Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-25T00:20:36.144Z Has data issue: false hasContentIssue false

Viral vector delivery of neurotrophic factors for Parkinson's disease therapy

Published online by Cambridge University Press:  13 May 2015

Martin J. Kelly
Affiliation:
Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
Gerard W. O'Keeffe
Affiliation:
Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
Aideen M. Sullivan*
Affiliation:
Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
*
Corresponding author: Dr Aideen Sullivan, Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland. E-mail: a.sullivan@ucc.ie
Rights & Permissions [Opens in a new window]

Abstract

Parkinson's disease (PD) is a neurodegenerative disorder characterised by the progressive loss of midbrain dopaminergic neurons, which causes motor impairments. Current treatments involve dopamine replacement to address the disease symptoms rather than its cause. Factors that promote the survival of dopaminergic neurons have been proposed as novel therapies for PD. Several dopaminergic neurotrophic factors (NTFs) have been examined for their ability to protect and/or restore degenerating dopaminergic neurons, both in animal models and in clinical trials. These include glial cell line-derived neurotrophic factor, neurturin, cerebral dopamine neurotrophic factor and growth/differentiation factor 5. Delivery of these NTFs via injection or infusion to the brain raises several practical problems. A new delivery approach for NTFs involves the use of recombinant viral vectors to enable long-term expression of these factors in brain cells. Vectors used include those based on adenoviruses, adeno-associated viruses and lentiviruses. Here we review progress to date on the potential of each of these four NTFs as novel therapeutic strategies for PD, as well as the challenges that have arisen, from pre-clinical analysis to clinical trials. We conclude by discussing recently-developed approaches to optimise the delivery of NTF-carrying viral vectors to the brain.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2015 

Dopaminergic neurotrophic factors (NTFs)

NTFs are proteins which promote the survival and healthy development of neurons; the effects of these factors are mediated via stimulation of multiple intracellular signalling pathways in these cells (for reviews see Refs Reference Gordon and Blobe1, Reference Saarma2, Reference Sariola and Saarma3). NTFs which act on dopaminergic neurons have been extensively studied as therapeutics for Parkinson's disease (PD), since this neurodegenerative disorder is caused by the death of dopaminergic neurons projecting from the substantia nigra in the midbrain to the caudate-putamen in the forebrain. Loss of these neurons results in the characteristic motor symptoms of PD-bradykinesia, rigidity and resting tremor. NTFs have potential to protect and restore degenerating dopaminergic neurons, thus slowing or halting the disease progression. NTFs which have potent dopaminergic neuroprotective effects include glial cell-line derived neurotrophic factor (GDNF), neurturin (NRTN) and growth/differentiation factor 5 (GDF5). These NTFs belong to the transforming growth factor-β (TGFβ) protein superfamily. The neurotrophic effects of GDNF, NRTN and GDF5 on the nigrostriatal pathway have been studied for more than a decade; both GDNF and NRTN have been tested in clinical trials (for review see Ref. Reference Sullivan and Toulouse4). However, these proteins are not the only NTFs being explored for their potential in PD. Cerebral dopamine neurotrophic factor (CDNF) and its protein paralogue, mesencephalic astrocyte-derived neurotrophic factor (MANF), have recently been demonstrated to confer neurotrophic and restorative actions on the nigrostriatal pathway (for review see Ref. Reference Lindholm and Saarma5).

NTFs exert their effects by binding to their individual receptors on the neuronal cell membrane, then triggering specific intracellular signalling cascades. For many TGFβ superfamily members, signalling is initiated through the ligand binding to a type-2 serine/threonine receptor. The resulting ligand–receptor complex recruits and binds to the corresponding type-1 serine/threonine receptor. Phosphorylation of the type-1 receptor at a glycine–serine-rich domain by the type-2 receptor promotes downstream activation of receptor-Smads (R-Smads). The R-Smads bind to a co-Smad, Smad-4, which facilitates entry of the complex into the nucleus and thus regulates the transcription of a variety of genes (for reviews see Refs Reference Gordon and Blobe1, Reference Hegarty, O'Keeffe and Sullivan6). In the case of GDF5, the type-1 and type-2 receptors most commonly used are bone morphogenetic protein receptor (BMPR)-1b and BMPR-2, respectively (Ref. Reference Nishitoh7). The resulting activated R-Smads are Smad-1, Smad-5 and Smad-8.

GDNF and NRTN signalling occurs via a different mechanism to that of GDF5 and other TGFβs. These two factors act by activating a receptor tyrosine kinase, Ret, after forming a ligand–receptor complex with a glycosylphosphatidylinositol-anchored GDNF family receptor α (GFRα). GDNF preferentially binds to GFRα1, while NRTN preferentially binds to GFRα2. Once Ret has been stimulated by the ligand–receptor complex, a Ret–Ret homodimer is formed and is auto-phosphorylated. Downstream intracellular signalling pathways activated by phosphorylated Ret include those involving mitogen-activated protein kinase, c-Jun N-terminal kinase (JNK), Src kinases, Akt-phosphoinositide-3 kinase and phospholipase Cγ (for reviews see Refs Reference Saarma2, Reference Sariola and Saarma3).

Potential of dopaminergic NTFs for treatment of PD

There is a pressing need for disease-modifying therapies for PD, since currently-used treatments focus on symptom management rather than on their cause. NTFs hold significant promise in this respect. The most widely-used pharmacological approach is administration of levodopa (L-DOPA), a precursor of dopamine, to replenish diminished levels of striatal dopamine. However, at least half of L-DOPA users experience drug-induced dyskinesia or other motor complications after about 5 years of therapy. Agents which are used in combination with L-DOPA to optimise its effects include DA agonists, peripheral aromatic amino acid decarboxylase inhibitors, monoamine oxidase B inhibitors and catechol-O-methyl transferase inhibitors (for reviews see Refs Reference Toulouse and Sullivan8, Reference Yokochi9). Alternatives to pharmacological treatment, for patients in which L-DOPA therapy does not work well or no longer works, include deep brain stimulation (DBS). DBS confers improvements in motor symptoms due to ablation of brain structures such as the thalamus or pallidum. This strategy is not without complications and is typically used as a last resort to manage motor symptoms (Ref. Reference Beric10). Like L-DOPA, DBS does not halt or slow the progression of the neurodegeneration. Thus, other approaches, including cell-based therapies and NTFs, are under intensive investigation as disease-modifying therapies.

The use of NTFs has the potential advantage that these factors may protect the remaining nigrostriatal dopaminergic neurons from delayed or further damage, rather than only treating the disease symptoms. This is crucial, as approximately 50% of these neurons have degenerated, corresponding to a loss of striatal input of about 80%, by the time that PD symptoms are clinically apparent (for review see Ref. Reference Samii, Nutt and Ransom11). Since approximately 90% of PD cases are of idiopathic origin, earlier detection of PD in the general population is a difficult challenge to overcome at present. Thus the use of NTFs as an intervention in the neurodegenerative process upon PD diagnosis has the potential to be a very beneficial approach.

Application of dopaminergic NTFs as recombinant proteins

GDNF

GDNF is the most extensively studied dopaminergic NTF to date. In vitro studies highlighted GDNF's neurotrophic effects on developing dopaminergic neurons (Refs Reference Krieglstein12, Reference Lin13, Reference Widmer14). GDNF treatment enhanced neuritic branching in cultured embryonic rat dopaminergic neurons (Ref. Reference Widmer14) and protected them against death induced by the neurotoxins 1-methyl-4-phenylpyridinium (MPP+) (Ref. Reference Hou, Lin and Mytilineou15) and 6-hydroxydopamine (6-OHDA) (Ref. Reference Eggert16), in vitro models of the neurodegeneration that occurs in PD. In vivo, GDNF has been shown to bestow significant neuroprotection on adult rodent midbrain dopaminergic neurons against 6-OHDA lesions in short-term (Refs Reference Bowenkamp17, Reference Hoffer18, Reference Kearns and Gash19, Reference Sauer, Rosenblad and Bjorklund20, Reference Shults, Kimber and Martin21) and long-term (Ref. Reference Sullivan, Opacka-Juffry and Blunt22) studies (for reviews see Refs Reference Sullivan and Toulouse4, Reference Bjorklund23). Such neuroprotection was also observed when GDNF was administered prior to or following injection of the neurotoxin l-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the mouse midbrain (Ref. Reference Tomac24). Delayed injection of GDNF after 6-OHDA lesions can confer significant restorative effects on the host striatum (Refs Reference Rosenblad, Martinez-Serrano and Bjorklund25, Reference Aoi26). Furthermore, GDNF treatment enhances the survival and function of embryonic rat dopaminergic grafts in 6-OHDA-lesioned rats (Refs Reference Granholm27, Reference Apostolides28, Reference Sullivan, Pohl and Blunt29, Reference Yurek30). Lastly, GDNF treatment can induce neuroprotective effects in monkey parkinsonian models (Refs Reference Gash31, Reference Gerhardt32, Reference Grondin33, Reference Iravani34, Reference Maswood35, Reference Miyoshi36).

Due to the encouraging performance of GDNF in both in vitro and animal studies, intracerebral application of this protein was examined in human clinical trials. Since NTFs do not cross the blood–brain barrier and are rapidly biometablised in vivo, clinical application requires them to be injected intracerebrally. In the initial clinical trial, recombinant human GDNF (Liatermin®; Amgen Incorporated) was infused via a intraventricular route in 38–50 patients undertaking a randomised, placebo-controlled double-blind trial (Ref. Reference Nutt37). No significant increases in the Unified Parkinson's Disease Rating Scale (UPDRS) scores were observed and participants experienced undesirable symptoms ranging from appetite loss to depression and paraesthesia (Ref. Reference Nutt37). The poor outcome of this trial may have been due to the fact that delivery was intraventricular. More positive results were achieved following GDNF administration directly to the brain parenchyma, in two open-label trials in the UK (Refs Reference Gill38, Reference Patel39) and the USA (Ref. Reference Slevin40). Both of these studies infused GDNF into the putamen, and observed significant overall improvements in the UPDRS scores for both on- and off-medication phases in patients with advanced PD. These patients did not suffer any significant side-effects, apart from a mild Lhermitte's sign (an electrical sensation that runs down the back and into the limbs). The success of these open-label trials led to the instigation of a randomised, placebo-controlled trial using intraputaminal delivery of GDNF (Ref. Reference Lang41). Unfortunately the promising therapeutic effects seen in the open-label trials were not reproduced. Furthermore, safety issues were raised, as some patients developed antibodies towards the exogenous human GDNF (Ref. Reference Tatarewicz42). In 2013, following a safety trial in six PD patients, the Bristol group initiated a phase 2 clinical trial involving 36 patients, implementing a new infusion strategy for GDNF protein. This ongoing trial uses an optimised delivery port and the researchers are hopeful that this will circumvent some of the problems encountered in the previous trials.

