Production and quality testing of multipotent mesenchymal stromal cell therapeutics for clinical use

I n 1867 Cohnheim proposed bone marrow (BM) as a source of fibroblasts involved in wound-healing processes. In the 1970s, Friedenstein reported the colonyforming unit fibrobast describing nonhematopoietic fibroblast-like cells from the BM with self-renewal capacity, multilineage differentiation potential in vitro, and hematopoietic support in vivo referring to them as stromal cells. Analogous to the hematopoietic system, Caplan popularized the term mesenchymal stem cells, and the mesodermal lineage differentiation prompted their clinical evaluation in cancer and osteogenesis imperfecta patients. Increasing interest stimulated a number of diverse studies yielding heterogeneous results. Minimal criteria, therefore, have been suggested to try to characterize MSCs, then depicted as “multipotent mesenchymal stromal cells,” based on a variety of properties including adherence (plastic-adherent fibroblastoid cells), cell marker expression (positive for CD105, CD73, CD90; negative for CD45, CD34, CD14 or CD11b, CD79 or CD19; and human leukocyte antigen [HLA] Class II), and differentiation in vitro (i.e., osteoblasts, chondrocytes, and adipocytes). MSC therapies have been applied in numerous clinical trials targeting various diseases such as ischemic (i.e., stroke, myocardial infarction, limb ischemia), pulmonary (i.e., respiratory distress syndrome, emphysema), and neurologic pathologies (i.e., amyotrophic lateral sclerosis, spinal cord injury), as well as skeletal degeneration (i.e., osteoarthritis, degenerative disk disease), with a specific focus on immunopathologies such as graft-versus-host disease (GvHD), Crohn disease, or multiple sclerosis. Additionally, MSCs have emerged as a versatile and promising candidate in the field of regenerative medicine and tissue engineering (e.g., to be seeded on scaffolds before implantation). Despite the increasing application of MSC-based therapies in a wide range of disease settings, sustainable translation and marketability may be limited, due to the complexity of MSC products. Variable tissue sources and heterogeneity between donors as well as within the MSC preparations (see below) are main drivers of the complexity that is hampering the identification of their molecular mechanism(s) of action (MoAs). Current concepts of the MoAs include production and/or secretion of cytokines and trophic factors, as well as secretion of microvesicles that contribute to the immunomodulatory and regenerative potential of MSC. Without clear evidence of specific MoAs that can be linked to clinical effects, it is difficult to establish consistent manufacturing conditions, quality criteria, and assays that predict efficacy. Potency is a therapeutically relevant biologic function that can be measured quantitatively by in vitro or in vivo assays that could be linked to properly controlled clinical efficacy data. Potency assays are critical to control for quality and consistency of MSC-based products; however, to date, there are few published data on potency assays for MSCs. The majority of proposed potency assays refer to their immunomodulatory and proangiogenic capabilities; however, a direct link to clinical efficacy is still lacking. We examine the current challenges and concepts as well as discuss possible future options for the production and quality testing of MSC therapeutics.

[1]  D. Strunk,et al.  Manufacturing Mesenchymal Stromal Cells for the Treatment of Graft-versus-Host Disease: A Survey among Centers Affiliated with the European Society for Blood and Marrow Transplantation , 2018, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[2]  J. Burns,et al.  Versatile graphene biosensors for enhancing human cell therapy. , 2018, Biosensors & bioelectronics.

[3]  C. Simmons,et al.  Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering , 2018, Stem cells international.

[4]  M. Ikeya,et al.  Generation and Applications of Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells , 2018, Stem cells international.

[5]  Anthony Atala,et al.  An Industry‐Driven Roadmap for Manufacturing in Regenerative Medicine , 2018, Stem cells translational medicine.

[6]  Robert Wilson Design of experiment in assessing robustness and for qualification of a cell-based potency assay. , 2018, Bioanalysis.

[7]  Q. Guan,et al.  Inducible indoleamine 2,3-dioxygenase 1 and programmed death ligand 1 expression as the potency marker for mesenchymal stromal cells. , 2018, Cytotherapy.

