Safety and tolerability of AAV8 delivery of a broadly neutralizing antibody in adults living with HIV: a phase 1, dose-escalation trial

Joseph P Casazza, Evan M Cale, Sandeep Narpala, Galina V Yamshchikov, Emily E Coates, Cynthia S Hendel, Laura Novik, LaSonji A Holman, Alicia T Widge, Preeti Apte, Ingelise Gordon, Martin R Gaudinski, Michelle Conan-Cibotti, Bob C Lin, Martha C Nason, Olga Trofymenko, Shinyi Telscher, Sarah H Plummer, Diane Wycuff, William C Adams, Janardan P Pandey, Adrian McDermott, Mario Roederer, Avery N Sukienik, Sijy O'Dell, Jason G Gall, Britta Flach, Travis L Terry, Misook Choe, Wei Shi, Xuejun Chen, Florence Kaltovich, Kevin O Saunders, Judy A Stein, Nicole A Doria-Rose, Richard M Schwartz, Alejandro B Balazs, David Baltimore, Gary J Nabel, Richard A Koup, Barney S Graham, Julie E Ledgerwood, John R Mascola, VRC 603 Study Team, Charla Andrews, Anita Arthur, Seemal F Awan, Allison Beck, Eugeania Burch, Maria C Burgos Florez, Nina M Berkowitz, Eli A Boritz, Kevin Carlton, Cora T Cartagena, Christina Carter, Grace L Chen, Pamela Costner, Jennifer Cunningham, Daniel C Douek, Aba M Eshun, Catina Evans, Renunda Hicks, Katherine V Houser, Justine Jones, Brenda Larkin, Lam Le, Floreliz Mendoza, Stephen Migueles, John Misasi, Thuy A Nguyen, Abidemi Ola, Karen Parker, Iris Pittman, La' Shawn Requilman, Ro Shauna Rothwell, Gretchen L Schieber, Jamie Saunders, Sandra Sitar, Colin Tran, Olga Trofymenko, Olga Vasilenko, Sana Waheed, Lingshu Wang, Xiaolin Wang, William Whalen, Pernell Williams, Richard L Wu, Kathy Zephir, Joseph P Casazza, Evan M Cale, Sandeep Narpala, Galina V Yamshchikov, Emily E Coates, Cynthia S Hendel, Laura Novik, LaSonji A Holman, Alicia T Widge, Preeti Apte, Ingelise Gordon, Martin R Gaudinski, Michelle Conan-Cibotti, Bob C Lin, Martha C Nason, Olga Trofymenko, Shinyi Telscher, Sarah H Plummer, Diane Wycuff, William C Adams, Janardan P Pandey, Adrian McDermott, Mario Roederer, Avery N Sukienik, Sijy O'Dell, Jason G Gall, Britta Flach, Travis L Terry, Misook Choe, Wei Shi, Xuejun Chen, Florence Kaltovich, Kevin O Saunders, Judy A Stein, Nicole A Doria-Rose, Richard M Schwartz, Alejandro B Balazs, David Baltimore, Gary J Nabel, Richard A Koup, Barney S Graham, Julie E Ledgerwood, John R Mascola, VRC 603 Study Team, Charla Andrews, Anita Arthur, Seemal F Awan, Allison Beck, Eugeania Burch, Maria C Burgos Florez, Nina M Berkowitz, Eli A Boritz, Kevin Carlton, Cora T Cartagena, Christina Carter, Grace L Chen, Pamela Costner, Jennifer Cunningham, Daniel C Douek, Aba M Eshun, Catina Evans, Renunda Hicks, Katherine V Houser, Justine Jones, Brenda Larkin, Lam Le, Floreliz Mendoza, Stephen Migueles, John Misasi, Thuy A Nguyen, Abidemi Ola, Karen Parker, Iris Pittman, La' Shawn Requilman, Ro Shauna Rothwell, Gretchen L Schieber, Jamie Saunders, Sandra Sitar, Colin Tran, Olga Trofymenko, Olga Vasilenko, Sana Waheed, Lingshu Wang, Xiaolin Wang, William Whalen, Pernell Williams, Richard L Wu, Kathy Zephir