NRTN

NRTN is a member of the GDNF family of proteins (Ref. Reference Kotzbauer43), and has been shown to exert neurotrophic effects on embryonic rat dopaminergic neurons in vitro, to the same extent as GDNF (Refs Reference Horger44, Reference Akerud45). Unlike those treated with GDNF, however, NRTN-supplemented neurons did not display extensive branching (Ref. Reference Akerud45). The impacts of NRTN on survival and branching of nigral dopaminergic neurons in a 6-OHDA rat model of PD reflected those observed in the in vitro study (Ref. Reference Akerud45). Another in vivo study showed that NRTN protected the function of nigrostriatal neurons, as expressed in terms of amphetamine-induced rotational asymmetry, following 6-OHDA-lesioning of the adult rat medial forebrain bundle (MFB) (Ref. Reference Horger44) or striatum (Refs Reference Rosenblad46, Reference Oiwa47). Rosenblad et al. (Ref. Reference Rosenblad46) observed that injection of NRTN into the striatum or ventricles conferred a lower level of dopaminergic neuroprotection than that achieved after intranigral injection, as reported by Horger et al. (Ref. Reference Horger44). The level of protection conferred was also lower than that achieved by intrastriatal GDNF, suggesting that NRTN has poorer solubility and diffusion properties in vivo (Ref. Reference Rosenblad46).

The neuroprotective effects of intraputaminal NRTN infusion have also been tested in primates. Chronic infusion of NRTN in MPTP-lesioned rhesus monkeys resulted in an improved parkinsonian motor score (Ref. Reference Grondin48), albeit a lower effect than that conferred by GDNF infusion in a previous study (Ref. Reference Grondin33). The number of dopaminergic neurons in the substantia nigra of NRTN-treated monkeys did not differ significantly from that of MPTP-only group (Ref. Reference Grondin48), again highlighting the issue of poor diffusion of NRTN (Ref. Reference Rosenblad46). Clinical trials of NRTN protein have not been conducted, probably because of the safety issues that arose during the GDNF trials (Refs Reference Lang41, Reference Tatarewicz42). However, trials using NRTN with adeno-associated virus (AAV) type-2 (AAV2) have been completed, as discussed below.

CDNF

Preliminary findings from an in vitro study of MANF (Ref. Reference Petrova49) led to the development of in vivo studies of its paralogue, CDNF, in animal models of PD. The initial study described MANF as an astrocyte-derived factor, which had selective protective actions on dopaminergic neurons in culture (Ref. Reference Petrova49). Thus, CDNF was proposed as a novel NTF for PD treatment. Subsequent analysis led to the categorisation of a new family of proteins consisting of CDNF and MANF (Refs Reference Lindholm50, Reference Parkash51). The neurotrophic potential of CDNF was then demonstrated in an in vivo rat model of PD, the intrastriatal 6-OHDA lesion (Ref. Reference Lindholm50). Unilateral intrastriatal CDNF given 6 h prior to 6-OHDA conferred neuroprotective effects at least as potent as those of GDNF (Ref. Reference Lindholm50). CDNF administration 4 weeks after striatal 6-OHDA lesioning resulted in partial neurorestoration of the nigrostriatal system (Ref. Reference Lindholm50). In mice, intrastriatal injection of CDNF just prior to, or 1 week after, peripheral MPTP injection, conferred dopaminergic neuroprotective or restorative properties, respectively (Ref. Reference Airavaara52). Chronic intrastriatal infusion of recombinant human CDNF over 2 weeks in 6-OHDA-lesioned rats induced cumulative improvements in motor behaviour and preservation of nigral dopaminergic neurons, to a greater extent than GDNF infusion (Ref. Reference Voutilainen53). This study also demonstrated that radiolabelled CDNF protein followed a similar retrograde transport profile to that of GDNF, being transported to the nigra when administered via intrastriatal infusion (Ref. Reference Voutilainen53). No chronic infusion approach of CDNF protein has so far been explored in primate models of PD or in clinical trials. Nevertheless, viral vector-mediated CDNF research is underway, with in vivo studies examining the effects of AAV2-mediated CDNF delivery to the nigrostriatal pathway in animal PD models, as discussed below (Refs Reference Bäck54, Reference Ren55).

GDF5

Like GDNF, the neurotrophic and neuroprotective effects of GDF5 on dopaminergic neurons have been demonstrated in vitro and in vivo. The first study showed that treatment with recombinant human GDF5 protected cultured rat dopaminergic neurons from MPP+-induced damage (Ref. Reference Krieglstein56). GDF5 treatment was further found to promote dopaminergic neuronal survival and stimulate their neurite outgrowth in vitro (Refs Reference O'Keeffe, Dockery and Sullivan57, Reference Costello58, Reference Hegarty, Sullivan and O'Keeffe59, Reference Hegarty60), properties which are of value to its therapeutic application. Pretreatment with recombinant human GDF5 had survival-promoting and functional effects on embryonic rat dopaminergic neurons after transplantation to the 6-OHDA-lesioned rat striatum; these neuroprotective effects were comparable with those seen after GDNF pretreatment (Ref. Reference Sullivan, Pohl and Blunt29). In adult rats, intracerebral injection of GDF5 protected nigrostriatal dopaminergic neurons against 6-OHDA lesions (Refs Reference Sullivan61, Reference Sullivan62). The efficacy of GDF5 was highest after injection into the striatum and/or nigra, in comparison with intraventricular delivery (Ref. Reference Sullivan62). Thus, although intraventricular administration of NTFs would be less invasive in a clinical setting, it appears that direct administration to the parenchyma is the most efficacious approach. For NTFs to be used clinically in PD patients, they would be administered after significant striatal denervation had already taken place. Thus, it is important to test the effects of potentially therapeutic factors when they are administered after a nigrostriatal lesion, in animal models. Promising results were reported in a study which achieved restoration of nigrostriatal function when recombinant GDF5 was administered at 1 or 2 weeks after intrastriatal 6-OHDA (Ref. Reference Hurley, Costello and Sullivan63). A greater effect was seen when GDF5 was administered after a 1-week delay than after 2 weeks (Ref. Reference Hurley, Costello and Sullivan63), highlighting the issue that timing is important in the clinical setting.

Potential for gene therapy in NTF therapy for PD

The more advanced the disease by the time of NTF administration, the less effective the therapy is likely to be. At least until earlier diagnosis of PD is possible, it is critical that NTF therapy is optimised to ensure the best possible clinical efficacy, even in the advanced disease state. Since administration of NTF proteins is hampered by their rapid metabolism in the brain, methods which ensure a consistent supply of NTF to dopaminergic neurons are desirable. Overexpression of NTFs using a gene therapy approach may be the optimal manner in which to ensure an adequate supply to the degenerating nigrostriatal neurons (Fig. 1). Gene therapy has previously been applied in patients with advanced-stage PD, to deliver the enzyme glutamic acid decarboxylase; this was the first double-blind clinical trial to show an effect of gene therapy in a neurological disease (Ref. Reference LeWitt64). Thus, the gene therapy approach has been proven to be safe and effective in PD.

Figure 1 Experimental strategy used to examine the efficacy of viral-mediated delivery of NTFs in preclinical models of PD. (a) PD results from a loss of dopaminergic neurons in the SN which leads to a reduction in striatal dopaminergic innervation. (b) Preclinical assessment of the neuroprotective efficacy of viral-mediated delivery systems involves stereotactic administration of the viral vector to the striatum or midbrain, prior to, at the same time, or after, stereotactic administration of a neurotoxin (e.g. 6-OHDA) to either the (SNpc), striatum (Str) or medial forebrain bundle (mfb). (c) Assessment of motor function after surgery, coupled with (d) post-mortem assessment of numbers of dopaminergic neurons and their striatal processes, is used to determine efficacy of viral-delivered NTFs.

Viral vectors used in gene therapy

Viruses are naturally efficient at introducing genes into target cells. There are several viral vectors which possess natural tropism for fully mature neurons; each type has individual benefits and shortfalls (Fig. 2). Viruses that have been used in development of vectors for NTF delivery include adenovirus (AdV), AAV and lentivirus (LV) (for reviews see Refs Reference Bjorklund23, Reference Karra and Dahm65, Reference White66). Each of these is capable of delivering their genome into nondividing cells such as neurons, supporting their use in neuronal gene therapy. However, size limits exist on the viral capacity to carry transgenes. AdV and LV viruses possess much larger capacities than AAV for transporting recombinant DNA. To provide an effective, efficient, long-term treatment approach, stable integration into the host DNA is sought. Unlike AAV and LV, AdV does not integrate into the host genome. AdV has another disadvantage in that it can induce dose-dependent inflammatory responses in hosts. With regards to vectors that can be integrated, LV vectors ensure much earlier protein expression than AAV vectors (hours versus several days; for review see Ref. Reference Karra and Dahm65). For AAV-delivered NTFs, this additional time needed for transgene expression must be considered when assessing the usefulness of the system to deliver NTFs in a clinical paradigm. Some LV vectors present a potential tumour-producing risk, due to the manner in which they integrate into the genome. AAV vectors, in contrast, are deemed to be safe for clinical use. Finally, AAV has a diffusion advantage over AdV and LV due to its particle size, which is 25 nm in diameter, compared with at least 70 nm for AdV particles, whereas LV particles are no less than 100 nm in diameter. While AdV has been the most frequently chosen virus to date for all clinical trials (for review see Ref. Reference Ginn67), AAV2 has been the virus of choice for developing gene therapeutics for PD.

