You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Bringing Advanced Therapies for Parkinson’s Disease to the Clinic: The Scientist’s Perspective

Abstract

After many years of preclinical development, cell and gene therapies have advanced from research tools in the lab to clinical-grade products for patients, and today they constitute more than a quarter of all new Phase I clinical trials for Parkinson’s disease. Whereas efficacy has been convincingly proven for many of these products in preclinical models, the field is now entering a new phase where the functionality and safety of these products will need to stand the test in clinical trials. If successful, these new products can have the potential to provide patients with a one-time administered treatment which may alleviate them from daily symptomatic dopaminergic medication.

INTRODUCTION: WHY DO WE NEED ADVANCED REGENERATIVE THERAPIES FOR PARKINSON’S DISEASE?

Ever since the discovery of Parkinson’s disease (PD) in the early 19th century, there has been intense focus on trying to identify the underlying cause of the disease as well as the mechanism of disease progression. However, while the development of early symptomatic treatments for the disease has been successful, we have yet to develop disease-modifying treatments to halt or slow progression of PD. Despite promising data from animal models, not a single disease-modifying therapy for PD has until now passed through Phase III clinical trial with positive outcome. Recently, there has been much anticipation to antibody therapies which can block propagation of a-Synuclein (a-Syn) pathology in the brain. However, in April 2020 Prothena/Roche announced that their therapy Prasinezumab had failed to meet the primary endpoint of reduction on the Unified Parkinson’s Disease rating scale (UPDRS) [1]. More phase II trial results are expected this year to uncover the clinical efficacy of a-Syn antibody technologies. The depressing conclusions so far likely reflect the fact that we still do not understand what triggers PD at its infancy, or why patients display such variable patterns of spread of pathology throughout the brain. Furthermore, the pathogenesis of PD is highly complex, involving damaging effects due to protein aggregation, inflammation, mitochondrial dysfunction, impaired autophagy and reactive oxygen species just to mention a few [2], and it may be naive to think that a single molecule can halt this multitude of deleterious processes. It is thought-provoking that the drugs which are currently showing most promising disease-slowing effects in clinical trials are GLP-1 agonists, which have an unclear mechanism of action proposed to involve modulation of several cell types including neurons, glia and immune cells [3, 4]. While refined and efficacious disease-modifying treatments are still struggling to reach the market, patients are in dire need of therapies which provide better quality of life and which are effective beyond the initial period after diagnosis. Compared to symptomatic dopamine (DA)-modifying medications, gene and cell therapies have the potential to provide more refined solutions to a complex problem by restoring neuronal signaling and in some cases even circuits in the diseased brain. As such, cell therapies such as DA cell replacement have the potential to significantly minimise the need for DA medications, relieving the patients not only of primary motor symptoms, but also of the severe side effects accompanying the ever-increasing doses of medication required to keep the progressing disease in check.

ADVANCED THERAPIES FOR PARKINSON’S DISEASE PASSING THROUGH THE PIPELINE

Until now, only few advanced therapeutic medicinal products (ATMPs) have been tested beyond initial Phase I/II clinical trials for PD. Overall, trials involving delivery of neurotrophic factors for supporting DA neuron survival, i.e., AAV-Neurturin (CERE-120) and GDNF infusion have shown disappointing outcomes on efficacy parameters [5, 6]. This likely reflects the fact that damaged DA neurons are difficult to rescue from cell death when disease pathology is ongoing. A novel gene therapy trial relying on supplying GBA1 to GBA mutation carriers is currently in Phase-I clinical trial, and will provide important information on whether GBA mutations alone are central in driving the progression of the disease in already diagnosed patients [7]. Other gene therapies are based on strategies to alter neurotransmitter production, either through hijacking non-DA neurons to produce DA by supplying key DA enzymes (ProSavin: AADC, TH and CH1) or by increasing inhibitory signals from the subthalmic nucleus (STN) through AAV-GAD expression. Both approaches have shown moderate efficacy on UPDRS scores in clinical trials and are being explored further in new trials [8, 9]. However, an initiated Phase-II gene therapy trial with AAV-AADC (VY-AADC02) was halted by the FDA in December 2020, and the future development of this product is currently uncertain [7, 10]. Evolving from these early efforts with ATMPs, more advanced products are being developed and cell and gene therapies now constitute 27% of all ongoing Phase I studies in PD (14 out of 51 trials by January 2020), thereby clearly marking a new era of advanced therapies moving in larger numbers from the lab into the clinic [7]. New on this stage is the emergence of DA cell replacement products based on pluripotent stem cells (PSCs), which have the potential to yield authentic and fully functional midbrain DA neurons, the type which is lost in PD.

