rational design of doravirine (dor): a review of ...natap.org/2018/idweek/poster60_545.pdf · wt...

1
Rational Design of Doravirine (DOR): A Review of Development From Bench to Patients BRIEF HISTORY OF THE NNRTI CLASS Early/mid 1990s: discovery of first NNRTIs: 1-(2-hydroxyethoxymethyl)-6-(phenylthio)thymine (HEPT) and tetrahydroimidazo[4,5,1-jkj][1,4]benzodiazepin-2(1H)-one and -thione (TIBO) Late 1990s: approval of the first NNRTIs: dipyridodiazepinones (eg, NVP in 1996), bis(heteroaryl) piperazines (eg, DLV in 1997) and benzoxazinones (eg, EFV in 1998) Six drugs in this class are currently approved by regulatory authorities (DLV*, EFV, ETR, NVP, RPV and DOR) Efavirenz (EFV), the most widely used NNRTI for over a decade, was removed from the preferred list in the US guidelines due to CNS tolerability issues Doravirine (DOR) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that has recently been approved (August 2018) as a single entity, and as a fixed-dose combination with lamivudine (3TC) and tenofovir disoproxil fumarate (TDF) (DOR/3TC/TDF) as once-daily oral treatment for HIV-1 infection in treatment-naïve adults NNRTI Drug Approvals to Date NVP ETV RPV DOR 1996 DLV* EFV 1997 1998 2011 2008 2018 Second Generation First Generation *Anticipate discontinuation in October 2018. DOR=doravirine; DVL=delavirdine; EFV=efavirenz; ETR=etravirine; NNRTI=nonnucleoside reverse transcriptase inhibitor; NVP=nevirapine; RPV=rilpivirine. MECHANISM OF ACTION AND PROPENSITY FOR RESISTANCE NNRTIs prevent HIV-1 replication by noncompetitively inhibiting reverse transcription of HIV-1 RNA catalyzed by reverse transcriptase (RT) NNRTIs bind to a unique, allosteric pocket on HIV-1 RT and induce conformational changes in the substrate-binding site, which interferes with DNA polymerase activity NNRTIs, a heterogenous group of chemical structures, bind to RT at a hydrophobic pocket in the p66 subunit Resistance to first-generation NNRTIs frequently led to treatment failure Understanding the mechanisms of NNRTI resistance, coupled with extensive structure-based modeling, led to the discovery of second-generation NNRTIs Of the second-generation agents, ETR is the only NNRTI approved to treat NNRTI treatment failures The flexible structure of ETR allows it to rotate within the NNRTI-binding pocket, permitting multiple interactions with RT even in the presence of NNRTI mutations; thus, maintaining activity 1 Due to the flexible binding mode, ETR was also shown to be more difficult to select for resistance in vitro; in clinical studies, 3 or more mutations were required to develop high-level resistance to ETR Flexible inhibitor Rigid inhibitor Torsional changes (wiggling) Reorientation and repositioning (jiggling) Steric hindrance Reprinted with permission from Das K et al. J Med Chem. 2004;47(10):2550-2260. Copyright (2004) American Chemical Society. STRUCTURE-BASED DRUG DESIGN COUPLED WITH RESISTANCE PROFILING WERE CRITICAL TO THE DORAVIRINE DISCOVERY EFFORT DOR discovery effort started from a series of novel diaryl ether NNRTIs exemplified by MK-1107, a precursor of DOR, with excellent activity vs wild-type (WT) HIV-1 and NNRTI-resistant viruses 2,3 In contrast to ETR, comparing X-ray crystal structures of MK-1107 and related