Figure 2 Distinct features of AV, AAV and LV vectors. AdV, AAV and LV vectors have all been explored for the delivery of NTFs in PD models. Panels (a–c) show the events involved from in vitro generation to stereotactic administration of each viral vector type. Shown also is an overview of the distinct features of each viral vector when applied to the brain in vivo.

Gene therapy approaches to deliver GDNF, NRTN, CDNF and GDF5

GDNF

GDNF-expressing vectors have been designed and tested in animal studies using AdV, LV and AAV (for review see Ref. Reference Bjorklund23). GDNF delivered by AdV vector to the substantia nigra (Refs Reference Choi-Lundberg68, Reference Mandel69) or striatum (Refs Reference Balemans and Van Hul70, Reference Bilang-Bleuel71, Reference Choi-Lundberg72) of rats with intrastriatal 6-OHDA lesions induced motor improvements and protection of nigral dopaminergic neurones. When administered into the substantia nigra after a 6-OHDA MFB lesion, AdV-delivered GDNF also resulted in significant motor improvements (Ref. Reference Lapchak73), whereas in the intrastriatal lesion model, it was effective when injected into the substantia nigra, but not the striatum (Ref. Reference Kozlowski74). In MPTP-treated mice, AdV-mediated GDNF delivery to the striatum prevented depletion of striatal dopamine levels (Ref. Reference Kojima75).

Administration of AAV–GDNF into the rat striatum, but not substantia nigra, 4 weeks prior to 6-OHDA lesion surgery resulted in significant improvements on amphetamine-induced rotational tests (Ref. Reference Kirik76). In marmosets, injection of AAV2–GDNF 3 weeks before 6-OHDA-lesion surgery induced significant reductions in amphetamine-induced rotations (Ref. Reference Eslamboli77). In these AAV2–GDNF-treated monkeys, dopaminergic neuronal sprouting was observed in the lesioned striatum, as well as significant sparing of dopaminergic neuronal cell bodies in the substantia nigra (Ref. Reference Eslamboli77). Both of these studies involved the injection of the therapeutic vector prior to administration of the dopaminergic toxin. However, in order for viral vectors to be therapeutically applicable, they need to be shown to induce restoration of nigrostriatal function when administered after the lesion.

LV-based vectors have also been used to deliver GDNF in animal models of PD, conferring neuroprotective effects on the nigrostriatal pathway of 6-OHDA-lesioned rodents (Refs Reference Bensadoun78, Reference Georgievska79) and MPTP-treated primates (Ref. Reference Kordower80). Intrastriatal delivery of LV-GDNF was effective in reducing 6-OHDA-induced damage to nigrostriatal neurons, protecting nigral dopaminergic neuronal cell bodies and resulting in sprouting of their lesioned axons along the nigrostriatal pathway (Ref. Reference Georgievska79). This study reported that LV-driven GDNF expression was maintained for at least 8 months in vivo, and that GDNF protein was anterogradely transported to the nigra from the intrastriatal injection site. Another study showed that LV-GDNF protects 6-OHDA-lesioned rats from deficits in an operant task which measures complex motor functions, as well as deficits in several tests of simple motor function (Ref. Reference Dowd81). LV-GDNF was also shown to induce reinnervation of the partially denervated striatum, and to restore motor function, when injected 4 weeks after intranigral 6-OHDA (Ref. Reference Brizard82).

In contrast to the promising results of studies that applied GDNF in 6-OHDA-lesioned rats, a recent study found that GDNF delivery by either AAV or LV vectors was not effective in preventing or minimising neurodegeneration caused by an AAV-α-synuclein (AAV-α-syn) vector (Ref. Reference Decressac83). Mutations in α-synuclein are associated with autosomal dominant PD (for reviews see Refs Reference Bekris, Mata and Zabetian84, Reference Deng and Yuan85). Overexpression of α-synuclein using AAV is a recently developed animal model of PD, in which protracted and progressive degeneration of the nigrostriatal pathway is observed (for review see Ref. Reference Ulusoy86). This model has an advantage over the 6-OHDA model as it mimics some of the pathology of the human disease. An LV-GDNF vector and an AAV-GDNF vector were separately tested in AAV-α-syn-treated adult rats (Ref. Reference Decressac83). LV-GDNF was administered 2 weeks prior to the AAV-α-syn injection, whereas AAV-GDNF was administered 3 weeks beforehand (Ref. Reference Decressac83). Target sites for the therapeutic vector injections included the rostral substantia nigra, within the striatum, or both (Ref. Reference Decressac83). Neither LV-GDNF nor AAV-GDNF significantly reduced the dopaminergic degeneration induced by the AAV-α-syn vector, despite the observed overexpression of GDNF from both vector systems (Ref. Reference Decressac83). A subsequent study showed that α-synuclein overexpression downregulates the expression of Nurr1, and its downstream target, the GDNF receptor Ret (Ref. Reference Decressac87). Further investigation of the cellular mechanisms underlying the neuropathology observed in the AAV-α-syn rat model is warranted, in order to ascertain the validity of this model for future studies on NTFs. AAV-mediated overexpression of α-synuclein may result in cytosolic levels of α-synuclein that exceed those seen in the human disease. Thus, it may be necessary to refine these models to more closely mimic the neuropathology of human PD. Clearly, the choice of animal model has a critical impact on whether a potential therapeutic may be considered for future clinical trials. The difference between the effects of GDNF treatment in the α-synuclein model and the more widely-used 6-OHDA and MPTP models mandates considerable thought in the validation of therapeutics for PD. Indeed, it is possible that GDNF may not be the best choice of NTF for PD therapy, since the expression of its receptor is downregulated by α-synuclein. Strategies to circumvent this limitation could involve attempts to upregulate Nurr1, in combination with optimised GDNF delivery to the nigrostriatal system, as discussed below. However, it may be that GDNF is not the best factor for clinical use, which strengthens the argument for investigation of other dopaminergic neurotrophic factors that act via receptors that are not affected by the disease pathology.

In terms of optimised delivery, several recent studies by the Bankiewicz group, using a system of convection-enhanced delivery (CED) to deliver AAV2-GDNF in animal models, have shown promise (Refs Reference Eberling88, Reference Johnston89, Reference Kells90, Reference Richardson91, Reference Su92, Reference Su93). The use of CED as an alternative approach of viral vector delivery is relatively novel (for review see Ref. Reference Salegio94), although this approach had initially been used to demonstrate the delivery of macromolecules nearly two decades ago (Ref. Reference Bobo95). Johnston et al. (Ref. Reference Johnston89) began preliminary testing of AAV2-GDNF via CED in aged rhesus macaques, while a parallel study used MPTP-lesioned rhesus macaques (Ref. Reference Eberling88). In the first study, the safety of this approach was confirmed, as was very effective delivery of GDNF protein to the target regions (Refs Reference Johnston89, Reference Su92). Low-dose, unilateral putaminal infusion of AAV2-GDNF slowed the decline in locomotor activity that is a normal feature of aging in these monkeys (Ref. Reference Johnston89). Efficient transport of GDNF to the caudate, as well as putamen, was achieved following intranigral infusion of the vector, indicating anterograde transport of GDNF along the nigrostriatal pathway. This study also found that AAV2-GDNF induced upregulation of tyrosine hydroxylase (TH) expression, significant increases in the size (but not number) of nigral dopaminergic cell bodies, and enhancement of dopaminergic function in the ipsilateral putamen (Ref. Reference Johnston89). Although this study was not performed in a PD model, the confirmation of safety and effective delivery in the aged monkey brain was an important step towards the clinical application of this delivery approach in PD patients, the majority of whom would be of advanced age. Encouraging results were also seen in the parallel study, which used MPTP lesions to induce a model of early stage PD in one hemisphere and advanced-stage PD in the other hemisphere, in rhesus monkeys (Ref. Reference Eberling88). Infusion of AAV2-GDNF via CED 4 months after establishment of the lesions induced significant improvements in clinical rating scale scores over the following nine months, compared with control animals that had received saline by CED (Ref. Reference Eberling88). A significant increase in dopaminergic function, as measured by PET, was observed in both mildly- and severely-lesioned hemispheres at 6 months post-treatment, compared with that in controls (Ref. Reference Eberling88). As with the companion study performed in aged monkeys, the safety of this GDNF delivery approach was confirmed (Ref. Reference Su92). This study, by showing the efficacy of AAV2-GDNF in already-lesioned animals, provides important support for the eventual clinical application of CED-mediated AAV2-GDNF in PD patients. The long-term neurorestorative capability of CED-mediated AAV2-GDNF in this group of MPTP-lesioned primates was verified after 24 months (Ref. Reference Kells90). The improvements in motor function and striatal dopaminergic function which had been recorded after 6 and 12 months in the earlier study (Ref. Reference Eberling88) were maintained for 24 months (Ref. Reference Kells90). Post-mortem analysis showed increases in TH-immunopositive fibre density and sprouting in the lesioned putamen (Ref. Reference Kells90). Importantly, no significant adverse effects were recorded in this study, indicating good tolerance of high-dose GDNF delivery to the putamen in the long term. The only concern was weight loss, and dyskinesia in a leg of one monkey, which the authors attribute to mispositioning of a cannula in this animal (Ref. Reference Kells90). Weight loss issues following GDNF treatment have been noted in other studies (for examples, see Refs Reference Iravani34, Reference Miyoshi36, Reference Nutt37, Reference Su92, Reference Manfredsson96, Reference Manfredsson97). This highlights the importance of optimal positioning of delivery cannulae for safe and accurate delivery of NTFs in future clinical studies. Another important point to be raised by the above studies is that anterograde transport of GDNF or AAV-2-GDNF particles appears to be the predominant manner in which GDNF is conveyed in the brain following intracerebral administration (Refs Reference Johnston89, Reference Kells90). This suggests that intraputaminal infusion of AAV2 vectors for delivery of NTFs is the most appropriate, as it will allow transport of the vector and/or factor to the nigra along striatonigral axonal projections, which are largely intact in PD patients, even when nigrostriatal projections have severely degenerated. It is worth noting that these studies by the Bankiewicz group investigated the regenerative effects of GDNF in the already-lesioned monkey brain, in contrast to the majority of studies, which examined the protective effects of NTFs delivered prior to the lesion. Although there have been some studies reporting the restorative effects of GDNF (Refs Reference Rosenblad, Martinez-Serrano and Bjorklund25, Reference Aoi26), CNTF (Refs Reference Lindholm50, Reference Airavaara52) and GDF5 (Ref. Reference Hurley, Costello and Sullivan63) in rodent PD models, there is a need to extend these studies to primate models. Optimisation and standardisation of study design in preclinical models will allow the results to be more applicable to clinical studies, which by necessity evaluate the restorative rather than protective effects of NTFs in the diseased brain.