Table 1

Pluripotent stem cell products in the clinical trial pipeline for PD

Cell productSite of development (Company/Institution)Immune matchingMoATrial sitePreclin. dataTrial startPatients included*Clinical data trial ID
ISC-hpNSC (Human parthenogenetic hESC-derived NSCs)International Stem Cell Corporation (ISCO)Allogeneic, non-matchedTrophic supportAustralia[13, 38, 39]201612N/A NCT02452723
Human parthenogenetic hESC-derived ventral midbrain progenitorsChinese Academy of Sciences, BeijingAllogeneic, HLA matched and non-matchedDA-CRTChina[40]2017(50)N/A NCT03119636
hiPSC-derived ventral midbrain progenitorsHarvard UniversityAutologousDA-CRTUSA[41]20171[29] IND No. 17145
hiPSC-derived ventral midbrain progenitorsKyoto UniversityAllogeneic, HLA matched and non-matchedDA-CRTJapan[17, 42, 43]2018(7)N/A UMIN000033564
MSK-DA01 (hESC-derived ventral midbrain progenitors)Weill Cornell/Memorial Sloan Kettering/BlueRock TherapeuticsAllogeneic, non-matchedDA-CRTUSA[16, 19, 20, 44]2021(10)N/A NCT04802733
STEM-PD (hESC-derived ventral midbrain progenitors)Lund University/Cambridge UniversityAllogeneic, non-matchedDA-CRTSweden UK[30, 45– 50]Est. 2022(8)N/A EudraCT 2021-001366-38

MoA, Mechanism of Action; NSC, Neural stem cell; DA-CRT, Dopamine cell replacement therapy; N/A, not available. *Patient numbers in brackets are total no. of planned patients in trials which are not yet completed. Patient numbers without brackets are completed, transplanted patients.

CLINICAL TRANSLATION OF DA CELL REPLACEMENT THERAPIES: WHERE ARE WE NOW?

The concept of DA cell replacement therapy has a rich clinical history based on allografting of human fetal ventral mesencephalic tissue, with estimates of over 300 PD patients transplanted over the past 30 years [11]. Patient outcomes have been variable, with some patients able to reduce or discontinue medications for years while others suffered from graft-induced dyskinesias. Two double-blinded, placebo-controlled trials failed to meet their endpoint which led to a re-evaluation of fetal cell therapies for Parkinson’s, eventually leading to the initiation of the EU-funded TRANSEURO trial using improved protocols for fetal tissue preparation and more rigorous patient selection [12]. This trial has recently completed transplantation of all 11 patients included in the trial with fetal tissue, and clinical results are expected during 2021/22 [12]. Along with earlier fetal cell work, results from this trial will provide an important framework for stem cell-based therapies to come.

Clinical trials using PSC-based therapies have only just begun (see summary in Table 1). The first PSC-based trial for PD (ISCO trial) was initiated in Australia in 2016, using parthenogenetic PSC-derived neural progenitor cells of a non-DA fate [13]. These cells are proposed to have a more indirect mechanism of action, working potentially through trophic support. However, multiple groups have shown through genetic engineering in preclinical models that it is specifically the midbrain DA neurons which are responsible for reverting symptoms in animal models of PD [14– 16].

Following this rationale, Jun Takahashi and colleagues launched the first PSC-based midbrain DA neuron cell replacement in Japan in 2018 using an allogenic iPSC approach [17, 18]. Just recently, a collaborative effort between Weill Cornell, Memorial Sloan Kettering, and BlueRock Therapeutics was granted permission by the FDA to advance to Phase 1 clinical trial in the US with an hESC-based midbrain DA product (MSK-DA01) originally developed by Lorenz Studer [19– 21]. This trial is anticipated to begin later this year. In an equivalent European effort, Malin Parmar and colleagues have provided strong proof of concept for their hESC-based product and are aiming to begin clinical trials in the EU in 2022. These groups working on PSC-based products have been meeting annually since 2014 in the research-based network GForce-PD with the aim of sharing experiences to bring the best treatments forward for the PD community. Building on these academically led advancements, three additional companies, Sumitomo Dainippon Pharma, FujiFilm Cellular Dynamics Inc. and Novo Nordisk have also announced the development of PSC-based cell products for treatment of PD.

