Aguilar: Grant/Research Support: AbbVie, Genentech, Gilead. J Heo: Advisory Board: Bristol-Meyers Squibb, Gilead Sciences; Research Support: Bristol-Meyers Squibb, Roche. S Wang, TI Ng, R Liu, CW Lin, and F Mensa are employees of AbbVie Inc. and may hold stock or stock opti ons.

ACKNOWLEDGEMENTS The authors would like to express their grati tude to the pati ents and their families who parti cipated in this study. Medical writi ng support was provided by Zoë Hunter, PhD, of AbbVie.

MATERIALS AND METHODSSTUDY DESIGN

GT 2, 4, 5, 6 N = 203

Day 0 Wk 40

Open-label Treatment

Wk 20Wk 8

G/P

300 mg/120 mg*

SVR12 assessment

*G/P is co-formulated and dosed once daily as three 100 mg/40 mg pills for a total dose of 300 mg/120 mg.

KEY INCLUSION CRITERIA

• ≥18 years of age

• BMI ≥18 kg/m2

• Chronic HCV GT2, 4, 5, or 6 infecti on with HCV RNA >1000 IU/mL

• Absence of cirrhosis documented by liver biopsy (eg, METAVIR <4 and ISHAK <5), transient elastography (FibroScan® ≤12.5 kPa) or serum markers (FibroTest® ≤0.48 + APRI <1)

• HCV treatment-naïve or

• Treatment-experienced with interferon (IFN) or pegIFN ± ribavirin (RBV), or sofosbuvir (SOF) + RBV ± pegIFN therapy

KEY EXCLUSION CRITERIA

• Prior HCV treatment experience with any direct-acti ng anti viral (DAA) other than SOF

• Co-infecti on with hepati ti s B or HIV

• ALT >10 × ULN, AST >10 × ULN, direct bilirubin >ULN, albumin <LLN or platelets <90 000 cells/mm3

ENDPOINTS AND ANALYSES

Safety Assessments

Primary Efficacy Endpoint

Secondary EfficacyEndpoints

The percentage of pa�ents with on-treatment virologic failure, post-treatment relapse and SVR4

The percentage of pa�ents with SVR12 (HCV RNA <25 IU/mL 12 weeks a�er the last dose of study drug)

The percentage of GT2-infected DAA-naïve pa�ents with SVR12 will be non-inferior to the historical 95% SVR12 rate (12 weeks SOF + RBV) if the lower confidence bound of the 2-sided 95% confidence interval is >89%

Adverse events (AEs) and laboratory abnormali�es were assessed in all pa�ents receiving at least 1 dose of study drug

Next genera�on sequencing (detec�on threshold = 15%) performed to iden�fy baseline polymorphisms and, treatment-emergent subs�tu�ons in NS3 and NS5A

Addi�onal Assessments

Table 1. Baseline Demographics and Disease Characteristi cs

Characteristic8 Week G/P

N = 203

Male, n (%) 98 (48)

Race, n (%)*

White 155 (76)

Black 21 (10)

Asian 23 (11)

Other 4 (2)

Age, median (range), years 55 (19-83)

BMI, median (range), kg/m2 26.8 (17.3–65.7)

Genotype, n (%)†

2 145 (71)

Subtype‡ a/b/c/q 21/103/3/1

4 46 (23)

Subtype‡ a/d/f/k/m/v/v–q 26/8/1/1/1/3/1

5 2 (1)§

6 10 (5)

Subtype‡ a/e/l 3/3/1

HCV Treatment-naïve, n (%) 176 (87)

Treatment-experienced, n (%) 27 (13)

IFN-based 21 (10)

SOF-based 6 (3)

HCV RNA, median (range), log10

IU/mL¶ 6.45 (0.75–7.62)

Baseline Fibrosis Stage, n (%)

F0–F1 170 (84)

F2 12 (6)

F3 21 (10)

Non-CC IL28B genotype, n (%) 115 (57)

PPI use 19 (9)

BMI, body mass index; IFN, interferon; SOF, sofosbuvir; HCV, hepati ti s C virus; PPI, proton pump inhibitor.*Race was self-reported. †Genotype determined by the Central Laboratory. ‡Subtype confi rmed by phylogeneti c analysis.§One pati ent with confi rmed GT5a subtype. ¶HCV RNA quanti fi ed using the Roche COBAS TaqMan RT-PCR assay, v2.0 or higher.

Table 2. Baseline Polymorphisms*

Sequence, n (%)GT2

N = 123GT4

N = 41GT5

N = 1GT6

N = 6

None 29 (24) 23 (56) 1 (100) 2 (33)

NS3 only 0 0 0 0

NS5A only 93 (76) 17 (42) 0 4 (67)

NS3 + NS5A 1 (0.8) 1 (2) 0 0

*Baseline polymorphisms detected at 15% next generati on sequencing threshold in samples that had sequences available for both targets (N) at a key subset of amino acid positi ons (NS3: 155, 156, 168; NS5A: 24, 28, 30, 31, 58, 92, 93).

• 42% (53/126) of pati ents with GT2 infecti on had M at NS5A amino acid positi on 31 at baseline; 96% (51/53) of these pati ents achieved SVR12

Table 3. Adverse Events

Event, n (%)8 Week G/P

N = 203

Any AE* 128 (63)

AEs leading to discontinuation 0

Serious AE 2 (1)†

AEs occurring in ≥10% total patients

Fatigue 37 (18)

Headache 28 (14)

Nausea 23 (11)

*Includes all AEs regardless of relati on to study drug. †One pati ent experienced cholecysti ti s (Day 30 of treatment) and 1 pati ent experienced urosepsis (post-treatment Day 15), both assessed as not related to study drug.

