BAX 335 hemophilia B gene therapy clinical trial results: potential impact of CpG sequences on gene expression

Barbara A Konkle, Christopher E Walsh, Miguel A Escobar, Neil C Josephson, Guy Young, Annette von Drygalski, Scott W J McPhee, R Jude Samulski, Ivan Bilic, Maurus de la Rosa, Birgit M Reipert, Hanspeter Rottensteiner, Friedrich Scheiflinger, John C Chapin, Bruce Ewenstein, Paul E Monahan, Barbara A Konkle, Christopher E Walsh, Miguel A Escobar, Neil C Josephson, Guy Young, Annette von Drygalski, Scott W J McPhee, R Jude Samulski, Ivan Bilic, Maurus de la Rosa, Birgit M Reipert, Hanspeter Rottensteiner, Friedrich Scheiflinger, John C Chapin, Bruce Ewenstein, Paul E Monahan

Abstract

Gene therapy has the potential to maintain therapeutic blood clotting factor IX (FIX) levels in patients with hemophilia B by delivering a functional human F9 gene into liver cells. This phase 1/2, open-label dose-escalation study investigated BAX 335 (AskBio009, AAV8.sc-TTR-FIXR338Lopt), an adeno-associated virus serotype 8 (AAV8)-based FIX Padua gene therapy, in patients with hemophilia B. This report focuses on 12-month interim analyses of safety, pharmacokinetic variables, effects on FIX activity, and immune responses for dosed participants. Eight adult male participants (aged 20-69 years; range FIX activity, 0.5% to 2.0%) received 1 of 3 BAX 335 IV doses: 2.0 × 1011; 1.0 × 1012; or 3.0 × 1012 vector genomes/kg. Three (37.5%) participants had 4 serious adverse events, all considered unrelated to BAX 335. No serious adverse event led to death. No clinical thrombosis, inhibitors, or other FIX Padua-directed immunity was reported. FIX expression was measurable in 7 of 8 participants; peak FIX activity displayed dose dependence (32.0% to 58.5% in cohort 3). One participant achieved sustained therapeutic FIX activity of ∼20%, without bleeding or replacement therapy, for 4 years; in others, FIX activity was not sustained beyond 5 to 11 weeks. In contrast to some previous studies, corticosteroid treatment did not stabilize FIX activity loss. We hypothesize that the loss of transgene expression could have been caused by stimulation of innate immune responses, including CpG oligodeoxynucleotides introduced into the BAX 335 coding sequence by codon optimization. This trial was registered at www.clinicaltrials.gov as #NCT01687608.

Conflict of interest statement

Conflict-of-interest disclosure: B.A.K. has received research funding from Bioverativ, Pfizer, Sangamo, Shire (a Takeda company), and Spark Therapeutics; and has received payment for consulting from BioMarin, Pfizer, Sanofi, and Spark Therapeutics. M.A.E. has received honoraria and consulting fees from Baxalta, a member of the Takeda group of companies, Bayer, CSL Behring, Genentech/Roche, Kedrion, Novo Nordisk, Pfizer, and Sanofi. G.Y. has received honoraria and consulting fees from Bayer, Bioverativ, CSL Behring, Genentech/Roche, Grifols, Kedrion, Novo Nordisk, Pfizer, Shire, a member of the Takeda group of companies, Spark, and UniQure. A.V.D. has received honoraria and consulting fees from Baxalta, Bayer, BioMarin, Bioverativ/Sanofi, CSL Behring, Hema Biologics, Novo Nordisk, Pfizer, Shire, a member of the Takeda group of companies, and uniQure; and is cofounder and member of the board of directors of Hematherix LLC., a biotech company that is developing superFV, a therapy for bleeding complications. S.W.J.M. was an employee of Asklepios and a co-owner of Chatham at the time of the study and may receive remuneration from future development of this program. R.J.S. is the founder and a shareholder at Asklepios BioPharmaceutical and Bamboo Therapeutics, Inc.; holds patents that have been licensed by the University of North Carolina to Asklepios BioPharmaceutical, for which he receives royalties; and has consulted for Baxter Healthcare and has received payment for speaking. I.B. is an employee of Baxalta Innovations GmbH, a member of the Takeda group of companies. M.d.l.R. was an employee of Baxalta Innovations GmbH, a member of the Takeda group of companies, at the time of the study. B.M.R. was an employee of Baxalta Innovations GmbH, a member of the Takeda group of companies, at the time of the study and is a Takeda stock owner. H.R. was an employee of Baxalta Innovations GmbH, a member of the Takeda group of companies, at the time of the study and is a Takeda stock owner. F.S. was an employee of Baxalta Innovations GmbH, a member of the Takeda group of companies, at the time of the study and is a Takeda stock owner. J.C.C. is an employee of Baxalta US Inc., a member of the Takeda group of companies, and is a Takeda stock owner. B.E. is an employee of Baxalta US Inc., a member of the Takeda group of companies. P.E.M. was an employee of the University of North Carolina and was subsequently an employee of Baxalta US Inc., a member of the Takeda group of companies, at the time of the study (with no conflicts to disclose related to this analysis); he holds patents that have been licensed to Asklepios BioPharmaceutical, for which he receives royalties; and he has received research support through the University of North Carolina from Asklepios BioPharmaceutical, Baxter Healthcare, and Novo Nordisk. The remaining authors declare no competing financial interests.

