1
Forgiveness of Antiretroviral Regimens: In Vitro HIV-1 Viral Breakthrough with 2-Drug versus 3-Drug Regimens Simulating Variable Adherence to Treatment Andrew Mulato, Rima Acosta, Stephen R. Yant, Tomas Cihlar, and Kirsten White Gilead Sciences, Inc., Foster City, CA, USA Background References © 2019 Gilead Sciences, Inc. All rights reserved. 10 th IAS Conference on HIV Science (IAS 2019), 21-24 July 2019, Mexico City, Mexico Poster TUPEB244 In vitro viral breakthrough selection experiments help define the resistance barrier of ARV drug combinations and the resistance mutations that emerge. Here, two series of breakthrough experiments were conducted starting with wild-type or low-level M184V HIV and antiretroviral regimens set at drug in vivo concentrations or concentrations mimicking short lapses in adherence (1 to 3 consecutive missed doses) Using wild-type HIV, BIC+FTC+TAF was better at preventing in vitro viral breakthrough and emergent drug resistance compared to the 2-drug combination of DTG+3TC BIC+FTC+TAF had viral breakthrough only in the wild-type experiments under the lowest drug concentrations tested (C min after missing 3 doses). Only one viral breakthrough had emergent resistance with M184I in RT. In total, 23/252 wells had breakthrough and 1 had emergent resistance In the wild-type experiments, DTG+3TC had 104/144 wells breakthrough and 15 of these had emergent resistance consisting of M184V/I and other mutations in RT, and other mutations in IN, including R263K and S153F, and one with resistance mutations in both RT and IN mutations. M184I has also been reported in DTG+3TC two-drug in vitro breakthrough experiments by another group 27 M184V causes resistance to FTC and 3TC and is one of the most frequent mutations to occur during virologic failure, is archived, and can be transmitted 29-31 . Pre- existing M184V at low levels in the inoculum had viral breakthroughs with DTG+3TC and high-level M184V with or without additional substitutions compared to BIC+FTC+TAF where no breakthrough occurred These in vitro studies suggest that the favorable pharmacology, antiviral synergy, and resistance profile provided by BIC+FTC+TAF may be more protective against virologic breakthrough and emergent resistance than DTG+3TC in the setting of non-adherence In vitro studies of the 'forgiveness' of ART regimens are important as they may help to predict regimen durability and risk of resistance in the real world, where sub- optimal adherence is more common Acknowledgements The authors thank Alex Thielen and Martin Däeumer at Seq-IT and Ross Martin, Xinan Liu, and Silvia Chang (Gilead Sciences, Inc.) for deep sequence analysis. Conclusions Methods Current guidelines for the treatment of HIV-1 infection recommend triple therapy consisting of an integrase strand transfer inhibitor (INSTI) plus 2 nucleoside/tide reverse transcriptase inhibitors (NRTIs) including the single-tablet regimen (STR) of bictegravir+emtricitabine+tenofovir alafenamide (BIC/FTC/TAF) 1,2 BIC/FTC/TAF is approved for the treatment of HIV infection, has a high barrier to resistance 3-10 , and may offer “forgiveness” (avoiding viral rebound and resistance in the setting of short-term non-adherence) Optimal adherence to daily oral antiretroviral therapy (ART) is important to minimize the emergence of drug resistance 4 . Whereas high adherence rates are achieved in clinical trials, treatment adherence rates in the real world are lower and not always predictable 11-13 A regimen’s barrier to resistance is both genetic and pharmacologic. Drugs with higher resistance barriers are preferred. Variations in minimal drug concentrations (C min ) can occur due to natural variation between individuals, drug-drug interactions, food effects, and missed doses Genetic Barrier: The number and sequence of mutations needed to generate resistance to a drug Pharmacokinetic Barrier: The concentration of drug needed to suppress the virus, clinical drug concentrations, and drug half-life Figure 1: Schematic of Antiviral Pharmacokinetics Figure 2. In Vitro Breakthrough Selection Methodology I. Regimen Forgiveness by In Vitro Breakthrough Selections: Constant Drugs levels at