Table 4. Laboratory Abnormaliti es

Event, n (%)8 Week G/P

N = 203

AST Grade 3 (>5–20 × ULN)* 1 (0.5)‡

ALT Grade 3 (>5–20 × ULN)† 0

Total Bilirubin Grade 3 (>3–10 × ULN)* 1 (0.5)

AST, aspartate aminotransferase; ALT, alanine aminotransferase; ULN, upper limit of normal. *Post-baseline increase in grade.†Post-baseline and post-nadir increase in grade. ‡AST elevati on associated with SAE of cholecysti ti s (described in Table 3) in same pati ent.

• The grade 3 bilirubin elevati on occurred on Day 29 of treatment in a pati ent who had previously experienced grade 2 elevati ons

– All total bilirubin elevati ons were predominately indirect – There were no associated post-nadir ALT elevati ons

• There were no grade 4 or higher lab abnormaliti es

Figure 1. Effi cacy A. SVR12, ITT Populati on

Overall GT2 GT4 GT5 GT60

20

40

60

80

100

%Pa

tien

tsW

ith

SVR

12

97 98 93 100 90

142145

4346

22

910

196203

BreakthroughRelapse

Discon�nuation

0: 0 0 0 0 2: 2 0 0 0 2: 1* 1† 0 0 3: 0 2 0 1Missing SVR12 data

*Pati ent disconti nued on Day 15 due to loss to follow-up. †Pati ent disconti nued on Day 36 due to non-compliance.

• Of the pati ents with missing SVR12 data, at the last study visit, 2 had achieved SVR4 and 1 had achieved SVR8

B. SVR12, mITT* Populati on

Overall GT2 GT4 GT5 GT60

20

40

60

80

100

%Pa

tien

tsW

ith

SVR

12

99 98 100 100 100

140142

4343

22

99

194196

*mITT, modifi ed intent-to-treat; excludes non-virologic failures and 2 pati ents in the GT2 treatment arm that were identi fi ed by phylogeneti c analysis as having GT1 infecti on.

• 8-week treatment of DAA-naïve GT2-infected pati ents with G/P yielded a SVR12 rate of 99% (135/137) that was non-inferior to the historical 95% SVR12 rate (12 weeks SOF/RBV)

Table 5. Characteristi cs of Pati ents With Virologic FailurePatient A* Patient B

Time of Failure Relapse, post-treatment Day 29 Relapse, post-treatment Day 55

Age/Race/Gender 56-year-old white female 55-year-old white male

Genotype/Subtype GT2a GT2a

IL28B Genotype C/C C/T

Fibrosis Stage F0–F1 F3

Baseline Viral Load 2 870 000 IU/mL 11 700 000 IU/mL

Prior Treatment Experience Experienced Experienced

Treatment compliant† Yes Yes

Baseline polymorphisms

NS3 None None

NS5A L31M L31M

Treatment-emergent substitutions at time of failure

NS3 None None

NS5A None None

*Pati ent had a medical history of gastric bypass. Exposure of GLE on Day 1 and Week 4 was >75% lower than the mean in pati ents in the same treatment arm; exposure of PIB was comparable to the other pati ents in the cohort. †Measured as the percentage of tablets taken relati ve to the total tablets expected to be taken during the treatment period; compliance achieved if percentage was ≥80%.

RESULTS

Page 2: SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

Drug-Drug Interactions Between Direct Acting Antivirals Glecaprevir (ABT-493) and Pibrentasvir (ABT-530) with Angiotensin II Receptor Blockers Losartan or Valsartan Matthew P. Kosloski, Sandeep Dutta, Qi Jiang, Betty Yao, Armen Asatryan, Federico Mensa, Jens Kort, Wei Liu AbbVie Inc., North Chicago, Illinois, United States

854

• Non-compartmental analysis was performed with Phoenix WinNonlin v6.3 including estimation of maximum plasma concentration (Cmax), area under the curve (AUC) from time zero to infinity (AUCinf; losartan, losartan carboxylic acid, and valsartan), AUC from time zero to 24 hours (AUC24; GLE and PIB), and trough concentrations (C24; GLE and PIB).

• The ratio of central values and 90% confidence intervals (CI) were calculated for log-transformed pharmacokinetic parameters on Day 10 (test) versus Day 1 (reference) in each arm for losartan, losartan carboxylic acid, or valsartan, and for Day 10 (test) versus Day 9 (reference) in each arm for GLE and PIB.

• Safety metrics including monitoring of adverse events, vital signs, physical examinations, ECGs, and laboratory tests were assessed throughout the study.

MAIN INCLUSION CRITERIA

• Healthy male and female subjects between 18 and 55 years old.

INTRODUCTION • The direct acting antiviral (DAA) combination of

glecaprevir (GLE; formerly ABT-493), an NS3/4A protease inhibitor discovered by AbbVie and Enanta, and pibrentasvir (PIB; formerly ABT-530), an NS5A inhibitor, achieved high sustained virologic response rates across chronic hepatitis C virus (HCV) genotype 1-6 infection in Phase 2 studies with a favorable safety profile. The once daily fixed dose combination of GLE/PIB 300 mg/120 mg is currently being studied in Phase 3 clinical studies.

• Angiotensin II receptor blockers (ARBs) including losartan and valsartan are used in the treatment of hypertension and other cardiovascular conditions, and are commonly administered in HCV infected subjects with such comorbidities.

• Losartan is a competitive inhibitor of the angiotensin II receptor type 1 (AT1) and forms an active acid metabolite that is 10-40 times more potent of an AT1 inhibitor than the parent compound.

• Valsartan undergoes limited metabolism to form a largely inactive primary metabolite with 1/200th the potency of the parent compound.

