Journal List > Yonsei Med J > v.58(2) > 1032109

Chang, Ahn, Sung, and Park: Stem Cell Therapy for Neonatal Disorders: Prospects and Challenges

Abstract

Despite recent advances in neonatal medicine, neonatal disorders, such as bronchopulmonary dysplasia and intraventricular hemorrhage in preterm neonates and hypoxic ischemic encephalopathy in term neonates, remain major causes of mortality and morbidities. Promising preclinical research results suggest that stem cell therapies represent the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies. This review focuses primarily on the potential role of stem cell therapy in the above mentioned neonatal disorders, highlighting the results of human clinical trials and the challenges that remain to be addressed for their safe and successful translation into clinical care of newborn infants.

INTRODUCTION

Despite recent advances in neonatal intensive care medicine, several neonatal disorders, including bronchopulmonary dysplasia (BPD)12 and severe intraventricular hemorrhage (IVH)3 in preterm neonates and hypoxic ischemic encephalopathy (HIE)4 in borderline preterm and term neonates, remain major causes of mortality and morbidities. Few effective therapies are currently available to ameliorate the injuries resulting from these disorders. Therefore, developing new safe and effective therapies to improve the outcomes of these intractable and devastating neonatal disorders is an urgent and considerable issue.
Recently, various preclinical studies have shown that stem cell therapy significantly attenuates injuries in newborn animal models of BPD,5 HIE,6 and IVH.7 Phase I clinical trials conducted in neonates with BPD,8 HIE,9 or severe IVH (NCT02274428) have shown stem cell treatments to be safe, feasible, and potentially efficacious. Overall, these findings suggest that stem cell therapy might represent the next breakthrough in therapy of these currently intractable and devastating neonatal disorders with complex multifactorial etiologies. This review summarizes recent advances in stem cell research for treating neonatal disorders of BPD, HIE, and IVH. We focus on the results of human clinical trials and the challenges, including safety and delivery of the optimal cells for the right patient with the right route at the right time and dose, that remain for the safe and successful translation of stem cell therapies into clinical care of newborn infants.

CLINICAL TRIALS DATA

Phase I clinical trial for BPD

Based on preclinical evidence showing the protective effects of mesenchymal stem cell (MSC) transplantation against neonatal hyperoxic lung injuries without short- and long-term toxicity or tumorigenicity,510111213 the first pioneering clinical study in preterm infants at risk of BPD has been conducted.8 In an open-label, single-center, phase I, dose-escalation clinical trial, Chang, et al.5 assessed the safety and feasibility of a single intratracheal (IT) transplantation of allogenic human umbilical cord blood (UCB)-derived MSCs for BPD. The MSCs were transplanted into nine very preterm infants at high risk for developing BPD, with a mean gestational age of 25 weeks (range: 24–26 weeks) and a mean birth weight of 793 g (range: 630–1030 g) at a mean age of 10 days (range: 7–14 days) after birth. The preterm infants were all on continuous ventilator support with no signs of imminent clinical improvement. Three patients received a low dose (1×107 cells/kg in 2 mL/kg of saline), and the remaining 6 patients received a high dose (2×107 cells/kg in 4 mL/kg of saline). The MSCs were administered intratracheally in two fractions into the left and right lungs via a gavage tube using the same method for administering surfactant. The treatment was well tolerated without any serious adverse effects or dose-limiting toxicity up to 84 days following transplantation. No significant differences in serious adverse effects were observed between the low- and high-dose groups. Levels of cytokines in tracheal aspirates at day 7 following treatment were significantly reduced, compared with those at baseline. Furthermore, BPD severity was significantly lower in transplant recipients, compared with the historical gestational age-, birth weight-, and respiratory severity score-matched control group. Although it is too early to draw any firm conclusion on therapeutic efficacy, the study indicates that IT transplantation of allogenic human UCB-derived MSCs in very preterm infants at highest risk for developing BPD is safe and feasible. A long-term follow-up study and a phase II, double-blind, randomized, controlled trial to assess therapeutic efficacy are underway.

