Abstract
The ability to develop cost-effective, scalable and robust bioprocesses for human pluripotent stem cells (hPSCs) will be key to their commercial success as cell therapies and tools for use in drug screening and disease modelling studies. This review outlines key process economic drivers for hPSCs and progress made on improving the economic and operational feasibility of hPSC bioprocesses. Factors influencing key cost metrics, namely capital investment and cost of goods, for hPSCs are discussed. Step efficiencies particularly for differentiation, media requirements and technology choice are amongst the key process economic drivers identified for hPSCs. Progress made to address these cost drivers in hPSC bioprocessing strategies is discussed. These include improving expansion and differentiation yields in planar and bioreactor technologies, the development of xeno-free media and microcarrier coatings, identification of optimal bioprocess operating conditions to control cell fate and the development of directed differentiation protocols that reduce reliance on expensive morphogens such as growth factors and small molecules. These approaches offer methods to further optimise hPSC bioprocessing in terms of its commercial feasibility.
Keywords: Bioprocess economics, Cell therapy, Regenerative medicine, Scale-up, Pluripotent stem cells
1 Introduction
Human pluripotent stem cells (hPSCs) have opened fresh avenues in modern medicine that have the potential to revolutionise healthcare, particularly since the first derivation of induced pluripotent stem cells (iPSCs) [1, 2]. Human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) have the capacity to differentiate into all mature cell types, making them attractive candidates for use as cell therapies [3]. Moreover, hPSCs also offer a unique, novel platform by which to augment, and even redefine, current drug discovery and drug screening programmes by the provision of a human in vitro tool on which to perform efficacy and toxicity screens for novel chemical entities (NCEs) [4, 5]. hiPSCs could also pave the way for personalised medicine through the medium of responders versus non-responder ‘trial in a dish’ models [6].
hPSCs can provide a cornerstone of the regenerative medicine industry via the provision of cell therapies for diseases with unmet clinical needs. hiPSCs in particular might provide a diagnostic tool capable of assuaging the high late-phase failure rate of NCEs in clinical trials [7, 8]. The market for stem cell research products exceeded $3bn at the end of 2013 (http://tinyurl.com/n26fe4z). hPSCs for use as research tools are currently marketed at $2000–$3000/vial [9]. However, the value of this market is likely to be incremental when considered against hPSC-derived cell therapies [10], which has the potential to tap into a multi-billion dollar global market [11]. If hPSCs are to achieve their full clinical and commercial potential, significant challenges must be overcome with regards to current abilities to produce hPSC-derived cells at commercially relevant scales.
At the forefront of current challenges to hPSC-derived therapies is the production of cells at a relevant quantity and quality to support their function. Details on the bioprocess techniques for hPSC therapies currently being manufactured for preclinical and clinical trials or for use as research tools are provided in Table 1. To date, hPSC-derived products in development have mainly consisted of retinal progenitor cells and pancreatic β-cells derived from hESCs [12–15], or a variety of cell lineages such as neurons, cardiomyocytes and hepatocytes derived from hPSCs for use as research tools [16]. Promising results have also been observed for cell therapies derived from hiPSCs, such as retinal pigment epithelial cells [17]. Table 2 indicates that most products in clinical development still depend on planar technologies. Traditional, planar technologies that offer reliable tools for laboratory-based protocols are labour-intensive and do not lend themselves to large scale, allogeneic processes [18]. Dose sizes reported for hPSC-derived cell therapy products currently range from 5 × 104 cells for indications such as macular degeneration to 108 cells for diseases such as diabetes (Table 2). Furthermore, it has been estimated that large doses, of around 109 cells, will be needed to treat conditions such as myocardial infarction and liver disease [19, 20]. Whilst this review focuses on process techniques for the production of single-cell type populations, therapies for certain disease types will necessitate transplantation of a functional tissue-like structure. Novel, organoid development techniques may therefore provide a method of production of tissue-like structure representing a variety of different organs from small seed populations of hPSCs. These have potential applications as either transplantable therapies or research tools [21–24].
Table 1.
Bioprocess development considerations for hPSC-derived products
Consideration | Example Criteria |
---|---|
Operational | Expansion yields (harvest densities) |
performance | Expansion folds |
Differentiation efficiencies | |
DSP yields | |
Purity | |
Resource utilisation | |
Scalability | |
Lot processing time | |
Economic | Capital investment |
Cost of goods (materials, labour, quality control and indirect) | |
Economies of scale – scale-up versus scale-out | |
Fresh versus frozen product transportation and storage | |
Process development costs | |
Supply chain replenishment | |
Product shelf-life | |
Reimbursement value | |
Quality control | cGMP and cGTP standards |
and regulatory | Process robustness and reproducibility |
compliance | Process validation, acceptable ranges of operation |
Product characterisation | |
Quality, consistency and source of raw materials | |
Automated versus manual processing | |
Safety | Contamination and containment |
Live human tissue handling | |
Patient safety – side-effects, risk of tumour formation | |
Flexibility | Process changes |
Manufacturing demand changes | |
Process bottlenecks | |
Process scalability |
Table 2.
Technologies used for expansion and differentiation of hPSC-derived cell products used in clinical trials or as research tools
Company | Indication | Target cell type | Dose size | Cell expansiona) details | Differentiation detailsa) | Source |
---|---|---|---|---|---|---|
Cellectis | Diabetes mellitus (type I) | Insulin producing β-cells | ND | SUB: hollow fiber, multicompartment perfusion bioreactor | SUB: hollow fiber, multicompartment perfusion bioreactor | [12] |
Advanced Cell Technology | Macular degeneration | Retinal pigment epithelial (RPE) cells | 5 × 104 | Planar: well-plates, MEF feeder layer, three passages | Planar: well-plates EB formation | [13] |
CellCure | Macular degeneration | RPE cells | 2 × 104 | Planar: well-plates xeno-free media | Planar: well-plates, 8-wk process, serum-free conditions | [14] |
Healios | Macular degeneration | RPE cells | 5 × 104 | Planar: plate-based custom designed automated process platform | Planar: plate-based custom designed automated process platform | [17] |
ViaCyte | Diabetes mellitus (types I and II) | Pancreatic β-cell precursors | 108 | Planar: multi-layer cell factories, 2-wk process, xeno-free media | Planar: plate-based aggregate differentiation, 2-wk process, Xeno-free media | [15] |
Geronb) | Spinal cord injuries | Oligodendrocyte progenitor cells | 2 × 106 | Planar: matrigel coatedT-flasks, 3- to 5-wk process | Planar: T-flasks, 6-wk process, growth factor-based protocol | [111] |
CellularDynamics International | hiPSC-derived cells for use as research tools | Cardiomyocytes, neurons, hepatocytes, | N/A | SUB: litre-scale, five passages, Xeno-free media endothelial cells | SUB: litre-scale, chemically defined conditions | [16] |
Technologies for expansion and differentiation operations are detailed alongside process durations and media where this information is available.
Geron's GRNOPC1 therapy was withdrawn from trials but has been included in this table for comparison.
Planar technologies may struggle to satisfy the global demand for hPSC-derived cell therapies requiring high dose sizes [20, 25]. Furthermore, there is widespread use of xenogeneic materials associated with traditional hPSC technologies, preventing their use in the production of clinical grade hPSC-derived cells. Differentiation strategies have often represented idiosyncratic protocols that have proven difficult to translate to robust bioprocess unit operations [26]. Media costs associated with hPSC processes are of further concern; many media supplements render the products of current hPSC processes prohibitively expensive for purpose. Additionally, studies into the poorly understood interactions between hPSCs and the microenvironment provided by media components and cell anchorage materials are only now beginning to take place; significant gaps exist in our knowledge of how the design of such materials affects hPSC activity [27].
Previous reviews in this field have discussed advances in the large-scale expansion and bioreactor-based culture of hPSCs (e.g. [25, 28–30]). Other studies have focused upon the impact of the design of microcarriers and anchorage materials on hPSC bioprocessing (e.g. [27, 31–33]). Herein, we aim to summarise key considerations and methods that might be employed in order to achieve cost-effective bioprocess design across a range of manufacturing scales. Key process economic metrics and drivers are outlined. A discussion of advancements in robust, GMP-based expansion and directed differentiation strategies is provided. Finally, a review of recent innovations in integrated bioprocess design is presented.