analogs bound to the WT and mutant RT enzymes led to a strategy of introducing cyclic constraints in the molecule to optimize the favorable interactions to retain activity against the key resistance mutants The favorable resistance profiles of key analogs, including DOR were validated using high- throughput resistance selection FROM MK-1107 TO DORAVIRINE Preorganized structures of MK-1107 and related analogs bound to mutant and WT RTs provided potent compounds with markedly improved antiviral activity against WT and NNRTI-resistant viruses that were comparable or superior to more flexible analogs Subsequent iterations of MK-1107 delivered the optimized analog: DOR Cl CN O N O F 3 C N NH N O N H O Cl O F F F Virus EC 50 (nM) a EFV 4 MK-1107 MK-4965 MK-7445 MK-6186 DOR 4 WT 41 34 41 77 64 19 K103N 1427 11 b 17 b 25 b 113 42 Y181C 80 23 b 28 b 81 b 335 25 K103N/Y181C 2943 NA 84 b 202 b 650 52 a Tested in 50% FBS; b Tested in 10% FBS. EC 50 =half maximal effective concentration; FBS=fasting blood sugar. 0 20 40 60 80 100 EFV EFV Select K103N; L100I Select V106A; L234I Day 96 Day 48 Resistance Profiling Resistance Selecon Crystallography L100 K103 Fold-Change EFV DOR DOR CNDO V106A/G190A/F227L K101E/Y181C/G190A K103N/Y181C/G190A K101E/G190A Y181C/G190A K103N/P225H K103N/G190A K103N/Y181C L100I/K103N H221Y E138K M230L K101P V179F V106A K101E G190A Y181I Y188L Y181C K103N G190S L100I MDRC4 0 20 40 60 80 100 DOR Fold-Change CNDO V106A/G190A/F227L K101E/Y181C/G190A K103N/Y181C/G190A K101E/G190A Y181C/G190A K103N/P225H K103N/G190A K103N/Y181C L100I/K103N H221Y E138K M230L K101P V179F V106A K101E G190A Y181I Y188L Y181C K103N G190S L100I MDRC4 X-ray Structure of DOR With Reverse Transcriptase (RT) Doravirine interacts with backbone of K103 (not side chain) No interaction with Y181, L100, E138, K101 but V106 is key 5 Reprinted with permission from Feng M, et al. Antimicrob Agents Chemother. 2015;59(1):590-598. Copyright (2015) American Society for Microbiology. Structural studies suggested a key hydrophobic interaction between the V106 side chain and the central phenyl ring of DOR 3 Resistance profiling studies with DOR and related analogs demonstrated that they were less effective against viruses that contained the triple NNRTI resistance mutation pattern, V106A/G190A/F227L 3 DOR resistance was slower to appear in vitro as compared with first-generation NNRTIs, such as EFV 5 DOR Displays Greater Activity vs RPV, EFV, and ETR Against WT and NNRTI-Resistant Viruses 6,7 NNRTI Antiviral Activity IC 50 (nM) a Protein Binding (%) WT K103N Y181C K103N/Y181C DOR 12 21 31 33 76.0 RPV 58 56 169 318 99.6 EFV 30 1173 90 3119 99.6 ETR 67 68 383 478 99.9 a In vitro multiple cycle infectivity assay conducted in the presence of 100% normal human serum. PRECLINICAL AND PHARMACOKINETIC STUDIES OF DORAVIRINE DOR exhibited favorable pharmacokinetic profiles in rats and dogs, suggesting the potential for once-daily dosing in humans 3 Preclinical studies also suggested the potential for DOR to be administered at low doses conducive to the development of a fixed-dose coformulation with other antiretroviral drugs 3 DOR is a substrate for CYP3A4, but unlike other NNRTIs, DOR does not impact the expression of CYP3A4 or other major