Elsevier

Cytotherapy

Volume 21, Issue 12, December 2019, Pages 1216-1233
Cytotherapy

Full-Length Article
Manufacturing Processes
Cryopreservation timing is a critical process parameter in a thymic regulatory T-cell therapy manufacturing protocol

https://doi.org/10.1016/j.jcyt.2019.10.011Get rights and content

Highlights

  • Tested GMP-compatible activation reagents and media for Treg expansion.

  • Tregs that return to resting size (∼8.5 μm) expand more upon restimulation.

  • Timing of Treg cryopreservation effects viability, phenotype and function.

  • Cryopreserved thymic Tregs could be used as an off-the-shelf cell therapy product.

Abstract

Regulatory T cells (Tregs) are a promising therapy for several immune-mediated conditions but manufacturing a homogeneous and consistent product, especially one that includes cryopreservation, has been challenging. Discarded pediatric thymuses are an excellent source of therapeutic Tregs with advantages including cell quantity, homogeneity and stability. Here we report systematic testing of activation reagents, cell culture media, restimulation timing and cryopreservation to develop a Good Manufacturing Practice (GMP)–compatible method to expand and cryopreserve Tregs. By comparing activation reagents, including soluble antibody tetramers, antibody-conjugated beads and artificial antigen-presenting cells (aAPCs) and different media, we found that the combination of Dynabeads Treg Xpander and ImmunoCult-XF medium preserved FOXP3 expression and suppressive function and resulted in expansion that was comparable with a single stimulation with aAPCs. Cryopreservation tests revealed a critical timing effect: only cells cryopreserved 1–3 days, but not >3 days, after restimulation maintained high viability and FOXP3 expression upon thawing. Restimulation timing was a less critical process parameter than the time between restimulation and cryopreservation. This systematic testing of key variables provides increased certainty regarding methods for in vitro expansion and cryopreservation of Tregs. The ability to cryopreserve expanded Tregs will have broad-ranging applications including enabling centralized manufacturing and long-term storage of cell products.

Introduction

Regulatory T cell (Treg) therapy is a promising approach to prevent or treat graft-versus-host disease following hematopoietic stem cell transplantation, graft rejection following solid organ transplantation, or autoimmune conditions [1]. Several phase 1 clinical trials have been completed [2], [3], [4], [5], [6], [7], [8], [9], [10], [11] and phase 1/2 clinical trials of Treg therapy are underway (reviewed in [12]). However, significant obstacles remain to the broad use of Treg-based therapies, including developing and optimizing manufacturing protocols to generate large numbers of cells with the desired phenotype and function, while limiting manufacturing complexity and cost [13].

We have recently shown that discarded pediatric thymuses are a feasible source of clinically applicable Tregs, with numerous advantages over blood- or cord blood–derived cells. Specifically, thymus-derived Tregs are abundant [14] and, because in the thymus CD25 expression on CD4+CD8 T cells exclusively marks Tregs [15], a homogeneous population of Tregs can be recovered by magnetic bead-based separation of CD25+ and CD8 cells. Because thymus-derived Tregs are naive and homogenous, they are also resistant to inflammatory cytokine-induced destabilization [14,16], making them a promising autologous cell therapy in children undergoing heart transplantation, or an allogeneic cell therapy in the setting of other diseases [13]. Because ∼1% of children are born with congenital heart defects, many of whom require surgery including removal of the thymus [17], thymuses are routinely discarded. This material could be handled using procedures similar to those used to collect pancreata for islet transplantation and then used as a source of material from which to isolate Tregs.

Multiple protocols have been developed to manufacture Tregs for clinical trials, with variations in almost all key process parameters, including media, activation reagents, interleukin (IL)-2 concentration, restimulation timing and culture duration [2,4,5,[8], [9], [10], [11],[18], [19], [20], [21], [22], [23], [24]]. Recently, a variety of new Good Manufacturing Practice (GMP)–compatible reagents to activate T cells has been released but there are no reported comparisons of the performance of these now commercially available products. The lack of systematic studies to define critical process parameters and optimal reagents leads to uncertainty about the best methods to isolate and expand Tregs for clinical use [13].

