Electrospinning 2017; 1:62–72

Research Article

Nima Khadem Mohtaram, Vahid Karamzadeh, Yousef Shafieyan, and Stephanie M. Willerth* Commercializing Electrospun Scaffolds for Pluripotent Stem Cell-based Tissue Engineering Applications https://doi.org/10.1515/esp-2017-0003 Received May 08, 2017; accepted July 05, 2017 1 Pluripotent Stem Cells and their

Abstract: Tissue engineering, the process of combining Promise for Tissue Engineering bioactive scaffolds often with cells to produce replace- Applications ments for damaged organs, represents an enormous mar- ket opportunity. This review critically evaluates the com- Scientists had long hypothesized about the existence of mercialization potential of electrospun scaffolds for ap- stem cells in the body before confirming their existence plications in stem cell biology, including tissue engineer- experimentally [1]. These stem cells possess two distinct ing. First, it provides an overview of pluripotent stem cells properties that distinguish them from other types of cells (PSCs) and their defining properties, pluripotency and the present in an organism [2]. The first property of a stem cell ability to self-renew. These cells serve as an important tool is the ability to become or differentiate into multiple types for engineering tissues, including for clinical applications. of cells. Their second property is the ability to self-renew, Next, we review the technique of and its meaning that they can produce more stem cells. Both prop- promise for fabricating substrates and scaf- erties make stem cells a valuable tool for engineering tis- folds for directing tissue formation from stem cells and sues. The process of tissue engineering combines cells, compare these scaffolds to existing technologies, such as biochemical cues, and biomaterial scaffolds as a way of hydrogels. We address the associated market for electro- producing substitutes for diseased or damaged tissues and spun scaffolds for PSCs and its potential for growth along it represents a lucrative market valued in the billions of with highlighting the importance of 3D cell culture sub- dollars that is growing rapidly [3]. In terms of the history strates for PSCs by analyzing the net capital invested in of how stem cells were discovered, the Canadian duo of Dr. this market and the associated growth rate. This review Ernest Till and Dr. James McCulloch first identified mes- finishes by detailing the current state of commercializing enchymal stem cells in the 1960s [4–6]. However, these electrospun scaffolds along with pathways for translating cells were limited in terms of the types of mature cells that these scaffolds from research laboratories into success- they could become as they were derived from a specific tis- ful start-up companies and the associated challenges with sue. Thus, scientists aimed to find a more versatile type of this process. stem cell. One of the most desirable properties a stem cell can Keywords: Pluripotent stem cells, Electrospinning, possess is pluripotency, meaning that these cells can pro- Nanofibers, Microfibers, Commercialization, Tissue en- duce any cell type found in the organism from which they gineering were derived. Pluripotent stem cell lines are considered to be undifferentiated, reflecting this property, and these cells differentiate into mature phenotypes. The isolation *Corresponding Author: Stephanie M. Willerth: Department and culture of pluripotent stem cells (PSCs) occurred much of Mechanical Engineering, University of Victoria, Victoria, BC, later than the initial discovery of stem cells with these cells Canada; Division of Medical Sciences, University of Victoria, Vic- being isolated from mouse embryos in 1981 [7]. These PSCs toria, BC, Canada; Centre for Biomedical Research, Victoria, BC, Canada; International Collaboration On Repair Discoveries, Vancou- were termed embryonic stem cells (ESCs), reflecting the ver, BC, Canada; Department of Biochemistry, University of British Columbia, Vancouver, BC, Canada; Email: [email protected]; Tel.: 250-721-7303; Fax: 250-721-6051 Nima Khadem Mohtaram, Yousef Shafieyan: CellFace Ltd, Vahid Karamzadeh: Department of Mechanical Engineering, Con- Toronto, ON, Canada cordia University, Montreal, QC, Canada

Open Access. © 2017 N. K. Mohtaram et al., published by De Gruyter Open. This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License Commercializing Electrospun Scaffolds for Tissue Engineering Applications Ë 63 fact that they were derived from the embryo, which is an the first clinical trial evaluating human iPSC-derived reti- early stage of development that mammals go through in nal pigment epithelial cells is underway [19]. More on the the process of becoming a mature organism. Deriving such translation of PSC-derived therapies for clinical applica- pluripotent stem cells from human embryos was challeng- tions can be found in a recent review from Nature Reviews ing – both in terms of finding sufficient donor embryos and Molecular Cell Biology [20]. successfully completing the isolation process. Thus, it was While their pluripotency and their ability to self-renew not until 1998 that human ESC lines were derived in the serve as attractive properties for applications in tissue en- Thomson lab at the University of Wisconsin-Madison [8]. gineering, pluripotent stem cell lines, including ESCs and The generation of human ESCs lines then made it possible iPSCs, possess several limitations. One major concern is to engineer any type of tissue from these pluripotent cell that their ability to replicate indefinitely can lead to tu- lines. mour formation both in in vitro and in vivo settings. Thus, Significant advances were then made in understand- it is important to ensure that a chosen tissue engineering ing how to derive mature functional cells from human strategy promotes sufficient and mature differentiation of ESCs, such cardiac tissue, bone, pancreas, and nervous PSCs and avoids the presence of undifferentiated cells in tissue. These advances have resulted in two ESC-derived the construct that is to be implanted, such as in the cell products being evaluated in clinical trials. The first prod- therapies currently being evaluated in clinical trials. Un- uct, oligodendrocyte progenitor cells derived from human differentiated ESCs can be depleted using selection tech- ESCs, are being evaluated by Asterias Therapeutics (for- niques like flow cytometry or antibiotic-mediated death in merly Geron Corporation) in Phase I clinical trial for treat- combination with genetically altered cell lines [21]. A pair ing spinal cord injury [9]. Another clinical trial has exam- of studies has indicated that iPSC lines can be selected ined the ability of human ESC-derived retinal pigmented that possess a reduced risk of tumour formation [22, 23]. epithelium to promote recovery in patients suffering from As these cells can be maintained indefinitely in culture, macular degeneration, which demonstrated promising re- confirming their genomic integrity with routine karyotyp- sults [10, 11]. Many neurological diseases and disorders ing will ensure that no harmful mutations have occurred in lack true cures, making them attractive targets for cell ther- their DNA. Another approach to generating safe iPSC lines apies. In terms of pre-clinical work, it is anticipated that a uses non-integrating microRNA technology developed by Phase I clinical trial will be launched to evaluate the abil- the Nagy lab at the University of Toronto, as the transient ity of human ESC-derived dopaminergic neurons to treat expression of the pluripotency genes results in cell lines Parkinson’s Disease in the near future [12]. Other promis- without the risk of tumor formation [24]. Overall, PSCs rep- ing targets for ESC-derived therapies include treating dia- resent a powerful tool for engineering tissues, which will betes and heart disease [13, 14]. be explained in the next section. However, several issues remain with the use of hESCs. The number of cell lines derived was limited under the Bush administration as the United States restricted fund- 2 Current Status of PSCs for Tissue ing for stem cell research. As a result, researchers began to investigate alternative ways of generating PSCs. In 2006, Engineering Applications Takahashi and Yamanaka made a breakthrough discovery when they determined that four transcription factors, pro- Early work from the Langer lab explored how combina- teins which regulate the properties of cells, could be used tions of polymer scaffolds and growth factors could be to directly reprogram mouse fibroblasts back into a state of used to engineer different types of tissue from human pluripotency [15]. They named the resulting cells induced ESCs [25]. Since then, numerous studies have explored pluripotent stem cells (iPSCs). This breakthrough was fol- how to engineer functional tissues from PSCs as these cells lowed by the generation of human iPSC lines from fibrob- can become any type of tissue found in the body. The ma- lasts [16, 17]. These cells generated excitement for several jor challenge is determining the necessary set of physical reasons. First, they now enabled the generation of pluripo- and chemical cues necessary to guide these cells to form tent stem cells from specific patients, reducing the poten- functional tissues [26]. Scientists often look to the chemi- tial for immune rejection if the cells were used therapeuti- cal cues and physical structures present during tissue for- cally. It was also now possible to generate iPSC lines from mation during the process of development as a starting patients suffering from diseases, allowing the investiga- point for designing proper scaffolding for directing the dif- tion into disease mechanisms and the ability to use these ferentiation of PSCs [27]. lines for applications in drug screening [18]. In Japan, 64 Ë N. K. Mohtaram et al.