2 Cell therapy bioprocess economics
When examining process options for hPSC manufacture, it is important to consider not only the operational performance but also the consequences on the economics, quality, regulatory compliance, safety and flexibility. These key considerations are summarised in Table 1. This review pays particular attention to progress made on improving the economic and operational feasibility of hPSC bioprocessing.
Reimbursement pressures have resulted in an increased awareness of the importance of estimating and improving manufacturing costs for stem cell products. This section discusses factors that influence two key cost metrics: fixed capital investment (FCI) and cost of goods (COG).
2.1 Capital investment
The FCI represents the cost to build a manufacturing facility ready for start-up. It includes the cost of the building with the fixed (non-disposable) equipment, piping, instrumentation and utilities installed. Estimates of facility costs are often made using factorial estimates. These are well established for traditional stainless steel biopharmaceutical facilities using the Lang Factor method [34], which involves multiplying the total equipment purchase cost by the ‘Lang factor’. At present, there are no published studies that have determined an appropriate factorial method for stem cell manufacturing facilities. The Lang factor is usually derived based on the analysis of costs of previous projects; as yet very few FCI benchmarks have been published for stem cell manufacturing facilities. Investment costs for stem cell facilities will also be influenced by the degree of open versus closed processing and the consequences on the cleanroom classification required and whether automated or manual process techniques are employed. Stem cell bioprocessing is also dependent on disposable or single-use process platforms such as T-flasks, CellStacks and single-use bioreactors (SUBs). To this end, a Lang factor method adapted for disposable-based biopharmaceutical facilities is currently the best method available by which to approximate the FCI associated with stem cell manufacturing [35]. Ongoing work at University College London is focused upon developing a method for estimating FCI that is specific to stem cell manufacturing facilities.
2.2 Cost of goods
The COG represents the cost to manufacture a stem cell product and comprises direct (e.g. materials) and indirect (e.g. maintenance) costs. Simaria et al. [20] summarise key factors that influence COG values for stem cell products; these include process efficiencies (e.g. harvest densities post-expansion, differentiation yields), technology choices (e.g. planar vs. microcarrier-based SUBs), and resources required and their unit costs (e.g. media, single-use vessels and labour). Economies of scale are a relevant factor as demand and lot size are varied as well as dose for cell therapies and required cell population sizes for cells as drug screening tools. Outputs are usually expressed as COG per cell population for screening tools or COG per dose for therapeutic applications.
Decision-support software can aid the design of cost-effective bioprocesses. However, to date, few published cost studies exist for stem cell bioprocessing. Commercially available flowsheeting software packages have been employed to cost stem cell process designs at fixed scales [36]. Simaria et al. [20] and Hassan et al. [37] present the development and application of decisional tools that integrate models for mass balancing, equipment sizing and bioprocess economics with optimisation algorithms for allogeneic mesenchymal stem cell (MSC) production. The tools were used to predict the most cost-effective upstream and downstream technologies for commercial MSC manufacture across a range of different scales and doses. The analyses presented in these works illustrate how such tools can be used to determine the scale at which planar technologies cease to be cost-effective in contrast to microcarrier-based SUBs, when downstream processing bottlenecks occur, as well as future required performance capabilities of promising technologies to close existing technology gaps and meet COG targets. This approach is being extended [38] to hPSC processes, which typically have additional process steps such as differentiation, so as to identify key economic drivers for drug screening and therapeutic applications, and predict technical innovations required to bridge the gaps constraining widespread application of hPSCs.
In addition to considering the costs of stem cell bioprocessing alternatives, it is also useful to capture the impact of uncertainties such as lot-to-lot variability and contamination risks, particularly when manual processing techniques are employed [39]. Stochastic modelling techniques, such as the Monte Carlo simulation method, have been used to evaluate process robustness under uncertainty in the biopharmaceutical sector [35, 40, 41]. Stochastic modelling has yet to be applied to iPSC processing, but will form an important part of future work in order to develop robust and cost-effective hPSC bioprocesses.
2.3 Process economic drivers
In order to achieve cost-effective bioprocesses for hPSCs, efforts need to focus on increasing the overall productivity and/or decreasing the overall production costs. Hence, critical process economic drivers for hPSC processes include expansion and differentiation yields as well as the cost of materials and labour. The remainder of this paper therefore focuses on progress made on the expansion and differentiation yields in planar and bioreactor technologies as well as the development of media and cell-anchorage materials for these unit operations.
3 hPSC bioprocessing strategies: Expansion
3.1 Planar culture systems for hPSC expansion
Until recently, planar hPSC culture platforms relied heavily on the use of xenogeneic growth substrates and non-human feeder layers, which risk contamination of hPSC-derived products. Furthermore, feeder layers differentially secrete signalling factors, resulting in poorly defined culture conditions that are unsuitable for empirical study of hPSC expansion [42]. The development of anchorage materials comprising of a mixture of synthetic and/or recombinant biological motifs have allowed hPSC culture to progress away from the use of feeder layers [31].
The labour-intensive nature of T-flasks limits their throughput and applicability to larger-scale processes [18, 25]. Systems that stack multiple culture chambers above one another vertically have enabled greater cell yields than traditional 2D culture methods at lower factory floor footprints [43]. The Cell Factory (ThermoFisher Scientific, Waltham, MA, USA), CellStack/ HyperStack (Corning, New York, NY, USA) and Xpansion (Pall Life Sciences, Port Washington, NY, USA) systems are examples of this. Inflated facility size requirements and subsequent capital investment costs associated with 2D culture scale-up are challenges facing companies hoping to produce hPSCs on a commercial scale [44, 45].
Automated, closed-process systems such as the CompacT SelecT (Sartorius AG, Gottingen, Germany), capable of handling 90 × T175 flasks simultaneously, and the Nunc Automatic Cell Factory Manipulator (ThermoFisher Scientific), capable of manipulating 4 × 40 layer vessels, may help increase the throughput of 2D hPSC bioprocess strategies [46] during expansion and differentiation. Automated systems allow processing to take place in smaller, lower clean rooms compared to manual processing. The closed processing offered by automation systems provides greater process control and reproducibility compared to manual processes [39].
Planar processing platforms will continue to have a place in commercial hPSC bioprocesses. This is particularly likely for the production of autologous cell therapies and patient-specific hPSC-derived cells for personalised medicine drug screening that necessitate a scale-out, rather than a scale-up approach to bioprocess design [43].
3.2 Three-dimensional culture systems for hPSC expansion
There are two main methods of 3D hPSC culture; the use of suspended microcarriers as adherent surfaces for stem cell growth [47], or growth of hPSCs as suspended aggregates in SUBs [48]. 3D hPSC culture systems allow online process monitoring, provide greater scalability potential and reduce facility size requirements when compared to planar technologies. Bioreactor systems also permit strict control of conditions during bioprocesses [49]. A challenge to implementation of 3D culture of hPSCs is exposure of cells to shear forces, which must be tightly controlled as they can impact upon hPSC fate determination [50, 51]. Development of xeno-free, defined media [52] is crucial to the robust bioprocessing of hPSCs. The simplicity of such media may reduce costs associated with hPSC expansion materials by 30–60% [53].
3.2.1 Microcarrier-based systems for hPSC expansion
Microcarriers are small beads or discs, which permit propagation or directed differentiation of hPSCs within a 3D bioreactor. hPSC studies investigating microcarriers have recently achieved expansion folds as high as 28-fold over 6 days [54]. Microcarrier-based expansion folds are often higher than those in 2D expansion studies across similar timescales [46, 47, 55–57]. Several recently published reviews include summary tables for SUB-based hPSC expansion studies (using both microcarrier and aggregate culture strategies [25, 28, 29].
A critical property of microcarriers is their high surface area to volume ratio, on which large populations of hPSC cells may be cultured in a relatively small vessel, alleviating the costs associated with expensive media and supplements necessary for hPSC bioprocessing [28, 56]. Microcarriers are able to support expansion and long-term self-renewal of hPSCs over multiple passages, proving the platform's capability to support production of clinically relevant cell numbers [47]. Microcarrier culture of hPSCs also results in cell colonies that generally have less than 10 layers [58]; thus concentration profiles of nutrients and signalling molecules are less likely to occur than in aggregate-based cultures.
