The presence of CpGs in AAV gene therapy vectors induces a plasmacytoid dendritic cell-like population very early after administration

Adeno-associated viral (AAV) vector-based gene transfer is a promising strategy to express transgenes to supplant defective endogenous protein function and/or modulate the course of disease. Today, five AAV-based products have been FDA approved and intense pre-clinical and clinical investigations are underway for their application to a broad range of diseases involving diverse tissues and systems [1].

Long-term, effective transgene expression can be antagonized by the induction of inflammatory B and T lymphocytes that recognize AAV capsid and/or transgene [2]. A long-known source of inflammation for facilitating lymphocyte activation are unmethylated CpG dinucleotides, which are present in variable abundance in natural and recombinant AAV genomes [3]. Unmethylated CpGs serve as bacteria- or virus-derived pathogen-associated molecular pattern ligands for Toll-like receptor (TLR)-9 [4], [5]. TLR9 is a pattern recognition receptor (PRR) that usually resides in the endosomal compartment and is principally expressed in B cells and plasmacytoid dendritic cells (pDCs) [6]. CpG-TLR9 stimulation results in the MyD88 adaptor-mediated activation of NF-κB and IRF7 to effect an innate response marked by the expression of diverse inflammatory cytokines and chemokines, including the Type-I interferons, TNF-α, IL-1β, IL-6, and CCL2 [7]. These factors in turn promote activation of B cells and T cells against AAV capsid and transgene product.

AAV1-human Factor IX (hFIX)administration with the TLR9 agonist oligodeoxynucleotide 1826 (ODN1826) in mice resulted in reduced hFIX expression and significant induction of anti-FIX antibody when compared to sole AAV-hFIX administration [8]. This associated with early (24 h) activation of lymph node and splenic conventional DCs (cDCs), pDCs, and B cells, largely manifested as upregulation of the co-stimulatory molecules CD40 and CD86. Further study noted the importance of Treg cells towards shaping antibody formation to transgene product [9]. In an independent study, CpG motif depletion in a FIX-expressing construct led to reduced neutralizing antibody levels to AAV8 capsid [10].

The impact of CpG-TLR9 signaling on T cell activation following AAV-mediated gene transfer has also been explored. Mice deficient in TLR9 expression showed reductions in nuclear beta-galactosidase (nLacZ) transgene-specific T cell proliferation and IFN-gamma (IFNγ) expression subsequent to AAVrh32.33nLacZ administration [11]. These mice exhibited improved nLacZ expression that associated with reduced inflammatory gene expression and decreased MHC-II expression in transduced muscle. Depleting CpGs in the vector led to similar observations in multiple studies [12], [13], [14], [15], implying that during in vivo AAV-mediated gene transfer, pDCs are stimulated to produce Type-I IFN after TLR9 recognition of CpGs present on the vector genome.

AAV vectors can be used to deliver therapeutic antibodies, targeting a wide range of pathologies [16], [17], [18]. We have been interested in designing vectors able to produce antibodies directed against elements of the contact system such as kallikrein (anti-plasma kallikrein antibodies, α-pKal Ab) for the treatment of hereditary angioedema (HAE) [19], [20]. While evaluating the kinetics of human α-pKal Ab expression from an AAV9 vector in mice, we confirmed that CpG depletion of the human antibody transgene sequence benefited expression in the long-term. Unexpectedly, we also observed a significant effect of CpG depletion as early as 24–48 h following vector administration. We show that a PDCAlo/−CD11b+ myeloid cell population that can bind to the antibody encoded by the transgene is amplified in the presence of CpGs and contributes to the early reduction of transgene product levels.

留言 (0)

沒有登入
gif