Abstract
Osteoarthritis is a common disease resulting in significant disability without approved disease-modifying treatment (other than total joint replacement). Stem cell-based therapy is being actively explored for the repair of cartilage lesions in the treatment and prevention of osteoarthritis. Embryonic stem cells are a very attractive source as they address many of the limitations inherent in autologous stem cells, such as variability in function and limited expansion. Over the past 20 years, there has been widespread interest in differentiating ESC into mesenchymal stem cells and chondroprogenitors with successful in vitro, ex vivo, and early animal studies. However, to date, none have progressed to clinical trials. In this review, we compare and contrast the various approaches to differentiating ESC; and discuss the benefits and drawbacks of each approach. Approaches relying on spontaneous differentiation are simpler but not as efficient as more targeted approaches. Methods replicating developmental biology are more efficient and reproducible but involve many steps in a complicated process. The small-molecule approach, arguably, combines the advantages of the above two methods because of the relative efficiency, reproducibility, and simplicity. To better understand the reasons for lack of progression to clinical applications, we explore technical, scientific, clinical, and regulatory challenges that remain to be overcome to achieve success in clinical applications.
Graphical Abstract
Graphical Abstract.
Significance Statement.
Over the past 10 years, there has been widespread interest in differentiating embryonic stem cells into mesenchymal cells. We review the various approaches to differentiating embryonic stem cells and compare and contrast the benefits and drawbacks of each approach. We also list technical, scientific, clinical, and regulatory challenges that remain to be overcome to achieve success in clinical applications. Osteoarthritis is a common disease resulting in significant disability and no approved disease-modifying treatment. This review will benefit readers interested in cell therapy for osteoarthritis, the present status of embryonic stem cell therapy, and the reasons for lack of progression to clinical applications
Cell-Based Repair of Cartilage: The Need for a Suitable Cell Source
Current clinical approaches to address cartilage defects can be classified into repair, replacement, or regeneration. The present standard of care is to induce repair via microfracture of the subchondral bone which encourages marrow cell infiltration into the defect resulting in the formation of fibrocartilage.1 Replacement involves autologous or allogeneic grafts of healthy cartilage or osteochondral tissue, usually in defects larger than those that can be treated with microfracture.2-4 Regeneration is achieved by harvesting, expanding, and subsequent implantation of chondrocytes in suspension (autologous chondrocyte implantation (ACI))5,6 or chondrocytes cultured on a scaffold system (matrix-induced ACI).7 Other cell sources explored include bone marrow aspirate concentrate (BMAC) and allogeneic cells such as umbilical cord MSCs (CartiStem).8,9 Several challenges remain for current cartilage treatment methods, most notably the lack of consistent hyaline cartilage formation and the relatively high re-operation rates for young patients (reviewed in ref. 10).
The clinical efficacy of cell-based cartilage repair is reliant on an effective cell source that is yet to be universally accepted.1,11 Several autologous cell sources with chondrogenic potential have their respective advantages and drawbacks. Terminally differentiated cells such as chondrocytes are attractive due to their inherent phenotype; however, limited cell expansion, loss of phenotype after multiple passages,12,13 harvest site morbidity,14,15 and requirements for enzymatic isolation13 tend to limit their clinical potential (Table 1).
Table 1.
Therapeutic approaches for cartilage lesions.
| ESC | MACI | De Novo | MSC | CartiStem | |
|---|---|---|---|---|---|
| Cell source | Embryonic | Arthroscopic biopsy of healthy cartilage | Juvenile allograft tissues | Bone marrow, adipose, synovial tissues | Umbilical cord blood |
| Harvest site morbidity | None | Arthroscopic biopsy | None | Needle aspiration; liposuction | None |
| Proliferation capacity | Very high | Limited | N/A | Limited to early passages | Very high |
| Chondrogenic differentiation potential | Good | Good | Not needed | Good | Good |
| Status in translation to clinical use | Preclinical | FDA approved | FDA approved | Permitted by FDA for certain applications | FDA approved |
| Potential for immune response | Yes | No | Yes | No | Yes |
| Commercialization/Potential for off-the-shelf therapy | Yes | No | Yes | No | Yes |
| Ethical considerations | Destruction of embryos | None | None | None | None |
| Regulatory challenges | High | FDA approved | FDA approved | Only permitted for minimally manipulated cells | FDA approved |
| Safety and potential risks in addition to surgical implantation | Risk of teratoma formation | Small surgical risk of biopsy | Minimal | Small surgical risk of needle aspiration | Minimal |
Adult-derived mesenchymal stem cells (MSC) are an alternate source of chondroprogenitors. MSC have cartilage-forming capacity and can be derived from bone marrow, adipose, synovial membrane, or other tissues with less invasive techniques.16-18 Conversely, site harvest morbidity can be an issue, there are limitations on extensive cell expansion before senescence and reduction in cartilage forming capacity,19,20 and adult MSC tend to undergo hypertrophic differentiation.21 Adult MSC are the most extensively studied stem cells in regenerative medicine with relatively few ethical, immunogenic, and teratogenic risks but with limitations on their ability to form cartilage tissue following extended culture.11,22 The clinical applications of adult MSC are therefore limited to using early passage cells.