In terms of tissue engineering, the most commonly types of topography. Chemical cues can be incorporated used scaffolds are hydrogels fabricated from biomateri- into the polymer solution to enable the resulting fibers als that are used to culture, expand, and differentiate to present bioactive factors in a controlled fashion [54– PSCs [28–31]. Hydrogels consist of cross-linked polymer 56]. Two variants of the electrospinning process exist – networks that contain high water content. These hydrogels solution and melt. In solution electrospinning, the poly- can be generated from both natural and synthetic poly- mer is dissolved in a liquid solvent [32] with other com- mers as their properties are selected to provide a support- ponents being mixed into polymer solution to further add ive environment for controlling PSC behavior. While hy- bio-functionalities, including small molecules and macro- drogels are the most commonly used scaffolds for engi- molecules, such as proteins [34, 48, 55, 57–64]. By con- neering tissues from PSCs, electrospinning has become an trast, melt electrospinning generates a liquid polymer by increasingly popular alternative fabrication technique for heating the material until it liquifies [53]. Both electrospin- generating such scaffolds. ning techniques have been successfully applied to the field of tissue engineering as way to produce a variety of tissue types [65–68]. 3 Fabrication of Fibrous Scaffolds using Solution and Melt 4 Tissue Engineering Applications Electrospinning of Fiber-Based Scaffolds Electrospinning has been used for diverse tissue engi- Solution electrospinning can produce engineered neering applications in research laboratories [32–45]. The nanofiber scaffolds with multiple functionalities, includ- technique spins highly charged polymer solutions into ing the controlled release of morphogens and the pre- nanofibers or microfibers. In addition to the voltage field sentation of novel topographical cues. These nanofiber used to charge the polymer, other parameters play a key scaffolds have been combined with PSCs to engineer a role in this process, including collection distance between variety of tissues, including neural, cardiac, and carti- the extruded polymer and the collector, the method of lage. The Willerth lab has done extensive work designing fiber collection, needle size used for extrusion and some and fabricating such multifunctional scaffolds for promot- other material-related parameters like molecular weight, ing the neuronal differentiation of PSCs [47, 59, 69, 70]. solution concentration, viscosity and the type of solvent For instance, Mohtaram et al. showed that encapsulation used to dissolve polymer or the melting point of the poly- of retinoic acid inside engineered nanofibers with differ- mer [46–50]. A wide variety of polymers have been electro- ent topographies enhanced the neuronal differentiation spun into scaffolds, including both natural and synthetic of mouse iPSCs while guiding the outgrowth of neurites materials [51]. In terms of synthetic materials, poly (lactic- from these cells along the scaffold topography [55]. In co-glycolic acid) (PLGA) and poly (caprolactone) are pop- terms of differentiating human iPSCs into neural tissue, ular choices for electrospinning. Natural biomaterials, in- aligned nanofiber scaffolds were encapsulated with the cluding proteins like , fibrin and ceramics, have protein glial derived neurotrophic factor (GDNF), which also been electrospun. However, not all types of polymers was remained bioactive as it promoted the differentiation can be electrospun as their intrinsic material properties of human iPSCs into neurons [48]. These scaffolds can dictate if and how the resulting fibers will form during the repair damaged cardiac tissue as reviewed recently [71]. fabrication process. The electrospinning process can also Such electrospun nanofiber scaffolds can direct pluripo- alter the bioactivity of natural proteins, which is an impor- tent stem cells to form functional cardiac tissue as shown tant consideration when using electrospinning to fabricate in a 2016 study [72]. Nanofiber scaffolds have also been scaffolds for PSC cultures. designed that promote the differentiation of PSCs into os- Electrospinning provides a unique opportunity to fab- teogenic lineages as a way to engineer bone tissue [73, 74]. ricate complex scaffolds that present the necessary chemi- Finally, nanofiber scaffolds generated using solution elec- cal and physical cues for promoting differentiation of both trospinning can also direct PSCs to form chondrocytes, ESCs and iPSCs [40, 44, 52, 53]. However, ensuring proper enabling the generation of cartilage [75]. simultaneous presentation of chemical and physical cues Other groups have used engineered nanofibers for using electrospinning can be challenging. The collection other applications in directing PSC behavior as detailed by method for these fibers can be altered to produce different Higuchi et al. who performed an in depth review on the Commercializing Electrospun Scaffolds for Tissue Engineering Applications Ë 65 use of electropsun materials for culturing and differenti- et al. used melt electrospun scaffolds with novel topogra- ating PSCs [76]. Besides directing stem cell differentiation, phy to direct neurite extension from differentiated neurons electrospun scaffolds can be used as a tool for expanding derived from human iPSCs [69]. These scaffolds relied on pluripotent stem cells. In particular, defined electrospun physical cues (in this case, fiber orientation) to differen- scaffolds can serve as alternative to animal derived matri- tiate PSCs. Although melt electrospinning avoids the two ces, like Matrigel, which is often used to coat 2D substrates most common issues associated with solution electrospin- for expanding undifferentiated PSCs. 3D hydrogels have ning, which are the use of toxic solvents and the lack of also been evaluated as a tool for PSC expansion as recently reproducibility, there are still challenges to be addressed. reviewed [77, 78]. One study determined that aligned and Examples include finding suitable polymers that can be random PCL, PLGA, and poly (L-lactic acid) (PLLA) elec- melted, how to incorporate bioactive molecules and drugs, trospun substrates supported the culture and expansion how to electrospin natural biomaterials, and being able to of human ESCs [79]. Recent work published in Acta Bio- generate fibers on the nanoscale. materialia showed that electrospun polystyrene scaffolds Overall, both solution and melt electrospun scaffolds could be used to culture undifferentiated hiPSCs under have shown great promise in the field of stem cell-based xeno-free conditions, providing an attractive alternative to tissue engineering. Such scaffolds can be produced in a Matrigel [80]. A recent Biomaterials papers demonstrated highly scalable manner and have utility for applications how solution electrospinning can generate fibrous scaf- in PSC culture and differentiation into functional tissue. folds that mimic the , enabling the ex- Such engineered products are in high demand as more re- pansion of human PSCs [81]. Such substrates address one search funding is being invested in the field of stem cell of the major challenges in stem cell biology: producing based tissue engineering. The next section addresses the sufficient quantities of cells for therapeutic applications. market associated with such products and discusses the Although solution electrospinning remains an excellent current state of the field and its future directions. technique for fabricating 2D and 3D substrates for expand- ing and differentiating PSCs, many challenges exist with this technique. These challenges include the use of organic 5 3D Substrates for PSC Culture toxic solvents, which can have negative effects on cell be- havior, and the difficulties of controlling the architecture As mentioned earlier, PSCs can differentiate into any cell of the electrospun scaffolds. Another issue is ensuring that type found in human body. However, the differentiation the mechanical properties of these scaffolds are consistent process requires several complicated factors and specific as they can vary during fabrication. These issues serve as environments or niches [92, 97–101]. These niches present the main obstacles to commercializing these technologies. biochemical, physical and mechanical signals to PSCs, Melt electrospinning provides an attractive alterna- which influence differentiation. Most current in vitro mod- tive to the more commonly used technique of solution els for cell cultures seed aggregates of PSCs onto 2D electrospinning [39, 45, 82]. It opens new possibilities in polystyrene plates where they are treated with chemical the field of engineered electrospun scaffolds. Melt elec- cues to induce differentiation (102). Such plates will not trospinning produces engineered microfibers with a high present or mimic the complex microenvironment of PSCs degree of controllability and repeatability. Dr. Paul Dal- in vivo, and the cells often remodel their architecture to ton and his research group pioneered this technique and adapt to these 2D environments. Therefore, it is necessary they have used melt electrospinning to fabricate scaffolds to construct 3D surfaces that mimic the naturally occurring for a variety of tissue engineering applications [37, 83–91]. environment where PSCs proliferate, grow and differenti- These applications include culturing fibroblasts and engi- ate. neering replacements for damaged tendons. The Willerth Current micro and nanofabrication techniques can be lab in collaboration with Dr. Martin Jun’s research group implemented to generate 2D and 3D structures possessing at Purdue University used a customized melt electrospin- the topography and surface features to support more phys- ning setup to fabricate electrospun scaffolds with novel to- iologically relevant 3D stem cell cultures [103, 104]. Among pographies for promoting the differentiation of PSCs into such fabrication techniques is electrospinning. Therefore, neural tissue [59, 69, 70, 92–96]. Such scaffolds can be de- modifying the surface of common 2D plates with electro- signed and fabricated to meet needs of the consumer by spun fibers opens a new avenue for tissue engineering ap- altering parameters like needle size, collection distance plications that involve the expansion and differentiation and voltage field, which can be tuned to meet specifica- tions provided by a customer [46]. For example, Mohtaram 66 Ë N. K. Mohtaram et al.