The common use of microcarrier coatings that contain animal-derived components, which are unsuitable for use in the manufacture of cell therapies, is a significant challenge to culture of hPSCs on microcarriers. Serum-free and feeder-free microcarrier platforms are available for hPSC-based processing, although many of these utilise the microcarrier coating, Matrigel, which is derived from murine origins [25]. Recombinant human proteins can now be used as a substitute for animal-derived microcarrier coatings in planar and 3D microcarrier cultures [59, 60]. However, such proteins can be difficult to isolate, expensive to produce, and prone to lot-to-lot variation. Synthetic substrates, which circumvent consistency issues associated with recombinant substrates, have been developed in planar conditions and successfully applied to microcarrier-based hPSC culture [61–63].
Xeno-free microcarrier coatings utilise polymers to mimic Matrigel and feeder layer properties in order to encourage attachment and self-renewal of hPSCs on microcarriers. Expansion folds on xeno-free microcarriers comparable to those coated with Matrigel have been reported [60, 63]. It has been proposed that positively charged microcarriers can be used to successfully support hPSC expansion at clinically relevant scales and similar cell concentrations and expansion folds to coated microcarriers were achieved [58]. Methods of xeno-free hPSC culture represent a regulatory compliant approach to the production of hPSCs for clinical applications; they also reduce additional expenses incurred by the use of supplementary serums. Development of xeno-free microcarrier coatings is one area where a quality-by-design (QbD) approach to product development has allowed elucidation of specific properties of microcarriers that affect hPSC self-renewal.
Harvesting of cells from microcarriers is usually carried out using enzymatic separation, which can add to material costs associated with microcarrier-based culture of hPSCs. Microcarriers coated in thermo-sensitive polymers that allow detachment of seeded cells obviate the need for dissociation enzymes, although studies in this area are still in their preliminary phases in this area [64].
Parallel to developing GMP-based hPSC expansion protocols, research has focused on optimising bioreactor conditions for dynamic hPSC expansion processes so as to increase achievable expansion folds and cell concentrations. This will help reduce COG associated with manufacture of hPSCs. Controlling the dissolved oxygen levels has been found to be critical during hPSC culture on microcarriers in SUBs; 2.5 higher expansion folds and ∼85% improvements in maximum cell concentrations were reported in a hypoxic environment when compared to uncontrolled conditions [57]. Furthermore, attachment of hPSCs to microcarriers as single cells can improve seeding efficiency from 30 to over 80% and reduce durations associated with microcarrier loading compared to clump seeding [63].
3.2.2 Aggregate suspension culture of hPSCs
hPSCs can be cultured as suspended aggregates in bioreactors. When hPSCs are grown as aggregates the rho-associated protein kinase inhibitor (ROCKi), Y-27632, is used to protect single cells from dissociation-induced apoptosis [65]. Each cell aggregate is treated as a de facto colony. Aggregate sizes must be controlled in suspension bioreactors to prevent differentiation of cells in larger colonies [66–68]. It has been reported that aggregate culture of stem cells increases the therapeutic potential and the differentiation efficiency of hPSCs via the sustainment of endogenous signalling within cell colonies [32]. Aggregate expansion of hPSCs also negates the need for expensive (and sometimes undefined) components of substrates upon which hPSCs are cultivated in adherent cultures [67]. Aggregate culture of hPSCs rely more heavily on the expensive media supplements (such as GFs) compared to microcarrier culture [54]. Several groups have proposed methods of hPSC culture through the use of cell aggregates with the potential to be scaled up in order to produce clinically relevant cell numbers [67–69]. Twenty-fivefold expansion has been achieved over 14 days during aggregate-based hPSC culture [49] and studies into expansion of hPSCs as aggregates yield similar expansion-folds when compared to microcarrier systems [70–72]. Long-term maintenance of hPSCs in aggregates in dynamic bioreactor conditions over several passages has also been proven to be feasible [49, 68]. Aggregate-based hPSC cultivation necessitates frequent manual interactions in order to control aggregate sizes [28, 71, 73], which will adversely affect labour costs and the robustness of aggregate-based hPSC bioprocesses.
Agitation rates can be used to successfully modulate uniform aggregate size in order to improve expansion of hPSCs as aggregates and reduce cell loss due to shear forces [53, 68]. This is also the case with microcarrier cultures, where impeller speeds of between 45 and 60 rpm were found to promote optimal cell population doubling times [74]. The effects of shear on hPSC self-renewal and lineage determination is an area of intensifying research, although currently this is a poorly understood area in terms of the effect of mechanical strain on hPSC fate determination [29, 75].
The importance of cell inoculation concentration has been demonstrated during aggregate culture of hPSCs in dynamic bioreactor conditions; seeding concentrations of 2–3 × 105 cells/mL were found to maximise viability of hPSCs [68, 76]. Single cell inoculation has also been estimated to reduce cell losses by up to 60% [68]. Cell concentrations of up to 3.4 × 106 hPSCs/mL have been achieved using dynamic, aggregate-based culture techniques [76]. This represents 1.9-fold improvement over maximum cell concentrations achieved in planar systems, although it is significantly lower than the maximum cell concentrations achieved in xeno-free hPSC cultures performed in microcarrier-based systems (6 × 106 cells/mL) [77].
A few aggregate hPSC expansion processes combine xeno-free conditions with defined media [53, 69, 71]. These investigations represent a valuable effort to remove media supplements that either introduce the risk of xenogeneic material to hPSCs or expose media to lot-to-lot variability, although early attempts resulted in relatively modest expansion folds [69].
4 hPSC bioprocessing strategies: Differentiation
4.1 Planar strategies for hPSC differentiation
Traditional stem cell differentiation protocols were designed around bench-scale research paradigms and little effort was made to incorporate reproducibility and process robustness into these experiments [78]. Directed differentiation strategies often involve exposing hPSCs to a cocktail of morphogens, at specific time-points throughout the differentiation process [79–82]. Similar to hPSC expansion, traditional differentiation strategies rely heavily upon the use of xenogeneic materials [83]. Planar differentiation protocols, that are free of xenogeneic material, have been reported [84]. Despite such progress, many differentiation protocols are inherently variable owing to the laboratory idiosyncrasies of individual technicians, thus reliable and robust differentiation processes are still in their infancy. Timescales and efficiencies also vary significantly between experiments even when the same cell type is targeted for production (Table 3).
Table 3.
Key performance characteristics of planar and bioreactor-based differentiation protocols
Derived cell-type | Method | Time (days) | Number of target cells per input hPSC (ratio) | Reported efficiency (%) | Max. cell concentration (cells/mL)a) | Refs. |
---|---|---|---|---|---|---|
Cardiomyocyte | 2D monolayer | 9 | ND | 64.8 ± 3.3 | 2.5–5 × 104 | [87] |
Cardiomyocyte | 2D EB formation | 60 | 0.81 | 10 ± 2 – 22 ± 4 | ND | [80] |
Hepatocytes | 2D EB formation | ND | ND | 50 ± 2 | 1–5 × 104 | [81] |
Hepatocytes | 2D monolayer | 14 | ND | 73 ± 18 | ND | [82] |
Motor neurons | 2D monolayer | 14 | ND | 33.6 ± 12 | ND | [79] |
Neural nociceptors | 2D monolayer | 15 | ND | 61 ± 2 | 1 × 104 | [88] |
Neurons | 2D monolayer | ∼7 | ND | ND | 4.5 × 104 | [84] |
Dopaminergic neurons | 2D monolayer | ∼28 | ND | 30 ± 2 | ND | [89] |
Neural progenitor cells | 2D monolayer | 6 | ND | 90 ± 1 | 5 × 104 | [91] |
Endoderm progenitors | 2D monolayer | 4 | ND | 73.2 ± 1.6 | 1.3 × 105 | [90] |
Cardiomyocytes | 2D EB formation | 16–18 | 70 | 87 ± 3.4 | 4.5 -6 × 104 | [97] |
Cardiomyocytes | SUB microcarriers | 16 | 0.33 | 15.7 ± 3.3 | 1.36 × 106 | [93] |
Haematopoietic cells | SUB microcarriers | 7 | 4.41 | ND | ND | [92] |
Cardiomyocytes | SUB cell aggregates | 18 | 23 | 100% beating aggregates | 4.3 × 105–5.2 × 105 | [96] |
Hepatocyte-like cells | SUB cell aggregates | 21 | ND | 18 ± 7 | 3–5 × 105 | [94](HLCs) |
ND, no data.
