Antibody neutralization poses a barrier to intravitreal adeno-associated viral vector gene delivery to non-human primates

MA Kotterman, L Yin, JM Strazzeri, JG Flannery… - Gene therapy, 2015 - nature.com
MA Kotterman, L Yin, JM Strazzeri, JG Flannery, WH Merigan, DV Schaffer
Gene therapy, 2015nature.com
Gene delivery vectors based on adeno-associated viruses (AAV) have exhibited promise in
both preclinical disease models and human clinical trials for numerous disease targets,
including the retinal degenerative disorders Leber's congenital amaurosis and
choroideremia. One general challenge for AAV is that preexisting immunity, as well as
subsequent development of immunity following vector administration, can severely inhibit
systemic AAV vector gene delivery. However, the role of neutralizing antibodies (NABs) in …
Abstract
Gene delivery vectors based on adeno-associated viruses (AAV) have exhibited promise in both preclinical disease models and human clinical trials for numerous disease targets, including the retinal degenerative disorders Leber’s congenital amaurosis and choroideremia. One general challenge for AAV is that preexisting immunity, as well as subsequent development of immunity following vector administration, can severely inhibit systemic AAV vector gene delivery. However, the role of neutralizing antibodies (NABs) in AAV transduction of tissues considered to be immune privileged, such as the eye, is unclear in large animals. Intravitreal AAV administration allows for broad retinal delivery, but is more susceptible to interactions with the immune system than subretinal administration. To assess the effects of systemic anti-AAV antibody levels on intravitreal gene delivery, we quantified the anti-AAV antibodies present in sera from non-human primates before and after intravitreal injections with various AAV capsids. Analysis showed that intravitreal administration resulted in an increase in anti-AAV antibodies regardless of the capsid serotype, transgene or dosage of virus injected. For monkeys injected with wild-type AAV2 and/or an AAV2 mutant, the variable that most significantly affected the production of anti-AAV2 antibodies was the amount of virus delivered. In addition, post-injection antibody titers were highest against the serotype administered, but the antibodies were also cross-reactive against other AAV serotypes. Furthermore, NAB levels in serum correlated with those in vitreal fluid, demonstrating both that this route of administration exposes AAV capsid epitopes to the adaptive immune system and that serum measurements are predictive of vitreous fluid NAB titers. Moreover, the presence of preexisting NAB titers in the serum of monkeys correlated strongly (R= 0.76) with weak, decaying or no transgene expression following intravitreal administration of AAV. Investigating anti-AAV antibody development will aid in understanding the interactions between gene therapy vectors and the immune system during ocular administration and can form a basis for future clinical studies applying intravitreal gene delivery.
nature.com