Table 1: Examples of Companies that Supply Tissue Culture Plates and their Annual Revenue. (Source: PitchBook Database as of April 25_th, 2017, provided by Ontario Bioscience Innovation Organization – http://www.OBIO.ca).

Company Name Headquarters No of Employees Annual Revenue (US$ m) Johnson & Johnson United States 126,400 71,890.00 Pfizer Inc United States 96,500 52,824.00 F. Hoffmann-La Roche Ltd Switzerland 94,052 52,573.80 Bayer AG Germany 115,200 51,751.65 Novartis AG Switzerland 118,393 49,436.00 Merck & Co Inc United States 68,000 39,807.00 Sanofi France 106,859 38,405.70 GlaxoSmithKline Plc United Kingdom 99,827 37,679.18 Fresenius SE & Co KGaA Germany 232,873 31,064.14 Gilead Sciences Inc United States 9,000 30,390.00 Bayer HealthCare AG Germany 60,700 30,365.06 AbbVie Inc United States 30,000 25,638.00 AstraZeneca Plc United Kingdom 59,700 23,002.00 Amgen Inc United States 19,200 22,991.00 Teva Pharmaceutical Israel 56,960 21,903.00 Industries Ltd Eli Lilly and Company United States 41,975 21,222.10 of PSCs that could eventually be used for therapeutic ap- Various clients in health and education sectors bene- plications as described in the previous section. fit from 3D cell culture technology. For instance, pharma- ceutical companies, biotechnology and life-science com- panies, hospitals, universities, laboratories, and research 6 The Current State of institutes are among the potential customers for 3D cell culture plates and associated projects. Biotechnology com- Commercializing 3D Electrospun panies can generate revenue by licensing their technol- Scaffolds and Associated ogy to one of the current cell culture plate manufacturers. Alternatively, start-up companies can pursue joint ven- Challenges tures with a nanofabrication partner or a cell culture plate producer, generating revenue by selling biomimetic sur- Many biotechnology companies use electrospinning to faces and plates through the sales and marketing network produce biomaterial scaffolds for stem cell research, med- of the plate manufacturer. There are currently numerous ical devices and regenerative medicine as an alternative to well-established 2D cell culture manufacturers. Table 1 using hydrogels. Electrospun scaffolds can provide a 3D lists the most important companies in the field along with niche for stem cells seeded upon or inside. However, the their annual revenue. Most of these companies produce 3D currently available scaffolds are limited to certain appli- cell culture systems based on hydrogels, making it easier cations and have not been readily adopted as the industry to translate promising technologies. Additionally, many standard. Several major challenges must be addressed. For small to medium-size companies are also active in the de- instance, none of the current 3D cell culture systems have sign and manufacture of 3D cell culture plates. However, replaced 2D tissue culture products in terms of the large- none of these companies currently offer biomimetic sur- scale market. Many of the current 3D systems have issues faces suitable for PSC culture and differentiation. in regards to scalability, reproducibility, sensitivity, and compatibility with high-throughput screening [105]. The market for 3D cell culture substrates was $438 million in 2013. This market is expected to grow to about $2.2 billion in 2019 based on a compound annual growth rate (CAGR) of 30.1%. Commercializing Electrospun Scaffolds for Tissue Engineering Applications Ë 67