[8]  F. Cerignoli,et al.  In vitro immunotherapy potency assays using real-time cell analysis , 2018, PloS one.

[9]  G. Gibson,et al.  Potency Analysis of Mesenchymal Stromal Cells Using a Combinatorial Assay Matrix Approach. , 2018, Cell reports.

[10]  M. Introna,et al.  Utility of routine evaluation of sterility of cellular therapy products with or without extensive manipulation: Best practices and clinical significance. , 2018, Cytotherapy.

[11]  G. Lucchini,et al.  Effective treatment of steroid and therapy-refractory acute graft-versus-host disease with a novel mesenchymal stromal cell product (MSC-FFM) , 2018, Bone Marrow Transplantation.

[12]  J. Vives,et al.  Optimisation of a potency assay for the assessment of immunomodulative potential of clinical grade multipotent mesenchymal stromal cells , 2018, Cytotechnology.

[13]  C. Librach,et al.  Angiogenic potency evaluation of cell therapy candidates by a novel application of the in vitro aortic ring assay , 2017, Stem Cell Research & Therapy.

[14]  M. Hoefnagel,et al.  Regulatory perspective on in vitro potency assays for human mesenchymal stromal cells used in immunotherapy. , 2017, Cytotherapy.

[15]  D. Sumstad,et al.  Impact of starting material (fresh versus cryopreserved marrow) on mesenchymal stem cell culture , 2017, Transfusion.

[16]  G. Lajoie,et al.  High Aldehyde Dehydrogenase Activity Identifies a Subset of Human Mesenchymal Stromal Cells with Vascular Regenerative Potential , 2017, Stem cells.

[17]  H. Hackstein,et al.  Immunosuppressive capacity of mesenchymal stem cells correlates with metabolic activity and can be enhanced by valproic acid , 2017, Stem Cell Research & Therapy.

[18]  M. Pittenger,et al.  Concise Review: MSC‐Derived Exosomes for Cell‐Free Therapy , 2017, Stem cells.

[19]  A. Majumdar,et al.  Development of a surrogate potency assay to determine the angiogenic activity of Stempeucel®, a pooled, ex-vivo expanded, allogeneic human bone marrow mesenchymal stromal cell product , 2017, Stem Cell Research & Therapy.

[20]  F. Catani,et al.  Potency Biomarker Signature Genes from Multiparametric Osteogenesis Assays: Will cGMP Human Bone Marrow Mesenchymal Stromal Cells Make Bone? , 2016, PloS one.

[21]  J. García-López,et al.  Design and validation of a consistent and reproducible manufacture process for the production of clinical-grade bone marrow-derived multipotent mesenchymal stromal cells. , 2016, Cytotherapy.

[22]  T. Ritter,et al.  Development of a flow cytometry-based potency assay for measuring the in vitro immunomodulatory properties of mesenchymal stromal cells. , 2016, Immunology letters.

[23]  A. Dietz,et al.  Identification and validation of multiple cell surface markers of clinical-grade adipose-derived mesenchymal stromal cells as novel release criteria for good manufacturing practice-compliant production , 2016, Stem Cell Research & Therapy.

[24]  G. Lucchini,et al.  Mesenchymal stromal cells from pooled mononuclear cells of multiple bone marrow donors as rescue therapy in pediatric severe steroid-refractory graft-versus-host disease: a multicenter survey , 2016, Haematologica.

[25]  R. Schäfer,et al.  Mesenchymal Stem/Stromal Cells in Regenerative Medicine: Can Preconditioning Strategies Improve Therapeutic Efficacy , 2016, Transfusion Medicine and Hemotherapy.

[26]  A. Ho,et al.  Indoleamine 2,3-dioxygenase mediates inhibition of virus-specific CD8(+) T cell proliferation by human mesenchymal stromal cells. , 2016, Cytotherapy.

[27]  P. Hematti Characterization of mesenchymal stromal cells: potency assay development , 2016, Transfusion.