© 2021 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Schematic drawing of the BAX 335 expression cassette. The recombinant vector genome of BAX 335 is designed to express a codon-optimized FIX Padua (R338L) cDNA from the liver-specific murine transthyretin (TTR) promoter/enhancer. The intron fragment from minute virus of mice (MVM) and the bovine growth hormone (BGH) polyadenylation (pA) sequence help improve expression. The expression cassette is flanked by 1 AAV2 wild-type inverted terminal repeat (ITR) sequence (3′ITR) and 1 mutated AAV2 ITR (MUT 5′ITR) to direct preferential replication and packaging of self-complementary (sc) vector DNA. The overall size of the resulting sc genome is 4806 nucleotides (nt). The BAX 335 nucleotide sequence including ITRs is provided in supplemental Figure 1.
Figure 2.
Figure 2.
FIX activity, FIX Padua levels, liver enzymes, and PBMC-mediated immune responses to AAV8 post–BAX 335 infusion for participants in dose cohort 2. FIX activity and FIX Padua activity levels post–BAX 335 infusion by participant in relation to bleeding episodes, administration of FIX replacement therapy, and prednisone dosing (participant 8) as well as liver enzyme markers of hepatotoxicity (ALT and aspartate aminotransferase [AST]) are shown. Study week represents nominal collection time (mean values used if ≥1 result in 1 week). The number of exogenous FIX infusions or bleeding episodes within 1 week is indicated. The lower limit of detection (LOD) for FIX Padua–specific activity (<0.020 U) is shown by the gray dashed line. Participant 8 received prophylactic treatment with prednisone. The duration of prednisone treatment is shown in the green bar chart, with the slope indicating dose tapering. The results from IFN-γ ELISpot assays are shown in the lower graph panel for each participant; the reaction of the participant’s PBMCs to AAV8 capsid peptides in 3 separate pools are plotted as the number of spots per million PBMCs. Values were considered positive if they were above the media control (background) by a factor of 2 and were >60 spots per million PBMCs (black dashed line). Maximum results across all pools were presented for each time point.
Figure 3.
Figure 3.
FIX activity, FIX Padua levels, liver enzyme levels, and PBMC-mediated immune responses to AAV8 post–BAX 335 infusion for participants in dose cohort 3. FIX and FIX Padua activity post–BAX 335 infusion by participant in relation to bleeding episodes, administration of FIX replacement therapy, and prednisone dosing (participants 6 and 7) as well as liver enzyme markers of hepatotoxicity (ALT and aspartate aminotransferase [AST]) are shown. Study week represents nominal collection time (mean values used if ≥1 result in 1 week). The number of exogenous FIX infusions or bleeding episodes within 1 week is indicated. Participants 6 and 7 received prednisone as a rescue treatment in response to a sudden loss of FIX expression. The duration of prednisone treatment is shown in the green bar charts, with the slope indicating dose tapering. FIX Padua–specific activity values were above the lower limit of detection (LOD) (<0.020 U), which is shown by the gray dashed line. The results from IFN-γ ELISpot assays are shown in the lower graph panel for each participant; the reaction of the participant’s PBMCs to AAV8 capsid peptides in 3 separate pools are plotted as the number of spots per million PBMCs. Values were considered positive if they were above the media control (background) by a factor of 2 and were >60 spots per million PBMCs (black dashed line). Maximum results across all pools were presented for each time point.
Figure 4.
Figure 4.
Peak FIX activity by participant, baseline CRM status, and dose cohort. FIX activity was measured in each participant during the 6 months after BAX 335 infusion. Participants, testing either positive or negative for circulating FIX antigen CRM at baseline, received a single infusion of BAX 335 at 1 of 3 doses. FIX measurements were determined by one-stage clotting assay (central laboratory), including measurements within 5 days of an exogenous FIX infusion and measurements from unscheduled study visits.
Figure 5.
Figure 5.
Anti-AAV8 NAb responses in mice treated with BAX 335 vs CpG-depleted vector constructs. Anti-AAV8 NAb responses, as a surrogate marker for immune activation via TLR9, indicate lower immunogenicity for CpG-depleted vectors. C57BL/6J mice (8-10 weeks old, n = 6-8 per group) were treated IV with identical doses (4 × 1012 vg/kg) of either one of two AAV8 vectors carrying an FIX Padua coding sequence with different numbers of CpG oligodeoxynucleotides (ODNs): BAX 335 (99 CpG ODNs); CpG-depleted candidate (0 CpG ODNs). Blood was collected 4 weeks later, and the magnitude of the resulting titer of anti-AAV8 NAbs was assayed as a marker of adaptive immune responses to CpG ODNs.