With the issue of optimal positioning of delivery cannulae in mind, studies by the Bankiewicz group explored magnetic resonance imaging-guided CED of AAV2-GDNF in a preclinical setting, using gadoteridol (Gd) to assist in real-time tracking of the infusions (Refs Reference Richardson91, Reference Su93, Reference Gimenez98). Gd was shown to be an accurate tracer for the distribution of GDNF protein after CED-administered AAV2-delivery (Refs Reference Richardson91, Reference Su93, Reference Gimenez98). Richardson et al. successfully tested the delivery of Gd using the ClearPoint System™ technology from SurgiVision Inc. (Ref. Reference Richardson99) and further validated this approach for AAV2-GDNF delivery (Ref. Reference Richardson91). Bankiewicz and co-workers have also carefully considered the surgical cannula design (Refs Reference Krauze100, Reference Sanftner101, Reference Yin, Forsayeth and Bankiewicz102) and insertion zones (Refs Reference Yin, Forsayeth and Bankiewicz102, Reference Yin103), to optimise the delivery of AAV vectors to the brain. Thus, this optimised method of CED-mediated delivery provides a useful tool to track the infusion of viral vectors to the brain, and has the potential to greatly enhance current delivery strategies for NTFs in PD. Since the main problems with the unsuccessful clinical trials to date appear to be related to insufficient or inappropriate delivery of the therapeutic factor to the target tissue, such advances in delivery methodologies should promote the drive towards the instigation of future trials.

Regulated delivery of the therapeutic gene of interest is of great value in PD, as it allows the opportunity to fine-tune the dose of NTF to be delivered, depending on the patients’ responses. This can be achieved by peripheral administration of a drug which can enter the brain and regulate transcription of the therapeutic gene encoded by the viral vector. Hadaczek et al. used this approach to control GDNF delivery from a modified AAV-2 vector, in which GDNF transcription was regulated by the administration of rapamycin (Ref. Reference Hadaczek104). The therapeutic implications of this system have yet to be evaluated, but this approach holds great promise for the future widespread clinical use of viral vector-mediated NTF delivery.

NRTN

An initial report showed that intracerebral injection of LV-NRTN conferred neuroprotective effects on the rat nigrostriatal system against intrastriatal 6-OHDA (Ref. Reference Fjord-Larsen105). Subsequently, AAV2-delivered NRTN, under the name CERE-120 (Ceregene Incorporated), was tested in 6-OHDA-lesioned rats (Refs Reference Bartus106, Reference Gasmi107, Reference Herzog108) and MPTP-lesioned monkeys (Refs Reference Bartus109, Reference Kordower110). Both young (Refs Reference Bartus109, Reference Herzog111) and aged nonparkinsonian monkeys (Refs Reference Bartus109, Reference Herzog112) were used to test the safety and in vivo expression profile of CERE-120. Intrastriatal CERE-120 2 weeks prior to 6-OHDA conferred neuroprotection of rat nigral dopaminergic neurons, as well as stable expression of NRTN protein, for at least 1 year, with no adverse effects (Ref. Reference Gasmi107). Monkeys which had received CERE-120 injections in both striatum and nigra 4 days after MPTP administration displayed significant improvements in parkinsonian clinical scores, as well as significant preservation of nigrostriatal dopaminergic neurons, during a 10-month study (Ref. Reference Kordower110). Both anterograde and retrograde transport of NRTN was detected by post-mortem analysis (Ref. Reference Kordower110). Significant increases in dopaminergic fibre density were found in the ipsilateral striatum of aged monkeys at eight months after intrastriatal CERE-120 (Ref. Reference Herzog112). A further study showed the safety of intrastriatal CERE-120 in primates, following extensive toxicological and pathological analysis (Ref. Reference Herzog111). One result of concern which emerged was that CERE-120 resulted in downregulation of TH expression in rat nigrostriatal dopaminergic neurons, albeit without affecting their structural integrity. However, this effect was not observed in monkeys and was deemed to be a rodent-specific nontoxic compensatory response to the increase in dopaminergic tone induced by NRTN (for review see Ref. Reference Bartus113), as had been previously reported for GDNF (Ref. Reference Georgievska, Kirik and Bjorklund114). Despite the variable statistical significance of these pre-clinical data, the indication that CERE-120 was safe, well-tolerated even at very high doses (for review see Ref. Reference Bartus113) and potentially efficacious in rodents and nonhuman primates ensured that it entered into clinical trials.

CERE-120 was tested in an open-label phase I clinical trial (Ref. Reference Marks115) and then in a double-blind, randomised, controlled phase II trial (Ref. Reference Marks116). The initial trial involved bilateral intraputaminal injection of one of two doses of CERE-120 to 12 patients suffering from advanced PD, with clinical follow-up for 1 year (Ref. Reference Marks115). No serious adverse effects were reported; transient anti-AAV2 antibodies were produced but caused no symptoms (Ref. Reference Marks115). The patients’ motor symptoms and quality-of-life indices showed variable improvement levels, with the main benefit being reductions in their off-medication UPDRS scores, in the time spent in ‘off’ and in dyskinesias (Ref. Reference Marks115). Intraputaminal CERE-120 was thus deemed to be a safe and potentially effective treatment for PD, and a phase II study was initiated by Ceregene. The phase II trial involved 58 patients; one-third of these was designated to the control group and received sham surgery (Ref. Reference Marks116). As in the phase I trial, some patients produced transient AAV2 antibodies, but none produced antibodies to NRTN (Ref. Reference Marks116). Thirteen (out of 38) of the CERE-120-treated patients reported serious side-effects, but these were attributed to the surgical procedure rather than to the treatment, since they were also experienced by four (out of 20) of the control group (Ref. Reference Marks116). The study also reported tumour formation in three of the CERE-120-treated patients, and in two control patients, which was not attributed to the surgical procedure (Ref. Reference Marks116). The CERE-120-treated patients did not experience significant improvements in their UPDRS scores at 12 months after their initial assessment, compared with those of the control group. Thus, the phase II trial did not meet its primary endpoint and was at that point deemed to be a failure (Ref. Reference Marks116). However, additional data presented from this study showed that the CERE-120-treated patients did show significant improvements in off-medication UPDRS score when assessed after 18 months, suggesting a delayed neurotrophic effect (Ref. Reference Marks116).

Post-mortem analysis on two patients who had received CERE-120 (and died of unrelated causes at 1.5 and 3 months after the trial) highlighted important differences in the distribution of AAV-2 mediated NRTN delivery to the human and animal brains (Refs Reference Bartus109, Reference Marks116). Bartus et al. examined the bioactivity and distribution of CERE-120 after intraputaminal delivery in young, aged and MPTP-treated rhesus monkeys, and compared these with the patterns found in the two post-mortem brains from the clinical trial (Ref. Reference Bartus109). They reported that in the PD patients, intraputaminal CERE-120 resulted in NRTN expression in ~15% of the putamen, but no expression in the substantia nigra. This is in contrast to their studies on the monkey brain, in which nigral expression of NRTN was seen after delivery solely to the putamen (Ref. Reference Bartus109). Any potential retrograde transport of NTN is hampered by its low level of diffusion and secretion, in contrast to GDNF, which is readily secreted and diffuses easily in brain parenchyma. It is likely that the restricted extent of putaminal transduction that was achieved in the Phase 2 trial, in combination with the limited diffusion of NTN protein, prevented any significant retrograde transport to the nigra. The lack of retrograde transport may be the reason for the absence of clinical benefit after intraputaminal delivery of CERE-120 in advanced-stage patients (Ref. Reference Marks116). This highlights the importance of early diagnosis of PD to enable treatment while the nigrostriatal pathway remains relatively intact; currently, CERE-120 delivery solely to the putamen may be of limited therapeutic use. Furthermore, the fact that lack of retrograde transport to the nigra after intrastriatal injection to the human PD brain was unexpected on the basis of the pre-clinical data demonstrates an important limitation of current animal models of PD in terms of their applicability to the clinical situation.

The deterioration of dopaminergic neuronal terminals in the caudate-putamen, and the consequential loss of dopaminergic input to this region, supports the concept of targeting the striatum with PD therapeutics. Furthermore, this area is considered a more easily accessible surgical target than the substantia nigra pars compacta (SNpc). However, the Ceregene group has queried the standard approach of solely targeting putaminal dopaminergic terminals, in order to treat degeneration of the nigrostriatal pathway. Having observed poor distribution of CERE-120 after striatal delivery in patients compared with nonhuman primates (Ref. Reference Bartus109), they examined the safety and practicality of targeting the SNpc (Ref. Reference Bartus106). This study showed that intranigral delivery of CERE-120 was safe and well-tolerated in a rat PD model, and extrapolated appropriate nigral dosages of CERE-120 for future clinical studies. This group further found greater neuroprotection and a larger distribution area of NRTN protein in 6-OHDA-lesioned rats after intranigral than after striatal injection of CERE-120 (Ref. Reference Herzog108). They concluded that targeting the nigra, in addition to the caudate-putamen, thus conferring simultaneous neurotrophic effects on both nigral dopaminergic cell bodies and their striatal terminals, would be more efficacious than striatal delivery alone (Ref. Reference Herzog108); they have adopted this strategy in their next clinical trial (Ref. Reference Bartus117). However, the targeting of the SNpc cannot be approached without caution. Three previous studies had reported significant weight loss in animals that had received a GDNF-expressing AAV2 vector into the SNpc (Refs Reference Su92, Reference Manfredsson96, Reference Manfredsson97). Thus, Bartus et al. compared an AAV2-GDNF reference dose against the range of CERE-120 doses being tested (Ref. Reference Bartus106). Weight loss was observed both in animals that received the AAV2-GDNF vector and those that had received the highest CERE-120 dose (Ref. Reference Bartus106). However, the authors attributed this weight loss to vector expression in mistargeted brain regions (Ref. Reference Bartus106). Such nonspecific targeting must be kept minimal, if NTF treatments are to have a significant therapeutic benefit to risk ratio.