C min and After Missing 1-3 Consecutive Doses (HIV-1 IIIb virus) Results Recently, controlled clinical trials in treatment-naïve individuals have shown efficacy using an INSTI + 1 NRTI (dolutegravir/lamivudine; DTG+3TC) but the question remains whether the decreased number of regimen components may be less forgiving than triple therapy; virologic failure on DTG/3TC with emergent resistance to both DTG and 3TC has been reported (ACTG 5353) 28 Objective To investigate BIC+FTC+TAF vs. DTG+3TC “forgiveness” by in vitro HIV-1 viral breakthrough selections and resistance emergence In cells infected with either wild-type or low-level M184V Drug exposures modeling full adherence and suboptimal adherence to ART Test Drug Concentrations (Table 1): BIC and DTG concentrations were calculated using their human plasma clinical trough concentrations (C min ) according to their prescribing information and adjusted for human plasma protein binding 15-17 . The TAF C min concentration generated tenofovir-diphosphate (TFV- DP) at its physiological concentration in PBMCs from TAF-treated individuals 18 . FTC and 3TC concentrations were set at their human plasma-free adjusted C min concentrations 18-20 . Missed Doses: To simulate 1, 2, or 3 consecutive missed doses (C min minus 1 dose, C min minus 2 doses, and C min minus 3 doses), drug concentrations were adjusted by their plasma half-lives for BIC and DTG and active metabolite half-lives for the NRTIs (TAF, FTC, and 3TC). C min – X doses determined as C min *(0.5^{24*X/ t 1/2 }). Drug concentrations were kept constant in each selection. Table 1. Drug Concentrations for Cell Culture Equivalents a. C min values are median values from United States prescribing information (USPI) b. BIC and DTG data generated in-house by standard equilibrium dialysis shift in human serum versus cell culture media 15 c. C min / Human Serum Shift d. t 1/2 for FTC, TAF, and 3TC represent intracellular half-life of the active di- or tri- phosphate metabolite 18-20 ; t 1/2 values for BIC and DTG from USPI Genotypic Analyses: Each well with viral breakthrough was sequenced by next generation sequencing (Seq-IT) and mutations were reported if present at >2%. Mutations were observed between 2.1% and 99% frequency per well. I. Time to Viral Breakthrough: C min and After Missing 1-3 Consecutive Doses (N=144) I. Resistance in Breakthrough Viruses at C min and After Missing 1-3 Consecutive Doses II. Regimen Forgiveness by In Vitro Breakthrough Selections: Using Low-Level M184V xxLAI Starting Virus Mixtures II. Resistance in Breakthrough Viruses: Low-Level M184V Starting Virus Table 2. Mechanisms of Forgiveness and Barrier to Resistance Mechanisms BIC+FTC+TAF DTG+3TC Drug Levels Long plasma or intracellular t 1/2 BIC or DTG 16-17 17 hr 14 hr FTC-TP or 3TC-TP 18-20 37 hr 17.5 hr TFV-DP 18 116 hr na Long IN/DNA dissociation t 1/2 for BIC or DTG 21-22 132 hr 71-78 hr Synergy and Mechanisms of Synergy Combination antiviral activity BIC + FTC or DTG + 3TC 15,23-25 Synergy Synergy BIC + TAF 15 Synergy na TFV + FTC 23-24 Synergy na TFV-chain-termination stabilized by Dead-End Complex with FTC-TP 25 Increased TFV Activity na Resistance Profile Phenotype of M184V BIC or DTG 16 Sensitive Sensitive FTC or 3TC 17-18 Resistant Resistant TFV (TAF) 26 Hypersensitive na BIC + FTC + TAF DTG + 3TC BIC FTC TAF DTG 3TC Clinical Dose (mg) 50 200 25 50 300 Molecular Weight (g/mol) 449.4 247.2 534.5 419.4 229.3 Clinical C min (µg/mL) a 2.61 0.096 0.008 1.11 0.042 Clinical C min (nM) 5808 388 15 2515 265 Human Serum Shift b 43.6 1.0 1.0 27.5 1.0 Cell Culture Equivalent C min (nM) c 133 388 15 91 265 t ½ (hr) d 17 37 116 14 17.5 Missing antiretroviral doses results in a predictable decrease of systemic exposures to each drug in the regimen based on their established clinical half-life Optimal Adherence With 1 – 3 Consecutive Missed Doses II. Time to Viral Breakthrough: Using Low-Level M184V xxLAI Starting Virus Mixtures 1. DHHS. Human Immunodeficiency Virus-1 Infection: Developing Antiretroviral Drugs for Treatment Guidance for Industry. https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/ Guidances/UCM355128. Published November 2015. Accessed March 2019. 