You are on page 1of 6

Journal of Parkinson’s Disease xx (2021) x–xx 1

DOI 10.3233/JPD-212685
IOS Press

1 Review

2 Bringing Advanced Therapies for


Parkinson’s Disease to the Clinic:

f
3

roo
4 The Scientist’s Perspective

rP
5 Mark Tomishimaa and Agnete Kirkebyb,c,∗
6
a BlueRock Therapeutics, New York, NY, USA
7
b Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
c Wallenberg Center for Molecular Medicine (WCMM) and Department of Experimental Medical Science,

Lund University, Lund, Sweden

tho
8
11

10 Accepted 14 June 2021


Pre-press 3 July 2021 Au
12 Abstract. After many years of preclinical development, cell and gene therapies have advanced from research tools in the
13 lab to clinical-grade products for patients, and today they constitute more than a quarter of all new Phase I clinical trials for
14 Parkinson’s disease. Whereas efficacy has been convincingly proven for many of these products in preclinical models, the
15 field is now entering a new phase where the functionality and safety of these products will need to stand the test in clinical
16 trials. If successful, these new products can have the potential to provide patients with a one-time administered treatment
d

17 which may alleviate them from daily symptomatic dopaminergic medication.

Keywords: ATMP, clinical trial, dopaminergic neurons, regenerative therapy, stem cells, transplantation
cte

18
rre

19 INTRODUCTION: WHY DO WE NEED treatments to halt or slow progression of PD. Despite 30

20 ADVANCED REGENERATIVE promising data from animal models, not a single 31

21 THERAPIES FOR PARKINSON’S disease-modifying therapy for PD has until now 32

22 DISEASE? passed through Phase III clinical trial with positive 33


co

outcome. Recently, there has been much anticipation 34

23 Ever since the discovery of Parkinson’s disease to antibody therapies which can block propagation 35

24 (PD) in the early 19th century, there has been intense of a-Synuclein (a-Syn) pathology in the brain. How- 36

25 focus on trying to identify the underlying cause of ever, in April 2020 Prothena/Roche announced that 37
Un

26 the disease as well as the mechanism of disease pro- their therapy Prasinezumab had failed to meet the 38

27 gression. However, while the development of early primary endpoint of UPDRS reduction from the first 39

28 symptomatic treatments for the disease has been suc- such a phase II antibody trial [1]. More phase II trial 40

29 cessful, we have yet to develop disease-modifying results are expected this year to uncover the clin- 41

ical efficacy of a-Syn antibody technologies. The 42

∗ Correspondence
depressing conclusions so far likely reflect the fact 43
to: Agnete Kirkeby, Wallenberg Center for
Molecular Medicine (WCMM), and Department of Experimen-
that we still do not understand what triggers PD 44

tal Medical Science, Lund University, 22184 Lund, Sweden. Tel.: at its infancy, or why patients display such vari- 45

+45 5168 5353; E-mail: agnete.kirkeby@med.lu.se. able patterns of spread of pathology throughout the 46

ISSN 1877-7171/$35.00 © 2021 – IOS Press. All rights reserved.


2 M. Tomishima and A. Kirkeby / The Scientist’s Perspective to PD Therapies

47 brain. Furthermore, the pathogenesis of PD is highly non-DA neurons to produce DA by supplying key 96

48 complex, involving damaging effects due to protein DA enzymes (ProSavin: AADC, TH and CH1) or 97

49 aggregation, inflammation, mitochondrial dysfunc- by increasing inhibitory signals from the subthalmic 98

50 tion, impaired autophagy and reactive oxygen species nucleus (STN) through AAV-GAD expression. Both 99

51 just to mention a few [2], and it may be naive to approaches have shown moderate efficacy on UPDRS 100

52 think that a single molecule can halt this multitude of scores in clinical trials and are being explored further 101

53 deleterious processes. It is thought-provoking that the in new trials [8, 9]. However, an initiated Phase-II 102

54 drugs which are currently showing most promising gene therapy trial with AAV-AADC (VY-AADC02) 103

f
55 disease-slowing effects in clinical trials are GLP- was halted by the FDA in December 2020, and the 104

roo
56 1 agonists, which have an unclear mechanism of future development of this product is currently uncer- 105

57 action proposed to involve modulation of several cell tain [7, 10]. Evolving from these early efforts with 106

58 types including neurons, glia and immune cells [3, ATMPs, more advanced products are being devel- 107

59 4]. While refined and efficacious disease-modifying oped and cell and gene therapies now constitute 27% 108