Another uncertainty in the Treg manufacturing process is the feasibility of cryopreserving expanded cells. Some studies reported that cryopreservation reduces Treg quality, resulting in decreased FOXP3 expression and impaired suppressive capacity [25,26]. Consequently, to date only a limited number of clinical trials have used cryopreserved Tregs [21,22,27]. Using fresh cells as a clinical treatment creates many logistical hurdles: release testing must be done in a shorter time and there is limited notice of manufacturing failure; there is less infusion time flexibility to accommodate changes in patient health status; and it is difficult to quickly produce cells for rapidly progressing diseases such as acute graft-versus-host disease. Finding ways to cryopreserve clinical-grade Tregs to be used as an "off-the-shelf" product would overcome these hurdles, enable centralized manufacturing and allow a single product to be used to treat multiple patients [28,29]. Indeed, the concept of using “off-the-shelf” allogeneic cells is seen as the future of cell therapy products.

To develop a protocol to manufacture clinical-grade thymus-derived Tregs, we have comprehensively compared multiple process parameters using GMP-compatible reagents. We also report a new protocol to cryopreserve expanded thymus-derived Tregs to enable their use as an “off-the-shelf” cell therapy product.

Section snippets

Study approval

Human research was approved by the University of British Columbia Research Ethics Board (H17-01490 and H18-02553), the University of Alberta Human Research Ethics Board (Pro00001408), and the University of Minnesota.

Cell isolation

Thymus tissue was collected during infant cardiac surgery at British Columbia Children's Hospital or University of Alberta Stollery Children's Hospital. Tissue was dissociated in RPMI medium (Thermo Fisher Scientific, Waltham, MA, USA) with 10% heat-inactivated fetal bovine serum

Development of clinical-grade thymic Treg isolation protocol

We previously showed that CD25+CD8 Tregs can be isolated from pediatric thymus tissue using manual dissociation with scissors, complement-mediated lysis to deplete CD8-expressing cells and magnetic bead–based positive selection of CD25+ cells [14]. To develop a protocol that would be appropriate for GMP manufacturing, we first tested alternative methods for thymus tissue processing, seeking a method that minimized manual steps, ideally in a closed system. Thymuses were collected and processed

Effect of cell culture media on thymic Treg expansion

In Figures 2 and 3, the activation reagents were tested in different cell culture media, raising the possibility that some of the observed differences could be due to the media. We selected the two best-performing cell-free activation reagents (as defined by the highest fold expansion and FOXP3 expression), Treg Xpander and CD3/CD28/CD2 T Cell Activator, and compared their effects in four different media. Specifically, cells were expanded in ImmunoCult-XF, X-Vivo 15 and OpTmizer with serum

Discussion

Here we report the first comprehensive testing of activation reagents, media, restimulation timing and cryopreservation conditions to optimize a GMP-compatible manufacturing protocol for Tregs isolated from discarded pediatric thymuses. We found large differences between the types of activation reagents and media, with the combination of Dynabeads Treg Xpander and ImmunoCult-XF medium resulting in the most efficient expansion of cells that retained high FOXP3 expression and suppressive

Declaration of Competing Interest

ThermoFisher provided in-kind reagents and STEMCELL Technology provided donations to support this project. M.K.L. has received research funding from Bristol Myers Squibb, Takeda, Pfizer, CRISPR Therapeutics and Sangamo for work not related to this project. J.M.P. has received research funding from Amgen, Bayer and Pfizer; he has patents pending with AbCellera and SonoSep Technologies, as well as ownership of SonoSep, all for work not related to this project. B.R.B. receives remuneration as an

Author Contributions

KNM: conceived, designed and conducted experiments, analyzed data, and wrote the manuscript. SI: conceived, designed and conducted experiments, analyzed data and critically reviewed the manuscript. KLH: conceived and designed experiments, analyzed data and critically reviewed the manuscript. REH: provided critical feedback throughout the project and critically reviewed the manuscript. MH and GZ conducted experiments and analyzed data. IED: provided critical feedback throughout the project and

Acknowledgments

The authors thank volunteers, patients and their parents for contribution of samples, as well as the surgical and cardiac clinic staff at the British Columbia Children's Hospital and the University of Alberta Stollery Children's Hospital who made this study possible; special thanks to Melanie Ganshorn, Allison Jamieson, Lyn Nguyen and Colleen Ring. We also thank ThermoFisher Inc for the provision of in-kind reagents.

