Suppression of human and simian immunodeficiency virus replication with the CCR5-specific antibody Leronlimab in two species

Xiao L Chang, Jason S Reed, Gabriela M Webb, Helen L Wu, Jimmy Le, Katherine B Bateman, Justin M Greene, Cleiton Pessoa, Courtney Waytashek, Whitney C Weber, Joseph Hwang, Miranda Fischer, Cassandra Moats, Oriene Shiel, Rachele M Bochart, Hugh Crank, Don Siess, Travis Giobbi, Jeffrey Torgerson, Rebecca Agnor, Lina Gao, Kush Dhody, Jacob P Lalezari, Ivo Sah Bandar, Alnor M Carnate, Alina S Pang, Michael J Corley, Scott Kelly, Nader Pourhassan, Jeremy Smedley, Benjamin N Bimber, Scott G Hansen, Lishomwa C Ndhlovu, Jonah B Sacha, Xiao L Chang, Jason S Reed, Gabriela M Webb, Helen L Wu, Jimmy Le, Katherine B Bateman, Justin M Greene, Cleiton Pessoa, Courtney Waytashek, Whitney C Weber, Joseph Hwang, Miranda Fischer, Cassandra Moats, Oriene Shiel, Rachele M Bochart, Hugh Crank, Don Siess, Travis Giobbi, Jeffrey Torgerson, Rebecca Agnor, Lina Gao, Kush Dhody, Jacob P Lalezari, Ivo Sah Bandar, Alnor M Carnate, Alina S Pang, Michael J Corley, Scott Kelly, Nader Pourhassan, Jeremy Smedley, Benjamin N Bimber, Scott G Hansen, Lishomwa C Ndhlovu, Jonah B Sacha

Abstract

The CCR5-specific antibody Leronlimab is being investigated as a novel immunotherapy that can suppress HIV replication with minimal side effects. Here we studied the virological and immunological consequences of Leronlimab in chronically CCR5-tropic HIV-1 infected humans (n = 5) on suppressive antiretroviral therapy (ART) and in ART-naïve acutely CCR5-tropic SHIV infected rhesus macaques (n = 4). All five human participants transitioned from daily combination ART to self-administered weekly subcutaneous (SC) injections of 350 mg or 700 mg Leronlimab and to date all participants have sustained virologic suppression for over seven years. In all participants, Leronlimab fully occupied CCR5 receptors on peripheral blood CD4+ T cells and monocytes. In ART-naïve rhesus macaques acutely infected with CCR5-tropic SHIV, weekly SC injections of 50 mg/kg Leronlimab fully suppressed plasma viremia in half of the macaques. CCR5 receptor occupancy by Leronlimab occurred concomitant with rebound of CD4+ CCR5+ T-cells in peripheral blood, and full CCR5 receptor occupancy was found in multiple anatomical compartments. Our results demonstrate that weekly, self-administered Leronlimab was safe, well-tolerated, and efficacious for long-term virologic suppression and should be included in the arsenal of safe, easily administered, longer-acting antiretroviral treatments for people living with HIV-1. Trial Registration: ClinicalTrials.gov Identifiers: NCT02175680 and NCT02355184.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: JBS and LCN have received compensation for serving on the scientific advisory board of CytoDyn, a company that may have commercial interests in the results of this research. JBS, HLW, and GMW have also received compensation for consulting for CytoDyn. The potential conflict of interest has been reviewed and managed by the Oregon Health & Science University. LCN has served as an advisor for Abbvie and ViiV Healthcare unrelated to this study. NP and SK are employees of CytoDyn, owner and developer of Leronlimab. KD is an employee of Amarex Clinical Research, a company that manages clinical trials and regulatory matters for CytoDyn. All other authors declare no competing interests. Authors Kush Dhody, Nader Pourhassan, and Cassandra Moats were unavailable to confirm their authorship contributions. On their behalf, the corresponding author has reported their contributions to the best of their knowledge.

