skip to main content


Title: Optimizing Cytotoxic T Cell Growth in a Centrifugal Bioreactor through Kinetic Growth Models
Cancer is the second leading cause of death globally and remains a significant issue in medicine. Immunotherapy treatments such as Chimeric Antigen Receptor T cell (CAR-T) therapies are becoming a more promising option because of their effectiveness in killing cancer cells without harming healthy tissue in the body. CAR-T therapies, however, are inaccessible to many due to the high cost—a result of inefficient cell expansion and manufacturing methods. To address this issue, we have developed the Centrifugal Fluidized Expansion (CentriFLEX) bioreactor that balances centrifugal and fluid forces, allowing the system to operate in perfusion and maintain a high cell density. Shown in past applications for similar cell types, the CentriFLEX can expand cultures up to 2.1 billion cells in an 11.4 mL chamber over the course of one week. Recently, we have used this system to expand bovine T cells as part of a collaboration with the College of Veterinary Medicine at Washington State University. Through the project, we conducted kinetic studies to model substrate consumption and metabolite production of bovine T cells and have enhanced the bioreactor design by making it more compact to fit entirely within a biosafety cabinet— mitigating contamination concerns. Current efforts have been spent determining the remaining parameters for the kinetic models and using such models to understand how the cells grow over time and in the space of a high-population density chamber. In this presentation, we will share how we use growth models that are based on a series of kinetic studies to predict substrate and metabolite levels over time in the bioreactor, allowing us to alter feed and dosing rates of medium and nutrients to maintain cell growth at the maximum specific growth rate.  more » « less
Award ID(s):
1645249
NSF-PAR ID:
10310313
Author(s) / Creator(s):
Date Published:
Journal Name:
Annual meeting American Institute of Chemical Engineers
ISSN:
0196-7282
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Cancer has been one of the most significant and critical challenges in the field of medicine. It is a leading cause of death both in the United States and worldwide. Common cancer treatments such as radiation and chemotherapy can be effective in destroying cancerous tissue but cause many detrimental side effects. Thus, recent years have seen new treatment methods that do not harm healthy tissue, including immunotherapy. Adoptive cell therapy (ACT) is one form of immunotherapy in which patients’ immune cells are modified to target cancer cells and then reintroduced into the body. ACT is promising, but most current treatments are inefficient and costly. Widespread implementation of ACT has been a difficult task due to the high treatment cost and inefficient methods currently used to expand the cells. Additionally, if the manufacturing process is not carefully controlled, it can result in the cells losing their cancer-killing ability after expansion. To address the need for an economically feasible culture process to expand immune cells for immunotherapy, our laboratory has designed a centrifugal bioreactor (CBR) expansion system. The CBR uses a balance of centrifugal forces and fluid forces, as shown in Figure 1, to quickly expand infected CD8+ T-cells from a bovine model up to high population densities. With other applications, the CBR has achieved cell densities as high as 1.8 x 108 cells/mL over 7 days in an 11.4-mL chamber. For this study, our goal is to begin validating the CBR by optimizing the growth of CEM (human lymphoblastic leukemia) cells, which are similar cell to cytotoxic T lymphocytes (CTLs). This can be accomplished by measuring kinetic growth parameters based on the concentrations of glucose and inhibitory metabolites in the culture. We hypothesize that by designing a kinetic model from static culture experiments, we can predict the parameters necessary to achieve peak CEM and eventually CTL growth in the CBR. We will report on kinetic growth studies in which different glucose concentrations are tested, and a maximum specific growth rate and Monod constant determined, as well as studies where varying levels of the inhibitory growth byproducts, lactate and ammonium, are added to the culture and critical inhibitor concentrations are determined. Another recent conceptual development for the design of the CBR is a real-time monitoring and feedback control system to regulate the cellular environment, based on levels of surface co-receptors and mRNA signaling within the culture. Prior studies have pinpointed T cell exhaustion as a significant issue in achieving successful immunotherapy, particularly in treatments for solid tumors; T cell exhaustion occurs during a period of chronic antigen stimulation when the cells lose their ability to target and kill cancer cells, currently theorized to be associated with particular inhibitory receptors and cytokines in the immune system. Designing a system with a fiber optic sensor that can monitor the cell state and use feedback control to regulate the pathways involved in producing these receptors will ensure the cells maintain cytotoxic properties during the expansion process within a Centrifugal Fluidized Expansion we call the CentriFLEX. In this presentation, we will also report on early results from development of this exhaustion monitoring system. In brief, achieving optimal kinetic models for the CBR system and methods to prevent T cell exhaustion has the potential to significantly enhance culture efficiency and availability of immunotherapy treatments. 
    more » « less
  2. T cell transfer immunotherapy is a highly effective cancer treatment in which the immune system’s inherent ability to fight cancer is amplified by increasing the amount of T cells that are deemed most active within a patient. T cells are a lymphocyte produced as an immune response to cancerous cells. Despite this advanced form of biological therapy, current T cell expansion methods are inefficient, resulting in high manufacturing costs, which brings question to the efficacy of T cell therapies. To address this issue, the recent development of a centrifugal bioreactor aims to rapidly expand T cells for cancer immunotherapy treatments at higher cell densities and in a shorter amount of time compared to current systems on the market. We hypothesize that by producing a mathematical model of a proof-of-concept T cell line to determine substrate consumption and metabolite production over time, we will be able to optimize growth of the cell line in the bioreactor. A series of three studies were performed to produce the growth model: (1) measuring yield coefficients of lactate, ammonium ion, and glucose, (2) determining the Monod constant and maximum specific growth rate, and (3) finding critical metabolite concentrations. To measure yield coefficients, T cells were grown in a 6-well plate at 1 x 105 cells/mL in 4 mL of medium with 100 uL samples taken and frozen each day over a 5-day period. At the end of the study, samples are thawed and used with lactate and ammonium assay kits for microplate reading to determine metabolite levels over time. To determine the Monod constant and maximum specific growth rate, T cells were grown in 12-well plates at pre-calculated varying glucose concentrations in 4 mL of medium in triplicates. Cells were counted for a minimum of six days to determine expansion over time to develop a linearized growth plot. To find critical metabolite concentrations, ammonium and lactate were added to glucose-free T cell medium at four different concentrations in triplicates utilizing a 12-well plate with a seeding density of 1 x 105 cells/mL in 4 mL of medium. The T cells then remained undisturbed in culture and were counted on day three. Once all parameters are determined, we can apply them to the growth model to determine levels of glucose, lactate, and ammonium as the T cells grow to high densities in the bioreactor and, as a result, optimize the manufacturing process for cancer immunotherapy treatments. 
    more » « less
  3. Abstract

    One of the current difficulties limiting the use of adoptive cell therapy (ACT) for cancer treatment is the lack of methods for rapidly expanding T cells. As described in the present report, we developed a centrifugal bioreactor (CBR) that may resolve this manufacturing bottleneck. The CBR operates in perfusion by balancing centrifugal forces with a continuous feed of fresh medium, preventing cells from leaving the expansion culture chamber while maintaining nutrients for growth. A bovine CD8 cytotoxic T lymphocyte (CTL) cell line specific for an autologous target cell infected with a protozoan parasite,Theileria parva, was used to determine the efficacy of the CBR for ACT purposes. Batch culture experiments were conducted to predict how CTLs respond to environmental changes associated with consumption of nutrients and production of toxic metabolites, such as ammonium and lactate. Data from these studies were used to develop a kinetic growth model, allowing us to predict CTL growth in the CBR and determine the optimal operating parameters. The model predicts the maximum cell density the CBR can sustain is 5.5 × 107 cells/mL in a single 11‐mL conical chamber with oxygen being the limiting factor. Experimental results expanding CTLs in the CBR are in 95% agreement with the kinetic model. The prototype CBR described in this report can be used to develop a CBR for use in cancer immunotherapy.

     
    more » « less
  4. Abstract

    Mesenchymal stromal cells (MSCs) have shown promise in regenerative medicine applications due in part to their ability to modulate immune cells. However, MSCs demonstrate significant functional heterogeneity in terms of their immunomodulatory function because of differences in MSC donor/tissue source, as well as non-standardized manufacturing approaches. As MSC metabolism plays a critical role in their ability to expand to therapeutic numbers ex vivo, we comprehensively profiled intracellular and extracellular metabolites throughout the expansion process to identify predictors of immunomodulatory function (T-cell modulation and indoleamine-2,3-dehydrogenase (IDO) activity). Here, we profiled media metabolites in a non-destructive manner through daily sampling and nuclear magnetic resonance (NMR), as well as MSC intracellular metabolites at the end of expansion using mass spectrometry (MS). Using a robust consensus machine learning approach, we were able to identify panels of metabolites predictive of MSC immunomodulatory function for 10 independent MSC lines. This approach consisted of identifying metabolites in 2 or more machine learning models and then building consensus models based on these consensus metabolite panels. Consensus intracellular metabolites with high predictive value included multiple lipid classes (such as phosphatidylcholines, phosphatidylethanolamines, and sphingomyelins) while consensus media metabolites included proline, phenylalanine, and pyruvate. Pathway enrichment identified metabolic pathways significantly associated with MSC function such as sphingolipid signaling and metabolism, arginine and proline metabolism, and autophagy. Overall, this work establishes a generalizable framework for identifying consensus predictive metabolites that predict MSC function, as well as guiding future MSC manufacturing efforts through identification of high-potency MSC lines and metabolic engineering.

     
    more » « less
  5. Abstract

    Despite substantial advancements in development of cancer treatments, lack of standardized and physiologically‐relevant in vitro testing platforms limit the early screening of anticancer agents. A major barrier is the complex interplay between the tumor microenvironment and immune response. To tackle this, a dynamic‐flow based 3D bioprinted multi‐scale vascularized breast tumor model, responding to chemo and immunotherapeutics is developed. Heterotypic tumors are precisely bioprinted at pre‐defined distances from a perfused vasculature, exhibit tumor angiogenesis and cancer cell invasion into the perfused vasculature. Bioprinted tumors treated with varying dosages of doxorubicin for 72 h portray a dose‐dependent drug response behavior. More importantly, a cell based immune therapy approach is explored by perfusing HER2‐targeting chimeric antigen receptor (CAR) modified CD8+T cells for 24 or 72 h. Extensive CAR‐T cell recruitment to the endothelium, substantial T cell activation and infiltration to the tumor site, resulted in up to ≈70% reduction in tumor volumes. The presented platform paves the way for a robust, precisely fabricated, and physiologically‐relevant tumor model for future translation of anti‐cancer therapies to personalized medicine.

     
    more » « less