OBJECTIVE

METHODS

RESULTS (continued)

• This study was designed to assess the pharmacokinetics (PK), safety, and tolerability of GLE and PIB coadministered with losartan or valsartan and evaluate the drug-drug interaction potential of these agents.

STUDY DESIGN

• Phase 1, single-center, non-fasting, open label study (Figure 1) in 12 healthy subjects. • All enrolled subjects completed the study and were

included in pharmacokinetic and safety analyses.

Figure 2. Arm 1: Losartan, Losartan Carboxylic Acid, GLE, and PIB Plasma Concentration-Time Profiles

CONCLUSIONS

DISCLOSURES

• GLE and PIB increased losartan, losartan carboxylic acid, and valsartan exposures; however, the increases were not considered clinically significant and no dose adjustment is required when GLE + PIB are coadministered with losartan or valsartan.

• GLE or PIB exposures were not affected by losartan or valsartan.

This study was funded by AbbVie. AbbVie contributed to the study design, research, and interpretation of data, writing, reviewing, and approving the publication. SD is a former AbbVie employee and may hold AbbVie stocks or options. All other authors are AbbVie employees and may hold AbbVie stocks or options.

Presented at the 2016 AASLD Liver Meeting, November 11–15, 2016, Boston, MA

SAFETY RESULTS • No serious adverse events occurred in the study. In Arm

1, a single adverse event of Grade 2 viral infection was experienced by one subject following coadministration of GLE, PIB, and losartan. In Arm 2, a single adverse event of Grade 1 rhinorrhoea was experienced by one subject during administration of GLE and PIB. No other adverse events were reported.

• No clinically significant vital signs, ECGs, or laboratory abnormalities were observed in the study.

Figure 1. Study Design

ARM 1 (N=12) ARM 2 (N=12) Mean ± SD Min – Max Mean ± SD Min – Max

Age (years) 37 ± 11 22 – 52 32 ± 7 22 – 48

Weight (kg) 82 ± 12 57 – 97 80 ± 12 60 – 101

Height (cm) 174 ± 8 162 – 183 174 ± 8 160 – 185

Sex 2 Females (17%), 10 Males (83%) 3 Females (25%), 9 Males (75%)

Race 5 White (42%), 6 Black (50%),

1 Multi-race (8%) 6 White (50%), 6 Black (50%)

Figure 4. Arm 2: Valsartan, GLE, and PIB Plasma Concentration-Time Profiles

Figure 3. Interactions Between GLE and PIB with Losartan

Figure 5. Interactions Between GLE and PIB with Valsartan

• Exposures were higher for losartan (↑Cmax 151% and ↑AUCinf 56%) when administered with multiple doses of GLE + PIB, relative to losartan alone.

• Losartan active carboxylic metabolite Cmax was 118% higher, while AUCinf was similar (≤ 14% difference).

• Based on losartan prescribing information, similar magnitudes of exposure increases caused by other drugs or in special populations did not require dose adjustment.

• GLE and PIB exposures were similar with and without losartan (≤17% difference).

• Valsartan exposures were slightly higher (↑Cmax 36% and ↑AUCinf 31%) when administered with GLE and PIB.

• Based on valsartan prescribing information, similar magnitudes of exposure increases in special populations did not require dose adjustment.

• GLE and PIB exposures were similar with and without losartan (≤15% difference), except GLE C24 which was 23% lower.

METHODS (continued)

• Intensive PK samples for determination of losartan, losartan carboxylic acid, or valsartan plasma concentrations were collected on Days 1 and 10 of each arm. Intensive PK samples to evaluate GLE and PIB concentrations were collected on Days 9 and 10 of each Arm.

RESULTS

Day 1 Day 2 Days 3 - 9 Day 10 Day 11

Arm 1 N=12

Losartan 50 mg, single dose (Day 1)

GLE 300 mg + PIB 120 mg QD (Days 3 – 11)

Losartan 50 mg, single dose (Day 10)

Arm 2 N=12

Valsartan 80 mg, single dose (Day 1)

GLE 300 mg + PIB 120 mg

QD (Days 3 – 11)

Valsartan 80 mg,

single dose (Day 10)

Table 1. Subject Demographics and Disposition

Time (h)

0 4 8 12 16 20 24 28 32 36 40 44 48

Vals

arta

n Pl

asm

a C

once

ntra

tion

(ng/

mL)

0

500

1000

1500

2000

2500

Day 1: Valsartan aloneDay 10: GLE + PIB + Valsartan

Time (h)

0 2 4 6 8 10 12 14 16 18 20 22 24

GLE

Pla

sma

Con

cent

ratio

n (n

g/m

L)

0

250

500

750

1000

1250

1500Day 9: GLE + PIB aloneDay 10: GLE + PIB + Valsartan

Time (h)

0 2 4 6 8 10 12 14 16 18 20 22 24

PIB

Pla

sma

Con

cent

ratio

n (n

g/m

L)

0

50

100

150

200

250

300

350

400

Day 9: GLE + PIB aloneDay 10: GLE + PIB + Valsartan

Time (h)

0 4 8 12 16 20 24

Losa

rtan

Pla

sma

Con

cent

ratio

n (n

g/m

L)

0

50

100

150

200

250

Day 1: Losartan aloneDay 10: GLE + PIB + Losartan

Time (h)

0 4 8 12 16 20 24 28 32 36 40 44 48

Losa

rtan

Car

boxy

lic A

cid

Plas

ma

Con

cent

ratio

n (n

g/m

L)

0

50

100

150

200

250

300

350

400

450

500 Day 1: Losartan aloneDay 10: GLE + PIB + Losartan

Time (h)

0 4 8 12 16 20 24

GLE

Pla

sma

Con

cent

ratio

n (n

g/m

L)

0

200

400

600

800

1000

1200

1400Day 9: GLE + PIB aloneDay 10: GLE + PIB + Losartan

Time (h)

0 4 8 12 16 20 24

PIB

Pla

sma

Con

cent

ratio

n (n

g/m

L)

0

50

100

150

200

250

300

Day 9: GLE + PIB aloneDay 10: GLE + PIB + Losartan

Page 3: SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

• Subjects received single doses of the GLE 300 mg + PIB 120 mg combination in Period 1 three hours prior to the start of hemodialysis and in Period 2 on the day prior to a scheduled hemodialysis session.