Abstract

Adeno-associated viral vector-mediated transfer of DNA coding for broadly neutralizing anti-HIV antibodies (bnAbs) offers an alternative to attempting to induce protection by vaccination or by repeated infusions of bnAbs. In this study, we administered a recombinant bicistronic adeno-associated virus (AAV8) vector coding for both the light and heavy chains of the potent broadly neutralizing HIV-1 antibody VRC07 (AAV8-VRC07) to eight adults living with HIV. All participants remained on effective anti-retroviral therapy (viral load (VL) <50 copies per milliliter) throughout this phase 1, dose-escalation clinical trial ( NCT03374202 ). AAV8-VRC07 was given at doses of 5 × 1010, 5 × 1011 and 2.5 × 1012 vector genomes per kilogram by intramuscular (IM) injection. Primary endpoints of this study were to assess the safety and tolerability of AAV8-VRC07; to determine the pharmacokinetics and immunogenicity of in vivo VRC07 production; and to describe the immune response directed against AAV8-VRC07 vector and its products. Secondary endpoints were to assess the clinical effects of AAV8-VRC07 on CD4 T cell count and VL and to assess the persistence of VRC07 produced in participants. In this cohort, IM injection of AAV8-VRC07 was safe and well tolerated. No clinically significant change in CD4 T cell count or VL occurred during the 1-3 years of follow-up reported here. In participants who received AAV8-VRC07, concentrations of VRC07 were increased 6 weeks (P = 0.008) and 52 weeks (P = 0.016) after IM injection of the product. All eight individuals produced measurable amounts of serum VRC07, with maximal VRC07 concentrations >1 µg ml-1 in three individuals. In four individuals, VRC07 serum concentrations remained stable near maximal concentration for up to 3 years of follow-up. In exploratory analyses, neutralizing activity of in vivo produced VRC07 was similar to that of in vitro produced VRC07. Three of eight participants showed a non-idiotypic anti-drug antibody (ADA) response directed against the Fab portion of VRC07. This ADA response appeared to decrease the production of serum VRC07 in two of these three participants. These data represent a proof of concept that adeno-associated viral vectors can durably produce biologically active, difficult-to-induce bnAbs in vivo, which could add valuable new tools to the fight against infectious diseases.

© 2022. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.

Figures

Fig. 1 ∣. Schematic representation of the…
Fig. 1 ∣. Schematic representation of the AAV8-VRC07 vector cassette.
The AAV8-VRC07 vector uses a single open reading frame design packaged between two inverted terminal repeats (ITRs) with a CASI promoter, a truncated woodchuck hepatitis post-translational regulatory element (WPRE) and a single SV40 poly-A sequence (SV40 polyA). The CASI promoter is 1.05 kb and consists of a CMV enhancer; a chicken β-actin promoter; a splice donor (SD) inserted between the β-actin promoter and a human ubiquitin C enhancer element; and a splice acceptor (SA). The VRC07 coding cassette contains the VRC07 IgG heavy chain (VRC07 IgG HC) and the VRC07 IgG light chain (VRC07 LC) coding sequences. Between the heavy chain and light chain sequences is an F2A self-processing sequence (2A), which contains a picornavirus-derived ribosomal skip sequence preceded by a furin cleavage site.
Fig. 2 ∣. VRC 603 CONSORT diagram.
Fig. 2 ∣. VRC 603 CONSORT diagram.
Study enrollment, AAV8-VRC07 administration, participant follow-up and data analysis are shown for the three study groups.
Fig. 3 ∣. Concentrations of post-product administration…