The trials mentioned above are based on allogeneic cell transplantation strategies. Induced pluripotent stem cell technology could provide an autologous approach with the benefit of genetic matching at the cost of time, logistic complexity and potential variability. For intracerebral therapies, it is not clear that autologous approaches will be advantageous in the immunoprivileged brain since fetal cell allografts have demonstrated robust and long-lived survival, up to 24 years without long term immune suppression [22– 28]. Relying on autologous grafting could limit the accessibility of stem cell therapies due to the cost and the complication of batch-to-batch variation in differentiation efficacy. Nevertheless, one cannot discount the possibility that the immune response to grafted allogeneic cells can limit efficacy. A recent single patient case led by the group of Kwang-Soo Kim at Harvard demonstrated proof of concept for the autologous transplantation approach [29]. The Kim group, as well as the US-based company Aspen Neuroscience, is now pursuing further clinical trials using autologous iPSCs for transplantation.

LOOKING TOWARDS THE FUTURE OF PSC-BASED CELL THERAPIES

It has taken around twenty years of intensive preclinical work to produce clinically acceptable pluripotent stem cell-derived human DA neurons that function efficiently in PD animal models. While this is great progress, much remains to be learned from clinical trials to improve cell products further. Careful work by the Parmar lab [30] has shown equivalence between fetal cell and PSC-based dopamine neurons in animal models of PD, but clinical data will be crucial to assess this equivalence in patients. Another unresolved question is whether transplanted DA neurons are “self-regulating.” That is, DA neurons might receive feedback from the brain that naturally balances and limits dopamine release. This is important since excessive dopamine release might result in dyskinesias (involuntary movement) [31]. Understanding this biology in humans will inform our strategy for patient dosing.

Another factor affecting dose is the observation that many neurons die after transplantation. It is not known why this occurs, and excess cells must be transplanted to account for this loss and assure adequate dosing. The delivery device itself creates an injury and a localized immune reaction, and large numbers of dead cells could alert the immune system to the transplanted cells. Beyond just the transplantation period itself, monitoring immune reactivity in allogeneic and autologous cell transplantation trials will be key to understanding the role of the immune response to grafted cells and is crucial for future cell therapy development. A better understanding of the immune response to grafts is needed to evaluate whether allogeneic cell sources can survive and escape rejection longterm, or if the autologous cells will provide a clinical advantage despite the many hurdles associated with their manufacturing. Many groups are further looking into the possibilities of using gene-edited allogeneic cell sources which can escape immune recognition for future universal use in patients to avoid the complexities and cost of making a personalized medicine [32– 35]. Such cell lines must however be used with caution since these cells might inadvertently function as host cells to amplify viruses or permit uncontrolled cell growth. As such, incorporation of “suicide genes” into such engineered cells would allow the clinical option to chemically ablate the grafted cells if required [36, 37].

One inconvenient truth learned from the fetal cell trials is that it can take several years after transplantation to achieve optimal clinical function from cell replacement therapies. This makes the choice of a clinical endpoint critical to assure the strongest clinical signal and reduced chance for placebo effects. Another key piece to the puzzle remains the patients themselves. PD has been increasingly recognized as a syndrome with a range of clinical courses. Increasing knowledge of PD subtypes and results from stem cell trials will allow us to better understand which patients will truly benefit from cell replacement therapies.

ACKNOWLEDGMENTS

The authors would like to thank Joachim Fruebis and Seth Ettenberg for critical and constructive comments to the manuscript.

CONFLICT OF INTEREST

Mark Tomishima holds several patent agreements and is an employee of BlueRock Therapeutics, a company that plans to commercially develop the DA01 product. Agnete Kirkeby holds several patent agreements and is a consultant for Novo Nordisk A/S on the development of the STEM-PD product.

REFERENCES

[1] 

Prothena ((2020) ) https://ir.prothena.com/news-releases/news-release-details/update-phase-2-pasadena-study-prasinezumab-prx002rg7935.

[2] 

Maiti P , Manna J , Dunbar GL ((2017) ) Current understanding of the molecular mechanisms in Parkinson’s disease: Targets for potential treatments. Transl Neurodegener 6: , 28.