2. DHHS. Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents. https://aidsinfo.nih.gov/guidelines/html/1/adult-andadolescent-arv/0. Published October 2018. Accessed March 2019. 3. Gallant J, et al. The Lancet (2017) 390(10107): 2063-2072 4. Sax PE, et al. The Lancet (2017) 390(10107): 2073-2082 5. Daar ES, et al. The Lancet HIV (2018) 5(7): e347-e356 6. Molina JM, et al. The Lancet HIV (2018) 5(7): e357-e365 7. Wohl D, et al. ID Week (2018) Presentation #74246 8. Stellbrink HJ, et al. HIV Glasgow (2018) Presentation #4185960 9. Kityo C, et al. CROI (2018) Presentation #500 10. Gaur AH, et al. CROI (2019) Presentation #2571 11. Cheng Y et al. Medicine (2018) 97(2):1-7 12. Kangethe, et al. J. Managed Care Pharm (2019) 25(1): 88-93 13. Tandon et al. Curr Med Res Opin (2018) 35(1): 63-71 14. Cahn P et al. The Lancet (2019) 393(10167): 143-155 15. Tsiang M, et al. Antimicrob Agents and Chemother (2016) 60(12): 7086-7097 16. BIKTARVY ® United States Prescribing Information 17. TIVICAY ® United States Prescribing Information 18. Dickinson L, et al. Antimicrob Agents and Chemother (2015) 59(10): 6080-6086 19. Else LJ et al. Antimicrob Agents and Chemother (2011) 56(3): 1427-1433 20. Yuen GJ et al. Antimicrob Agents and Chemother (2004) 48(1): 176-182 21. Hightower K, et al. Antimicrob Agents and Chemother (2015) 55(10):4552-4559 22. White K, et al. CROI (2017) Presentation #893 23. Laskey S, et al. JCI Insight (2016) 1(19):e90033 24. Borroto-Esoda K, et al. Antiv Ther (2006) 11(3): 377-384 25. Kulkarni R, et al. Antivir Res (2014) 101: 131-135 26. Deval J, et al. J Biol Chem (2004) 279(1):509-516 27. Yoshinaga T, et al. Antimicrob Agents and Chemother (2019) 63(30): e02104-18 28. Taiwo BO, et al. Clin Infect Dis (2018) 66(11):1689-97 29. Castro H, et al., JID (2013) 208(9): 1459-63 30. Wainberg MA, et al., JAC (2011) 66(10): 2346-9 31. Jain V, et al., JID (2011) 203(8): 1174-81 Viral Breakthrough Selections (Figure 2): In vitro viral breakthrough selection experiments should be interpreted as a comparative in vitro study; clinical trials of missed doses for these ARV combinations have not been conducted. MT-2 cells were infected with HIV-1 (IIIb or clonal xxLAI strain containing varying levels of M184V, an FTC and 3TC resistance mutation). Infected cells were cultured in the presence of fixed concentrations of BIC+FTC+TAF or DTG+3TC, split every 3-4 days with fresh media containing drug, and monitored for viral breakthrough by cytopathic effect for up to 5 weeks. Supernatants containing breakthrough virus were collected and stored at the time of cytopathic effect. Statistical comparisons for experiments using the same starting virus backbone were made using Fisher’s exact test. Gilead Sciences, Inc. 333 Lakeside Drive Foster City, CA 94404 1-800-445-3235 I. Fixed Drug Concentrations C min minus 1 dose C min minus 2 doses C min minus 3 doses C min HIV: IIIb M184V/I K219N/R V75I V118I T215A K70R R263K* G140E S153F* P145S A128T L74M 1 2 3 1 2 3 NRTI -R INSTI -R Virus with Resistance (N) BIC+FTC+TAF DTG+3TC 4 4 (Resistance in 1/144) (Resistance in 15/144) *R263K and S153F have been previously selected by DTG and cause reduced susceptibility to DTG † M184V and M184I cause high -level resistance to FTC and 3TC and increased sensitivity to TAF; occurred only once for BIC+FTC+TAF and was in the C min after 3 missed doses experiments † M184V and M184I cause high-level resistance to FTC and 3TC; all breakthroughs with M184V were >68% *S153Y has been previously selected by DTG and causes reduced susceptibility to DTG *R263K and S153F have been previously selected by DTG and cause reduced susceptibility to DTG The well with R263K in IN also had T215A and K219R present. M184V M184V+M184I+E157K M184V+E157K M184V+E138K M184V+D67N M184V+S153Y* 5 10 15 0.4% M184V inoculum 4% M184V inoculum Virus with Resistance (N) BIC+FTC+TAF DTG+3TC 20 (Resistance in 0/72) (Resistance in 40/72) M184V 5 10 15 20 M184V+M50I All 15 breakthroughs had M184V as the majority species All 25 breakthroughs had M184V as the majority species NRTI and INSTI -R NRTI and INSTI -R NRTI -R only NRTI -R only 3-4 days Plating 37°C 5% CO2

