Skip to main content
Brain Circulation logoLink to Brain Circulation
. 2016 Oct 18;2(3):141–145. doi: 10.4103/2394-8108.192524

Nonhuman primate model in clinical modeling of diseases for stem cell therapy

Gourav R Choudhury 1, Jeffrey Kim 1, Patrice A Frost 1, Raul A Bastarrachea 1, Marcel M Daadi 1,2,3,
PMCID: PMC6126269  PMID: 30276291

Abstract

Nonhuman primates (NHPs) are alike humans in size, behavior, physiology, biochemistry, and immunology. Given close similarities to humans, the NHP model offers exceptional opportunities to understand the biological mechanisms and translational applications with direct relevance to human conditions. Here, we evaluate the opportunities and limitations of NHPs as animal models for translational regenerative medicine. NHP models of human disease propose exceptional opportunities to advance stem cell-based therapy by addressing pertinent translational concerns related to this research. Nonetheless, the value of these primates must be carefully assessed, taking into account the expense of specialized equipment and requirement of highly specialized staff. Well-designed initial fundamental studies in small animal models are essential before translating research into NHP models and eventually into human trials. In addition, we suggest that applying a directed and collaborative approach, as seen in the evolution of stroke NHP models, will greatly benefit the translation of stem cell therapy in other NHP disease models.

Keywords: Myocardial infarction, nonhuman primate models, Parkinson's disease, stem cell therapy, STEPS, stroke, translational studies, Type-1 diabetes

Introduction

Human and nonhuman primates (NHPs) have many similar attributes, such as behavior, physiology, anatomy, biochemistry, organ mechanisms, and immunology.[1]

Research suggests that NHP animal models possess the capability to link the translational research between small animal models and humans. NHP models of human disease propose exceptional opportunities to advance stem cell-based therapy by addressing pertinent translational concerns associated with this research. These translational aspects include the application of autologous/allogeneic-induced pluripotent stem cell (iPSC)-derived cellular products, concerns related to the immune response, delivery techniques in a clinical setting, and the evaluation of candidate cell line profiles when transplanted. NHP models offer unique opportunities to evaluate the complexity of the biochemical, physiological, behavioral, and imaging end points that are pertinent to current human conditions. Given the expense of specialized equipment and requirement of a highly specialized staff, the value of using these primates must be carefully assessed. Well-designed, less resource demanding, initial fundamental studies in small animal models, such as rodents, are essential before translating research into NHP models and ultimately into human trials. In this current report, we suggest that a robust dialog within each disease-specific research community focused on the development of relevant NHP models will greatly benefit the advance of the translation of stem cell research.

Nonhuman Primate Disease Models for Stem Cell-Based Therapy

One of the most relevant NHP models is the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced NHP model of Parkinson's disease (PD). It provides significant preclinical opportunities that will ultimately demonstrate the safety and efficacy necessary to translate novel treatments into human trials. Like in humans,[2,3] MPTP-treated NHPs exhibit most difficult symptoms of sporadic PD, such as static tremor,[4] that can only be recognized in certain PD monkey models. In addition, MPTP injections in NHPs result in side effects to anti-parkinsonian drugs as experienced by humans with PD, such as dyskinesias with Levodopa Pharmacotherapy,[5] cumulate the major constituent, α-synuclein, of Lewy bodies, the pathological trademark of PD,[6] and exhibit cognitive disruptions.[7] NHP models of PD have already been instrumental to our understanding of the differences in autologous versus allogeneic PSC-based therapy, their ability to innervate the putamen, and the appropriate differentiation profile of dopaminergic neurons.[8,9,10,11,12,13,14]

Numerous research facilities have focused on various stem cell-based regenerative therapies for PD. This will yield important translational data to prove the potential of this therapy and recognize the possible clinical obstacles that must be addressed. As seen in Phase I studies, PD patients receiving neural grafts have shown an increase in their quality of life,[15,16] suggesting proof of the concept. However, the next hurdle to overcome remains the challenge of developing a predictable delivery system and sustainable stem cell populations that adhere to strict regulatory measures. Proven safety and efficacy of the therapy in NHP will effectively translate the product to commercial use and provide way to the development of a standardized treatment.