Phase I clinical trial for HIE

To date, no clinical trial using MSCs for HIE has been reported, and one phase I clinical trial using autologous UCB cells has been reported.9 Cotten, et al.9 assessed the safety and feasibility of autologous UCB cell transplantation in a phase I clinical trial of 23 neonates with HIE who met criteria and received concurrent hypothermia treatment. In that open-label study, intravenous infusion of non-cryopreserved, autologous, volume-and red blood cell-reduced UCB cells, up to four doses of 1–5×107 cells per dose, was found to be feasible and well tolerated. With appropriate isolation and processing, collection of UCB was successful even with a small volume of cord blood (range: 3–178 mL) to achieve the goal dose for each infusion for HIE in near-term and term infants with hypothermia. No significant adverse reactions, cardiopulmonary compromise, or infections associated with the transfusion were observed, and UCB recipients showed better 1-year survival rates (74%) with Bayley scores >85 than concurrent cooled infants (41%). Collectively, these findings suggest that collection, preparation, and intravenous infusion of fresh, non-cryopreserved, autologous UCB cells for clinical use in infants with HIE are likely safe and feasible. Randomized, double-blind, controlled clinical trials in the future will be required to evaluate the therapeutic efficacy of autologous UCB cell therapy for HIE.

Phase I clinical trial for IVH

Recently, a phase I, dose-escalating clinical study on the safety and feasibility of human UCB-derived MSC transplantation in preterm infants with severe IVH (i.e., grade≥3) (NCT02274428) has been conducted in light of promising preclinical evidence demonstrating the protective effects of MSC transplantation in a newborn rat model of severe IVH.7 This study was an open-label, single-center clinical trial to assess the safety and feasibility of a single intraventricular transplantation of allogenic human UCB-derived MSCs within 7 days after detection of severe IVH (grade≥3) under ultrasound guidance in preterm infants. Of a total of nine preterm infants with severe IVH, three were given a low dose (5×106 cells/kg in 1 mL/kg of saline); after confirming that no dose-limiting toxicity or serous adverse events were associated with transplantation, the remaining six infants were given a high dose (1×107 cells/kg in 2 mL/kg of saline). The primary outcome measures were unsuspected death or anaphylactic shock within 6 h after MSC transplantation, and the secondary outcome measures were death or hydrocephalus requiring shunt surgery up to 1 year of age. These enrolled infants are currently undergoing follow-up care for long-term adverse outcomes and assessment of neurologic health. We are also planning to conduct a double-blind, randomized, phase IIa, controlled clinical trial to test the therapeutic efficacy of MSC transplantation for currently intractible and devastating severe IVH in premature infants.

PROSPECTS AND CHALLENGES FOR CLINICAL TRANSLATION

Clinical translation of stem cell therapies could represent a paradigm shift in neonatal medicine. However, barriers to the clinical implementation thereof, including delivering optimal cells for the right patient with the right route at the right time and dose and long-term safety of MSC transplantation, need to be resolved for successful clinical translation of stem cell therapies in the near future. Current research on each barrier is briefly summarized and discussed in the following sections.