References

    1. Mannucci PM, Tuddenham EG. The hemophilias—from royal genes to gene therapy. N Engl J Med. 2001;344(23):1773-1779.
    1. Srivastava A, Brewer AK, Mauser-Bunschoten EP, et al. ; Treatment Guidelines Working Group on Behalf of the World Federation Of Hemophilia . Guidelines for the management of hemophilia. Haemophilia. 2013;19(1):e1-e47.
    1. Manno CS, Pierce GF, Arruda VR, et al. . Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response [published correction appears in Nat Med. 2006;12(5):592]. Nat Med. 2006;12(3):342-347.
    1. Nathwani AC, Rosales C, McIntosh J, et al. . Long-term safety and efficacy following systemic administration of a self-complementary AAV vector encoding human FIX pseudotyped with serotype 5 and 8 capsid proteins. Mol Ther. 2011;19(5):876-885.
    1. Nathwani AC, Reiss UM, Tuddenham EG, et al. . Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med. 2014;371(21):1994-2004.
    1. George LA, Sullivan SK, Giermasz A, et al. . Hemophilia B gene therapy with a high-specific-activity factor IX variant. N Engl J Med. 2017;377(23):2215-2227.
    1. Miesbach W, Meijer K, Coppens M, et al. . Gene therapy with adeno-associated virus vector 5-human factor IX in adults with hemophilia B. Blood. 2018;131(9):1022-1031.
    1. Nathwani AC, Davidoff AM, Tuddenham EGD. Advances in gene therapy for hemophilia. Hum Gene Ther. 2017;28(11):1004-1012.
    1. Jiang H, Couto LB, Patarroyo-White S, et al. . Effects of transient immunosuppression on adenoassociated, virus-mediated, liver-directed gene transfer in rhesus macaques and implications for human gene therapy. Blood. 2006;108(10):3321-3328.
    1. Wang L, Calcedo R, Bell P, et al. . Impact of pre-existing immunity on gene transfer to nonhuman primate liver with adeno-associated virus 8 vectors. Hum Gene Ther. 2011;22(11):1389-1401.
    1. Vandamme C, Adjali O, Mingozzi F. Unraveling the complex story of immune responses to AAV vectors trial after trial. Hum Gene Ther. 2017;28(11):1061-1074.
    1. Nathwani AC, Tuddenham EG, Rangarajan S, et al. . Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med. 2011;365(25):2357-2365.
    1. Monahan PE, Sun J, Gui T, et al. . Employing a gain-of-function factor IX variant R338L to advance the efficacy and safety of hemophilia B human gene therapy: preclinical evaluation supporting an ongoing adeno-associated virus clinical trial. Hum Gene Ther. 2015;26(2):69-81.
    1. Wu Z, Sun J, Zhang T, et al. . Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose. Mol Ther. 2008;16(2):280-289.
    1. Arruda VR, Doshi BS, Samelson-Jones BJ. Emerging therapies for hemophilia: controversies and unanswered questions. F1000 Res. 2018;7:F1000 Faculty Rev-489.
    1. Simioni P, Tormene D, Tognin G, et al. . X-linked thrombophilia with a mutant factor IX (factor IX Padua). N Engl J Med. 2009;361(17):1671-1675.