Type-1 diabetes mellitus (T1D) is an autoimmune disease that results in the permanent destruction of a specific cell type, the β-cells of the pancreatic islets of Langerhans; therefore, it represents an ideal candidate for cell replacement therapy. Indeed, islet transplantation started in 1894 with much improvement in the 1990s[17] with a landmark paper reporting the insulin therapy independence in seven out of seven T1D patients.[18] Clinical centers with experience in the transplantation procedure achieved 80% insulin independence in 80% of patients during the 1st year posttransplantation.[19] However, there was a high incidence of failure in long-term associated mainly with poor pancreatic graft and complications from permanent immunosuppression. Other limitations and challenges within the current approach are the reliance on organ donations, tissues processing for isolating quality-controlled islet cells, lifelong immunosuppression, and the unreasonable long waiting list for patients. Thus, stem cell-based therapeutic strategies may eliminate or lower the severity of immunosuppression through autologous or stem cell microencapsulation, both promising approaches for T1D cell therapy. In the cell microencapsulation strategy, insulin-producing cells are loaded into a semipermeable capsule made from biodegradable materials that can allow for the secretion of insulin while providing the cells with nutrients and protection from the host immune system.[20,21]

A relevant NHP model to address these clinical outcome concerns is crucial. A NHP model of T1D, the streptozotocin (STZ) NHP model of T1D, has been developed.[22] In this animal model, arterial and venous tether catheters enable continuous measurement of glucose, C-peptide, and other various biochemical measurements. STZ is administered intravenously to ablate the pancreatic β-cells and to induce diabetes. In this model, a comprehensive therapeutic strategy was implemented including a tether system, permanent indwelling catheter implants, an optimal hydration protocol, and pain and anxiety management. Glucose levels post-STZ administration were monitored moment by moment with continuous intravenous insulin therapy. Among the advantages of the STZ NHP model of T1D is that clinically relevant doses of stem cells and routes of delivery may be tested.

A recent study in this STZ-baboon model used an ultrasound-targeted microbubble destruction approach, in which nonviral gene therapy was targeted to pancreatic islets. STZ hyperglycemic-induced conscious tethered baboons received a gene cocktail comprising cyclin D2, CDK, and GLP1, which normalized intravenous glucose tolerance test curves. Immunohistochemistry demonstrated evidence of islet regeneration and restoration of β-cell mass.[22] STZ targets GLUT2 glucose transporter on β-cells and ablates them through the release of reactive oxygen species and alkylation of DNA.[23] Despite the ability to destroy β-cells, STZ is not necessarily representative of the autoimmune condition seen in T1D.[24] In this regard, transgenesis in NHPs[25,26] may play an important role in developing relevant models that closely mimic the autoimmune component of T1D.

Human pluripotent stem cells have indisputable cardiomyocyte (CM)-generating abilities and have been extensively investigated for repair of the injured heart. Studies of human embryonic stem cell-derived CMs (hESC-CMs) in small animal models have shown favorable effects of stem cell therapy. The human heart has a limited capacity for regeneration after injury. NHP models of myocardial infarction are well established and essential for addressing critical preclinical questions before the initiation of clinical trials. Myocardial infarction is induced in NHP using a coronary catheter that engages the main coronary artery, a guide wire, and an angioplasty balloon. The angioplasty balloon is inserted into the anterior descending artery and inflated to block circulation for a predetermined period, usually 90 min followed by reperfusion. This animal model has been used to evaluate hESC-CMs and reprogramed somatic cells (iPSC) for their regenerative capacity of the heart.[27,28,29]

