Abstract

Cell therapies, potent populations of cells used to treat disease and injury, can be strategically manufactured with biomaterial intervention to improve clinical translation. In this viewpoint, we discuss biomaterial design and integration into cell manufacturing steps to achieve three main goals: scale-up, phenotype control, and selection of potent cells. Material properties can be engineered to influence the cell–biomaterial interface and, therefore, impart desirable cell behavior such as growth, secretory activity, and differentiation. Future directions for the field should capitalize on the combinatorial design of biomaterial properties to yield highly specific and potent cell populations. Furthermore, future biomaterials could contribute to novel high-throughput cell separation technologies that can individually select the most therapeutically relevant cells within a produced batch.
1. Introduction
1.1. Cell Therapies in the Clinic
Cell therapies, viable and dynamic cell populations that can be transferred into the body to prevent or treat disease, are of high interest to the research and clinical community following the USFDA approval of the first products in 2010.1,2 Unlike small molecule pharmaceutics, cell therapies capitalize on sophisticated mechanisms of action that can influence and integrate with multiple simultaneous healing processes. With this potential and due to the convergence of biomaterials, stem cell biology, immunotherapy, and gene editing research fields, there are currently thousands of active clinical trials for cell therapies.3−5 More than 10,000 open clinical trials, of which 214 are in phase IV, were identified following the search for “cell therapy” on the ClinicalTrials.gov database as of 2024. Although many clinical trials involving cell-based therapeutics have shown promising results, reproducible manufacturing of highly potent cells at scale remains a major limitation.3,6−8
1.2. Autologous and Allogenic Cell Therapies
Therapeutic cells can be derived from the patient or come from donor tissue. Autologous transplantation uses cells from the patient. As such, the likelihood for immune rejection is reduced,3 but often the cells must be culture expanded prior to reintroduction to the patient.3 Allogenic cells, or those that come from another person, benefit from the ability to store and/or bank the cells and therefore provide immediate availability for the treatment of many patients. As such, allogenic transplantation is better suited for urgent medical situations and has become a major research emphasis with regards to commercial-scale manufacturing.3,9
1.3. Scalable Cell Culture
Large-scale manufacturing of therapeutic cells remains a bottleneck to clinical translation and eventually commercialization.4,6,9−11 Therapeutic cell doses vary but are on the order of 109 cells per patient.6 Due to these large dose requirements, conventional 2D culture flasks demonstrate limitations associated with large footprints and large process volumes at scale.6,7 As such, several 3D culture platforms have been adopted to address these shortcomings.
In this viewpoint, we will discuss bioreactor design for adherent cell culture systems since there is a direct interaction between cells and engineered biomaterial surfaces in this case. Several 3D hollow-fiber systems have been cleared by the FDA for clinical use.7 The use of microcarriers (microparticles of approximately 125–300 μm diameter12 used to increase surface area for cell attachment) suspended in a stir-tank reactor provide an alternative approach and can seamlessly incorporate therapeutic specific biomaterial design.6,7 To date, microparticle design has primarily focused on increasing cell proliferation rates to capitalize on yield with minimal emphasis on potency engineering.4−6 Nevertheless, biomaterials, as coatings in hollow-fiber reactors or as microcarriers, provide a great opportunity to synergize both phenotypic control and cell growth. However, the optimization of biomaterials for specific cell phenotypes remains a challenge.
1.4. Engineering Cell Potency
Therapeutic cell potency will be defined for the purposes of this viewpoint as the capacity of a cell to beneficially regulate the healing process. This can be achieved through enhanced paracrine signaling, differentiation, or tissue remodeling and therefore defines the ideal phenotypic state post manufacturing.3,7,13,14 The cell phenotype can be engineered genetically, usually by a viral vector, or directly through the use of transcription factors or biomaterials.15,16 More specifically, direct control of cell potency does not involve driving the cells to pass through a pluripotent stem cell stage.15,17−19 Genetic engineering of cell potency is a powerful regenerative medicine tool that can yield highly specific cell fates; however, it can lead to epigenetic remodeling and tumor formation.15,20 As such, genetic engineering will only be briefly discussed in this viewpoint. Direct engineering of cell potency, on the other hand, provides an alternative approach to cell fate control that has a low risk for epigenetic remodeling.15 While transcription factors were initially the standard for direct engineering of cell potency, the efficiency is low.16 Biomaterials, however, can provide physical and/or biochemical cues to increase the efficiency of direct engineering of cell potency, either through reprogramming or shifts in the phenotypic state.15−18 Although biomaterials have been used to engineer more theoretically potent cell populations, potency variability remains a translational challenge.
1.5. Heterogeneity in Cell Culture
Finally, in order to observe consistent therapeutic outcomes, variation in the cell phenotype should be minimized. Functional heterogeneity can exist within a single population of therapeutic cells or exist as batch-to-batch differences.21−23 Initial cell populations may vary in phenotype and efficacy based on parameters such as donor age, donor sex, donor condition, or cell source.3 For example, it is well-known that cellular senescence is highly correlated with age.24 Not only may cells from an elderly donor lead to a different therapeutic outcome compared to a young donor, but also the fraction of senescent cells within the donor population can have an entirely unique and deleterious secretome, called the senescent-associated secretory phenotype (SASP).24,25 While heterogeneity can partially be attributed to stochasticity, cells are also very sensitive to microenvironmental cues. As such, phenotypic changes can arise and compound during many manufacturing steps.3,21,26 Again, biomaterials provide an approach to reducing cell heterogeneity either through direct sorting/filtering of cell populations or indirectly through cell phenotype control. Selection of a subpopulation of cells at the end of the manufacturing process provides a methodology to reduce variability in therapeutic efficacy; however, many of these techniques have not been fully integrated into the manufacturing pipeline.
1.6. Viewpoint Goal
This viewpoint will summarize current biomaterial strategies as well as highlight challenges that can be further addressed by biomaterials scientists in order to scale-up the manufacture of therapeutic cells and improve cell potency and specificity (Figure 1). In this viewpoint, advances in the culture of induced pluripotent stem cells (iPSCs) and mesenchymal stromal cells (MSCs) will be used as examples, but the general principles discussed can be extended to all clinically relevant cell types.
Figure 1.
Biomaterial integration into the cell therapy manufacturing pipeline. Biomaterials can be used to both facilitate commercial scale-up of cells and impart cell potency to increase clinical efficacy. For adherent cell culture, cells can be expanded on microcarriers in suspension to reach clinically relevant lot sizes (1012 cells). The properties of the microcarrier (stiffness/elasticity, ligand functionalization, and geometry/topography) can be independently controlled to further facilitate cell growth and increase the potency of the source cells. The potency can take the form of directed differentiation of progenitor cells or modulation of the cell’s secretory phenotype. Following expansion and phenotype control, heterogeneity in the cell population can be reduced with various cell sorting technologies. Obtaining a homogeneous population of therapeutic cells will improve the reproducibility of potency outcomes.
2. Biomaterials for Scalable Cell Culture – Increased Cell Yield
Clinical translation of cell therapies demands cost-effective scale-up of adherent cell manufacturing to achieve commercially relevant lot sizes.6,7,11,27−29 A single therapeutic dose is on the order of 109 cells, and therefore, for multiple dose therapies and homogeneity of outcome, lot sizes should ideally be on the order of 1012 cells.6,30 Conventional planar technologies, although modified to contain multiple layers per vessel, have significant cost limitations at the scale needed to produce commercial-sized lots due to a low surface area to volume ratio (SA:V). As such, 3D systems, e.g., hollow-fiber and microcarrier suspension bioreactors, have been designed to maximize the SA:V (upward of a 100-fold increase) and therefore reduce both the footprint and cost. Furthermore, biomaterial design can be leveraged in 3D bioreactor systems to maximize the expansion rate through material properties.11 Of the various 3D systems, microcarrier-based suspension bioreactors provide a promising approach to capitalize on both optimized SA:V culture geometries and biomaterial intervention for improved growth kinetics.
