This application seeks funding for studies designed to dissect T cell responses to the capsid antigens of adeno-associated viruses (AAV), different types of AAV vectors and transgene products of such vectors. The goal of this renewal application is to further our understanding on detrimental immune responses that arise upon AAV gene transfer and interfere with sustained expression of therapeutic proteins.
In aim 1 we will determine how long CDS""""""""^ T cells recognize capsid epitopes of different AAV vectors using methods developed during the current funding period.
In aim 2 we will characterize CD8* T cell responses to the transgene product of double-stranded (ds)AAV vectors. As we showed during the last funding period, transgene product-specific CD8'^ T cells induced in mice by single-stranded (ss)AAV vectors are functionally Impaired. In more recent studies we observed that dsAAV vectors stimulate by far more potent CDS* T cell responses at lower doses, which appear to be functional unlike those Induced by ssAAV vectors. We will conduct additional studies to assess immune responses to transgene products of dsAAV vectors and to elucidate if and to what degree such responses interfer with sustained gene transfer.
In aim 3 we will characterize T cell responses to natural AAV infections in non-human primates (NHPs) and in humans. As we showed during the current funding period, both humans and NHPs have memory T cells to AAV capsid. Frequencies of such T cells are very robust in NHPs and markedly lower in humans. This initially appears to contradict the basic hypothesis of this program that pre-existing memory CDS* T cells to AAV capsid are reactivated upon AAV-mediated gene transfer and then cause rejection of cells transduced with AAV vectors expressing a therapeutic protein, as this potential problem was only encountered in humans but not In NHPs. Our hypothesis is that AAV capsid-specific CDS* T cells from humans and NHPs differ in their functionality and that those from humans can mount an effective recall response, while those from rhesus macaques are unable to do so, as will be investigated In depth using samples from NHPs (aim 3.1.), healthy human adults (aim 3.2.1.) and children convalescent from an infection with an adenovirus and potentially AAV (aim 3.2.2). An additional puzzling finding from a clinical AAV gene transfer patient will be followed up in aim 3.2.3. In this trial a patient was treated by hepatic infusion of an AAV2-hF.IX vector while under Immunosuppression (IS). He developed an increase In circulating AAV capsid-specific T cells following gene transfer without showing evidence of liver cell destruction. The pathways underlying this increase of apparently dysfunctional AAV capsid-specific CDS* T cells will be elucidated further with samples from transplant patients taken before and/after initiation of IS.

Public Health Relevance

Gene replacement therapy has the potential to cure inherited diseases. Viral vectors such as AAV vectors are highly suited to deliver genes, but, nevertheless, may not result in sustained transgene expression in humans, as antigens ofthe viral vector and a potentially foreign transgene product may elicit adaptive immune responses that cause rejection of vector-transduced cells. Understanding such immune responses is a pre-requisite to devise avenues to circumvent them, which may then allow for sustained AAV-mediated gene transfer and the associated benefits to human subjects suffering from genetic diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Program Projects (P01)
Project #
5P01HL078810-09
Application #
8502305
Study Section
Heart, Lung, and Blood Initial Review Group (HLBP)
Project Start
Project End
Budget Start
2013-07-01
Budget End
2014-06-30
Support Year
9
Fiscal Year
2013
Total Cost
$381,861
Indirect Cost
$145,265
Name
Wistar Institute
Department
Type
DUNS #
075524595
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
High, Katherine A; Anguela, Xavier M (2016) Adeno-associated viral vectors for the treatment of hemophilia. Hum Mol Genet 25:R36-41
Vercauteren, Koen; Hoffman, Brad E; Zolotukhin, Irene et al. (2016) Superior In vivo Transduction of Human Hepatocytes Using Engineered AAV3 Capsid. Mol Ther 24:1042-1049
Wang, Xiaomei; Terhorst, Cox; Herzog, Roland W (2016) In vivo induction of regulatory T cells for immune tolerance in hemophilia. Cell Immunol 301:18-29
Sharma, Rajiv; Anguela, Xavier M; Doyon, Yannick et al. (2015) In vivo genome editing of the albumin locus as a platform for protein replacement therapy. Blood 126:1777-84
Biswas, Moanaro; Terhorst, Cox; Herzog, Roland W (2015) Treg: tolerance vs immunity. Oncotarget 6:19956-7
Biswas, Moanaro; Sarkar, Debalina; Kumar, Sandeep R P et al. (2015) Synergy between rapamycin and FLT3 ligand enhances plasmacytoid dendritic cell-dependent induction of CD4+CD25+FoxP3+ Treg. Blood 125:2937-47
Rogers, Geoffrey L; Suzuki, Masataka; Zolotukhin, Irene et al. (2015) Unique Roles of TLR9- and MyD88-Dependent and -Independent Pathways in Adaptive Immune Responses to AAV-Mediated Gene Transfer. J Innate Immun 7:302-14
Rogers, Geoffrey L; Herzog, Roland W (2015) Gene therapy for hemophilia. Front Biosci (Landmark Ed) 20:556-603
Hui, Daniel J; Edmonson, Shyrie C; Podsakoff, Gregory M et al. (2015) AAV capsid CD8+ T-cell epitopes are highly conserved across AAV serotypes. Mol Ther Methods Clin Dev 2:15029
Wang, Xiaomei; Su, Jin; Sherman, Alexandra et al. (2015) Plant-based oral tolerance to hemophilia therapy employs a complex immune regulatory response including LAP+CD4+ T cells. Blood 125:2418-27

Showing the most recent 10 out of 74 publications