Elsevier

Cytotherapy

Volume 19, Issue 7, July 2017, Pages 784-797
Cytotherapy

Review Article
Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy

https://doi.org/10.1016/j.jcyt.2017.03.076Get rights and content

Abstract

Mesenchymal stromal cells (MSCs) are multipotent cells derived from various tissues that can differentiate into several cell types. MSCs are able to modulate the response of immune cells of the innate and adaptive immune system. Because of these multimodal properties, the potential use of MSCs for immunotherapies is currently explored in various clinical indications. Due to the diversity of potential MSC medicinal products at the level of cell source, manufacturing process and indication, distinct functionality tests may be needed to ensure the quality for each of the different products. In this review, we focus on in vitro potency assays proposed for characterization and release of different MSC medicinal products. We discuss the most used functional assays, as presented in scientific advices and literature, highlighting specific advantages and limitations of the various assays. Currently, the most proposed and accepted potency assay for release is based on in vitro inhibition of T cell proliferation or other functionalities. However, for some products, assays based on other MSC or responder cell properties may be more appropriate. In all cases, the biological relevance of the proposed assay for the intended clinical activity should be substantiated with appropriate product-specific (non-)clinical data. In case practical considerations prevent the use of the ideal potency assay at release, use of a surrogate marker or test could be considered if correlation with functionality has been demonstrated. Nevertheless, as the field of MSC immunology is evolving, improvements can be expected in relevant assays and consequently in guidance related to potency testing.

Introduction

Mesenchymal stromal cells (MSCs) are multipotent cells that can differentiate into several cell types. MSCs can be isolated from various tissues and although their numbers are low and decrease with age, their potential to expand ex vivo may allow production of sufficient amounts for therapeutic use [1]. Human tissues from which MSCs have been isolated include bone marrow, adipose tissue, placental tissue, umbilical cords and embryonic stem cells [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11], [12]. Importantly, these MSCs differ in their expression of surface markers and in their functional capacity after stimulation with proinflammatory mediators [2], [13]. As a consequence, comparison of the clinical effects of the various MSC products is hampered. To create a broader consensus for more uniform characterization of MSCs, the International Society for Cellular Therapy (ISCT) proposed three minimal criteria to define human MSCs: (i) MSCs should be plastic-adherent when maintained in standard culture conditions; (ii) MSCs should express CD105, CD73 and CD90 and should lack CD45, CD34, CD14/CD11b, CD79α/CD19 and human leukocyte antigen (HLA)-DR expression; and (iii) MSCs should be able to differentiate (in vitro) into osteoblasts, adipocytes and chondroblasts [14].

However, the nonspecificity of these criteria has been a topic of discussion ever since [15], [16]. One should at least bear in mind that, in spite of above minimal criteria, MSCs remain heterologous populations of cells with a variety of gene expression profiles, differentiation and expansion potential and phenotype, which are influenced by tissue origin, cell isolation and expansion procedures [14], [17], [18], [19].

In vivo, MSCs play a crucial role in peripheral tissue homeostasis (including blood vessels) and maintenance of the (hematopoietic) stem cell niche, mainly due to their potential to differentiate into various cell types and their secretion of several growth-promoting factors [20], [21]. It has been shown that MSCs preferentially home to damaged tissue, where they are thought to exert their biological action by direct cell-cell interactions through their surface receptors, but also by producing soluble factors [18], [22], [23], [24].

Apart from their tissue homeostasis and regeneration capacities, MSCs also have immunomodulatory abilities with potential therapeutic applications. This review article focuses on the in vitro testing of the functionality of these MSC medicinal products.

Section snippets

Immunomodulation by MSCs

The general immunomodulatory role of MSCs involves the orchestration of immunologic tolerance, next to the role of regulatory T and B cells and innate suppressor cells [25]. MSCs function via direct suppression of the activation, proliferation and effector functions of proinflammatory cells and the stimulation of various anti-inflammatory cell types to indirectly augment immune response regulation [20]. The most prominent MSC functions are exerted locally, although also some systemic effects

Clinical application

As said, MSCs have a diversity of physiological functions including the ability to migrate to inflamed tissue, differentiate into various cell types and secrete anti-inflammatory and tissue-renewing factors. Due to their broad immunoregulatory potential, clinical application of these cells is explored for a wide range of disorders with high immune activation, including graft-versus-host disease (GvHD), transplant rejection and autoimmunity [26]. Because MSCs harbor low immunogenicity (despite

In vitro assays to test for MSC functionality

As said, three minimal criteria for defining human MSCs have been proposed [14]. In addition, Wuchter et al. posed several criteria for GMP-grade MSCs [74]. However, although the criteria address MSC functionality, no concluding recommendations regarding potency assays are provided. This is in line with the notion that every MSC product is unique and that quality control assays should be justified on a case-by-case basis. Nevertheless, the following overview of potency assays to test for MSC

Selection and design of potency assays

Despite the common use of immune cell inhibition and suppressor cell activation assays, there is still a lot of uncertainty with respect to which assay would be most appropriate to assess potency of MSC products. For instance, using a potency assay combining both MLR and mitogen stimulation, Ketterl et al. observed that some MSC batches showed little suppression in the mitogen-stimulated assay, while showing a much higher suppression in the MLR [5]. This would indicate that a specific batch

Future perspectives and conclusion

The majority of MSC medicinal products in clinical development are indicated for GvHD or transplant rejection. Nevertheless, several MSC products are currently tested for treatment of chronic inflammation in autoimmune diseases, for example, systemic lupus erythematosus, systemic sclerosis, type 1 diabetes mellitus, multiple sclerosis, Crohn's disease and rheumatoid arthritis [2], [19], [21], [32], [88], [89]. Although most potential MSC products for these indications are still in experimental

Acknowledgments

We thank Dr. Carla Herberts for useful comments and suggestions on the manuscript. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Disclosure of interests: The authors have no commercial, proprietary or financial interest in the products described in this article.