    1. Chang J, Jin J, Lollar P, et al. . Changing residue 338 in human factor IX from arginine to alanine causes an increase in catalytic activity. J Biol Chem. 1998;273(20):12089-12094.
    1. Bali V, Bebok Z. Decoding mechanisms by which silent codon changes influence protein biogenesis and function. Int J Biochem Cell Biol. 2015;64:58-74.
    1. Inouye S, Sahara-Miura Y, Sato J, Suzuki T. Codon optimization of genes for efficient protein expression in mammalian cells by selection of only preferred human codons. Protein Expr Purif. 2015;109:47-54.
    1. Kimchi-Sarfaty C, Schiller T, Hamasaki-Katagiri N, Khan MA, Yanover C, Sauna ZE. Building better drugs: developing and regulating engineered therapeutic proteins. Trends Pharmacol Sci. 2013;34(10):534-548.
    1. Mauro VP, Chappell SA. A critical analysis of codon optimization in human therapeutics. Trends Mol Med. 2014;20(11):604-613.
    1. Grieger JC, Samulski RJ. Adeno-associated virus vectorology, manufacturing, and clinical applications. Methods Enzymol. 2012;507:229-254.
    1. Grieger JC, Soltys SM, Samulski RJ. Production of recombinant adeno-associated virus vectors using suspension HEK293 cells and continuous harvest of vector from the culture media for GMP FIX and FLT1 clinical vector. Mol Ther. 2016;24(2):287-297.
    1. Weber A, Engelmaier A, Voelkel D, et al. . Development of methods for the selective measurement of the single amino acid exchange variant coagulation factor IX Padua. Mol Ther Methods Clin Dev. 2018;10:29-37.
    1. Kruzik A, Koppensteiner H, Fetahagic D, et al. . Detection of biologically relevant low-titer neutralizing antibodies against adeno-associated virus require sensitive in vitro assays. Hum Gene Ther Methods. 2019;30(2):35-43.
    1. Mingozzi F, Maus MV, Hui DJ, et al. . CD8(+) T-cell responses to adeno-associated virus capsid in humans. Nat Med. 2007;13(4):419-422.
    1. Faust SM, Bell P, Cutler BJ, et al. . CpG-depleted adeno-associated virus vectors evade immune detection. J Clin Invest. 2013;123(7):2994-3001.
    1. Butterfield JSS, Biswas M, Shirley JL, et al. . TLR9-activating CpG-B ODN but not TLR7 agonists triggers antibody formation to Factor IX in muscle gene transfer. Hum Gene Ther Methods. 2019;30(3):81-92.
    1. Kudla G, Lipinski L, Caffin F, Helwak A, Zylicz M. High guanine and cytosine content increases mRNA levels in mammalian cells. PLoS Biol. 2006;4(6):e180.
    1. Kircher M, Witten DM, Jain P, O’Roak BJ, Cooper GM, Shendure J. A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet. 2014;46(3):310-315.
    1. Ferreira RC, Freitag DF, Cutler AJ, et al. . Functional IL6R 358Ala allele impairs classical IL-6 receptor signaling and influences risk of diverse inflammatory diseases. PLoS Genet. 2013;9(4):e1003444.
    1. McFarland-Mancini MM, Funk HM, Paluch AM, et al. . Differences in wound healing in mice with deficiency of IL-6 versus IL-6 receptor. J Immunol. 2010;184(12):7219-7228.