[28]  K. Bieback,et al.  Characterization of mesenchymal stem or stromal cells: tissue sources, heterogeneity, and function , 2016, Transfusion.

[29]  Peter W Zandstra,et al.  Quality cell therapy manufacturing by design , 2016, Nature Biotechnology.

[30]  A. Schambach,et al.  Massive Clonal Selection and Transiently Contributing Clones During Expansion of Mesenchymal Stem Cell Cultures Revealed by Lentiviral RGB‐Barcode Technology , 2016, Stem cells translational medicine.

[31]  T. Bliss,et al.  High-Resolution Microfluidic Single-Cell Transcriptional Profiling Reveals Clinically Relevant Subtypes among Human Stem Cell Populations Commonly Utilized in Cell-Based Therapies , 2016, Front. Neurol..

[32]  M. Toungouz,et al.  The Immunomodulatory Potential of Mesenchymal Stromal Cells: A Story of a Regulatory Network , 2016, Journal of immunotherapy.

[33]  S. Ferguson,et al.  An in vitro expansion score for tissue‐engineering applications with human bone marrow‐derived mesenchymal stem cells , 2016, Journal of tissue engineering and regenerative medicine.

[34]  Ivan Martin,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. , 2016, Cytotherapy.

[35]  L. Turchetto,et al.  Quality Control Assays for Clinical-Grade Human Mesenchymal Stromal Cells: Methods for ATMP Release. , 2016, Methods in molecular biology.

[36]  L. Turchetto,et al.  Quality Control Assays for Clinical-Grade Human Mesenchymal Stromal Cells: Validation Strategy. , 2016, Methods in molecular biology.

[37]  M. Pittenger,et al.  Unraveling the Mesenchymal Stromal Cells' Paracrine Immunomodulatory Effects. , 2016, Transfusion medicine reviews.

[38]  K. Keyvanfar,et al.  Quantitative activation suppression assay to evaluate human bone marrow-derived mesenchymal stromal cell potency. , 2015, Cytotherapy.

[39]  A. Reinisch,et al.  A robust potency assay highlights significant donor variation of human mesenchymal stem/progenitor cell immune modulatory capacity and extended radio-resistance , 2015, Stem Cell Research & Therapy.

[40]  Vivek Tanavde,et al.  Research using Mesenchymal Stem/Stromal Cells: quality metric towards developing a reference material. , 2015, Cytotherapy.

[41]  P. Kruzliak,et al.  Validation of shortened 2-day sterility testing of mesenchymal stem cell-based therapeutic preparation on an automated culture system , 2015, Cell and Tissue Banking.

[42]  Michael Raghunath,et al.  Establishing Criteria for Human Mesenchymal Stem Cell Potency , 2015, Stem cells.

[43]  M. Boutros,et al.  Functional fingerprinting of human mesenchymal stem cells using high-throughput RNAi screening , 2015, Genome Medicine.

[44]  M. Mcgrogan,et al.  Cell injury-induced release of FGF2: relevance to intracerebral mesenchymal stromal cell transplantations. , 2015 .

[45]  TorreMaria Luisa,et al.  Ex Vivo Expanded Mesenchymal Stromal Cell Minimal Quality Requirements for Clinical Application , 2015 .

[46]  K. Hoffmann,et al.  Cryopreservation does not alter main characteristics of Good Manufacturing Process-grade human multipotent mesenchymal stromal cells including immunomodulating potential and lack of malignant transformation. , 2015, Cytotherapy.

[47]  John M Centanni,et al.  A reproducible immunopotency assay to measure mesenchymal stromal cell-mediated T-cell suppression. , 2015, Cytotherapy.

[48]  B. Thompson,et al.  Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. , 2015, The Lancet. Respiratory medicine.

[49]  J. Trouvin,et al.  The challenges of potency assay development for cell-based medicinal products in Europe , 2015 .

[50]  D. Prockop,et al.  TSG-6 as a biomarker to predict efficacy of human mesenchymal stem/progenitor cells (hMSCs) in modulating sterile inflammation in vivo , 2014, Proceedings of the National Academy of Sciences.