[3] 

McFarthing K , Larson D , Simuni T ((2020) ) Clinical Trial Highlights - GLP-1 agonists. J Parkinsons Dis 10: , 355–368.

[4] 

Grieco M , Giorgi A , Gentile MC , d’Erme M , Morano S , Maras B , Filardi T ((2019) ) Glucagon-like peptide-1: A focus on neurodegenerative diseases. Front Neurosci 13: , 1112.

[5] 

Marks WJ Jr. , Bartus RT , Siffert J , Davis CS , Lozano A , Boulis N , Vitek J , Stacy M , Turner D , Verhagen L , Bakay R , Watts R , Guthrie B , Jankovic J , Simpson R , Tagliati M , Alterman R , Stern M , Baltuch G , Starr PA , Larson PS , Ostrem JL , Nutt J , Kieburtz K , Kordower JH , Olanow CW ((2010) ) Gene delivery of AAV2-neurturin for Parkinson’s disease: A double-blind, randomised, controlled trial. Lancet Neurol 9: , 1164–1172.

[6] 

Whone A , Luz M , Boca M , Woolley M , Mooney L , Dharia S , Broadfoot J , Cronin D , Schroers C , Barua NU , Longpre L , Barclay CL , Boiko C , Johnson GA , Fibiger HC , Harrison R , Lewis O , Pritchard G , Howell M , Irving C , Johnson D , Kinch S , Marshall C , Lawrence AD , Blinder S , Sossi V , Stoessl AJ , Skinner P , Mohr E , Gill SS ((2019) ) Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain 142: , 512–525.

[7] 

McFarthing K , Buff S , Rafaloff G , Dominey T , Wyse RK , Stott SRW ((2020) ) Parkinson’s disease drug therapies in the clinical trial pipeline: 2020. J Parkinsons Dis 10: , 757–774.

[8] 

Niethammer M , Tang CC , LeWitt PA , Rezai AR , Leehey MA , Ojemann SG , Flaherty AW , Eskandar EN , Kostyk SK , Sarkar A , Siddiqui MS , Tatter SB , Schwalb JM , Poston KL , Henderson JM , Kurlan RM , Richard IH , Sapan CV , Eidelberg D , During MJ , Kaplitt MG , Feigin A ((2017) ) Long-term follow-up of a randomized AAV2-GAD gene therapy trial for Parkinson’s disease. JCI Insight 2: , e90133.

[9] 

Palfi S , Gurruchaga JM , Lepetit H , Howard K , Ralph GS , Mason S , Gouello G , Domenech P , Buttery PC , Hantraye P , Tuckwell NJ , Barker RA , Mitrophanous KA ((2018) ) Long-term follow-up of a Phase I/II Study of ProSavin, a lentiviral vector gene therapy for Parkinson’s disease. Hum Gene Ther Clin Dev 29: , 148–155.

[10] 

https://www.globenewswire.com/fr/news-release/2021/02/02/2168590/0/en/Voyager-Therapeutics-Provides-Update-on-NBIb-1817-VY-AADC-Gene-Therapy-Program.html.

[11] 

Kim TW , Koo SY , Studer L ((2020) ) Pluripotent stem cell therapies for parkinson disease: Present challenges and future opportunities. Front Cell Dev Biol 8: , 729.

[12] 

Barker RA , consortium T ((2019) ) Designing stem-cell-based dopamine cell replacement trials for Parkinson’s disease. Nat Med 25: , 1045–1053.

[13] 

Garitaonandia I , Gonzalez R , Christiansen-Weber T , Abramihina T , Poustovoitov M , Noskov A , Sherman G , Semechkin A , Snyder E , Kern R ((2016) ) Neural stem cell tumorigenicity and biodistribution assessment for Phase I clinical trial in Parkinson’s disease. Sci Rep 6: , 34478.

[14] 

Aldrin-Kirk P , Heuer A , Wang G , Mattsson B , Lundblad M , Parmar M , Bjorklund T ((2016) ) DREADD modulation of transplanted DA neurons reveals a novel parkinsonian dyskinesia mechanism mediated by the serotonin 5-HT6 receptor. Neuron 90: , 955–968.

[15] 

Chen Y , Xiong M , Dong Y , Haberman A , Cao J , Liu H , Zhou W , Zhang SC ((2016) ) Chemical control of grafted human PSC-Derived neurons in a mouse model of Parkinson’s disease. Cell Stem Cell 18: , 817–826.