Alternative cell sources are embryonic stem cells (ESC) and induced pluripotent stem cells (iPSCs), which have been used to generate MSC-like cells with cartilage tissue forming capacity (Tables 1-6). Embryonic stem cell-derived mesenchymal stem cells (ES-MSC) are a potentially universal source for cell-based cartilage regeneration, overcoming the inherent variability and limited availability of cells in autologous applications. Furthermore, these ES-MSC possess immunosuppressive and enhanced proliferative capacity in comparison to adult MSC.23
Table 6.
Mechanical stressor approach.
| Cells used | Applied stress | Reference |
|---|---|---|
| 7AC5 | 24-h static compression (0.05 MPa) | McKee et al58 |
Table 3.
Developmental approach.
| Cells used | Factors used | References |
|---|---|---|
| HUES1, HUES7, and HUES8 |
Primitive stream/Mesendoderm: WNT3A, Act-A, BMP4, FGF2, Mesoderm: FGF2, BMP4, NT4 |
Oldershaw et al.36 |
| H1, H9, and HES3 | Mesoderm: BMP4, VEGF, and bFGF, ActA. FACS: CD326- CD56+ and CD73+ CD105+ CD34− cells | Evseenko et al.34 |
| H1, UCLA3, HIPS23 | Mesoderm: bFGF, ActA, Noggin and Wnt3a. MACS: CD166+CD146+PDGFR-a+KDRneg | Wu et al40 |
| ESI-017 |
Mesoderm: bFGF, Wnt3a, ActA, Noggin MACS: CD326+ and CD309+ depletion |
Petrigliano et al41 |
| HUES1, MAN7, human fibroblast-derived iPSC | See: Oldershaw et al.36 | Cheng et al.32 (modification of Oldershaw et al.36) |
| HUES1, MAN7 | Substitution of BMP2 | Wang et al.37 (modification of Oldershaw et al.36) |
| 409B2, 604B1, HDF-11, KF4009-1 iPSCs | Adjusted Oldershaw & Umeda protocols | Yamashita et al.76 |
| H9 | Primitive Streak: BMP4 | Faial et al.49 |
| HES2, H7 |
Primitive Streak: ActA, BMP4, FGF Mesoderm: DM, FGF, |
Craft et al.38 |
| hiPSC (ATCC, BJFF, and STAN) |
Primitive Streak: Act, CHIR99021, FGF2 Mesoderm: SB505124, CHIR99021 FGF2, DM Chondroprogenitor: SB505124, WntC59, PD173074, DM, Purmorphamine, BMP4 |
Wu et al.39 |
| H1 |
Mesoderm (Days 1-3): FGF2, ActA, Wnt3a (Day 3-7): Add Noggin, remove ActA Chondrogenic (Day 11-14):FGF2, SHH, BMP4 (Day 14+): IGF2, FGF2, LIF, TGFb1, BMP4 |
Ferguson et al.35 |
There are obvious ethical, regulatory, and clinical concerns associated with the use of ESC for cartilage tissue regeneration.24 The major issues arise from the generation of these cells under conditions involving exposure to animal cells and products, undesirable differentiation, local or systemic biodistribution, or teratoma formation. ESC lines that have been developed using xeno-free processes may be more promising for clinical use. In this review, we focus on methods used to derive ES-MSC cell lines with cartilage forming potential, assess the strengths and weaknesses of these methods, and list the existing challenges for translating ES-MSC to a clinical therapy for cartilage repair.