Figures

Fig 1. Leronlimab maintained virologic suppression in…
Fig 1. Leronlimab maintained virologic suppression in humans for over seven years.
(A) Study outline for CD01 and CD01-Extension studies (ClinicalTrials.gov: NCT02175680 and NCT02355184). One-week prior to ART interruption, cohort 1 (n = 2; red) and cohort 2 (n = 3; blue) received weekly SC 350 mg injections. After weeks 246–249, cohort 2 switched to weekly SC 700 mg. Longitudinal HIV-1 RNA copies/mL in plasma for (B) cohort 1 and (C) cohort 2. (D) Total number and (E) mean (±SEM) HIV-1 RNA copies/mL of viral blips (>40 copies/mL). (B-C, E) Horizontal dashed line denotes assay limit of detection (LOD) of 40 copies/mL; undetected viral loads graphed at LOD. (F-G) Left graph shows longitudinal CD4+ T-cell counts in the blood and right graph shows mean (±SD) for all CD4+ T-cell timepoints based on the treatment dose for cohort 1 (E) and cohort 2 (F). (F-G) Two horizontal dotted lines represent the normal range for CD4+ T-cells. Gray and orange boxes denote 350 and 700 mg injections, respectively. (E, G) Two-tailed unpair t test.
Fig 2. Analyses of long-term Leronlimab treatment…
Fig 2. Analyses of long-term Leronlimab treatment in humans.
Blood was collected from 01–037, 01–038, 01–057, 01–061, and 01–064 at weeks 304, 301, 297, 299, and 297, respectively. (A) Integrated HIV-1 DNA copies in CD4+ T-cells. (B) Leronlimab concentration in the plasma. Horizontal dashed line denotes LOD (0.0226 μg/mL). (C) CCR5 RO by Leronlimab on CD4+ T-cells, CD8+ T-cells, and CD14+ monocytes. Intracellular cytokine staining of CD95+ memory (D) CD4+ and (E) CD8+ TM-cells stimulated with HCMV (IE1 and PP65) and HIV (Gag and Nef) peptides. Positive responses were determined by Boolean gating with CD69+TFN-α+ and/or CD69+IFN-γ+.
Fig 3. Leronlimab suppressed SHIV SF162P3 viremia…
Fig 3. Leronlimab suppressed SHIVSF162P3 viremia in macaques.
Macaques were infected intravenously with 1,000 TCID50 SHIVSF162P3. Leronlimab-treated group (n = 4, green) received weekly SC 50 mg/kg starting at week-3 post-infection and untreated controls (n = 5, black). (A) Study outline. (B) Longitudinal CCR5 RO levels by Leronlimab on blood CD4+CCR5+T-cell. (C) Longitudinal plasma concentration; horizontal dashed line denotes LOD (0.0226 μg/mL). (D) Longitudinal SHIVSF162P3 RNA copies/mL in plasma. Horizontal dashed line denotes LOD (50 copies/mL); undetectable viral loads graphed at LOD. (E) Longitudinal fold change from week-0 (baseline) for CD4+CCR5+ T-cell percentage in blood. (D-E) Weekly P-values can be found in S4 Table. (F-G) Mean (±SEM) cell-associated SHIVSF162P3 (F) vDNA and (G) vRNA at week-3 (pre-treatment) and week-15 (necropsy). Horizontal dashed lines denote LOD (7 copies/106 cells). PBMC = peripheral blood mononuclear cell, AxLN = axillary lymph node, MesLN = mesenteric lymph node. (F-G) P-values calculated by two-way repeated-measures ANOVA with Tukey-Kramer adjustment. Gray box represents period of Leronlimab injections.
Fig 4. Tissue penetrance by Leronlimab.
Fig 4. Tissue penetrance by Leronlimab.
Tissues were collected at week-15 (necropsy). Tissue concentration in A) non-brain and (B) brain tissues. (C) CCR5 RO by Leronlimab on CD4+ T-cells. Panels A-C show mean (±SEM). (D) Representative flow cytometry plots showing the costaining of anti-CCR5 (clone 3A9) and Leronlimab (by anti-human IgG4, clone HP-6025) on CD4+ T-cells from control (38224) and Leronlimab-treated (35778) macaques. PBMC = peripheral blood mononuclear cell, Ax LN = axillary lymph node, Ing LN = inguinal lymph node, Mes LN = mesenteric lymph node, BAL = bronchoalveolar lavage.