60 treatments are still struggling to reach the market, of all ongoing Phase I studies in PD (14 out of 51 tri- 109

rP
61 patients are in dire need of therapies which pro- als by January 2020), thereby clearly marking a new 110

62 vide better quality of life and which are effective era of advanced therapies moving in larger numbers 111

63 beyond the initial period after diagnosis. Compared from the lab into the clinic [7]. New on this stage 112

64 to symptomatic dopamine (DA)-modifying medica- is the emergence of DA cell replacement products 113

tho
65 tions, gene and cell therapies have the potential to based on pluripotent stem cells (PSCs), which have 114

66 provide more refined solutions to a complex prob- the potential to yield authentic and fully functional 115

67 lem by restoring neuronal signaling and in some midbrain DA neurons, the type which is lost in PD. 116

68 cases even circuits in the diseased brain. As such,


69 cell therapies such as DA cell replacement have the
Au
70 potential to significantly minimise the need for DA CLINICAL TRANSLATION OF DA CELL 117

71 medications, relieving the patients not only of pri- REPLACEMENT THERAPIES: WHERE 118

72 mary motor symptoms, but also of the severe side ARE WE NOW? 119

73 effects accompanying the ever-increasing doses of


74 medication required to keep the progressing disease The concept of DA cell replacement therapy 120

in check. has a rich clinical history based on allografting of


d

75 121

human fetal ventral mesencephalic tissue, with esti- 122


cte

mates of over 300 PD patients transplanted over 123

76 ADVANCED THERAPIES FOR the past 30 years [11]. Patient outcomes have been 124

77 PARKINSON’S DISEASE PASSING variable, with some patients able to reduce or dis- 125

78 THROUGH THE PIPELINE continue medications for years while others suffered 126

from graft-induced dyskinesias. Two double-blinded, 127


rre

79 Until now, only few advanced therapeutic medic- placebo-controlled trials failed to meet their endpoint 128

80 inal products (ATMPs) have been tested beyond which led to a re-evaluation of fetal cell therapies for 129

81 initial Phase I/II clinical trials for PD. Overall, tri- Parkinson’s, eventually leading to the initiation of the 130

82 als involving delivery of neurotrophic factors for EU-funded TRANSEURO trial using improved pro- 131
co

83 supporting DA neuron survival, i.e., AAV-Neurturin tocols for fetal tissue preparation and more rigorous 132

84 (CERE-120) and GDNF infusion have shown dis- patient selection [12]. This trial has recently com- 133

85 appointing outcomes on efficacy parameters [5, 6]. pleted transplantation of all 11 patients included in the 134

86 This likely reflects the fact that damaged DA neu- trial with fetal tissue, and clinical results are expected 135
Un

87 rons are difficult to rescue from cell death when during 2021/22 [12]. Along with earlier fetal cell 136

88 disease pathology is ongoing. A novel gene therapy work, results from this trial will provide an important 137

89 trial relying on supplying GBA1 to GBA mutation framework for stem cell-based therapies to come. 138

90 carriers is currently in Phase-I clinical trial, and Clinical trials using PSC-based therapies have 139

91 will provide important information on whether GBA only just begun (see summary in Table 1). The first 140

92 mutations alone are central in driving the progres- PSC-based trial for PD (ISCO trial) was initiated 141

93 sion of the disease in already diagnosed patients [7]. in Australia in 2016, using parthenogenetic PSC- 142

94 Other gene therapies are based on strategies to alter derived neural progenitor cells of a non-DA fate 143

95 neurotransmitter production, either through hijacking [13]. These cells are proposed to have a more 144
M. Tomishima and A. Kirkeby / The Scientist’s Perspective to PD Therapies 3

indirect mechanism of action, working potentially

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
145

EudraCT 2021-001366-38
through trophic support. However, multiple groups 146
Clinical data trial ID

have shown through genetic engineering in preclin- 147

UMIN000033564
ical models that it is specifically the midbrain DA 148

IND No. 17145


NCT02452723

NCT03119636

NCT04802733
neurons which are responsible for reverting symp- 149

toms in animal models of PD [14–16]. 150

Following this rationale, Jun Takahashi and col-


N/A

N/A

N/A

N/A

N/A
151
[29]

leagues launched the first PSC-based midbrain DA 152

f
neuron cell replacement in Japan in 2018 using an
Patients included∗

153

roo
allogenic iPSC approach [17, 18]. Just recently, a 154

collaborative effort between Weill Cornell, Memo-


(50)