[16] 

Steinbeck JA , Choi SJ , Mrejeru A , Ganat Y , Deisseroth K , Sulzer D , Mosharov EV , Studer L ((2015) ) Optogenetics enables functional analysis of human embryonic stem cell-derived grafts in a Parkinson’s disease model. Nat Biotechnol 33: , 204–209.

[17] 

Doi D , Magotani H , Kikuchi T , Ikeda M , Hiramatsu S , Yoshida K , Amano N , Nomura M , Umekage M , Morizane A , Takahashi J ((2020) ) Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson’s disease. Nat Commun 11: , 3369.

[18] 

Takahashi J ((2020) ) iPS cell-based therapy for Parkinson’s disease: A Kyoto trial. Regen Ther 13: , 18–22.

[19] 

Kim TW , Piao J , Koo SY , Kriks S , Chung SY , Betel D , Socci ND , Choi SJ , Zabierowski S , Dubose BN , Hill EJ , Mosharov EV , Irion S , Tomishima MJ , Tabar V , Studer L ((2021) ) Biphasic activation of WNT signaling facilitates the derivation of midbrain dopamine neurons from hESCs for translational use. Cell Stem Cell 28: , 343–355 e345.

[20] 

Piao J , Zabierowski S , Dubose BN , Hill EJ , Navare M , Claros N , Rosen S , Ramnarine K , Horn C , Fredrickson C , Wong K , Safford B , Kriks S , El Maarouf A , Rutishauser U , Henchcliffe C , Wang Y , Riviere I , Mann S , Bermudez V , Irion S , Studer L , Tomishima M , Tabar V ((2021) ) Preclinical efficacy and safety of a human embryonic stem cell-derived midbrain dopamine progenitor product, MSK-DA01. Cell Stem Cell 28: , 217–229 e217.

[21] 

Takahashi J ((2021) ) Clinical trial for Parkinson’s disease gets a green light in the US. Cell Stem Cell 28: , 182–183.

[22] 

Li W , Englund E , Widner H , Mattsson B , van Westen D , Latt J , Rehncrona S , Brundin P , Bjorklund A , Lindvall O , Li JY ((2016) ) Extensive graft-derived dopaminergic innervation is maintained 24 years after transplantation in the degenerating parkinsonian brain. Proc Natl Acad Sci U S A 113: , 6544–6549.

[23] 

Hallett PJ , Cooper O , Sadi D , Robertson H , Mendez I , Isacson O ((2014) ) Long-term health of dopaminergic neuron transplants in Parkinson’s disease patients. Cell Rep 7: , 1755–1761.

[24] 

Kordower JH , Chu Y , Hauser RA , Freeman TB , Olanow CW ((2008) ) Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease.. Nat Med 14: , 504–506.

[25] 

Li JY , Englund E , Holton JL , Soulet D , Hagell P , Lees AJ , Lashley T , Quinn NP , Rehncrona S , Bjorklund A , Widner H , Revesz T , Lindvall O , Brundin P ((2008) ) Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 14: , 501–503.

[26] 

Li JY , Englund E , Widner H , Rehncrona S , Bjorklund A , Lindvall O , Brundin P ((2010) ) Characterization of Lewy body pathology in 12- and 16-year-old intrastriatal mesencephalic grafts surviving in a patient with Parkinson’s disease. Mov Disord 25: , 1091–1096.

[27] 

Mendez I , Vinuela A , Astradsson A , Mukhida K , Hallett P , Robertson H , Tierney T , Holness R , Dagher A , Trojanowski JQ , Isacson O ((2008) ) Dopamine neurons implanted into people with Parkinson’s disease survive without pathology for 14 years. Nat Med 14: , 507–509.

[28] 

Freed CR , Zhou W , Breeze RE ((2011) ) Dopamine cell transplantation for Parkinson’s disease: The importance of controlled clinical trials. Neurotherapeutics 8: , 549–561.

[29] 

Schweitzer JS , Song B , Herrington TM , Park TY , Lee N , Ko S , Jeon J , Cha Y , Kim K , Li Q , Henchcliffe C , Kaplitt M , Neff C , Rapalino O , Seo H , Lee IH , Kim J , Kim T , Petsko GA , Ritz J , Cohen BM , Kong SW , Leblanc P , Carter BS , Kim KS ((2020) ) Personalized iPSC-derived dopamine progenitor cells for Parkinson’s disease. N Engl J Med 382: , 1926–1932.