The right cells

Choosing the right cells for transplantation is the most critical issue for successful clinical translation of stem cell therapy. As of today, MSCs have been most extensively investigated, since they are easily obtainable, do not have the ethical or safety concerns (e.g., tumorigenic potential) of embryonic stem cells, and their therapeutic efficacy has consistently been demonstrated in animal models of BPD,51012131415 HIE,6 and IVH.71617 Although no specific markers for MSCs exist, as of now, MSCs have several unusual characteristics: 1) fibroblast-like morphology, 2) adherence to plastic, 3) positive expression of CD73, CD90, and CD105, 4) negative expression of CD45, CD34, CD14, CD11b, and HLA DR, and 5) differentiation capacity into various cell lineages including adipocytes, chondrocytes, and osteoblasts.18
Use of allogenic versus autologous MSCs is one issue to be addressed. MSCs are known to be immune-privileged due to their lack of MHC II antigens and their ability to inhibit the proliferation and function of immune cells, such as dendritic cells, NK cells, and T and B lymphocytes,1920 and these features allow for easier allogenic therapy.21 In addition to their immune tolerance, donor MSCs do not engraft, and they fade away after therapeutic paracrine protection,22 such that immunosuppression of the recipient might not be necessary. Therefore, allogenic transplantation of MSCs could safely be applied to all neonates in need of this therapy. Furthermore, considering the time-consuming and costly isolation and expansion processes for autologous MSCs, allogenic therapy might have a logistic advantage, because they are ready to use “off the shelf” in the clinical setting.
As MSCs are broadly distributed in the body, determining the tissue source of MSCs is another issue that needs to be resolved. Gestational tissues, such as the placenta, amniotic fluid, UCB, Wharton's jelly, and the umbilical cord, are medical waste and usually discarded at birth. Therefore, MSCs obtained from gestational tissues are particularly attractive due to their easy obtainability and lack of significant ethical concerns. Furthermore, MSCs derived from gestational tissues have been shown to exhibit lower immunogenicity,2324 higher proliferative capacity,2526 and higher paracrine potency than adult tissue-derived MSCs.27 UCB-derived MSCs have been found to offer better in vivo therapeutic efficacy in attenuating hyperoxic lung injuries and increased paracrine potency, compared to fat-derived MSCs, in newborn rats.15 Overall, these findings suggest that gestational tissues, rather than adult tissues, might be a promising source for obtaining exogenous human MSCs for future clinical use in treating neonates with intractable disorders.1415
Production of a high-quality, standardized, clinical-grade product using good manufacturing practice (GMP) criteria is another critical issue for the success of MSCs in clinical settings.28 Allogenic human UCB-derived MSCs manufactured in strict compliance with GMP criteria and approved by both Korean and US FDAs were used for phase I/II clinical trials for BPD conducted in Korea8 and the USA (NCT02381366) and for a phase I clinical trial for severe IVH (NCT02274428). Important quality control criteria include the risk for malignant transformation, as well as aging, of the manufactured MSCs.8 We have observed karyotypic stability and no senescence up to the 11th passage of human UCB-derived MSCs, and sixth-passage MSCs were used for the clinical trial to avoid these potential risks.8 Standardization of isolation and expansion procedures, mini-mized batch variability, increased cell potency, and mass production of clinical-grade products in GMP-compliant facilities would be necessary for future clinical introduction of other cell types, including endothelial progenitor cells and amnion epithelial cells, as well as cell-derived products, such as exosomes, that show beneficial effects in models of neonatal disorders.

The right patients

Despite preclinical and clinical evidence demonstrating potential benefits, cautious risk-benefit evaluations must be made before implementing routine clinical use of MSC transplantation in vulnerable preterm infants. Therefore, identifying newborn infants at highest risk for developing intractable diseases and the ensuing mortality and morbidities is very important for applying these potentially beneficial rescue/preventive strategies.
As younger gestational age and prolonged respiratory support are two prime predictors of subsequent development of BPD in extremely preterm infants,29 extremely preterm infants at 24–26 weeks gestation needing continuous ventilator support without clinical improvements within 24 hours were enrolled in the phase I clinical trial conducted by Chang, et al.8 However, clinical courses can widely vary, and not all of these high-risk extremely preterm infants develop BPD. Additional clinical predictors and/or biomarkers30 will be necessary to identify infants that will ultimately progress to BPD in order to avoid unnecessary treatment exposure.
Currently, although hypothermia is the only clinically available treatment for neonatal HIE, it is only partially effective, especially in severe cases.3132 Therefore, early identification using not only categorical neurologic examination, but also biomarkers of neuronal injury33 in neonates with severe HIE who ultimately might benefit from stem cell therapy, in addition to hypothermia,6 is crucial for better therapeutic outcomes.
More than 50% of preterm infants with severe (grade≥3) IVH die or develop posthemorrhagic hydrocephalus (PHH), which requires shunt surgery in up to 70% of cases.34 Therefore, severe IVH in preterm infants might be a good therapeutic indication of stem cell therapy. Further studies will be necessary to clarify whether biomarkers of neuronal injury in cerebrospinal fluid35 can predict the progress of PHH and poor neurodevelopmental outcomes, thus stratifying the preterm infants with severe IVH who might most benefit from stem cell therapy.