2.1. Biomaterial-Based Microcarriers
As mentioned, microcarriers are used in suspension culture to increase the surface area for cell attachment, therefore maximizing cell growth during each passage. For many cell types, long-term expansion can lead to the development of adverse phenotypes such as increased cellular senescence or loss of multipotency.31,32 Therefore, maximizing the cell density is essential. Not only can microcarriers be engineered from different materials to optimize cell growth kinetics, but also microcarrier material properties can be used to enhance the effects of media additives.11 The minimization of expensive culture media additives provides another avenue for direct manufacturing cost reduction.33,34
2.1.1. Nonporous and Macroporous Microcarriers
Broadly, microcarriers can be classified as either non- or macroporous. Several commercially available nonporous microcarriers exist such as Cytodex (cross-linked dextran matrix) and Synthemax (United States Patent Class VI polystyrene material).27 Mouse embryonic stem cells (ESCs) cultured on dextran microcarriers and human MSCs cultured on polystyrene carriers were shown to have a 200-fold and 1000-fold expansion, respectively, while maintaining their stemness.35,36
Compared to porous microcarriers, nonporous microcarriers allow easier cell seeding and harvesting; however, larger culture times are needed to achieve the same expansion due to limited cell–cell contact early after seeding.27 Like nonporous microcarriers, there are several commercially available macroporous microcarriers (e.g., CultiSpher-S). Mouse ESCs cultured on cross-linked gelatin microcarriers led to a 439-fold expansion in 6 days while maintaining pluripotency.37 Human MSCs cultured on gelatin-cross-linked microcarriers achieved a 1000-fold expansion, similar to Synthemax microcarriers; however, the 1000-fold expansion occurred 10 days faster.38 Macroporous microcarriers provide a larger surface area for cell adherence and migration than nonporous microcarriers. They also provide a structure that can protect against shear-induced phenotypic changes caused by impellers that can be used in suspension bioreactors.27 However, cell harvesting is more challenging and leads to a reduced cell yield. Therefore, while suspension bioreactor design can address the demands for commercial scale-up, the collection of cultured cells for downstream processing should be considered during process development.
2.1.2. Degradable Microcarriers
For therapeutic approaches involving direct injection into a tissue defect, delivery of cells on microcarriers is a suitable option, and detachment of the cells prior to injection can be avoided. However, for cell therapies administered through alternative routes, cell detachment and isolation is necessary. Harvesting cells without inducing cell damage or phenotypic changes however remains a challenge.27 For example, traditional harvesting procedures utilize recombinant proteases to detach cells from their substrate that can lead to the damage of cell membrane proteins and receptors.39 As such, degradable microcarriers that are sensitive to pH, temperature, or enzymes have been engineered to better harvest cells.40 One such system using gelatin methacryloyl (GelMA) illustrated that a 5 min soft-digestion step (0.005% TrypZEAN, 10-fold lower than the recommended working concentration) could completely recover the human iPSC derived MSCs, maintain >95% viability, and preserve the immunomodulatory activity of the cells.41
2.2. Opportunities and Challenges
Although there has been progress in microcarrier design for the commercial scale-up of cell therapies, there is still an opportunity to exploit cell–biomaterial interactions to (1) optimize microcarrier formulations to enhance proliferation for specific cell types, (2) reduce costs associated with expensive media supplements, (3) increase cell expansion through the reduction of cellular senescence, and (4) increase the potency by artificially selecting desirable cell phenotypes. Furthermore, biomaterial advances mostly have been explored in the context of planar systems, but much of this work has not yet been translated to microcarrier systems.
Going forward, microcarrier research in the context of cell therapy manufacturing can capitalize on combinatorial biomaterial screening platforms to select for candidate microcarrier formulations.42−46 For example, hydrogel arrays using surfaces patterned through differential wettability have demonstrated the ability to probe both substrate stiffness and peptide immobilization on human MSC proliferation.42 It was found that 5 kPa polyethylene glycol (PEG)-based hydrogels with 4 mM CRGDS peptide led to the largest increase in human MSC proliferation.42 This same array setup was also used to screen for substrate properties that facilitate the activation of iPSC-derived vascular endothelial cells and illustrate the potential for a single platform technology to optimize microcarrier design for multiple different cells.43
Furthermore, similar technologies can screen for cell behavior other than proliferation, such as secretory activity and differentiation potential, which will be discussed later in this viewpoint. In an approach to reduce the serum content during the manufacture of MSCs, culture on heparin/collagen multilayers was found to support equivalent growth in 2% serum compared to conventional culture in 10% serum.47 Recently, layer-by-layer assembly of heparin/collagen surfaces was partially translated to 3D microcarrier systems.48,49 Similar to the planar study, it was demonstrated that the 2% serum condition supported equivalent growth to conventional culture in 10% serum.48 Similarly, polyethylene glycol coating of polystyrene microcarriers facilitated equivalent growth in serum-free media to growth on uncoated microcarriers in 10% FBS.50,51 Although promising, additional work is required to characterize the effects of surface coating of microcarriers on long-term expansion in a bioreactor.
Tuning the mechanical properties of hydrogel microcarriers was found to influence cellular senescence in MSCs.25 Serial culture on 100 kPa hydrogel surfaces reduced cellular senescence, characterized by β-galactosidase activity, compared to culture on conventional tissue culture plastic.25 Currently, only a few studies have begun to explore the MSC culture on hydrogel microcarriers as a mechanism to reduce cellular senescence. Again, translation of 2D hydrogel design into 3D systems presents a great opportunity to address the limitation of extended cell culture at scale.
Finally, hydrogel microcarrier design can be modulated to drive shifts in a cell’s secretory profile. For example, human MSCs, genetically modified to express the interleukin-1 receptor antagonist (IL-1Ra), cultured on microcarriers softer than commercial polystyrene microcarriers showed a marked increase in IL-1Ra secretion.52 While commercial microcarrier formulations are focused on proliferation, the biomaterials field can further improve microcarrier design by incorporating engineering principles aimed to modulate a cell’s phenotypic state, which will be emphasized in the next section.
3. Biomaterials for Controlling Cell Phenotypic Expression – Increased Cell Potency and Specificity
Cell–biomaterial interactions can be used to direct cells toward favorable functional states, increasing their specificity and therefore their potency.3,5 As mentioned in the Introduction, cell potency is unique to the disease and treatment mechanism and therefore can be controlled many ways such as through the modulation of the secretory behavior of the cells or by directed differentiation. Biomaterial control of both will be further discussed in this section.