drug-metabolizing enzymes or transporters 8 No clinically meaningful interactions in humans between DOR (with tenofovir disoproxil fumarate [TDF]) and atorvastatin, oral contraceptives, or pantoprazole 9,10 No clinically meaningful effect of food on DOR 11 Disposition of DOR 12 Cl N O N O F F F N NH N O Cl N O N O O O F F F M9 N HN N CYP3A-Mediated Metabolism ~15% Absorbed Drug Doravirine 100 mg Renal Clearance ~65% >55% Negligible Biliary Clearance Negligible Phase 2 Metabolism Main (12.9%) Metabolite SAFETY AND TOLERABILITY Quantitative EEG methods were developed in preclinical species and validated in humans with EFV to derisk potential CNS toxicity of novel NNRTIs Quantitative preclinical electroencephalography (EEG) studies of MK-6186, a closely related analog of DOR, found no measurable central nervous system (CNS) toxicity Adverse CNS effects are observed with EFV Mechanism(s) responsible for the adverse CNS effects of EFV remain unclear, but several clinical studies have demonstrated associated changes in brain EEG and sleep architecture 13 In clinical studies, DOR has been shown to have fewer neuropsychiatric AEs vs EFV with a CNS profile indistinguishable from DRV 14,15 Neuro Target In Vitro Activities K i or IC 50 <10μM EFV RPV DOR Target K i (μM) Target K i (μM) Target K i (μM) Norepinephrine transporter 4.1 Monoamine oxidase (MAO-A) 2.3 (IC 50 ) Serotonin (5-HT 2B ) 1.6 Dopamine transporter 5.6 Phosphodiesterase (PDE3) 8.0 (IC 50 ) Progesterone (PR-B) 2.3 Protein Ser/Thr Kinase MAPK3 (ERK1) 2.4 (IC 50 ) Serotonin (5-HT 2A ) 1.1 Protein Ser/Thr Kinase, MARK3 3.1 (IC 50 ) Serotonin (5-HT 2B ) 6.3 Protein Ser/Thr Kinase, PKA, nonselective 2.6 (IC 50 ) Serotonin (5-HT 2C ) 0.34 Protein Ser/Thr Kinase, PKC, nonselective 2.6 (IC 50 ) Serotonin (5-HT 6 ) 0.28 Adenosine A 1 4.1 Androgen AR 4.0 Adenosine A 2 3.8 Adenosine transporter 0.59 Adrenergic β 1 6.6 Adrenergic β 3 6.2 Norepinephrine transporter 6.6 Cannabinoid (CB 1 ) 1.7 Histamine (H 4 ) 2.3 Progesterone PR-B 0.43 Serotonin (5-HT 2B ) 1.1 Androgen receptor 4.5 Prostanoid thromboxane A 2 (TP) 4.9 Prostanoid FP 4.6 (IC 50 ) CLINICAL TRIAL OBSERVATIONS The clinical development program for DOR and the combination, DOR/3TC/TDF includes: 36 phase 1 trials 3 phase 2b trials: P007 (NCT01632345), DRIVE-BEYOND (NCT02629822), DRIVE-CLEAR (NCT02652260) 3 phase 3 trials: DRIVE-FORWARD (NCT02275780), 14 DRIVE-AHEAD (NCT02403674), 15 DRIVE-SHIFT (NCT02397096) DRIVE-FORWARD: DOR was noninferior to darunavir plus ritonavir (DRV+r), both given with 2 nucleoside reverse transcriptase inhibitors (NRTIs) in treatment-naïve patients 14 HIV-1 RNA <50 copies/mL at Week 48: 83.8% DOR vs 79.9% DRV+r; difference = 3.9% (95% CI: -1.6 to 9.4) CNS tolerability was comparable, with a more favorable lipid profile for DOR DRIVE-AHEAD: DOR/3TC/TDF was non-inferior to EFV/FTC/TDF 15 HIV-1 RNA <50 copies/mL at Week 48 : 84.3% DOR vs 80.8% EFV; difference = 3.5% (95% CI: -2.0 to 9.0) DOR had significantly fewer neuropsychiatric events and a more favorable lipid profile compared with EFV Proportion of Participants With HIV-1 RNA <50 copies/mL at Week 48 (FDA Snapshot Approach). A. DRIVE-FORWARD 14 ; B. DRIVE-AHEAD 15 0 0 20 40 60 80 100 4 8 16 24 Time (weeks) Difference (95% CI): 3.9 (–1.6, 9.4) Proportion of participants (%) 36 48 80% 84% DOR DRV+r A. 0 0 20 40 60 80 100 B. 4 8 16 24 Time (weeks) Difference (95% CI): 3.5 (–2.0, 9.0) Proportion of participants (%) 36 48 81% 84% DOR EFV DRIVE-AHEAD: Proportion of Participants With Predefined Neuropsychiatric Adverse Events a at Week 48 15 8.8 12.1 4.4 4.1 0.3 37.1 25.5 8.2 6.6 1.1 0.0 5.0 10.0 15.0 20.0 25.0 30.0 35.0 40.0 Proportion of patients (%) DOR EFV Dizziness Sleep disorders/ disturbances Altered sensorium Depression and suicide/ self-injury Psychosis/ psychotic disorders P<0.001 P<0.001 P=0.033 a Statistical testing for parameters other than dizziness and sleep disorders/disturbances, and altered sensorium were not prespecified. CONCLUSIONS Two decades of research on NNRTIs Numerous failures but wealth of preclinical and clinical experience Helped to build a logical, step-by-step approach to DOR development that addressed many of the critical liabilities of the NNRTI class DOR was rationally designed Unique NNRTI Distinct resistance profile Excellent tolerability Noninferior efficacy to EFV and DRV+r Superior neuropsychiatric profile compared with EFV Superior lipid profile compared with DRV+r and EFV References 1. Das K, Clark AD Jr, Lewi PJ, et al. J Med Chem . 2004;47(10):2550-2560. 2. Tucker TJ, Saggar S, Sisko JT, et al. Bioorg Med Chem Lett . 2008;18(9):2959-2966. 3. Côté B, Burch JD, Asante-Appiah E, et al. Bioorg Med Chem Lett . 2014;24(3):917-922. 4. Lai M, Feng M, Falgueyret, J, et al. Antimicrob Agents Chemother . 2014;58(3):1652-1663. 5. Feng M, Wang D, Grobler JA, et al. Antimicrob Agents Chemother . 2015;59(1):590-598. 6. Lai M, Sachs N, Xu M, et al. Abstract presented at: Conference on Retroviruses and Opportunistic Infections (CROI). February 22-25, 2016; Boston, MA. Abstract #506. 7. Feng M, Sachs NA, Xu M, et al. Antimicrob Agents Chemother . 2016;60(4):2241-2247. 8. Anderson MS, Gilmartin J, Cilissen C, et al. Antivir Ther . 2015;20(4):397-405. 9. Khalilieh S, Yee KL, Sanchez RI, et al. Abstract presented at: International AIDS Society (IAS) Conference on HIV Science; July 23-26, 2017; Paris, France. Poster MOPEB0334. Available at: http://programme.ias2017.org/Abstract/Abstract/3996. Accessed February 18, 2018. 10. Khalilieh S, Yee KL, Sanchez RI, et al. Antimicrob Agents Chemother . 2017;61(2). 11. Behm MO, Yee KL, Liu R, et al. Clin Drug Investig . 2017;37(6):571-579. 12. Sanchez RI, Fillgrove KL, Yee KL, et al. Xenobiotica . 2018:1-11. 13. Gallego L, Barreiro P, del Río R, et al. Clin Infect Dis . 2004;38(3):430-432. 14. Molina JM, Squires K, Sax P, et al. Lancet HIV . 2018;5(5):e211-e220. 15. Orkin C, Squires K, Molina J-M, et al. Clin Infect Dis . In press. Disclosures Carey Hwang, Ming-Tain Lai, and Daria Hazuda are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. The authors wish to thank Meredith Rogers, MS, CMPP, The Lockwood Group (Stamford, CT) for writing and editorial assistance. Carey Hwang, MD, PhD; Ming-Tain Lai, PhD; Daria Hazuda, PhD Merck & Co., Inc., Kenilworth, NJ, USA Abstract #545 Copyright © 2018 Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc. , Kenilworth, N.J., U.S.A. All Rights Reserved. https://bit.ly/2w4GUzG Presented at IDWeek; San Francisco, CA; October 3–7, 2018.