Methods to Derive MSC-Like Cells from ESC
Several approaches, with varying degrees of complexity, have been described to differentiate embryonic stem cells (ESC) to an MSC-like or a chondroprogenitor phenotype for cartilage repair. The simplest approaches leverage the spontaneous induction of differentiation in ESC. While these methods may be easier to implement, the mechanisms are not well understood or have not been fully characterized. More elaborate methods simulate the key developmental signaling events, involve co-culturing with chondrocytes, applying mechanical stimulation, or enriching selective populations via cell sorting. An overview of the various approaches is provided in Fig. 1. Details of cell sources and differentiation factors are listed in Tables 2-6.
Figure 1.
Overview of methods used to derive MSC-like cells. Spontaneous differentiation protocols involve long-term culture for emergence and selection of an MSC phenotype; developmental differentiation emulates the progression of embryonic development of mesoderm and can be combined with cell selection via flow activated cell sorting (FACS); Small-molecules target temporal blocking of ALK-5 signaling; coculture of ESC with chondrocytes leverages paracrine signaling; and mechanical stimulation activates differentiation pathways. Evidence of differentiation is typically screened using flow cytometry, gene expression profiling, and histologic analysis of 3D cultures. Differentiated cell lines are initially screened in small animal models. For clinical application, proof of concept and safety studies need to be conducted in large animal models. EB = embryoid bodies.
Table 2.
Spontaneous approach.
| Cells used | Substrate | References |
|---|---|---|
| H9 | Standard TC Culture | Olee et al.25 |
| SA167, SA002.5, SA461 | Gelatin | de Peppo et al.80 |
| SA001, SA002, SA002.5, AS034, AS034.1.1, SA121, SA167, SA348, SA461, SA611. | Gelatin | Karlsson et al.81 |
| H9 | Gelatin | Nakagawa et al.26 |
| HUES9 | Gelatin | Hwang et al.82 |
| Umbilical cord-derived iPSC | Gelatin | Rim et al.83 |
| Adipose & Fibroblast-derived iPSC | Matrigel | Nejadnik et al.27 |
| Fibroblast-derived iPSC | MEFs | Guzzo and Drissi84 |
| H1, WA1 | Matrigel | Lee et al.30 |
| H1, H7, H9 | Matrigel | Trivedi and Hematti85 |
| H9 | MEFs | Gibson et al.31 |
| Nasal mucosa MSC-derived iPSC | Geltrex | Jakob et al.28 |
| QCTS-hESC-2 | Vitronectin | Xing et al.29 |
Spontaneous Differentiation
ESC in 2D culture spontaneously differentiate. We derived MSC-like cells by culturing mechanically dissected ESC colonies on standard tissue culture plastic.25 MSC-markers (CD73, CD90, CD44, and CD105) began to appear by passage 3; and by passage 9, the percentage of MSC-like cells had increased to 95%. These ES-MSC cells underwent chondrogenic differentiation in high density cultures and repaired articular defects in ex vivo human cartilage explants. Others have reported success with enhancing spontaneous differentiation by selective pressure favoring fast growing cells with the ability to attach and proliferate on gelatin, vitronectin, Matrigel, or Geltrex coatings.26-29 Culture on cell feeder layers such as murine bone marrow stromal cells (OP9) or irradiated mouse embryonic fibroblasts can also induce differentiation into ES-MSC.30,31
Developmental Approaches
The developmental approach involves a more directed recapitulation of the steps in embryonic development, namely the formation of primitive streak and paraxial mesoderm. Primitive streak differentiation is usually achieved by exposing ESC to several molecules such as Activin A, BMP2, BMP4, FGF2, and Wnt3a.32-38 Differentiation toward a paraxial mesoderm fate is induced by adding bFGF and follistatin, inhibiting BMP receptors with noggin or small-molecule dorsomorphin (DM), or inhibiting activin receptor-like kinase (ALK).32,33,35-39 The resulting cells are expanded into fibroblastic cells with an MSC-like phenotype in monolayer on standard tissue culture plastic or on substrates such as gelatin and fibronectin. Although more complicated, these developmental approaches appear to be robust, reproducible, well characterized, and feasible in inducing a mesenchymal phenotype and providing a source of chondroprogenitor cells. Chondrogenesis has been documented via expression of chondrogenic genes, and histologic analysis of 3D cultures in vitro or in vivo after subcutaneous implantation into immune deficient mice, or osteochondral defects in mice or rats.