(10)
(7)

(8)
12

155
1

rial Sloan Kettering, and BlueRock Therapeutics was 156

granted permission by the FDA to advance to Phase 157

1 clinical trial in the US with an hESC-based mid- 158

rP
Trial start

Est. 2021

Est. 2022

brain DA product (MSK-DA01) originally developed 159


2016

2017

2017

2018

by Lorenz Studer [19–21]. This trial is anticipated to 160

begin later this year. In an equivalent European effort, 161

Malin Parmar and colleagues have provided strong


[16, 19, 20, 44]
Pluripotent stem cell products in the clinical trial pipeline for PD

162
Preclin. data

[13, 38, 39]

[17, 42, 43]

[30, 45–50]

tho
proof of concept for their hESC-based product and are 163
[40]

[41]

aiming to begin clinical trials in the EU in 2022. These 164

groups working on PSC-based products have been 165

meeting annually since 2014 in the research-based 166


Sweden UK

network GForce-PD with the aim of sharing expe- 167


Australia
Trial site

Au
China

Japan
USA

USA

riences to bring the best treatments forward for the 168

PD community. Building on these academically led 169


Table 1

advancements, three additional companies, Sumit- 170


Trophic support

omo Dainippon Pharma, FujiFilm Cellular Dynamics 171


are not yet completed. Patient numbers without brackets are completed, transplanted patients.
DA-CRT

DA-CRT

DA-CRT

DA-CRT

DA-CRT

Inc. and Novo Nordisk have also announced the 172


MoA

development of PSC-based cell products for treat-


d

173

ment of PD. 174


cte

The trials mentioned above are based on allogeneic 175

cell transplantation strategies. Induced pluripotent


Immune matching

176
Allogeneic, HLA

Allogeneic, HLA

stem cell technology could provide an autologous


non-matched

non-matched

non-matched

non-matched

non-matched
matched and

matched and

177
Autologous
Allogeneic,

Allogeneic,

Allogeneic,

approach with the benefit of genetic matching at 178

the cost of time, logistic complexity and potential 179


rre

variability. For intracerebral therapies, it is not clear 180

that autologous approaches will be advantageous in 181


BlueRock Therapeutics

the immunoprivileged brain since fetal cell allografts 182


(Company/Institution)

Cambridge University
Weill Cornell/Memorial
International Stem Cell
Corporation (ISCO)

have demonstrated robust and long-lived survival, up


Chinese Academy of

183
Site of development

Sciences, Beijing

Harvard University

co
Sloan Kettering/

to 24 years without long term immune suppression


Kyoto University

Lund University/

184

[22–28]. Relying on autologous grafting could limit 185

the accessibility of stem cell therapies due to the cost 186

and the complication of batch-to-batch variation in 187


Un

differentiation efficacy. Nevertheless, one cannot dis- 188

count the possibility that the immune response to 189


STEM-PD (hESC-derived

grafted allogeneic cells can limit efficacy. A recent


(hESC-derived ventral
Human parthenogenetic

midbrain progenitors)
hESC-derived ventral

190
hESC-derived NSCs)

midbrain progenitors

midbrain progenitors

midbrain progenitors
hiPSC-derived ventral

hiPSC-derived ventral
ISC-hpNSC (Human

single patient case led by the group of Kwang-Soo 191


ventral midbrain
parthenogenetic

Kim at Harvard demonstrated proof of concept for the 192


progenitors)

autologous transplantation approach [29]. The Kim


Cell product

MSK-DA01

193

group, as well as the US-based company Aspen Neu- 194

roscience, is now pursuing further clinical trials using 195

autologous iPSCs for transplantation.


4 M. Tomishima and A. Kirkeby / The Scientist’s Perspective to PD Therapies

196 LOOKING TOWARDS THE FUTURE OF transplantation to achieve optimal clinical function 247

197 PSC-BASED CELL THERAPIES from cell replacement therapies. This makes the 248

choice of a clinical endpoint critical to assure the 249

198 It has taken around twenty years of intensive strongest clinical signal and reduced chance for 250

199 preclinical work to produce clinically acceptable placebo effects. Another key piece to the puzzle 251

200 pluripotent stem cell-derived human DA neurons that remains the patients themselves. PD has been increas- 252

201 function efficiently in PD animal models. While this ingly recognized as a syndrome with a range of 253