In planar studies cell concentrations per mL have been estimated based on cell densities and recommended working volume for vessels used in these studies as no cell concentrations are provided in studies of this type.
The use of small molecules within differentiation protocols has helped to improve their reproducibility via reduced use of recombinant growth factors [85, 86]. Several groups have created highly simplified protocols, whilst still improving the efficiency and processing times of 2D differentiation processes, by replacing growth factors with small molecules in the preliminary stages of differentiation protocols [84, 87, 88]. A protocol with the specified aim of creating a differentiation process capable of creating dopaminergic neurons for transplantation in T-flasks has been developed [89]. This strategy enabled the production of cryopreservable dopaminergic neurons with a high level of efficiency, allowing better control of time management in within the differentiation process. This approach is well served to reduce bottlenecks in the downstream phases of autologous hPSC processes. Further advances have proven it is possible to derive progenitor cells, suitable for transplantation as cell therapies at high efficiencies [90] and in xeno-free and small-molecule free, defined conditions [91]. Negation of the need for small molecules and growth factors also has the potential to drastically reduce the cost of differentiation procedures.
4.2 Bioreactor-based systems for hPSC differentiation
Concentrated research into bioreactor-based differentiation strategies has stemmed from the need to translate differentiation from a lab-scale area of research into processes capable of producing industrially relevant cell numbers in a reproducible manner. A number of studies in which hPSCs have been successfully differentiated in bioreactor conditions have been carried out, either attached to microcarriers [92] or in the form of cell aggregates [93, 94]. Differentiation strategies developed in 3D bioreactors, particularly stirred-tank vessels, lend themselves to large-scale processes far better than their planar counterparts as labour-intensive tasks, such as media exchanges, can be fully automated in such vessels, which also offer processing advantages such as online environmental monitoring and control. Research carried out on SUB-based differentiation of mPSCs suggests that the use of spinner flasks resulted in a 12-fold reduction of the man hours spent in the laboratory when compared to planar techniques [95].
hPSCs can be differentiated towards a number of clinically relevant lineages in SUBs including cardiac [96], haematopoietic [92], neuronal [77] and hepatocyte-like [94] (see Table 3 for summary). Bioreactor-based differentiation will be necessary in order to produce certain hPSC-derived cell products at commercially relevant scales, however it must be considered that such processes will only be made more cost-effective by making concurrent improvements in differentiation efficiencies and through the reduction of expensive media supplements in such protocols. Bioreactor-based differentiation would benefit from the translation of highly efficient protocols demonstrated in planar systems [88, 91, 97] to SUB systems. Such protocols have the potential to result in differentiation efficiencies that are higher than those achieved with SUB-based bioreactors alone. A novel, microparticle-based approach to morphogen delivery to PSC aggregates was reported as a method to achieve up to a 12-fold reduction in morphogen use during bioreactor-based differentiation protocols [98]. Such systems provide a valuable method by which to reduce material costs associated with SUB-based hPSC culture.
One question arising from the birth of SUB-based hPSC differentiation is how a dynamic, controlled environment might be harnessed to augment processing strategies in this area [51]. One of the reasons for the lack of characterisation with regards to the effects of shear on hPSCs, is that different dynamic culture systems result in different shear profiles. Thus, drawing comparisons across separate studies is difficult [75]. However, scale-down studies suggest that shear stress during early hPSC differentiation promotes mesodermal, endothelial and haematopoietic phenotypes even when the presence of morphogens promoting these lineages were absent [99–101]. Interestingly, in early stages of differentiation, hPSCs lineage determination seems to be insensitive to the magnitude of shear stress; however in later stages, progenitor cell activity appears to be more magnitude-sensitive [100, 102]. Shear forces have been shown to partially negate the need for costly media supplements in published studies [99, 100]; broadening our knowledge of the way that shear stress impacts upon hPSC culture must be seen as an important factor in bioprocess optimisation. Hypoxic environments, which can be tightly controlled within SUBs, have also been shown to enhance differentiation of hPSCs towards both ectoderm and mesoderm cell lineages [96, 103].
Novel, microfluidic systems could be a key tool in elucidating the effects of specific environmental parameters on hPSC propagation and differentiation. Microfluidic bioreactors provide an ultra-scale down, high throughput platform by which to study single cells or colonies in strictly defined conditions [104–107]. The cellular processes governing hPSC fate are complex and cannot be attributed to a single given parameter. Microfluidic devices are well placed, as a low cost development platform, to enhance our understanding of how defined microenvironmental conditions can affect hPSC activity [108].
The effect of the biochemical properties of microcarriers on hPSC fate determination is poorly understood. It has also been suggested that the mechanical properties of microcarriers, such as their stiffness and size can also be investigated and optimised for specific purposes [27]. Rational design of microcarriers could provide an optimized bioprocess platform with which to manufacture specific hPSC-derived cell lineages. This would enable better control of cells’ microenvironment and thus allow more efficient differentiation processes that do not rely as heavily on expensive media supplements as current platforms do.
5 Integrated bioprocess design strategies for hPSCs
Novel, integrated hPSC bioprocesses, whereby multiple unit operations are carried out using continuous culture strategies, are being explored as an alternative to segregated bioprocess strategies. Integrated bioprocesses negate the need for labour-intensive processes that usually take place following hPSC expansion such as harvest and transfer of cells prior to differentiation. Processing hPSCs in this manner can help to avoid process bottlenecks and increase throughputs of hPSC product manufacture. Additionally, integrated iPSC bioprocess protocols offer greater containment capabilities, reducing the potential for contamination within the bioprocess. Several studies in which hPSC expansion and differentiation are carried out as a single unit operation using continuous culture strategies exist [67, 74, 77]. Figure 1A illustrates how these strategies differ from segregated culture and differentiation of hPSCs. Only a handful of investigations into this relatively new area of hPSC bioprocess research have been published and these are summarised in Table 4.
Figure 1.
hPSC bioprocess strategies and their characteristics: (A) Planar processing strategies (top panels) rely on multi-layer vessels. Development of SUBs for PSC culture and differentiation has allowed development of 3D strategies, which are suitable for large-scale allogeneic bioprocesses (middle panels). Integrated bioprocesses allow hPSC expansion and differentiation to be carried out as a single unit operation in the same bioreactor (bottom panels). (B) Traditional and integrated autologous iPSC bioprocess strategies: Tradtional planar strategies neccessitate the need for use of well-plates and T-Flasks. Novel, automated bioreactor systems such as the Ambr system (Sartorius AG) may allow implementation of bioprocess strategies whereby reprogramming, iPSC expansion and differentiation are carried out within a single bioreactor. (C) Segregated and integrated allogeneic bioprocess strategies: Tradtional allogneic bioprocess strategies utilise multi-layer, planar technologies. Segregated bioprocessing stratagies make use of separate SUBs for hPSC expansion and differentiation. Integrated bioprocessing allows hPSC expansion and differentiation to be carried out within a single bioreactor as a single unit opearation.
Table 4.
Key performance characteristics of studies investigating the integrated expansion and differentiation of hPSCs
Culture conditions | Cell type (iPSC/ESC) | Target cell type | Expansion max.cell density (cells/mL) (fold expansion) | Differentiation max. cell density (cells/mL) (fold expansion) | Reported differentiation efficiency (%) | Process time (days) | Target cells produced per input hPSC | Xeno-free (Y/N) | Refs. |
---|---|---|---|---|---|---|---|---|---|
Microcarrier (DE53 Whatman) | hiPSC | Neural progenitor cells (NPCs) | 6.1 × 106 (20) | 1.1 × 106 (16.6) | 78 ± 4.7 | 25 | 333 | N | [77] |
Microcarrier (DE53 Whatman) | hESC | NPCs | 4.3 × 106 (21.3) | 1 × 106 (17.7) | 83 ± 8.5 | 23 | 371 | N | [77] |
Aggregate culture | hESC | Definitive endoderm progenitors (DEPs) | ND (5000)a) | ND (23.5) | > 80% | 22 | 65, 000a) | N | [67] |
Microcarrier (collagen-coated hyclone) | hESC | DEPs | 1 × 106 (34–45) | 4 × 105 (ND) | 84.2 ± 2.3% | 12 | 4 | N | [74] |
Parameters given for both expansion and differentiation where available. ND = No data given.
hESCs underwent four rounds of expansion during this study, as opposed to one round of expansion in other studies shown here. This may contribute to the disparity in performance parameters between this study and others shown here. ((Please start this sentence as a new line))
Early investigations incorporating expansion and differentiation focused on overcoming the technical hurdles of carrying out two different operations in one integrated process step; as such only modest yields of target cells were achieved [74]. Recent studies have sought to optimise integrated iPSC bioprocesses via strategies such as the determination of optimal aggregate size during hPSC culture and differentiation [67]. Switching feeding regimes from once to twice per day was found to double the achievable cell density during the expansion phase of an integrated bioprocess, although process economic analysis comparing the two approaches was not offered [77]. The reported expansion folds and differentiation efficiencies for integrated bioprocesses compare well with separated systems of a similar nature (Table 4).