The characterization of an NHP model of focal ischemia reperfusion with a defined syndrome, impaired arm function, and finger dexterity in the context of the current NHP models has recently been reported.[30] In this review, we take the opportunity to address another relevant model, that is the transient global ischemia (TGI).[31,32] Noting its difference from the clinical condition of brain ischemia, the induction of the TGI model may be a more appropriate model for cardiac arrest. However, the cell death events that precede an initial blood flow disruption are indicative of stroke-induced cell loss in distinct brain regions, particularly the hippocampus. Furthermore, the behavioral deficits evaluated in TGI animal models, such as memory decline, mirror the cognitive impairment assessed in clinical stroke patients.[33] The NHP model offers numerous opportunities, particularly the applicability to cerebral ischemia pathology.

NHP models can also offer important information for stroke rehabilitation that complement findings from rodent models. In a recent set of guidelines, the Stroke Therapy Academic Industry Roundtable (STAIR) has emphasized the necessity for NHP models of stroke for preclinical progress of neuroprotective therapies.[34] These guidelines were prompted by effective results in the laboratory that failed to produce beneficial results in the clinic. Certain features of stroke are consistent only with humans and NHPs, including the subcortical white matter injury and the internal capsule where the descending cortical pathways are found. Likewise, rats do not display the advanced development of the direct cortico-motorneuronal pathway, which dictates the precise motor skills impacted by stroke. Recent studies have described NHP models of stroke with excellent imaging, behavioral, and pathophysiological characterization[35,36,37,38,39,40,41] and have demonstrated the value of these models in preclinical advancement of therapeutic mediation.[42,43,44] The goal of NHP models is to accurately predict results, identify challenges, and increase the probability of successful therapy in human clinical trials. Various NHP species have demonstrated and validated the presence of ischemic neuronal loss in discrete regions of the brain after focal or global ischemia, and thus they would be crucial in understanding the pathophysiology of stroke and in stem cell translational research.

Guidelines Proposed to Enforce Uniformity and Advance Further Research

Stem cell Therapeutics as an Emerging Paradigm for Stroke (STEPS)[45,46,47,48] has established guidelines that will provide the foundation for investigating the translation of cell-based therapeutics from the laboratory to the clinic by using animal stroke models. As emphasized in the STEPS, testing the potential of anti-stroke therapeutics is critically dependent on the appropriate use of species and applicable stroke animal models, such as NHPs. While larger animals are favorable, the lack of a reputable NHP stroke model leads to an emphasis on safety rather than efficacy as an appropriate outcome measure. Although the STEPS welcomes different, available types of surgical methods, it highlights the importance of the “end point” rather than the “technique employed” to produce the stroke. The conditions of the stroke model should mirror the human disease state very closely to improve the success of translating therapeutics from the laboratory to the clinic.[46,47]

A standard-of-care therapy (i.e., tissue plasminogen activator (tPA), rehabilitation therapy) must be established as a control group for translational studies. Currently, small animals such as rodents and rabbits receive such “best in class” control, yet accommodating this treatment to NHPs remains a challenge. The major obstacle in extending such control group to NHPs is the difficulty of standardizing the tPA treatment and rehabilitation therapy in these larger animal models. A solution to this would require NHP research personnel with expertise in this specific treatment and therapy.

Consideration must be given to the experimental therapeutic mechanism of action (MOA) in choosing the potential therapeutic windows for the various species utilized for stroke modeling. Subsequently, the stroke model and the species will be chosen that are deemed suitable to examine the MOA. For example, if the neuroprotective stage is targeted, the therapeutic window is expected to be during the acute stage of stroke. On the other hand, if the therapeutic MOA targets the neurorestorative stage instead, the window will most likely be within the subacute and chronic stages of the stroke. Small species have generally been utilized for examining therapeutic windows ranging from acute, subacute, and chronic stages of stroke. The use of smaller species can be attributed to the much less laborious postoperative care. Similarly, to humans and depending on the extent of the lesion, larger animal models of stroke, including NHPs, may require extensive postoperative monitoring and care. In addition, further studies are required to validate the standardization of NHP stroke models, particularly the chronic stage. This may restrict their use in examining therapeutic windows during the acute and subacute stages of stroke.

