Skip to main content

OPINION article

Front. Aging Neurosci., 07 December 2020
Sec. Parkinson’s Disease and Aging-related Movement Disorders
This article is part of the Research Topic Regeneration and Brain Repair View all 11 articles

The Future of GDNF in Parkinson's Disease

  • 1Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, United States
  • 2The Michael J. Fox Foundation for Parkinson's Research, New York, NY, United States
  • 3Department of Neurology and Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
  • 4Department of Neurosurgery, Emory University, Atlanta, GA, United States
  • 5Virscio, Inc., New Haven, CT, United States
  • 6Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
  • 7Department of Neuroscience, University of Florida, Gainesville, FL, United States

Introduction

At the 2019 annual meeting for American Society of Neural Therapy and Repair (ASNTR) a special panel assembled to discuss the future of neurotrophic factor delivery in Parkinson's disease (PD), particularly those factors belonging to the Glial cell line-derived neurotrophic factor family of ligands (GFLs; GDNF and Neurturin). The panel consisted of representatives from academia, industry, and non-profit organizations with primary backgrounds in neurology or neurosurgery and the impetus for the assembly was data from the a recent GDNF clinical trial (Whone A. et al., 2019; Whone A. L. et al., 2019) that utilized an enhanced method of protein infusion to facilitate improved spread of GDNF. Despite preclinical success, this trial, as all previously published trials, failed to demonstrate clinical efficacy (Whone A. L. et al., 2019), leaving the field wondering if there is a future for these neurotrophic factors in PD (Kirkeby and Barker, 2019). This opinion piece will summarize the discussion and the overarching recommendations from the meeting.

Recent Results From GDNF Trials

Over the last few decades there have been numerous clinical trials utilizing central delivery of GDNF or neurturin via direct protein infusion or overexpression using viral vector-based gene therapy (Merola et al., 2010) with the latest trial reporting dosing of the first patient in September 2020 [Brain Neurotherapy Bio; adeno-associated virus (AAV)-GDNF]. Despite preliminary reports of efficacy in the open-label phase of trials, placebo-controlled studies have failed to replicate any favorable outcomes (e.g., Marks et al., 2008, 2010). Although the reasons behind these apparent failures are unknown, one of the issues may be lack of sufficient target engagement—either via poor diffusion in protein delivery trials (Salvatore et al., 2006) or poor transduction in viral vector trials (Bartus et al., 2011). To that end, the most recent trial rationalized that improving delivery with convection enhanced delivery (CED) might overcome the limitation of insufficient putamenal and nigral drug coverage and achieve improvements in motor function (Whone A. et al., 2019). Regrettably, the results from this trial closely resembled those seen in previous trials—improved 18F-DOPA uptake in the absence of clinical improvements. Here, we discuss what additional potential inadequacies have confounded various clinical trials and whether any rational hope remains in regard to utilizing this family of growth factors in the treatment of PD.

Lost in Translation

The most obvious question that remains is whether neurotrophic factors such as GDNF truly hold disease modifying potential for PD. A wealth of preclinical data supports the notion that GDNF may prevent or retard nigrostriatal degeneration. Moreover, preclinical and human clinical data clearly indicate that GFLs are, in fact, CNS dopaminergic trophic factors. Therefore, treatment with GFLs should promote survival of striatal dopamine (DA) innervation and thereby improve the motor symptoms of PD. So why have clinical trials utilizing this approach largely failed?

One key aspect of GDNF and similar factors is that the therapeutic mode of action is not fully defined, and that the degenerating PD brain may be resistant to the neuroprotective potential of these proteins. The lack of clarity on GDNF's mechanism of action may be causing issues in appropriate model selection for preclinical therapeutic testing. For instance, GDNF preclinical data is largely based on acute, toxin-induced models–such as 6-hydroxydopamine and MPTP. While administration of GDNF under these settings have provided both neuroprotection and neurorestoration, the same claims of GDNF efficacy have not been substantiated in other models of PD—such as in alpha-synuclein (α-syn) overexpression models (Decressac et al., 2011). In fact, work in the AAV α-syn overexpression model shows that GDNF exhibits no neuroprotective effect (Decressac et al., 2011).

There is some controversy as to the cause of this resistance to neuroprotection. On one hand, the lack of GDNF-induced beneficial effects in the α-syn model has been argued to be due to the downregulation of Nurr1 and its downstream product, the GDNF receptor component receptor tyrosine kinase (RET) (Decressac et al., 2012), although other groups have failed to reproduce the downregulation of Nurr1. Importantly, some key caveats to α-syn overexpression models lie in the finding that α-syn mRNA is in fact decreased in PD and few have reported changes in Nurr1 and RET in human disease (Chu et al., 2006; Su et al., 2017). Moreover, this and other α-syn models of PD overexpress α-syn by 4–10 times normally seen in human studies. Although animal models provide valuable insight into certain disease processes, it is clear that the PD field suffers from a lack of clinically-predictive animal models that faithfully recapitulate all key aspects of parkinsonian neurodegeneration and disease progression. Thus, until we have models more encompassing of the etiopathological features of PD, future reports of preclinical efficacy, or the lack thereof, must be interpreted with caution.

In regard to translation, it is also important to note that the chief risk-factor in PD is age (Collier et al., 2011), yet a majority of preclinical studies have largely neglected this crucial variable. However, it is clear that age alters the local environment, and confers impediments in delivery modalities such as viral vector transduction (Polinski et al., 2016), amongst others. In addition, many intracellular processes change with aging. Thus, performing the preclinical experimentation in models whose age corresponds to that of the average human patient is as important as choosing the model with the most appropriate pathological insult. Nonetheless, it is important to note that GDNF retains at least some function with advanced age as intraputamenal infusion into aged monkeys reduces age-related motoric deficits (Maswood et al., 2002).

Another equally important variable is disease duration. It is well-known that PD patients with longer duration of disease have more disease related complications, and such patients are the target for surgical experimental therapeutics for valid reasons of clinical morbidity. However, this variable introduces a critical barrier to translating animal studies to humans as animal studies do not recapitulate the longevity of disease duration as they are cost prohibitive. Also, in the open label studies as well as in the blinded placebo controlled studies, the average age of onset of disease was much younger than the average age (<50) of onset of PD (Gill et al., 2003; Slevin et al., 2005; Lang et al., 2006). This younger age of onset for PD represents a unique subpopulation as has been recognized by many contemporary researchers (Mehanna and Jankovic, 2019; Espay et al., 2020). This raises the important question if such younger onset patients are the best candidates and if they are indeed chosen based on preclinical age equivalency, then, the GDNF intervention must be performed much earlier in the course of their illness (the mean duration of illness was 10 years in these studies). This raises important ethical issues of risk vs. benefits in early onset PD subjects from invasive neurosurgical interventions. Early onset PD patients have a slower disease progression trajectory and so are treated effectively with pharmacotherapy during this “honeymoon” period that lasts well over 5 years. Yet, based on preclinical testing data, treating these patients earlier within 5 years of disease onset may be the best possible use of GDNF. The only possible ethical solution to this conundrum is to reduce the risks of the neurosurgical intervention. Developing less invasive and more safe methods for intracranial delivery of either GDNF protein or GDNF delivery vectors will allow testing such therapies in early disease in such younger patients with ethical equipoise.

Have We Performed the Right Clinical Trial?

One question in the neurotrophic factor field has always been that of the timing of neurotrophic factor administration. Studies that utilized toxin-induced models clearly demonstrated that GDNF must be administered prior to, or during the insult, in order to achieve efficacy. Administration later may enhance the dopaminergic tone of nigral neurons, but does not provide neuroprotection (Mandel et al., 1997, 1999; Salvatore et al., 2004; Manfredsson et al., 2009a). There is clear evidence from human trials that GFLs can induce DA dendritic sprouting (Love et al., 2005; Kordower et al., 2013) or F-dopa uptake (Gill et al., 2003; Whone A. L. et al., 2019). Therefore, if the theorem that increasing striatal DA should confer therapeutic benefit is correct, then it may be that GFLs are biologically effective but have not reached a necessary threshold of striatal DA regeneration to achieve this benefit. Intervention at earlier stages of disease when more of the nigrostriatal DA pathway is intact or has not undergone plastic changes due to ongoing degeneration, should give GFLs a greater opportunity to reach this threshold of striatal regeneration to provide clinical benefit.

Nevertheless, during the early days of neurotrophic factor delivery, questions regarding the integrity of the nigrostriatal system during disease progression remained. It was not until recently that histopathological characterization by Kordower et al. clearly delineated the significant degree of nigrostriatal denervation during the years immediately following diagnosis (Kordower et al., 2013). Thus, the GFL clinical trials to date targeting late stage patients in Hoehn & Yahr stage 3–4 (where reports have been available) clinical trials are seemingly at odds with the preclinical studies administering the intervention when the nigrostriatal system is mostly intact. To that fact, how many times have we heard variations on the statement “you cannot save what is no longer there”? Thus, the only way to reconcile the field is to test the neuroprotective potential of GDNF and neurturin in early stage patients. During our discussion, it was proposed that the quintessential clinical trial would be performed in patients with unilateral onset, prior to contralateral progression. This, of course, yet again brings up the question about patient safety and advocacy.

Safety of GFLs

Following diagnosis, PD progresses slowly on average, and pharmacological restoration of the dopaminergic tone in the caudate/putamen (e.g., with Sinemet) provides a fairly lengthy “honeymoon period.” Thus, performing a rather complex neurosurgical procedure at a time shortly following diagnosis is not to be taken lightly. This provides an ethical dilemma whereby a clinician is faced with a patient that will maintain acceptable quality of life for some time, yet the disease will progress relentlessly albeit asymmetrically. At this point, how can you justify the testing of an invasive therapeutic paradigm that remains unproven in PD? Neurosurgical experience and advances would support lowering intervention thresholds. Safety data from a wealth of procedures with an indwelling lead/cannula–such as deep brain stimulation (DBS) where a number of anatomical locations, including deep structures, have been targeted—support the lower thresholds as a relatively low rate of serious adverse events are now reported (Budman et al., 2018). In line with DBS safety, striatal intraparenchymal fetal mesencephalic transplantation has demonstrated that the neurosurgical procedure itself is very safe (Lindvall, 2015). Still, despite all the current improvements with stereotactic neurosurgical techniques, one can argue that the risks of a neurosurgical intervention do not match up with the risks associated with currently effective pharmacotherapy in early stages of PD. Therefore, the justification to perform such a procedure must provide disproportionately high benefit to the risks or the risks themselves must get mitigated via the use of less invasive methods of delivery.

Moreover, even if currently optimized surgical methods are used with the least possible adverse events, there are still open questions as to the safety of GDNF itself. Although all indications from preclinical research into neurotrophic factors belonging to this family of proteins suggest that GDNF is safe, perhaps the most compelling data comes from the long-term safety profile of Neurturin, GDNF and other gene therapy-based candidates in human clinical trials (Tenenbaum and Humbert-Claude, 2017; Chu et al., 2020). Nevertheless, there could be consequences of long-term activity with the possibility that secondary issues such as aberrant sprouting of neurons (Georgievska et al., 2002) may lead to a new set of symptoms. Certainly, the use of a clinical approach that allows for cessation of protein delivery (i.e., cannulation/pump infusion, or regulated vectors) would provide a safety mechanism whereby treatment could be halted in the case of an adverse event.

Disease Diagnosis and Tracking of Progression

Most panelists agreed that GFL delivery could be clinically therapeutic if treatment were initiated earlier in disease progression for PD patients. However, even if all clinicians would agree that the delivery of GFLs was of a similar risk to pharmacological treatment (an agreement that is not in place at present), it is currently impossible to reliably detect very early PD (Rizzo et al., 2016). Despite being easily recognized in the public eye, PD is a rather complex disorder, and early diagnosis is not unequivocal (Berg et al., 2018). In fact, a diagnosis of early PD is extremely uncertain, especially when performed outside of a specialty movement disorders Center of Excellence. For instance, other neurodegenerative disorders such as multiple system atrophy (Krismer and Wenning, 2017) and progressive supranuclear palsy (Owolabi, 2013) can often times be misdiagnosed as PD early in the course of disease (Tolosa et al., 2006). This is obviously a complication that makes clinical trial design for early PD increasingly complex. Along the same lines, disease modifying clinical trials in PD, especially early in the disease, are hampered by the fact that there are no good metrics whereby one can measure progression without very large sample sizes or utilizing exceptionally long trial periods. Moreover, trials thus far have been powered to detect improvement in the Unified Parkinson Disease Rating Scale (UPDRS) when perhaps we should be looking for stabilization in decline. Finally, PD is also an extremely heterogenous disorder: Progression rates vary widely, there is heterogeneity in the predominant symptom (e.g., tremor-dominant vs. gait/balance-dominant) that may not respond the same to GDNF, or may not be homogeneous enough for current progression markers (like UPDRS), to detect changes, Thus, as crucial as future biomarkers are in PD for earlier definitive diagnosis (Parnetti et al., 2019) and to track progression, they will be equally important to enable definitive clinical trials in early disease. Such a shift in treatment paradigms would have the greatest impact for PD in the immediate future.

Alternatives to GFLs

Although GDNF and neurturin undoubtedly has undergone the highest scrutiny of all potential trophic factors in PD, there are alternatives worth mentioning. Damage to the striatum results in increased astrocytic production of ciliary neurotrophic factor (CNTF), which belongs to the interleukin-6 family of neuropoietic cytokines. CNTF signaling occurs via a variety of heteroreceptors complexes following binding to CNTF receptor alpha (CNTFRα) (Schuster et al., 2003). Although the exact signaling mechanism is unknown, increased CNTF can protect DA neurons from toxicity, both via direct interaction with neurons as well as by reducing the inflammatory potential of microglia (Hagg and Varon, 1993; Nam et al., 2015; Baek et al., 2018). The closely related trophic factors mesencephalic astrocyte-derived neurotrophic factor (MANF) (Petrova et al., 2003) and cerebral/conserved dopamine neurotrophic factor (CDNF) (Lindholm et al., 2007) similarly provide neuroprotection in various animal models of PD (Airavaara et al., 2012). The exact mode of action of these proteins is unknown, although neuroprotection seems to be, at least in part, conferred via modulation of endoplasmic reticulum stress and autophagy (Zhang et al., 2018). A clinical trial is currently ongoing (ClinicalTrials.gov Identifier: NCT03775538) assessing the safety of putamenal delivery of CDNF (Huttunen and Saarma, 2019) and anecdotal reports suggests that the treatment has been well-tolerated. Finally, small molecule GDNF family receptor (GFR) agonists are being investigated as a potential alternative to the invasive neurosurgical approach otherwise required (Ivanova et al., 2018). However, GFR receptors are heavily expressed in organs throughout the body [The Human Protein Atlas (Uhlen et al., 2015)]. For example, an intracerebroventricularly delivered GDNF trial was halted due to side-effects (Nutt et al., 2003) which is likely due to GDNF's actions in hypothalamus (Manfredsson et al., 2009b). Thus, GDNF administration for PD likely requires site-specific putamenal delivery rendering this strategy the rare case where intraparenchymal delivery is more advantageous than a global small molecule paradigm. Nonetheless, regardless of the therapeutic modality one chooses, the same critical GFL safety factors discussed above apply.

Conclusions

In summary, the panel participants, as well as the audience, expressed cautious optimism for the future of neurotrophic factors, maintaining that GDNF remains a highly promising target in the treatment of PD progression. The preclinical data remain strong, and we simply may not have unleashed the full potential of these proteins, because they have thus not been properly delivered and tested in the context of human disease at feasible points of intervention. Surely, recent improvements such as enhanced vector biodistribution (Kanaan et al., 2017; Davidsson et al., 2019) and less invasive delivery techniques such as focused ultrasound-assisted delivery (Noroozian et al., 2019), are moving us closer to the reinvention of clinical trials. Nevertheless, the path forward is not clear cut, and with current means at our disposal, the execution of early stage clinical trials may not be feasible. It is very possible that the repeated failure to find positive GFL-based clinical trial outcomes mar the field and effectively prohibit future trials from being proposed due lack of financial interests and/or negative public perception. What will the threshold be for investing in new and redesigned trials that are likely to be more expensive than those in the past? In essence, the future of GFL treatment to intervene in the progression of PD symptoms is dependent on significant improvements to preclinical models, improvements to clinical striatal delivery methods, discovery of alternate less invasive methods, improvements to very early PD diagnosis, and especially improvements to PD clinical trial design that would facilitate the prosecution of conclusive clinical trials.

Author Contributions

FM and RM wrote the first version of this manuscript. All authors contributed to the editing of subsequent drafts.

Conflict of Interest

DW was employed by Virscio, Inc.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Airavaara, M., Harvey, B. K., Voutilainen, M. H., Shen, H., Chou, J., Lindholm, P., et al. (2012). CDNF protects the nigrostriatal dopamine system and promotes recovery after MPTP treatment in mice. Cell Transpl. 21, 1213–1223. doi: 10.3727/096368911X600948

PubMed Abstract | CrossRef Full Text | Google Scholar

Baek, J. Y., Jeong, J. Y., Kim, K. I., Won, S. Y., Chung, Y. C., Nam, J. H., et al. (2018). Inhibition of microglia-derived oxidative stress by ciliary neurotrophic factor protects dopamine neurons in vivo from MPP(+) neurotoxicity. Int. J. Mol. Sci. 19:3543. doi: 10.3390/ijms19113543

PubMed Abstract | CrossRef Full Text | Google Scholar

Bartus, R. T., Herzog, C. D., Chu, Y., Wilson, A., Brown, L., Siffert, J., et al. (2011). Bioactivity of AAV2-neurturin gene therapy (CERE-120): differences between Parkinson's disease and nonhuman primate brains. Mov. Disord. 26, 27–36. doi: 10.1002/mds.23442

PubMed Abstract | CrossRef Full Text | Google Scholar

Berg, D., Adler, C. H., Bloem, B. R., Chan, P., Gasser, T., Goetz, C. G., et al. (2018). Movement disorder society criteria for clinically established early Parkinson's disease. Mov. Disord. 33, 1643–1646. doi: 10.1002/mds.27431

PubMed Abstract | CrossRef Full Text | Google Scholar

Budman, E., Deeb, W., Martinez-Ramirez, D., Pilitsis, J. G., Peng-Chen, Z., Okun, M. S., et al. (2018). Potential indications for deep brain stimulation in neurological disorders: an evolving field. Eur. J. Neurol. 25, 434–e30. doi: 10.1111/ene.13548

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, Y., Bartus, R. T., Manfredsson, F., Olanow, C. W., and Kordower, J. H. (2020). Long-term post-mortem studies following neurturin gene therapy in patients with advanced Parkinson's disease. Brain 143, 960–975. doi: 10.1093/brain/awaa020

PubMed Abstract | CrossRef Full Text | Google Scholar

Chu, Y., Le, W., Kompoliti, K., Jankovic, J., Mufson, E. J., and Kordower, J. H. (2006). Nurr1 in Parkinson's disease and related disorders. J. Comp. Neurol. 494, 495–514. doi: 10.1002/cne.20828

PubMed Abstract | CrossRef Full Text | Google Scholar

Collier, T. J., Kanaan, N. M., and Kordower, J. H. (2011). Ageing as a primary risk factor for Parkinson's disease: evidence from studies of non-human primates. Nat. Rev. Neurosci. 12, 359–366. doi: 10.1038/nrn3039

PubMed Abstract | CrossRef Full Text | Google Scholar

Davidsson, M., Wang, G., Aldrin-Kirk, P., Cardoso, T., Nolbrant, S., Hartnor, M., et al. (2019). A systematic capsid evolution approach performed in vivo for the design of AAV vectors with tailored properties and tropism. Proc. Natl. Acad. Sci. U.S.A. 116, 27053–27062. doi: 10.1073/pnas.1910061116

PubMed Abstract | CrossRef Full Text | Google Scholar

Decressac, M., Kadkhodaei, B., Mattsson, B., Laguna, A., Perlmann, T., and Bjorklund, A. (2012). Alpha-synuclein-induced down-regulation of Nurr1 disrupts GDNF signaling in nigral dopamine neurons. Sci. Transl. Med. 4:163ra156. doi: 10.1126/scitranslmed.3004676

PubMed Abstract | CrossRef Full Text | Google Scholar

Decressac, M., Ulusoy, A., Mattsson, B., Georgievska, B., Romero-Ramos, M., Kirik, D., et al. (2011). GDNF fails to exert neuroprotection in a rat alpha-synuclein model of Parkinson's disease. Brain 134, 2302–2311. doi: 10.1093/brain/awr149

PubMed Abstract | CrossRef Full Text | Google Scholar

Espay, A. J., Kalia, L. V., Gan-Or, Z., Williams-Gray, C. H., Bedard, L., and Rowe, S. M., et al. (2020). Disease modification and biomarker development in parkinson disease: revision or reconstruction? Neurology 94, 481–494. doi: 10.1212/WNL.0000000000009107

CrossRef Full Text | Google Scholar

Georgievska, B., Kirik, D., and Bjorklund, A. (2002). Aberrant sprouting and downregulation of tyrosine hydroxylase in lesioned nigrostriatal dopamine neurons induced by long-lasting overexpression of glial cell line derived neurotrophic factor in the striatum by lentiviral gene transfer. Exp. Neurol. 177, 461–474. doi: 10.1006/exnr.2002.8006

PubMed Abstract | CrossRef Full Text | Google Scholar

Gill, S. S., Patel, N. K., Hotton, G. R., O'Sullivan, K., McCarter, R., Bunnage, M., et al. (2003). Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat. Med. 9, 589–595. doi: 10.1038/nm850

PubMed Abstract | CrossRef Full Text | Google Scholar

Hagg, T., and Varon, S. (1993). Ciliary neurotrophic factor prevents degeneration of adult rat substantia nigra dopaminergic neurons in vivo. Proc. Natl. Acad. Sci. U.S.A. 90, 6315–6319. doi: 10.1073/pnas.90.13.6315

PubMed Abstract | CrossRef Full Text | Google Scholar

Huttunen, H. J., and Saarma, M. (2019). CDNF protein therapy in Parkinson's disease. Cell Transpl. 28, 349–366. doi: 10.1177/0963689719840290

PubMed Abstract | CrossRef Full Text | Google Scholar

Ivanova, L., Tammiku-Taul, J., Sidorova, Y., Saarma, M., and Karelson, M. (2018). Small-molecule ligands as potential GDNF family receptor agonists. ACS Omega 3, 1022–1030. doi: 10.1021/acsomega.7b01932

CrossRef Full Text | Google Scholar

Kanaan, N. M., Sellnow, R. C., Boye, S. L., Coberly, B., Bennett, A., Agbandje-McKenna, M., et al. (2017). Rationally engineered AAV capsids improve transduction and volumetric spread in the CNS. Mol. Ther. Nucleic Acids. 8, 184–197. doi: 10.1016/j.omtn.2017.06.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Kirkeby, A., and Barker, R. A. (2019). Parkinson disease and growth factors - is GDNF good enough? Nat. Rev. Neurol. 15, 312–314. doi: 10.1038/s41582-019-0180-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Kordower, J. H., Olanow, C. W., Dodiya, H. B., Chu, Y., Beach, T. G., Adler, C. H., et al. (2013). Disease duration and the integrity of the nigrostriatal system in Parkinson's disease. Brain 136, 2419–2431. doi: 10.1093/brain/awt192

PubMed Abstract | CrossRef Full Text | Google Scholar

Krismer, F., and Wenning, G. K. (2017). Multiple system atrophy: insights into a rare and debilitating movement disorder. Nat. Rev. Neurol. 13, 232–243. doi: 10.1038/nrneurol.2017.26

PubMed Abstract | CrossRef Full Text | Google Scholar

Lang, A. E., Gill, S., Patel, N. K., Lozano, A., Nutt, J. G., Penn, R., et al. (2006). Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol. 59, 459–466. doi: 10.1002/ana.20737

PubMed Abstract | CrossRef Full Text | Google Scholar

Lindholm, P., Voutilainen, M. H., Lauren, J., Peranen, J., Leppanen, V. M., Andressoo, J. O., et al. (2007). Novel neurotrophic factor CDNF protects and rescues midbrain dopamine neurons in vivo. Nature 448, 73–77. doi: 10.1038/nature05957

PubMed Abstract | CrossRef Full Text | Google Scholar

Lindvall, O. (2015). Treatment of Parkinson's disease using cell transplantation. Philos. Trans. R Soc. Lond. B Biol. Sci. 370:20140370. doi: 10.1098/rstb.2014.0370

PubMed Abstract | CrossRef Full Text | Google Scholar

Love, S., Plaha, P., Patel, N. K., Hotton, G. R., Brooks, D. J., and Gill, S. S. (2005). Glial cell line-derived neurotrophic factor induces neuronal sprouting in human brain. Nat. Med. 11, 703–704. doi: 10.1038/nm0705-703

PubMed Abstract | CrossRef Full Text | Google Scholar

Mandel, R. J., Snyder, R. O., and Leff, S. E. (1999). Recombinant adeno-associated viral vector-mediated glial cell line-derived neurotrophic factor gene transfer protects nigral dopamine neurons after onset of progressive degeneration in a rat model of Parkinson's disease. Exp. Neurol. 160, 205–214. doi: 10.1006/exnr.1999.7203

PubMed Abstract | CrossRef Full Text | Google Scholar

Mandel, R. J., Spratt, S. K., Snyder, R. O., and Leff, S. E. (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. Proc. Natl. Acad. Sci. U.S.A. 94, 14083–14088. doi: 10.1073/pnas.94.25.14083

PubMed Abstract | CrossRef Full Text | Google Scholar

Manfredsson, F., Okun, M. S., and Mandel, R. J. (2009a). Gene therapy for neurological disorders: challenges and future prospects for the use of growth factors for the treatment of Parkinson's disease. Curr. Genet. Ther. 9, 375–388. doi: 10.2174/156652309789753400

PubMed Abstract | CrossRef Full Text | Google Scholar

Manfredsson, F., Tumer, N., Erdos, B., Landa, T., Broxson, C. S., Sullivan, L. F., et al. (2009b). Nigrostriatal rAAV-mediated GDNF overexpression induces robust weight loss in a rat model of age-related obesity. Mol. Ther. 17, 980–991. doi: 10.1038/mt.2009.45

PubMed Abstract | CrossRef Full Text | Google Scholar

Marks, W. J. Jr., Bartus, R. T., Siffert, J., Davis, C. S., Lozano, A., Boulis, N., et al. (2010). Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol. 9, 1164–1172. doi: 10.1016/S1474-4422(10)70254-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Marks, W. J. Jr., Ostrem, J. L., Verhagen, L., Starr, A., Larson, S., Bakay, R. A., 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 Neurol. 7, 400–408. doi: 10.1016/S1474-4422(08)70065-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Maswood, N., Grondin, R., Zhang, Z., Stanford, J. A., Surgener, S., Gash, D. M., et al. (2002). Effects of chronic intraputamenal infusion of glial cell line-derived neurotrophic factor (GDNF) in aged rhesus monkeys. Neurobiol. Aging. 23, 881–889. doi: 10.1016/S0197-4580(02)00022-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Mehanna, R., and Jankovic, J. (2019). Young-onset Parkinson's disease: its unique features and their impact on quality of life. Parkinson. Relat. Disord. 65, 39–48. doi: 10.1016/j.parkreldis.2019.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Merola, A., Van Laar, A., Lonser, R., and Bankiewicz, K. (2010). Gene therapy for Parkinson's disease: contemporary practice and emerging concepts. Expert. Rev. Neurother. 20, 577–590. doi: 10.1080/14737175.2020.1763794

PubMed Abstract | CrossRef Full Text | Google Scholar

Nam, J. H., Park, E. S., Won, S. Y., Lee, Y. A., Kim, K. I., Jeong, J. Y., et al. (2015). TRPV1 on astrocytes rescues nigral dopamine neurons in Parkinson's disease via CNTF. Brain 138, 3610–3622. doi: 10.1093/brain/awv297

PubMed Abstract | CrossRef Full Text | Google Scholar

Noroozian, Z., Xhima, K., Huang, Y., Kaspar, B. K., Kugler, S., Hynynen, K., et al. (2019). MRI-guided focused ultrasound for targeted delivery of rAAV to the brain. Methods Mol. Biol. 1950, 177–197. doi: 10.1007/978-1-4939-9139-6_10

PubMed Abstract | CrossRef Full Text | Google Scholar

Nutt, J. G., Burchiel, K. J., Comella, C. L., Jankovic, J., Lang, A. E., Laws, E. R. Jr., et al. (2003). Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 60, 69–73. doi: 10.1212/WNL.60.1.69

PubMed Abstract | CrossRef Full Text | Google Scholar

Owolabi, L. (2013). Progressive supranuclear palsy misdiagnosed as Parkinson's disease: a case report and review of literature. Ann. Med. Health Sci. Res. 3 (Suppl. 1):S44–47. doi: 10.4103/2141-9248.121221

PubMed Abstract | CrossRef Full Text | Google Scholar

Parnetti, L., Gaetani, L., Eusebi, P., Paciotti, S., Hansson, O., El-Agnaf, O., et al. (2019). CSF and blood biomarkers for Parkinson's disease. Lancet Neurol. 18, 573–586. doi: 10.1016/S1474-4422(19)30024-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Petrova, P., Raibekas, A., Pevsner, J., Vigo, N., Anafi, M., Moore, M. K., et al. (2003). MANF: a new mesencephalic, astrocyte-derived neurotrophic factor with selectivity for dopaminergic neurons. J. Mol. Neurosci. 20, 173–188. doi: 10.1385/JMN:20:2:173

PubMed Abstract | CrossRef Full Text | Google Scholar

Polinski, N. K., Manfredsson, F., Benskey, M. J., Fischer, D. L., Kemp, C. J., Steece-Collier, K., et al. (2016). Impact of age and vector construct on striatal and nigral transgene expression. Mol. Ther. Methods Clin. Dev. 3:16082. doi: 10.1038/mtm.2016.82

PubMed Abstract | CrossRef Full Text | Google Scholar

Rizzo, G., Copetti, M., Arcuti, S., Martino, D., Fontana, A., and Logroscino, G. (2016). Accuracy of clinical diagnosis of Parkinson disease: a systematic review and meta-analysis. Neurology 86, 566–576. doi: 10.1212/WNL.0000000000002350