Fig. 3 ∣. Concentrations of post-product administration of AAV8-VRC07 plasma DNA.
Longitudinal plasma AAV8-VRC07 DNA from 2 weeks after injection until two sequential longitudinal samples of less than 200 copies per milliliter of AAV8-VRC07 plasma DNA were found. Participants A–C received IM injections of 5 × 1010 vg kg−1 of AAV8-VRC07; Participants D and E received 5 × 1011 vg kg−1; and Participants F–H received 2.5 × 1012 vg kg−1.
Fig. 4 ∣. Longitudinal serum VRC07 concentration…
Fig. 4 ∣. Longitudinal serum VRC07 concentration and tier 1 and tier 2 ADA from immediately before IM injection of AAV8-VRC07 to 80–156 weeks after IM injection of 5 × 1010, 5 × 1011 and 2.5 × 1012 vg kg−1.
Participants without tier 1 and tier 2 ADA are shown in the left column; participants with tier 1 and tier 2 ADA are shown in the right column. Serum VRC07 concentrations for different participants are as shown by colored lined as indicated in the figure legend. Tier 1 ADA is as indicated by black lines, with tier 2 ADA as indicated by solid colored bars over the time at which they were identified. No tier 3 ADA was identified for any participants. For VRC07 concentration data, all data points represent the results of a single assay performed in triplicate. Tier 2 and tier 3 ADA responses represent the average of a single assay done in quadruplicate.
Fig. 5 ∣. Longitudinal neutralization data and…
Fig. 5 ∣. Longitudinal neutralization data and the effect of VRC07 paratope binding by 5C9 on neutralization by purified IgG from study participants.
a, Neutralization of five different tier 2 pseudoviruses as shown in the figure legend by purified IgG for the first seven trial participant enrolled in VRC 603. Participants A–C received 5 × 1010 vg kg−1; Participants D and E received 5 × 1011 vg kg−1; and Participants F and G received 2.5 × 1012 vg kg−1 of AAV8-VRC07 by IM injection. Longitudinal IgG IC50s are reported as μg ml−1 of purified IgG on the left y axis for each pseudoviral strain. VRC07 concentration as determined by 5C9 titration is reported as ng ml−1 of 1 mg ml−1 of purified IgG on the right y axis. Data points represent single determinations. Data points in the shaded area are below the limits of accurate quantitation. b, Neutralization curves adjusted for the concentration of VRC07 in purified IgG serum samples are shown for Participant C at 0, 6, 12, 24 and 52 weeks after IM injection of AAV8-VRC07, as indicated in the figure legend. Neutralization curves for purified in vitro produced VRC07 for each pseudoviral species are overlaid on each plot. Data points for neutralization assays of ex vivo produced VRC07 represent the average of six replicate assays; error bars are ± s.d. Data points for in vitro produced VRC07 represent a single assay, These data are representative of data for Participants A, B and D–G. c, Representative neutralization curve showing the failure of in vitro produced VRC07 to neutralize pseudoviral strain CAP210.E8.SG3 at weeks 6,12, 28 and 52. Each data point represents a single determination. d, From left to right, neutralization of TZBD by purified IgG from Participant C containing the amount of VRC07 indicated on the x axis from week 28 purified IgG; neutralization of TZBD by purified in vitro produced VRC07; and neutralization of TZBD by in vitro produced VRC13, in the presence or absence of 10 μg ml−1 of the VRC07 paratope-binding monoclonal antibody 5C9, as shown in the figure legend. These data are representative of neutralization curves for Participants A, B and D–G. Each data point represents a single determination.