Ceregene's most recent open-label trial assessed the safety and efficacy of simultaneously targeting the SNpc and striatum. Patients received a total of four nigral doses and six putaminal doses of CERE-120 and were followed clinically for 2 years (Ref. Reference Bartus117). The intraputaminal dose was fourfold greater than previous trials, on the basis of safety data from the two preceding clinical trials in conjunction with dose-escalation studies in animals. Furthermore, the number of bolus injections was decreased from eight in the initial trial to three, and the infusion rate was increased. Analysis of data from the open-label, dose-escalation safety arm of this study in six patients reported no incidences of medical, immunological or psychiatric complications, for up to 24 months (Ref. Reference Bartus117). Due to the success of this safety trial, 51 patients were involved in the placebo-controlled double-blind phase IIb part of this study; half of these underwent sham surgery (Ref. Reference Bartus118). Although some of the key secondary end-point measures did show a significant effect of treatment, this trial did not meet the predetermined end-point of significant improvements in motor-off scores, compared with the sham surgery group (for review see Ref. Reference Bartus, Weinberg and Samulski119). Also, significant motor-off benefits were observed in patients that had been diagnosed within 5 years prior to surgery, compared with those who were diagnosed at least 10 years prior (for review see Ref. Reference Bartus, Weinberg and Samulski119). This finding highlights the necessity for earlier intervention approaches of NTF therapy in progressive neurological diseases such as PD, if significant clinical benefits are to be achieved.

CDNF

Two in vivo studies have so far examined the neurotrophic effects of CDNF via AAV2-mediated delivery in rats (Refs Reference Bäck54, Reference Ren55). AAV2-CDNF conferred beneficial effects upon the nigrostriatal pathway to a certain extent in both studies (Refs Reference Bäck54, Reference Ren55). Bäck et al. compared the ability of AAV2-CDNF to protect dopaminergic neurons with that of AAV2-GDNF (Ref. Reference Bäck54). Intrastriatal injection of AAV2-CDNF in rats protected nigral dopamine neurons, and improved motor function, when administered 2 weeks before intrastriatal 6-OHDA, to a comparable extent to those of AAV2-GDNF (Ref. Reference Bäck54). Rats which received AAV2-CDNF did not display sprouting of dopaminergic neuronal terminals, in contrast to those that had received AAV2-GDNF. This is probably due to different distribution patterns of the proteins, as GDNF immunoreactivity was widespread, whereas CDNF was detected only within cells (Ref. Reference Bäck54). Intraneuronal confinement of CDNF protein following AAV-mediated delivery is probably the reason for the lack of neuronal sprouting, since such sprouting was found following infusion of CDNF protein into the striatal parenchyma (Ref. Reference Voutilainen53). This restricted expression of CDNF protein may explain the neuroprotective effects of AAV-CDNF being localised to the central part of the nigra, in contrast to the dopaminergic neuronal protection throughout the nigra that was conferred by AAV-GDNF (Ref. Reference Bäck54). Another study reported significant improvements in motor behaviour and rescue of nigral dopaminergic neurons and their striatal terminals, in rats that had received intrastriatal injection of AAV2-CDNF 6 weeks after intrastriatal 6-OHDA (Ref. Reference Ren55). Some animals from that study were tested after 1 year and displayed long-term motor improvements (Ref. Reference Ren55). Both of these studies indicate that the AAV2 system is a good candidate to test the delivery and effects of CDNF in other models of PD. A recent paper showed that combined administration of LV-CDNF and LV-MANF into the nigra, but not into the striatum, had significant protective effects on DA neuronal survival, and on amphetamine-induced rotational behaviour, in the 6-OHDA rat model of PD (Ref. Reference Cordero-Llana120). Furthermore, this study found that joint nigral overexpression of both factors had synergistic neuroprotective effects, suggesting that combined delivery of neurotrophic factors is a good avenue of exploration in the quest for an optimal neuroprotective therapy for PD.

GDF5

With regards to virus-mediated GDF5 delivery, research has been directed mainly at tendon repair, due to GDF5's properties as a bone morphogenetic protein. Two studies reported therapeutic improvements after AAV-mediated GDF5 delivery in rodent models of tendon injury (Refs Reference Rickert121, Reference Basile122), proposing a nonsurgical approach for treatment of tendon injuries. Advances towards the use of viral vector-mediated GDF5 delivery to the brain for PD were made in two studies on 6-OHDA-lesioned rats (Refs Reference Costello58, Reference O'Sullivan, Harrison and Sullivan123). The first transplanted GDF5-overexpressing embryonic rat dopaminergic neurons into the striatum of rats with striatal 6-OHDA lesions, and reported significant improvements in motor function in these rats, over those which had received control transplants (Ref. Reference O'Sullivan, Harrison and Sullivan123). This study provided proof-of-principle that GDF5 overexpression conferred therapeutic benefits, without any adverse effects, in a PD model. The second study reported improvements in motor function and increases in survival of transplanted dopaminergic neurons, following co-transplantation with a GDF5-overexpressing cell line, in rats with 6-OHDA lesions of the MFB or striatum (Ref. Reference Costello58). Since viral vector-mediated GDF5 delivery has already been validated outside the nervous system, and since GDF5 overexpression by transplanted cells has shown efficacy in rat models of PD, it is timely for investigations of viral vector-driven GDF5 in PD models. We are currently investigating the efficacy of AAV- and LV-mediated delivery of GDF5 in 6-OHDA and AAV-α-syn rat models of PD. (For a summary of the current start-of-the-art and stage of development of viral-mediated NTFs for PD, see Fig. 3.)

Figure 3 Summary of the stages of development of the various NTFs under development in viral-vector-mediated therapies for PD.

Strategies for optimising NTF delivery in PD

Careful consideration must be given to the methodologies used to deliver optimised therapeutic agents in PD patients. The ideal delivery should be safe, efficient accurate and minimally invasive. Since NTFs do not cross the blood–brain barrier, this barrier must be bypassed via direct injection or infusion into the brain (for reviews see Refs Reference Zhu124, Reference White66). This intracranial approach generally induces some trauma to the brain and has the risk of introducing infections. Alternative NTF delivery methods are being explored. For example, intranasal delivery, which has worked with other recombinant viral vectors (for review see Ref. Reference Lochhead and Thorne125), has been tested for GDF5 protein delivery to the brain (Ref. Reference Hanson126). Although the majority of the administered dose did not diffuse further than the olfactory epithelium, the nanomolar amount of GDF5 that travelled to the midbrain was deemed to be a potentially therapeutic concentration (Ref. Reference Hanson126). Future studies may support the use of intranasal delivery for viral vectors delivering NTFs. Any such strategies must take into consideration the anatomical differences between the rodent and human nasal cavity. The olfactory epithelium covers a much larger proportion of the nasal cavities in rodents than in primates (Ref. Reference Harkema, Carey and Wagner127), and thus it would be critical to test this route of delivery in nonhuman primates to determine the viability of this approach. Other propositions include the use of capsules designed through nanotechnology, thereby removing the need for viruses, or compression of vectors into nanoparticles to enable nuclear entry via nuclear pores (for review see Ref. Reference Pervaiz and Irshad128). It will be some time before the first nanotechnologically-delivered NTF clinical trial is conducted, and the long-term effects of using these nanomaterials can be assessed.

A final point on target selection is the consideration of whether intraputaminal vector treatments, such as AAV2-GDNF and CERE-120, are transported from the striatum to the substantia nigra in an anterograde or retrograde manner. Bartus et al. attributed the lack of CERE-120 transport to SNpc as failure of retrograde transport efficacy in the degenerating neurons (Ref. Reference Bartus109). It was on this basis that studies involving CERE-120 infusions into both striatum and substantia nigra were initiated (for review see Ref. Reference Bartus113). However, Bankiewicz and co-workers showed that nigral cell bodies can still receive intraputaminal AAV2-GDNF via anterograde transport by nondopaminergic neurons of the striatonigral pathway (for review Ref. Reference Kells, Forsayeth and Bankiewicz129). Their rationale is that targeting the nigra with NTFs carries considerable health and stereotactic risks, and that the intraputaminal CED approach alone is enough to therapeutically support the nigrostriatal neurons (Ref. Reference Su92). One risk with efficient transfer of NTF protein by anterograde trafficking is that areas additional to the target area may receive the therapeutic protein, which may cause off-target side-effects. If the upcoming AAV2-GDNF clinical trial yields more efficacious results than the CERE-120 clinical trial (Ref. Reference Bartus118), then it may be worth investigating whether CERE-120 could be more effectively delivered via intraputaminal CED. This could then be similarly applied to future viral vector-mediated delivery of other NTFs, such as CDNF and GDF5.

Conclusions

Gene therapy has contributed considerable advancements to the ongoing development of neuroprotective therapeutics for PD. Initial clinical trials involved administering GDNF protein, with mixed results (Refs Reference Nutt37, Reference Gill38, Reference Patel39, Reference Slevin40, Reference Lang41, Reference Tatarewicz42). The appearance of GDNF antibodies in some patients (Refs Reference Lang41, Reference Tatarewicz42) highlighted a need to approach NTF therapy differently. This led to testing of viral vector-mediated delivery of NTFs, with AAV2 vectors seemingly the most appropriate in the clinical scenario. Research on viral vector delivery of GDNF, and its optimisation via the use of CED, has led to the development of a protocol which will be implemented in an upcoming GDNF clinical trial (Ref. Reference Richardson91). Only minor benefits have been observed following viral vector-mediated NRTN delivery (Refs Reference Marks115, Reference Marks116, Reference Bartus117, Reference Bartus118), but trials incorporating optimised methodologies are ongoing. Regarding other dopaminergic NTFs, viral vector-delivered CDNF has been tested in rodents (Refs Reference Bäck54, Reference Ren55) and GDF5 has been delivered using cell-based methods in rodents. It will be some time before these two NTFs are tested as potential PD therapeutics at the clinical level.