Table 2: Examples of Companies that Provide Stem Cells and their Annual Revenue. (Source: PitchBook Database as of April 25_th, 2017, provided by Ontario Bioscience Innovation Organization, http://www.OBIO.ca).

Company Name Company Financing Status Growth Primary Industry Group Rate Directlifeglobal Corporation 3.19% Pharmaceuticals and Biotechnology Silene Biotech Accelerator/Incubator Backed 2.73% Healthcare Services Stemmatters Venture Capital-Backed 2.33% Pharmaceuticals and Biotechnology Smart Cells International Venture Capital-Backed 1.82% Pharmaceuticals and Biotechnology Cell Medica Venture Capital-Backed 1.80% Pharmaceuticals and Biotechnology Nobleresearch Corporation 1.75% Pharmaceuticals and Biotechnology Publishers Stem Cell Therapy Corporation 1.72% Pharmaceuticals and Biotechnology Procedure Boston Biomedical Formerly VC-Backed 1.01% Pharmaceuticals and Biotechnology Animal Cell Therapies Angel-Backed 0.94% Pharmaceuticals and Biotechnology Hubrecht Institute Corporation 0.93% Pharmaceuticals and Biotechnology Nexcelom Bioscience Corporation 0.93% Pharmaceuticals and Biotechnology Biobest Laboratories Private Equity-Backed 0.88% Retail Candiolo Cancer Corporation 0.81% Healthcare Devices and Supplies Institute Irccs

Figure 1: Total Capital Invested in the Stem Cell Market in North America. (Source: PitchBook Database as of April 25_th, 2017, provided by Ontario Bioscience Innovation Organization, http://www.OBIO.ca).

7 The Market for 3D Electrospun pography. Table 2 contains examples of companies in- volved with stem cell research along with their growth rate Scaffolds and Stem Cell Related and financing status. Most of the market focuses onstem Products cell research, with less focus on cancer stem cells as ev- idenced by low product availability. Figure 1 shows the rapid growth in the total capital invested in stem cells re- Currently, stem cell research is one of the most common search. In 2016, it reached around $16 billion. This mar- applications of 3D cell cultures, and it has accounted for ket is expected to reach USD $18,329.37 million by 2020 the largest demand for 3D cell culture substrates. For the due to increased spending in research and development. purposes of this review, we will consider electrospun scaf- The methods for developing assays and related tools will folds to provide such a 3D environment due to their to- progress and become more streamlined, and drug devel- 68 Ë N. K. Mohtaram et al.

Figure 2: Total Capital Invested in the Market of Nanofibers in North America. (Source: PitchBook Database as of April 25_th, 2017, provided by Ontario Bioscience Innovation Organization, http://www.OBIO.ca). opers will be able to identify problems with drug candi- nique of electrospinning is quite similar, translating these date leads at earlier stages of development, reducing the technologies to market has been challenging. cost of drug discovery. This issue is one of the most press- In terms of demand for electrospun scaffolds, Figure 2 ing faced by small biopharmaceutical companies as over shows the total capital investment in North American com- 90% of the compounds identified for neurological diseases panies that sell nanofiber-based products, such as Inter- in pre-clinical screens fail during clinical trials [106]. Glob- face Biologics. Innovative, cutting edge, and promising ally, the regenerative medicine market is poised for intense technologies can be firstly designed and implemented in growth as companies turn to regenerative medicine to treat research laboratories. Intellectual property offices at Uni- unmet medical needs. Cell therapy products account for versities and research institutes have their own regula- 76% of regenerative medicine products currently in devel- tions and these offices work extensively with principal in- opment in Europe, Japan and the U.S. The role that 3D cell vestigators, as well as graduate students, to file provisional culture will play in regenerative medicine remains largely patents, protecting their intellectual property. Once these undefined, but can be compared to the testing of biolog- technologies are patented, research labs can perform the ics during process development. If so, the data from the necessary experiments to provide further proof-of-concept research applications will set the stage for later develop- data to support the claims made in the patents. This pro- ment in the field of 3D cell culture substrates pertaining to cess represents the initial steps necessary for commercial- therapeutics. ization of engineered scaffolds. Collaborative projects in tissue engineering have pro- duced numerous promising results as detailed earlier in 8 Translating Technology from this review which eventually support the commercial- ization of such technology. Patents and their associated Research Laboratories into publications serve as the main conceptual assets of start Start-up Companies up companies working in this field. Once these assests are in place, start up companies can approach local an- gel investors to further increase the chance of bringing Electrospun scaffolds are easily fabricated in research such products to large scale markets. Research laborato- laboratories. Most of electrospinning commercially avail- ries, hospitals, life science and pharmaceutical companies able electrospinning set-up companies, such as Spray- serve as the potential customers for engineered and cus- base, provide customized systems. Electrospinning setups tom designed scaffolds. Such products are referred to as include a high voltage power supply, a computer numeri- now-products since this field is extremely new and still cal control (CNC) machine, a computer, needles, syringes, faces serious challenges when it comes to successful com- and customized collectors. These components are readily mercialization. These challenges include the scale-up is- available in the commercial marketplace. While the tech- sues, regulatory affairs, and finding potential investigators to develop the products for market. Commercializing Electrospun Scaffolds for Tissue Engineering Applications Ë 69