References

    1. Fuchs SP & Desrosiers RC Promise and problems associated with the use of recombinant AAV for the delivery of anti-HIV antibodies. Mol. Ther. Methods Clin. Dev 3, 16068 (2016).
    1. Rerks-Ngarm S et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med 361, 2209–2220 (2009).
    1. Gift SK, Leaman DP, Zhang L, Kim AS & Zwick MB Functional stability of HIV-1 envelope trimer affects accessibility to broadly neutralizing antibodies at its apex. J. Virol 91, e01216–17 (2017).
    1. Torrents de la Pena A et al. Improving the immunogenicity of native-like HIV-1 envelope trimers by hyperstabilization. Cell Rep. 20, 1805–1817 (2017).
    1. Klein JS & Bjorkman PJ Few and far between: how HIV may be evading antibody avidity. PLoS Pathog. 6, e1000908 (2010).
    1. Schiller J & Chackerian B Why HIV virions have low numbers of envelope spikes: implications for vaccine development. PLoS Pathog. 10, e1004254 (2014).
    1. Burton DR & Mascola JR Antibody responses to envelope glycoproteins in HIV-1 infection. Nat. Immunol 16, 571–576 (2015).
    1. Pancera M et al. Structural basis for diverse N-glycan recognition by HIV-1-neutralizing V1–V2-directed antibody PG16. Nat. Struct. Mol. Biol 20, 804–813 (2013).
    1. Wei X et al. Antibody neutralization and escape by HIV-1. Nature 422, 307–312 (2003).
    1. Hartley O, Klasse PJ, Sattentau QJ & Moore JP V3: HIV’s switch-hitter. AIDS Res Hum. Retroviruses 21, 171–189 (2005).
    1. Bonsignori M et al. Antibody-virus co-evolution in HIV infection: paths for HIV vaccine development. Immunol. Rev 275, 145–160 (2017).
    1. Korber B et al. Evolutionary and immunological implications of contemporary HIV-1 variation. Br. Med. Bull 58, 19–42 (2001).
    1. Huang J et al. Identification of a CD4-binding-site antibody to HIV that evolved near-pan neutralization breadth. Immunity 45, 1108–1121 (2016).
    1. Huang J et al. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491, 406–412 (2012).
    1. Mouquet H et al. Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc. Natl Acad. Sci. USA 109, E3268–3277 (2012).
    1. Sok D et al. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc. Natl Acad. Sci. USA 111, 17624–17629 (2014).
    1. Wu X HIV broadly neutralizing antibodies: VRC01 and beyond. Adv. Exp. Med. Biol 1075, 53–72 (2018).
    1. Liu J et al. Antibody-mediated protection against SHIV challenge includes systemic clearance of distal virus. Science 353, 1045–1049 (2016).
    1. Mascola JR et al. Protection of macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J. Virol 73, 4009–4018 (1999).
    1. Rudicell RS et al. Enhanced potency of a broadly neutralizing HIV-1 antibody in vitro improves protection against lentiviral infection in vivo. J. Virol 88, 12669–12682 (2014).
    1. Saunders KO et al. Sustained delivery of a broadly neutralizing antibody in nonhuman primates confers long-term protection against simian/human immunodeficiency virus infection. J. Virol 89, 5895–5903 (2015).
    1. Caskey M et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 522, 487–491 (2015).
    1. Lynch RM et al. Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci. Transl. Med 7, 319ra206 (2015).
    1. Mendoza P et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 561, 479–484 (2018).
    1. Burton DR & Hangartner L Broadly neutralizing antibodies to HIV and their role in vaccine design. Annu Rev. Immunol 34, 635–659 (2016).
    1. Kwong PD, Mascola JR & Nabel GJ Rational design of vaccines to elicit broadly neutralizing antibodies to HIV-1. Cold Spring Harb. Perspect. Med 1, a007278 (2011).
    1. Balazs AB et al. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature 481, 81–84 (2011).
    1. Johnson PR et al. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat. Med 15, 901–906 (2009).
    1. Sharon D & Kamen A Advancements in the design and scalable production of viral gene transfer vectors. Biotechnol. Bioeng 115, 25–40 (2018).
    1. Daya S & Berns KI Gene therapy using adeno-associated virus vectors. Clin. Microbiol. Rev 21, 583–593 (2008).
    1. Duan D et al. Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue. J. Virol 72, 8568–8577 (1998).
    1. Nowrouzi A et al. Integration frequency and intermolecular recombination of rAAV vectors in non-human primate skeletal muscle and liver. Mol. Ther 20, 1177–1186 (2012).
    1. Penaud-Budloo M et al. Adeno-associated virus vector genomes persist as episomal chromatin in primate muscle. J. Virol 82, 7875–7885 (2008).
    1. Brady JM, Baltimore D & Balazs AB Antibody gene transfer with adeno-associated viral vectors as a method for HIV prevention. Immunol. Rev 275, 324–333 (2017).
    1. Schnepp BC & Johnson PR Adeno-associated virus delivery of broadly neutralizing antibodies. Curr. Opin. HIV AIDS 9, 250–256 (2014).
    1. Calcedo R, Vandenberghe LH, Gao G, Lin J & Wilson JM Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J. Infect. Dis 199, 381–390 (2009).
    1. Srivastava A In vivo tissue-tropism of adeno-associated viral vectors. Curr. Opin. Virol 21, 75–80 (2016).
    1. Balazs AB et al. Vectored immunoprophylaxis protects humanized mice from mucosal HIV transmission. Nat. Med 20, 296–300 (2014).