• Intensive pharmacokinetic samples for determination of GLE and PIB plasma concentrations were collected up to 24 hours after dosing in each period. Additionally, arterial (predialyzer) and venous (postdialyzer) blood samples were collected during dialysis.

• Non-compartmental analysis was performed with Phoenix WinNonlin v6.3 including estimation of maximum plasma concentration (Cmax) and area under the curve (AUC) from time zero to the last sampling point (AUCt), AUC during dialysis for arterial (AUCarterial) or venous (AUCvenous) samples, apparent oral clearance (CL/F), and clearance due to dialysis (CLD). CLD was derived from differences in arterial and venous exposures during dialysis.

• Unbound fractions of GLE and PIB were assessed ex vivo in plasma samples collected prior to the start of and immediately after dialysis.

• The ratio of central values and 90% confidence intervals (CI) were calculated for log-transformed pharmacokinetic parameters in Period 1 (Day of dialysis) versus Period 2 (Non-dialysis day) for GLE and PIB.

• Safety was evaluated throughout the study with assessment of adverse events, vital signs, ECGs and clinical laboratory tests.

MAIN INCLUSION CRITERIA

• Male and female subjects between 18 and 75 years old with ESRD receiving hemodialysis for at least 1 month.

INTRODUCTION

• The direct acting antiviral (DAA) combination of glecaprevir (GLE; formerly ABT-493), an NS3/4A protease inhibitor discovered by AbbVie and Enanta, and pibrentasvir (PIB; formerly ABT-530), an NS5A inhibitor, achieved high sustained virologic response rates across chronic hepatitis C virus (HCV) genotype 1-6 infection in Phase 2 studies with a favorable safety profile, and a once daily fixed dose combination of GLE/PIB 300 mg/120 mg is currently being evaluated in Phase 3 studies.

• In a Phase 1 study conducted in non-HCV infected subjects, GLE and PIB exposure as determined by AUCinf demonstrated a maximum increase of 56% and 46%, respectively, in subjects with end stage renal disease (ESRD) not on dialysis relative to normal subjects. Cmax was similar across groups (≤ 25% difference).1

• Exposure changes in GLE and PIB in non-HCV infected subjects with renal impairment were not considered clinically significant and based on these findings, dose-adjustment of GLE or PIB is not required in subjects with any degree of renal impairment not on dialysis.

OBJECTIVE

METHODS

METHODS (CONTINUED) RESULTS

• This study evaluated the impact of hemodialysis on the pharmacokinetics and safety of GLE and PIB in ESRD subjects requiring dialysis.

STUDY DESIGN

• Phase 1, non-fasting, open label study (Figure 1) in N=8 subjects with ESRD requiring hemodialysis.

• All enrolled subjects completed the study and were included in pharmacokinetic and safety analyses.

• The typical hemodialysis session ended approximately 7 hours after dosing of study drug in Period 1, or 4 hours after the start of dialysis.

Table 1. Subject Demographics and Disposition

• Concentration-time profiles were similar for GLE and PIB in arterial and venous plasma samples collected during dialysis

CONCLUSIONS

REFERENCES

DISCLOSURES

• GLE and PIB exposures were not affected by hemodialysis.

• No dose-adjustment is needed when GLE and PIB are administered in subjects with renal impairment, with or without dialysis.

1. Kosloski MP, Dutta S, Zhao W, Pugatch D, Mensa F, Kort J, Liu W. “Pharmacokinetics, Safety, and Tolerability of Next Generation Direct Acting Antivirals ABT-493 and ABT-530 in Subjects with Renal Impairment.” Presented at: EASL International Liver Congress; April 13-17, 2016; Barcelona Spain. Poster THU-230.

This study was funded by AbbVie. AbbVie contributed to the study design, research, and interpretation of data, writing, reviewing, and approving the publication. SD is a former AbbVie employee and may hold AbbVie stocks or options. All other authors are AbbVie employees and may hold AbbVie stocks or options.

Presented at the 2016 AASLD Liver Meeting, November 11–15, 2016, Boston, MA

Hemodialysis Does Not Affect the Pharmacokinetics of Glecaprevir (ABT-493) or Pibrentasvir (ABT-530) Matthew P. Kosloski, Sandeep Dutta, Weihan Zhao, David Pugatch, Federico Mensa, Jens Kort, Wei Liu AbbVie Inc., North Chicago, Illinois, United States

Period 1

Washout

≥ 7 Days

Period 2

Day 1 Day 2 Day 1 Day 2

DIALYSIS DIALYSIS

GLE + PIB GLE + PIB

Mean ± SD (N = 8) Min – Max

Age (years) 57 ± 9 47 – 73

Weight (kg) 80 ± 23 52 – 122

Height (cm) 170 ± 9 156 – 183

Sex 2 Females (25%), 6 Males (75%)

Race 8 Black (100%)

Table 2. Geometric Mean (Mean, CV%) GLE and PIB Pharmacokinetic Parameters

Pharmacokinetic

Parameter (units)

