AAV vectors are exceptionally efficient for gene delivery to the brain by direct intraparenchymal infusion, where they mediate continuous transgene expression perhaps for the lifetime of the experimental animal. These exceptional properties of existing AAV vectors are the basis for ongoing or planned clinical trials for neurological diseases where focal gene delivery is therapeutically effective. However development of gene therapy approaches for many other neurological diseases will require global gene delivery to the CNS. Here we will use in vivo selection of an AAV capsid library and molecular grafting to develop new CNS- targeted AAV vectors with such capability. In the first approach we will use in vivo selection of an AAV capsid library with a diversity of 5x109 clones generated by DNA shuffling of AAV1, 2, 5, 8, 9, and rh10 Cap genes, to identify new brain- and spinal cord-tropic AAV capsids after intravenous or ICV delivery in adult animals. The library will be infused via the tail vein or the cerebral lateral ventricles of adult mice and one month later we will isolate DNA from the brain and spinal cord for PCR amplification of tissue-resident AAV Cap genes. These will be cloned back into the original library plasmid backbone and used to produce more AAV virions for subsequent rounds of in vivo selection. We will sequence 10 AAV Cap genes per round of selection. Once we determine convergence of tissue resident AAV Cap genes for each target, we will prepare recombinant AAV vectors encoding firefly luciferase (Fluc) using the newly selected capsids and infuse them into adult animals to determine their biodistribution. We will use bioluminescence imaging to assess distribution and kinetics of gene expression followed by biochemical quantification of Fluc activity in different organs and histological assessment of transduced cell distribution in brain and spinal cord. In the second approach we will combine AAV vectors with different proteins and peptides previously shown to be highly efficient in ferrying liposomes, enzymes, and siRNAs across the BBB after intravascular infusion. For this we will use chimeric AAV capsids carrying a 14 amino acid biotin acceptor peptide that allows for incorporation of biotin into the AAV capsid during packaging. As brain-targeting molecules we will use Streptavidin fused to a single-chain antibody specific for the mouse Transferrin receptor (TfR), or peptides derived from human ApoB-100, or Rabies virus glycoprotein. The biodistribution of the new AAV-molecular conjugates after intravenous infusion will be assessed as above. The therapeutic efficacy of all new CNS-targeted AAV vectors will be tested in a mouse model of GM1-gangliosidosis, which is a lysosomal storage disease that severely affects the CNS. We expect this new generation of CNS-targeted AAV vectors to foster the development of highly effective gene therapy approaches to treat many childhood, adult, and geriatric neurological diseases currently beyond the reach of modern medicine

Public Health Relevance

The purpose of this proposal is to develop a new generation of AAV vectors capable of targeting the brain via the vasculature. Achieving global gene delivery to the adult CNS remains the 'holy grail'of neural gene therapy, and it is likely necessary to develop effective therapies for many neurological diseases. Here we will investigate two approaches to develop new CNS-targeted AAV vectors: 1) In vivo selection of an AAV capsid library via intravascular and intracerebroventricular delivery in adult animals;2) AAV-molecular conjugates incorporating brain-tropic proteins and peptides. This new generation of AAV vectors may provide the means to develop highly effective gene therapy approaches to treat many childhood, adult, and geriatric neurological diseases currently beyond the reach of modern medicine.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS066310-04
Application #
8327777
Study Section
Special Emphasis Panel (ZMH1-ERB-L (05))
Program Officer
Porter, John D
Project Start
2009-09-01
Project End
2014-08-31
Budget Start
2012-09-01
Budget End
2014-08-31
Support Year
4
Fiscal Year
2012
Total Cost
$322,420
Indirect Cost
$126,420
Name
University of Massachusetts Medical School Worcester
Department
Neurology
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
GuhaSarkar, Dwijit; Neiswender, James; Su, Qin et al. (2017) Intracranial AAV-IFN-? gene therapy eliminates invasive xenograft glioblastoma and improves survival in orthotopic syngeneic murine model. Mol Oncol 11:180-193
GuhaSarkar, Dwijit; Su, Qin; Gao, Guangping et al. (2016) Systemic AAV9-IFN? gene delivery treats highly invasive glioblastoma. Neuro Oncol 18:1508-1518
Choudhury, Sourav R; Fitzpatrick, Zachary; Harris, Anne F et al. (2016) In Vivo Selection Yields AAV-B1 Capsid for Central Nervous System and Muscle Gene Therapy. Mol Ther 24:1247-57
Choudhury, Sourav R; Harris, Anne F; Cabral, Damien J et al. (2016) Widespread Central Nervous System Gene Transfer and Silencing After Systemic Delivery of Novel AAV-AS Vector. Mol Ther 24:726-35
Stoica, Lorelei; Ahmed, Seemin S; Gao, Guangping et al. (2013) Gene transfer to the CNS using recombinant adeno-associated virus. Curr Protoc Microbiol Chapter 14:Unit14D.5
Batista, Ana Rita; Sena-Esteves, Miguel; Saraiva, Maria João (2013) Hepatic production of transthyretin L12P leads to intracellular lysosomal aggregates in a new somatic transgenic mouse model. Biochim Biophys Acta 1832:1183-93
Bowers, William J; Breakefield, Xandra O; Sena-Esteves, Miguel (2011) Genetic therapy for the nervous system. Hum Mol Genet 20:R28-41
Breakefield, Xandra O; Sena-Esteves, Miguel (2010) Healing genes in the nervous system. Neuron 68:178-81