Synthetic and natural biomaterials have a wide range of tunable properties that can be used to control cellular perturbations during manufacturing. Several studies have shown that cells are most similar to their in vivo phenotype when they are cultured on biomaterials resembling their native physiological niche.53,54 Therefore, cell niche engineering strategies have been largely motivated by our understanding of spatiotemporal and microenvironmental cues that direct cell behavior during wound healing and tissue remodeling.53 Matrix properties such as stiffness, shape, composition, and topography can be sensed by cells through cell surface receptors and therefore influence the cell state.5,25,55
3.1. Secretome Modulation
During healing, biological processes are often orchestrated by a complex interplay of multiple cytokines and growth factors and therefore would benefit from multimodal therapeutic intervention. Accordingly, exploiting the secretory activity of delivered stem cells has become a common therapeutic approach.56−58 The potency of the delivered stem cells can be enhanced through targeted shifts in their secretome profile using biomaterials.4,5,58,59 Mesenchymal stromal cells (MSCs) are of particular clinical interest due to their high secretory activity aiding in their immunomodulatory, angiogenic, and trophic properties.5,25,60,61 Their versatile secretion profiles have therefore been used to treat a diverse set of diseases and injury such as critical limb ischemia and Crohn’s disease.61,62
3.1.1. Stiffness/Elasticity
With regards to human MSCs, mechanical cues of the substrate are known to yield unique secretory profiles.25,56,61,63−65 One potential mechanism for this observation is that increased matrix stiffness leading to increase cytoskeletal tension can facilitate the translocation of transcription regulators into the nucleus and initiate a cascade of downstream gene expression.61 In one example, MSCs cultured on polyethylene glycol diacrylate (PEGDA) hydrogels functionalized with integrin-engaging RGD peptides demonstrated a significant upregulation of a number of paracrine factors on 30 kPa gels compared to 100 kPa gels.25 Subsequent coculture of the secretome from MSCs cultured on 30 kPa gels with human umbilical cord endothelial cells initiated vessel network formation, demonstrating bioactivity of the secreted factors.25
In addition to the secretion of proregenerative factors, stiffness can be used to direct the immunomodulatory properties of MSCs. In one study, peptoids of different secondary structures were used as hydrogel cross-linkers to tune the stiffness.66 The softer (∼1.5 kPa) hydrogels resulted in increased immunoregulatory cytokine (e.g., IL-6, MCP-1, and M-CSF) secretion and gene expression.66 Similar results were reported for MSC spheroids encapsulated in 5 kPa alginate gels.67
In order to recapitulate all aspects of mechanical properties, viscoelastic gels have been synthesized using a combination of covalent and ionic linkers. For example, only cultures of MSCs on low stiffness (∼1 kPa) and energy dissipating (tan δ > 1) substrates were found to lead to a substantial increase in the secretion of several growth factors (SDF-1a, BDNF, and bNGF) and result in increased hematopoietic stem cell proliferation.68
3.1.2. Ligand Functionalization
The inclusion of ligands to biomaterials was first explored as an approach to promote attachment, but incorporated ligands also have the potential to bias the MSC secretome.3,5,25,61 For example, adding GFOGER, a collagen I mimic that can specifically bind to integrin α5β1, led to increased MSC secretion of cytokines IL-8 and IL-6 that are responsible for osteoprogenitor differentiation and controlled levels of osteoclast resorption.69−71 The delivery of MSCs loaded in GFOGER functionalized hydrogels led to a substantial increase in bone formation in a murine radial segmental defect as characterized by microcomputed tomography (microCT).69
Like stiffness, the surface chemistry of biomaterials can be used to alter the immunomodulatory properties of the MSCs. It was found that both αV/α5 and α2/β1 integrins are essential for MSC immunomodulation when cultured on fibrin and collagen hydrogels, respectively.72
3.1.3. Geometry/Topography
Broadly, increased MSC secretory activity has been shown in both scaffolds and aggregate culture geometries.5,61 Tuning of scaffold porosity and substrate surface patterning can direct MSC clustering and increase secretion activities through cell–cell interactions.5,61 The myogenic potential of the MSC secretome for myoblast differentiation has been shown to be linked to the porosity of the scaffold. MSCs cultured in macroporous scaffolds and nanoporous hydrogels were found to have distinct secretion profiles that consequently result in differential paracrine effects on myoblasts.70 In this study, it was found that the N-cadherin mediated cell–cell interaction was physically blocked in the nanoporous gel and led to a decrease in myogenesis of myoblasts sensing the MSC secretome.70
3.2. Differentiation Control
For other clinical applications, cell potency can be increased through differentiation of stem cells into a more mature and specialized cell type. Although many therapeutic considerations for MSCs involve directing their differentiation,73 the differentiation of iPSCs, another promising cell type for cell therapies, will be the focus of this section. Inducing pluripotency strategically allows for downstream biomaterial directed differentiation into somatic cells for cell transplantation specific to diseases ranging from neurological to cardiovascular.73,74
iPSCs are used as precursors to manufacture both progenitor and differentiated somatic cells in many ongoing clinical trials.3,75−77 A particular promise of iPSCs is their ability to serve as a foundational building block that can continuously expand and then be subsequently differentiated to address the disease of interest.77 Unfortunately, iPSC-based therapies developed with traditional directed differentiation protocols comprised of soluble factors are still limited by unstable lineage stabilization, heterogeneous phenotype presentation, and scalability concerns.76 However, similar to secretome modulation, directed differentiation can be controlled through both biochemical and biomechanical cues and therefore invites the use of engineered biomaterials to address these limitations.
3.2.1. Stiffness/Elasticity
Substrate stiffness can influence the iPSC cell fate. In studies where iPSCs were cultured on poly(vinyl alcohol-co-itaconic acid) (P-IA) hydrogels at stiffnesses greater than 30 kPa, iPSCs were shown to undergo long-term expansion while preserving pluripotency.75 However, lowering the substrate stiffness to match the stiffness of muscle (1–10 kPa) or the brain and lung (<1 kPa) can direct the differentiation of iPSCs.75,76 As such, control of the mechanical environment of iPSCs may provide an inexpensive and easy method to switch between expansion and the directed cell fate.
3.2.2. Ligand Functionalization
Patterned physiochemical cues provide another biomaterial approach to the direct differentiation of iPSCs. More specifically, substrates with tethered inductive moieties have been shown to facilitate neurogenesis, myofibrillogenesis, osteogenesis, and hepatogenesis of iPSCs.76 For example, the surface modification of an alginate and poly(c-glutamic acid) composite scaffold with transactivator of transcription (TAT)-VHL peptides, a peptide known to stimulate neurite outgrowth, directly stimulated seeded iPSCs and facilitated neurogenesis.78
3.2.3. Geometry/Topography
Naturally occurring ECM nanotopography influences the local migration, polarization, and other functions of surrounding cells and as such can be used to drive similar processes in vitro.76,79 For example, polydimethylsiloxane (PDMS) substrates with nanoscale channels (350 nm width) were shown to elevate the expression of neuronal markers.80 The observed increase in neuronal differentiation was, in part, shown to be an effect of the topographical features influencing the contact guidance and alignment of seeded iPSCs.80
3.3. Gene Modification
Genetic editing of cell-based therapies involves the delivery of genetic material such as encoding DNA, mRNA, miRNA, small interfering RNA (siRNA), or small hairpin RNA (shRNA) through either viral or nonviral vectors.3,81 Viral vector limitations involve immunogenicity, mutagenesis, and batch variation; however, they benefit from high transfection efficiency and long-term gene expression.81 In contrast to viral gene vectors, nonviral gene vectors can avoid integration into the host genome after transfection but suffer from lower transfection efficiency and a shorter gene expression duration.81
While efforts for engineering cell potency (improving secretion activities or differentiation) through gene modification are still advancing, we only briefly introduce the approach here to highlight that it is an alternative to direct cell programming. For example, genetically modifying MSCs to yield a conditionally active AKT MSC variant was illustrated to substantially increase vascular endothelial growth factor (VEGF) secretion and highlights the potential of genetic engineering of the secretome.56,82 Furthermore, several studies have shown that lentivirus encoding pro-neuronal and subtype-specific transcription factors can drive human fibroblasts into a multipotent transient state that can eventually differentiate into induced neurons.83,84 Strategic biomaterial design through the use of inorganic materials, lipid/lipid-like materials, and polymeric materials has been a major focus to improve the transfection efficiency of nonviral gene vectors.81
3.4. Opportunities and Challenges
Although there have been advances in using biomaterials to engineer cell specificity, there is still a need to better control phenotype to obtain optimally potent cells. Broadly, biomaterials require further optimization to encourage the secretion of specific factors and the suppression of others or to promote the secretion of transgene-encoded proteins. Additionally, biomaterials need further tuning to increase differentiation rates and specificity as well as the efficiency of vector uptake.
To address these remaining challenges, the incorporation of materials that allow for independent control of matrix stiffness and viscoelasticity into cell manufacturing platforms should be addressed. For example, Adu-Berchie et al. have developed a collagen-based ECM mimic with independently tunable mechanical properties (slow relaxing noncovalent collagen interactions and fast relaxing covalent bonds made from a click reaction between norbornene and tetrazine moieties) to generate functionally distinct T-cell populations.85 However, incorporation into a cell manufacturing platform, and more specifically a microcarrier system, has yet to be studied.