In summary, NTFs can play an integral part in the search for a viable treatment approach to PD. Current treatment options can only manage PD symptoms for a limited period of time. At the very minimum, NTFs could delay the onset of symptoms and slow disease progression. Since NTF proteins have a limited half-life in vivo and it is impractical to administer several applications of a NTF indefinitely, gene therapy presents huge potential for long-term and targeted treatment of PD.

Acknowledgements

The authors declare no conflict of interest.

References

1 Gordon, K.J. and Blobe, G.C. (2008) Role of transforming growth factor-beta superfamily signaling pathways in human disease. Biochimica et Biophysica Acta (BBA) – Reviews on Cancer 1782, 197-228 CrossRefGoogle ScholarPubMed
2 Saarma, M. (2000) GDNF – a stranger in the TGF-beta superfamily? European Journal of Biochemistry 267, 6968-6971 CrossRefGoogle ScholarPubMed
3 Sariola, H. and Saarma, M. (2003) Novel functions and signalling pathways for GDNF. Journal of Cell Science 116, 3855-3862 Google Scholar
4 Sullivan, A.M. and Toulouse, A. (2011) Neurotrophic factors for the treatment of Parkinson's disease. Cytokine & Growth Factor Reviews 22, 157-165 Google Scholar
5 Lindholm, P. and Saarma, M. (2010) Novel CDNF/MANF family of neurotrophic factors. Developmental Neurobiology 70, 360-371 CrossRefGoogle ScholarPubMed
6 Hegarty, S.V., O'Keeffe, G.W. and Sullivan, A.M. (2013) BMP-Smad 1/5/8 signalling in the development of the nervous system. Progress in Neurobiology 109, 28-41 CrossRefGoogle ScholarPubMed
7 Nishitoh, H. et al. (1996) Identification of type I and type II serine/threonine kinase receptors for growth/differentiation factor-5. Journal of Biological Chemistry 271, 21345-21352 Google Scholar
8 Toulouse, A. and Sullivan, A.M. (2008) Progress in Parkinson's disease-where do we stand? Progress in Neurobiology 85, 376-392 CrossRefGoogle ScholarPubMed
9 Yokochi, M. (2009) Reevaluation of levodopa therapy for the treatment of advanced Parkinson's disease. Parkinsonism & Related Disorders 15(Suppl 1), S25-S30 CrossRefGoogle ScholarPubMed
10 Beric, A. et al. (2001) Complications of deep brain stimulation surgery. Stereotactic and Functional Neurosurgery 77, 73-78 Google Scholar
11 Samii, A., Nutt, J.G. and Ransom, B.R. (2004) Parkinson's disease. Lancet 363, 1783-1793 Google Scholar
12 Krieglstein, K. et al. (1995) TGF-beta superfamily members promote survival of midbrain dopaminergic neurons and protect them against MPP+ toxicity. EMBO Journal 14, 736-742 CrossRefGoogle ScholarPubMed
13 Lin, L.F. et al. (1993) GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 260, 1130-1132 CrossRefGoogle ScholarPubMed
14 Widmer, H.R. et al. (2000) Glial cell line-derived neurotrophic factor stimulates the morphological differentiation of cultured ventral mesencephalic calbindin- and calretinin-expressing neurons. Experimental Neurology 164, 71-81 CrossRefGoogle ScholarPubMed
15 Hou, J.G., Lin, L.F. and Mytilineou, C. (1996) Glial cell line-derived neurotrophic factor exerts neurotrophic effects on dopaminergic neurons in vitro and promotes their survival and regrowth after damage by 1-methyl-4-phenylpyridinium. Journal of Neurochemistry 66, 74-82 CrossRefGoogle ScholarPubMed
16 Eggert, K. et al. (1999) Glial cell line-derived neurotrophic factor protects dopaminergic neurons from 6-hydroxydopamine toxicity in vitro. Neuroscience Letters 269, 178-182 CrossRefGoogle ScholarPubMed
17 Bowenkamp, K.E. et al. (1995) Glial cell line-derived neurotrophic factor supports survival of injured midbrain dopaminergic neurons. Journal of Comparative Neurology 355, 479-489 CrossRefGoogle ScholarPubMed
18 Hoffer, B.J. et al. (1994) Glial cell line-derived neurotrophic factor reverses toxin-induced injury to midbrain dopaminergic neurons in vivo. Neuroscience Letters 182, 107-111 CrossRefGoogle ScholarPubMed
19 Kearns, C.M. and Gash, D.M. (1995) GDNF protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Research 672, 104-111 Google Scholar
20 Sauer, H., Rosenblad, C. and Bjorklund, A. (1995) Glial cell line-derived neurotrophic factor but not transforming growth factor beta 3 prevents delayed degeneration of nigral dopaminergic neurons following striatal 6-hydroxydopamine lesion. Proceedings of the National Academy of Sciences of the United States of America 92, 8935-8939 Google Scholar
21 Shults, C.W., Kimber, T. and Martin, D. (1996) Intrastriatal injection of GDNF attenuates the effects of 6-hydroxydopamine. Neuroreport 7, 627-631 Google Scholar
22 Sullivan, A.M., Opacka-Juffry, J. and Blunt, S.B. (1998) Long-term protection of the rat nigrostriatal dopaminergic system by glial cell line-derived neurotrophic factor against 6-hydroxydopamine in vivo. European Journal of Neuroscience 10, 57-63 Google Scholar
23 Bjorklund, A. et al. (2000) Towards a neuroprotective gene therapy for Parkinson's disease: use of adenovirus, AAV and lentivirus vectors for gene transfer of GDNF to the nigrostriatal system in the rat Parkinson model. Brain Research 886, 82-98 Google Scholar
24 Tomac, A. et al. (1995) Protection and repair of the nigrostriatal dopaminergic system by GDNF in vivo. Nature 373, 335-339 Google Scholar
25 Rosenblad, C., Martinez-Serrano, A. and Bjorklund, A. (1998) Intrastriatal glial cell line-derived neurotrophic factor promotes sprouting of spared nigrostriatal dopaminergic afferents and induces recovery of function in a rat model of Parkinson's disease. Neuroscience 82, 129-137 Google Scholar
26 Aoi, M. et al. (2000) The effect of intrastriatal single injection of GDNF on the nigrostriatal dopaminergic system in hemiparkinsonian rats: behavioral and histological studies using two different dosages. Neuroscience Research 36, 319-325 Google Scholar
27 Granholm, A.C. et al. (1997) Glial cell line-derived neurotrophic factor improves survival of ventral mesencephalic grafts to the 6-hydroxydopamine lesioned striatum. Experimental Brain Research 116, 29-38 CrossRefGoogle Scholar
28 Apostolides, C. et al. (1998) Glial cell line-derived neurotrophic factor improves intrastriatal graft survival of stored dopaminergic cells. Neuroscience 83, 363-372 CrossRefGoogle ScholarPubMed
29 Sullivan, A.M., Pohl, J. and Blunt, S.B. (1998) Growth/differentiation factor 5 and glial cell line-derived neurotrophic factor enhance survival and function of dopaminergic grafts in a rat model of Parkinson's disease. European Journal of Neuroscience 10, 3681-3688 Google Scholar
30 Yurek, D.M. (1998) Glial cell line-derived neurotrophic factor improves survival of dopaminergic neurons in transplants of fetal ventral mesencephalic tissue. Experimental Neurology 153, 195-202 Google Scholar
31 Gash, D.M. et al. (1996) Functional recovery in parkinsonian monkeys treated with GDNF. Nature 380, 252-255 Google Scholar
32 Gerhardt, G.A. et al. (1999) GDNF improves dopamine function in the substantia nigra but not the putamen of unilateral MPTP-lesioned rhesus monkeys. Brain Research 817, 163-171 CrossRefGoogle Scholar
33 Grondin, R. et al. (2002) Chronic, controlled GDNF infusion promotes structural and functional recovery in advanced parkinsonian monkeys. Brain 125, 2191-2201 CrossRefGoogle ScholarPubMed
34 Iravani, M.M. et al. (2001) GDNF reverses priming for dyskinesia in MPTP-treated, L-DOPA-primed common marmosets. European Journal of Neuroscience 13, 597-608 CrossRefGoogle ScholarPubMed
35 Maswood, N. et al. (2002) Effects of chronic intraputamenal infusion of glial cell line-derived neurotrophic factor (GDNF) in aged Rhesus monkeys. Neurobiology of Aging 23, 881-889 CrossRefGoogle ScholarPubMed
36 Miyoshi, Y. et al. (1997) Glial cell line-derived neurotrophic factor-levodopa interactions and reduction of side effects in parkinsonian monkeys. Annals of Neurology 42, 208-214 Google Scholar
37 Nutt, J.G. et al. (2003) Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 60, 69-73 Google Scholar
38 Gill, S.S. et al. (2003) Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nature Medicine 9, 589-595 CrossRefGoogle ScholarPubMed
39 Patel, N.K. et al. (2005) Intraputamenal infusion of glial cell line-derived neurotrophic factor in PD: a two-year outcome study. Annals of Neurology 57, 298-302 CrossRefGoogle ScholarPubMed
40 Slevin, J.T. et al. (2005) Improvement of bilateral motor functions in patients with Parkinson disease through the unilateral intraputaminal infusion of glial cell line-derived neurotrophic factor. Journal of Neurosurgery 102, 216-222 CrossRefGoogle ScholarPubMed
41 Lang, A.E. et al. (2006) Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Annals of Neurology 59, 459-466 Google Scholar
42 Tatarewicz, S.M. et al. (2007) Development of a maturing T-cell-mediated immune response in patients with idiopathic Parkinson's disease receiving r-metHuGDNF via continuous intraputaminal infusion. Journal of Clinical Immunology 27, 620-627 Google Scholar
43 Kotzbauer, P.T. et al. (1996) Neurturin, a relative of glial-cell-line-derived neurotrophic factor. Nature 384, 467-470 CrossRefGoogle ScholarPubMed
44 Horger, B.A. et al. (1998) Neurturin exerts potent actions on survival and function of midbrain dopaminergic neurons. Journal of Neuroscience 18, 4929-4937 CrossRefGoogle ScholarPubMed