[51]  Asha A. Nair,et al.  High‐Resolution Molecular Validation of Self‐Renewal and Spontaneous Differentiation in Clinical‐Grade Adipose‐Tissue Derived Human Mesenchymal Stem Cells , 2014, Journal of cellular biochemistry.

[52]  B. Nilsson,et al.  Cryopreserved Mesenchymal Stromal Cells Display Impaired Immunomodulatory and Therapeutic Properties ? , 2022 .

[53]  Yaojiong Wu,et al.  Epigenetic changes of mesenchymal stem cells in three-dimensional (3D) spheroids , 2014, Journal of cellular and molecular medicine.

[54]  ViswanathanSowmya,et al.  Soliciting strategies for developing cell-based reference materials to advance mesenchymal stromal cell research and clinical translation. , 2014 .

[55]  J. Daniels,et al.  Challenges in the development of a reference standard and potency assay for the clinical production of RAFT tissue equivalents for the cornea. , 2014, Regenerative medicine.

[56]  R. Puri,et al.  MSC-based product characterization for clinical trials: an FDA perspective. , 2014, Cell stem cell.

[57]  J. James,et al.  Can we fix it? Evaluating the potential of placental stem cells for the treatment of pregnancy disorders. , 2014, Placenta.

[58]  A. Hmadcha,et al.  Standard requirement of a microbiological quality control program for the manufacture of human mesenchymal stem cells for clinical use. , 2014, Stem cells and development.

[59]  E. Alici,et al.  Towards an advanced therapy medicinal product based on mesenchymal stromal cells isolated from the umbilical cord tissue: quality and safety data , 2014, Stem Cell Research & Therapy.

[60]  K. Tarte,et al.  Immunological characterization of multipotent mesenchymal stromal cells--The International Society for Cellular Therapy (ISCT) working proposal. , 2013, Cytotherapy.

[61]  K. Tarte,et al.  Risk of tumorigenicity in mesenchymal stromal cell-based therapies--bridging scientific observations and regulatory viewpoints. , 2013, Cytotherapy.

[62]  Joelle Dulong,et al.  Clinical-grade mesenchymal stromal cells produced under various good manufacturing practice processes differ in their immunomodulatory properties: standardization of immune quality controls. , 2013, Stem cells and development.

[63]  H. Northoff,et al.  Phenotype, donor age and gender affect function of human bone marrow-derived mesenchymal stromal cells , 2013, BMC Medicine.

[64]  Mélanie Gadelorge,et al.  Production of mesenchymal stromal/stem cells according to good manufacturing practices: a review , 2013, Stem Cell Research & Therapy.

[65]  M. Neubauer,et al.  Mesenchymal stromal cell characteristics vary depending on their origin. , 2013, Stem cells and development.

[66]  L. Moroni,et al.  Human mesenchymal stem cells: A bank perspective on the isolation, characterization and potential of alternative sources for the regeneration of musculoskeletal tissues , 2013, Journal of cellular physiology.

[67]  P. de Coppi,et al.  Therapeutic potential of amniotic fluid stem cells. , 2013, Current stem cell research & therapy.

[68]  D. Covas,et al.  Differential expression of AURKA and AURKB genes in bone marrow stromal mesenchymal cells of myelodysplastic syndrome: correlation with G-banding analysis and FISH. , 2013, Experimental hematology.

[69]  Jacques Galipeau,et al.  The mesenchymal stromal cells dilemma--does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graft-versus-host disease represent a death knell or a bump in the road? , 2013, Cytotherapy.

[70]  Christopher A Bravery,et al.  Potency assay development for cellular therapy products: an ISCT review of the requirements and experiences in the industry. , 2013, Cytotherapy.

[71]  D. Phinney Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy , 2012, Journal of cellular biochemistry.

[72]  M. Toungouz,et al.  Characterization and functionality of the CD200-CD200R system during mesenchymal stromal cell interactions with T-lymphocytes. , 2012, Immunology letters.