The translation of ligand density effects on cell phenotypes to the cell manufacturing context is limited. Work has been done to directly study the effect of adhesive ligand density on gene expression of MSCs and yielded in the discovery of a highly specific MSC phenotype that can promote hematopoietic stem cell differentiation through cytokine secretion when cultured on alginate modified gels with a 1500 μM GGGRGDSP peptide density compared to 150 μM.86 Moreover, this study also explored how transcriptional programs were affected by combinations of ligand density, material stiffness, and stress–relaxation through RNA-seq.86 This methodology is a promising framework that can elucidate how to engineer distinct biophysical features into cells.
With regard to topography/geometry control of cell phenotype, there are still limitations pertaining to the integration of discoveries from microphysiological system assembly. Microphysiological systems recapitulate human physiology to recreate key biological processes and heavily rely on modeling exact tissue architectures.87 For example, iPSC-derived neural progenitor cells can differentiate to express more synapse promoting proteins such as synaptophysin based on a surface patterned groove.88−90 Integration of surface patterns into microcarriers can lead to improved directed differentiation of phenotype expression.
While general trends between material properties and cell phenotype have been identified, highly specific phenotypic states could benefit from combinatorial design or be obtained from stepwise phenotype shifts. Biomaterial arrays and smart materials that can transition between different material states have the potential to help direct optimized cell potency. Furthermore, better predictive models of how stiffness, ligand density, and topography collectively interact to change cell phenotype can be an outcome of combinatorial studies and shorten the timeline for biomaterial design.86
4. Biomaterials for the Selection of Cells Post Culture – Decreased Heterogeneity
To harness the full potential of cell-based therapeutics, quality attributes such as cell identity, purity, and potency should be rigorously controlled throughout the biomanufacturing process. However, in many cases, it may be impossible to achieve full homogeneity of the product without some type of postexpansion selection and filtering. Currently many high-throughput devices have been developed that can separate cells based on size and deformability.91,92 However, if the goal is to isolate cells based on the expression of characteristic surface markers, new separation approaches need to be further developed.
One such approach would be the use of an affinity-based separations technique that can filter cells based on surface markers characteristic of the desirable cell phenotype.93 Cell-binding ligands have been immobilized onto solid substrates, polymer carriers, as well as magnetic particles (MACS) or fluorescent markers (FACS) that can be separated by an electromagnetic field.93 More recently, affinity ligands have been displayed on the channels of microfluidic devices.93,94
Although iPSC sorting based on the surface markers can increase homogeneity and potency, studies have shown that there is not a correlation between surface markers and potency for MSCs.95−97 As such, isolation of potent MSC subtypes is a major limitation. Material advances, such as polymers that change conformation in the presence of a particular secreted protein, might capture desirable MSC phenotypes. However, single cell sensing in a high-throughput manner remains a major technical challenge.
Currently, cell separation techniques post scale-up are not often implemented, and instead, cells classified as minimally or nonpotent are checked for potential deleterious effects. Even if these cells are viewed as therapeutically inert, their inclusion in therapies can reduce the number of potent cells per dose, thus affecting clinical efficacy. The development of cell sorting technologies based on direct potency metrics, therefore, should be a major focus of the field.
5. Conclusions and Future Directions
To date, there have been numerous biomaterial interventions that can increase cell potency, scale-up therapeutic cell manufacturing, and reduce cell heterogeneity (Table 1). However, the integration of all of these principles into a single manufacturing pipeline has remained a challenge. Furthermore, the reproducibility and predictability of cell-based therapies have severely impacted clinical translation. In order to advance reproducibility, both heterogeneity within the initial cell population and heterogeneity that compounds during processing steps should be addressed with more sophisticated cell isolation techniques.
Table 1. Biomaterials for the Manufacturing of Adherent Cells.
The integration of biomaterial cell potency control and scale-up via microcarriers provides a great opportunity to minimize costs and yield more reproducible and potent cells. However, as mentioned above, there can be trade-offs between biomaterial-induced cell expansion and biomaterial-induced potency control. Current integration of these techniques would require sequential operation, where a stiffer microcarrier could be used for increasing cell proliferation and a subsequent culture on softer hydrogel microcarriers could be used for differentiation or secretome control. As an effort to bridge these processing steps, the incorporation of stimuli-responsive hydrogel materials that can adjust their mechanical properties in response to an introduced stimuli could be a promising approach aimed to maximize expansion and potency control within a single bioreactor system in a continuous manner.98−102
Even with the many advances in cell potency control, one of the main limitations of translation is the mismatch between theoretical and perceived clinical potency. Inconsistent clinical potency can arise from many factors such as an immunogenic response and clearance of the delivered cells or fast phenotypic shifts in the delivered cells due to the extracellular environment at the site of transplantation.11,103 Poor potency can be linked to uncharacterized modes of action and a lack of predictability. As such, a concerted effort to integrate biomaterial recapitulation of the delivery site as a method of screening for in vivo efficacy can be used as a feedback/optimization tool to better engineer highly potency cells. Moreover, selecting for phenotypes that require large activation transition states to enter into a new phenotype may allow for maintained and predictable potency post transplantation.
Finally, biomaterial advances will need to be interfaced with sensors and/or imaging platforms that can detect subcellular markers for desired phenotypes. As a potential first step, noninvasive imaging could be integrated with culture platforms to characterize and/or sort cells based on morphology changes correlated with unique cell phenotypes.104,105 While new biomaterials technologies are being developed to screen and filter cells as a method of quality control, essential for clinical translation, limitations exist with high-throughput operation and single cell sensing, and both should be a major consideration with regard to future development of these technologies.
Overall, the use of biomaterials in cell manufacturing provides an innovative and exciting means to directly interface with and thus control cell behavior, with the eventual goal of manufacturing highly potent cell therapies that can treat a wide range of diseases.
Author Contributions
# R.C.M. and J.S.T. contributed equally. The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. CRediT: Ryan Cree Miller conceptualization, writing - original draft; Johnna S. Temenoff conceptualization, writing - review & editing.
This research was supported by the National Science Foundation (Grant no. 1648035, Engineering Research Center for Cell Manufacturing Technologies), as well as by the Marcus Foundation, the Georgia Research Alliance, and Georgia Tech Foundation through their support of the Marcus Center for Cell Characterization and Manufacturing (MC3M) at Georgia Tech.
The authors declare no competing financial interest.