The right route

Various preclinical and clinical studies of BPD, HIE, and IVH have attempted to determine the optimal route for MSC translation using several routes, including local intraventricular,736 intrathecal,37 intranasal,38 or IT5 administration and systemic intraperitoneal5 or intravenous939 administration.
Although intranasal delivery of MSCs is minimally invasive, it did not attenuate hyperoxic lung injuries in mice40 and showed less therapeutic efficacy requiring more than five-fold dose in attenuating brain injuries in HIE.4142 Therefore, more preclinical studies in larger animals, such as piglets or primates, verifying the therapeutic efficacy of intranasal delivery of stem cells are necessary for successful clinical translation.
As systemically transplanted MSCs migrate and localize toward injured tissue under chemotactic guidance,4344 the convenient and minimally-invasive approaches of systemic intravenous or intraperitoneal administration might be therapeutically advantageous, compared with the more invasive approaches of local intraventricular or IT administration, especially in very unstable newborn infants who may not tolerate more invasive local injection of stem cells. However, systemically transplanted MSCs might have limitations in crossing the blood brain barrier17 and could also be retained in other organs, such as the lungs, liver, spleen, and kidneys.45 Thus, better therapeutic efficacy was observed with local administration of MSCs, compared to four- to five-fold higher doses delivered by systemic injection, in newborn animal models of IVH17 and BPD.39 More-over, because the anterior fontanelle is open in newborn infants, and all preterm infants at high risk for BPD are already intubated to receive ventilator support, further invasive procedures would not be necessary for local delivery of stem cells in these neonates. Based on this preclinical evidence, MSCs were delivered intratracheally or intraventricularly in the first phase I clinical trials of BPD8 and IVH,46 respectively.

The right timing

Determining the optimal timing for MSC transplantation is another major issue that remains to be clarified for future clinical translation. For BPD, Pierro, et al.11 found that administration of MSCs or conditioned media not only at P4 for prevention, but also at P14 for rescue approaches prevented and rescued hyperoxic alveolar growth arrest. However, significant attenuation of hyperoxic lung injuries with MSC transplantation was observed only early (P3) but not late (P10), and no synergies were observed with combined early and late MSC transplantation in the study by Chang, et al.12 In the study by van Haaften, et al.,47 MSC transplantation at P4 for prevention, but not at P14 for regeneration, significantly attenuated neonatal hyperoxic lung injuries. Collectively, these data suggest that the therapeutic time window for MSC transplantation for BPD is narrow. Thus, in a clinical setting, early identification of preterm infants at the highest risk for developing BPD and transplantation of MSCs as early as possible within the first few postnatal days seem favorable over later administration in preterm infants with established BPD.
The therapeutic time window of MSCs for neonatal brain injury shows wide variability, ranging from the first hours to 10 days after the insult,748495051 and this wide variation might be attributable to differences in animal models and the severity of brain insult. As the neuroprotective effects of MSCs might be mediated primarily by their paracrine anti-inflammatory and antiapoptotic properties,7 MSCs must be transplanted during the very acute phase after brain injury for their neuroprotection to be effective. In our recent study conducted to optimize the timing of MSC transplantation for severe IVH,52 intraventricular transplantation of MSCs early at 2 days, but not at 7 days, after induction of severe IVH significantly attenuated the development of PHH; impaired behavioral function tests; increased apoptosis, astrogliosis, and inflammatory responses; and reduced corpus callosum thickness and brain myelination associated with IVH. This phenomenon was also observed in other disease models, including murine graft-versus-host disease models and experimental autoimmune encephalomyelitis models.4953 Overall, the therapeutic time window for stem cell therapy might be narrow; thus, transplantation of MSCs as close as possible to the time of brain insult might provide better therapeutic outcomes. Further studies will be necessary to clarify this.