Fluorescence-activated (FACS) and magnetic-activated cell sorting (MACS) have also been implemented to select mesoderm and pre-chondrogenic populations after mesoderm and mesenchymal induction.34,40,41 Petrigliano et al.41 used MACS to purify hESC-derived mesodermal cells and exclude epithelial (CD326+) and cardiovascular mesodermal (CD309+) cells. Cartilage regenerative capacity was shown by implanting ESC-derived chondrospheres or cell-seeded porcine collagen I/III membranes into full thickness chondral lesions in minipigs. After 6 months, the resultant repair tissue contained proteoglycans and collagen type II, evidence of stratification with a superficial region and good integration with the surrounding host tissue. In general, sorting with FACS and MACS results in a greater percentage of MSC-like cells (>95%), although the net cell population can be small (15-25% of the starting ESC).
Developmental approaches have been widely studied, are especially informative for understanding ESC differentiation, and have led to more targeted methods to generate chondroprogenitors.
Small Molecules
Small molecules can accelerate the derivation of chondroprogenitors, bypassing some of the steps in developmental approaches. Studies have used the ALK-inhibitor SB431542 (SB) to specifically drive ESCs to a mesoderm fate and derive MSC-like cells with cartilage forming capacity.42-44 FGF and TGFβ pathways are essential to maintain ESCs in a pluripotent state.45-47 To induce ESC differentiation, FGF is replaced with SB to inhibit SMAD 2/3 signaling by binding to the ATP-binding domains of activin receptor-like kinase (ALK) receptors 4, 5, and 7, while increasing the SMAD 1/5/8 signaling pathway.48 BMP4 expression increases after SB treatment, which is required for mesodermal fate.49
Inactivating ALK receptors also reduces NANOG promoter activity and leads to a loss of OCT4, SOX2, and NANOG expression (typically needed to maintain ESC in an undifferentiated state).44,45,50 The collective influence of SB treatment on differentiation pathways therefore induces a mesoderm fate.
The main differences among studies reporting the use of SB are duration of exposure and differentiation of ESC as embryoid bodies (EBs) or single cells in monolayer culture. Although all these methods led to emergence of MSC-like cell phenotypes, the efficiency of generating MSC and the requirement for EBs versus monolayer expansion must be weighed against the level of evidence for chondrogenesis. Mahmood et al. exposed EBs to SB before seeding on fibronectin42; Chen et al. did not require any specific coating of culture plates44; and Sanchez et al. avoided using EBs but required cell sorting.43 Differences in the reporting of mesenchymal cell markers, however, make it challenging to compare the purity of MSC populations among these methods.
Other small molecules that have been used to produce chondroprogenitors are a glycogen synthase kinase 3 inhibitor (CHIR99021), a retinoid acid receptor agonist (TTNPB), and a combination of bromodomain-containing protein 4 (BRD4) inhibitors (LLY-507 and AZD5153)51,52 (Table 4).
Table 4.
Small-molecule approach.
| Cells used | Small molecules | References |
|---|---|---|
| HUES9 | SB431542 | Mahmood et al42 |
| H9, AND1, AND2, SHEF1; iPSC (iPS-CB-CD34#2, iAND-4, iMSUH001) | SB431542 | Sánchez et al43 |
| Mel1, HES3; iPSC (MR90CL2 and ES4CL1) | SB431542 | Chen et al44 |
| H9, Mel1 | SB431542, BIO, Noggin | Umeda et al.86 |
| hiPSC (7F3955, PB001, PB004) | CHIR99021, TTNPB | Kawata et al.51 |
| H1, hiPSC | LLY-507 and AZD5153 | Zhang et al.52 |
Co-culture
Co-culturing with chondrocytes can also direct differentiation in pluripotent cells.53-56 Co-culture with primary human or bovine chondrocytes in culture well inserts induced chondrogenic differentiation in pluripotent cells.55-57 ESC or iPSC co-cultured with irradiated human chondrocytes in high-density pellet culture or encapsulated in cellulose and alginate have also exhibited successful chondrogenic commitment resulting in matrix deposition and morphology typical of cartilage.54 Xenogenic cell sources have also been explored for chondrogenic coculture, although unlikely to be suitable for clinical translation (Table 5).
Table 5.