Upload: others

Post on 08-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Rational Design of Doravirine (DOR): A Review of ...natap.org/2018/IDWeek/POSTER60_545.pdf · WT K103N Y181C K103N/Y181C Protein Binding (%) DOR 12 21 31 33 76.0 RPV 58 56 169 318

Rational Design of Doravirine (DOR): A Review of Development From Bench to Patients

BRIEF HISTORY OF THE NNRTI CLASS • Early/mid 1990s: discovery of first NNRTIs: 1-(2-hydroxyethoxymethyl)-6-(phenylthio)thymine (HEPT) and tetrahydroimidazo[4,5,1-jkj][1,4]benzodiazepin-2(1H)-one and -thione (TIBO)

• Late 1990s: approval of the first NNRTIs: dipyridodiazepinones (eg, NVP in 1996), bis(heteroaryl)piperazines (eg, DLV in 1997) and benzoxazinones (eg, EFV in 1998)

• Six drugs in this class are currently approved by regulatory authorities (DLV*, EFV, ETR, NVP, RPV and DOR)

• Efavirenz (EFV), the most widely used NNRTI for over a decade, was removed from the preferred list in the US guidelines due to CNS tolerability issues

• Doravirine (DOR) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that has recently been approved (August 2018) as a single entity, and as a fixed-dose combination with lamivudine (3TC) and tenofovir disoproxil fumarate (TDF) (DOR/3TC/TDF) as once-daily oral treatment for HIV-1 infection in treatment-naïve adults

NNRTI Drug Approvals to DateNVP ETV RPV DOR

1996

DLV* EFV

1997 1998 20112008 2018

SecondGeneration

FirstGeneration

*Anticipate discontinuation in October 2018.DOR=doravirine; DVL=delavirdine; EFV=efavirenz; ETR=etravirine; NNRTI=nonnucleoside reverse transcriptase inhibitor; NVP=nevirapine; RPV=rilpivirine.

MECHANISM OF ACTION AND PROPENSITY FOR RESISTANCE

• NNRTIs prevent HIV-1 replication by noncompetitively inhibiting reverse transcription of HIV-1 RNA catalyzed by reverse transcriptase (RT)

• NNRTIs bind to a unique, allosteric pocket on HIV-1 RT and induce conformational changes in the substrate-binding site, which interferes with DNA polymerase activity

− NNRTIs, a heterogenous group of chemical structures, bind to RT at a hydrophobic pocket in the p66 subunit − Resistance to first-generation NNRTIs frequently led to treatment failure

• Understanding the mechanisms of NNRTI resistance, coupled with extensive structure-based modeling, led to the discovery of second-generation NNRTIs

• Of the second-generation agents, ETR is the only NNRTI approved to treat NNRTI treatment failures

− The flexible structure of ETR allows it to rotate within the NNRTI-binding pocket, permitting multiple interactions with RT even in the presence of NNRTI mutations; thus, maintaining activity1

− Due to the flexible binding mode, ETR was also shown to be more difficult to select for resistance in vitro; in clinical studies, 3 or more mutations were required to develop high-level resistance to ETR

Flexibleinhibitor

Rigidinhibitor

Torsional changes(wiggling)

Reorientation andrepositioning (jiggling)

Steric hindrance

Reprinted with permission from Das K et al. J Med Chem. 2004;47(10):2550-2260.Copyright (2004) American Chemical Society.

STRUCTURE-BASED DRUG DESIGN COUPLED WITH RESISTANCE PROFILING WERE CRITICAL TO THE

DORAVIRINE DISCOVERY EFFORT • DOR discovery effort started from a series of novel diaryl ether NNRTIs exemplified by MK-1107, a precursor of DOR, with excellent activity vs wild-type (WT) HIV-1 and NNRTI-resistant viruses2,3

• In contrast to ETR, comparing X-ray crystal structures of MK-1107 and related analogs bound to the WT and mutant RT enzymes led to a strategy of introducing cyclic constraints in the molecule to optimize the favorable interactions to retain activity against the key resistance mutants

• The favorable resistance profiles of key analogs, including DOR were validated using high-throughput resistance selection

FROM MK-1107 TO DORAVIRINE • Preorganized structures of MK-1107 and related analogs bound to mutant and WT RTs provided potent compounds with markedly improved antiviral activity against WT and NNRTI-resistant viruses that were comparable or superior to more flexible analogs

• Subsequent iterations of MK-1107 delivered the optimized analog: DOR

Cl CN

ON

O

F3CN NH

NON

H

OCl

O

F

FF

Virus

EC50 (nM)a

EFV4 MK-1107 MK-4965 MK-7445 MK-6186 DOR4

WT 41 34 41 77 64 19

K103N 1427 11b 17b 25b 113 42

Y181C 80 23b 28b 81b 335 25

K103N/Y181C 2943 NA 84b 202b 650 52aTested in 50% FBS; bTested in 10% FBS.EC50=half maximal effective concentration; FBS=fasting blood sugar.

