Cabotegravir for the prevention of HIV-1 in women: results from HPTN 084, a phase 3, randomised clinical trial

Sinead Delany-Moretlwe, James P Hughes, Peter Bock, Samuel Gurrion Ouma, Portia Hunidzarira, Dishiki Kalonji, Noel Kayange, Joseph Makhema, Patricia Mandima, Carrie Mathew, Elizabeth Spooner, Juliet Mpendo, Pamela Mukwekwerere, Nyaradzo Mgodi, Patricia Nahirya Ntege, Gonasagrie Nair, Clemensia Nakabiito, Harriet Nuwagaba-Biribonwoha, Ravindre Panchia, Nishanta Singh, Bekezela Siziba, Jennifer Farrior, Scott Rose, Peter L Anderson, Susan H Eshleman, Mark A Marzinke, Craig W Hendrix, Stephanie Beigel-Orme, Sybil Hosek, Elizabeth Tolley, Nirupama Sista, Adeola Adeyeye, James F Rooney, Alex Rinehart, William R Spreen, Kimberly Smith, Brett Hanscom, Myron S Cohen, Mina C Hosseinipour, HPTN 084 study group, Aida Asmelash, Alice Sehurutshi, Allan Baguma, Anita Marais, Barbarah Kawoozo, Bongiwe Prudence Malinga, Brenda Gati Mirembe, Brenda Okech, Bryan Esterhuizen, Caroline Murombedzi, Daphne Gadama, Eldinah Hwengwere, Elizabeth Roos, Elizabeth S Magada, Emily Shava, Estelle Piwowar-Manning, Eunice Tahuringana, Felix Gs Muhlanga, Francesca Conradie, Frank Angira, Gertrude Nanyonjo, Girisha Kistnasami, Hazzie Mvula, Ishana Naidoo, Jaco Horak, Jane Jere, Jeeva Moodley, Katie Shin, Kerry Nel, Kevin Bokoch, Lilian Birungi, Lynda Emel, Maletsatsi Monametsi, Marvelous Sibanda, Mercy Mutambanengwe, Miria Chitukuta, Moleen Matimbira, Muchaneta Bhondai-Mhuri, Ncamsile Sibisi, Neetha Morar, Netsai Mudzonga, Paul Natureeba, Paul Richardson, Petina Musara, Pippa Macdonald, Rejoice Nkambule, Repelang Mosime, Rhonda White, Ribka Berhanu, Ritha Ncube-Sihlongonyane, Rogers Sekabira, Samantha Siva, Saresha Pillay, Shamelle Govender, Sheiala Bamweyana, Siyabonga Nzimande, Steve Innes, Sufia Dadabhai, Taraz Samandari, Tchangani Tembo, Thandie Lungu Mabedi, Thandiwe Chirenda, Tinashe Chidemo, Victor Mudhune, Vikesh Naidoo, Wadzanai Samaneka, Yaw Agyei, Yeukai Musodza, Yolandie Fourie, Zakir Gaffoor, Sinead Delany-Moretlwe, James P Hughes, Peter Bock, Samuel Gurrion Ouma, Portia Hunidzarira, Dishiki Kalonji, Noel Kayange, Joseph Makhema, Patricia Mandima, Carrie Mathew, Elizabeth Spooner, Juliet Mpendo, Pamela Mukwekwerere, Nyaradzo Mgodi, Patricia Nahirya Ntege, Gonasagrie Nair, Clemensia Nakabiito, Harriet Nuwagaba-Biribonwoha, Ravindre Panchia, Nishanta Singh, Bekezela Siziba, Jennifer Farrior, Scott Rose, Peter L Anderson, Susan H Eshleman, Mark A Marzinke, Craig W Hendrix, Stephanie Beigel-Orme, Sybil Hosek, Elizabeth Tolley, Nirupama Sista, Adeola Adeyeye, James F Rooney, Alex Rinehart, William R Spreen, Kimberly Smith, Brett Hanscom, Myron S Cohen, Mina C Hosseinipour, HPTN 084 study group, Aida Asmelash, Alice Sehurutshi, Allan Baguma, Anita Marais, Barbarah Kawoozo, Bongiwe Prudence Malinga, Brenda Gati Mirembe, Brenda Okech, Bryan Esterhuizen, Caroline Murombedzi, Daphne Gadama, Eldinah Hwengwere, Elizabeth Roos, Elizabeth S Magada, Emily Shava, Estelle Piwowar-Manning, Eunice Tahuringana, Felix Gs Muhlanga, Francesca Conradie, Frank Angira, Gertrude Nanyonjo, Girisha Kistnasami, Hazzie Mvula, Ishana Naidoo, Jaco Horak, Jane Jere, Jeeva Moodley, Katie Shin, Kerry Nel, Kevin Bokoch, Lilian Birungi, Lynda Emel, Maletsatsi Monametsi, Marvelous Sibanda, Mercy Mutambanengwe, Miria Chitukuta, Moleen Matimbira, Muchaneta Bhondai-Mhuri, Ncamsile Sibisi, Neetha Morar, Netsai Mudzonga, Paul Natureeba, Paul Richardson, Petina Musara, Pippa Macdonald, Rejoice Nkambule, Repelang Mosime, Rhonda White, Ribka Berhanu, Ritha Ncube-Sihlongonyane, Rogers Sekabira, Samantha Siva, Saresha Pillay, Shamelle Govender, Sheiala Bamweyana, Siyabonga Nzimande, Steve Innes, Sufia Dadabhai, Taraz Samandari, Tchangani Tembo, Thandie Lungu Mabedi, Thandiwe Chirenda, Tinashe Chidemo, Victor Mudhune, Vikesh Naidoo, Wadzanai Samaneka, Yaw Agyei, Yeukai Musodza, Yolandie Fourie, Zakir Gaffoor