GLE PIB

Period 1,

Day of Dialysis

Period 2,

Non-Dialysis Day

Period 1,

Day of Dialysis

Period 2,

Non-Dialysis Day

Cmax (ng/mL) 671 (883, 69) 723 (1050, 101) 128 (150, 48) 156 (193, 54)

Tmaxs (h)a 3.6 (2.0 to 9.0) 2.5 (2.0 to 5.0) 5.5 (3.3 to 9.0) 5.0 (3.0 to 6.0)

AUCt (ng∙h/mL) 3010 (3820, 69) 2840 (4090, 102) 1020 (1180, 47) 1120 (1360, 54)

AUCarterial (ng∙h/mL) 1580 (2000, 69) -- 358 (457, 54) --

AUCvenous (ng∙h/mL) 1560 (1980, 67) -- 377 (481, 52) --

CLD (L/h)b (0.576, 119) -- (0.00336, 283) --

CL/F (L/h)b,c (125, 74) (144, 94) (148, 90) (152, 117) a. Median (Minimum to Maximum); b. (Mean, CV%); c. Calculated as Dose/AUCt

Figure 4. The Effect of Hemodialysis on GLE and PIB Pharmacokinetics

• GLE and PIB exposures were similar (≤ 18% difference) when GLE + PIB were administered three hours prior to the start of hemodialysis or on a non-dialysis day.

SAFETY RESULTS • Adverse events were rare; a single Grade 1

adverse event was reported.

• No clinically significant vital signs, ECGs, or laboratory abnormalities were observed in the study.

• Mean values of CLD represented a minimal portion of CL/F for GLE (< 1%) and PIB (< 0.005%).

• Fraction of unbound drug pre- or post- dialysis was similar and ranged between 2.5 to 2.9% (GLE) or 0.019 to 0.029% (PIB).

Figure 1 Study Design

Time (h)

0 3 6 9 12 15 18 21 24

GL

E C

on

ce

ntr

ati

on

(n

g/m

L)

0

200

400

600

800

1000 Dialysis DayNon-Dialysis Day

Hemodialysis begun

(Period 1 Day 1)

Hemodialysis ended

(Period 1 Day 1)

Time (h)

0 3 6 9 12 15 18 21 24

PIB

Co

nc

en

tra

tio

n (

ng

/mL

)

0

50

100

150

200 Dialysis DayNon-Dialysis Day

Hemodialysis begun

(Period 1 Day 1)

Hemodialysis ended

(Period 1 Day 1)

Time Since Dialysis Start (h)

0 1 2 3 4

GL

E C

on

ce

ntr

ati

on

(n

g/m

L)

0

200

400

600

800

1000

Arterial PlasmaVenous Plasma

Time Since Dialysis Start (h)

0 1 2 3 4

PIB

Co

nc

en

tra

tio

n (

ng

/mL

)

0

50

100

150

200

Arterial Plasma Venous Plasma

Figure 3. Mean GLE and PIB Arterial and Venous Plasma Concentrations During Hemodialysis

Figure 2. Mean GLE and PIB Plasma Concentration-Time Profiles

• Maximum concentrations of GLE and PIB reached by 2.5 and 5 hours post dose on non-dialysis day. Therefore, starting dialysis 3-hours post dose in Period 1 ensured the assessment of maximum dialysis effect on GLE and PIB exposure.

Central Value Ratio and 90% Confidence Interval

0.1 1 10

PIB

GLE

CmaxAUCt

0.93 (0.58 - 1.50)

0.82 (0.63 - 1.05)

1.06 (0.71 - 1.59)

0.91 (0.73 - 1.14)

855

Page 4: SURVEYOR-II, Part 4: Glecaprevir/Pibrentasvir Demonstrates ... ·

Analysis of HCV Variants in the MAGELLAN-I Study (Part 1): ABT-493 and ABT-530 Combinati on Therapy of Genotype 1-Infected Pati ents Who Had Failed Prior Direct Acti ng Anti viral-Containing RegimensTeresa Ng, Tami Pilot-Mati as, Rakesh Tripathi, Gretja Schnell, Preethi Krishnan, Thomas Reisch, Jill Beyer, Tanya Dekhtyar, Michelle Irvin, Liangjun Lu, Armen Asatryan, Federico Mensa, Jens Kort, Christi ne CollinsAbbVie Inc., North Chicago, Illinois, United States

SUMMARY

High SVR12 rate among 50 DAA-experienced HCV GT1-infected pati ents without cirrhosis

– 2 pati ents experienced virologic failure; 2 pati ents were lost-to-follow-up

– RBV did not appear to increase SVR12 rate

Resistance analysis of pati ent baseline samples – No signifi cant diff erence in frequency of baseline polymorphisms in NS3 and/or NS5A based on 2% or 15% NGS detecti on threshold

– Most pati ents (≥80%) had at least 1 baseline polymorphism in NS3 or NS5A

High SVR12 rate was achieved (46/48 pati ents, mITT) despite the high prevalence of baseline polymorphisms, including in pati ents with NS5A polymorphisms

MAGELLAN-I Part 2 is evaluati ng a RBV-free regimen of GLE/PIB in a larger group of DAA-experienced pati ents with GT1, 4, 5, or 6 infecti on, including compensated cirrhoti cs

BACKGROUNDHCV DIRECT ACTING ANTIVIRAL DAA TREATMENT FAILURE

• Although DAA therapies achieve high SVR rates in most HCV-infected pati ents, treatment failure may occur

• DAA-treatment failure is oft en associated with viral resistance due to – Baseline amino acid polymorphisms – Treatment-emergent amino acid substi tuti ons