020406080

100EFV

EFVSelectK103N; L100I

SelectV106A; L234I

Day 96

Day 48

Resistance Profiling

Resistance Selection Crystallography

L100

K103

Fold

-Cha

nge

EFVDOR

DOR

CN

DO

V10

6A/G

190A

/F22

7LK

101E

/Y18

1C/G

190A

K10

3N/Y

181C

/G19

0AK

101E

/G19

0AY

181C

/G19

0AK

103N

/P22

5HK

103N

/G19

0AK

103N

/Y18

1CL1

00I/K

103N

H22

1YE

138K

M23

0LK

101P

V17

9FV

106A

K10

1EG

190A

Y18

1IY

188L

Y18

1CK

103N

G19

0SL1

00I

MD

RC

4

020406080

100DOR

Fold

-Cha

nge

CN

DO

V10

6A/G

190A

/F22

7LK

101E

/Y18

1C/G

190A

K10

3N/Y

181C

/G19

0AK

101E

/G19

0AY

181C

/G19

0AK

103N

/P22

5HK

103N

/G19

0AK

103N

/Y18

1CL1

00I/K

103N

H22

1YE

138K

M23

0LK

101P

V17

9FV

106A

K10

1EG

190A

Y18

1IY

188L

Y18

1CK

103N

G19

0SL1

00I

MD

RC

4

X-ray Structure of DOR With Reverse Transcriptase (RT)

Doravirine interacts with backbone of K103 (not side chain)No interaction with Y181, L100, E138, K101 but V106 is key5

Reprinted with permission from Feng M, et al. Antimicrob Agents Chemother. 2015;59(1):590-598. Copyright (2015) American Society for Microbiology.

• Structural studies suggested a key hydrophobic interaction between the V106 side chain and the central phenyl ring of DOR3

• Resistance profiling studies with DOR and related analogs demonstrated that they were less effective against viruses that contained the triple NNRTI resistance mutation pattern, V106A/G190A/F227L3

• DOR resistance was slower to appear in vitro as compared with first-generation NNRTIs, such as EFV5

DOR Displays Greater Activity vs RPV, EFV, and ETR Against WT and NNRTI-Resistant Viruses6,7

NNRTIAntiviral Activity IC50 (nM)a

Protein Binding (%)WT K103N Y181C K103N/Y181CDOR 12 21 31 33 76.0RPV 58 56 169 318 99.6EFV 30 1173 90 3119 99.6ETR 67 68 383 478 99.9

aIn vitro multiple cycle infectivity assay conducted in the presence of 100% normal human serum.

PRECLINICAL AND PHARMACOKINETIC STUDIES OF DORAVIRINE

• DOR exhibited favorable pharmacokinetic profiles in rats and dogs, suggesting the potential for once-daily dosing in humans3

• Preclinical studies also suggested the potential for DOR to be administered at low doses conducive to the development of a fixed-dose coformulation with other antiretroviral drugs3

• DOR is a substrate for CYP3A4, but unlike other NNRTIs, DOR does not impact the expression of CYP3A4 or other major drug-metabolizing enzymes or transporters8

− No clinically meaningful interactions in humans between DOR (with tenofovir disoproxil fumarate [TDF]) and atorvastatin, oral contraceptives, or pantoprazole9,10