1961; 14(2):213-22. 9 Conclusions and Future [6] Becker AJ, McCulloch EA, Till JE. Cytological demonstration of Recommendations the clonal nature of spleen colonies derived from transplanted mouse marrow cells. 1963. [7] Evans MJ, Kaufman MH. Establishment in culture of pluripoten- On-going pre-clinical and clinical trials are evaluating the tial cells from mouse embryos. Nature. 1981; 292(5819):154-6. use of stem cells to combat serious illnesses [107, 108]. [8] Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Many challenges are associated with stem cell therapies, Swiergiel JJ, Marshall VS, et al. Embryonic stem cell lines de- including achieving survival of these cells along with ef- rived from human blastocysts. science. 1998; 282(5391):1145-7. fectively controlling their differentiation into the desired [9] Biotherapeutic A. Asterias Biotherapeutics Receives Safety Clearance to Begin Administering the Highest Dose of AST-OPC1 phenotypes both in vitro and in vivo. 3D engineered elec- in the SCiStar Phase 1/2a Clinical Trial in Cervical Spinal Cord In- trospun biomaterial scaffolds can regulate the fate of PSCs jury Patients. 2016 for applications in tissue engineering and regenerative [10] Schwartz SD, Regillo CD, Lam BL, Eliott D, Rosenfeld PJ, Gregori medicine. Two classes of electrospun scaffolds and their NZ, et al. Human embryonic stem cell-derived retinal pigment role in regulating the fate of PSCs were discussed in this epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: follow-up of two open-label review along with a detailed discussion on the market for phase 1/2 studies. The Lancet. 2015; 385(9967):509-16. such products. Many existing start-up companies aim to [11] Song WK, Park K-M, Kim H-J, Lee JH, Choi J, Chong SY, et al. produce electrospun scaffolds which provide a relevant Treatment of macular degeneration using embryonic stem cell- microenvironment for stem cell studies, along with drug derived retinal pigment epithelium: preliminary results in Asian discovery and research. Electrospinning tech- patients. Stem cell reports. 2015; 4(5):860-72. nology can bridge the gap between traditional in vitro 2D [12] Buzhor E, Leshansky L, Blumenthal J, Barash H, Warshawsky D, Mazor Y, et al. Cell-based therapy approaches: the hope for in- cell culture systems and animal models, opening new av- curable diseases. Regen Med. 2014; 9(5):649-72. enues in commercialization of these products. [13] Moon S, Bae D, Jung T, Chung E, Jeong Y, Park S, et al. From Bench to Market: Preparing Human Pluripotent Stem Cells De- Acknowledgement: The authors acknowledge support rived Cardiomyocytes for Various Applications. International from the NSERC Discovery Grant program (S.M.W.), the journal of stem cells. 2017; 10(1):1. [14] Loo LSW, Lau HH, Jasmen JB, Lim CS, Teo AKK. An arduous jour- Canada Research Chairs program (S.M.W.), and the Stem ney from human pluripotent stem cells to functional pancreatic Cell Network (S.M.W). They would also like to acknowl- β-cells. Diabetes, Obesity and Metabolism. 2017. edge the CellFace Ltd. board members and the MaRS Dis- [15] Takahashi K, Yamanaka S. Induction of pluripotent stem cells covery District, and the Ontario Bioscience Innovation from mouse embryonic and adult fibroblast cultures by defined Organization (OBIO) for their online resources for en- factors. cell. 2006; 126(4):663-76. trepreneurs. We also would like to thank Keiran Letwin [16] Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human and Antonio Ranaudo for their constructive feedback on fibroblasts by defined factors. cell. 2007; 131(5):861-72. this manuscript. [17] Okita K, Ichisaka T, Yamanaka S. Generation of germline- competent induced pluripotent stem cells. nature. 2007; 448(7151):313-7. [18] Inoue H, Nagata N, Kurokawa H, Yamanaka S. iPS cells: a References game changer for future medicine. The EMBO journal. 2014; 33(5):409-17. [1] Ramalho-Santos M, Willenbring H. On the origin of the term [19] Fields M, Cai H, Gong J, Del Priore L. Potential of Induced Pluripo- “stem cell”. Cell stem cell. 2007; 1(1):35-8. tent Stem Cells (iPSCs) for Treating Age-Related Macular Degen- [2] Till JE, McCulloch EA, Siminovitch L. A stochastic model of stem eration (AMD). Cells. 2016; 5(4):44. cell proliferation, based on the growth of spleen colony-forming [20] Trounson A, DeWitt ND. Pluripotent stem cells progressing cells. Proceedings of the National Academy of Sciences. 1964; to the clinic. Nature Reviews Molecular Cell Biology. 2016; 51(1):29-36. 17(3):194-200. [3] Vacanti JP, Langer R. Tissue engineering: the design and fabri- [21] Odorico J, Pedersen R, Zhang S-C. Human embryonic stem cells: cation of living replacement devices for surgical reconstruction Garland Science; 2004. and transplantation. The lancet. 1999; 354:S32-S4. [22] Miura K, Okada Y, Aoi T, Okada A, Takahashi K, Okita K, et al. [4] McCulloch EA, Till JE. The radiation sensitivity of normal mouse Variation in the safety of induced pluripotent stem cell lines. Na- bone marrow cells, determined by quantitative marrow trans- ture biotechnology. 2009; 27(8):743-5. plantation into irradiated mice. Radiation research. 1960; [23] Tsuji O, Miura K, Okada Y, Fujiyoshi K, Mukaino M, Nagoshi 13(1):115-25. N, et al. Therapeutic potential of appropriately evaluated safe- [5] Till JE, McCulloch EA. A direct measurement of the radiation sen- induced pluripotent stem cells for spinal cord injury. Proceed- sitivity of normal mouse bone marrow cells. Radiation research. ings of the National Academy of Sciences. 2010; 107(28):12704- 9. 70 Ë N. K. Mohtaram et al.

