1,782
Views
23
CrossRef citations to date
0
Altmetric
Editorial

How far are we from the clinical use of placental-derived mesenchymal stem cells?

, MD PhD, , MD, , MD PhD & , PhD

Abstract

In recent years, multiple studies have investigated the biology and clinical applications of mesenchymal stem cells (MSCs), trying to define their markers, and elucidate their effects in animal models. MSCs are available from different tissues, and the use of placental-derived MSCs (PMSCs) for treating a variety of disorders is on the forefront. Herein, we discuss the most recent findings regarding the standardization of their isolation procedure and phenotype, along with advantages and limitations of their use. We also discuss the safety of the placental cell products, including the issue of senescence and mutagenesis of PMSCs, and efficacy from preclinical studies.

1. Mesenchymal stem cells in regenerative medicine

Mesenchymal stem cells (MSCs) were identified in 1970 in the bone marrow (BM) as fibroblast-like cells secreting factors involved in hematopoiesis Citation[1] and with adipogenic, chondrogenic and osteogenic differentiation potential. Notably, MSCs can also be isolated from different placental tissues, including fetal membranes Citation[2], different regions of the umbilical cord Citation[3] and amniotic fluid Citation[4].

There have been significant attempts to define markers, biological properties and therapeutic applications of MSCs Citation[5]. MSCs are nowadays the most widely explored cell type for therapy due to their renowned regenerative and immune-regulatory properties Citation[5]. The current need is to standardize in vitro assays in order to predict the potential therapeutic effects of different MSCs from different sources and to optimize protocols for clinical applications.

2. The issue regarding MSC sources: different sources, different uses?

MSCs are considered immune-privileged for their low expression of class II major histocompatibility complex and co-stimulatory molecules Citation[2]. BM-MSCs possess immunomodulatory capacities Citation[5], even though previous reports have shown that they can induce an immune response Citation[6]. MSCs are available from different sources, thus possibly expanding their potential applications. However, this raises the question regarding which is the ideal source, considering also differentiation and functional properties of MSCs Citation[7,8]. A major issue is the need to standardize a single isolation procedure for large-scale in vitro expansion, taking also into consideration whether it is possible to obtain enough cells from a single donor, or if it is necessary to use cells from multiple donors. Another concern is the choice between using allogeneic or autologous MSCs in clinical trials. The latter would allow better standardization and reduce production costs, since autologous cells are not always exploitable for efficient ex vivo expansion. As pointed out by Zhu and colleagues Citation[9], this procedure has the intrinsic risk of genetic and epigenetic mutations that may lead to malignant transformation of MSCs derived from several tissues of different animal species. Deposition of epigenetic mutations was also discovered within placental-derived MSCs (PMSCs); however, these alterations did not translate into a malignant phenotype. This relevant issue has to be unraveled before any clinical application is pursued.

3. Placental mesenchymal stem/stromal cells

Human term placenta is an organ composed of different tissues. Cells with mesenchymal stem/stromal characteristics can be isolated from different regions of the fetal part of human term placenta, namely the amniotic membrane (human amniotic membrane-derived MSCs) and chorionic mesenchymal region (human chorionic mesenchymal stroma-derived MSCs), and also from the chorionic villi and regions of the umbilical cord Citation[10]. Herein, PMSCs will be used as a general term and is referred to MSCs isolated from the placenta, not taking into consideration the specific region from which they were isolated.

3.1 Advantages and limitations

The advantages of placenta as an MSC source include its easy procurement with no harm to the mother and baby, high cell recovery, ethical acceptance, and interestingly, the possible higher stemness potential related to the early embryological origin of the placenta Citation[11] and the unique immunomodulatory characteristics of placental cells Citation[2]. Similar to MSCs from other sources, those derived from the human amniotic membrane inhibit T-cell proliferation in vitro Citation[12] and in vivo Citation[13], and MSCs from different placental regions have also been shown to strongly inhibit the generation, maturation and function of monocyte-derived dendritic cells Citation[14]. Interestingly, the conditioned medium (CM) obtained from the culture of MSCs from the amniotic membrane possesses the same in vitro Citation[8] and in vivo potential Citation[15]. To the contrary, BM-MSCs and their CM exert immunomodulatory potential only if the cells are cultured in the presence of activating conditions Citation[8,16]. This raises an interesting and advantageous point regarding the peculiar immunomodulatory characteristics unique to PMSCs, which constitutes an underlying mechanism of their therapeutic potential based on paracrine effects or on released factors, both of which have been proven in different experimental models of inflammation and fibrosis Citation[17]. On the other hand, the differentiation capability of PMSCs has been proven in vitro, but it is more questioned in vivo Citation[2].