45 Akerud, P. et al. (1999) Differential effects of glial cell line-derived neurotrophic factor and neurturin on developing and adult substantia nigra dopaminergic neurons. Journal of Neurochemistry 73, 70-78 CrossRefGoogle ScholarPubMed
46 Rosenblad, C. et al. (1999) Protection and regeneration of nigral dopaminergic neurons by neurturin or GDNF in a partial lesion model of Parkinson's disease after administration into the striatum or the lateral ventricle. European Journal of Neuroscience 11, 1554-1566 CrossRefGoogle ScholarPubMed
47 Oiwa, Y. et al. (2002) Dopaminergic neuroprotection and regeneration by neurturin assessed by using behavioral, biochemical and histochemical measurements in a model of progressive Parkinson's disease. Brain Research 947, 271-283 CrossRefGoogle Scholar
48 Grondin, R. et al. (2008) Intraputamenal infusion of exogenous neurturin protein restores motor and dopaminergic function in the globus pallidus of MPTP-lesioned rhesus monkeys. Cell Transplantation 17, 373-381 Google Scholar
49 Petrova, P. et al. (2003) MANF: a new mesencephalic, astrocyte-derived neurotrophic factor with selectivity for dopaminergic neurons. Journal of Molecular Neuroscience 20, 173-188 Google Scholar
50 Lindholm, P. et al. (2007) Novel neurotrophic factor CDNF protects and rescues midbrain dopamine neurons in vivo. Nature 448, 73-77 CrossRefGoogle ScholarPubMed
51 Parkash, V. et al. (2009) The structure of the conserved neurotrophic factors MANF and CDNF explains why they are bifunctional. Protein Engineering Design and Selection 22, 233-241 Google Scholar
52 Airavaara, M. et al. (2012) CDNF protects the nigrostriatal dopamine system and promotes recovery after MPTP treatment in mice. Cell Transplantation 21, 1213-1223 Google Scholar
53 Voutilainen, M.H. et al. (2011) Chronic infusion of CDNF prevents 6-OHDA-induced deficits in a rat model of Parkinson's disease. Experimental Neurology 228, 99-108 Google Scholar
54 Bäck, S. et al. (2013) Gene therapy with AAV2-CDNF provides functional benefits in a rat model of Parkinson's disease. Brain and Behaviour 3, 75-88 Google Scholar
55 Ren, X. et al. (2013) AAV2-mediated striatum delivery of human CDNF prevents the deterioration of midbrain dopamine neurons in a 6-hydroxydopamine induced parkinsonian rat model. Experimental Neurology 248, 148-156 Google Scholar
56 Krieglstein, K. et al. (1995) Trophic and protective effects of growth/differentiation factor 5, a member of the transforming growth factor-beta superfamily, on midbrain dopaminergic neurons. Journal of Neuroscience Research 42, 724-732 Google Scholar
57 O'Keeffe, G.W., Dockery, P. and Sullivan, A.M. (2004) Effects of growth/differentiation factor 5 on the survival and morphology of embryonic rat midbrain dopaminergic neurones in vitro. Journal of Neurocytology 33, 479-488 Google Scholar
58 Costello, D.J. et al. (2012) Transplantation of novel human GDF5-expressing CHO cells is neuroprotective in models of Parkinson's disease. Journal of Cellular and Molecular Medicine 16, 2451-2460 CrossRefGoogle ScholarPubMed
59 Hegarty, S.V., Sullivan, A.M. and O'Keeffe, G.W. (2013) BMP2 and GDF5 induce neuronal differentiation through a Smad dependant pathway in a model of human midbrain dopaminergic neurons. Molecular and Cellular Neuroscience 56, 263-271 Google Scholar
60 Hegarty, S.V. et al. (2014) Canonical BMP-Smad signalling promotes neurite growth in rat midbrain dopaminergic neurons. Neuromolecular Medicine 16, 473-489 Google Scholar
61 Sullivan, A.M. et al. (1997) Growth/differentiation factor 5 protects nigrostriatal dopaminergic neurones in a rat model of Parkinson's disease. Neuroscience Letters 233, 73-76 Google Scholar
62 Sullivan, A.M. et al. (1999) Neuroprotective effects of growth/differentiation factor 5 depend on the site of administration. Brain Research 818, 176-179 Google Scholar
63 Hurley, F.M., Costello, D.J. and Sullivan, A.M. (2004) Neuroprotective effects of delayed administration of growth/differentiation factor-5 in the partial lesion model of Parkinson's disease. Experimental Neurology 185, 281-289 Google Scholar
64 LeWitt, P.A. et al. (2011) AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial. Lancet Neurology 10, 309-319 Google Scholar
65 Karra, D. and Dahm, R. (2010) Transfection techniques for neuronal cells. Journal of Neuroscience 30, 6171-6177 CrossRefGoogle ScholarPubMed
66 White, E. (2012) Molecular neurosurgery: vectors and vector delivery strategies. British Journal of Neurosurgery 26, 798-808 Google Scholar
67 Ginn, S.L. et al. (2013) Gene therapy clinical trials worldwide to 2012 – an update. Journal of Gene Medicine 15, 65-77 Google Scholar
68 Choi-Lundberg, D.L. et al. (1997) Dopaminergic neurons protected from degeneration by GDNF gene therapy. Science 275, 838-841 CrossRefGoogle ScholarPubMed
69 Mandel, R.J. et al. (1997) Midbrain injection of recombinant adeno-associated virus encoding rat glial cell line-derived neurotrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson's disease in rats. Proceedings of the National Academy of Sciences of the United States of America 94, 14083-14088 Google Scholar
70 Balemans, W. and Van Hul, W. (2002) Extracellular regulation of BMP signaling in vertebrates: a cocktail of modulators. Developmental Biology 250, 231-250 CrossRefGoogle ScholarPubMed
71 Bilang-Bleuel, A. et al. (1997) Intrastriatal injection of an adenoviral vector expressing glial-cell-line-derived neurotrophic factor prevents dopaminergic neuron degeneration and behavioral impairment in a rat model of Parkinson disease. Proceedings of the National Academy of Sciences of the United States of America 94, 8818-8823 Google Scholar
72 Choi-Lundberg, D.L. et al. (1998) Behavioral and cellular protection of rat dopaminergic neurons by an adenoviral vector encoding glial cell line-derived neurotrophic factor. Experimental Neurology 154, 261-275 CrossRefGoogle ScholarPubMed
73 Lapchak, P.A. et al. (1997) Adenoviral vector-mediated GDNF gene therapy in a rodent lesion model of late stage Parkinson's disease. Brain Research 777, 153-160 CrossRefGoogle Scholar
74 Kozlowski, D.A. et al. (2000) Delivery of a GDNF gene into the substantia nigra after a progressive 6-OHDA lesion maintains functional nigrostriatal connections. Experimental Neurology 166, 1-15 Google Scholar
75 Kojima, H. et al. (1997) Adenovirus-mediated transduction with human glial cell line-derived neurotrophic factor gene prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopamine depletion in striatum of mouse brain. Biochemical and Biophysical Research Communications 238, 569-573 CrossRefGoogle ScholarPubMed
76 Kirik, D. et al. (2000) Long-term rAAV-mediated gene transfer of GDNF in the rat Parkinson's model: intrastriatal but not intranigral transduction promotes functional regeneration in the lesioned nigrostriatal system. Journal of Neuroscience 20, 4686-4700 Google Scholar
77 Eslamboli, A. et al. (2003) Recombinant adeno-associated viral vector (rAAV) delivery of GDNF provides protection against 6-OHDA lesion in the common marmoset monkey (Callithrix jacchus). Experimental Neurology 184, 536-548 Google Scholar
78 Bensadoun, J.C. et al. (2000) Lentiviral vectors as a gene delivery system in the mouse midbrain: cellular and behavioral improvements in a 6-OHDA model of Parkinson's disease using GDNF. Experimental Neurology 164, 15-24 Google Scholar
79 Georgievska, B. et al. (2002) Neuroprotection in the rat Parkinson model by intrastriatal GDNF gene transfer using a lentiviral vector. Neuroreport 13, 75-82 Google Scholar
80 Kordower, J.H. et al. (2000) Neurodegeneration prevented by lentiviral vector delivery of GDNF in primate models of Parkinson's disease. Science 290, 767-773 Google Scholar
81 Dowd, E. et al. (2005) Lentivector-mediated delivery of GDNF protects complex motor functions relevant to human Parkinsonism in a rat lesion model. European Journal of Neuroscience 22, 2587-2595 Google Scholar
82 Brizard, M. et al. (2006) Functional reinnervation from remaining DA terminals induced by GDNF lentivirus in a rat model of early Parkinson's disease. Neurobiology of Disease 21, 90-101 Google Scholar
83 Decressac, M. et al. (2011) GDNF fails to exert neuroprotection in a rat alpha-synuclein model of Parkinson's disease. Brain 134, 2302-2311 Google Scholar
84 Bekris, L.M., Mata, I.F. and Zabetian, C.P. (2010) The genetics of Parkinson disease. Journal of Geriatric Psychiatry and Neurology 23, 228-242 Google Scholar
85 Deng, H. and Yuan, L. (2014) Genetic variants and animal models in SNCA and Parkinson disease. Ageing Research Reviews 15C, 161-176 Google Scholar
86 Ulusoy, A. et al. (2010) Viral vector-mediated overexpression of alpha-synuclein as a progressive model of Parkinson's disease. Progress in Brain Research 184, 89-111 CrossRefGoogle ScholarPubMed
87 Decressac, M. et al. (2012) alpha-Synuclein-induced down-regulation of Nurr1 disrupts GDNF signaling in nigral dopamine neurons. Science Translational Medicine 4, 163ra156 Google Scholar
88 Eberling, J.L. et al. (2009) Functional effects of AAV2-GDNF on the dopaminergic nigrostriatal pathway in parkinsonian rhesus monkeys. Human Gene Therapy 20, 511-518 Google Scholar
89 Johnston, L.C. et al. (2009) Clinically relevant effects of convection-enhanced delivery of AAV2-GDNF on the dopaminergic nigrostriatal pathway in aged rhesus monkeys. Human Gene Therapy 20, 497-510 CrossRefGoogle ScholarPubMed
