Elsevier

Cytotherapy

Volume 18, Issue 9, September 2016, Pages 1197-1208
Cytotherapy

Cell manufacture
Design and validation of a consistent and reproducible manufacture process for the production of clinical-grade bone marrow–derived multipotent mesenchymal stromal cells

https://doi.org/10.1016/j.jcyt.2016.05.012Get rights and content

Abstract

Background

Multipotent mesenchymal stromal cells (MSC) have achieved a notable prominence in the field of regenerative medicine, despite the lack of common standards in the production processes and suitable quality controls compatible with Good Manufacturing Practice (GMP). Herein we describe the design of a bioprocess for bone marrow (BM)–derived MSC isolation and expansion, its validation and production of 48 consecutive batches for clinical use.

Methods

BM samples were collected from the iliac crest of patients for autologous therapy. Manufacturing procedures included: (i) isolation of nucleated cells (NC) by automated density-gradient centrifugation and plating; (ii) trypsinization and expansion of secondary cultures; and (iii) harvest and formulation of a suspension containing 40 ± 10 × 106 viable cells. Quality controls were defined as: (i) cell count and viability assessment; (ii) immunophenotype; and (iii) sterility tests, Mycoplasma detection, endotoxin test and Gram staining.

Results

A 3-week manufacturing bioprocess was first designed and then validated in 3 consecutive mock productions, prior to producing 48 batches of BM-MSC for clinical use. Validation included the assessment of MSC identity and genetic stability. Regarding production, 139.0 ± 17.8 mL of BM containing 2.53 ± 0.92 × 109 viable NC were used as starting material, yielding 38.8 ± 5.3 × 106 viable cells in the final product. Surface antigen expression was consistent with the expected phenotype for MSC, displaying high levels of CD73, CD90 and CD105, lack of expression of CD31 and CD45 and low levels of HLA-DR. Tests for sterility, Mycoplasma, Gram staining and endotoxin had negative results in all cases.

Discussion

Herein we demonstrated the establishment of a feasible, consistent and reproducible bioprocess for the production of safe BM-derived MSC for clinical use.

Introduction

Recent scientific advances preclude that regenerative medicine will become an essential component of medical care in the near future [1], [2]. The high number and scope of currently active clinical trials will likely make new treatments available to patients in a growing wave of medicinal cell-based products [3], [4]. As a consequence of developments in this emerging field, a specific regulatory framework introducing new concepts such as Advanced Therapy Medical Products (ATMP) has developed on methodologies taken from traditional pharmaceutical drugs [5], which are governed in Europe by the Directive 2001/83/EC and Regulation 726/2004, amended by Regulation 1394/2007.

Among all adult somatic cells proposed for clinical application, multipotent mesenchymal stromal cells (MSC) have reached a major prominence. In particular, MSC isolated from bone marrow (BM) display a number of biological properties that qualify them for clinical use, either based on their stem or stromal properties [6], [7]. These include migratory, homing and differentiation potential toward mesodermal lineages [8], [9], [10], as well as a powerful paracrine activity involving regulation of inflammation, immune response and tissue regeneration [11], [12]. Such pleiotropic activity has served as rationale in several hundred clinical trials worldwide, mostly in the autologous therapy setting. However, the major weakness is the lack of common standards in cell production processes and their quality controls, thus challenging the reliability of meta-analyses conducted on data from those clinical trials. In this sense, adherence to white papers and compliance with voluntary accreditation schemes and mandatory Good Scientific Practice (GxP) regulations may play a major role in the understanding and comparability of clinical results when using MSC-based medicines [13], [14], [15], [16].

In the present work, we described the design and validation of a Good Manufacturing Practice (GMP)-compliant manufacture process within a small academic laboratory and the subsequent production of 48 consecutive GMP-compliant batches of BM-MSC for use in a Phase I/IIa clinical trial for the treatment of chronic osteoarthritis and compassionate treatments [17], thus confirming the feasibility, consistency and reproducibility of the bioprocess, and safety of the ATMP. It includes a description of quality controls, key technical aspects and areas of potential improvement on standardization and quality assurance.

Section snippets

Production of clinical-grade BM-MSC

Within the context of a prospective, open-label, single-dose, single-arm Phase I–IIa clinical trial (Eudra-CT, 2009-016449-24; ClinicalTrials.gov identifier, NCT01227694) conducted by Institut de Teràpia Regenerativa Tissular (ITRT) at the Hospital Quirón Teknon (HQT) from October 2010 to June 2012, and compassionate treatments related to the clinical study, a cell-based therapy product for clinical use was manufactured in GMP-certified facilities (Banc de Sang i Teixits) [18], [19].