• No clinically meaningful effect of food on DOR11

Disposition of DOR12Cl N

ON

O

FF

F

N NH

NO

Cl N

ON

O O

OFF

F M9

NHN

N

CYP3A-Mediated Metabolism

~15%

Absorbed Drug

Doravirine 100 mg

Renal Clearance

~65%

>55%

Negligible Biliary ClearanceNegligible Phase 2 Metabolism

Main (12.9%)Metabolite

SAFETY AND TOLERABILITY • Quantitative EEG methods were developed in preclinical species and validated in humans with EFV to derisk potential CNS toxicity of novel NNRTIs

• Quantitative preclinical electroencephalography (EEG) studies of MK-6186, a closely related analog of DOR, found no measurable central nervous system (CNS) toxicity

• Adverse CNS effects are observed with EFV − Mechanism(s) responsible for the adverse CNS effects of EFV remain unclear, but several clinical studies have demonstrated associated changes in brain EEG and sleep architecture13

• In clinical studies, DOR has been shown to have fewer neuropsychiatric AEs vs EFV with a CNS profile indistinguishable from DRV14,15

Neuro Target In Vitro ActivitiesKi or IC50 <10μM

EFV RPV DORTarget Ki (μM) Target Ki (μM) Target Ki (μM)Norepinephrine transporter 4.1 Monoamine oxidase (MAO-A) 2.3 (IC50) Serotonin (5-HT2B) 1.6Dopamine transporter 5.6 Phosphodiesterase (PDE3) 8.0 (IC50)Progesterone (PR-B) 2.3 Protein Ser/Thr Kinase MAPK3 (ERK1) 2.4 (IC50)Serotonin (5-HT2A) 1.1 Protein Ser/Thr Kinase, MARK3 3.1 (IC50)Serotonin (5-HT2B) 6.3 Protein Ser/Thr Kinase, PKA, nonselective 2.6 (IC50)Serotonin (5-HT2C) 0.34 Protein Ser/Thr Kinase, PKC, nonselective 2.6 (IC50)Serotonin (5-HT6) 0.28 Adenosine A1 4.1Androgen AR 4.0 Adenosine A2 3.8

Adenosine transporter 0.59Adrenergic β1 6.6Adrenergic β3 6.2

Norepinephrine transporter 6.6Cannabinoid (CB1) 1.7

Histamine (H4) 2.3Progesterone PR-B 0.43Serotonin (5-HT2B) 1.1Androgen receptor 4.5

Prostanoid thromboxane A2 (TP) 4.9Prostanoid FP 4.6 (IC50)

CLINICAL TRIAL OBSERVATIONS • The clinical development program for DOR and the combination, DOR/3TC/TDF includes:

− 36 phase 1 trials − 3 phase 2b trials: P007 (NCT01632345), DRIVE-BEYOND (NCT02629822), DRIVE-CLEAR (NCT02652260) − 3 phase 3 trials: DRIVE-FORWARD (NCT02275780),14 DRIVE-AHEAD (NCT02403674),15 DRIVE-SHIFT (NCT02397096)

• DRIVE-FORWARD: DOR was noninferior to darunavir plus ritonavir (DRV+r), both given with 2 nucleoside reverse transcriptase inhibitors (NRTIs) in treatment-naïve patients14

− HIV-1 RNA <50 copies/mL at Week 48: 83.8% DOR vs 79.9% DRV+r; difference = 3.9% (95% CI: -1.6 to 9.4) − CNS tolerability was comparable, with a more favorable lipid profile for DOR

• DRIVE-AHEAD: DOR/3TC/TDF was non-inferior to EFV/FTC/TDF15 − HIV-1 RNA <50 copies/mL at Week 48 : 84.3% DOR vs 80.8% EFV; difference = 3.5% (95% CI: -2.0 to 9.0) − DOR had significantly fewer neuropsychiatric events and a more favorable lipid profile compared with EFV

Proportion of Participants With HIV-1 RNA <50 copies/mL at Week 48 (FDA Snapshot Approach). A. DRIVE-FORWARD14; B. DRIVE-AHEAD15

00

20

40

60

80

100

4 8 16 24Time (weeks)

Difference (95% CI):3.9 (–1.6, 9.4)

Pro

porti

on o

f par

ticip

ants

(%)

36 48

80%

84%

DORDRV+r

A.