The NIH's National Institute of Neurological Disorders and Stroke has advanced the integration of RIGOR guidelines for translational research, along with recommendations by STAIR and STEPS, to standardize stroke research procedures worldwide. The STAIR, STEPS, and RIGOR guidelines are similar in highlighting the importance of good laboratory practices (GLPs), such as the requirement for all animal modeling studies to uphold treatment conditions such as blinding, randomization, consideration of sex and age variables, and complete power and statistical analysis. In addition, when proposing translational applications and manuscripts, one must clearly present these GLP practices.[48] Translational Stroke Research and the Journal of Neurology and Neurophysiology have recognized this policy in their submissions. Although many animal models exist, finding optimal animal models is key to enhance the successful translation of novel therapeutics from the laboratory to the clinic. Equally significant to this is asserting a transparency of any person's conflict of interest that may possibly suggest bias in the study.[49]

Future Direction for Nonhuman Primate Models in Translational Medicine

Given their close similarities to humans, the NHP model offers unique opportunities to understand biological mechanisms and translational applications with direct relevance to human conditions. Still, the value of using these primates must be carefully assessed, taking into account the expense of specialized equipment and requirement of highly specialized personnel. As such, carrying out initial fundamental studies in small animal models such as rodents may prove to be more efficient before translating such research to NHP models and eventually to first-in-human trials.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