202 is great progress, much remains to be learned from clinical courses. Increasing knowledge of PD sub- 254

f
203 clinical trials to improve cell products further. Careful types and results from stem cell trials will allow us 255

roo
204 work by the Parmar lab [30] has shown equiva- to better understand which patients will truly benefit 256

205 lence between fetal cell and PSC-based dopamine from cell replacement therapies. 257

206 neurons in animal models of PD, but clinical data


207 will be crucial to assess this equivalence in patients. ACKNOWLEDGMENTS 258
208 Another unresolved question is whether transplanted

rP
209 DA neurons are “self-regulating.” That is, DA neu- The authors would like to thank Joachim Frue- 259
210 rons might receive feedback from the brain that bis and Seth Ettenberg for critical and constructive 260
211 naturally balances and limits dopamine release. This comments to the manuscript. 261
212 is important since excessive dopamine release might

tho
213 result in dyskinesias (involuntary movement) [31].
CONFLICT OF INTEREST 262
214 Understanding this biology in humans will inform
215 our strategy for patient dosing.
Mark Tomishima holds several patent agreements 263
216 Another factor affecting dose is the observation
and is an employee of BlueRock Therapeutics, a com- 264
217 that many neurons die after transplantation. It is not
pany that plans to commercially develop the DA01
Au 265
218 known why this occurs, and excess cells must be
product. Agnete Kirkeby holds several patent agree- 266
219 transplanted to account for this loss and assure ade-
ments and is a consultant for Novo Nordisk A/S on 267
220 quate dosing. The delivery device itself creates an
the development of the STEM-PD product. 268
221 injury and a localized immune reaction, and large
222 numbers of dead cells could alert the immune system
to the transplanted cells. Beyond just the transplan- REFERENCES
d

223 269

224 tation period itself, monitoring immune reactivity in


[1] Prothena (2020) https://ir.prothena.com/news-releases/ne 270
cte

225 allogeneic and autologous cell transplantation trials


ws-release-details/update-phase-2-pasadena-study- 271
226 will be key to understanding the role of the immune prasinezumab-prx002rg7935. 272
227 response to grafted cells and is crucial for future [2] Maiti P, Manna J, Dunbar GL (2017) Current understanding 273

228 cell therapy development. A better understanding of of the molecular mechanisms in Parkinson’s disease: Targets 274
for potential treatments. Transl Neurodegener 6, 28. 275
229 the immune response to grafts is needed to evalu- [3] McFarthing K, Larson D, Simuni T (2020) Clinical Trial
rre

276
230 ate whether allogeneic cell sources can survive and Highlights - GLP-1 agonists. J Parkinsons Dis 10, 355-368. 277
231 escape rejection longterm, or if the autologous cells [4] Grieco M, Giorgi A, Gentile MC, d’Erme M, Morano S, 278

232 will provide a clinical advantage despite the many Maras B, Filardi T (2019) Glucagon-like peptide-1: A focus 279
on neurodegenerative diseases. Front Neurosci 13, 1112. 280
233 hurdles associated with their manufacturing. Many [5] Marks WJ, Jr., Bartus RT, Siffert J, Davis CS, Lozano A, 281
co

234 groups are further looking into the possibilities of Boulis N, Vitek J, Stacy M, Turner D, Verhagen L, Bakay 282

235 using gene-edited allogeneic cell sources which can R, Watts R, Guthrie B, Jankovic J, Simpson R, Tagliati 283

236 escape immune recognition for future universal use in M, Alterman R, Stern M, Baltuch G, Starr PA, Larson 284
PS, Ostrem JL, Nutt J, Kieburtz K, Kordower JH, Olanow 285
237 patients to avoid the complexities and cost of making CW (2010) Gene delivery of AAV2-neurturin for Parkin-
Un

286
238 a personalized medicine [32–35]. Such cell lines must son’s disease: A double-blind, randomised, controlled trial. 287

239 however be used with caution since these cells might Lancet Neurol 9, 1164-1172. 288

240 inadvertently function as host cells to amplify viruses [6] Whone A, Luz M, Boca M, Woolley M, Mooney L, Dharia 289
S, Broadfoot J, Cronin D, Schroers C, Barua NU, Longpre 290
241 or permit uncontrolled cell growth. As such, incorpo- L, Barclay CL, Boiko C, Johnson GA, Fibiger HC, Harri- 291
242 ration of “suicide genes” into such engineered cells son R, Lewis O, Pritchard G, Howell M, Irving C, Johnson 292