References
- Kirouac D. C.; Zandstra P. W. The Systematic Production of Cells for Cell Therapies. Cell Stem Cell 2008, 3 (4), 369–381. 10.1016/j.stem.2008.09.001. [DOI] [PubMed] [Google Scholar]
- Wang L. L. W.; Janes M. E.; Kumbhojkar N.; Kapate N.; Clegg J. R.; Prakash S.; Heavey M. K.; Zhao Z.; Anselmo A. C.; Mitragotri S. Cell therapies in the clinic. Bioeng Transl Med. 2021, 6 (2), e10214 10.1002/btm2.10214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abdeen A. A.; Saha K. Manufacturing Cell Therapies Using Engineered Biomaterials. Trends Biotechnol 2017, 35 (10), 971–982. 10.1016/j.tibtech.2017.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherian D. S.; Bhuvan T.; Meagher L.; Heng T. S. P. Biological Considerations in Scaling Up Therapeutic Cell Manufacturing. Front Pharmacol 2020, 11, 654. 10.3389/fphar.2020.00654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doron G.; Temenoff J. S. Culture Substrates for Improved Manufacture of Mesenchymal Stromal Cell Therapies. Adv. Healthc Mater. 2021, 10 (15), 2100016 10.1002/adhm.202100016. [DOI] [PubMed] [Google Scholar]
- Roh K. H.; Nerem R. M.; Roy K. Biomanufacturing of Therapeutic Cells: State of the Art, Current Challenges, and Future Perspectives. Annu. Rev. Chem. Biomol 2016, 7, 455–478. 10.1146/annurev-chembioeng-080615-033559. [DOI] [PubMed] [Google Scholar]
- Aijaz A.; Li M.; Smith D.; Khong D.; LeBlon C.; Fenton O. S.; Olabisi R. M.; Libutti S.; Tischfield J.; Maus M. V.; et al. Biomanufacturing for clinically advanced cell therapies. Nat. Biomed Eng. 2018, 2 (6), 362–376. 10.1038/s41551-018-0246-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dwarshuis N. J.; Parratt K.; Santiago-Miranda A.; Roy K. Cells as advanced therapeutics: State-of-the-art, challenges, and opportunities in large scale biomanufacturing of high-quality cells for adoptive immunotherapies. Adv. Drug Deliver Rev. 2017, 114, 222–239. 10.1016/j.addr.2017.06.005. [DOI] [PubMed] [Google Scholar]
- Pigeau G. M.; Csaszar E.; Dulgar-Tulloch A. Commercial Scale Manufacturing of Allogeneic Cell Therapy. Front Med-Lausanne 2018, 5, 233. 10.3389/fmed.2018.00233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fuzeta M. D.; Branco A. D. D.; Fernandes-Platzgummer A.; da Silva C. L.; Cabral J. M. S. Addressing the Manufacturing Challenges of Cell-Based Therapies. Adv. Biochem Eng. Biotechnol. 2020, 171, 225–278. 10.1007/10_2019_118. [DOI] [PubMed] [Google Scholar]
- Bashor C. J.; Hilton I. B.; Bandukwala H.; Smith D. M.; Veiseh O. Engineering the next generation of cell-based therapeutics. Nat. Rev. Drug Discov 2022, 21 (9), 655–675. 10.1038/s41573-022-00476-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rafiq Q. A.; Coopman K.; Nienow A. W.; Hewitt C. J. Systematic microcarrier screening and agitated culture conditions improves human mesenchymal stem cell yield in bioreactors. Biotechnol J. 2016, 11 (4), 473–486. 10.1002/biot.201400862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowdell M. W.; Weil B. Bringing function to the forefront of cell therapy: how do we demonstrate potency?. Front Immunol 2023, 14, 1226841 10.3389/fimmu.2023.1226841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robb K. P.; Fitzgerald J. C.; Barry F.; Viswanathan S. Mesenchymal stromal cell therapy: progress in manufacturing and assessments of potency. Cytotherapy 2019, 21 (3), 289–306. 10.1016/j.jcyt.2018.10.014. [DOI] [PubMed] [Google Scholar]
- Kim E.; Tae G. Direct reprogramming and biomaterials for controlling cell fate. Biomater Res. 2016, 20 (39), 2055–7124. 10.1186/s40824-016-0086-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H. F.; Yang Y. C.; Liu J. D.; Qian L. Direct cell reprogramming: approaches, mechanisms and progress. Nat. Rev. Mol. Cell Bio 2021, 22 (6), 410–424. 10.1038/s41580-021-00335-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chinnadurai R.; Viswanathan S.; Moll G. Editorial: Next generation MSC therapy manufacturing, potency and mechanism of action analysis. Frontiers in Immunology 2023, 14, 1192636 10.3389/fimmu.2023.1192636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castilla-Casadiego D. A.; Reyes-Ramos A. M.; Domenech M.; Almodovar J. Effects of Physical, Chemical, and Biological Stimulus on h-MSC Expansion and Their Functional Characteristics. Ann. Biomed Eng. 2020, 48 (2), 519–535. 10.1007/s10439-019-02400-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J.; Du Y. Y.; Deng H. K. Direct Lineage Reprogramming: Strategies, Mechanisms, and Applications. Cell Stem Cell 2015, 16 (2), 119–134. 10.1016/j.stem.2015.01.013. [DOI] [PubMed] [Google Scholar]
- Kitada T.; DiAndreth B.; Teague B.; Weiss R. Programming gene and engineered-cell therapies with synthetic biology. Science 2018, 359 (6376), 651. 10.1126/science.aad1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marklein R. A.; Lam J.; Guvendiren M.; Sung K. E.; Bauer S. R. Functionally-Relevant Morphological Profiling: A Tool to Assess Cellular Heterogeneity. Trends Biotechnol 2018, 36 (1), 105–118. 10.1016/j.tibtech.2017.10.007. [DOI] [PubMed] [Google Scholar]
- Phinney D. G. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. J. Cell Biochem 2012, 113 (9), 2806–2812. 10.1002/jcb.24166. [DOI] [PubMed] [Google Scholar]
- Etzrodt M.; Endele M.; Schroeder T. Quantitative Single-Cell Approaches to Stem Cell Research. Cell Stem Cell 2014, 15 (5), 546–558. 10.1016/j.stem.2014.10.015. [DOI] [PubMed] [Google Scholar]
- Childs B. G.; Durik M.; Baker D. J.; van Deursen J. M. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 2015, 21 (12), 1424–1435. 10.1038/nm.4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogle M. E.; Doron G.; Levy M. J.; Temenoff J. S. Hydrogel Culture Surface Stiffness Modulates Mesenchymal Stromal Cell Secretome and Alters Senescence. Tissue Eng. Pt A 2020, 26 (23–24), 1259–1271. 10.1089/ten.tea.2020.0030. [DOI] [PubMed] [Google Scholar]
- Miller R. C.; Lee J. H.; Kim Y. J.; Han H. S.; Kong H. Y. J. In-Drop Thermal Cycling of Microcrystal Assembly for Senescence Control (MASC) with Minimal Variation in Efficacy. Adv. Funct Mater. 2023, 33 (37), 2302232 10.1002/adfm.202302232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen R. Y.; Li L.; Feng L.; Luo Y. X.; Xu M. G.; Leong K. W.; Yao R. Biomaterial-assisted scalable cell production for cell therapy. Biomaterials 2020, 230, 119627 10.1016/j.biomaterials.2019.119627. [DOI] [PubMed] [Google Scholar]
- Naing M. W.; Williams D. J. Three-dimensional culture and bioreactors for cellular therapies. Cytotherapy 2011, 13 (4), 391–399. 10.3109/14653249.2011.556352. [DOI] [PubMed] [Google Scholar]
- Ge C.; Selvaganapathy P. R.; Geng F. Advancing our understanding of bioreactors for industrial-sized cell culture: health care and cellular agriculture implications. Am. J. Physiol-Cell Ph 2023, 325 (3), C580–C591. 10.1152/ajpcell.00408.2022. [DOI] [PubMed] [Google Scholar]
- Campbell A.; Brieva T.; Raviv L.; Rowley J.; Niss K.; Brandwein H.; Oh S.; Karnieli O. Concise Review: Process Development Considerations for Cell Therapy. Stem Cells Transl Med. 2015, 4 (10), 1155–1163. 10.5966/sctm.2014-0294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miclau K.; Hambright W. S.; Huard J.; Stoddart M. J.; Bahney C. S. Cellular expansion of MSCs: Shifting the regenerative potential. Aging Cell 2023, 22 (1), e13759 10.1111/acel.13759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu Y. J.; Li T.; Ding Y. L.; Sun L. X.; Tu T.; Zhu W.; Hu J. B.; Sun X. C. Changes in mesenchymal stem cells following long-term culture. Mol. Med. Rep 2016, 13 (6), 5207–5215. 10.3892/mmr.2016.5169. [DOI] [PubMed] [Google Scholar]
- Karnieli O.; Friedner O. M.; Allickson J. G.; Zhang N.; Jung S.; Fiorentini D.; Abraham E.; Eaker S. S.; Yong T. K.; Chan A.; et al. A consensus introduction to serum replacements and serum-free media for cellular therapies. Cytotherapy 2017, 19 (2), 155–169. 10.1016/j.jcyt.2016.11.011. [DOI] [PubMed] [Google Scholar]
- ten Ham R. M.T.; Nievaart J. C.; Hoekman J.; Cooper R. S.; Frederix G. W.J.; Leufkens H. G.M.; Klungel O. H.; Ovelgonne H.; Hoefnagel M. H.N.; Turner M. L.; Mountford J. C. Estimation of manufacturing development costs of cell-based therapies: a feasibility study. Cytotherapy 2021, 23 (8), 730–739. 10.1016/j.jcyt.2020.12.014. [DOI] [PubMed] [Google Scholar]
- Alfred R.; Radford J.; Fan J.; Boon K.; Krawetz R.; Rancourt D.; Kallos M. S. Efficient Suspension Bioreactor Expansion of Murine Embryonic Stem Cells on Microcarriers in Serum-Free Medium. Biotechnol. Prog. 2011, 27 (3), 811–823. 10.1002/btpr.591. [DOI] [PubMed] [Google Scholar]
- Hervy M.; Weber J. L.; Pecheul M.; Dolley-Sonneville P.; Henry D.; Zhou Y.; Melkoumian Z. Long Term Expansion of Bone Marrow-Derived hMSCs on Novel Synthetic Microcarriers in Xeno-Free, Defined Conditions. PLoS One 2014, 9 (3), e92120 10.1371/journal.pone.0092120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melero-Martin J. M.; Dowling M. A.; Smith M.; Al-Rubeai M. Expansion of chondroprogenitor cells on macroporous microcarriers as an alternative to conventional monolayer systems. Biomaterials 2006, 27 (15), 2970–2979. 10.1016/j.biomaterials.2006.01.023. [DOI] [PubMed] [Google Scholar]
- Yuan Y. F.; Kallos M. S.; Hunter C.; Sen A. Improved expansion of human bone marrow-derived mesenchymal stem cells in microcarrier-based suspension culture. J. Tissue Eng. Regen M 2014, 8 (3), 210–225. 10.1002/term.1515. [DOI] [PubMed] [Google Scholar]
- Yang H. S.; Jeon O.; Bhang S. H.; Lee S. H.; Kim B. S. Suspension Culture of Mammalian Cells Using Thermosensitive Microcarrier That Allows Cell Detachment Without Proteolytic Enzyme Treatment. Cell Transplant 2010, 19 (9), 1123–1132. 10.3727/096368910X516664. [DOI] [PubMed] [Google Scholar]
- Derakhti S.; Safiabadi-Tali S. H.; Amoabediny G.; Sheikhpour M. Attachment and detachment strategies in microcarrier-based cell culture technology: A comprehensive review. Mat Sci. Eng. C-Mater. 2019, 103, 109782 10.1016/j.msec.2019.109782. [DOI] [PubMed] [Google Scholar]
- Rogers R. E.; Haskell A.; White B. P.; Dalal S.; Lopez M.; Tahan D.; Pan S. M.; Kaur G.; Kim H.; Barreda H.; et al. A scalable system for generation of mesenchymal stem cells derived from induced pluripotent cells employing bioreactors and degradable microcarriers. Stem Cell Transl Med. 2021, 10 (12), 1650–1665. 10.1002/sctm.21-0151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le N. N. T.; Zorn S.; Schmitt S. K.; Gopalan P.; Murphy W. L. Hydrogel arrays formed via differential wettability patterning enable combinatorial screening of stem cell behavior. Acta Biomater 2016, 34, 93–103. 10.1016/j.actbio.2015.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belair D. G.; Schwartz M. P.; Knudsen T.; Murphy W. L. Human iPSC-derived endothelial cell sprouting assay in synthetic hydrogel arrays. Acta Biomater 2016, 39, 12–24. 10.1016/j.actbio.2016.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jongpaiboonkit L.; King W. J.; Lyons G. E.; Paguirigan A. L.; Warrick J. W.; Beebe D. J.; Murphy W. L. An adaptable hydrogel array format for 3-dimensional cell culture and analysis. Biomaterials 2008, 29 (23), 3346–3356. 10.1016/j.biomaterials.2008.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lei R. X.; Akins E. A.; Wong K. C. Y.; Repina N. A.; Wolf K. J.; Dempsey G. E.; Schaffer D. V.; Stahl A.; Kumar S. Multiwell Combinatorial Hydrogel Array for High-Throughput Analysis of Cell-ECM Interactions. Acs Biomater Sci. Eng. 2021, 7 (6), 2453–2465. 10.1021/acsbiomaterials.1c00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo J.; Shin J. Y.; Leijten J.; Jeon O.; Camci-Unal G.; Dikina A. D.; Brinegar K.; Ghaemmaghami A. M.; Alsberg E.; Khademhosseini A. High-throughput approaches for screening and analysis of cell behaviors. Biomaterials 2018, 153, 85–101. 10.1016/j.biomaterials.2017.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cifuentes S. J.; Priyadarshani P.; Castilla-Casadiego D. A.; Mortensen L. J.; Almodóvar J.; Domenech M. Heparin/collagen surface coatings modulate the growth, secretome, and morphology of human mesenchymal stromal cell response to interferon-gamma. J. Biomed Mater. Res. A 2021, 109 (6), 951–965. 10.1002/jbm.a.37085. [DOI] [PubMed] [Google Scholar]
- Cifuentes S. J.; Theran-Suarez N. A.; Rivera-Crespo C.; Velez-Roman L.; Thacker B.; Glass C.; Domenech M. Heparan sulfate-collagen surface multilayers support serum-free microcarrier culture of mesenchymal stem cells. Acs Biomater Sci. Eng. 2024, 10 (9), 5739–5751. 10.1021/acsbiomaterials.4c01008. [DOI] [PubMed] [Google Scholar]
- Timsina H.; McTyer J.; Rao R. R.; Almodovar J. A comparative evaluation of layer-by-layer assembly techniques for surface modification of microcarriers used in human mesenchymal stromal cell manufacturing. Biotechnol J. 2022, 17 (8), 2100605 10.1002/biot.202100605. [DOI] [PubMed] [Google Scholar]
- Dias A. D.; Elicson J. M.; Murphy W. L. Microcarriers with synthetic hydrogel surfaces for stem cell expansion. Adv. Healthc Mater. 2017, 6 (16), 1700072 10.1002/adhm.201700072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin S.; Cao Y. Hydrogels for large-scale expansion of stem cells. Acta Biomater 2021, 128, 1–20. 10.1016/j.actbio.2021.03.026. [DOI] [PubMed] [Google Scholar]
- Doron G.; Wood L. B.; Guldberg R. E.; Temenoff J. S. Poly(ethylene glycol)-Based Hydrogel Microcarriers Alter Secretory Activity of Genetically Modified Mesenchymal Stromal Cells. Acs Biomater Sci. Eng. 2023, 9 (11), 6282–6292. 10.1021/acsbiomaterials.3c00954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shakiba N.; Jones R. D.; Weiss R.; Del Vecchio D. Context-aware synthetic biology by controller design: Engineering the mammalian cell. Cell Syst 2021, 12 (6), 561–592. 10.1016/j.cels.2021.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tewary M.; Shakiba N.; Zandstra P. W. Stem cell bioengineering: building from stem cell biology. Nat. Rev. Genet 2018, 19 (10), 595–614. 10.1038/s41576-018-0040-z. [DOI] [PubMed] [Google Scholar]
- Wang L.; Wang C. Y.; Wu S.; Fan Y. B.; Li X. M. Influence of the mechanical properties of biomaterials on degradability, cell behaviors and signaling pathways: current progress and challenges. Biomater Sci-Uk 2020, 8 (10), 2714–2733. 10.1039/D0BM00269K. [DOI] [PubMed] [Google Scholar]
- Daneshmandi L.; Shah S.; Jafari T.; Bhattacharjee M.; Momah D.; Saveh-Shemshaki N.; Lo K. W. H.; Laurencin C. T. Emergence of the Stem Cell Secretome in Regenerative Engineering. Trends Biotechnol 2020, 38 (12), 1373–1384. 10.1016/j.tibtech.2020.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kol S.; Ley D.; Wulff L. T.; Decker M.; Arnsdorf J.; Schoffelen S.; Hansen A. H.; Jensen T. L.; Gutierrez J. M.; Chiang A. W. T.; et al. Multiplex secretome engineering enhances recombinant protein production and purity. Nat. Commun. 2020, 11 (1), 1908. 10.1038/s41467-020-15866-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu K. Z.; Veenendaal T.; Wiendels M.; Ruiz-Zapata A. M.; van Laar J.; Kyranas R.; Enting H.; van Cranenbroek B.; Koenen H. J. P. M.; Mihaila S. M.; et al. Synthetic Extracellular Matrices as a Toolbox to Tune Stem Cell Secretome. Acs Appl. Mater. Inter 2020, 12 (51), 56723–56730. 10.1021/acsami.0c16208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drzeniek N. M.; Kahwaji N.; Schlickeiser S.; Reinke P.; Geissler S.; Volk H. D.; Gossen M. Immuno-engineered mRNA combined with cell adhesive niche for synergistic modulation of the MSC secretome. Biomaterials 2023, 294, 121971 10.1016/j.biomaterials.2022.121971. [DOI] [PubMed] [Google Scholar]
- Vilar A.; Hodgson-Garms M.; Kusuma G. D.; Donderwinkel I.; Carthew J.; Tan J. L.; Lim R.; Frith J. E. Substrate mechanical properties bias MSC paracrine activity and therapeutic potential. Acta Biomater 2023, 168, 144–158. 10.1016/j.actbio.2023.06.041. [DOI] [PubMed] [Google Scholar]
- Wechsler M. E.; Rao V. V.; Borelli A. N.; Anseth K. S. Engineering the MSC Secretome: A Hydrogel Focused Approach. Adv. Healthc Mater. 2021, 10 (7), 2001948 10.1002/adhm.202001948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levy O.; Kuai R.; Siren E. M. J.; Bhere D.; Milton Y.; Nissar N.; De Biasio M.; Heinelt M.; Reeve B.; Abdi R.; et al. Shattering barriers toward clinically meaningful MSC therapies. Sci. Adv. 2020, 6 (30), eaba6884 10.1126/sciadv.aba6884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ranganath S. H.; Levy O.; Inamdar M. S.; Karp J. M. Harnessing the Mesenchymal Stem Cell Secretome for the Treatment of Cardiovascular Disease. Cell Stem Cell 2012, 10 (3), 244–258. 10.1016/j.stem.2012.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang C.; Yan J.; Yao Z. C.; Zhang C.; Li X. W.; Mao H. Q. Effects of Mesenchymal Stem Cell-Derived Paracrine Signals and Their Delivery Strategies. Adv. Healthc Mater. 2021, 10 (7), 2001689 10.1002/adhm.202001689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doron G.; Wood L. B.; Guldberg R. E.; Temenoff J. S. Poly(ethylene glycol)-Based Hydrogel Microcarriers Alter Secretory Activity of Genetically Modified Mesenchymal Stromal Cells. Acs Biomater Sci. Eng. 2023, 9 (11), 6282–6292. 10.1021/acsbiomaterials.3c00954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu F. D.; Pishesha N.; Poon Z.; Kaushik T.; Van Vliet K. J. Material Viscoelastic Properties Modulate the Mesenchymal Stem Cell Secretome for Applications in Hematopoietic Recovery. Acs Biomater Sci. Eng. 2017, 3 (12), 3292–3306. 10.1021/acsbiomaterials.7b00644. [DOI] [PubMed] [Google Scholar]
- Clark A. Y.; Martin K. E.; García J. R.; Johnson C. T.; Theriault H. S.; Han W. M.; Zhou D. W.; Botchwey E. A.; García A. J. Integrin-specific hydrogels modulate transplanted human bone marrow-derived mesenchymal stem cell survival, engraftment, and reparative activities. Nat. Commun. 2020, 11 (1), 114. 10.1038/s41467-019-14000-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith J. K. IL-6 and the dysregulation of immune, bone, muscle, and metabolic homeostasis during spaceflight. Npj Microgravity 2018, 4, 24. 10.1038/s41526-018-0057-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitaura H.; Marahleh A.; Ohori F.; Noguchi T.; Shen W. R.; Qi J. W.; Nara Y.; Pramusita A.; Kinjo R.; Mizoguchi I. Osteocyte-Related Cytokines Regulate Osteoclast Formation and Bone Resorption. Int. J. Mol. Sci. 2020, 21 (14), 5169. 10.3390/ijms21145169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qazi T. H.; Mooney D. J.; Duda G. N.; Geissler S. Biomaterials that promote cell-cell interactions enhance the paracrine function of MSCs. Biomaterials 2017, 140, 103–114. 10.1016/j.biomaterials.2017.06.019. [DOI] [PubMed] [Google Scholar]
- Mao A. S.; Shin J. W.; Mooney D. J. Effects of substrate stiffness and cell-cell contact on mesenchymal stem cell differentiation. Biomaterials 2016, 98, 184–191. 10.1016/j.biomaterials.2016.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwee B. J.; Lam J.; Akue A.; KuKuruga M. A.; Zhang K. Y.; Gu L.; Sung K. E. Functional heterogeneity of IFN-γ-licensed mesenchymal stromal cell immunosuppressive capacity on biomaterials. P Natl. Acad. Sci. USA 2021, 118 (35), e2105972118 10.1073/pnas.2105972118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hockemeyer D.; Jaenisch R. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 2016, 18 (5), 573–586. 10.1016/j.stem.2016.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trounson A.; DeWitt N. D. Pluripotent stem cells progressing to the clinic. Nat. Rev. Mol. Cell Bio 2016, 17 (3), 194–200. 10.1038/nrm.2016.10. [DOI] [PubMed] [Google Scholar]
- Poorna M. R.; Jayakumar R.; Chen J. P.; Mony U. Hydrogels: A potential platform for induced pluripotent stem cell culture and differentiation. Colloid Surface B 2021, 207, 111991 10.1016/j.colsurfb.2021.111991. [DOI] [PubMed] [Google Scholar]
- Tong Z.; Solanki A.; Hamilos A.; Levy O.; Wen K.; Yin X.; Karp J. M. Application of biomaterials to advance induced pluripotent stem cell research and therapy. EMBO J. 2015, 34 (8), 987–1008. 10.15252/embj.201490756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madrid M.; Sumen C.; Aivio S.; Saklayen N. Autologous Induced Pluripotent Stem Cell-Based Cell Therapies: Promise, Progress, and Challenges. Curr. Protoc 2021, 1 (3), e88 10.1002/cpz1.88. [DOI] [PubMed] [Google Scholar]
- Kuo Y. C.; Chung C. Y. TATVHL peptide-grafted alginate/poly(γ-glutamic acid) scaffolds with inverted colloidal crystal topology for neuronal differentiation of iPS cells. Biomaterials 2012, 33 (35), 8955–8966. 10.1016/j.biomaterials.2012.08.073. [DOI] [PubMed] [Google Scholar]
- Kuo Y. C.; Rajesh R. Guided differentiation and tissue regeneration of induced pluripotent stem cells using biomaterials. J. Taiwan Inst Chem. E 2017, 77, 41–53. 10.1016/j.jtice.2017.04.043. [DOI] [Google Scholar]
- Pan F.; Zhang M.; Wu G. M.; Lai Y. K.; Greber B.; Schöler H. R.; Chi L. F. Topographic effect on human induced pluripotent stem cells differentiation towards neuronal lineage. Biomaterials 2013, 34 (33), 8131–8139. 10.1016/j.biomaterials.2013.07.025. [DOI] [PubMed] [Google Scholar]