00

20

40

60

80

100B.

4 8 16 24Time (weeks)

Difference (95% CI):3.5 (–2.0, 9.0)

Pro

porti

on o

f par

ticip

ants

(%)

36 48

81%

84%

DOREFV

DRIVE-AHEAD: Proportion of Participants With Predefined Neuropsychiatric Adverse Eventsa at Week 4815

8.8

12.1

4.4 4.1

0.3

37.1

25.5

8.26.6

1.10.0

5.0

10.0

15.0

20.0

25.0

30.0

35.0

40.0

Pro

porti

on o

f pat

ient

s (%

)

DOREFV

Dizziness Sleepdisorders/

disturbances

Alteredsensorium

Depressionand suicide/self-injury

Psychosis/psychoticdisorders

P<0.001

P<0.001

P=0.033

aStatistical testing for parameters other than dizziness and sleep disorders/disturbances, and altered sensorium were not prespecified.

CONCLUSIONS • Two decades of research on NNRTIs

− Numerous failures but wealth of preclinical and clinical experience − Helped to build a logical, step-by-step approach to DOR development that addressed many of the critical liabilities of the NNRTI class

• DOR was rationally designed − Unique NNRTI − Distinct resistance profile − Excellent tolerability − Noninferior efficacy to EFV and DRV+r − Superior neuropsychiatric profile compared with EFV − Superior lipid profile compared with DRV+r and EFV

References1. Das K, Clark AD Jr, Lewi PJ, et al. J Med Chem. 2004;47(10):2550-2560.2. Tucker TJ, Saggar S, Sisko JT, et al. Bioorg Med Chem Lett. 2008;18(9):2959-2966.3. Côté B, Burch JD, Asante-Appiah E, et al. Bioorg Med Chem Lett. 2014;24(3):917-922.4. Lai M, Feng M, Falgueyret, J, et al. Antimicrob Agents Chemother. 2014;58(3):1652-1663.5. Feng M, Wang D, Grobler JA, et al. Antimicrob Agents Chemother. 2015;59(1):590-598.6. Lai M, Sachs N, Xu M, et al. Abstract presented at: Conference on Retroviruses and Opportunistic

Infections (CROI). February 22-25, 2016; Boston, MA. Abstract #506.7. Feng M, Sachs NA, Xu M, et al. Antimicrob Agents Chemother. 2016;60(4):2241-2247.8. Anderson MS, Gilmartin J, Cilissen C, et al. Antivir Ther. 2015;20(4):397-405.9. Khalilieh S, Yee KL, Sanchez RI, et al. Abstract presented at: International AIDS Society (IAS)

Conference on HIV Science; July 23-26, 2017; Paris, France. Poster MOPEB0334. Available at: http://programme.ias2017.org/Abstract/Abstract/3996. Accessed February 18, 2018.

10. Khalilieh S, Yee KL, Sanchez RI, et al. Antimicrob Agents Chemother. 2017;61(2).11. Behm MO, Yee KL, Liu R, et al. Clin Drug Investig. 2017;37(6):571-579.12. Sanchez RI, Fillgrove KL, Yee KL, et al. Xenobiotica. 2018:1-11.13. Gallego L, Barreiro P, del Río R, et al. Clin Infect Dis. 2004;38(3):430-432.14. Molina JM, Squires K, Sax P, et al. Lancet HIV. 2018;5(5):e211-e220.15. Orkin C, Squires K, Molina J-M, et al. Clin Infect Dis. In press.

DisclosuresCarey Hwang, Ming-Tain Lai, and Daria Hazuda are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA.

The authors wish to thank Meredith Rogers, MS, CMPP, The Lockwood Group (Stamford, CT) for writing and editorial assistance.

Carey Hwang, MD, PhD; Ming-Tain Lai, PhD; Daria Hazuda, PhDMerck & Co., Inc., Kenilworth, NJ, USA

Abstract #545

Copyright © 2018 Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, N.J., U.S.A. All Rights Reserved.

https://bit.ly/2w4GUzG

Presented at IDWeek; San Francisco, CA; October 3–7, 2018.