  • 1.Bluemel J, Korte S, Schenck E, Weinbauer G. San Diego, CA: Academic Press; 2015. The Nonhuman Primate in Nonclinical Drug Development and Safety Assessment. [Google Scholar]
  • 2.Davis GC, Williams AC, Markey SP, Ebert MH, Caine ED, Reichert CM, et al. Chronic Parkinsonism secondary to intravenous injection of meperidine analogues. Psychiatry Res. 1979;1:249–54. doi: 10.1016/0165-1781(79)90006-4. [DOI] [PubMed] [Google Scholar]
  • 3.Langston JW, Ballard P, Tetrud JW, Irwin I. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science. 1983;219:979–80. doi: 10.1126/science.6823561. [DOI] [PubMed] [Google Scholar]
  • 4.Bergman H, Raz A, Feingold A, Nini A, Nelken I, Hansel D, et al. Physiology of MPTP tremor. Mov Disord. 1998;13(Suppl 3):29–34. doi: 10.1002/mds.870131305. [DOI] [PubMed] [Google Scholar]
  • 5.Bédard PJ, Di Paolo T, Falardeau P, Boucher R. Chronic treatment with L-DOPA, but not bromocriptine induces dyskinesia in MPTP-parkinsonian monkeys. Correlation with [3H] spiperone binding. Brain Res. 1986;379:294–9. doi: 10.1016/0006-8993(86)90783-3. [DOI] [PubMed] [Google Scholar]
  • 6.Purisai MG, McCormack AL, Langston WJ, Johnston LC, Di Monte DA. Alpha-synuclein expression in the substantia nigra of MPTP-lesioned non-human primates. Neurobiol Dis. 2005;20:898–906. doi: 10.1016/j.nbd.2005.05.028. [DOI] [PubMed] [Google Scholar]
  • 7.Schneider JS, Kovelowski CJ., 2nd Chronic exposure to low doses of MPTP. I. Cognitive deficits in motor asymptomatic monkeys. Brain Res. 1990;519:122–8. doi: 10.1016/0006-8993(90)90069-n. [DOI] [PubMed] [Google Scholar]
  • 8.Emborg ME, Liu Y, Xi J, Zhang X, Yin Y, Lu J, et al. Induced pluripotent stem cell-derived neural cells survive and mature in the nonhuman primate brain. Cell Rep. 2013;3:646–50. doi: 10.1016/j.celrep.2013.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Daadi MM, Grueter BA, Malenka RC, Redmond DE, Jr, Steinberg GK. Dopaminergic neurons from midbrain-specified human embryonic stem cell-derived neural stem cells engrafted in a monkey model of Parkinson's disease. PLoS One. 2012;7:e41120. doi: 10.1371/journal.pone.0041120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Takagi Y, Takahashi J, Saiki H, Morizane A, Hayashi T, Kishi Y, et al. Dopaminergic neurons generated from monkey embryonic stem cells function in a Parkinson primate model. J Clin Invest. 2005;115:102–9. doi: 10.1172/JCI21137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, et al. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature. 2011;480:547–51. doi: 10.1038/nature10648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Morizane A, Doi D, Kikuchi T, Okita K, Hotta A, Kawasaki T, et al. Direct comparison of autologous and allogeneic transplantation of iPSC-derived neural cells in the brain of a non-human primate. Stem Cell Reports. 2013;1:283–92. doi: 10.1016/j.stemcr.2013.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Doi D, Morizane A, Kikuchi T, Onoe H, Hayashi T, Kawasaki T, et al. Prolonged maturation culture favors a reduction in the tumorigenicity and the dopaminergic function of human ESC-derived neural cells in a primate model of Parkinson's disease. Stem Cells. 2012;30:935–45. doi: 10.1002/stem.1060. [DOI] [PubMed] [Google Scholar]
  • 14.Emborg ME, Zhang Z, Joers V, Brunner K, Bondarenko V, Ohshima S, et al. Intracerebral transplantation of differentiated human embryonic stem cells to hemiparkinsonian monkeys. Cell Transplant. 2013;22:831–8. doi: 10.3727/096368912X647144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kefalopoulou Z, Politis M, Piccini P, Mencacci N, Bhatia K, Jahanshahi M, et al. Long-term clinical outcome of fetal cell transplantation for Parkinson disease: Two case reports. JAMA Neurol. 2014;71:83–7. doi: 10.1001/jamaneurol.2013.4749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mendez I, Viñuela A, Astradsson A, Mukhida K, Hallett P, Robertson H, et al. Dopamine neurons implanted into people with Parkinson's disease survive without pathology for 14 years. Nat Med. 2008;14:507–9. doi: 10.1038/nm1752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ricordi C, Strom TB. Clinical islet transplantation: Advances and immunological challenges. Nat Rev Immunol. 2004;4:259–68. doi: 10.1038/nri1332. [DOI] [PubMed] [Google Scholar]