243 would allow the clinical option to chemically ablate D, Kinch S, Marshall C, Lawrence AD, Blinder S, Sossi V, 293
Stoessl AJ, Skinner P, Mohr E, Gill SS (2019) Randomized 294
244 the grafted cells if required [36, 37].
trial of intermittent intraputamenal glial cell line-derived 295
245 One inconvenient truth learned from the fetal neurotrophic factor in Parkinson’s disease. Brain 142, 296
246 cell trials is that it can take several years after 512-525. 297
M. Tomishima and A. Kirkeby / The Scientist’s Perspective to PD Therapies 5

298 [7] McFarthing K, Buff S, Rafaloff G, Dominey T, Wyse RK, [21] Takahashi J (2021) Clinical trial for Parkinson’s disease gets 363
299 Stott SRW (2020) Parkinson’s disease drug therapies in the a green light in the US. Cell Stem Cell 28, 182-183. 364
300 clinical trial pipeline: 2020. J Parkinsons Dis 10, 757-774. [22] Li W, Englund E, Widner H, Mattsson B, van Westen D, Latt 365
301 [8] Niethammer M, Tang CC, LeWitt PA, Rezai AR, Leehey J, Rehncrona S, Brundin P, Bjorklund A, Lindvall O, Li JY 366
302 MA, Ojemann SG, Flaherty AW, Eskandar EN, Kostyk SK, (2016) Extensive graft-derived dopaminergic innervation is 367
303 Sarkar A, Siddiqui MS, Tatter SB, Schwalb JM, Poston KL, maintained 24 years after transplantation in the degenerating 368
304 Henderson JM, Kurlan RM, Richard IH, Sapan CV, Eidel- parkinsonian brain. Proc Natl Acad Sci U S A 113, 6544- 369
305 berg D, During MJ, Kaplitt MG, Feigin A (2017) Long-term 6549. 370
306 follow-up of a randomized AAV2-GAD gene therapy trial [23] Hallett PJ, Cooper O, Sadi D, Robertson H, Mendez I, 371
307 for Parkinson’s disease. JCI Insight 2, e90133. Isacson O (2014) Long-term health of dopaminergic neu- 372

f
308 [9] Palfi S, Gurruchaga JM, Lepetit H, Howard K, Ralph GS, ron transplants in Parkinson’s disease patients. Cell Rep 7, 373

roo
309 Mason S, Gouello G, Domenech P, Buttery PC, Hantraye P, 1755-1761. 374
310 Tuckwell NJ, Barker RA, Mitrophanous KA (2018) Long- [24] Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow 375
311 term follow-up of a Phase I/II Study of ProSavin, a lentiviral CW (2008) Lewy body-like pathology in long-term embry- 376
312 vector gene therapy for Parkinson’s disease. Hum Gene Ther onic nigral transplants in Parkinson’s disease. Nat Med 14, 377
313 Clin Dev 29, 148-155. 504-506. 378
314 [10] https://www.globenewswire.com/fr/news-release/2021/02/ [25] Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, 379

rP
315 02/2168590/0/en/Voyager-Therapeutics-Provides-Update- Lashley T, Quinn NP, Rehncrona S, Bjorklund A, Widner 380
316 on-NBIb-1817-VY-AADC-Gene-Therapy-Program.html. H, Revesz T, Lindvall O, Brundin P (2008) Lewy bodies in 381
317 [11] Kim TW, Koo SY, Studer L (2020) Pluripotent stem cell grafted neurons in subjects with Parkinson’s disease suggest 382
318 therapies for parkinson disease: Present challenges and host-to-graft disease propagation. Nat Med 14, 501-503. 383
319 future opportunities. Front Cell Dev Biol 8, 729. [26] Li JY, Englund E, Widner H, Rehncrona S, Bjorklund A, 384
320 [12] Barker RA, consortium T (2019) Designing stem-cell-based Lindvall O, Brundin P (2010) Characterization of Lewy 385

tho
321 dopamine cell replacement trials for Parkinson’s disease. body pathology in 12- and 16-year-old intrastriatal mes- 386
322 Nat Med 25, 1045-1053. encephalic grafts surviving in a patient with Parkinson’s 387
323 [13] Garitaonandia I, Gonzalez R, Christiansen-Weber T, disease. Mov Disord 25, 1091-1096. 388
324 Abramihina T, Poustovoitov M, Noskov A, Sherman G, [27] Mendez I, Vinuela A, Astradsson A, Mukhida K, Hallett P, 389
325 Semechkin A, Snyder E, Kern R (2016) Neural stem cell Robertson H, Tierney T, Holness R, Dagher A, Trojanowski 390
326 tumorigenicity and biodistribution assessment for Phase I JQ, Isacson O (2008) Dopamine neurons implanted into 391
Au
327 clinical trial in Parkinson’s disease. Sci Rep 6, 34478. people with Parkinson’s disease survive without pathology 392
328 [14] Aldrin-Kirk P, Heuer A, Wang G, Mattsson B, Lundblad for 14 years. Nat Med 14, 507-509. 393
329 M, Parmar M, Bjorklund T (2016) DREADD modulation [28] Freed CR, Zhou W, Breeze RE (2011) Dopamine cell 394
330 of transplanted DA neurons reveals a novel parkinsonian transplantation for Parkinson’s disease: The importance of 395
331 dyskinesia mechanism mediated by the serotonin 5-HT6 controlled clinical trials. Neurotherapeutics 8, 549-561. 396
332 receptor. Neuron 90, 955-968. [29] Schweitzer JS, Song B, Herrington TM, Park TY, Lee N, 397
333 [15] Chen Y, Xiong M, Dong Y, Haberman A, Cao J, Liu H, Zhou Ko S, Jeon J, Cha Y, Kim K, Li Q, Henchcliffe C, Kaplitt 398
d

334 W, Zhang SC (2016) Chemical control of grafted human M, Neff C, Rapalino O, Seo H, Lee IH, Kim J, Kim T, 399
335 PSC-Derived neurons in a mouse model of Parkinson’s dis- Petsko GA, Ritz J, Cohen BM, Kong SW, Leblanc P, Carter 400
cte

336 ease. Cell Stem Cell 18, 817-826. BS, Kim KS (2020) Personalized iPSC-derived dopamine 401
337 [16] Steinbeck JA, Choi SJ, Mrejeru A, Ganat Y, Deisseroth progenitor cells for Parkinson’s disease. N Engl J Med 382, 402
338 K, Sulzer D, Mosharov EV, Studer L (2015) Optogenet- 1926-1932. 403
339 ics enables functional analysis of human embryonic stem [30] Grealish S, Diguet E, Kirkeby A, Mattsson B, Heuer A, 404
340 cell-derived grafts in a Parkinson’s disease model. Nat Bramoulle Y, Van Camp N, Perrier AL, Hantraye P, Bjork- 405
341 Biotechnol 33, 204-209. lund A, Parmar M (2014) Human ESC-derived dopamine 406
rre

342 [17] Doi D, Magotani H, Kikuchi T, Ikeda M, Hiramatsu S, neurons show similar preclinical efficacy and potency to 407
343 Yoshida K, Amano N, Nomura M, Umekage M, Morizane fetal neurons when grafted in a rat model of Parkinson’s 408
344 A, Takahashi J (2020) Pre-clinical study of induced pluripo- disease. Cell Stem Cell 15, 653-665. 409
345 tent stem cell-derived dopaminergic progenitor cells for [31] Greene PE, Fahn S, Eidelberg D, Bjugstad KB, Breeze RE, 410
346 Parkinson’s disease. Nat Commun 11, 3369. Freed CR (2021) Persistent dyskinesias in patients with fetal 411
co

347 [18] Takahashi J (2020) iPS cell-based therapy for Parkinson’s tissue transplantation for Parkinson disease. NPJ Parkinsons 412
348 disease: A Kyoto trial. Regen Ther 13, 18-22. Dis 7, 38. 413
349 [19] Kim TW, Piao J, Koo SY, Kriks S, Chung SY, Betel D, [32] Deuse T, Hu X, Gravina A, Wang D, Tediashvili G, De C, 414
350 Socci ND, Choi SJ, Zabierowski S, Dubose BN, Hill EJ, Thayer WO, Wahl A, Garcia JV, Reichenspurner H, Davis 415
351 Mosharov EV, Irion S, Tomishima MJ, Tabar V, Studer L MM, Lanier LL, Schrepfer S (2019) Hypoimmunogenic 416
Un