Abstract

Background: Oral pre-exposure prophylaxis has been introduced in more than 70 countries, including many in sub-Saharan Africa, but women experience considerable barriers to daily pill-taking, such as stigma, judgement, and the fear of violence. Safe and effective long-acting agents for HIV prevention are needed for women. We aimed to evaluate the safety and efficacy of injectable cabotegravir compared with daily oral tenofovir diphosphate plus emtricitabine (TDF-FTC) for HIV prevention in HIV-uninfected women.

Methods: HPTN 084 was a phase 3, randomised, double-blind, double-dummy, active-controlled, superiority trial in 20 clinical research sites in seven countries in sub-Saharan Africa. Participants were eligible for enrolment if they were assigned female sex at birth, were aged 18-45 years, reported at least two episodes of vaginal intercourse in the previous 30 days, were at risk of HIV infection based on an HIV risk score, and agreed to use a long-acting reversible contraceptive method. Participants were randomly assigned (1:1) to either active cabotegravir with TDF-FTC placebo (cabotegravir group) or active TDF-FTC with cabotegravir placebo (TDF-FTC group). Study staff and participants were masked to study group allocation, with the exception of the site pharmacist who was responsible for study product preparation. Participants were prescribed 5 weeks of daily oral product followed by intramuscular injections every 8 weeks after an initial 4-week interval load, alongside daily oral pills. Participants who discontinued injections were offered open-label daily TDF-FTC for 48 weeks. The primary endpoints of the study were incident HIV infection in the intention-to-treat population, and clinical and laboratory events that were grade 2 or higher in all women who had received at least one dose of study product. This study is registered with ClinicalTrials.gov, NCT03164564.

Findings: From Nov 27, 2017, to Nov 4, 2020, we enrolled 3224 participants (1614 in the cabotegravir group and 1610 in the TDF-FTC group). Median age was 25 years (IQR 22-30); 1755 (54·7%) of 3209 had two or more partners in the preceding month. 40 incident infections were observed over 3898 person-years (HIV incidence 1·0% [95% CI 0·73-1·40]); four in the cabotegravir group (HIV incidence 0·2 cases per 100 person-years [0·06-0·52]) and 36 in the TDF-FTC group (1·85 cases per 100 person-years [1·3-2·57]; hazard ratio 0·12 [0·05-0·31]; p<0·0001; risk difference -1·6% [-1·0% to -2·3%]. In a random subset of 405 TDF-FTC participants, 812 (42·1%) of 1929 plasma samples had tenofovir concentrations consistent with daily use. Injection coverage was 93% of the total number of person-years. Adverse event rates were similar across both groups, apart from injection site reactions, which were more frequent in the cabotegravir group than in the TDF-FTC group (577 [38·0%] of 1519 vs 162 [10·7%] of 1516]) but did not result in injection discontinuation. Confirmed pregnancy incidence was 1·3 per 100 person-years (0·9-1·7); no congenital birth anomalies were reported.

Interpretation: Although both products for HIV prevention were generally safe, well tolerated, and effective, cabotegravir was superior to TDF-FTC in preventing HIV infection in women.

Funding: National Institute of Allergy and Infectious Diseases, ViiV Healthcare, and the Bill & Melinda Gates Foundation. Additional support was provided through the National Institute of Mental Health, the National Institute on Drug Abuse, and the Eunice Kennedy Shriver National Institute of Child Health and Human Development. ViiV Healthcare and Gilead Sciences provided pharmaceutical support.

Conflict of interest statement

Declaration of interests PLA has received fees from Merck, ViiV Healthcare, and Gilead Sciences, and research support from Gilead Sciences paid to his institution. AR, WRS, and KS are employees of ViiV Healthcare. JFR is an employee of Gilead Sciences. All other authors declare no competing interests.