  • 18.Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343:230–8. doi: 10.1056/NEJM200007273430401. [DOI] [PubMed] [Google Scholar]
  • 19.Shapiro AM, Ricordi C, Hering B. Edmonton's islet success has indeed been replicated elsewhere. Lancet. 2003;362:1242. doi: 10.1016/S0140-6736(03)14526-6. [DOI] [PubMed] [Google Scholar]
  • 20.Agulnick AD, Ambruzs DM, Moorman MA, Bhoumik A, Cesario RM, Payne JK, et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo . Stem Cells Transl Med. 2015;4:1214–22. doi: 10.5966/sctm.2015-0079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Vegas AJ, Veiseh O, Gürtler M, Millman JR, Pagliuca FW, Bader AR, et al. Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat Med. 2016;22:306–11. doi: 10.1038/nm.4030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Frost PA, Chen S, Mezzles MJ, Voruganti VS, Nava-Gonzalez EJ, Arriaga-Cazares HE, et al. Successful pharmaceutical-grade streptozotocin (STZ)-induced hyperglycemia in a conscious tethered baboon (Papio hamadryas) model. J Med Primatol. 2015;44:202–17. doi: 10.1111/jmp.12182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Szkudelski T. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res. 2001;50:537–46. [PubMed] [Google Scholar]
  • 24.Mordes JP, Bortell R, Blankenhorn EP, Rossini AA, Greiner DL. Rat models of type 1 diabetes: Genetics, environment, and autoimmunity. ILAR J. 2004;45:278–91. doi: 10.1093/ilar.45.3.278. [DOI] [PubMed] [Google Scholar]
  • 25.Chan AW, Chong KY, Martinovich C, Simerly C, Schatten G. Transgenic monkeys produced by retroviral gene transfer into mature oocytes. Science. 2001;291:309–12. doi: 10.1126/science.291.5502.309. [DOI] [PubMed] [Google Scholar]
  • 26.Wolfgang MJ, Golos TG. Nonhuman primate transgenesis: Progress and prospects. Trends Biotechnol. 2002;20:479–84. doi: 10.1016/s0167-7799(02)02052-8. [DOI] [PubMed] [Google Scholar]
  • 27.Shiba Y, Fernandes S, Zhu WZ, Filice D, Muskheli V, Kim J, et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature. 2012;489:322–5. doi: 10.1038/nature11317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Blin G, Nury D, Stefanovic S, Neri T, Guillevic O, Brinon B, et al. A purified population of multipotent cardiovascular progenitors derived from primate pluripotent stem cells engrafts in postmyocardial infarcted nonhuman primates. J Clin Invest. 2010;120:1125–39. doi: 10.1172/JCI40120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chong JJ, Yang X, Don CW, Minami E, Liu YW, Weyers JJ, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature. 2014;510:273–7. doi: 10.1038/nature13233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.McEntire CR, Choudhury GR, Torres A, Steinberg GK, Redmond DE, Jr, Daadi MM. Impaired arm function and finger dexterity in a nonhuman primate model of stroke: Motor and cognitive assessments. Stroke. 2016;47:1109–16. doi: 10.1161/STROKEAHA.115.012506. [DOI] [PubMed] [Google Scholar]
  • 31.Tabuchi E, Endo S, Ono T, Nishijo H, Kuze S, Kogure K. Hippocampal neuronal damage after transient forebrain ischemia in monkeys. Brain Res Bull. 1992;29:685–90. doi: 10.1016/0361-9230(92)90139-o. [DOI] [PubMed] [Google Scholar]
  • 32.Hara K, Yasuhara T, Matsukawa N, Maki M, Masuda T, Yu G, et al. Hippocampal CA1 cell loss in a non-human primate model of transient global ischemia: A pilot study. Brain Res Bull. 2007;74:164–71. doi: 10.1016/j.brainresbull.2007.06.014. [DOI] [PubMed] [Google Scholar]
  • 33.Yukie M, Yamaguchi K, Yamashima T. Impairments in recognition memory for object and for location after transient brain ischemia in monkeys. Rev Neurosci. 2006;17:201–14. doi: 10.1515/revneuro.2006.17.1-2.201. [DOI] [PubMed] [Google Scholar]
  • 34.Stroke Therapy Academic Industry Roundtable (STAIR). Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke. 1999;30:2752–8. doi: 10.1161/01.str.30.12.2752. [DOI] [PubMed] [Google Scholar]