    1. Saunders KO et al. Broadly neutralizing human immunodeficiency virus type 1 antibody gene transfer protects nonhuman primates from mucosal simian-human immunodeficiency virus infection. J. Virol 89, 8334–8345 (2015).
    1. Welles HC et al. Vectored delivery of anti-SIV envelope targeting mAb via AAV8 protects rhesus macaques from repeated limiting dose intrarectal swarm SIVsmE660 challenge. PLoS Pathog. 14, e1007395 (2018).
    1. Martinez-Navio JM et al. Adeno-associated virus delivery of anti-HIV monoclonal antibodies can drive long-term virologic suppression. Immunity 50, 567–575 e565 (2019).
    1. Priddy FH et al. Adeno-associated virus vectored immunoprophylaxis to prevent HIV in healthy adults: a phase 1 randomised controlled trial. Lancet HIV 6, e230–e239 (2019).
    1. Szymczak AL et al. Correction of multi-gene deficiency in vivo using a single ‘self-cleaving’ 2A peptide-based retroviral vector. Nat. Biotechnol 22, 589–594 (2004).
    1. Manno CS et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat. Med 12, 342–347 (2006).
    1. Rangarajan S et al. AAV5-Factor VIII gene transfer in severe hemophilia A. N. Engl. J. Med 377, 2519–2530 (2017).
    1. Jefferis R & Lefranc MP Human immunoglobulin allotypes: possible implications for immunogenicity. MAbs 1, 332–338 (2009).
    1. Ledgerwood JE et al. Safety, pharmacokinetics and neutralization of the broadly neutralizing HIV-1 human monoclonal antibody VRC01 in healthy adults. Clin. Exp. Immunol 182, 289–301 (2015).
    1. Sarzotti-Kelsoe M et al. Optimization and validation of the TZM-bl assay for standardized assessments of neutralizing antibodies against HIV-1. J. Immunol. Methods 409, 131–146 (2014).
    1. Nathwani AC et al. Long-term safety and efficacy of Factor IX gene therapy in hemophilia B. N. Engl. J. Med 371, 1994–2004 (2014).
    1. Lisowski L, Tay SS & Alexander IE Adeno-associated virus serotypes for gene therapeutics. Curr. Opin. Pharm 24, 59–67 (2015).
    1. Fuchs SP et al. AAV-delivered antibody mediates significant protective effects against SIVmac239 challenge in the absence of neutralizing activity. PLoS Pathog. 11, e1005090 (2015).
    1. Fuchs SP, Martinez-Navio JM, Rakasz EG, Gao G & Desrosiers RC Liver-directed but not muscle-directed AAV-antibody gene transfer limits humoral immune responses in rhesus monkeys. Mol. Ther. Methods Clin. Dev 16, 94–102 (2020).
    1. Bar KJ et al. Effect of HIV antibody VRC01 on viral rebound after treatment interruption. N. Engl. J. Med 375, 2037–2050 (2016).
    1. Cale EM et al. Neutralizing antibody VRC01 failed to select for HIV-1 mutations upon viral rebound. J. Clin. Invest 130, 3299–3304 (2020).
    1. Crowell TA et al. Safety and efficacy of VRC01 broadly neutralising antibodies in adults with acutely treated HIV (RV397): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet HIV 6, e297–e306 (2019).
    1. Cunningham CK et al. Safety, tolerability, and pharmacokinetics of the broadly neutralizing human immunodeficiency virus (HIV)-1 monoclonal antibody VRC01 in HIV-exposed newborn infants. J. Infect. Dis 222, 628–636 (2020).
    1. Riddler SA et al. Randomized clinical trial to assess the impact of the broadly neutralizing HIV-1 monoclonal antibody VRC01 on HIV-1 persistence in individuals on effective ART. Open Forum Infect. Dis 5, ofy242 (2018).
    1. Gaudinski MR et al. Safety and pharmacokinetics of the Fc-modified HIV-1 human monoclonal antibody VRC01LS: a phase 1 open-label clinical trial in healthy adults. PLoS Med. 15, e1002493 (2018).
    1. Gaudinski MR et al. Safety and pharmacokinetics of broadly neutralising human monoclonal antibody VRC07-523LS in healthy adults: a phase 1 dose-escalation clinical trial. Lancet HIV 6, e667–e679 (2019).
    1. Caskey M et al. Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nat. Med 23, 185–191 (2017).
    1. Fang J et al. Stable antibody expression at therapeutic levels using the 2A peptide. Nat. Biotechnol 23, 584–590 (2005).
    1. Zhou T et al. Multidonor analysis reveals structural elements, genetic determinants, and maturation pathway for HIV-1 neutralization by VRC01-class antibodies. Immunity 39, 245–258 (2013).
    1. Fang J et al. An antibody delivery system for regulated expression of therapeutic levels of monoclonal antibodies in vivo. Mol. Ther 15, 1153–1159 (2007).
    1. Schambach A et al. Woodchuck hepatitis virus post-transcriptional regulatory element deleted from X protein and promoter sequences enhances retroviral vector titer and expression. Gene Ther. 13, 641–645 (2006).
    1. Casazza JP et al. Therapeutic vaccination expands and improves the function of the HIV-specific memory T-cell repertoire. J. Infect. Dis 207, 1829–1840 (2013).
    1. Prabhakaran M et al. A sensitive method to quantify HIV-1 antibodies in mucosal samples. J. Immunol. Methods 491, 112995 (2021).
    1. Seaman MS et al. Optimization and qualification of a functional anti-drug antibody assay for HIV-1 bnAbs. J. Immunol. Methods 479, 112736 (2020).
    1. Pandey JP et al. Immunoglobulin genes and immunity to HSV1 in Alzheimer’s disease. J. Alzheimers Dis 70, 917–924 (2019).
    1. Schanfield M & van Logem E in Handbook of Experimental Immunology Vol. 94 (ed. Weir D) 1–18 (Blackwell, 1986).

Source: PubMed

3
订阅