To date, no studies have produced an integrated process for hPSCs from derivation all the way through to differentiation. This has been achieved with mouse iPSCs (miPSCs) in a SUB [109]. To our knowledge, there are only two studies exhibiting ‘suspension culture reprogrammed iPSCs’, both of which deal with miPSCs [109, 110]. Translation of integrated miPSC production techniques described by Baptista et al. [110] to hiPSC processing may be particularly useful in the production of autologous stem cell therapies, where continuous derivation of large numbers hiPSCs would help reduce the bottlenecks bought about by cellular reprogramming. Moreover, it may provide a ‘black box’ platform to derive, expand and differentiate a patient's cells in a single, contained unit that could be installed at point-of-care centres for relevant disease types. Novel, controlled miniature bioreactor systems, such as the ambr15 (Sartorius AG), might offer a suitable platform for autologous hiPSC product manufacture, where limited cell numbers are required and scale-out strategies necessitate alternatives to large-scale bioreactors (Fig. 1B and 1C).
6 Concluding remarks
The fulfilment of the clinical potential of hPSCs depends on the development of scalable, robust, GMP-compliant bioprocesses. Concentrated efforts to develop rigorously designed bioprocesses with a greater emphasis placed on QbD will continue to enhance process understanding with respects to defining critical quality attributes and identifying key process variables that must be controlled. hPSC-derived products are subject to strict economic boundaries owing to the nature of global healthcare systems. Future work must build upon the promise of works reviewed in this paper in order to make delivering such products on budget an achievable goal. Novel approaches discussed in this review offer methods to further optimise hPSC bioprocessing platforms.
Michael J. Jenkins is a doctoral research student in Biochemical Engineering at University College London (UCL) in the UK. Michael's project, in collaboration with Neusentis Ltd., focuses on cost-effective bioprocess design for hPSC-derived cell products including cell therapies and drug screening tools. His research involves the creation of novel decisional tools that combine process economics analysis, bioprocess optimisation and uncertainty analyses of hPSC bioprocesses. He obtained his Master's degree in Biochemical Engineering at UCL, with a year as an exchange student within the Biotechnology Department at Lund University, Sweden.
Suzanne S. Farid is Professor in Bioprocess Systems Engineering at the Advanced Centre for Biochemical Engineering at University College London (UCL) in the UK and Co-Director of the EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies. She leads research on ‘Bioprocess Decisional Tools’ that has pioneered the development of algorithms at the process–business interface to facilitate cost-effective bioprocess design, capacity planning and R&D portfolio management. She sits on the UK BioIndustry Association Manufacturing Advisory Committee and is a Fellow of the IChemE. She obtained her Bachelor's and Ph.D. degrees in Biochemical Engineering with UCL and Lonza Biologics.
Acknowledgments
Financial support from the UK Engineering and Physical Sciences Research Council (EPSRC) (grant reference number: EP/G034656/1) and Neusentis Ltd. is gratefully acknowledged. UCL hosts the EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies with Imperial College London and a consortium of industrial and government partners. The authors are very grateful to Sally Hassan in the Department of Biochemical Engineering at UCL for her contributions towards Table 1.
The authors declare no financial or commercial conflict of interest.
Glossary
- COG
cost of goods
- cGMP
current good manufacturing practice
- cGTP
current good tissue practice
- FCI
fixed capital investment
- hPSC
human pluripotent stem cell
- QbD
quality by design
- SUB
single-use bioreactor
References
- 1.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 2.Takahashi K, Tanabe K, Ohnuki M, Narita M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 3.Robinton DA, Daley GQ. The promise of induced pluripotent stem cells in research and therapy. Nature. 2012;481:295–305. doi: 10.1038/nature10761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ebert AD, Svendsen CN. Human stem cells and drug screening: Opportunities and challenges. Nat. Rev. Drug Discov. 2010;9:367–372. doi: 10.1038/nrd3000. [DOI] [PubMed] [Google Scholar]
- 5.Grskovic M, Javaherian A, Strulovici B, Daley GQ. Induced pluripotent stem cells –Opportunities for disease modelling and drug discovery. Nat. Rev. Drug Discov. 2011;10:915–929. doi: 10.1038/nrd3577. [DOI] [PubMed] [Google Scholar]
- 6.Kiskinis E, Eggan K. Progress toward the clinical application of patient-specific pluripotent stem cells. J. Clin. Invest. 2010;120:51–59. doi: 10.1172/JCI40553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wu SM, Hochedlinger K. Harnessing the potential of induced pluripotent stem cells for regenerative medicine. Nat. Cell Biol. 2011;13:497–505. doi: 10.1038/ncb0511-497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zeevi-Levin N, Itskovitz-Eldor J, Binah O. Cardiomyocytes derived from human pluripotent stem cells for drug screening. Pharmacol. Ther. 2012;134:180–188. doi: 10.1016/j.pharmthera.2012.01.005. [DOI] [PubMed] [Google Scholar]
- 9.Rajamohan D, Matsa E, Kalra S, Crutchley J, et al. Current status of drug screening and disease modelling in human pluripotent stem cells. Bioessays. 2013;35:281–298. doi: 10.1002/bies.201200053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.McKernan R, McNeish J, Smith D. Pharma's developing interest in stem cells. Cell Stem Cell. 2010;6:517–520. doi: 10.1016/j.stem.2010.05.012. [DOI] [PubMed] [Google Scholar]
- 11.Smith DM. Assessing commercial opportunities for autologous and allogeneic cell-based products. Regen. Med. 2012;7:721–732. doi: 10.2217/rme.12.40. [DOI] [PubMed] [Google Scholar]
- 12.Sivertsson L, Synnergren J, Jensen J, Bjorquist P, Ingelman-Sundberg M. Hepatic differentiation and maturation of human embryonic stem cells cultured in a perfused three-dimensional bioreactor. Stem Cells Dev. 2013;22:581–594. doi: 10.1089/scd.2012.0202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Schwartz SD, Hubschman JP, Heilwell G, Franco-Cardenas V, et al. Embryonic stem cell trials for macular degeneration: A preliminary report. Lancet. 2012;379:713–720. doi: 10.1016/S0140-6736(12)60028-2. [DOI] [PubMed] [Google Scholar]
- 14.Idelson M, Alper R, Obolensky A, Ben-Shushan E, et al. Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. Cell Stem Cell. 2009;5:396–408. doi: 10.1016/j.stem.2009.07.002. [DOI] [PubMed] [Google Scholar]
- 15.Schulz TC, Young HY, Agulnick AD, Babin MJ, et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE. 2012;7:e37004. doi: 10.1371/journal.pone.0037004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Parker, C., Novel application and requirements for the broad adoption of iPSC-derived cellular materials. World Stem Cells Regenerative Medicine Congress 2014, London, UK, 2014.
- 17. Kagimoto, H. T. S., Debating the first iPS cell-derived therapy human trial for AMD. World Stem Cells Regenerative Medicine Congress 2014, London, UK, 2014.