[24] Clancy JL, Patel HR, Hussein SM, Tonge PD, Cloonan N, Corso AJ, plications: a critically selected survey. Journal of Biomaterials et al. Small RNA changes en route to distinct cellular states of Science, Polymer Edition. 2016; 27(2):157-76. induced pluripotency. Nature communications. 2014; 5. [43] Cheng J, Jun Y, Qin J, Lee S-H. Electrospinning versus microflu- [25] Levenberg S, Huang NF, Lavik E, Rogers AB, Itskovitz-Eldor J, idic spinning of functional fibers for biomedical applications. Langer R. Differentiation of human embryonic stem cells on Biomaterials. 2016. three-dimensional polymer scaffolds. Proceedings of the Na- [44] Khalf A, Madihally SV. Recent advances in multiaxial electro- tional Academy of Sciences. 2003; 100(22):12741-6. spinning for drug delivery. European Journal of Pharmaceutics [26] Willerth SM, Sakiyama-Elbert SE. Combining stem cells and bio- and Biopharmaceutics. 2016. material scaffolds for constructing tissues and cell delivery. [45] Willerth SM. Melt electrospinning in tissue engineering. In: Kny 2008. E, Uyar T, Khenoussi N, editors. Electrospun materials for tissue [27] Matthys OB, Hookway TA, McDevitt TC. Design Principles for En- engineering and biomedical applications: research, design and gineering of Tissues from Human Pluripotent Stem Cells. Cur- commercialization: Elsevier 2017. rent Stem Cell Reports. 2016; 2(1):43-51. [46] Ko J, Mohtaram NK, Ahmed F, Montgomery A, Carlson M, Lee PC, [28] Assuncao-Silva RC, Gomes ED, Sousa N, Silva NA, Salgado AJ. et al. Fabrication of poly (ϵ-caprolactone) microfiber scaffolds Hydrogels and Cell Based Therapies in Spinal Cord Injury Re- with varying topography and mechanical properties for stem generation. Stem Cells Int. 2015; 2015:948040. cell-based tissue engineering applications. Journal of Biomate- [29] Yang J, Zhang YS, Yue K, Khademhosseini A. Cell-Laden Hydro- rials Science, Polymer Edition. 2014; 25(1):1-17. gels for Osteochondral and Cartilage Tissue Engineering. Acta [47] Ko J, Ahsani V, Yao SX, Mohtaram NK, Lee PC, Jun MB. Fab- Biomaterialia. 2017. ricating and controlling PCL electrospun microfibers using fil- [30] Sepantafar M, Maheronnaghsh R, Mohammadi H, Rajabi-Zeleti ament feeding melt electrospinning technique. Journal of Mi- S, Annabi N, Aghdami N, et al. Stem cells and injectable hydro- cromechanics and Microengineering. 2016; 27(2):025007. gels: synergistic therapeutics in myocardial repair. Biotechnol- [48] Mohtaram N, Ko J, Agbay A, Rattray D, Neill P, Rajwani A, ogy advances. 2016; 34(4):362-79. et al. Development of a glial cell-derived neurotrophic factor- [31] Enam S, Jin S. Substrates for clinical applicability of stem cells. releasing artificial dura for neural tissue engineering applica- World journal of stem cells. 2015; 7(2):243. tions. Journal of Materials Chemistry B. 2015; 3(40):7974-85. [32] Li WJ, Laurencin CT, Caterson EJ, Tuan RS, Ko FK. Electrospun [49] Beachley V, Wen X. Effect of electrospinning parameters on the nanofibrous structure: a novel scaffold for tissue engineering. nanofiber diameter and length. Materials Science and Engineer- Journal of biomedical materials research. 2002; 60(4):613-21. ing: C. 2009; 29(3):663-8. [33] Shukla S, Brinley E, Cho HJ, Seal S. Electrospinning of hydrox- [50] Agarwal S, Wendorff JH, Greiner A. Progress in the field ofelec- ypropyl cellulose fibers and their application in synthesis of trospinning for tissue engineering applications. Advanced Ma- nano and submicron tin oxide fibers. Polymer. 2005 Dec 12; terials. 2009; 21(32-33):3343-51. 46(26):12130-45. [51] Manea L, Hristian L, Leon A, Popa A, editors. Recent advances of [34] Liao IC, Chew SY, Leong KW. Aligned core-shell nanofibers deliv- basic materials to obtain electrospun polymeric nanofibers for ering bioactive proteins. Nanomedicine : nanotechnology, biol- medical applications. IOP Conference Series: Materials Science ogy, and medicine. 2006 Dec; 1(4):465-71. and Engineering; 2016: IOP Publishing. [35] Jiang HL, Hu YQ, Zhao PC, Li Y, Zhu KJ. Modulation of protein re- [52] Cheng J, Jun Y,Qin J, Lee S-H. Electrospinning versus microfluidic lease from biodegradable core-shell structured fibers prepared spinning of functional fibers for biomedical applications. Bio- by coaxial electrospinning. Journal of Biomedical Materials Re- materials. 2017; 114:121-43. search Part B-Applied Biomaterials. 2006 Oct; 79B(1):50-7. [53] Uyar T, Kny E. Electrospun Materials for Tissue Engineering and [36] Sill TJ, von Recum HA. Electrospinning: applications in drug de- Biomedical Applications: Research, Design and Commercializa- livery and tissue engineering. Biomaterials. [Research Support, tion: Woodhead Publishing; 2017. Non-U.S. Gov’t Review]. 2008 May; 29(13):1989-2006. [54] Xie J, Willerth SM, Li X, Macewan MR, Rader A, Sakiyama-Elbert [37] Dalton PD, Joergensen NT, Groll J, Moeller M. Patterned melt SE, et al. The differentiation of embryonic stem cells seeded electrospun substrates for tissue engineering. Biomed Mater. on electrospun nanofibers into neural lineages. Biomaterials. 2008 Sep; 3(3):034109. 2009; 30(3):354-62. [38] Xie J, Macewan MR, Willerth SM, Li X, Moran DW, Sakiyama- [55] Mohtaram NK, Ko J, Montgomery A, Carlson M, Sun L, Wong A, Elbert SE, et al. Conductive Core-Sheath Nanofibers and Their et al. Multifunctional electrospun scaffolds for promoting neu- Potential Application in Neural Tissue Engineering. Adv Funct ronal differentiation of induced pluripotent stem cells. Journal Mater. 2009 Jul 24; 19(14):2312-8. of Biomaterials and Tissue Engineering. 2014; 4(11):906-14. [39] Brown TD, Dalton PD, Hutmacher DW. Direct writing by way of [56] Agbay A, Mohtaram NK, Willerth SM. Controlled release of glial melt electrospinning. Adv Mater. 2011 Dec 15; 23(47):5651-7. cell line-derived neurotrophic factor from poly (ϵ-caprolactone) [40] Muerza-Cascante ML, Haylock D, Hutmacher DW, Dalton PD. microspheres. Drug delivery and translational research. 2014; Melt electrospinning and its technologization in tissue engi- 4(2):159-70. neering. Tissue Engineering Part B: Reviews. 2014; 21(2):187- [57] Sahoo S, Ang LT, Goh JC, Toh SL. delivery through 202. electrospun nanofibers in scaffolds for tissue engineering ap- [41] Schaub NJ, Johnson CD, Cooper B, Gilbert RJ. Electrospun Fibers plications. J Biomed Mater Res A. 2010 Jun 15; 93(4):1539-50. for Spinal Cord Injury Research and Regeneration. J Neuro- [58] Mohtaram NK, Ko J, King C, Sun L, Muller N, Jun MB, et al. Elec- trauma. 2015 Dec 9. trospun biomaterial scaffolds with varied topographies for neu- [42] Villarreal-Gómez LJ, Cornejo-Bravo JM, Vera-Graziano R, Grande ronal differentiation of human-induced pluripotent stem cells. J D. Electrospinning as a powerful technique for biomedical ap- Biomed Mater Res A. 2015 Aug; 103(8):2591-601. Commercializing Electrospun Scaffolds for Tissue Engineering Applications Ë 71