Forgiveness of Antiretroviral Regimens: In Vitro HIV-1 ... · antiretroviral regimens set at drug in vivo concentrations or concentrations mimicking short lapses in adherence (1 to

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Forgiveness of Antiretroviral Regimens: In Vitro HIV-1 ... · antiretroviral regimens set at drug in vivo concentrations or concentrations mimicking short lapses in adherence (1 to

Forgiveness of Antiretroviral Regimens: In Vitro HIV-1 Viral Breakthrough with 2-Drug versus 3-Drug Regimens Simulating Variable Adherence to Treatment

Andrew Mulato, Rima Acosta, Stephen R. Yant, Tomas Cihlar, and Kirsten WhiteGilead Sciences, Inc., Foster City, CA, USA

Background

References

© 2019 Gilead Sciences, Inc. All rights reserved.10th IAS Conference on HIV Science (IAS 2019), 21-24 July 2019, Mexico City, Mexico

Poster TUPEB244

♦ In vitro viral breakthrough selection experiments help define the resistance barrier of ARV drug combinations and the resistance mutations that emerge. Here, two series of breakthrough experiments were conducted starting with wild-type or low-level M184V HIV and antiretroviral regimens set at drug in vivo concentrations or concentrations mimicking short lapses in adherence (1 to 3 consecutive missed doses)

♦ Using wild-type HIV, BIC+FTC+TAF was better at preventing in vitro viral breakthrough and emergent drug resistance compared to the 2-drug combination of DTG+3TC

– BIC+FTC+TAF had viral breakthrough only in the wild-type experiments under the lowest drug concentrations tested (Cmin after missing 3 doses). Only one viral breakthrough had emergent resistance with M184I in RT. In total, 23/252 wells had breakthrough and 1 had emergent resistance

– In the wild-type experiments, DTG+3TC had 104/144 wells breakthrough and 15 of these had emergent resistance consisting of M184V/I and other mutations in RT, and other mutations in IN, including R263K and S153F, and one with resistance mutations in both RT and IN mutations. M184I has also been reported in DTG+3TC two-drug in vitro breakthrough experiments by another group27

♦ M184V causes resistance to FTC and 3TC and is one of the most frequent mutations to occur during virologic failure, is archived, and can be transmitted29-31. Pre-existing M184V at low levels in the inoculum had viral breakthroughs with DTG+3TC and high-level M184V with or without additional substitutions compared to BIC+FTC+TAF where no breakthrough occurred