The right dose

In light of the preclinical data showing wide variation in doses and the resultant therapeutic efficacy according to the route of stem cell administration in animal models of BPD,1339 HIE49 and IVH,17 the injury site and route of administration seem to be two major determinants of the optimal dose for MSC transplantation. In light of preclinical evidence showing dose-dependent protection with MSC transplantation,1349 we tested the safety and feasibility of a single IT transplantation of 1×107 cells/kg for low dose or 2×107 cells/kg for high dose in our pioneering phase I clincical trial for BPD8 and a single intraventricular transplatation of 5×106 cells/kg for low dose or 1×107 cells/kg for high dose in a phase I clinical trial for severe IVH;46 no dose-limiting toxicity or serious adverse events were observed with either dose. Taken together, these findings suggest that injury site and route of administration are major determinants for the optimal dose of MSCs in transplantation. Based on these findings, further preclinical clinical studies to determine the optimal dose of MSCs for maximal clinical benefit in newborn infants affected by currently intractable and devastating disorders are anticipated.

Safety of MSC transplantation

Considering the early postnatal age of newborn infants who are potential candidates for stem cell therapy, a longitudinal study showing not only therapeutic efficacy, but also both short-and long-term safety in an animal model is essential for successful clinical translation. Due to its short life span, a rodent model is most suitable because both short- and long-term research is feasible in a short time frame. In addition to the persistent protective effects of intranasal transplantation of MSCs against hypoxic ischemic brain injuries in newborn mice up to 14 months after transplant, no abnormalities including neoplasia were observed in the nasal turbinates, brain, or other organs.54 Similarly, the protective and beneficial effects of IT transplantation of MSCs were sustained without any long-term adverse effects, such as tumorigenicity, in rats up to 70 days10 or 6 months of age,11 comparable to human adolescence and mid-adulthood, respectively. Furthermore, in our previous phase I clinical trial,8 IT transplantation of MSCs was safe and feasible, and no adverse outcomes including tumorigenicity have been reported in follow up of these infants until 2 years of corrected age and in more than 350 clinical studies of MSC transplantation conducted worldwide. The absence of long-term adverse effects including tumorigenicity after MSC transplantation might be attributable to the absent engraftment of the transplanted MSCs;11 thus, they exert their therapeutic function by a “hit and run” mechanism.55 Overall, the preclinical data indicating the sustained protective effects of MSC transplantation without any short- or long-term adverse effects warrant the translation of MSC transplantation into clinical trials for treatment of intractable disorders in newborn infants.

CONCLUSION

Stem cell therapy for currently intractable and devastating neonatal disorders, such as BPD, HIE, and IVH, might be a potential paradigm shift in neonatal medicine. Exciting progress in preclinical and clinical studies has brought human stem cell therapy for newborn infants one step closer to clinical translation. However, the resolution of several issues, such as safety, up-scaling of manufacturing processes, standardization methods, clinical indications, timing, dosage, and a better understanding of protective mechanisms, are required to permit safe clinical translation of stem cell therapy for newborn infants in the near future. It is essential to proceed step-by-step, rather than in haste, with relentless efforts to overcome these obstacles and make stem cell therapy the next breakthrough in the therapy of these intractable neonatal disorders.

ACKNOWLEDGEMENTS

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (NRF-2014R1A1A3051220), the Korean Healthcare Technology R&D Project, Ministry of Health, Welfare and Family Affairs, Republic of Korea (HR14C0008, HI16C1061).

Notes

The authors have no financial conflicts of interest.