[59] Agbay A, Edgar JM, Robinson M, Styan T, Wilson K, Schroll J, et al. [75] Liu J, Nie H, Xu Z, Niu X, Guo S, Yin J, et al. The effect of 3D nanofi- Biomaterial Strategies for Delivering Stem Cells as a Treatment brous scaffolds on the chondrogenesis of induced pluripotent for Spinal Cord Injury. Cells Tissues Organs. 2016; 202(1-2):42- stem cells and their application in restoration of cartilage de- 51. fects. PloS one. 2014; 9(11):e111566. [60] Kim TG, Lee DS, Park TG. Controlled protein release from elec- [76] Higuchi A, Kumar SS, Ling Q-D, Alarfaj AA, Munusamy MA, Muru- trospun biodegradable fiber mesh composed of poly(epsilon- gan K, et al. Polymeric design of cell culture materials that guide caprolactone) and poly(ethylene oxide). International Journal of the differentiation of human pluripotent stem cells. Progress in Pharmaceutics. [Research Support, Non-U.S. Gov’t]. 2007 Jun Polymer Science. 2017; 65:83-126. 29; 338(1-2):276-83. [77] Yang JJ, Liu JF, Kurokawa T, Kitada K, Gong JP. Hydrogels as [61] Maretschek S, Greiner A, Kissel T. Electrospun biodegradable feeder-free scaffolds for long-term self-renewal of mouse in- nanofiber nonwovens for controlled release of proteins. Journal duced pluripotent stem cells. Journal of tissue engineering and of controlled release : oflcial journal of the Controlled Release regenerative medicine. 2015; 9(4):375-88. Society. 2008 Apr 21; 127(2):180-7. [78] Cosson S, Otte EA, Hezaveh H, Cooper-White JJ. Concise review: [62] Piras AM, Chiellini F, Chiellini E, Nikkola L, Ashammakhi N. New tailoring bioengineered scaffolds for stem cell applications in Multicomponent Bioerodible Electrospun Nanofibers for Dual- tissue engineering and regenerative medicine. Stem cells trans- controlled Drug Release. Journal of Bioactive and Compatible lational medicine. 2015; 4(2):156-64. Polymers. 2008; 23(5):423-43. [79] Kumar D, Dale TP,Yang Y, Forsyth NR. Self-renewal of human em- [63] Valmikinathan CM, Defroda S, Yu XJ. Polycaprolactone and bryonic stem cells on defined synthetic electrospun nanofibers. Bovine Serum Albumin Based Nanofibers for Controlled Re- Biomedical Materials. 2015; 10(6):065017. lease of Nerve Growth Factor. Biomacromolecules. 2009 May; [80] Leong MF, Lu HF, Lim TC, Du C, Ma NK, Wan AC. Electrospun 10(5):1084-9. polystyrene scaffolds as a synthetic substrate for xeno-free ex- [64] Weng L, Xie J. Smart electrospun nanofibers for controlled drug pansion and differentiation of human induced pluripotent stem release: recent advances and new perspectives. Current phar- cells. Acta biomaterialia. 2016; 46:266-77. maceutical design. 2015; 21(15):1944-59. [81] Liu L, Kamei K-i, Yoshioka M, Nakajima M, Li J, Fujimoto N, et [65] Li M, Mondrinos MJ, Gandhi MR, Ko FK, Weiss AS, Lelkes PI. Elec- al. Nano-on-micro fibrous extracellular matrices for scalable ex- trospun protein fibers as matrices for tissue engineering. Bio- pansion of human ES/iPS cells. Biomaterials. 2017; 124:47-54. materials. 2005; 26(30):5999-6008. [82] Hutmacher DW, Dalton PD. Melt electrospinning. Chem Asian J. [66] da Silva MLA, Martins A, Costa-Pinto AR, Costa P,Faria S, Gomes 2011 Jan 3; 6(1):44-56. M, et al. Cartilage Tissue Engineering Using Electrospun PCL [83] Dalton PD, Lleixa Calvet J, Mourran A, Klee D, Moller M. Nanofiber Meshes and MSCs. Biomacromolecules. 2010 Dec; Melt electrospinning of poly-(ethylene glycol-block-epsilon- 11(12):3228-36. caprolactone). Biotechnol J. 2006 Sep; 1(9):998-1006. [67] Chung S, Ingle NP,Montero GA, Kim SH, King MW. Bioresorbable [84] Dalton PD, Lleixà Calvet J, Mourran A, Klee D, Möller M. Melt elastomeric vascular tissue engineering scaffolds via melt spin- electrospinning of poly-(ethylene glycol-block-ϵ-caprolactone). ning and electrospinning. Acta Biomater. 2010 Jun; 6(6):1958- Biotechnology journal. 2006; 1(9):998-1006. 67. [85] Dalton PD, Grafahrend D, Klinkhammer K, Klee D, Möller M. Elec- [68] Meinel AJ, Germershaus O, Luhmann T, Merkle HP, Meinel L. trospinning of polymer melts: phenomenological observations. Electrospun matrices for localized drug delivery: Current tech- Polymer. 2007; 48(23):6823-33. nologies and selected biomedical applications. European Jour- [86] Farrugia BL, Brown TD, Upton Z, Hutmacher DW, Dalton PD, nal of Pharmaceutics and Biopharmaceutics. 2012 Feb 11. Dargaville TR. Dermal fibroblast infiltration of poly(epsilon- [69] Mohtaram NK, Ko J, King C, Sun L, Muller N, Jun MBG, et caprolactone) scaffolds fabricated by melt electrospinning ina al. Electrospun biomaterial scaffolds with varied topographies direct writing mode. Biofabrication. 2013 Jun; 5(2):025001. for neuronal differentiation of human-induced pluripotent stem [87] Dalton PD, Muerza-Cascante ML, Hutmacher DW. Design and cells. Journal of Biomedical Materials Research Part A. 2015; Fabrication of Scaffolds via Melt Electrospinning for Applica- 103(8):2591-601. tions in Tissue Engineering. In: Mitchell GR, editor. Electro- [70] Pedde RD, Mirani B, Navaei A, Styan T, Wong S, Mehrali M, et spinning: Principles, practice and possibilities: Royal Society of al. Emerging Biofabrication Strategies for Engineering Complex Chemistry; 2015. Tissue Constructs. Advanced Materials. 2017. [88] Hochleitner G, Jüngst T, Brown TD, Hahn K, Moseke C, Jakob F, [71] Kim P-H, Cho J-Y. Myocardial tissue engineering using electro- et al. Additive manufacturing of scaffolds with sub-micron fil- spun nanofiber composites. BMB reports. 2016; 49(1):26. aments via melt electrospinning writing. Biofabrication. 2015; [72] Tang Y, Liu L, Li J, Yu L, Wang L, Shi J, et al. Induction and 7(3):035002. differentiation of human induced pluripotent stem cells into [89] Muerza-Cascante ML, Haylock D, Hutmacher DW, Dalton PD. functional cardiomyocytes on a compartmented monolayer of Melt electrospinning and its technologization in tissue engi- gelatin nanofibers. Nanoscale. 2016; 8(30):14530-40. neering. Tissue Eng Part B Rev. 2015 Apr; 21(2):187-202. [73] Deng Y,Yang Y,Wei S. Peptide-decorated nanofibrous niche aug- [90] Visser J, Melchels FP, Jeon JE, van Bussel EM, Kimpton LS, ments in vitro directed osteogenic conversion of human pluripo- Byrne HM, et al. Reinforcement of hydrogels using three- tent stem cells. Biomacromolecules. 2017. dimensionally printed microfibres. Nat Commun. 2015; 6:6933. [74] Xie J, Peng C, Zhao Q, Wang X, Yuan H, Yang L, et al. Osteogenic [91] Chen F, Hochleitner G, Woodfield T, Groll J, Dalton PD, Ams- differentiation and bone regeneration of iPSC-MSCs supported den BG. Additive Manufacturing of a Photo-Cross-Linkable by a biomimetic nanofibrous scaffold. Acta biomaterialia. 2016; Polymer via Direct Melt Electrospinning Writing for Produc- 29:365-79. ing High Strength Structures. Biomacromolecules. 2016 Jan 11; 72 Ë N. K. Mohtaram et al.