- 18.Placzek MR, Chung IM, Macedo HM, Ismail S, et al. Stem cell bioprocessing: Fundamentals and principles. J. R. Soc. Interface. 2009;6:209–232. doi: 10.1098/rsif.2008.0442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Mason C, Dunnill P. Quantities of cells used for regenerative medicine and some implications for clinicians and bioprocessors. Regen. Med. 2009;4:153–157. doi: 10.2217/17460751.4.2.153. [DOI] [PubMed] [Google Scholar]
- 20.Simaria AS, Hassan S, Varadaraju H, Rowley J, et al. Allogeneic cell therapy bioprocess economics and optimization: Single-use cell expansion technologies. Biotechnol. Bioeng. 2014;111:69–83. doi: 10.1002/bit.25008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lancaster MA, Knoblich JA. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 2014;345:283. doi: 10.1126/science.1247125. [DOI] [PubMed] [Google Scholar]
- 22.Lancaster MA, Renner M, Martin CA, Wenzel D, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373–379. doi: 10.1038/nature12517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Takasato M, Er PX, Becroft M, Vanslambrouck JM, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat. Cell Biol. 2014;16:118–126. doi: 10.1038/ncb2894. [DOI] [PubMed] [Google Scholar]
- 24.Nakano T, Ando S, Takata N, Kawada M, et al. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell. 2012;10:771–785. doi: 10.1016/j.stem.2012.05.009. [DOI] [PubMed] [Google Scholar]
- 25.Want AJ, Nienow AW, Hewitt CJ, Coopman K. Large-scale expansion and exploitation of pluripotent stem cells for regenerative medicine purposes: Beyond the T flask. Regen. Med. 2012;7:71–84. doi: 10.2217/rme.11.101. [DOI] [PubMed] [Google Scholar]
- 26.Klimanskaya I, Rosenthal N, Lanza R. Derive and conquer: Sourcing and differentiating stem cells for therapeutic applications. Nat. Rev. Drug Discov. 2008;7:131–142. doi: 10.1038/nrd2403. [DOI] [PubMed] [Google Scholar]
- 27.Sart S, Agathos SN, Li Y. Engineering stem cell fate with biochemical and biomechanical properties of microcarriers. Biotechnol. Prog. 2013;29:1354–1366. doi: 10.1002/btpr.1825. [DOI] [PubMed] [Google Scholar]
- 28.Serra M, Brito C, Correia C, Alves PM. Process engineering of human pluripotent stem cells for clinical application. Trends Biotechnol. 2012;30:350–359. doi: 10.1016/j.tibtech.2012.03.003. [DOI] [PubMed] [Google Scholar]
- 29.Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol. Adv. 2013;31:1600–1623. doi: 10.1016/j.biotechadv.2013.08.009. [DOI] [PubMed] [Google Scholar]
- 30.Chen AK, Reuveny S, Oh SK. Application of human mesenchymal and pluripotent stem cell microcarrier cultures in cellular therapy: Achievements and future direction. Biotechnol. Adv. 2013;31:1032–1046. doi: 10.1016/j.biotechadv.2013.03.006. [DOI] [PubMed] [Google Scholar]
- 31.Villa-Diaz LG, Ross AM, Lahann J, Krebsbach PH. Concise review: The evolution of human pluripotent stem cell culture: From feeder cells to synthetic coatings. Stem Cells. 2013;31:1–7. doi: 10.1002/stem.1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sart S, Schneider YJ, Li Y, Agathos SN. Stem cell bioprocess engineering towards cGMP production and clinical applications. Cytotechnology. 2014;66:709–722. doi: 10.1007/s10616-013-9687-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lund AW, Yener B, Stegemann JP, Plopper GE. The natural and engineered 3D microenvironment as a regulatory cue during stem cell fate determination. Tissue Eng. B Rev. 2009;15:371–380. doi: 10.1089/ten.teb.2009.0270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lang HJ. Simplified approach to preliminary cost estimates. Chem. Eng. 1948;55:112–113. [Google Scholar]
- 35.Pollock J, Ho SV, Farid SS. Fed-batch and perfusion culture processes: Economic, environmental, and operational feasibility under uncertainty. Biotechnol. Bioeng. 2013;110:206–219. doi: 10.1002/bit.24608. [DOI] [PubMed] [Google Scholar]
- 36.Darkins CL, Mandenius C-F. Design of large-scale manufacturing of induced pluripotent stem cell derived cardiomyocytes. Chem. Eng. Res. Des. 2014;92:1142–1152. [Google Scholar]
- 37. Hassan, S., Simaria, A. S., Varadaraju, H., Rowley, J., Farid, S. S., A tool to predict the economic benefit of alternative technologies for allogeneic cell therapy manufacture at commercial scale. 245th American Chemical Society (ACS) National Meeting, New Orleans, Louisiana, USA, 2013.
- 38. Farid, S. S., Hassan, S., Simaria, A. S., Varadaraju, H., Warren, K., Can the cell therapy sector achieve acceptable cost of goods at the commercial scale? 20th ISCT Annual Meeting, Paris, France, 2014.
- 39.Veraitch FS, Scott R, Wong JW, Lye GJ, Mason C. The impact of manual processing on the expansion and directed differentiation of embryonic stem cells. Biotechnol. Bioeng. 2008;99:1216–1229. doi: 10.1002/bit.21673. [DOI] [PubMed] [Google Scholar]
- 40.Farid SS, Washbrook J, Titchener-Hooker NJ. Decision-support tool for assessing biomanufacturing strategies under uncertainty: Stainless steel versus disposable equipment for clinical trial material preparation. Biotechnol. Prog. 2005;21:486–497. doi: 10.1021/bp049692b. [DOI] [PubMed] [Google Scholar]
- 41.Lim AC, Washbrook J, Titchener-Hooker NJ, Farid SS. A computer-aided approach to compare the production economics of fed-batch and perfusion culture under uncertainty. Biotechnol. Bioeng. 2006;93:687–697. doi: 10.1002/bit.20757. [DOI] [PubMed] [Google Scholar]
- 42.Eiselleova L, Peterkova I, Neradil J, Slaninova I, et al. Comparative study of mouse and human feeder cells for human embryonic stem cells. Int. J. Dev. Biol. 2008;52:353–363. doi: 10.1387/ijdb.082590le. [DOI] [PubMed] [Google Scholar]
- 43.Rowley J, Abraham E, Campbell A, Brandwein H, Oh S. Meeting lot-size challenges of manufacturing adherent cells for therapy. BioProcess Int. 2012;10:16–22. [Google Scholar]
- 44.Rodrigues CAV, Fernandes TG, Diogo MM, da Silva CL, Cabral JMS. Stem cell cultivation in bioreactors. Biotechnol. Adv. 2011;29:815–829. doi: 10.1016/j.biotechadv.2011.06.009. [DOI] [PubMed] [Google Scholar]
- 45.Brandenburger R, Burger S, Campbell A, Fong T, et al. Cell therapy bioprocessing. BioProcess Int. 2011;9:30–37. [Google Scholar]
- 46.Thomas RJ, Anderson D, Chandra A, Smith NM, et al. Automated, scalable culture of human embryonic stem cells in feeder-free conditions. Biotechnol. Bioeng. 2009;102:1636–1644. doi: 10.1002/bit.22187. [DOI] [PubMed] [Google Scholar]
- 47.Oh SKW, Chen AK, Mok Y, Chen X, et al. Long-term microcarrier suspension cultures of human embryonic stem cells. Stem Cell Res. 2009;2:219–230. doi: 10.1016/j.scr.2009.02.005. [DOI] [PubMed] [Google Scholar]
- 48.Gerecht-Nir S, Cohen S, Itskovitz-Eldor J. Bioreactor cultivation enhances the efficiency of human embryoid body (hEB) formation and differentiation. Biotechnol. Bioeng. 2004;86:493–502. doi: 10.1002/bit.20045. [DOI] [PubMed] [Google Scholar]
- 49.Olmer R, Lange A, Selzer S, Kasper C, et al. Suspension culture of human pluripotent stem cells in controlled, stirred bioreactors. Tissue Eng. C Methods. 2012;18:772–784. doi: 10.1089/ten.tec.2011.0717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chisti Y. Hydrodynamic damage to animal cells. Crit. Rev. Biotechnol. 2001;21:67–110. doi: 10.1080/20013891081692. [DOI] [PubMed] [Google Scholar]
- 51.Leung HW, Chen A, Choo AB, Reuveny S, Oh SK. Agitation can induce differentiation of human pluripotent stem cells in microcarrier cultures. Tissue Eng. C Methods. 2011;17:165–172. doi: 10.1089/ten.TEC.2010.0320. [DOI] [PubMed] [Google Scholar]
- 52.Chen G, Gulbranson DR, Hou Z, Bolin JM, et al. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods. 2011;8:424–429. doi: 10.1038/nmeth.1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wang Y, Chou BK, Dowey S, He C, et al. Scalable expansion of human induced pluripotent stem cells in the defined xeno-free E8 medium under adherent and suspension culture conditions. Stem Cell Res. 2013;11:1103–1116. doi: 10.1016/j.scr.2013.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Marinho PA, Vareschini DT, Gomes IC, Paulsen Bda S, et al. Xeno-free production of human embryonic stem cells in stirred microcarrier systems using a novel animal/human-component-free medium. Tissue Eng. C Methods. 2013;19:146–155. doi: 10.1089/ten.TEC.2012.0141. [DOI] [PubMed] [Google Scholar]
- 55.Phillips BW, Horne R, Lay TS, Rust WL, et al. Attachment and growth of human embryonic stem cells on microcarriers. J. Biotechnol. 2008;138:24–32. doi: 10.1016/j.jbiotec.2008.07.1997. [DOI] [PubMed] [Google Scholar]
- 56.Fernandes AM, Marinho PA, Sartore RC, Paulsen BS, et al. Successful scale-up of human embryonic stem cell production in a stirred microcarrier culture system. Braz. J. Med. Biol. Res. 2009;42:515–522. doi: 10.1590/s0100-879x2009000600007. [DOI] [PubMed] [Google Scholar]
- 57.Serra M, Brito C, Sousa MF, Jensen J, et al. Improving expansion of pluripotent human embryonic stem cells in perfused bioreactors through oxygen control. J. Biotechnol. 2010;148:208–215. doi: 10.1016/j.jbiotec.2010.06.015. [DOI] [PubMed] [Google Scholar]
- 58.Chen AK, Chen X, Lim YM, Reuveny S, Oh SK. Inhibition of ROCK-myosin II signaling pathway enables culturing of human pluripotent stem cells on microcarriers without extracellular matrix coating. Tissue Eng. C Methods. 2014;20:227–238. doi: 10.1089/ten.TEC.2013.0191. [DOI] [PubMed] [Google Scholar]
- 59.Rodin S, Domogatskaya A, Strom S, Hansson EM, et al. Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nat. Biotechnol. 2010;28:611–615. doi: 10.1038/nbt.1620. [DOI] [PubMed] [Google Scholar]
- 60.Chen AK, Chen X, Choo AB, Reuveny S, Oh SK. Critical microcarrier properties affecting the expansion of undifferentiated human embryonic stem cells. Stem Cell Res. 2011;7:97–111. doi: 10.1016/j.scr.2011.04.007. [DOI] [PubMed] [Google Scholar]
- 61.Melkoumian Z, Weber JL, Weber DM, Fadeev AG, et al. Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells. Nat. Biotechnol. 2010;28:606–610. doi: 10.1038/nbt.1629. [DOI] [PubMed] [Google Scholar]
- 62.Villa-Diaz LG, Nandivada H, Ding J, Nogueira-de-Souza NC, et al. Synthetic polymer coatings for long-term growth of human embryonic stem cells. Nat. Biotechnol. 2010;28:581–583. doi: 10.1038/nbt.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fan Y, Hsiung M, Cheng C, Tzanakakis ES. Facile engineering of xeno-free microcarriers for the scalable cultivation of human pluripotent stem cells in stirred suspension. Tissue Eng. A. 2014;20:588–599. doi: 10.1089/ten.tea.2013.0219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Tamura A, Kobayashi J, Yamato M, Okano T. Temperature-responsive poly(N-isopropylacrylamide)-grafted microcarriers for large-scale non-invasive harvest of anchorage-dependent cells. Biomaterials. 2012;33:3803–3812. doi: 10.1016/j.biomaterials.2012.01.060. [DOI] [PubMed] [Google Scholar]
- 65.Watanabe K, Ueno M, Kamiya D, Nishiyama A, et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol. 2007;25:681–686. doi: 10.1038/nbt1310. [DOI] [PubMed] [Google Scholar]
- 66.Krawetz R, Taiani JT, Liu S, Meng G, et al. Large-scale expansion of pluripotent human embryonic stem cells in stirred-suspension bioreactors. Tissue Eng. C Methods. 2010;16:573–582. doi: 10.1089/ten.TEC.2009.0228. [DOI] [PubMed] [Google Scholar]
- 67.Ungrin MD, Clarke G, Yin T, Niebrugge S, et al. Rational bioprocess design for human pluripotent stem cell expansion and endoderm differentiation based on cellular dynamics. Biotechnol. Bioeng. 2012;109:853–866. doi: 10.1002/bit.24375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Abbasalizadeh S, Larijani MR, Samadian A, Baharvand H. Bioprocess development for mass production of size-controlled human pluripotent stem cell aggregates in stirred suspension bioreactor. Tissue Eng. C Methods. 2012;18:831–851. doi: 10.1089/ten.TEC.2012.0161. [DOI] [PubMed] [Google Scholar]
- 69.Chen VC, Couture SM, Ye J, Lin Z, et al. Scalable GMP compliant suspension culture system for human ES cells. Stem Cell Res. 2012;8:388–402. doi: 10.1016/j.scr.2012.02.001. [DOI] [PubMed] [Google Scholar]
- 70.Zweigerdt R, Olmer R, Singh H, Haverich A, Martin U. Scalable expansion of human pluripotent stem cells in suspension culture. Nat. Protoc. 2011;6:689–700. doi: 10.1038/nprot.2011.318. [DOI] [PubMed] [Google Scholar]
- 71.Amit M, Laevsky I, Miropolsky Y, Shariki K, et al. Dynamic suspension culture for scalable expansion of undifferentiated human pluripotent stem cells. Nat. Protoc. 2011;6:572–579. doi: 10.1038/nprot.2011.325. [DOI] [PubMed] [Google Scholar]
- 72.Olmer R, Haase A, Merkert S, Cui W, et al. Long term expansion of undifferentiated human iPS and ES cells in suspension culture using a defined medium. Stem Cell Res. 2010;5:51–64. doi: 10.1016/j.scr.2010.03.005. [DOI] [PubMed] [Google Scholar]
- 73.Singh H, Mok P, Balakrishnan T, Rahmat SN, Zweigerdt R. Up-scaling single cell-inoculated suspension culture of human embryonic stem cells. Stem Cell Res. 2010;4:165–179. doi: 10.1016/j.scr.2010.03.001. [DOI] [PubMed] [Google Scholar]
- 74.Lock LT, Tzanakakis ES. Expansion and differentiation of human embryonic stem cells to endoderm progeny in a microcarrier stirred-suspension culture. Tissue Eng. A. 2009;15:2051–2063. doi: 10.1089/ten.tea.2008.0455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Fridley KM, Kinney MA, McDevitt TC. Hydrodynamic modulation of pluripotent stem cells. Stem Cell Res. Ther. 2012;3:45. doi: 10.1186/scrt136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kehoe DE, Jing DH, Lock LT, Tzanakakis ES. Scalable stirred-suspension bioreactor culture of human pluripotent stem cells. Tissue Eng. A. 2010;16:405–421. doi: 10.1089/ten.tea.2009.0454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bardy J, Chen AK, Lim YM, Wu S, et al. Microcarrier suspension cultures for high-density expansion and differentiation of human pluripotent stem cells to neural progenitor cells. Tissue Eng. C Methods. 2013;19:166–180. doi: 10.1089/ten.TEC.2012.0146. [DOI] [PubMed] [Google Scholar]
- 78.Mordwinkin N, Burridge P, Wu J. A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J. Cardiovasc. Transl. Res. 2013;6:22–30. doi: 10.1007/s12265-012-9423-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Karumbayaram S, Novitch BG, Patterson M, Umbach JA, et al. Directed differentiation of human-induced pluripotent stem cells generates active motor neurons. Stem Cells. 2009;27:806–811. doi: 10.1002/stem.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhang J, Wilson GF, Soerens AG, Koonce CH, et al. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ. Res. 2009;104:e30–e41. doi: 10.1161/CIRCRESAHA.108.192237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ghodsizadeh A, Taei A, Totonchi M, Seifinejad A, et al. Generation of liver disease-specific induced pluripotent stem cells along with efficient differentiation to functional hepatocyte-like cells. Stem Cell Rev. 2010;6:622–632. doi: 10.1007/s12015-010-9189-3. [DOI] [PubMed] [Google Scholar]
- 82.Sullivan GJ, Hay DC, Park IH, Fletcher J, et al. Generation of functional human hepatic endoderm from human induced pluripotent stem cells. Hepatology. 2010;51:329–335. doi: 10.1002/hep.23335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Martinez Y, Dubois-Dauphin M, Krause KH. Generation and applications of human pluripotent stem cells induced into neural lineages and neural tissues. Front. Physiol. 2012;3:47. doi: 10.3389/fphys.2012.00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Surmacz B, Fox H, Gutteridge A, Lubitz S, Whiting P. Directing differentiation of human embryonic stem cells toward anterior neural ectoderm using small molecules. Stem Cells. 2012;30:1875–1884. doi: 10.1002/stem.1166. [DOI] [PubMed] [Google Scholar]
- 85.Zhu S, Wei W, Ding S. Chemical strategies for stem cell biology and regenerative medicine. Annu. Rev. Biomed. Eng. 2011;13:73–90. doi: 10.1146/annurev-bioeng-071910-124715. [DOI] [PubMed] [Google Scholar]
- 86.Li W, Jiang K, Ding S. Concise review: A chemical approach to control cell fate and function. Stem Cells. 2012;30:61–68. doi: 10.1002/stem.768. [DOI] [PubMed] [Google Scholar]
- 87.Burridge PW, Thompson S, Millrod MA, Weinberg S, et al. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLoS ONE. 2011:e18293. doi: 10.1371/journal.pone.0018293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chambers SM, Qi Y, Mica Y, Lee G, et al. Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors. Nat. Biotechnol. 2012;30:715–720. doi: 10.1038/nbt.2249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Liu Q, Pedersen OZ, Peng J, Couture LA, et al. Optimizing dopaminergic differentiation of pluripotent stem cells for the manufacture of dopaminergic neurons for transplantation. Cytotherapy. 2013;15:999–1010. doi: 10.1016/j.jcyt.2013.03.006. [DOI] [PubMed] [Google Scholar]
- 90. Diekmann, U., Lenzen, S., Naujok, O., A reliable and efficient protocol for human pluripotent stem cell differentiation into the definitive endoderm based on dispersed single cells. Stem Cells Dev 2014, DOI: [DOI] [PubMed]
- 91.Lippmann ES, Estevez-Silva MC, Ashton RS. Defined human pluripotent stem cell culture enables highly efficient neuroepithelium derivation without small molecule inhibitors. Stem Cells. 2014;32:1032–1042. doi: 10.1002/stem.1622. [DOI] [PubMed] [Google Scholar]
- 92.Lu SJ, Kelley T, Feng Q, Chen A, et al. 3D microcarrier system for efficient differentiation of human pluripotent stem cells into hematopoietic cells without feeders and serum [corrected] Regen. Med. 2013;8:413–424. doi: 10.2217/rme.13.36. [DOI] [PubMed] [Google Scholar]
- 93.Lecina M, Ting S, Choo A, Reuveny S, Oh S. Scalable platform for human embryonic stem cell differentiation to cardiomyocytes in suspended microcarrier cultures. Tissue Eng. C Methods. 2010;16:1609–1619. doi: 10.1089/ten.TEC.2010.0104. [DOI] [PubMed] [Google Scholar]
- 94.Vosough M, Omidinia E, Kadivar M, Shokrgozar MA, et al. Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev. 2013;22:2693–2705. doi: 10.1089/scd.2013.0088. [DOI] [PubMed] [Google Scholar]
- 95.Zwi-Dantsis L, Mizrahi I, Arbel G, Gepstein A, Gepstein L. Scalable production of cardiomyocytes derived from c-Myc free induced pluripotent stem cells. Tissue Eng. A. 2011;17:1027–1037. doi: 10.1089/ten.TEA.2010.0235. [DOI] [PubMed] [Google Scholar]
- 96.Niebruegge S, Nehring A, Bar H, Schroeder M, et al. Cardiomyocyte production in mass suspension culture: Embryonic stem cells as a source for great amounts of functional cardiomyocytes. Tissue Eng. A. 2008;14:1591–1601. doi: 10.1089/ten.tea.2007.0247. [DOI] [PubMed] [Google Scholar]
- 97.Weng Z, Kong CW, Ren L, Karakikes I, et al. A simple, cost-effective but highly efficient system for deriving ventricular cardiomyocytes from human pluripotent stem cells. Stem Cells Dev. 2014;23:1704–1716. doi: 10.1089/scd.2013.0509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bratt-Leal AM, Nguyen AH, Hammersmith KA, Singh A, McDevitt TC. A microparticle approach to morphogen delivery within pluripotent stem cell aggregates. Biomaterials. 2013;34:7227–7235. doi: 10.1016/j.biomaterials.2013.05.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wolfe RP, Leleux J, Nerem RM, Ahsan T. Effects of shear stress on germ lineage specification of embryonic stem cells. Integr. Biol. (Camb.) 2012;4:1263–1273. doi: 10.1039/c2ib20040f. [DOI] [PubMed] [Google Scholar]
- 100.Wolfe RP, Ahsan T. Shear stress during early embryonic stem cell differentiation promotes hematopoietic and endothelial phenotypes. Biotechnol. Bioeng. 2013;110:1231–1242. doi: 10.1002/bit.24782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ahsan T, Nerem RM. Fluid shear stress promotes an endothelial-like phenotype during the early differentiation of embryonic stem cells. Tissue Eng. A. 2010;16:3547–3553. doi: 10.1089/ten.tea.2010.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Adamo L, Naveiras O, Wenzel PL, McKinney-Freeman S, et al. Biomechanical forces promote embryonic haematopoiesis. Nature. 2009;459:1131–1135. doi: 10.1038/nature08073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bae D, Mondragon-Teran P, Hernandez D, Ruban L, et al. Hypoxia enhances the generation of retinal progenitor cells from human induced pluripotent and embryonic stem cells. Stem Cells Dev. 2012;21:1344–1355. doi: 10.1089/scd.2011.0225. [DOI] [PubMed] [Google Scholar]
- 104.Cimetta E, Figallo E, Cannizzaro C, Elvassore N, Vunjak-Novakovic G. Micro-bioreactor arrays for controlling cellular environments: Design principles for human embryonic stem cell applications. Methods. 2009;47:81–89. doi: 10.1016/j.ymeth.2008.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wang D, Bodovitz S. Single cell analysis: The new frontier in ‘omics’. Trends Biotechnol. 2010;28:281–290. doi: 10.1016/j.tibtech.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Prabhakarpandian B, Shen MC, Pant K, Kiani MF. Microfluidic devices for modeling cell–cell and particle–cell interactions in the microvasculature. Microvasc. Res. 2011;82:210–220. doi: 10.1016/j.mvr.2011.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Reichen M, Macown RJ, Jaccard N, Super A, et al. Microfabricated modular scale-down device for regenerative medicine process development. PLoS ONE. 2012;7:e52246. doi: 10.1371/journal.pone.0052246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Macown RJ, Veraitch FS, Szita N. Robust, microfabricated culture devices with improved control over the soluble microenvironment for the culture of embryonic stem cells. Biotechnol. J. 2014;9:805–813. doi: 10.1002/biot.201300245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fluri DA, Tonge PD, Song H, Baptista RP, et al. Derivation, expansion and differentiation of induced pluripotent stem cells in continuous suspension cultures. Nat. Methods. 2012;9:509–516. doi: 10.1038/nmeth.1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Baptista RP, Fluri DA, Zandstra PW. High density continuous production of murine pluripotent cells in an acoustic perfused bioreactor at different oxygen concentrations. Biotechnol. Bioeng. 2013;110:648–655. doi: 10.1002/bit.24717. [DOI] [PubMed] [Google Scholar]
- 111.Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS. Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia. 2005;49:385–396. doi: 10.1002/glia.20127. [DOI] [PubMed] [Google Scholar]