  • 35.Marshall JW, Ridley RM. Assessment of cognitive and motor deficits in a marmoset model of stroke. ILAR J. 2003;44:153–60. doi: 10.1093/ilar.44.2.153. [DOI] [PubMed] [Google Scholar]
  • 36.Freret T, Bouet V, Toutain J, Saulnier R, Pro-Sistiaga P, Bihel E, et al. Intraluminal thread model of focal stroke in the non-human primate. J Cereb Blood Flow Metab. 2008;28:786–96. doi: 10.1038/sj.jcbfm.9600575. [DOI] [PubMed] [Google Scholar]
  • 37.Fukuda S, del Zoppo GJ. Models of focal cerebral ischemia in the nonhuman primate. ILAR J. 2003;44:96–104. doi: 10.1093/ilar.44.2.96. [DOI] [PubMed] [Google Scholar]
  • 38.Nudo RJ, Larson D, Plautz EJ, Friel KM, Barbay S, Frost SB. A squirrel monkey model of poststroke motor recovery. ILAR J. 2003;44:161–74. doi: 10.1093/ilar.44.2.161. [DOI] [PubMed] [Google Scholar]
  • 39.Huang J, Mocco J, Choudhri TF, Poisik A, Popilskis SJ, Emerson R, et al. A modified transorbital baboon model of reperfused stroke. Stroke. 2000;31:3054–63. doi: 10.1161/01.str.31.12.3054. [DOI] [PubMed] [Google Scholar]
  • 40.Kuge Y, Yokota C, Tagaya M, Hasegawa Y, Nishimura A, Kito G, et al. Serial changes in cerebral blood flow and flow-metabolism uncoupling in primates with acute thromboembolic stroke. J Cereb Blood Flow Metab. 2001;21:202–10. doi: 10.1097/00004647-200103000-00003. [DOI] [PubMed] [Google Scholar]
  • 41.Virley D, Hadingham SJ, Roberts JC, Farnfield B, Elliott H, Whelan G, et al. A new primate model of focal stroke: Endothelin-1-induced middle cerebral artery occlusion and reperfusion in the common marmoset. J Cereb Blood Flow Metab. 2004;24:24–41. doi: 10.1097/01.WCB.0000095801.98378.4A. [DOI] [PubMed] [Google Scholar]
  • 42.Bahjat FR, Williams-Karnesky RL, Kohama SG, West GA, Doyle KP, Spector MD, et al. Proof of concept: Pharmacological preconditioning with a Toll-like receptor agonist protects against cerebrovascular injury in a primate model of stroke. J Cereb Blood Flow Metab. 2011;31:1229–42. doi: 10.1038/jcbfm.2011.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cook DJ, Teves L, Tymianski M. Treatment of stroke with a PSD-95 inhibitor in the gyrencephalic primate brain. Nature. 2012;483:213–7. doi: 10.1038/nature10841. [DOI] [PubMed] [Google Scholar]
  • 44.Roitberg BZ, Mangubat E, Chen EY, Sugaya K, Thulborn KR, Kordower JH, et al. Survival and early differentiation of human neural stem cells transplanted in a nonhuman primate model of stroke. J Neurosurg. 2006;105:96–102. doi: 10.3171/jns.2006.105.1.96. [DOI] [PubMed] [Google Scholar]
  • 45.Borlongan CV. Cell therapy for stroke: Remaining issues to address before embarking on clinical trials. Stroke. 2009;40(3 Suppl):S146–8. doi: 10.1161/STROKEAHA.108.533091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stem Cell Therapies as an Emerging Paradigm in Stroke Participants. Stem Cell Therapies as an Emerging Paradigm in Stroke (STEPS): Bridging basic and clinical science for cellular and neurogenic factor therapy in treating stroke. Stroke. 2009;40:510–5. doi: 10.1161/STROKEAHA.108.526863. [DOI] [PubMed] [Google Scholar]
  • 47.Chopp M, Steinberg GK, Kondziolka D, Lu M, Bliss TM, Li Y, et al. Who's in favor of translational cell therapy for stroke: STEPS forward please? Cell Transplant. 2009;18:691–3. doi: 10.3727/096368909X470883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Borlongan CV, Chopp M, Steinberg GK, Bliss TM, Li Y, Lu M, et al. Potential of stem/progenitor cells in treating stroke: The missing steps in translating cell therapy from laboratory to clinic. Regen Med. 2008;3:249–50. doi: 10.2217/17460751.3.3.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lapchak PA. Scientific rigor recommendations for optimizing the clinical applicability of translational research. J Neurol Neurophysiol. 2012;3:pii: e111. doi: 10.4172/2155-9562.1000e111. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Brain Circulation are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES