Research paper
Single-pass, closed-system rapid expansion of lymphocyte cultures for adoptive cell therapy

https://doi.org/10.1016/j.jim.2009.04.009Get rights and content

Abstract

Adoptive cell therapy (ACT) for metastatic melanoma involves the ex vivo expansion and re-infusion of tumor infiltrating lymphocytes (TIL) obtained from resected specimens. With an overall objective response rate of 56%, this T-cell immunotherapy provides an appealing alternative to other therapies, including conventional therapies with lower response rates. However, there are significant regulatory and logistical concerns associated with the ex vivo activation and large-scale expansion of these cells. The best current practice uses a rapid expansion protocol (REP) consisting of an ex vivo process that occurs in tissue culture flasks (T-flasks) and gas-permeable bags, utilizes OKT3 (anti-CD3 monoclonal antibody), recombinant human interleukin-2, and irradiated peripheral blood mononuclear cells to initiate rapid lymphocyte growth. A major limitation to the widespread delivery of therapy to large numbers of melanoma patients is the open system in which a REP is initiated. To address this problem, we have investigated the initiation, expansion and harvest at clinical scale of TIL in a closed-system continuous perfusion bioreactor. Each cell product met all safety criteria for patient treatment and by head-to-head comparison had a similar potency and phenotype as cells grown in control T-flasks and gas-permeable bags. However, the currently available bioreactor cassettes were limited in the total cell numbers that could be generated. This bioreactor may simplify the process of the rapid expansion of TIL under stringent regulatory conditions thereby enabling other institutions to pursue this form of ACT.

Introduction

Adoptive cell therapy (ACT) involves the ex vivo isolation and expansion of antigen-specific lymphocytes for the purpose of autologous infusion. The applications of this therapy for cancer have included the treatment of Epstein–Barr virus (EBV) induced malignancies and metastatic melanoma (Rosenberg et al., 2008). Experimental protocols conducted at the National Cancer Institute-Surgery Branch (NCI-SB) have demonstrated the feasibility of inducing tumor regression in patients with metastatic melanoma following the infusion of tumor infiltrating lymphocytes (TIL). After isolation of TIL from resected metastatic lesions, rapid expansion in culture and re-administration to late-stage melanoma patients, these lymphocytes, when administered following a short course of lymphodepleting chemotherapy, achieved objective responses by RECIST criteria (Response Evaluation Criteria in Solid Tumors) in 52 out of 93 patients (56%) (Dudley et al., 2008).

Patients enrolled in initial clinical trials received an average dose of 6.0 × 1010 TIL per infusion. In order to generate this sizable cell product over a short period of time, TIL underwent a rapid expansion ex vivo, using anti-CD3 antibody, exogenous interleukin-2, and irradiated peripheral blood mononuclear “feeder” cells. This rapid expansion process (REP) was started in T-flasks and completed in gas-permeable (3-liter) bags. The cell expansion, viability, and performance achieved with current REP technology have been excellent for phase I/II developmental studies, and have allowed flexibility in the establishment of standard operating procedures. However, these current REP protocols present several limitations to the eventual widespread implementation of adoptive cell therapy. First, the REP in flasks/gas-permeable bags is an “open” system that necessitates multiple exposures of the cell product to the external environment for feeding, sampling, manipulation, and the transfer of TIL from flasks to bags. Such frequent manipulation allows for the possible contamination of the cells. Secondly, the use of numerous containers introduces the possibility of vessel-to-vessel variability. Third, this system is labor intensive requiring the devoted attention of skilled laboratory technicians.

Development of an optimal method of rapid expansion for widespread clinical use would involve a “closed” system thereby minimizing the chance of in-process contamination while maintaining comparable cell yields and requiring minimum expertise and labor. Several bioprocessors such as the Wave® and hollow-fiber bioreactors have been tried (Hami et al., 2004, Levine, 2008). These systems have proven useful for some clinical applications with lymphocytes, but none that involve the initiation of large-scale in vitro expansions with antigen presenting cells or feeder cells. We report here our investigations of a bioreactor developed at Aastrom Biosciences (Ann Arbor, MI) (Koller et al., 1993, Koller et al., 1998, Guardino et al., 2006). It has been previously employed in the ex vivo expansion of umbilical cord blood cells, bone marrow stem cells, and for clinical production of dendritic cell vaccines. These cell products have then been used to treat patients with chronic myelogenous leukemia, bone marrow suppression following high-dose chemotherapy, and for dendritic cell-based immunotherapy of multiple myeloma and other cancers.