• Amino acid substi tuti ons in diff erent HCV targets persist for diff erent periods of ti me – NS3 substi tuti ons usually become undetectable within 1 year of post-treatment1

– NS5A substi tuti ons may persist beyond 2 years of post-treatment1,2

– Some NS5B substi tuti ons may persist beyond 1 year of post-treatment1

• Pati ents with baseline NS5A polymorphisms may achieve lower SVR rates when treated with NS5A-containing DAA regimens

– LDV/SOF: 60% SVR12 in GT1a (33% if Y93H/N were present)3,4

– GZR/EBR: 70% SVR12 in GT1a5,6

– SOF/VEL: 88% SVR12 in GT37

AbbVie’s NEXT GENERATION HCV DAAS

• Glecaprevira (GLE; ABT-493): NS3/4A protease inhibitor (PI) • Pibrentasvir (PIB; ABT-530): NS5A inhibitor • In vitro, GLE and PIB have each demonstrated8,9

– Potent anti viral acti vity against major HCV genotypes – High barrier to development of resistance – Potent anti viral acti vity against common substi tuti ons that confer resistance to previous generati ons of HCV PIs or NS5A inhibitors

Table 1. Anti viral Acti vity of GLE and PIB

DAA

Stable HCV Replicon EC50

GT1a GT1b GT2a JFH-1 GT2b GT3a GT4a GT5a GT6a

GLE (nM)a 0.85 0.94 2.2 4.6 1.9 2.8 0.12b 0.86

PIB (pM) 1.8 4.3 5.0 1.9 2.1 1.9 1.4 2.8aGLE was identi fi ed by AbbVie and Enanta.bTransient replicon results.

OBJECTIVES• MAGELLAN-I (Part 1) study: Retreatment of HCV GT1-infected pati ents who had failed prior

DAA therapy• Resistance analysis: Evaluati on of viral sequences from HCV GT1-infected pati ents who were

treated with the combinati on of GLE and PIB in MAGELLAN-I (Part 1) study – Assessed prevalence of NS3 and NS5A amino acid polymorphisms in baseline samples collected before GLE/PIB treatment

Since pati ents’ HCV amino acid sequences prior to all previous DAA therapies were unknown, an amino acid detected in a pati ent baseline sample that was diff erent than the reference sequence was defi ned as a polymorphism

– Identi fi ed and characterized treatment-emergent substi tuti ons

METHODS• Clinical samples

– Next generati on sequencing (NGS) was performed on the HCV NS3/4A and NS5A genes from baseline and post-baseline pati ent samples

– Amino acid polymorphisms/substi tuti ons (NGS detecti on threshold of 2% or 15%) at the following signature resistance-associated positi ons for NS3/4A protease or NS5A inhibitor class were analyzed

NS3• GT1a: 36, 43, 54, 55, 56, 80, 107, 122, 132, 155, 156, 158, 168, and 170

• GT1b: 36, 54, 55, 56, 80, 107, 122, 155, 156, 158, 168, 170, and 175

NS5A• GT1a: 24, 28, 29, 30, 31, 32, 58, 62, 92, and 93

• GT1b: 24, 28, 29, 30, 31, 32, 54, 58, 62, 92, and 93

• In vitro studies – Resistance analyses were performed by introducing NS3 or NS5A substi tuti ons, as appropriate, into the corresponding wild-type replicons and testi ng their drug suscepti bility in transient replicon assays

• Prior DAAs used: – NS3/4A PIs: Asunaprevir (ASV); boceprevir; (BOC); faldaprevir (FDV); paritaprevir (PTV); simeprevir (SMV); sovaprevir (SVP); telaprevir (TVR); vedroprevir (VDV)

– NS5A inhibitors: Daclatasvir (DCV); ledipasvir (LDV); ombitasvir (OBV); odalasvir (ODV); ravidasvir (RDV); samatasvir (SAM)

– NS5B polymerase inhibitors: Beclabuvir (BCV); dasabuvir (DSV); deleobuvir (DBV); sofosbuvir (SOF); VX-222; TMC-647055

MAGELLAN-I (Part 1) Study Design • MAGELLAN-I (Part 1) is an open-label, multi center, randomized phase 2 trial in

DAA-experienced pati ents with GT1 infecti on and no cirrhosis

0 24 Weeks

Arm A GT1

N = 6 GLE

36

Arm B GT1

N = 22

Treatment Period Post-Treatment Period

SVR12

SVR12

12

Arm C GT1

N = 22

SVR12

PIB

GLE PIB

GLE PIB RBV

200 mg 80 mg

300 mg 120 mg

300 mg 120 mg 800 mg

Prio

r H

CV D

AA

Tre

atm

ent

Expe

rien

ced

*

*RBV (ribavirin) dosed once-daily.

• Total GT1 pati ents = 50 – GT1a pati ents = 42; GT1b pati ents = 8

Presented at the 67th Annual Meeti ng of the American Associati on for the Study of Liver Diseases, November 11–15, 2016, Boston, Massachusett s

849

REFERENCES 1. Krishnan P, et al. J Hepatol. 2015;62:S220.