References

1. Bland RD. Neonatal chronic lung disease in the post-surfactant era. Biol Neonate. 2005; 88:181–191.
crossref
2. Bhandari A, Panitch HB. Pulmonary outcomes in bronchopulmonary dysplasia. Semin Perinatol. 2006; 30:219–226.
crossref
3. Vohr BR, Wright LL, Dusick AM, Mele L, Verter J, Steichen JJ, et al. Neurodevelopmental and functional outcomes of extremely low birth weight infants in the national institute of child health and human development neonatal research network, 1993-1994. Pediatrics. 2000; 105:1216–1226.
crossref
4. Johnston MV. Hypoxic and ischemic disorders of infants and children. Lecture for 38th meeting of Japanese Society of Child Neurology, Tokyo, Japan, July 1996. Brain Dev. 1997; 19:235–239.
crossref
5. Chang YS, Oh W, Choi SJ, Sung DK, Kim SY, Choi EY, et al. Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2009; 18:869–886.
crossref
6. Park WS, Sung SI, Ahn SY, Yoo HS, Sung DK, Im GH, et al. Hypothermia augments neuroprotective activity of mesenchymal stem cells for neonatal hypoxic-ischemic encephalopathy. PLoS One. 2015; 10:e0120893.
crossref
7. Ahn SY, Chang YS, Sung DK, Sung SI, Yoo HS, Lee JH, et al. Mesenchymal stem cells prevent hydrocephalus after severe intraventricular hemorrhage. Stroke. 2013; 44:497–504.
crossref
8. Chang YS, Ahn SY, Yoo HS, Sung SI, Choi SJ, Oh WI, et al. Mesenchymal stem cells for bronchopulmonary dysplasia: phase 1 dose-escalation clinical trial. J Pediatr. 2014; 164:966–972.
crossref
9. Cotten CM, Murtha AP, Goldberg RN, Grotegut CA, Smith PB, Goldstein RF, et al. Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr. 2014; 164:973–979.
crossref
10. Ahn SY, Chang YS, Kim SY, Sung DK, Kim ES, Rime SY, et al. Longterm (postnatal day 70) outcome and safety of intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells in neonatal hyperoxic lung injury. Yonsei Med J. 2013; 54:416–424.
crossref
11. Pierro M, Ionescu L, Montemurro T, Vadivel A, Weissmann G, Oudit G, et al. Short-term, long-term and paracrine effect of human umbilical cord-derived stem cells in lung injury prevention and repair in experimental bronchopulmonary dysplasia. Thorax. 2013; 68:475–484.
crossref
12. Chang YS, Choi SJ, Ahn SY, Sung DK, Sung SI, Yoo HS, et al. Timing of umbilical cord blood derived mesenchymal stem cells transplantation determines therapeutic efficacy in the neonatal hyperoxic lung injury. PLoS One. 2013; 8:e52419.
crossref
13. Chang YS, Choi SJ, Sung DK, Kim SY, Oh W, Yang YS, et al. Intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells dose-dependently attenuates hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2011; 20:1843–1854.
crossref
14. Ahn SY, Chang YS, Park WS. Stem cell therapy for bronchopulmonary dysplasia: bench to bedside translation. J Korean Med Sci. 2015; 30:509–513.
crossref
15. Ahn SY, Chang YS, Sung DK, Yoo HS, Sung SI, Choi SJ, et al. Cell type-dependent variation in paracrine potency determines therapeutic efficacy against neonatal hyperoxic lung injury. Cytotherapy. 2015; 17:1025–1035.
crossref
16. Ahn SY, Chang YS, Sung DK, Sung SI, Ahn JY, Park WS. Pivotal role of brain derived neurotrophic factor secreted by mesenchymal stem cells in severe intraventricular hemorrhage in the newborn rats. Cell Transplant. 2016; 08. 16. [Epub]. DOI: 10.3727/096368916X692861.
17. Ahn SY, Chang YS, Sung DK, Sung SI, Yoo HS, Im GH, et al. Optimal route for mesenchymal stem cells transplantation after severe intraventricular hemorrhage in newborn rats. PLoS One. 2015; 10:e0132919.
crossref
18. Spencer ND, Gimble JM, Lopez MJ. Mesenchymal stromal cells: past, present, and future. Vet Surg. 2011; 40:129–139.
crossref