♦ These in vitro studies suggest that the favorable pharmacology, antiviral synergy, and resistance profile provided by BIC+FTC+TAF may be more protective against virologic breakthrough and emergent resistance than DTG+3TC in the setting of non-adherence

♦ In vitro studies of the 'forgiveness' of ART regimens are important as they may help to predict regimen durability and risk of resistance in the real world, where sub-optimal adherence is more common

AcknowledgementsThe authors thank Alex Thielen and Martin Däeumer at Seq-IT and Ross Martin, Xinan Liu, and Silvia Chang (Gilead Sciences, Inc.) for deep sequence analysis.

Conclusions

Methods

♦ Current guidelines for the treatment of HIV-1 infection recommend triple therapy consisting of an integrase strand transfer inhibitor (INSTI) plus 2 nucleoside/tide reverse transcriptase inhibitors (NRTIs) including the single-tablet regimen (STR) of bictegravir+emtricitabine+tenofovir alafenamide (BIC/FTC/TAF) 1,2

♦ BIC/FTC/TAF is approved for the treatment of HIV infection, has a high barrier to resistance 3-10, and may offer “forgiveness” (avoiding viral rebound and resistance in the setting of short-term non-adherence)

♦ Optimal adherence to daily oral antiretroviral therapy (ART) is important to minimize the emergence of drug resistance4. Whereas high adherence rates are achieved in clinical trials, treatment adherence rates in the real world are lower and not always predictable11-13

♦ A regimen’s barrier to resistance is both genetic and pharmacologic. Drugs with higher resistance barriers are preferred. Variations in minimal drug concentrations (Cmin) can occur due to natural variation between individuals, drug-drug interactions, food effects, and missed doses

– Genetic Barrier: The number and sequence of mutations needed to generate resistance to a drug

– Pharmacokinetic Barrier: The concentration of drug needed to suppress the virus, clinical drug concentrations, and drug half-life

Figure 1: Schematic of Antiviral Pharmacokinetics

Figure 2. In Vitro Breakthrough Selection Methodology

I. Regimen Forgiveness by In Vitro Breakthrough Selections: Constant Drugs levels at Cmin and After Missing 1-3 Consecutive Doses (HIV-1 IIIb virus)

Results

♦ Recently, controlled clinical trials in treatment-naïve individuals have shown efficacy using an INSTI + 1 NRTI (dolutegravir/lamivudine; DTG+3TC) but the question remains whether the decreased number of regimen components may be less forgiving than triple therapy; virologic failure on DTG/3TC with emergent resistance to both DTG and 3TC has been reported (ACTG 5353)28

Objective ♦ To investigate BIC+FTC+TAF vs. DTG+3TC “forgiveness” by in vitro HIV-1 viral

breakthrough selections and resistance emergence – In cells infected with either wild-type or low-level M184V – Drug exposures modeling full adherence and suboptimal adherence to ART

♦ Test Drug Concentrations (Table 1): BIC and DTG concentrations were calculated using their human plasma clinical trough concentrations (Cmin) according to their prescribing information and adjusted for human plasma protein binding15-17. The TAF Cmin concentration generated tenofovir-diphosphate (TFV-DP) at its physiological concentration in PBMCs from TAF-treated individuals18. FTC and 3TC concentrations were set at their human plasma-free adjusted Cmin concentrations18-20.

♦ Missed Doses: To simulate 1, 2, or 3 consecutive missed doses (Cmin minus 1 dose, Cmin minus 2 doses, and Cmin minus 3 doses), drug concentrations were adjusted by their plasma half-lives for BIC and DTG and active metabolite half-lives for the NRTIs (TAF, FTC, and 3TC). Cmin – X doses determined as Cmin*(0.5^{24*X/ t1/2}). Drug concentrations were kept constant in each selection.