References (89)

  • S. Glennie et al.

    Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells

    Blood

    (2005)
  • R. Ramasamy et al.

    The immunosuppressive effects of human bone marrow-derived mesenchymal stem cells target T cell proliferation but not its effector function

    Cell Immunol

    (2008)
  • K. Le Blanc et al.

    HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells

    Exp Hematol

    (2003)
  • M. Najar et al.

    Characterization and functionality of the CD200-CD200R system during mesenchymal stromal cell interactions with T-lymphocytes

    Immunol Lett

    (2012)
  • P. Kebriaei et al.

    Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease

    Biol Blood Marrow Transplant

    (2009)
  • J.J. Ruitenberg et al.

    Rapid assessment of in vitro expanded human regulatory T cell function

    J Immunol Methods

    (2011)
  • M. Najar et al.

    Impact of different mesenchymal stromal cell types on T-cell activation, proliferation and migration

    Int Immunopharmacol

    (2013)
  • YangH. et al.

    Human umbilical cord-derived mesenchymal stem cells suppress proliferation of PHA-activated lymphocytes in vitro by inducing CD4+CD25highCD45RA+ regulatory T cell production and modulating cytokine secretion

    Cell Immunol

    (2016)
  • D.D. Bloom et al.

    A reproducible immunopotency assay to measure mesenchymal stromal cell-mediated T-cell suppression

    Cytotherapy

    (2015)
  • R.N. Bárcia et al.

    Umbilical cord tissue–derived mesenchymal stromal cells maintain immunomodulatory and angiogenic potencies after cryopreservation and subsequent thawing

    Cytotherapy

    (2017)
  • A. Bartholomew et al.

    Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo

    Exp Hematol

    (2002)
  • L. von Bahr et al.

    Long-term complications, immunologic effects, and role of passage for outcome in mesenchymal stromal cell therapy

    Biol Blood Marrow Transplant

    (2012)
  • E. Valencic et al.

    The immunosuppressive effect of Wharton's jelly stromal cells depends on the timing of their licensing and on lymphocyte activation

    Cytotherapy

    (2010)
  • P. Wuchter et al.

    Standardization of Good Manufacturing Practice-compliant production of bone marrow-derived human mesenchymal stromal cells for immunotherapeutic applications

    Cytotherapy

    (2015)
  • D.H. Munn et al.

    Indoleamine 2,3 dioxygenase and metabolic control of immune responses

    Trends Immunol

    (2013)
  • A.L. Mellor et al.

    Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation?

    Immunol Today

    (1999)
  • T.B. Prigozhina et al.

    Mesenchymal stromal cells lose their immunosuppressive potential after allotransplantation

    Exp Hematol

    (2008)
  • J. Galipeau et al.

    International society for cellular therapy perspective on immune functional assays for mesenchymal stromal cells as potency release criterion for advanced phase clinical trials

    Cytotherapy

    (2016)
  • J. Galipeau et al.

    The challenge of defining mesenchymal stromal cell potency assays and their potential use as release criteria

    Cytotherapy

    (2015)
  • L.L. Kenney et al.

    Humanized mouse models for transplant immunology

    Am J Transplant

    (2016)
  • I. Ullah et al.

    Human mesenchymal stem cells—current trends and future prospective

    Biosci Rep

    (2015)
  • P.J. Tsai et al.

    Undifferentiated Wharton's jelly mesenchymal stem cell transplantation induces insulin-producing cell differentiation and suppression of T-cell-mediated autoimmunity in nonobese diabetic mice

    Cell Transplant

    (2015)
  • R.N. Bárcia et al.

    What makes umbilical cord tissue-derived mesenchymal stromal cells superior immunomodulators when compared to bone marrow derived mesenchymal stromal cells?

    Stem Cells Int

    (2015)
  • N. Ketterl et al.

    A robust potency assay highlights significant donor variation of human mesenchymal stem/progenitor cell immune modulatory capacity and extended radio-resistance

    Stem Cell Res Ther

    (2015)
  • P. Laranjeira et al.

    Human bone marrow-derived mesenchymal stromal cells differentially inhibit cytokine production by peripheral blood monocytes subpopulations and myeloid dendritic cells

    Stem Cells Int

    (2015)
  • P. Laranjeira et al.

    Effect of human bone marrow mesenchymal stromal cells on cytokine production by peripheral blood naive, memory, and effector T cells

    Stem Cell Res Ther

    (2015)
  • E.A. Kimbrel et al.

    Mesenchymal stem cell population derived from human pluripotent stem cells displays potent immunomodulatory and therapeutic properties

    Stem Cells Dev

    (2014)
  • B. Puissant et al.

    Immunomodulatory effect of human adipose tissue-derived adult stem cells: comparison with bone marrow mesenchymal stem cells

    Br J Haematol

    (2005)
  • P. Hematti

    Characterization of mesenchymal stromal cells: potency assay development

    Transfusion

    (2016)
  • P. Bianco et al.

    The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine

    Nat Med

    (2013)
  • K. Le Blanc et al.

    Immunomodulation by mesenchymal stem cells and clinical experience

    J Intern Med

    (2007)
  • A. Uccelli et al.

    Mesenchymal stem cells in health and disease

    Nat Rev Immunol

    (2008)
  • J. Tolar et al.

    Concise review: hitting the right spot with mesenchymal stromal cells

    Stem Cells

    (2010)
  • ZhaoS. et al.

    Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders

    Stem Cells Dev

    (2010)
  • Cited by (89)

    View all citing articles on Scopus
    View full text