2. Dvory-Sobol H, et al. J Hepatol. 2015;62:S221.

3. Zeuzem S, et al. Hepatology. 2015;62:254A.

4. Lawitz E, et al. J Hepatol. 2015;62:S192.

5. ZEPATIER prescribing informati on.

6. Jacobson IM, et al. AASLD 2015.

7. Foster, et al. N. Engl. J Med. 2015;373(27):2608–17.

8. Ng TI, et al. Abstract 636. 21st CROI 2014.

9. Ng TI, et al. Abstract 639. 21st CROI 2014.

10. Lenz O, et al. AAC. 2010;54:1878–87.

11. Hazuda D, et al. HepDART 2013.

12. Susser S, et al. Hepatology. 2009;50:1709–18.

13. McPhee F, et al. AAC. 2012; 56:3670–81.

14. Howe A, et al. Clin Infec Diseases. 2014;59:1657–65.

15. Chase R, et al. 8th IAPAC 2013.

16. Pilot-Mati as T, et al. AAC. 2015;59:988–97.

17. Wong KA, et al. AAC. 2013;57:6333–40.

18. Cheng G, et al. AAC. 2016;60:1847–53.

19. Krishnan P, et al. AAC. 2015;59:979–87.

20. Fridell RA, et al. AAC. 2010;54:3641–50.

21. Wang C, et al. AAC. 2013;57:2054–65.

22. Cheng G, et al. 48th EASL 2013.

23. Liu R, et al. AAC. 2015;59:6922–9.

DISCLOSURESAll authors are employees of AbbVie. The design, study conduct, and fi nancial support for this research were provided by AbbVie. AbbVie parti cipated in the interpretati on of data, review, and approval of the publicati on.

GLE IS ACTIVE AGAINST COMMON GT1 NS3 SUBSTITUTIONSFigure 1A. GLE Demonstrates an Improved Resistance Profi le Relati ve to 1st Generati on NS3/4A PIs

0.1

1

10

100

1000

≥10000

1a W

TV

36M

Q80

KR1

55K

A15

6TD

168A

D16

8E

D16

8V

1b W

T

T54A

R155

K

A15

6T

A15

6V

D16

8A

D16

8V

V17

0A

EC50

(nM

) GLESMVBOCTVR

0.1

1

10

100

000

000

1aW

TV

36M

Q80

KR1

55K

156T

168A

168E

68V

WT

4A 5K 6T V A

EC50

(nM

)

GLE: Glecaprevir; SMV: Simeprevir10,11; BOC: Boceprevir10,11,12; TVR: Telaprevir.10,13

Figure 1B. GLE Demonstrates an Improved Resistance Profi le Relati ve to 2nd Generati on NS3/4A PIs

0.1

1

10

100

1000

10000

1a W

TV

36M

Q80

K

R155

K

A15

6T

D16

8A

D16

8E

D16

8V

1b W

T

T54A

R155

K

A15

6T

A15

6V

D16

8A

D16

8V

V17

0A

EC50

(nM

) GLE

GZR

PTV

GLE: Glecaprevir; GZR: Grazoprevir11,14,15; PTV: Paritaprevir.16

• GLE is potent against common GT1 NS3 substi tuti ons at amino acid positi ons 80, 155, and 168• A156T/V in GT1 confer resistance to GLE, but these substi tuti ons have low viral fi tness and are rarely detected clinically

PIB IS ACTIVE AGAINST COMMON GT1 NS5A SUBSTITUTIONSFigure 2A. PIB Demonstrates an Improved Resistance Profi le Relati ve to 1st Generati on NS5A Inhibitors

1

10

100

1000

10000

≥1000001a

WT

M28

T

M28

V

Q30

E

Q30

H

Q30

R

L31M

L31V

Y93C

Y93H

Y93N

1b W

T

L31V

Y93H

Y93N

EC50

(pM

) PIB

LDV

OBV

DCV

PIB: Pibrentasvir; LDV: Ledipasvir17,18; OBV: Ombitasvir19; DCV: Daclatasvir.20,21

Figure 2B. PIB Demonstrates an Improved Resistance Profi le Relati ve to 2nd Generati on NS5A Inhibitors

1

10

100

1000

10000

100000

1a W

T

M28

T

M28

V

Q30

E

Q30

H

Q30

R

L31M

L31V

Y93C

Y93H

Y93N

1b W

T

L31V

Y93H

Y93N

EC50

(pM

) PIB

VEL

EBR

PIB: Pibrentasvir; VEL: Velpatasvir22; EBR: Elbasvir.23

• PIB is highly acti ve against common GT1 NS5A substi tuti ons at amino acid positi ons 28, 30, 31, and 93 that confer resistance to 1st and 2nd generati on NS5A inhibitors• Of note, PIB retains acti vity against GT1 Y93 substi tuti ons; many 1st and 2nd generati on NS5A inhibitors have signifi cantly lower acti vity against these substi tuti ons

MAGELLAN I PART 1 Figure 3. SVR12 Rate by ITT and mITT Analysis

SV

R1

2,

%P

ati

en

ts

0

1 0

2 0

3 0

4 0

5 0

6 0

7 0

8 0

9 0

1 0 0

1 0 0

6

6

9 5

1 9

2 2

2 1

2 2

8 6

6

6

1 9

2 0

2 1

2 2

1 0 0 9 5 9 5

IT T A n a ly s is m IT T A n a ly s is

G L E

P I B

R B V

2 0 0 3 0 0 3 0 0

8 0 1 2 0 1 2 0

8 0 0

2 0 0 3 0 0 3 0 0

8 0 1 2 0 1 2 0

8 0 0

B r e a k t h r o u g h

L T F U

0 0 1

0 0 2 *

0 0 1

R e la p s e 0 1 0 0 1 0

A B C A B C

*Two GT1a pati ents were lost to follow-up; both had undetectable HCV RNA at post-treatment Week 8.ITT: intent-to-treat; mITT: modifi ed intent-to-treat; SVR12: sustained virologic response at post-treatment Week 12; LTFU: lost-to-follow-up.