Copyright © 2022 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial design TDF-FTC=tenofovir disoproxil fumarate plus emtricitabine.
Figure 2
Figure 2
Trial profile TDF-FTC=tenofovir disoproxil fumarate plus emtricitabine. *One person could have had more than one reason for exclusion.
Figure 3
Figure 3
Cumulative HIV incidence by study group Kaplan-Meier estimates of HIV infection are shown. Four HIV infections were observed in the cabotegravir group (HIV incidence 0·20 per 100 person-years [95% CI 0·06–0·52]) and 36 in the TDF-FTC group (1·85 per 100 person-years [1·3–2·57]). Participants in the cabotegravir group had an 88% lower risk of HIV infection than those in the TDF-FTC group (hazard ratio 0·12 [0·05–0·31]; p

Figure 4

Pharmacological and virological data for…

Figure 4

Pharmacological and virological data for HIV infections in the cabotegravir group The timing…

Figure 4
Pharmacological and virological data for HIV infections in the cabotegravir group The timing of key events for the four infections in the cabotegravir group are shown (participants A1, B1, B2, and DX). Group A includes infections that occurred before enrolment; group B includes infections that occurred with no recent exposure to cabotegravir; and group D includes infections that occurred in participants with appropriately timed cabotegravir injections. Participant DX acquired the infection during the step 2 injection phase but is called DX because three of nine injections were delayed. PA-IC90=protein-adjusted concentration required for 90% viral inhibition. LLoQ= lower limit of quantitation. BLQ=below the limit of quantitation.
Figure 4
Figure 4
Pharmacological and virological data for HIV infections in the cabotegravir group The timing of key events for the four infections in the cabotegravir group are shown (participants A1, B1, B2, and DX). Group A includes infections that occurred before enrolment; group B includes infections that occurred with no recent exposure to cabotegravir; and group D includes infections that occurred in participants with appropriately timed cabotegravir injections. Participant DX acquired the infection during the step 2 injection phase but is called DX because three of nine injections were delayed. PA-IC90=protein-adjusted concentration required for 90% viral inhibition. LLoQ= lower limit of quantitation. BLQ=below the limit of quantitation.