[73]  Rochelle Cutrone,et al.  Development of a surrogate angiogenic potency assay for clinical-grade stem cell production. , 2012, Cytotherapy.

[74]  Byung-Soo Kim,et al.  Transplantation of cord blood mesenchymal stem cells as spheroids enhances vascularization. , 2012, Tissue engineering. Part A.

[75]  T. Redmer,et al.  Replicative aging and differentiation potential of human adipose tissue-derived mesenchymal stromal cells expanded in pooled human or fetal bovine serum. , 2012, Cytotherapy.

[76]  T. Young,et al.  The influence of spheroid formation of human adipose-derived stem cells on chitosan films on stemness and differentiation capabilities. , 2012, Biomaterials.

[77]  R. Romieu-Mourez,et al.  Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing , 2011, Cytotherapy.

[78]  Katie C. Russell,et al.  Clonal analysis of the proliferation potential of human bone marrow mesenchymal stem cells as a function of potency , 2011, Biotechnology and bioengineering.

[79]  Arnold I Caplan,et al.  The MSC: an injury drugstore. , 2011, Cell stem cell.

[80]  M. Najar,et al.  A rapid, simple, and reproducible method for the isolation of mesenchymal stromal cells from Wharton's jelly without enzymatic treatment. , 2011, Stem cells and development.

[81]  M. Yarmush,et al.  A mesenchymal stem cell potency assay. , 2011, Methods in molecular biology.

[82]  Soichiro Saito,et al.  Use of BAC array CGH for evaluation of chromosomal stability of clinically used human mesenchymal stem cells and of cancer cell lines , 2011, Human Cell.

[83]  Nikolay Bazhanov,et al.  Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties , 2010, Proceedings of the National Academy of Sciences.

[84]  Katie C. Russell,et al.  In Vitro High‐Capacity Assay to Quantify the Clonal Heterogeneity in Trilineage Potential of Mesenchymal Stem Cells Reveals a Complex Hierarchy of Lineage Commitment , 2010, Stem cells.

[85]  T. Moccetti,et al.  A practical approach for the validation of sterility, endotoxin and potency testing of bone marrow mononucleated cells used in cardiac regeneration in compliance with good manufacturing practice , 2009, Journal of Translational Medicine.

[86]  L. Isola,et al.  Adult human mesenchymal stem cells added to corticosteroid therapy for the treatment of acute graft-versus-host disease. , 2009, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[87]  A. Reinisch,et al.  Rapid large-scale expansion of functional mesenchymal stem cells from unmanipulated bone marrow without animal serum. , 2008, Tissue engineering. Part C, Methods.

[88]  Peter R. Brink,et al.  Culturing of Human Mesenchymal Stem Cells as Three-dimensional Aggregates Induces Functional Expression of CXCR4 That Regulates Adhesion to Endothelial Cells* , 2008, Journal of Biological Chemistry.

[89]  R. Consolini,et al.  Isolation of multipotent mesenchymal stem cells from umbilical cord blood , 2006 .

[90]  D. Prockop,et al.  Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. , 2006, Cytotherapy.

[91]  O. Lee,et al.  Isolation of multipotent mesenchymal stem cells from umbilical cord blood. , 2004, Blood.

[92]  H. Lorenz,et al.  Multilineage cells from human adipose tissue: implications for cell-based therapies. , 2001, Tissue engineering.

[93]  Arnold I. Caplan,et al.  Mesenchymal Stem Cells , 1991, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[94]  H. Broxmeyer,et al.  Characterization of human bone marrow fibroblast colony-forming cells (CFU-F) and their progeny. , 1980, Blood.

[95]  A. Friedenstein,et al.  STROMAL CELLS RESPONSIBLE FOR TRANSFERRING THE MICROENVIRONMENT OF THE HEMOPOIETIC TISSUES: Cloning In Vitro and Retransplantation In Vivo , 1974, Transplantation.

[96]  A. Friedenstein,et al.  Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. , 1968, Transplantation.

[97]  Reflection paper on stem cell-based medicinal products , 2022 .