    1. Xu B, Chen Q, Yue C, et al. . Prognostic value of IL-6R mRNA in lung adenocarcinoma and squamous cell carcinoma. Oncol Lett. 2018;16(3):2935-2948.
    1. Nault JC, Datta S, Imbeaud S, et al. . Recurrent AAV2-related insertional mutagenesis in human hepatocellular carcinomas. Nat Genet. 2015;47(10):1187-1193.
    1. Nault JC, Mami I, La Bella T, et al. . Wild-type AAV insertions in hepatocellular carcinoma do not inform debate over genotoxicity risk of vectorized AAV. Mol Ther. 2016;24(4):660-661.
    1. Rangarajan S, Walsh L, Lester W, et al. . AAV5-Factor VIII gene transfer in severe hemophilia. A. N Engl J Med. 2017;377(26):2519-2530.
    1. Ertl HCJ, High KA. Impact of AAV capsid-specific T-cell responses on design and outcome of clinical gene transfer trials with recombinant adeno-associated viral vectors: an evolving controversy. Hum Gene Ther. 2017;28(4):328-337.
    1. Barber GN. Cytoplasmic DNA innate immune pathways. Immunol Rev. 2011;243(1):99-108.
    1. Iwasaki A, Medzhitov R. Control of adaptive immunity by the innate immune system. Nat Immunol. 2015;16(4):343-353.
    1. Finn JD, Nichols TC, Svoronos N, et al. . The efficacy and the risk of immunogenicity of FIX Padua (R338L) in hemophilia B dogs treated by AAV muscle gene therapy. Blood. 2012;120(23):4521-4523.
    1. Nathwani AC, Gray JT, Ng CY, et al. . Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood. 2006;107(7):2653-2661.
    1. Suwanmanee T, Hu G, Gui T, et al. . Integration-deficient lentiviral vectors expressing codon-optimized R338L human FIX restore normal hemostasis in Hemophilia B mice. Mol Ther. 2014;22(3):567-574.
    1. Nathwani AC, Gray JT, McIntosh J, et al. . Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary AAV vector results in stable therapeutic expression of human FIX in nonhuman primates. Blood. 2007;109(4):1414-1421.
    1. McIntosh J, Lenting PJ, Rosales C, et al. . Therapeutic levels of FVIII following a single peripheral vein administration of rAAV vector encoding a novel human factor VIII variant. Blood. 2013;121(17):3335-3344.
    1. Zhu J, Huang X, Yang Y. The TLR9-MyD88 pathway is critical for adaptive immune responses to adeno-associated virus gene therapy vectors in mice. J Clin Invest. 2009;119(8):2388-2398.
    1. Shao W, Earley LF, Chai Z, et al. . Double-stranded RNA innate immune response activation from long-term adeno-associated virus vector transduction. JCI Insight. 2018;3(12):3.
    1. Martino AT, Suzuki M, Markusic DM, et al. . The genome of self-complementary adeno-associated viral vectors increases Toll-like receptor 9-dependent innate immune responses in the liver. Blood. 2011;117(24):6459-6468.
    1. Kuranda K, Jean-Alphonse P, Leborgne C, et al. . Exposure to wild-type AAV drives distinct capsid immunity profiles in humans. J Clin Invest. 2018;128(12):5267-5279.
    1. Wright JF. Codon modification and PAMPs in clinical AAV vectors: the tortoise or the hare? Mol Ther. 2020;28(3):701-703.

Source: PubMed

3
Tilaa