90 Kells, A.P. et al. (2010) Regeneration of the MPTP-lesioned dopaminergic system after convection-enhanced delivery of AAV2-GDNF. Journal of Neuroscience 30, 9567-9577 Google Scholar
91 Richardson, R.M. et al. (2011) Interventional MRI-guided putaminal delivery of AAV2-GDNF for a planned clinical trial in Parkinson's disease. Molecular Therapy 19, 1048-1057 Google Scholar
92 Su, X. et al. (2009) Safety evaluation of AAV2-GDNF gene transfer into the dopaminergic nigrostriatal pathway in aged and parkinsonian rhesus monkeys. Human Gene Therapy 20, 1627-1640 Google Scholar
93 Su, X. et al. (2010) Real-time MR imaging with Gadoteridol predicts distribution of transgenes after convection-enhanced delivery of AAV2 vectors. Molecular Therapy 18, 1490-1495 Google Scholar
94 Salegio, E.A. et al. (2012) Guided delivery of adeno-associated viral vectors into the primate brain. Advanced Drug Delivery Reviews 64, 598-604 Google Scholar
95 Bobo, R.H. et al. (1994) Convection-enhanced delivery of macromolecules in the brain. Proceedings of the National Academy of Sciences of the United States of America 91, 2076-2080 CrossRefGoogle ScholarPubMed
96 Manfredsson, F.P. et al. (2009) Tight Long-term dynamic doxycycline responsive nigrostriatal GDNF using a single rAAV vector. Molecular Therapy 17, 1857-1867 Google Scholar
97 Manfredsson, F.P. et al. (2009) Nigrostriatal rAAV-mediated GDNF overexpression induces robust weight loss in a rat model of age-related obesity. Molecular Therapy 17, 980-991 Google Scholar
98 Gimenez, F. et al. (2011) Image-guided convection-enhanced delivery of GDNF protein into monkey putamen. NeuroImage 54(Suppl 1), S189-S195 Google Scholar
99 Richardson, R.M. et al. (2011) Novel platform for MRI-guided convection-enhanced delivery of therapeutics: preclinical validation in nonhuman primate brain. Stereotactic and Functional Neurosurgery 89, 141-151 Google Scholar
100 Krauze, M.T. et al. (2005) Reflux-free cannula for convection-enhanced high-speed delivery of therapeutic agents. Journal of Neurosurgery 103, 923-929 Google Scholar
101 Sanftner, L.M. et al. (2005) AAV2-mediated gene delivery to monkey putamen: evaluation of an infusion device and delivery parameters. Experimental Neurology 194, 476-483 Google Scholar
102 Yin, D., Forsayeth, J. and Bankiewicz, K.S. (2010) Optimized cannula design and placement for convection-enhanced delivery in rat striatum. Journal of Neuroscience Methods 187, 46-51 Google Scholar
103 Yin, D. et al. (2011) Optimal region of the putamen for image-guided convection-enhanced delivery of therapeutics in human and non-human primates. NeuroImage 54(Suppl 1), S196-S203 Google Scholar
104 Hadaczek, P. et al. (2011) Evaluation of an AAV2-based rapamycin-regulated glial cell line-derived neurotrophic factor (GDNF) expression vector system. PloS ONE 6, e27728 Google Scholar
105 Fjord-Larsen, L. et al. (2005) Efficient in vivo protection of nigral dopaminergic neurons by lentiviral gene transfer of a modified Neurturin construct. Experimental Neurology 195, 49-60 Google Scholar
106 Bartus, R.T. et al. (2011) Properly scaled and targeted AAV2-NRTN (neurturin) to the substantia nigra is safe, effective and causes no weight loss: support for nigral targeting in Parkinson's disease. Neurobiology of Disease 44, 38-52 CrossRefGoogle Scholar
107 Gasmi, M. et al. (2007) AAV2-mediated delivery of human neurturin to the rat nigrostriatal system: long-term efficacy and tolerability of CERE-120 for Parkinson's disease. Neurobiology of Disease 27, 67-76 Google Scholar
108 Herzog, C.D. et al. (2013) Enhanced neurotrophic distribution, cell signaling and neuroprotection following substantia nigral versus striatal delivery of AAV2-NRTN (CERE-120). Neurobiology of Disease 58, 38-48 Google Scholar
109 Bartus, R.T. et al. (2011) Bioactivity of AAV2-neurturin gene therapy (CERE-120): differences between Parkinson's disease and nonhuman primate brains. Movement Disorders 26, 27-36 Google Scholar
110 Kordower, J.H. et al. (2006) Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Annals of Neurology 60, 706-715 Google Scholar
111 Herzog, C.D. et al. (2008) Transgene expression, bioactivity, and safety of CERE-120 (AAV2-neurturin) following delivery to the monkey striatum. Molecular Therapy 16, 1737-1744 Google Scholar
112 Herzog, C.D. et al. (2007) Striatal delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic nigrostriatal system in aged monkeys. Movement Disorders 22, 1124-1132 Google Scholar
113 Bartus, R.T. et al. (2013) Advancing neurotrophic factors as treatments for age-related neurodegenerative diseases: developing and demonstrating “clinical proof-of-concept” for AAV-neurturin (CERE-120) in Parkinson's disease. Neurobiology of Aging 34, 35-61 Google Scholar
114 Georgievska, B., Kirik, D. and Bjorklund, A. (2004) Overexpression of glial cell line-derived neurotrophic factor using a lentiviral vector induces time- and dose-dependent downregulation of tyrosine hydroxylase in the intact nigrostriatal dopamine system. Journal of Neuroscience 24, 6437-6445 Google Scholar
115 Marks, W.J. Jr et al. (2008) Safety and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-neurturin) to patients with idiopathic Parkinson's disease: an open-label, phase I trial. Lancet Neurology 7, 400-408 Google Scholar
116 Marks, W.J. Jr et al. (2010) Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurology 9, 1164-1172 Google Scholar
117 Bartus, R.T. et al. (2013) Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients. Neurology 80, 1698-1701 Google Scholar
118 Bartus, R.T. (2013) CERE-120 (AAV2-NRTN) for the treatment of Parkinson's disease: experience from 4 clinical trials and human autopsy data. Proceedings of the 16th Annual Meeting of the American Society for Gene and Cell Therapy, Salt Lake City, USA.Google Scholar
119 Bartus, R.T., Weinberg, M.S. and Samulski, R.J. (2014) Parkinson's disease gene therapy: success by design meets failure by efficacy. Molecular Therapy 22, 487-497 Google Scholar
120 Cordero-Llana, Ó et al. (2015) Enhanced efficacy of the CDNF/MANF family by combined intranigral overexpression in the 6-OHDA rat model of Parkinson's disease. Molecular Therapy 23, 244-254 Google Scholar
121 Rickert, M. et al. (2005) Adenovirus-mediated gene transfer of growth and differentiation factor-5 into tenocytes and the healing rat Achilles tendon. Connective Tissue Research 46, 175-183 CrossRefGoogle ScholarPubMed
122 Basile, P. et al. (2008) Freeze-dried tendon allografts as tissue-engineering scaffolds for Gdf5 gene delivery. Molecular Therapy 16, 466-473 Google Scholar
123 O'Sullivan, D.B., Harrison, P.T. and Sullivan, A.M. (2010) Effects of GDF5 overexpression on embryonic rat dopaminergic neurones in vitro and in vivo. Journal of Neural Transmission 117, 559-572 Google Scholar
124 Zhu, J. et al. (2012) Intranasal administration: a potential solution for cross-BBB delivering neurotrophic factors. Histology and Histopathology 27, 537-548 Google Scholar
125 Lochhead, J.J. and Thorne, R.G. (2012) Intranasal delivery of biologics to the central nervous system. Advanced Drug Delivery Reviews 64, 614-628 Google Scholar
126 Hanson, L.R. et al. (2012) Intranasal delivery of growth differentiation factor 5 to the central nervous system. Drug Delivery 19, 149-154 Google Scholar
127 Harkema, J.R., Carey, S.A. and Wagner, J.G. (2006) The nose revisited: a brief review of the comparative structure, function, and toxicologic pathology of the nasal epithelium. Toxicologic Pathology 34, 252-269 Google Scholar
128 Pervaiz, S. and Irshad, S. (2012) Role of nanobiotechnology in Parkinson's disease. Pakistan Journal of Biochemistry and Molecular Biology 34, 35-43 Google Scholar
129 Kells, A.P., Forsayeth, J. and Bankiewicz, K.S. (2012) Glial-derived neurotrophic factor gene transfer for Parkinson's disease: anterograde distribution of AAV2 vectors in the primate brain. Neurobiology of Disease 48, 228-235 Google Scholar
Figure 0

Figure 1 Experimental strategy used to examine the efficacy of viral-mediated delivery of NTFs in preclinical models of PD. (a) PD results from a loss of dopaminergic neurons in the SN which leads to a reduction in striatal dopaminergic innervation. (b) Preclinical assessment of the neuroprotective efficacy of viral-mediated delivery systems involves stereotactic administration of the viral vector to the striatum or midbrain, prior to, at the same time, or after, stereotactic administration of a neurotoxin (e.g. 6-OHDA) to either the (SNpc), striatum (Str) or medial forebrain bundle (mfb). (c) Assessment of motor function after surgery, coupled with (d) post-mortem assessment of numbers of dopaminergic neurons and their striatal processes, is used to determine efficacy of viral-delivered NTFs.

Figure 1

Figure 2 Distinct features of AV, AAV and LV vectors. AdV, AAV and LV vectors have all been explored for the delivery of NTFs in PD models. Panels (a–c) show the events involved from in vitro generation to stereotactic administration of each viral vector type. Shown also is an overview of the distinct features of each viral vector when applied to the brain in vivo.

Figure 2

Figure 3 Summary of the stages of development of the various NTFs under development in viral-vector-mediated therapies for PD.