References (51)

  • S. Parmar et al.

    Third-party umbilical cord blood–derived regulatory T cells prevent xenogenic graft-versus-host disease

    Cytotherapy

    (2014)
  • S. Parmar et al.

    Ex vivo fucosylation of third-party human regulatory T cells enhances anti–graft-versus-host disease potency in vivo

    Blood

    (2015)
  • H. Fraser et al.

    A Rapamycin-Based GMP-Compatible Process for the Isolation and Expansion of Regulatory T Cells for Clinical Trials

    Molecular therapy Methods & clinical development

    (2018)
  • A. Alsuliman et al.

    A robust, good manufacturing practice–compliant, clinical-scale procedure to generate regulatory T cells from patients with amyotrophic lateral sclerosis for adoptive cell therapy

    Cytotherapy

    (2016)
  • H. Torikai et al.

    Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors

    Molecular Therapy

    (2016)
  • D.H. McKenna et al.

    Optimization of cGMP purification and expansion of umbilical cord blood-derived T-regulatory cells in support of first-in-human clinical trials

    Cytotherapy

    (2017)
  • R. Zeiser et al.

    Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells

    Blood

    (2008)
  • C.-T. Huang et al.

    Role of LAG-3 in Regulatory T Cells

    Immunity

    (2004)
  • A.L. Putnam et al.

    Clinical Grade Manufacturing of Human Alloantigen-Reactive Regulatory T Cells for Use in Transplantation

    American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons

    (2013)
  • I. Caramalho et al.

    Human regulatory T-cell development is dictated by Interleukin-2 and -15 expressed in a non-overlapping pattern in the thymus

    Journal of Autoimmunity

    (2015)
  • P. Hoffmann et al.

    Only the CD45RA+ subpopulation of CD4+CD25high T cells gives rise to homogeneous regulatory T-cell lines upon in vitro expansion

    Blood

    (2006)
  • M. Miyara et al.

    Functional Delineation and Differentiation Dynamics of Human CD4+ T Cells Expressing the FoxP3 Transcription Factor

    Immunity

    (2009)
  • E.J. Woods et al.

    Off the shelf cellular therapeutics: factors to consider during cryopreservation and storage of human cells for clinical use

    Cytotherapy

    (2016)
  • Y. Luo et al.

    The state of T cells before cryopreservation: effects on post-thaw proliferation and function

    Cryobiology

    (2017)
  • M. Gliwiński et al.

    Cell-Based Therapies with T Regulatory Cells

    BioDrugs: clinical immunotherapeutics, biopharmaceuticals and gene therapy

    (2017)
  • Cited by (16)

    • Consequences of adjusting cell density and feed frequency on serum-free expansion of thymic regulatory T cells

      2022, Cytotherapy
      Citation Excerpt :

      Thus, it is important to address the challenges of developing manufacturing protocols using non-cell-based activation reagents and serum-free media. The authors have shown that pediatric thymuses discarded during cardiac surgery are an abundant source of Tregs that can be isolated using magnetic selection and are well suited for clinical use as a result of their homogeneity, long telomeres and resistance to cytokine-mediated destabilization [26–28]. The authors previously developed a 13-day protocol to expand and cryopreserve thymic Tregs using non-cell-based activation reagents and serum-free medium, with preserved viability, FOXP3 expression and suppressive function [28].

    • Engineered antigen-specific regulatory T cells for autoimmune skin conditions

      2021, Autoimmunity Reviews
      Citation Excerpt :

      In human patients however, progressive disease periods are interspersed with periods of inactivity, providing melanocyte stem cells with an opportunity to differentiate and repopulate the depigmented lesions. For the intermittent treatment of vitiligo, it will be beneficial to store autologous CAR Tregs for later use [183]. By virtue of their antigen specificity, these Tregs might provide better safety profiles and decrease the risk of generalized immunosuppression.

    View all citing articles on Scopus
    View full text