19. Parad RB, Davis JM, Lo J, Thomas M, Marlow N, Calvert S, et al. Prediction of respiratory outcome in extremely low gestational age infants. Neonatology. 2015; 107:241–248.
crossref
20. Parsons PE, Eisner MD, Thompson BT, Matthay MA, Ancukiewicz M, Bernard GR, et al. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit Care Med. 2005; 33:1–6.
crossref
21. Noël D, Djouad F, Bouffi C, Mrugala D, Jorgensen C. Multipotent mesenchymal stromal cells and immune tolerance. Leuk Lymphoma. 2007; 48:1283–1289.
crossref
22. Waszak P, Alphonse R, Vadivel A, Ionescu L, Eaton F, Thébaud B. Preconditioning enhances the paracrine effect of mesenchymal stem cells in preventing oxygen-induced neonatal lung injury in rats. Stem Cells Dev. 2012; 21:2789–2797.
crossref
23. Rocha V, Wagner JE Jr, Sobocinski KA, Klein JP, Zhang MJ, Horowitz MM, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. Eurocord and international bone marrow transplant registry working committee on alternative donor and stem cell sources. N Engl J Med. 2000; 342:1846–1854.
crossref
24. Le Blanc K. Immunomodulatory effects of fetal and adult mesenchymal stem cells. Cytotherapy. 2003; 5:485–489.
crossref
25. Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006; 24:1294–1301.
crossref
26. Yang SE, Ha CW, Jung M, Jin HJ, Lee M, Song H, et al. Mesenchymal stem/progenitor cells developed in cultures from UC blood. Cytotherapy. 2004; 6:476–486.
crossref
27. Amable PR, Teixeira MV, Carias RB, Granjeiro JM, Borojevic R. Protein synthesis and secretion in human mesenchymal cells derived from bone marrow, adipose tissue and Wharton's jelly. Stem Cell Res Ther. 2014; 5:53.
crossref
28. Mendicino M, Bailey AM, Wonnacott K, Puri RK, Bauer SR. MSCbased product characterization for clinical trials: an FDA perspective. Cell Stem Cell. 2014; 14:141–145.
crossref
29. Laughon MM, Langer JC, Bose CL, Smith PB, Ambalavanan N, Kennedy KA, et al. Prediction of bronchopulmonary dysplasia by postnatal age in extremely premature infants. Am J Respir Crit Care Med. 2011; 183:1715–1722.
crossref
30. D'Angio CT, Ambalavanan N, Carlo WA, McDonald SA, Skogstrand K, Hougaard DM, et al. Blood cytokine profiles associated with distinct patterns of bronchopulmonary dysplasia among extremely low birth weight infants. J Pediatr. 2016; 174:45–51.
31. Azzopardi DV, Strohm B, Edwards AD, Dyet L, Halliday HL, Juszczak E, et al. Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med. 2009; 361:1349–1358.
crossref
32. Shankaran S, Laptook AR, Ehrenkranz RA, Tyson JE, McDonald SA, Donovan EF, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med. 2005; 353:1574–1584.
crossref
33. Chalak LF, Sánchez PJ, Adams-Huet B, Laptook AR, Heyne RJ, Rosenfeld CR. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr. 2014; 164:468–474.
crossref
34. Murphy BP, Inder TE, Rooks V, Taylor GA, Anderson NJ, Mogridge N, et al. Posthaemorrhagic ventricular dilatation in the premature infant: natural history and predictors of outcome. Arch Dis Child Fetal Neonatal Ed. 2002; 87:F37–F41.
crossref
35. Merhar S. Biomarkers in neonatal posthemorrhagic hydrocephalus. Neonatology. 2012; 101:1–7.
crossref
36. Spaggiari GM, Capobianco A, Becchetti S, Mingari MC, Moretta L. Mesenchymal stem cell-natural killer cell interactions: evidence that activated NK cells are capable of killing MSCs, whereas MSCs can inhibit IL-2-induced NK-cell proliferation. Blood. 2006; 107:1484–1490.
crossref
37. Fang B, Wang H, Sun XJ, Li XQ, Ai CY, Tan WF, et al. Intrathecal transplantation of bone marrow stromal cells attenuates blood-spinal cord barrier disruption induced by spinal cord ischemia-reperfusion injury in rabbits. J Vasc Surg. 2013; 58:1043–1052.
crossref