352 (2021) Biphasic activation of WNT signaling facilitates the derivatives of induced pluripotent stem cells evade immune 417
353 derivation of midbrain dopamine neurons from hESCs for rejection in fully immunocompetent allogeneic recipients. 418
354 translational use. Cell Stem Cell 28, 343-355 e345. Nat Biotechnol 37, 252-258. 419
355 [20] Piao J, Zabierowski S, Dubose BN, Hill EJ, Navare M, [33] Gornalusse GG, Hirata RK, Funk SE, Riolobos L, Lopes 420
356 Claros N, Rosen S, Ramnarine K, Horn C, Fredrickson C, VS, Manske G, Prunkard D, Colunga AG, Hanafi LA, 421
357 Wong K, Safford B, Kriks S, El Maarouf A, Rutishauser U, Clegg DO, Turtle C, Russell DW (2017) HLA-E-expressing 422
358 Henchcliffe C, Wang Y, Riviere I, Mann S, Bermudez V, pluripotent stem cells escape allogeneic responses and lysis 423
359 Irion S, Studer L, Tomishima M, Tabar V (2021) Preclinical by NK cells. Nat Biotechnol 35, 765-772. 424
360 efficacy and safety of a human embryonic stem cell-derived [34] Han X, Wang M, Duan S, Franco PJ, Kenty JH, Hedrick 425
361 midbrain dopamine progenitor product, MSK-DA01. Cell P, Xia Y, Allen A, Ferreira LMR, Strominger JL, Melton 426
362 Stem Cell 28, 217-229 e217. DA, Meissner TB, Cowan CA (2019) Generation of 427
6 M. Tomishima and A. Kirkeby / The Scientist’s Perspective to PD Therapies

428 hypoimmunogenic human pluripotent stem cells. Proc Natl progenitors restore motor function in Parkinson’s disease 480
429 Acad Sci U S A 116, 10441-10446. models. J Clin Invest 130, 904-920. 481
430 [35] Xu H, Wang B, Ono M, Kagita A, Fujii K, Sasakawa N, Ueda [42] Morizane A, Kikuchi T, Hayashi T, Mizuma H, Takara S, 482
431 T, Gee P, Nishikawa M, Nomura M, Kitaoka F, Takahashi Doi H, Mawatari A, Glasser MF, Shiina T, Ishigaki H, Itoh 483
432 T, Okita K, Yoshida Y, Kaneko S, Hotta A (2019) Targeted Y, Okita K, Yamasaki E, Doi D, Onoe H, Ogasawara K, 484
433 disruption of HLA genes via CRISPR-Cas9 generates iPSCs Yamanaka S, Takahashi J (2017) MHC matching improves 485
434 with enhanced immune compatibility. Cell Stem Cell 24, engraftment of iPSC-derived neurons in non-human pri- 486
435 566-578 e567. mates. Nat Commun 8, 385. 487
436 [36] Ciceri F, Bonini C, Stanghellini MT, Bondanza A, Traver- [43] Kikuchi T, Morizane A, Doi D, Onoe H, Hayashi T, 488
437 sari C, Salomoni M, Turchetto L, Colombi S, Bernardi M, Kawasaki T, Saiki H, Miyamoto S, Takahashi J (2011) 489

f
438 Peccatori J, Pescarollo A, Servida P, Magnani Z, Perna SK, Survival of human induced pluripotent stem cell-derived 490

roo
439 Valtolina V, Crippa F, Callegaro L, Spoldi E, Crocchiolo R, midbrain dopaminergic neurons in the brain of a pri- 491
440 Fleischhauer K, Ponzoni M, Vago L, Rossini S, Santoro A, mate model of Parkinson’s disease. J Parkinsons Dis 1, 492
441 Todisco E, Apperley J, Olavarria E, Slavin S, Weissinger 395-412. 493
442 EM, Ganser A, Stadler M, Yannaki E, Fassas A, Anagnos- [44] Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, 494
443 topoulos A, Bregni M, Stampino CG, Bruzzi P, Bordignon Carrillo-Reid L, Auyeung G, Antonacci C, Buch A, Yang 495
444 C (2009) Infusion of suicide-gene-engineered donor lym- L, Beal MF, Surmeier DJ, Kordower JH, Tabar V, Studer 496

rP
445 phocytes after family haploidentical haemopoietic stem-cell L (2011) Dopamine neurons derived from human ES cells 497
446 transplantation for leukaemia (the TK007 trial): A non- efficiently engraft in animal models of Parkinson’s disease. 498
447 randomised phase I-II study. Lancet Oncol 10, 489-500. Nature 480, 547-551. 499
448 [37] Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser [45] Grealish S, Heuer A, Cardoso T, Kirkeby A, Jonsson M, 500
449 A, Martinez C, Straathof K, Liu E, Durett AG, Grilley B, Johansson J, Bjorklund A, Jakobsson J, Parmar M (2015) 501
450 Liu H, Cruz CR, Savoldo B, Gee AP, Schindler J, Krance Monosynaptic tracing using modified rabies virus reveals 502