[30] 

Grealish S , Diguet E , Kirkeby A , Mattsson B , Heuer A , Bramoulle Y , Van Camp N , Perrier AL , Hantraye P , Bjorklund A , Parmar M ((2014) ) Human ESC-derived dopamine neurons show similar preclinical efficacy and potency to fetal neurons when grafted in a rat model of Parkinson’s disease. Cell Stem Cell 15: , 653–665.

[31] 

Greene PE , Fahn S , Eidelberg D , Bjugstad KB , Breeze RE , Freed CR ((2021) ) Persistent dyskinesias in patients with fetal tissue transplantation for Parkinson disease. NPJ Parkinsons Dis 7: , 38.

[32] 

Deuse T , Hu X , Gravina A , Wang D , Tediashvili G , De C , Thayer WO , Wahl A , Garcia JV , Reichenspurner H , Davis MM , Lanier LL , Schrepfer S ((2019) ) Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat Biotechnol 37: , 252–258.

[33] 

Gornalusse GG , Hirata RK , Funk SE , Riolobos L , Lopes VS , Manske G , Prunkard D , Colunga AG , Hanafi LA , Clegg DO , Turtle C , Russell DW ((2017) ) HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol 35: , 765–772.

[34] 

Han X , Wang M , Duan S , Franco PJ , Kenty JH , Hedrick P , Xia Y , Allen A , Ferreira LMR , Strominger JL , Melton DA , Meissner TB , Cowan CA ((2019) ) Generation of hypoimmunogenic human pluripotent stem cells. Proc Natl Acad Sci U S A 116: , 10441–10446.

[35] 

Xu H , Wang B , Ono M , Kagita A , Fujii K , Sasakawa N , Ueda T , Gee P , Nishikawa M , Nomura M , Kitaoka F , Takahashi T , Okita K , Yoshida Y , Kaneko S , Hotta A ((2019) ) Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility.566-578 e. Cell Stem Cell 24: , 567.

[36] 

Ciceri F , Bonini C , Stanghellini MT , Bondanza A , Traversari C , Salomoni M , Turchetto L , Colombi S , Bernardi M , Peccatori J , Pescarollo A , Servida P , Magnani Z , Perna SK , Valtolina V , Crippa F , Callegaro L , Spoldi E , Crocchiolo R , Fleischhauer K , Ponzoni M , Vago L , Rossini S , Santoro A , Todisco E , Apperley J , Olavarria E , Slavin S , Weissinger EM , Ganser A , Stadler M , Yannaki E , Fassas A , Anagnostopoulos A , Bregni M , Stampino CG , Bruzzi P , Bordignon C ((2009) ) Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): A non-randomised phase I-II study. Lancet Oncol 10: , 489–500.

[37] 

Di Stasi A , Tey SK , Dotti G , Fujita Y , Kennedy-Nasser A , Martinez C , Straathof K , Liu E , Durett AG , Grilley B , Liu H , Cruz CR , Savoldo B , Gee AP , Schindler J , Krance RA , Heslop HE , Spencer DM , Rooney CM , Brenner MK ((2011) ) Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 365: , 1673–1683.

[38] 

Gonzalez R , Garitaonandia I , Poustovoitov M , Abramihina T , McEntire C , Culp B , Attwood J , Noskov A , Christiansen-Weber T , Khater M , Mora-Castilla S , To C , Crain A , Sherman G , Semechkin A , Laurent LC , Elsworth JD , Sladek J , Snyder EY , Redmond DE Jr. , Kern RA ((2016) ) Neural stem cells derived from human parthenogenetic stem cells engraft and promote recovery in a nonhuman primate model of Parkinson’s disease. Cell Transplant 25: , 1945–1966.

[39] 

Gonzalez R , Garitaonandia I , Crain A , Poustovoitov M , Abramihina T , Noskov A , Jiang C , Morey R , Laurent LC , Elsworth JD , Snyder EY , Redmond DE Jr. , Semechkin R ((2015) ) Proof of concept studies exploring the safety and functional activity of human parthenogenetic-derived neural stem cells for the treatment of Parkinson’s disease. Cell Transplant 24: , 681–690.