38. Donega V, Nijboer CH, van Velthoven CT, Youssef SA, de Bruin A, van Bel F, et al. Assessment of long-term safety and efficacy of intranasal mesenchymal stem cell treatment for neonatal brain injury in the mouse. Pediatr Res. 2015; 78:520–526.
crossref
39. Sung DK, Chang YS, Ahn SY, Sung SI, Yoo HS, Choi SJ, et al. Optimal route for human umbilical cord blood-derived mesenchymal stem cell transplantation to protect against neonatal hyperoxic lung injury: gene expression profiles and histopathology. PLoS One. 2015; 10:e0135574.
crossref
40. Liu L, Mao Q, Chu S, Mounayar M, Abdi R, Fodor W, et al. Intranasal versus intraperitoneal delivery of human umbilical cord tissuederived cultured mesenchymal stromal cells in a murine model of neonatal lung injury. Am J Pathol. 2014; 184:3344–3358.
crossref
41. Grade S, Weng YC, Snapyan M, Kriz J, Malva JO, Saghatelyan A. Brain-derived neurotrophic factor promotes vasculature-associated migration of neuronal precursors toward the ischemic striatum. PLoS One. 2013; 8:e55039.
crossref
42. Hudgins RJ, Boydston WR, Hudgins PA, Morris R, Adler SM, Gilreath CL. Intrathecal urokinase as a treatment for intraventricular hemorrhage in the preterm infant. Pediatr Neurosurg. 1997; 26:281–287.
crossref
43. Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J, et al. Bone marrow-derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol. 2005; 33:145–152.
crossref
44. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003; 100:8407–8411.
crossref
45. Aslam M, Baveja R, Liang OD, Fernandez-Gonzalez A, Lee C, Mitsialis SA, et al. Bone marrow stromal cells attenuate lung injury in a murine model of neonatal chronic lung disease. Am J Respir Crit Care Med. 2009; 180:1122–1130.
crossref
46. Ahn SY, Chang YS, Park WS. Stem cell for neonatal brain disorders. Neonatology. 2016; 109:377–383.
47. van Haaften T, Byrne R, Bonnet S, Rochefort GY, Akabutu J, Bouchentouf M, et al. Airway delivery of mesenchymal stem cells prevents arrested alveolar growth in neonatal lung injury in rats. Am J Respir Crit Care Med. 2009; 180:1131–1142.
crossref
48. Comi AM, Cho E, Mulholland JD, Hooper A, Li Q, Qu Y, et al. Neural stem cells reduce brain injury after unilateral carotid ligation. Pediatr Neurol. 2008; 38:86–92.
crossref
49. Donega V, van Velthoven CT, Nijboer CH, van Bel F, Kas MJ, Kavelaars A, et al. Intranasal mesenchymal stem cell treatment for neonatal brain damage: long-term cognitive and sensorimotor improvement. PLoS One. 2013; 8:e51253.
crossref
50. Mosna F, Sensebé L, Krampera M. Human bone marrow and adipose tissue mesenchymal stem cells: a user's guide. Stem Cells Dev. 2010; 19:1449–1470.
crossref
51. Yasuhara T, Hara K, Maki M, Xu L, Yu G, Ali MM, et al. Mannitol facilitates neurotrophic factor up-regulation and behavioural recovery in neonatal hypoxic-ischaemic rats with human umbilical cord blood grafts. J Cell Mol Med. 2010; 14:914–921.
crossref
52. Park WS, Sung SI, Ahn SY, Sung DK, Im GH, Yoo HS, et al. Optimal timing of mesenchymal stem cell therapy for neonatal intraventricular hemorrhage. Cell Transplant. 2016; 25:1131–1144.
crossref
53. Yañez R, Lamana ML, García-Castro J, Colmenero I, Ramírez M, Bueren JA. Adipose tissue-derived mesenchymal stem cells have in vivo immunosuppressive properties applicable for the control of the graft-versus-host disease. Stem Cells. 2006; 24:2582–2591.
crossref
54. Walter LM, Biggs SN, Nisbet LC, Weichard AJ, Muntinga M, Davey MJ, et al. Augmented cardiovascular responses to episodes of repetitive compared with isolated respiratory events in preschool children with sleep-disordered breathing. Pediatr Res. 2015; 78:560–566.
crossref
55. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014; 32:252–260.
crossref
TOOLS
Similar articles