Definition and validation of the bioprocess

A bioprocess aimed at producing BM-MSC yields in the range of 80–100 × 106 was designed to supply enough cells for clinical use and performance of Quality Controls (QC). This was achieved by seeding cells at low density in a primary culture after gradient separation of NC from BM, and further expansion through a second passage. The culture time spanned for approximately 21 days and 8–13 CPD (Figure 1; Table II). No evidence of turbidity was observed in any step of the 3 runs assayed in the

Discussion

A bioprocess for the manufacture of BM-MSC was successfully designed, validated and used in the production of 48 consecutive GMP-compliant batches for clinical application. This approach is compatible with small GMP facilities, as the ones that may be typically found in academic institutions or hospitals. As far as we are concerned, this is one of the few articles describing the complete procedure including validation, manufacturing and quality assessment of MSC-based products for clinical

Acknowledgments

The authors would like to acknowledge A. Pla, J.J. Cairó, F. Gòdia, R. Soler and L. Orozco for their support and advice; M. Caminal, L.Vidal, N. de la Fuente and S. Barbosa for technical assistance; and M. Rosal, A. Rojo, C. León and E. Morón from “Estabulari de l'Institut de Recerca” (Hospital de la Vall d'Hebron, Barcelona, Spain) for their careful assistance with animal management. This work was supported by “Ministerio de Economía y Competitividad” (grant number IPT-300000-2010-0017), “

References (28)

  • C.K. Schneider et al.

    Challenges with advanced therapy medicinal products and how to meet them

    Nat Rev Drug Discov

    (2010)
  • D.J. Prockop

    Marrow stromal cells as stem cells for nonhematopoietic tissues

    Science

    (1997)
  • A.J. Friedenstein et al.

    Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers

    Cell Tissue Kinet

    (1987)
  • RenG. et al.

    Concise review: mesenchymal stem cells and translational medicine: emerging issues

    Stem Cells Transl Med

    (2012)
  • Cited by (41)

    • A pilot study of circulating levels of TGF-β1 and TGF-β2 as biomarkers of bone healing in patients with non-hypertrophic pseudoarthrosis of long bones

      2022, Bone Reports
      Citation Excerpt :

      No previous sample size calculation was made because of the pilot nature of this study. Patients were randomised to either one of the two study treatments described next: 1) Treatment A (experimental): mechanical stabilisation (if required) associated with a Tissue Engineering Product (TEP composed of ex vivo expanded autologous Mesenchymal Stromal Cells (MSC) loaded onto allogeneic cancellous bone graft described previously (Prat et al., 2018; García de Frutos et al., 2020) and prepared in accordance with current Good Manufacturing Practices as reported elsewhere (Codinach et al., 2016; Vives et al., 2021; García-Muñoz and Vives, 2021); and 2) Treatment B (control): mechanical stabilisation, if required, associated with autologous iliac crest graft (gold standard) (Figs. 1 and 2). After treatment, patients were followed for a period of 12 months with monthly control radiographs (Rx) until 6 months and then at 9 and 12 months, and computerised tomography (CT) at 12 months.

    • Considerations on chemistry, manufacturing, and control of stem cell products for Investigational New Drug application in China

      2020, Biologicals
      Citation Excerpt :

      In the early stages of development, the lab-scale manufacturing process is often adopted, for example using small-scale dishes/flasks to produce cells with limited numbers. The reproducibility of such manufacturing process, control of aseptic operations, product quality maintenance is difficult to ensure in this occasion [4]. For clinical batch production, it is recommended to optimize the process, adjust the scale and maintain the batch-to batch quality consistency to meet the clinical trial need.

    • Strategies for large-scale expansion of clinical-grade human multipotent mesenchymal stromal cells

      2020, Biochemical Engineering Journal
      Citation Excerpt :

      Although autologous hSer has been reported to support MSC expansion, it would be difficult to procure it in sufficient quantities to generate clinically relevant numbers of MSC [48,49]. Allogeneic human AB serum would circumvent this issue as several donor serums are pooled to eliminate donor-specific differences and be produced in a large-scale manner but AB serum is scarce and human serum B (hSerB) can be used instead, performing as well as FBS [6,7,44,50]. It has also been reported that human platelet lysate (hPL) or platelet-rich plasma (PRP) have considerable growth-promoting properties for MSC while maintaining their differentiation potential and immunomodulatory properties [51,52].

    View all citing articles on Scopus
    View full text