Elsevier

Neurobiology of Disease

Volume 48, Issue 2, November 2012, Pages 228-235
Neurobiology of Disease

Review
Glial-derived neurotrophic factor gene transfer for Parkinson's disease: Anterograde distribution of AAV2 vectors in the primate brain

https://doi.org/10.1016/j.nbd.2011.10.004Get rights and content

Abstract

Delivery of neurotrophic factors to treat neurodegenerative diseases has not been efficacious in clinical trials despite their known potency for promoting neuronal growth and survival. Direct gene delivery to the brain offers an approach for establishing sustained expression of neurotrophic factors but is dependent on accurate surgical procedures to target specific anatomical regions of the brain. Serotype-2 adeno-associated viral (AAV2) vectors have been investigated in multiple clinical studies for neurological diseases without adverse effects; however the absence of significant clinical efficacy after neurotrophic factor gene transfer has been largely attributed to insufficient coverage of the target region. Our pre-clinical development of AAV2-glial-derived neurotrophic factor (GDNF) for Parkinson's disease involved real-time image guided delivery and optimization of delivery techniques to maximize gene transfer in the putamen. We have demonstrated that AAV2 vectors are anterogradely transported in the primate brain with GDNF expression observed in the substantia nigra after putaminal delivery in both intact and nigrostriatal lesioned primates. Direct midbrain delivery of AAV2-GDNF resulted in extensive anterograde transport to multiple brain regions and significant weight loss.

Introduction

An important therapeutic innovation with the potential to dramatically inhibit progression of the motor deficit in Parkinson's disease is the delivery of potent neurotrophic factors directly into the degenerated region of the brain. Delivery of a gene therapy vector or therapeutic agent into the brain via direct injection offers an effective method for bypassing the blood brain barrier and targeting a specific anatomical site. However, the large size and complex architecture of the human brain often prevents scalable translation of small animal experimental methods involving direct brain delivery into effective medical procedures. Previous clinical studies investigating the direct delivery of recombinant glial-derived neurotrophic factor (GDNF) protein into the putamen did not yield significant efficacy (Patel and Gill, 2007). The reason for this disappointing outcome is still unclear, but sub-optimal distribution of infused GDNF protein is suspected. The generation of anti-GDNF antibodies in some subjects, along with reported cerebellar toxicity in Rhesus macaques after cessation of prolonged putamen infusion of GDNF, diminished enthusiasm for this approach (Hovland et al., 2007, Lang et al., 2006). In contrast, gene therapy has become a more favored approach primarily because targeting of much lower levels of growth factor only to the nigrostriatal neurons seems to provide a way of harnessing the potency of neurotrophic factors while limiting the potential side effects that might be triggered through inadvertent escape of the neurotrophic factor into other parts of the brain or into peripheral circulation. Adeno-associated viral (AAV) vectors have emerged as the current vehicle of choice for neurological gene transfer with multiple clinical trials completed or initiated. Over 100 patients have received direct cranial delivery of AAV serotype 2 (AAV2) vectors in clinical trials for Parkinson's disease without any serious adverse events attributable to the vector (Christine et al., 2009, Kaplitt et al., 2007, LeWitt et al., 2011, Marks et al., 2008, Marks et al., 2010, Muramatsu et al., 2009, Muramatsu et al., 2010). The recently completed AAV2-GAD (glutamic acid decarboxylase) gene therapy trial for advanced Parkinson's disease is the first phase 2 double-blinded clinical study to demonstrate the efficacy of gene therapy in a neurological disorder (LeWitt et al., 2011).

The exploration of this technology, however, has revealed complex parameters that must be controlled in order to achieve efficacy without unacceptable side effects. The difficulty of achieving the requisite level of transgene expression over sufficient areas of the striatum to exert a positive effect in Parkinson's disease patients was encountered in a 58-patient controlled (sham surgery) phase 2 trial of AAV2-Neurturin (CERE-120). This trial was sufficiently powered to overcome the anticipated placebo effect prevalent in Parkinson's disease studies. In this study, a total of 16 vector injections were made via 8 needle passes that resulted in only about 15% coverage of the putamen, based on post-mortem analysis of 2 participants in the study who had died from unrelated causes (Bartus et al., 2011). This result suggested that perhaps inadequate distribution of vector within the putamen was responsible for the lack of clinical effect. In contrast, AAV2-mediated putaminal expression of aromatic l-amino acid decarboxylase (AADC), the rate-limiting enzyme required for conversion of l-dopa into dopamine, was substantial as shown by PET with the AADC-specific substrate, 6-[18F]-fluoro-metatyrosine (FMT) (Christine et al., 2009, Eberling et al., 2007, Muramatsu et al., 2010). The use of only two reflux-resistant infusion cannulae per hemisphere in the AAV2-hAADC study enabled pressurized convection-enhanced delivery (CED) that increases parenchymal distribution surrounding the cannula tip. Distribution of the vector infusate in these patients was visualized by postoperative MR imaging within 7 hours of vector delivery and was calculated to cover 22% of the putamen volume (Valles et al., 2010). Although the coverage of the putamen does not appear to be substantially different between the two delivery procedures adopted for these clinical trials, the CED approach required a smaller number of infusions and, therefore, lowered the risk of adverse events associated with intracranial cannula placement. It is, however, evident from these early studies that, without a standardized delivery system that can be implemented in multicenter studies and reliably provide a larger distribution of vector within the target brain structure, it will not be possible to fully evaluate the efficacy of therapeutic gene therapy vectors. Even in the successful AAV2-GAD study where gene transfer was targeted to a relatively small deep brain nucleus, the subthalamic nucleus, it was found that precise targeting was critical to clinical benefits, and no motor improvements were observed when the cannula was not correctly positioned (LeWitt et al., 2011).