Studies on PMSCs are exponentially growing, but are still in their early stages. Thus, in order to envision clinical applications, a few unresolved issues need to be considered, some of which are common to MSCs from other sources. For example, MSC heterogeneity and impact of culture conditions on cell characteristics and potentiality is a big issue in the stem cell field. There is a need to have specific markers, which can unambiguously define phenotype and mechanism of action. Other issues that need to be addressed are associated to the fact that the placenta is a recent MSC source, thus the standardization of isolation procedures, safety and stability, tumorigenicity, and efficacy assessments based on studies in different animal models, are still important concerns, and these will be discussed in the following sections.

3.2 Standardization of the isolation procedure

The isolation of MSCs from different placental regions employs either explant culture Citation[2] or enzymatic digestion with various enzymes, such as dispase and collagenase, in combination with DNase. MSCs possess characteristic spindle, fibroblast-like shape in culture and the classical phenotype, also reported for MSCs from other sources, as defined at the First International Workshop on PMSCs, namely expression of CD90, CD105 and CD73, lack of CD45, CD34, CD14, and HLA-DR.

However, PMSCs (like MSCs from other tissues) are highly heterogeneous, and include different populations and different subtypes Citation[2,18]. The differences in phenotype and properties between MSCs isolated from different sources are currently being investigated, but for human term placenta we should also consider that these characteristics may vary between regions of the same organ Citation[10,19]. This should be taken into account when investigating the effects of culture conditions on phenotype and safety. Furthermore, as previously demonstrated, the culture of placenta-derived cells could carry maternal contamination. This constitutes another important issue to be considered, which requires very sensitive tests to determine and/or control fetal or maternal origin after cell culture and expansion Citation[20,21].

3.3 Safety of PMSCs

MSCs are considered as an advanced therapy medicinal product, which include medical products based on genes (gene therapy), cells (cell therapy) and tissues (tissue engineering), and thus are subjected to the same regulation as conventional chemical pharmaceutical drugs.

Indeed, translating basic stem cell research into routine therapies is a complex, multi-step process. The challenge is to manage the expected therapeutic benefits with the potential risks while complying with existing guidelines and regulations. Undoubtedly, the most important point to consider is the safety of these cell products.

Currently, the clinical trials website (www.clinicaltrials.gov) reports 15 trials based on the use of ‘cells derived from placenta’ including those with unknown status. Within the eight completed clinical trials, two were carried out by academic groups and six by private companies. Results obtained from the clinical trial in completed studies have been published. Unfortunately, no details are provided regarding cell manufacturing, including the development of a multi-step controlled and validated process, neither about the quality control approach which should be appropriately designed during and after manufacturing operations Citation[22,23]. This example demonstrates how the safety of some cell products can be overshadowed with respect to their therapeutic efficacy. As previously mentioned, risks and safety should be the main focus in stem cell research and cell therapy today, and this applies also for PMSCs. Therefore, what are the crucial aspects to be considered in order to ensure a safe PMSC product, in addition to improving standardized isolation protocols, as discussed above? We believe the following points should be of interest for the future of PMSCs. First, the purity of the cell product should be determined. This should consider the possible fetal/maternal contamination and also negative clinical effects resulting from engraftment of undesired cell types into an ectopic tissue. A combination of validated analytical methods should define the acceptable percentage of contaminants cells a priori. Second, the tumorigenic potential should surely be considered as a safety evaluation. It is well established that genetic changes can occur during cell culture. Stem cells in particular accumulate chromosomal aberrations, especially at high passages Citation[24], which possibly lead to malignant transformation. Can a specific culture condition, a supplement, cytokine, or xenobiotic-free medium modify and compromise cell stability? These modifications should be thoroughly characterized and the risks assessed before any clinical application. For example, in an allogeneic setting (as is the case for PMSCs), the need for extensive cell quantity expansion in order to obtain enough cells to treat several patients increases the risk of accumulating genetic and epigenetic abnormalities. MSC expansion is associated with a systematic modulation of the epigenetic chromatin structure at specific sites in the genome, but the precise regulation of this process is yet unknown Citation[9]. Finally, senescence should also be taken into account. Cells in culture undergo replicative senescence but chromosomal aberrations are not always detected simply by karyotyping or by single nucleotide polymorphism-microarrays. At early passages, the fibroblastoid colony forming unit frequency and the differentiation potential of MSCs decline significantly, indicating that many cells within the MSC compartment have a restricted proliferative capacity Citation[24].