PubMed Abstract | CrossRef Full Text | Google Scholar

Salvatore, M. F., Ai, Y., Fischer, B., Zhang, A. M., Grondin, R. C., Zhang, Z., et al. (2006). Point source concentration of GDNF may explain failure of phase II clinical trial. Exp. Neurol. 202, 497–505. doi: 10.1016/j.expneurol.2006.07.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Salvatore, M. F., Zhang, J. L., Large, D. M., Wilson, E., Gash, C. R., Thomas, T. C., et al. (2004). Striatal GDNF administration increases tyrosine hydroxylase phosphorylation in the rat striatum and substantia nigra. J. Neurochem. 90, 245–254. doi: 10.1111/j.1471-4159.2004.02496.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Schuster, B., Kovaleva, M., Sun, Y., Regenhard, P., Matthews, V., Grotzinger, J., et al. (2003). Signaling of human ciliary neurotrophic factor (CNTF) revisited. The interleukin-6 receptor can serve as an alpha-receptor for CTNF. J. Biol. Chem. 278, 9528–9535. doi: 10.1074/jbc.M210044200

PubMed Abstract | CrossRef Full Text | Google Scholar

Slevin, J. T., Gerhardt, G. A., Smith, C. D., Gash, D. M., Kryscio, R., and Young, B. (2005). Improvement of bilateral motor functions in patients with Parkinson disease through the unilateral intraputaminal infusion of glial cell line-derived neurotrophic factor. J. Neurosurg. 102, 216–222. doi: 10.3171/jns.2005.102.2.0216

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, X., Fischer, D. L., Li, X., Bankiewicz, K., Sortwell, C. E., and Federoff, H. J. (2017). Alpha-synuclein mRNA is not increased in sporadic PD and alpha-synuclein accumulation does not block GDNF signaling in parkinson's disease and disease models. Mol. Ther. 25, 2231–2235. doi: 10.1016/j.ymthe.2017.04.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Tenenbaum, L., and Humbert-Claude, M. (2017). Glial cell line-derived neurotrophic factor gene delivery in parkinson's disease: a delicate balance between neuroprotection, trophic effects, and unwanted compensatory mechanisms. Front. Neuroanat. 11:29. doi: 10.3389/fnana.2017.00029

PubMed Abstract | CrossRef Full Text | Google Scholar

Tolosa, E., Wenning, G., and Poewe, W. (2006). The diagnosis of Parkinson's disease. Lancet Neurol. 5, 75–86. doi: 10.1016/S1474-4422(05)70285-4

CrossRef Full Text | Google Scholar

Uhlen, M., Fagerberg, L., Hallstrom, B. M., Lindskog, C., Oksvold, P., Mardinoglu, A., et al. (2015). Proteomics. Tissue-based map of the human proteome. Science 347:1260419. doi: 10.1126/science.1260419

PubMed Abstract | CrossRef Full Text | Google Scholar

Whone, A., Luz, M., Boca, M., Woolley, M., Mooney, L., Dharia, S., et al. (2019). Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson's disease. Brain 142, 512–525. doi: 10.1093/brain/awz023

PubMed Abstract | CrossRef Full Text | Google Scholar

Whone, A. L., Boca, M., Luz, M., Woolley, M., Mooney, L., Dharia, S., et al. (2019). Extended treatment with glial cell line-derived neurotrophic factor in Parkinson's Disease. J. Parkinsons Dis. 9, 301–313. doi: 10.3233/JPD-191576

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Z., Shen, Y., Luo, H., Zhang, F., Peng, D., Jing, L., et al. (2018). MANF protects dopamine neurons and locomotion defects from a human alpha-synuclein induced Parkinson's disease model in C. elegans by regulating ER stress and autophagy pathways. Exp Neurol. 308, 59–71. doi: 10.1016/j.expneurol.2018.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Parkinson's disease, gene therapy (GT), neurotrophic factor, neurturin, clinical trial (2.172), GDNF (glial cell line-derived neurotrophic factor)

Citation: Manfredsson FP, Polinski NK, Subramanian T, Boulis N, Wakeman DR and Mandel RJ (2020) The Future of GDNF in Parkinson's Disease. Front. Aging Neurosci. 12:593572. doi: 10.3389/fnagi.2020.593572

Received: 12 August 2020; Accepted: 12 October 2020;
Published: 07 December 2020.

Edited by:

Sofia Grade, Austrian Academy of Sciences (OeAW), Austria

Reviewed by:

Aideen M. Sullivan, University College Cork, Ireland
Agnete Kirkeby, University of Copenhagen, Denmark

Copyright © 2020 Manfredsson, Polinski, Subramanian, Boulis, Wakeman and Mandel. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fredric P. Manfredsson, fredric.manfredsson@barrowneuro.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.