tho
451 RA, Heslop HE, Spencer DM, Rooney CM, Brenner MK early and extensive circuit integration of human embryonic 503
452 (2011) Inducible apoptosis as a safety switch for adoptive stem cell-derived neurons. Stem Cell Rep 4, 975-983. 504
453 cell therapy. N Engl J Med 365, 1673-1683. [46] Kirkeby A, Nolbrant S, Tiklova K, Heuer A, Kee N, Cardoso 505
454 [38] Gonzalez R, Garitaonandia I, Poustovoitov M, Abramihina T, Ottosson DR, Lelos MJ, Rifes P, Dunnett SB, Grealish S, 506
455 T, McEntire C, Culp B, Attwood J, Noskov A, Christiansen- Perlmann T, Parmar M (2017) Predictive markers guide dif- 507
456 Weber T, Khater M, Mora-Castilla S, To C, Crain A, ferentiation to improve graft outcome in clinical translation 508
Au
457 Sherman G, Semechkin A, Laurent LC, Elsworth JD, Sladek of hESC-based therapy for Parkinson’s disease. Cell Stem 509
458 J, Snyder EY, Redmond DE, Jr., Kern RA (2016) Neural Cell 20, 135-148. 510
459 stem cells derived from human parthenogenetic stem cells [47] Nolbrant S, Heuer A, Parmar M, Kirkeby A (2017) Gener- 511
460 engraft and promote recovery in a nonhuman primate model ation of high-purity human ventral midbrain dopaminergic 512
461 of Parkinson’s disease. Cell Transplant 25, 1945-1966. progenitors for in vitro maturation and intracerebral trans- 513
462 [39] Gonzalez R, Garitaonandia I, Crain A, Poustovoitov M, plantation. Nat Protoc 12, 1962-1979. 514
463 Abramihina T, Noskov A, Jiang C, Morey R, Laurent LC, [48] Cardoso T, Adler AF, Mattsson B, Hoban DB, Nolbrant S, 515
d

464 Elsworth JD, Snyder EY, Redmond DE, Jr., Semechkin R Wahlestedt JN, Kirkeby A, Grealish S, Bjorklund A, Parmar 516
465 (2015) Proof of concept studies exploring the safety and M (2018) Target-specific forebrain projections and appro- 517
cte

466 functional activity of human parthenogenetic-derived neu- priate synaptic inputs of hESC-derived dopamine neurons 518
467 ral stem cells for the treatment of Parkinson’s disease. Cell grafted to the midbrain of parkinsonian rats. J Comp Neurol 519
468 Transplant 24, 681-690. 526, 2133-2146. 520
469 [40] Wang YK, Zhu WW, Wu MH, Wu YH, Liu ZX, Liang [49] Adler AF, Cardoso T, Nolbrant S, Mattsson B, Hoban DB, 521
470 LM, Sheng C, Hao J, Wang L, Li W, Zhou Q, Hu BY Jarl U, Wahlestedt JN, Grealish S, Bjorklund A, Parmar 522
471 (2018) Human clinical-grade parthenogenetic ESC-derived M (2019) hESC-derived dopaminergic transplants integrate 523
rre

472 dopaminergic neurons recover locomotive defects of non- into basal ganglia circuitry in a preclinical model of Parkin- 524
473 human primate models of Parkinson’s disease. Stem Cell son’s disease. Cell Rep 28, 3462-3473 e3465. 525
474 Rep 11, 171-182. [50] Tiklova K, Nolbrant S, Fiorenzano A, Bjorklund AK, 526
475 [41] Song B, Cha Y, Ko S, Jeon J, Lee N, Seo H, Park KJ, Lee Sharma Y, Heuer A, Gillberg L, Hoban DB, Cardoso T, 527
476 IH, Lopes C, Feitosa M, Luna MJ, Jung JH, Kim J, Hwang Adler AF, Birtele M, Lunden-Miguel H, Volakakis N, 528
co

477 D, Cohen BM, Teicher MH, Leblanc P, Carter BS, Kor- Kirkeby A, Perlmann T, Parmar M (2020) Single cell tran- 529
478 dower JH, Bolshakov VY, Kong SW, Schweitzer JS, Kim scriptomics identifies stem cell-derived graft composition 530
479 KS (2020) Human autologous iPSC-derived dopaminergic in a model of Parkinson’s disease. Nat Commun 11, 2434. 531
Un

You might also like