References

    1. Antiretroviral Therapy Cohort C. Survival of HIV-positive patients starting antiretroviral therapy between 1996 and 2013: a collaborative analysis of cohort studies. Lancet HIV. 2017;4:e349–e356. doi: 10.1016/S2352-3018(17)30066-8
    1. HIV drug resistance report 2017. Geneva: World Health Organization; 2017.
    1. Phillips AN, Stover J, Cambiano V, Nakagawa F, Jordan MR, Pillay D, et al.. Impact of HIV Drug Resistance on HIV/AIDS-Associated Mortality, New Infections, and Antiretroviral Therapy Program Costs in Sub-Saharan Africa. J Infect Dis. 2017;215:1362–1365. doi: 10.1093/infdis/jix089
    1. Global action plan on HIV drug resistance 2017–2021. Geneva: World Health Organization. 2017.
    1. Wijting I, Rokx C, Boucher C, van Kampen J, Pas S, de Vries-Sluijs T, et al.. Dolutegravir as maintenance monotherapy for HIV (DOMONO): a phase 2, randomised non-inferiority trial. Lancet HIV. 2017;4:e547–e554. doi: 10.1016/S2352-3018(17)30152-2
    1. Caskey M, Schoofs T, Gruell H, Settler A, Karagounis T, Kreider EF, et al.. Antibody 10–1074 suppresses viremia in HIV-1-infected individuals. Nat Med. 2017;23:185–191. doi: 10.1038/nm.4268
    1. Samson M, Libert F, Doranz BJ, Rucker J, Liesnard C, Farber CM, et al.. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature. 1996;382:722–725. doi: 10.1038/382722a0
    1. Allen AG, Chung CH, Atkins A, Dampier W, Khalili K, Nonnemacher MR, et al.. Gene Editing of HIV-1 Co-receptors to Prevent and/or Cure Virus Infection. Front Microbiol. 2018;9:2940. doi: 10.3389/fmicb.2018.02940
    1. Thompson MA. The return of PRO 140, a CCR5-directed mAb. Curr Opin HIV AIDS. 2018;13:346–353. doi: 10.1097/COH.0000000000000479
    1. Olson WC, Rabut GE, Nagashima KA, Tran DN, Anselma DJ, Monard SP, et al.. Differential inhibition of human immunodeficiency virus type 1 fusion, gp120 binding, and CC-chemokine activity by monoclonal antibodies to CCR5. J Virol. 1999;73:4145–4155. doi: 10.1128/JVI.73.5.4145-4155.1999
    1. Tilton JC, Wilen CB, Didigu CA, Sinha R, Harrison JE, Agrawal-Gamse C, et al.. A maraviroc-resistant HIV-1 with narrow cross-resistance to other CCR5 antagonists depends on both N-terminal and extracellular loop domains of drug-bound CCR5. J Virol. 2010;84:10863–10876. doi: 10.1128/JVI.01109-10
    1. Westby M, Smith-Burchnell C, Mori J, Lewis M, Mosley M, Stockdale M, et al.. Reduced maximal inhibition in phenotypic susceptibility assays indicates that viral strains resistant to the CCR5 antagonist maraviroc utilize inhibitor-bound receptor for entry. J Virol. 2007;81:2359–2371. doi: 10.1128/JVI.02006-06
    1. Jacobson JM, Saag MS, Thompson MA, Fischl MA, Liporace R, Reichman RC, et al.. Antiviral activity of single-dose PRO 140, a CCR5 monoclonal antibody, in HIV-infected adults. J Infect Dis. 2008;198:1345–1352. doi: 10.1086/592169
    1. Dhody K, Pourhassan N, Kazempour K, Green D, Badri S, Mekonnen H, et al.. PRO 140, a monoclonal antibody targeting CCR5, as a long-acting, single-agent maintenance therapy for HIV-1 infection. HIV Clinical Trials. 2018;19:85–93. doi: 10.1080/15284336.2018.1452842
    1. Jacobson JM, Lalezari JP, Thompson MA, Fichtenbaum CJ, Saag MS, Zingman BS, et al.. Phase 2a study of the CCR5 monoclonal antibody PRO 140 administered intravenously to HIV-infected adults. Antimicrob Agents Chemother. 2010;54:4137–4142. doi: 10.1128/AAC.00086-10