3.4 PMSC efficacy

The clinical trials mentioned above are Phase I and II, thus they are mostly aimed at safety evaluation. In regard to efficacy, lessons can be learned from preclinical studies, which have shown great promise. PMSCs have shown to be able to restore the impaired or even lost function and delay the degenerative process in models of debilitating neurological diseases such as Parkinson’s disease Citation[25], Alzheimer’s disease Citation[26,27] and multiple sclerosis Citation[28]. Furthermore, other studies employing PMSCs and their derivatives in animal models of lung disease have demonstrated reduced inflammation and fibrosis Citation[15,29,30]. Other groups have shown disease attenuation after treatment with PMSCs in animal models of myocardial infarction Citation[31,32], and also in autoimmune diseases such as rheumatoid arthritis Citation[13]. Furthermore, preclinical studies have shown that PMSCs may also be a helpful in liver diseases, as suggested by their potential to differentiate toward functional hepatocyte-like cells in vitro Citation[33], and also owing to their immune modulatory properties. Importantly, preclinical studies have and will continue to pave the way for further studies focusing on the mechanism of actions underlying disease amelioration.

4. Concluding remarks

In conclusion, there is plenty of evidence demonstrating efficacy of placenta-derived cells or CM derived from these cells in diseases with underlying inflammatory and fibrotic abnormalities Citation[15,17,30]. Attempts to improve and standardize methods have been reported, however much remains to be defined. It is unclear whether the current knowledge is sufficient to adequately assess the safety of new cell therapies in a comprehensive manner. The current concept of safety is not unambiguous, but we are surely evolving. The safety of a product will go hand in hand with the increase of knowledge on these cells. Importantly, cell biologists, therapy providers, clinicians and regulatory agencies need to interact to define the acceptable risk associated with PMSCs treatment, and to establish a framework for accurate risk assessment.

An obvious question arises regarding the need for an alternative source of MSCs, such as placenta, and if placental cells could target specific diseases compared to MSCs from other sources. Indeed, placenta as a source of other stem cells, besides MSCs, could reserve some interesting promises, while placenta as an MSC source is fascinating if we foresee to produce off-the-shelf products from biological waste. If the safety and efficacy of PMSCs can be proven comparable, or better than, MSC from other sources, then placenta could become an important, unforeseen cell source, not only of hematopoietic stem cells (as demonstrated by placental cord blood), but also of MSCs.

Declaration for interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Bibliography

  • Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 1970;3:393-403
  • Parolini O, Alviano F, Bagnara GP, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international workshop on placenta derived stem cells. Stem Cells 2008;26:300-11
  • Corrao S, La Rocca G, Lo Iacono M, et al. Umbilical cord revisited: from Wharton’s jelly myofibroblasts to mesenchymal stem cells. Histol Histopathol 2013;28:1235-44
  • Parolini O, Soncini M, Evangelista M, et al. Amniotic membrane and amniotic fluid-derived cells: potential tools for regenerative medicine? Regen Med 2009;4:275-91
  • Fierabracci A, Del Fattore A, Luciano R, Muraca M. Recent advances in mesenchymal stem cell immunomodulation. The role of microvesicles. Cell Transplant 2015;24. 10.3727/096368913X675728
  • Uccelli A, Moretta L, Pistoia V. Immunoregulatory function of mesenchymal stem cells. Eur J Immunol 2006;36:2566-73
  • Kmiecik G, Spoldi V, Silini A, Parolini O. Current view on osteogenic differentiation potential of mesenchymal stromal cells derived from placental tissues. Stem Cell Rev 2014. 10.1007/s12015-014-9569-1
  • Rossi D, Pianta S, Magatti M, et al. Characterization of the conditioned medium from amniotic membrane cells: prostaglandins as key effectors of its immunomodulatory activity. PLoS One 2012;7:e46956
  • Zhu Y, Song X, Wang J, et al. Placental mesenchymal stem cells of fetal origin deposit epigenetic alterations during long-term culture under serum-free condition. Expert Opin Biol Ther 2014;1-18
  • Parolini O, De D, Rodrigues MF, et al. Placental stem/progenitor cells: isolation and characterization, In: Atala A, Murphy SV, editors. Perinatal stem cells. Springer Science, New York; 2014. p. 373
  • Parolini O, Soncini M. Placenta as a source of stem cells and as a key organ for fetomaternal tolerance. In: Bhattacharya N, Stubblefield P, editors Regenerative medicine using pregnancy-specific biological substances. Springer-Verlag, London; 2011: p. 460
  • Magatti M, De Munari S, Vertua E, et al. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cells 2008;26:182-92
  • Parolini O, Souza-Moreira L, O’Valle F, et al. Therapeutic effect of human amniotic membrane-derived cells on experimental arthritis and other inflammatory disorders. Arthritis Rheumatol 2014;66:327-39
  • Magatti M, De Munari S, Vertua E, et al. Amniotic mesenchymal tissue cells inhibit dendritic cell differentiation of peripheral blood and amnion resident monocytes. Cell Transplant 2009;18:899-914
  • Cargnoni A, Piccinelli EC, Ressel L, et al. Conditioned medium from amniotic membrane-derived cells prevents lung fibrosis and preserves blood gas exchanges in bleomycin-injured mice-specificity of the effects and insights into possible mechanisms. Cytotherapy 2014;16:17-32
  • Groh ME, Maitra B, Szekely E, et al. Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp Hematol 2005;33:928-34
  • Silini A, Parolini O, Huppertz B, Lang I. Soluble factors of amnion-derived cells in treatment of inflammatory and fibrotic pathologies. Curr Stem Cell Res Ther 2013;8:6-14
  • Pozzobon M, Piccoli M, De Coppi P. Stem cells from fetal membranes and amniotic fluid: markers for cell isolation and therapy. Cell Tissue Bank 2014;15:199-211
  • Wegmeyer H, Broske AM, Leddin M, et al. Mesenchymal stromal cell characteristics vary depending on their origin. Stem Cells Dev 2013;22:2606-18
  • Soncini M, Vertua E, Gibelli L, et al. Isolation and characterization of mesenchymal cells from human fetal membranes. J Tissue Eng Regen Med 2007;1:296-305
  • Heazlewood CF, Vertua E, Gibelli L, et al. High incidence of contaminating maternal cell overgrowth in human placental mesenchymal stem/stromal cell cultures: a systematic review. Stem Cells Transl Med 2014;3:1305-11
  • Mayer L, Pandak WM, Melmed GY, et al. Safety and tolerability of human placenta-derived cells (PDA001) in treatment-resistant Crohn’s disease: a Phase I study. Inflamm Bowel Dis 2013;19:754-60
  • Chambers DC, Enever D, Ilic N, et al. A phase 1b study of placenta-derived mesenchymal stromal cells in patients with idiopathic pulmonary fibrosis. Respirology 2014;19:1013-18
  • Schellenberg A, Lin Q, Schüler H, et al. Replicative senescence of mesenchymal stem cells causes DNA-methylation changes which correlate with repressive histone marks. Aging 2011;3:873-88
  • Park S, Kim E, Koh SE, et al. Dopaminergic differentiation of neural progenitors derived from placental mesenchymal stem cells in the brains of Parkinson’s disease model rats and alleviation of asymmetric rotational behavior. Brain Res 2012;1466:158-66
  • Yun HM, Kim HS, Park KR, et al. Placenta-derived mesenchymal stem cells improve memory dysfunction in an Abeta1-42-infused mouse model of Alzheimer’s disease. Cell Death Dis 2013;4:e958
  • Kim KS, Kim HS, Park JM, et al. Long-term immunomodulatory effect of amniotic stem cells in an Alzheimer’s disease model. Neurobiol Aging 2013;34:2408-20
  • Fisher-Shoval Y, Barhum Y, Sadan O, et al. Transplantation of placenta-derived mesenchymal stem cells in the EAE mouse model of MS. J Mol Neurosci 2012;48:176-84
  • Cargnoni A, Gibelli L, Tosini A, et al. Transplantation of allogeneic and xenogeneic placenta-derived cells reduces bleomycin-induced lung fibrosis. Cell Transplant 2009;18:405-22
  • Cargnoni A, Ressel L, Rossi D, et al. Conditioned medium from amniotic mesenchymal tissue cells reduces progression of bleomycin-induced lung fibrosis. Cytotherapy 2012;14:153-61
  • Ventura C, Cantoni S, Bianchi F, et al. Hyaluronan mixed esters of butyric and retinoic Acid drive cardiac and endothelial fate in term placenta human mesenchymal stem cells and enhance cardiac repair in infarcted rat hearts. J Biol Chem 2007;282:14243-52
  • Tsuji H, Miyoshi S, Ikegami Y, et al. Xenografted human amniotic membrane-derived mesenchymal stem cells are immunologically tolerated and transdifferentiated into cardiomyocytes. Circ Res 2010;106:1613-23
  • Vaghjiani V, Vaithilingam V, Tuch B, et al. Deriving hepatocyte-like cells from placental cells for transplantation. Curr Stem Cell Res Ther 2013;8:15-24

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.