[40] 

Wang YK , Zhu WW , Wu MH , Wu YH , Liu ZX , Liang LM , Sheng C , Hao J , Wang L , Li W , Zhou Q , Hu BY ((2018) ) Human clinical-grade parthenogenetic ESC-derived dopaminergic neurons recover locomotive defects of nonhuman primate models of Parkinson’s disease. Stem Cell Rep 11: , 171–182.

[41] 

Song B , Cha Y , Ko S , Jeon J , Lee N , Seo H , Park KJ , Lee IH , Lopes C , Feitosa M , Luna MJ , Jung JH , Kim J , Hwang D , Cohen BM , Teicher MH , Leblanc P , Carter BS , Kordower JH , Bolshakov VY , Kong SW , Schweitzer JS , Kim KS ((2020) ) Human autologous iPSC-derived dopaminergic progenitors restore motor function in Parkinson’s disease models. J Clin Invest 130: , 904–920.

[42] 

Morizane A , Kikuchi T , Hayashi T , Mizuma H , Takara S , Doi H , Mawatari A , Glasser MF , Shiina T , Ishigaki H , Itoh Y , Okita K , Yamasaki E , Doi D , Onoe H , Ogasawara K , Yamanaka S , Takahashi J ((2017) ) MHC matching improves engraftment of iPSC-derived neurons in non-human primates. Nat Commun 8: , 385.

[43] 

Kikuchi T , Morizane A , Doi D , Onoe H , Hayashi T , Kawasaki T , Saiki H , Miyamoto S , Takahashi J ((2017) ) Survival of human induced pluripotent stem cell-derived midbrain dopaminergic neurons in the brain of a primate model of Parkinson’s disease. J Parkinsons Dis 1: , 395–412.

[44] 

Kriks S , Shim JW , Piao J , Ganat YM , Wakeman DR , Xie Z , Carrillo-Reid L , Auyeung G , Antonacci C , Buch A , Yang L , Beal MF , Surmeier DJ , Kordower JH , Tabar V , Studer L ((2011) ) Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease. Nature 480: , 547–551.

[45] 

Grealish S , Heuer A , Cardoso T , Kirkeby A , Jonsson M , Johansson J , Bjorklund A , Jakobsson J , Parmar M ((2015) ) Monosynaptic tracing using modified rabies virus reveals early and extensive circuit integration of human embryonic stem cell-derived neurons. Stem Cell Rep 4: , 975–983.

[46] 

Kirkeby A , Nolbrant S , Tiklova K , Heuer A , Kee N , Cardoso T , Ottosson DR , Lelos MJ , Rifes P , Dunnett SB , Grealish S , Perlmann T , Parmar M ((2017) ) Predictive markers guide differentiation to improve graft outcome in clinical translation of hESC-based therapy for Parkinson’s disease. Cell Stem Cell 20: , 135–148.

[47] 

Nolbrant S , Heuer A , Parmar M , Kirkeby A ((2017) ) Generation of high-purity human ventral midbrain dopaminergic progenitors for in vitro maturation and intracerebral transplantation. Nat Protoc 12: , 1962–1979.

[48] 

Cardoso T , Adler AF , Mattsson B , Hoban DB , Nolbrant S , Wahlestedt JN , Kirkeby A , Grealish S , Bjorklund A , Parmar M ((2018) ) Target-specific forebrain projections and appropriate synaptic inputs of hESC-derived dopamine neurons grafted to the midbrain of parkinsonian rats. J Comp Neurol 526: , 2133–2146.

[49] 

Adler AF , Cardoso T , Nolbrant S , Mattsson B , Hoban DB , Jarl U , Wahlestedt JN , Grealish S , Bjorklund A , Parmar M ((2019) ) hESC-derived dopaminergic transplants integrate into basal ganglia circuitry in a preclinical model of Parkinson’s disease. Cell Rep 28: , 3462–3473 e3465.

[50] 

Tiklova K , Nolbrant S , Fiorenzano A , Bjorklund AK , Sharma Y , Heuer A , Gillberg L , Hoban DB , Cardoso T , Adler AF , Birtele M , Lunden-Miguel H , Volakakis N , Kirkeby A , Perlmann T , Parmar M ((2020) ) Single cell transcriptomics identifies stem cell-derived graft composition in a model of Parkinson’s disease. Nat Commun 11: , 2434.