    1. Sorstedt E, Nilsson S, Blaxhult A, Gisslen M, Flamholc L, Sonnerborg A, et al.. Viral blips during suppressive antiretroviral treatment are associated with high baseline HIV-1 RNA levels. BMC Infect Dis. 2016;16:305. doi: 10.1186/s12879-016-1628-6
    1. Chang XL, Wu HL, Webb GM, Tiwary M, Hughes C, Reed JS, et al.. CCR5 Receptor Occupancy Analysis Reveals Increased Peripheral Blood CCR5+CD4+ T Cells Following Treatment With the Anti-CCR5 Antibody Leronlimab. Frontiers in Immunology. 2021;12. doi: 10.3389/fimmu.2021.794638
    1. Phillips KA, Bales KL, Capitanio JP, Conley A, Czoty PW, t Hart BA, et al.. Why primate models matter. American journal of primatology. 2014;76:801–827. doi: 10.1002/ajp.22281
    1. Chang XL, Webb GM, Wu HL, Greene JM, Abdulhaqq S, Bateman KB, et al.. Antibody-based CCR5 blockade protects Macaques from mucosal SHIV transmission. Nat Commun. 2021;12:3343. doi: 10.1038/s41467-021-23697-6
    1. Trkola A, Ketas TJ, Nagashima KA, Zhao L, Cilliers T, Morris L, et al.. Potent, broad-spectrum inhibition of human immunodeficiency virus type 1 by the CCR5 monoclonal antibody PRO 140. Journal of virology. 2001;75:579–588. doi: 10.1128/JVI.75.2.579-588.2001
    1. Ho SH, Trunova N, Gettie A, Blanchard J, Cheng-Mayer C. Different mutational pathways to CXCR4 coreceptor switch of CCR5-using simian-human immunodeficiency virus. J Virol. 2008;82:5653–5656. doi: 10.1128/JVI.00145-08
    1. Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014;5:520. doi: 10.3389/fimmu.2014.00520
    1. Martinez-Picado J, Deeks SG. Persistent HIV-1 replication during antiretroviral therapy. Curr Opin HIV AIDS. 2016;11:417–423. doi: 10.1097/COH.0000000000000287
    1. Thompson CG, Rosen EP, Prince HMA, White N, Sykes C, de la Cruz G, et al.. Heterogeneous antiretroviral drug distribution and HIV/SHIV detection in the gut of three species. Sci Transl Med. 2019;11. doi: 10.1126/scitranslmed.aap8758
    1. Bacchetti P, Deeks SG, McCune JM. Breaking free of sample size dogma to perform innovative translational research. Sci Transl Med. 2011;3:87ps24. doi: 10.1126/scitranslmed.3001628
    1. Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, et al.. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature. 2013;503:224–228. doi: 10.1038/nature12744
    1. Dornadula G, Zhang H, VanUitert B, Stern J, Livornese L Jr., Ingerman MJ, et al.. Residual HIV-1 RNA in blood plasma of patients taking suppressive highly active antiretroviral therapy. JAMA. 1999;282:1627–1632. doi: 10.1001/jama.282.17.1627
    1. Gupta P, Collins KB, Ratner D, Watkins S, Naus GJ, Landers DV, et al.. Memory CD4(+) T cells are the earliest detectable human immunodeficiency virus type 1 (HIV-1)-infected cells in the female genital mucosal tissue during HIV-1 transmission in an organ culture system. J Virol. 2002;76:9868–9876. doi: 10.1128/jvi.76.19.9868-9876.2002
    1. Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, et al.. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 2009;360:692–698. doi: 10.1056/NEJMoa0802905
    1. Symons J, Vandekerckhove L, Hutter G, Wensing AM, van Ham PM, Deeks SG, et al.. Dependence on the CCR5 coreceptor for viral replication explains the lack of rebound of CXCR4-predicted HIV variants in the Berlin patient. Clin Infect Dis. 2014;59:596–600. doi: 10.1093/cid/ciu284