Table 1. Drug Concentrations for Cell Culture Equivalents

a. Cmin values are median values from United States prescribing information (USPI)b. BIC and DTG data generated in-house by standard equilibrium dialysis shift in human serum versus cell culture

media15

c. Cmin / Human Serum Shiftd. t1/2 for FTC, TAF, and 3TC represent intracellular half-life of the active di- or tri- phosphate metabolite18-20; t1/2 values

for BIC and DTG from USPI

♦ Genotypic Analyses: Each well with viral breakthrough was sequenced by next generation sequencing (Seq-IT) and mutations were reported if present at >2%. Mutations were observed between 2.1% and 99% frequency per well.

I. Time to Viral Breakthrough: Cmin and After Missing 1-3 Consecutive Doses (N=144)

I. Resistance in Breakthrough Viruses at Cmin and After Missing 1-3 Consecutive Doses

II. Regimen Forgiveness by In Vitro Breakthrough Selections: Using Low-Level M184V xxLAI Starting Virus Mixtures

II. Resistance in Breakthrough Viruses: Low-Level M184V Starting Virus

Table 2. Mechanisms of Forgiveness and Barrier to Resistance

Mechanisms BIC+FTC+TAF DTG+3TC

Drug Levels

Long plasma or intracellular t1/2

BIC or DTG16-17 17 hr 14 hr FTC-TP or 3TC-TP18-20 37 hr 17.5 hr TFV-DP18 116 hr na

Long IN/DNA dissociation t1/2 for BIC or DTG21-22 132 hr 71-78 hr

Synergy and Mechanisms of Synergy

Combination antiviral activity BIC + FTC or DTG + 3TC15,23-25 Synergy Synergy BIC + TAF15 Synergy na TFV + FTC23-24 Synergy na TFV-chain-termination stabilized by Dead-End Complex with FTC-TP25

Increased TFV Activity na

Resistance Profile

Phenotype of M184V BIC or DTG16 Sensitive Sensitive FTC or 3TC17-18 Resistant Resistant TFV (TAF)26 Hypersensitive na

BIC + FTC + TAF DTG + 3TCBIC FTC TAF DTG 3TC

Clinical Dose (mg) 50 200 25 50 300

Molecular Weight (g/mol) 449.4 247.2 534.5 419.4 229.3

Clinical Cmin (µg/mL)a 2.61 0.096 0.008 1.11 0.042

Clinical Cmin (nM) 5808 388 15 2515 265

Human Serum Shiftb 43.6 1.0 1.0 27.5 1.0

Cell Culture Equivalent Cmin (nM)c 133 388 15 91 265

t ½ (hr)d 17 37 116 14 17.5

• Missing antiretroviral doses results in a predictable decrease of systemic exposures to each drug in the regimen based on their established clinical half-life

Optimal Adherence With 1 – 3 Consecutive Missed Doses

II. Time to Viral Breakthrough: Using Low-Level M184V xxLAI Starting Virus Mixtures

1. DHHS. Human Immunodeficiency Virus-1 Infection: Developing Antiretroviral Drugs for Treatment Guidance for Industry. https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM355128. Published November 2015. Accessed March 2019.

2. DHHS. Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents. https://aidsinfo.nih.gov/guidelines/html/1/adult-andadolescent-arv/0. Published October 2018. Accessed March 2019.

3. Gallant J, et al. The Lancet (2017) 390(10107): 2063-2072 4. Sax PE, et al. The Lancet (2017) 390(10107): 2073-20825. Daar ES, et al. The Lancet HIV (2018) 5(7): e347-e356 6. Molina JM, et al. The Lancet HIV (2018) 5(7): e357-e3657. Wohl D, et al. ID Week (2018) Presentation #742468. Stellbrink HJ, et al. HIV Glasgow (2018) Presentation #41859609. Kityo C, et al. CROI (2018) Presentation #50010. Gaur AH, et al. CROI (2019) Presentation #257111. Cheng Y et al. Medicine (2018) 97(2):1-712. Kangethe, et al. J. Managed Care Pharm (2019) 25(1): 88-9313. Tandon et al. Curr Med Res Opin (2018) 35(1): 63-7114. Cahn P et al. The Lancet (2019) 393(10167): 143-15515. Tsiang M, et al. Antimicrob Agents and Chemother (2016) 60(12): 7086-709716. BIKTARVY® United States Prescribing Information17. TIVICAY® United States Prescribing Information18. Dickinson L, et al. Antimicrob Agents and Chemother (2015) 59(10): 6080-608619. Else LJ et al. Antimicrob Agents and Chemother (2011) 56(3): 1427-143320. Yuen GJ et al. Antimicrob Agents and Chemother (2004) 48(1): 176-18221. Hightower K, et al. Antimicrob Agents and Chemother (2015) 55(10):4552-455922. White K, et al. CROI (2017) Presentation #89323. Laskey S, et al. JCI Insight (2016) 1(19):e9003324. Borroto-Esoda K, et al. Antiv Ther (2006) 11(3): 377-38425. Kulkarni R, et al. Antivir Res (2014) 101: 131-13526. Deval J, et al. J Biol Chem (2004) 279(1):509-51627. Yoshinaga T, et al. Antimicrob Agents and Chemother (2019) 63(30): e02104-1828. Taiwo BO, et al. Clin Infect Dis (2018) 66(11):1689-9729. Castro H, et al., JID (2013) 208(9): 1459-6330. Wainberg MA, et al., JAC (2011) 66(10): 2346-931. Jain V, et al., JID (2011) 203(8): 1174-81

♦ Viral Breakthrough Selections (Figure 2): In vitro viral breakthrough selection experiments should be interpreted as a comparative in vitro study; clinical trials of missed doses for these ARV combinations have not been conducted. MT-2 cells were infected with HIV-1 (IIIb or clonal xxLAI strain containing varying levels of M184V, an FTC and 3TC resistance mutation). Infected cells were cultured in the presence of fixed concentrations of BIC+FTC+TAF or DTG+3TC, split every 3-4 days with fresh media containing drug, and monitored for viral breakthrough by cytopathic effect for up to 5 weeks. Supernatants containing breakthrough virus were collected and stored at the time of cytopathic effect. Statistical comparisons for experiments using the same starting virus backbone were made using Fisher’s exact test.

Gilead Sciences, Inc.333 Lakeside Drive

Foster City, CA 94404 1-800-445-3235

I. Fixed Drug Concentrations

Cmin minus 1 doseCmin minus 2 doses

Cmin minus 3 doses

CminHIV: IIIb

M184V/I K219N/RV75I V118I T215AK70R

R263K* G140ES153F* P145S A128TL74M

123

123

NRTI-R

INSTI-R

Viru

s w

ith R

esis

tanc

e (N

)

BIC+FTC+TAF DTG+3TC

4

4

(Resistance in 1/144) (Resistance in 15/144)

*R263K and S153F have been previously selected by DTG and cause reduced susceptibility to DTGThe well with R263K in IN also had T215A and K219R present.

† M184V and M184I cause high-level resistance to FTC and 3TC and increased sensitivity to TAF; occurred only once for BIC+FTC+TAF and was in the Cmin after 3 missed doses experiments

† M184V and M184I cause high-level resistance to FTC and 3TC; all breakthroughs with M184V were >68%*S153Y has been previously selected by DTG and causes reduced susceptibility to DTG

*R263K and S153F have been previously selected by DTG and cause reduced susceptibility to DTGThe well with R263K in IN also had T215A and K219R present.

M184V M184V+M184I+E157K M184V+E157K

M184V+E138KM184V+D67N M184V+S153Y*

51015

0.4% M184V inoculum

4% M184V inoculumVi

rus

with

Res

ista

nce

(N)

BIC+FTC+TAF DTG+3TC

20

(Resistance in 0/72) (Resistance in 40/72)

M184V†

5101520

M184V+M50I

• All 15 breakthroughs had M184V as the majority species

• All 25 breakthroughs had M184V as the majority species

NRTI and INSTI-R

NRTI and INSTI-R

NRTI-R only

NRTI-R only

3-4 daysPlating37°C

5% CO2