References

    1. UNAIDS Confronting inequalities: lessons for pandemic responses from 40 years of AIDS. July 14, 2021.
    1. WHO . World Health Organization; Geneva: 2015. Policy brief: pre-exposure prophylaxis (PrEP): WHO expands recommendation on oral pre-exposure prophylaxis of HIV infection (PrEP)
    1. Celum CL, Delany-Moretlwe S, Baeten JM, et al. HIV pre-exposure prophylaxis for adolescent girls and young women in Africa: from efficacy trials to delivery. J Int AIDS Soc. 2019;22(suppl 4)
    1. Velloza J, Khoza N, Scorgie F, et al. The influence of HIV-related stigma on PrEP disclosure and adherence among adolescent girls and young women in HPTN 082: a qualitative study. J Int AIDS Soc. 2020;23
    1. Cottrell ML, Yang KH, Prince HM, et al. A translational pharmacology approach to predicting outcomes of preexposure prophylaxis against HIV in men and women using tenofovir disoproxil fumarate with or without emtricitabine. J Infect Dis. 2016;214:55–64.
    1. Andrews CD, Yueh YL, Spreen WR, et al. A long-acting integrase inhibitor protects female macaques from repeated high-dose intravaginal SHIV challenge. Sci Transl Med. 2015;7
    1. Radzio J, Spreen W, Yueh YL, et al. The long-acting integrase inhibitor GSK744 protects macaques from repeated intravaginal SHIV challenge. Sci Transl Med. 2015;7
    1. Markowitz M, Frank I, Grant RM, et al. Safety and tolerability of long-acting cabotegravir injections in HIV-uninfected men (ECLAIR): a multicentre, double-blind, randomised, placebo-controlled, phase 2a trial. Lancet HIV. 2017;4:e331–e340.
    1. Landovitz RJ, Li S, Grinsztejn B, et al. Safety, tolerability, and pharmacokinetics of long-acting injectable cabotegravir in low-risk HIV-uninfected individuals: HPTN 077, a phase 2a randomized controlled trial. PLoS Med. 2018;15
    1. Landovitz RJ, Donnell D, Clement ME, et al. Cabotegravir for HIV prevention in cisgender men and transgender women. N Engl J Med. 2021;385:595–608.
    1. Balkus JE, Brown E, Palanee T, et al. An empiric HIV risk scoring tool to predict HIV-1 acquisition in African women. J Acquir Immune Defic Syndr. 2016;72:333–343.
    1. Eshleman SH, Fogel JM, Piwowar-Manning E, et al. Characterization of HIV infections in women who received injectable cabotegravir or tenofovir disoproxil fumarate/emtricitabine for HIV prevention: HPTN 084. J Infect Dis (in press).
    1. Castillo-Mancilla JR, Zheng JH, Rower JE, et al. Tenofovir, emtricitabine, and tenofovir diphosphate in dried blood spots for determining recent and cumulative drug exposure. AIDS Res Hum Retroviruses. 2013;29:384–390.
    1. Anderson PL, Liu AY, Castillo-Mancilla JR, et al. Intracellular tenofovir-diphosphate and emtricitabine-triphosphate in dried blood spots following directly observed therapy. Antimicrob Agents Chemother. 2017;62:e01710–e01717.
    1. Hendrix CW, Andrade A, Bumpus NN, et al. Dose frequency ranging pharmacokinetic study of tenofovir-emtricitabine after directly observed dosing in healthy volunteers to establish adherence benchmarks (HPTN 066) AIDS Res Hum Retroviruses. 2016;32:32–43.
    1. Donnell D, Baeten JM, Bumpus NN, et al. HIV protective efficacy and correlates of tenofovir blood concentrations in a clinical trial of PrEP for HIV prevention. J Acquir Immune Defic Syndr. 2014;66:340–348.
    1. Marzinke MA, Grinsztejn B, Fogel JM, et al. Characterization of human immunodeficiency virus (HIV) infection in cisgender men and transgender women who have sex with men receiving injectable cabotegravir for HIV prevention: HPTN 083. J Infect Dis. 2021;224:1581–1592.
    1. US Department of Health and Human Services Division of AIDS (DAIDS) table for grading the severity of adult and pediatric adverse events, corrected version 2.1. July, 2017.
    1. Emerson SS, Fleming TR. Parameter estimation following group sequential hypothesis testing. Biometrika. 1990;77:875–892.
    1. Corey L, Gilbert PB, Juraska M, et al. Two randomized trials of neutralizing antibodies to prevent HIV-1 acquisition. N Engl J Med. 2021;384:1003–1014.
    1. Gray GE, Bekker LG, Laher F, et al. Vaccine efficacy of ALVAC-HIV and bivalent subtype C gp120-MF59 in adults. N Engl J Med. 2021;384:1089–1100.
    1. Palanee-Phillips T, Baeten J, Heller K, et al. HIV incidence remains high in women in South Africa: data from the ECHO trial. 10th International AIDS Society Conference on HIV Science; July 21–24, 2019 (abstr LBPEC23).
    1. Pillay D, Stankevitz K, Lanham M, et al. Factors influencing uptake, continuation, and discontinuation of oral PrEP among clients at sex worker and MSM facilities in South Africa. PLoS One. 2020;15
    1. Bärnighausen K, Geldsetzer P, Matse S, et al. Qualitative accounts of PrEP discontinuation from the general population in Eswatini. Cult Health Sex. 2021;23:1198–1214.
    1. Quaife M, Eakle R, Cabrera Escobar MA, et al. Divergent preferences for HIV prevention: a discrete choice experiment for multipurpose HIV prevention products in South Africa. Med Decis Making. 2018;38:120–133.
    1. Minnis AM, Browne EN, Boeri M, et al. Young women's stated preferences for biomedical HIV prevention: results of a discrete choice experiment in Kenya and South Africa. J Acquir Immune Defic Syndr. 2019;80:394–403.
    1. Laher F, Salami T, Hornschuh S, et al. Willingness to use HIV prevention methods among vaccine efficacy trial participants in Soweto, South Africa: discretion is important. BMC Public Health. 2020;20
    1. Marrazzo JM, Ramjee G, Richardson BA, et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N Engl J Med. 2015;372:509–518.
    1. Baeten JM, Brown ER, Hillier SL. Dapivirine vaginal ring for HIV-1 prevention. N Engl J Med. 2017;376:995–996.
    1. Venter WDF, Moorhouse M, Sokhela S, et al. Dolutegravir plus two different prodrugs of tenofovir to treat HIV. N Engl J Med. 2019;381:803–815.
    1. Landovitz RJ, Zangeneh SZ, Chau G, et al. Cabotegravir is not associated with weight gain in human immunodeficiency virus-uninfected individuals in HPTN 077. Clin Infect Dis. 2020;70:319–322.
    1. Eshleman S, Fogel JM, Halvas EK, et al. CAB-LA PrEP: early detection of HIV infection may reduce InSTI resistance risk. 29th Conference on Retroviruses and Opportunistic Infections; Feb 12–16, 2022 (abstr Oral-08).

Source: PubMed

Подписаться