Therefore, one of the greatest challenges in the development of therapeutic gene therapy for the treatment neurological disorders is to gain control over the distribution of gene transfer within the central nervous system (CNS). Neurotrophic factors play a critical role in maintaining the CNS and have long been proposed as therapeutic agents for neurodegenerative diseases. However, despite the promise that neurotrophic factors offer for neurodegenerative diseases, including nerve growth factor for Alzheimer's disease (Cattaneo et al., 2008), no neurotrophic factor has established clinical efficacy in a placebo-controlled trial. The challenges in delivering proteins, such as neurotrophic factors, to the brain are evident in the large number of animal and human studies that have focused on developing GDNF, a potent growth factor for dopaminergic neurons, into a treatment for Parkinson's disease (Kirik et al., 2004). Sub-optimal distribution has been blamed for the lack of clinical efficacy in investigations of GDNF or its homolog, Neurturin, to inhibit the progression of motor deficits in Parkinson's disease (Lang et al., 2006, Marks et al., 2010, Nutt et al., 2003). Therefore, a crucial component of our ongoing AAV2-GDNF gene therapy program for Parkinson's disease has been the parallel development of a real-time image-guided neurosurgical drug delivery system. This platform technology significantly enhances the accuracy of targeting deep brain structures and maximizes distribution of vector within the parenchyma surrounding the delivery catheter to ensure optimal coverage of the target structure. Repetitive magnetic resonance (MR) imaging during infusions into the non-human primate (NHP) brain provided real-time feedback of vector distribution that enabled us to optimize the cannula design and prescribe infusion protocols (i.e. infusion rates, volumes and cannula positioning) that minimize the risk of off-target distribution or backflow while maximizing target coverage (Yin et al., 2010, Yin et al., 2011). In collaboration with MRI Interventions (Irvine, CA, USA; formally SurgiVision Inc.), we have developed a reflux-resistant cannula with a 3-mm fused silica distal tip that is compatible with the FDA-approved Clearpoint® intra-operative MR trajectory guidance frame (Richardson et al., 2011a).

An unanticipated consequence of enhanced target coverage has been the repeated observation of AAV2-mediated transduction of neurons in anatomical areas of the primate brain not directly targeted but known to receive axonal projections originating from the site of vector infusion (Kells et al., 2009). A retrospective analysis of our recent studies shows significant evidence for anterograde, but not retrograde, transport of AAV2 vectors in the NHP brain. This anterograde trafficking of AAV2 may be advantageous when the secondary region is also a valid target for gene delivery. However, transgene expression within non-targeted areas of the brain may equally lead to serious adverse events such as the weight loss that occurred after intracerebroventricular delivery of GDNF protein to Parkinson's disease patients (Nutt et al., 2003).

Section snippets

Anterograde distribution of AAV2 in the non-human primate brain

Anterograde distribution of AAV2 vectors in the NHP brain was initially observed after delivery of AAV2-thymidine kinase into the putamen (Hadaczek et al., 2006), and after infusion of AAV2-GFP (green fluorescent protein) into the thalamus (Kells et al., 2009). Neither of the proteins produced by these vectors are secreted, and therefore only directly transduced cells contain thymidine kinase or GFP. The thalamic infusions of AAV2-GFP resulted in extensive GFP expression within layer V and VI

AAV2-GDNF distribution in the dopamine-depleted brain

Axonal trafficking has direct implications for treating basal ganglia disorders such as Parkinson's disease where neuro-protective and neuro-restorative treatments have frequently focused on the maintenance or restoration of dopaminergic activity in the caudate-putamen rather than on the vulnerable dopaminergic neuronal cell bodies residing in the ventral midbrain. The prevailing hypothesis has been that delivery of GDNF, a potent neurotrophic factor for dopaminergic neurons (Lin et al., 1993),