- Uludag H.; Ubeda A.; Ansari A. At the Intersection of Biomaterials and Gene Therapy: Progress in Non-viral Delivery of Nucleic Acids. Front Bioeng Biotech 2019, 7, 131. 10.3389/fbioe.2019.00131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song S. H.; Lee M. O.; Lee J. S.; Jeong H. C.; Kim H. G.; Kim W. S.; Hur M.; Cha H. J. Genetic modification of human adipose-derived stem cells for promoting wound healing. J. Dermatol Sci. 2012, 66 (2), 98–107. 10.1016/j.jdermsci.2012.02.010. [DOI] [PubMed] [Google Scholar]
- Mertens J.; Marchetto M. C.; Bardy C.; Gage F. H. Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience. Nat. Rev. Neurosci 2016, 17 (7), 424–437. 10.1038/nrn.2016.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Son E. Y.; Ichida J. K.; Wainger B. J.; Toma J. S.; Rafuse V. F.; Woolf C. J.; Eggan K. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell 2011, 9 (3), 205–218. 10.1016/j.stem.2011.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adu-Berchie K.; Liu Y. T.; Zhang D. K. Y.; Freedman B. R.; Brockman J. M.; Vining K. H.; Nerger B. A.; Garmilla A.; Mooney D. J. Generation of functionally distinct T-cell populations by altering the viscoelasticity of their extracellular matrix. Nat. Biomed Eng. 2023, 7 (11), 1374. 10.1038/s41551-023-01052-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darnell M.; O’Neil A.; Mao A.; Gu L.; Rubin L. L.; Mooney D. J. Material microenvironmental properties couple to induce distinct transcriptional programs in mammalian stem cells. P Natl. Acad. Sci. USA 2018, 115 (36), E8368–E8377. 10.1073/pnas.1802568115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roth A.; Human microphysiological systems for drug development. Science 2021, 373 (6561), 1304–1306. 10.1126/science.abc3734. [DOI] [PubMed] [Google Scholar]
- Emperador Melero J.; Nadadhur A. G.; Schut D.; Weering J. V.; Heine V. M.; Toonen R. F.; Verhage M. Differential Maturation of the Two Regulated Secretory Pathways in Human iPSC-Derived Neurons. Stem Cell Rep 2017, 8 (3), 659–672. 10.1016/j.stemcr.2017.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burbulla L. F.; Beaumont K. G.; Mrksich M.; Krainc D. Micropatterning Facilitates the Long-Term Growth and Analysis of iPSC-Derived Individual Human Neurons and Neuronal Networks. Adv. Healthc Mater. 2016, 5 (15), 1894–1903. 10.1002/adhm.201500900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Autar K.; Guo X. F.; Rumsey J. W.; Long C. J.; Akanda N.; Jackson M.; Narasimhan N. S.; Caneus J.; Morgan D.; Hickman J. J. A functional hiPSC-cortical neuron differentiation and maturation model and its application to neurological disorders. Stem Cell Rep 2022, 17 (1), 96–109. 10.1016/j.stemcr.2021.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun L. J.; Yang W. U.; Cai S. X.; Chen Y. B.; Chu H. H.; Yu H. B.; Wang Y. C.; Liu L. Q. Recent advances in microfluidic technologies for separation of biological cells. Biomed Microdevices 2020, 22 (3), 55. 10.1007/s10544-020-00510-7. [DOI] [PubMed] [Google Scholar]
- Nasiri R.; Shamloo A.; Ahadian S.; Amirifar L.; Akbari J.; Goudie M. J.; Lee K.; Ashammakhi N.; Dokmeci M. R.; Di Carlo D.; et al. Microfluidic-Based Approaches in Targeted Cell/Particle Separation Based on Physical Properties: Fundamentals and Applications. Small 2020, 16 (29), 2000171 10.1002/smll.202000171. [DOI] [PubMed] [Google Scholar]
- Bacon K.; Lavoie A.; Rao B. M.; Daniele M.; Menegatti S. Past, Present, and Future of Affinity-based Cell Separation Technologies. Acta Biomater 2020, 112, 29–51. 10.1016/j.actbio.2020.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cowell T. W.; Valera E.; Jankelow A.; Park J.; Schrader A. W.; Ding R.; Berger J.; Bashir R.; Han H. S. Rapid, multiplexed detection of biomolecules using electrically distinct hydrogel beads. Lab Chip 2020, 20 (13), 2274–2283. 10.1039/D0LC00243G. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naji A.; Suganuma N.; Espagnolle N.; Yagyu K.-i.; Baba N.; Sensebe L.; Deschaseaux F. Rationale for Determining the Functional Potency of Mesenchymal Stem Cells in Preventing Regulated Cell Death for Therapeutic Use. Stem Cell Transl Med. 2017, 6 (3), 713–719. 10.5966/sctm.2016-0289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samsonraj R. M.; Rai B.; Sathiyanathan P.; Puan K. J.; Rötzschke O.; Hui J. H.; Raghunath M.; Stanton L. W.; Nurcombe V.; Cool S. M. Establishing Criteria for Human Mesenchymal Stem Cell Potency. Stem Cells 2015, 33 (6), 1878–1891. 10.1002/stem.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee W. C.; Shi H.; Poon Z. Y.; Nyan L. M.; Kaushik T.; Shivashankar G. V.; Chan J. K. Y.; Lim C. T.; Han J.; Van Vliet K. J. Multivariate biophysical markers predictive of mesenchymal stromal cell multipotency. P Natl. Acad. Sci. USA 2014, 111 (42), E4409–E4418. 10.1073/pnas.1402306111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doron G.; Pearson J. J.; Guldberg R. E.; Temenoff J. S. Development and characterization of Factor Xa-responsive materials for applications in cell culture and biologics delivery. J. Biomed Mater. Res. A 2023, 111 (5), 634–643. 10.1002/jbm.a.37513. [DOI] [PubMed] [Google Scholar]
- Augustine R.; Kalva N.; Kim H. A.; Zhang Y.; Kim I. pH-Responsive Polypeptide-Based Smart Nano-Carriers for Theranostic Applications. Molecules 2019, 24 (16), 2961. 10.3390/molecules24162961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doberenz F.; Zeng K.; Willems C.; Zhang K.; Groth T. Thermoresponsive polymers and their biomedical application in tissue engineering - a review. J. Mater. Chem. B 2020, 8 (4), 607–628. 10.1039/C9TB02052G. [DOI] [PubMed] [Google Scholar]
- Katz J. S.; Burdick J. A. Light-Responsive Biomaterials: Development and Applications. Macromol. Biosci 2010, 10 (4), 339–348. 10.1002/mabi.200900297. [DOI] [PubMed] [Google Scholar]
- Hu Q. Y.; Katti P. S.; Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale 2014, 6 (21), 12273–12286. 10.1039/C4NR04249B. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou T.; Yuan Z. N.; Weng J. Y.; Pei D. Q.; Du X.; He C.; Lai P. L. Challenges and advances in clinical applications of mesenchymal stromal cells. J. Hematol Oncol 2021, 14 (1), 24. 10.1186/s13045-021-01037-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandel M. E.; Kim E.; Lee Y. J.; Tracy G.; Chung H. J.; Popescu G. Multiscale Assay of Unlabeled Neurite Dynamics Using Phase Imaging with Computational Specificity. Acs Sensors 2021, 6 (5), 1864–1874. 10.1021/acssensors.1c00100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kandel M. E.; He Y. C. R.; Lee Y. J.; Chen T. H. Y.; Sullivan K. M.; Aydin O.; Saif M. T. A.; Kong H.; Sobh N.; Popescu G. Phase imaging with computational specificity (PICS) for measuring dry mass changes in sub-cellular compartments. Nat. Commun. 2020, 11 (1), 6256. 10.1038/s41467-020-20062-x. [DOI] [PMC free article] [PubMed] [Google Scholar]