Table 2. Prior DAA Treatment Regimens

NS3/4A PINS5A

InhibitorNS5B

Inhibitor OthersTotal No. of Patients

Treated

BOC PR 10

TVR PR 8

LDV SOF 8

SMV SOF ± RBV 8

PTV/r OBV DSV ± RBV 4

FDV RDV DBV ± RBV 4

SMV SAM ± TMC-647055 ± RBV 3

DCV ± PR 2

ASV DCV ± BCV ± PR 2

Others * 6

*Other DAA-containing regimens with combinati ons/DAAs not listed above.PR, peginterferon plus ribavirin; RBV, ribavirin; r, ritonavir.

• Most common prior DAA regimens – BOC + PR, TVR + PR, LDV + SOF (Harvoni), and SMV/SOF ± RBV

• All pati ents who failed LDV + SOF (Harvoni) or SMV/SOF ± RBV achieved SRV12 in this study• 4 pati ents previously failed ≥2 diff erent DAA-containing regimens

– 75% (3/4) of them achieved SVR12 in this study

Table 3. Two Pati ents Experienced Virologic Failure

Patient IDPrior

Treatment AbbVie Regimen Subtype Time PointNS3 Polymorphism/

Substitutiona

GLE EC

50b (-fold)

NS5A Polymorphism/Substitutiona

PIB EC

50b (-fold)

Ac DCV;TVR + PR

GLE/PIB + RBV(Arm B)

1aBaseline None — L31M, H58D 23

Relapse A156V NA Q30R, L31M, H58D >1000

Bd OBV/PTV/r + DSV + RBV

GLE/PIB(Arm C)

1aBaseline Y56H, D168A/T 39e M28V, Q30R, H58C Pending

Breakthrough V36M, Y56H, D168A 162 M28G, Q30R, H58C 942

aPolymorphisms/substi tuti ons detected at 2% NGS detecti on threshold. bEC50 of single or linked polymorphism/substi tuti on (if >1 polymorphism/substi tuti on was detected in the same target). cPati ent received 2 diff erent regiments previously: DCV as the fi rst regimen, and TVR + PR as the second regimen. dPati ent had Crohn’s disease, was on immunosuppressive therapy, and had a prior ileocolectomy. eEC50 value shown is for Y56H + D168A; EC50 value for Y56H + D168T is pending. NA: Not available due to low replicati on effi ciency of the substi tuti on in vitro. DCV: Daclatasvir; TVR: telaprevir; PR: peginterferon plus ribavirin; OBV: ombitasvir; PTV: paritaprevir; r: ritonavir; DSV: dasabuvir; RBV: ribavirin.

Figure 4. Baseline NS3 Polymorphisms

2% 2% 2% 2%

46%

4%8% 8%

7%*13%*2% 2%

0 0

44%

4%2%

8%

2%* 13%*

0

5

10

15

20

25

No.

of S

ubje

cts

Wit

h Po

lym

orph

ism

s

2% NGS Threshold15% NGS Threshold

N = 50

*Percentage relati ve to the total number of GT1a pati ents (n = 42), or GT1b pati ents (n = 8), as appropriate.NGS: Next generati on sequencing.

• At 2% NGS detecti on threshold, most common NS3 baseline amino acid polymorphisms were at positi ons

– Q80 (46%), R155 (8%), and D168 (8%)

Figure 5. Baseline NS5A Polymorphisms

6%

12%

28%

14%75%*

10%*

13%*4%

2%

16%

4% 4%

22%

14%75%*

7%*

13%* 2% 2%

16%

0

5

10

15

20

25

2% NGS Threshold15% NGS Threshold

No.

of S

ubje

cts

Wit

h Po

lym

orph

ism

s

N = 50

*Percentage relati ve to the total number of GT1a pati ents (n = 42), or GT1b pati ents (n = 8), as appropriate.NGS: Next generati on sequencing.

• At 2% NGS detecti on threshold, most common NS5A baseline amino acid polymorphisms were at positi ons

– Q30 (28%), Y93 (16%), L31 (14%), M28 (12%), and GT1b Q54 (75%)*

Figure 6. Pati ents With Multi ple Polymorphisms

0

1

2

3

4

5

6

No.

of P

olym

orph

ism

s in

N

S3 a

nd/o

r N

S5A

NS5A Polymorphism*NS3 Polymorphism*

No. of Pa�ents 7 4 3 1 6 1 2 1 2 1

*Polymorphisms detected at 2% NGS detecti on threshold.

Most pati ents had multi ple (2 or more) baseline polymorphisms

• 14% (7/50) had no baseline polymorphism in NS3 or NS5A• 30% (15/50) had 1 baseline polymorphism in NS3 or NS5A• 56% (28/50) had multi ple (2 or more) baseline polymorphisms in NS3 and/or NS5A

– Most common baseline polymorphism combinati ons 2 NS5A polymorphisms (14%, 7/50)

1 NS3 + 2 NS5A polymorphisms (12%, 6/50)

• 6 pati ents had 4 or more baseline polymorphisms – 83% (5/6) of these pati ents achieved SVR12

Figure 7. Prevalence of Baseline Polymorphisms (N=50)

2% NGS Threshold

15% NGS Threshold

2

3

1

7

7

5

3

33

Arm A (n = 6) Arm B (n = 22) Arm C (n = 22)

No.

of P

a�en

ts w

ith

or w

itho

ut P

olym

orph

ism

s

6

7

5

4

NS3+NS5A NS3 only NS5A only None

9

5

3

5

10

5

3

4

NGS: Next generati on sequencing.

• No signifi cant diff erence in the number of pati ents with at least 1 baseline polymorphism in NS3 or NS5A based on 2% or 15% NGS detecti on threshold

– 86% (43/50 at 2% NGS detecti on threshold) – 80% (40/50 at 15% NGS detecti on threshold)

• More than 50% of pati ents had baseline polymorphisms in NS5A

RESULTS