Anterograde distribution of AAV2-GDNF in the aged primate brain

Early in our pre-clinical development program, we performed a safety and tolerability study of AAV2-GDNF in aged Rhesus macaques (over 20 years of age, n = 17) that investigated direct delivery to the putamen or midbrain (Johnston et al., 2009, Su et al., 2009). Aged primates typically have reduced dopaminergic function compared to younger animals and, in addition to being a relevant model for assessing safety of targeted gene transfer in a setting relevant to Parkinson's disease, they can afford

Conclusion

Multiple non-clinical primate and rodent studies have been conducted that demonstrate that direct delivery of AAV2-GDNF to the putamen induces sustained GDNF expression in the basal ganglia at levels that can both protect and restore nigrostriatal function. However, until recently, a safe and reliable clinical procedure has not been advanced that would result in the broad distribution of GDNF throughout the putamen and substantia nigra that we believe is necessary for clinical efficacy. We have

References (48)

  • S. Muramatsu et al.

    A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson's disease

    Mol. Ther.

    (2010)
  • R.M. Richardson et al.

    Interventional MRI-guided Putaminal Delivery of AAV2-GDNF for a Planned Clinical Trial in Parkinson's Disease

    Mol. Ther.

    (2011)
  • I. Stromberg et al.

    Glial cell line-derived neurotrophic factor is expressed in the developing but not adult striatum and stimulates developing dopamine neurons in vivo

    Exp. Neurol.

    (1993)
  • X. Su et al.

    Real-time MR imaging with Gadoteridol predicts distribution of transgenes after convection-enhanced delivery of AAV2 vectors

    Mol. Ther.

    (2010)
  • M.S. Airaksinen et al.

    The GDNF family: signalling, biological functions and therapeutic value

    Nat. Rev. Neurosci.

    (2002)
  • R.T. Bartus et al.

    Bioactivity of AAV2-neurturin gene therapy (CERE-120): differences between Parkinson's disease and nonhuman primate brains

    Mov. Disord.

    (2011)
  • A. Cattaneo et al.

    Towards non invasive nerve growth factor therapies for Alzheimer's disease

    J. Alzheimers Dis.

    (2008)
  • D.L. Choi-Lundberg et al.

    Dopaminergic neurons protected from degeneration by GDNF gene therapy

    Science

    (1997)
  • C.W. Christine et al.

    Safety and tolerability of putaminal AADC gene therapy for Parkinson disease

    Neurology

    (2009)
  • B. Connor et al.

    Differential effects of glial cell line-derived neurotrophic factor (GDNF) in the striatum and substantia nigra of the aged Parkinsonian rat

    Gene Ther.

    (1999)
  • J.L. Eberling et al.

    PET 6-[F]fluoro-L-m-tyrosine Studies of Dopaminergic Function in Human and Nonhuman Primates

    Front. Hum. Neurosci.

    (2007)
  • D.M. Gash et al.

    Functional recovery in parkinsonian monkeys treated with GDNF

    Nature

    (1996)
  • A.C. Granholm et al.

    Glial cell line-derived neurotrophic factor is essential for postnatal survival of midbrain dopamine neurons

    J. Neurosci.

    (2000)
  • R. Grondin et al.

    Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys

    Brain

    (2002)
  • Cited by (47)

    • The molecular pathology of neurodegenerative and psychiatric disorders

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    • Imaging of Gene and Cell-Based Therapies: Basis and Clinical Trials

      2021, Molecular Imaging: Principles and Practice
    • Parkinson's disease: Mechanisms, translational models and management strategies

      2019, Life Sciences
      Citation Excerpt :

      Strategies harness the capacities of adeno-associated virus vector serotype 2 (AAV2) and lenti-vector expression system to deliver the foreign gene into the host genome. The trials involved multiple approaches to prevent neurodegeneration and associated functional outcomes through delivering (i) neurotransmitter gamma-aminobutyric acid producing enzyme genes (AAV2-GAD-65, AAV2-GAD-67), (ii) L-dopa converting enzyme, aromatic L-amino acid decarboxylase (AAV2-AADC), (iii) Tricistronic lenti-vector to transfer three genes, including tyrosine hydroxylase (TH), GTP cyclohydrolase 1 (GTP-CH1) involving dopamine synthesis and AADC, and (iv) genes providing trophic support such as neuturin (AAV2-NRTN) and glia cell-derived neurotrophic factor (AAV2-GDNF) into putamen and SN, bilaterally in majority of trials [194–197]. Gene therapy provides a potential approach to deal and restore the detected mutation in the PD associated genes, even before the onset of PD manifestations.

    View all citing articles on Scopus
    View full text