17(1):208-14. [100] Preston M, Sherman LS. Neural Stem Cell Niches: Critical Roles [92] Willerth SM. Biomimetic strategies for replicating the neural for the Hyaluronan-Based Extracellular Matrix in Neural Stem stem cell niche. Current Opinion in Chemical Engineering. 2017; Cell Proliferation and Differentiation. Frontiers in bioscience 15:8-14. (Scholar edition). 2012; 3:1165. [93] Ko J, Bhullar S, Mohtaram N, Willerth S, Jun M. Using mathe- [101] Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dy- matical modeling to control topographical properties of poly (ϵ- namic microenvironment for stem cell niche. Biochimica et Bio- caprolactone) melt electrospun scaffolds. Journal of Microme- physica Acta (BBA)-General Subjects. 2014; 1840(8):2506-19. chanics and Microengineering. 2014; 24(6):065009. [102] Knight E, Przyborski S. Advances in 3D cell culture technolo- [94] Ko J, Mohtaram NK, Ahmed F, Montgomery A, Carlson M, Lee gies enabling tissue-like structures to be created in vitro. Jour- PC, et al. Fabrication of poly (-caprolactone) microfiber scaf- nal of anatomy. 2015; 227(6):746-56. folds with varying topography and mechanical properties for [103] Krishna L, Dhamodaran K, Jayadev C, Chatterjee K, Shetty R, stem cell-based tissue engineering applications. J Biomater Sci Khora S, et al. Nanostructured scaffold as a determinant of stem Polym Ed. 2014; 25(1):1-17. cell fate. Stem Cell Research & Therapy. 2016; 7(1):188. [95] Mohtaram NK, Ko J, Montgomery A, Carlson M, Sun L, Wong A, et [104] Mashinchian O, Turner L-A, Dalby MJ, Laurent S, Shokrgozar al. Fabrication of encapsulated nanofibers with varied topogra- MA, Bonakdar S, et al. Regulation of stem cell fate by nanoma- phies for the neuronal differentiation of mouse induced pluripo- terial substrates. Nanomedicine. 2015; 10(5):829-47. tent stem cells. Biofabrication. 2014(Under review). [105] Picollet-D’hahan N, Dolega ME, Liguori L, Marquette C, Le Gac [96] Ko J, Mohtaram NK, Lee PC, Willerth SM, Jun MB. Mathemat- S, Gidrol X, et al. A 3D toolbox to enhance physiological rele- ical model for predicting topographical properties of poly (ϵ- vance of human tissue models. Trends in Biotechnology. 2016; caprolactone) melt electrospun scaffolds including the effects 34(9):757-69. of temperature and linear transitional speed. Journal of Mi- [106] Moscicki RA, Tandon P. Drug-Development Challenges for cromechanics and Microengineering. 2015; 25(4):045018. Small Biopharmaceutical Companies. New England Journal of [97] Conover JC, Notti RQ. The neural stem cell niche. Cell and tissue Medicine. 2017; 376(5):469-74. research. 2008; 331(1):211-24. [107] Raspa A, Pugliese R, Maleki M, Gelain F. Recent therapeutic ap- [98] Lund AW, Yener B, Stegemann JP, Plopper GE. The natural and proaches for spinal cord injury. Biotechnol Bioeng. 2015 Jul 1. engineered 3D microenvironment as a regulatory cue during [108] Squillaro T, Peluso G, Galderisi U. Clinical trials with mes- stem cell fate determination. Tissue Eng Part B Rev. 2009 Sep; enchymal stem cells: an update. Cell transplantation. 2016; 15(3):371-80. 25(5):829-48. [99] Fisher OZ, Khademhosseini A, Langer R, Peppas NA. Bioinspired materials for controlling stem cell fate. Acc Chem Res. 2010 Mar 16; 43(3):419-28.