We report here the initiation, completion, and preliminary optimization of the rapid expansion of multiple TIL samples in this closed-system perfusion bioreactor. In addition, we evaluate the antigen-specificity, potency, viability, sterility, and phenotype of TIL produced in the bioreactor system when compared to static culture conditions in T-flasks and gas-permeable culture bags.

Section snippets

Tumor infiltrating lymphocytes (TIL)

The TIL cultures used for this set of experiments were generated by employing a variety of techniques that have been described previously (Dudley et al., 2003). Briefly, TIL cultures were created following the overnight enzymatic digestion of a tumor specimen, by physical disaggregation using a Medimachine (Becton-Dickenson) with subsequent lymphocyte enrichment on a ficoll-step gradient, or from 1–2 mm tumor fragments. The age of all TIL cultures expanded and used in this set of experiments

TIL with known antigen-specificity can be expanded in a closed-system bioreactor

Adoptive cell therapy depends on the large-scale expansion of highly active lymphocyte lines, such as TIL, which has traditionally only been accomplished using T-flasks and gas-permeable bags. To evaluate the potential of the bioreactor, antigen-specific TIL from an HLA-A2+ patient (TIL 2784-1F2 d17) underwent a complete fourteen-day rapid expansion from start (inoculation) to finish (harvest). Based on the surface area of the cell bed, a bioreactor was inoculated with a feeders-to-TIL ratio of

Discussion

TIL-based ACT for patients with melanoma, as well as other T-cell based immunotherapies, offers a promising new modality that can mediate curative responses in patients with refractory cancers. These therapies depend on reliable methods to generate safe and effective cell products, but the methods currently available generally require highly trained staff and/or open culture systems, and therefore notably constrain the wider clinical evaluation of these promising therapies. One potential

Acknowledgements

We thank Naia Venturi for bioengineering expertise as required for configuration and operation of the bioreactor system, Tom Deaver for logistical support and Dr. Ronnda Bartel for critical review of the manuscript.

First page preview

First page preview
Click to open first page preview

Cited by (41)

  • Objectives, benefits and challenges of bioreactor systems for the clinical-scale expansion of T lymphocyte cells

    2021, Biotechnology Advances
    Citation Excerpt :

    Although it is possible to obtain such populations with multiple vessels and heavy labour work, patients will benefit from faster and more reliable processes to ensure cells will be ready on time and as soon as possible. REP are therefore commonly used in combination with bioreactor systems, regardless of specific designs and geometry (Bohnenkamp et al., 2002; Chakraborty et al., 2013; Donia et al., 2014; Klapper et al., 2009; Trickett et al., 2002), even if sometimes only partially (for instance only IL-2 or anti-CD3 antibodies addition and not a complete factor-enriched environment) (Knazek et al., 1990; Malone et al., 2001; Tran et al., 2007; Vera et al., 2010; Wang et al., 2015). In the present paper, the maximum cell density and maximum yield that can be achieved in a specific type of device will be discussed.

  • Technology Transfer: From Science to Process Development for Cell Therapy Manufacturing

    2019, Encyclopedia of Tissue Engineering and Regenerative Medicine: Volumes 1-3
  • Technology transfer: From science to process development for cell therapy manufacturing

    2019, Encyclopedia of Tissue Engineering and Regenerative Medicine
  • Cancer immunotherapy utilizing gene-modified T cells: From the bench to the clinic

    2015, Molecular Immunology
    Citation Excerpt :

    At present, there have been few studies comparing the efficacy of CAR T cells using these different genetic modification approaches, however in order for adoptive immunotherapy to progress, the optimal genetic modification and culturing technique needs to be determined. Although, adoptive immunotherapy currently costs approximately $50,000 per patient, it is anticipated that due to the development of closed systems (Klapper et al., 2009) and following the initial investment into the construction of good manufacturing practice grade facilities, the overall cost of adoptive immunotherapy may significantly decrease. Furthermore, it is hoped that once CAR T cells are an established treatment for cancer, the use of cell banks, similar to those currently being used for Epstein–Barr virus specific T cells (Leen et al., 2013) can be used to store cryopreserved allogeneic CAR T cells.

View all citing articles on Scopus
1

These two authors contributed equally to this work.

2

Current Address: The University of Michigan, Michigan Nanotechnology Institute for Medicine and Biological Sciences, 1150 West Medical Center Drive, Ann Arbor, Michigan, 48109, USA.

View full text