    1. Bednar MM, Hauser BM, Zhou S, Jacobson JM, Eron JJ Jr., Frank I, et al.. Diversity and Tropism of HIV-1 Rebound Virus Populations in Plasma Level After Treatment Discontinuation. J Infect Dis. 2016;214:403–407. doi: 10.1093/infdis/jiw172
    1. Caskey M. Broadly neutralizing antibodies for the treatment and prevention of HIV infection. Curr Opin HIV AIDS. 2020;15:49–55. doi: 10.1097/COH.0000000000000600
    1. Fatkenheuer G, Pozniak AL, Johnson MA, Plettenberg A, Staszewski S, Hoepelman AI, et al.. Efficacy of short-term monotherapy with maraviroc, a new CCR5 antagonist, in patients infected with HIV-1. Nat Med. 2005;11:1170–1172. doi: 10.1038/nm1319
    1. Shapiro MB, Cheever T, Malherbe DC, Pandey S, Reed J, Yang ES, et al.. Single-dose bNAb cocktail or abbreviated ART post-exposure regimens achieve tight SHIV control without adaptive immunity. Nat Commun. 2020;11:70. doi: 10.1038/s41467-019-13972-y
    1. Uzzan M, Tokuyama M, Rosenstein AK, Tomescu C, SahBandar IN, Ko HM, et al.. Anti-alpha4beta7 therapy targets lymphoid aggregates in the gastrointestinal tract of HIV-1-infected individuals. Sci Transl Med. 2018;10. doi: 10.1126/scitranslmed.aau4711
    1. Hansen SG, Piatak M Jr., Ventura AB, Hughes CM, Gilbride RM, Ford JC, et al.. Immune clearance of highly pathogenic SIV infection. Nature. 2013;502:100–104. doi: 10.1038/nature12519
    1. Naing A, Infante J, Goel S, Burris H, Black C, Marshall S, et al.. Anti-PD-1 monoclonal antibody MEDI0680 in a phase I study of patients with advanced solid malignancies. J Immunother Cancer. 2019;7:225. doi: 10.1186/s40425-019-0665-2
    1. Mummidi S, Bamshad M, Ahuja SS, Gonzalez E, Feuillet PM, Begum K, et al.. Evolution of human and non-human primate CC chemokine receptor 5 gene and mRNA. Potential roles for haplotype and mRNA diversity, differential haplotype-specific transcriptional activity, and altered transcription factor binding to polymorphic nucleotides in the pathogenesis of HIV-1 and simian immunodeficiency virus. J Biol Chem. 2000;275:18946–18961. doi: 10.1074/jbc.M000169200
    1. Procopio FA, Fromentin R, Kulpa DA, Brehm JH, Bebin AG, Strain MC, et al.. A Novel Assay to Measure the Magnitude of the Inducible Viral Reservoir in HIV-infected Individuals. EBioMedicine. 2015;2:874–883. doi: 10.1016/j.ebiom.2015.06.019
    1. Cline AN, Bess JW, Piatak M Jr., Lifson JD. Highly sensitive SIV plasma viral load assay: practical considerations, realistic performance expectations, and application to reverse engineering of vaccines for AIDS. J Med Primatol. 2005;34:303–312. doi: 10.1111/j.1600-0684.2005.00128.x
    1. Bimber BN, Burwitz BJ, O’Connor S, Detmer A, Gostick E, Lank SM, et al.. Ultradeep pyrosequencing detects complex patterns of CD8+ T-lymphocyte escape in simian immunodeficiency virus-infected macaques. J Virol. 2009;83:8247–8253. doi: 10.1128/JVI.00897-09
    1. Bimber BN, Dudley DM, Lauck M, Becker EA, Chin EN, Lank SM, et al.. Whole-genome characterization of human and simian immunodeficiency virus intrahost diversity by ultradeep pyrosequencing. J Virol. 2010;84:12087–12092. doi: 10.1128/JVI.01378-10
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26:589–595. doi: 10.1093/bioinformatics/btp698
    1. Nelson EK, Piehler B, Eckels J, Rauch A, Bellew M, Hussey P, et al.. LabKey Server: an open source platform for scientific data integration, analysis and collaboration. BMC Bioinformatics. 2011;12:71. doi: 10.1186/1471-2105-12-71

Source: PubMed

3
Prenumerera