Coculture approach.
| Cells used | Coculture cell type | References |
|---|---|---|
| H1 | Chondrocytes | Vats et al56 |
| BG02 | Chondrocytes | Hwang et al55 |
| hESC HS306, hiPSC (UEFhfiPSC1.4) | Chondrocytes | Qu et al87 |
| SA167, AS034, AS034.1 | Irradiated chondrocytes | Bigdeli et al.54 |
| OA chondrocyte-derived hiPSC | Chondrocytes | Wei et al57 |
| H1, H9 | OP9 | Barberi et al53 |
| H1, H9, H13 | OP9 | Vodyanik et al88 |
| R1 (murine), FVB/N (murine) | Limb bud progenitor cells (murine) | Sui et al89 |
| ES-D3 GL (murine) | Hepatic cells HEPA-1C1c7 (murine) | Lee et al90 |
| A2B iPSC | Irradiated chondrocytes | Nguyen et al91 |
Mechanical Stress
Mechanical stimulation has major effects on cell proliferation and differentiation and has been used to produce chondroprogenitors. Twenty-four hours of static compression applied to murine ESC in a PDMS scaffold upregulated chondrogenic genes COL2A1, SOX9, and ACAN and downregulated hypertrophic chondrogenic markers RUNX2, COL10A1, and MMP13, and the pluripotent marker OCT4.58 Mechanical stimulation and selective control of matrix elasticity are therefore emerging areas of interest in the production of chondroprogenitors and may provide additional selectivity for the desired phenotype.
Assessing Chondrogenesis
In vitro, ex vivo, and in vivo models are commonly used to assess chondrogenesis and cartilage regeneration. In vitro models typically consist of high-density pellet cultures, cell-seeded scaffolds, or cell encapsulation in hydrogels under chondrogenic conditions. Ex vivo explants can additionally assess integration of neotissue with host tissue which is one of the major factors limiting clinical success.25 Establishing chondrogenic potential in human explants also overcomes a potential weakness of implanting human cells in animal models. Chondrogenesis has been assessed in vivo by implantation of pellet cultures,31,55 or cell-seeded hydrogels,32,37 into osteochondral defects32,37 or subcutaneous tissue.38,42
Validation in large animal models represents a major gap in the translation of ES-MSC for cartilage regeneration. One notable exception, Petrigliano et al.,41 implanted ESC derived chondrocytes into full thickness cartilage defects in Yucatan minipigs with encouraging results. Such validation of cartilage regeneration and healing in large mammals such as porcine, ovine, and caprine will be necessary before translation to clinical trials.
While the assessment of chondrogenesis is widely described, the differences in models, measurement techniques, and analysis make it difficult to compare efficacy among different studies. There is therefore a critical need for standardization of chondrogenic methods for clinical translation. Finally, we lack validation of these models and outcome measures. Why do cells that generate apparently compelling histology in vitro fail to do so in vivo? In all likelihood, the majority of negative animal results are not reported.
Translation of ESC-Derived Chondroprogenitors to the Clinic
Clinical translation of any cell therapy involves definitive demonstration of safety and efficacy in preclinical models, followed by clinical trials in a phased approach. This process involves strict compliance with the rules and regulations as required by the corresponding government agencies. In the US, for example, the Food and Drug Administration (FDA) specifies adherence to current Good Manufacturing Practices (cGMP) for all cell-lines intended for use in clinical trials. Meeting the criteria for the final release of cell-based products requires extensive testing to ensure sterility, identity, purity, potency, and genetic stability (Reviewed in59; ISSCR Guidelines 2016). Even stricter criteria are necessary for ESC-derived cell therapy due to the additional safety concerns of teratoma formation, undesired differentiation, proliferation out of the implantation site, local and systemic biodistribution, and off-target effects.60
Preclinical Requirements for ESC-Derived Cell Therapy
The FDA has published general guidelines for clinical use of cartilage repair products that are valuable to design ESC-based cell therapy for clinical trials (https://www.fda.gov/media/82562/download). Appropriate and convincing preclinical data supporting safety and efficacy of the proposed therapy are the primary criteria. Additional considerations must be made during experimental design to improve the translation of results into a new drug application (NDA). Cartilage repair in animal models should show proof of biological response, durability, toxicology, dose response in a cartilage lesion of clinically relevant size and location, and with appropriate endpoints that will inform subsequent clinical trials. These criteria are most applicable in a large animal model in which the size, depth, and location of the cartilage lesion can be analogous to human patients and long-term durability (at least 1 year) can be convincingly documented. The animal model should reflect these considerations and strive to replicate the clinical features of human patients in clinical trials.
While the FDA guidelines are broadly applicable to all cartilage repair products, ESC-based cell therapy bears an additional level of burden, particularly in terms of risk for teratoma formation, off-target effects, and allogeneic response.60 These safety studies must be carefully designed and executed as the clinical risks have yet to be fully established.
Derivation and Culture of ESC
A major concern related to derivation of earlier ESC lines was exposure to animal-derived materials, such as mouse embryonal fibroblasts (MEF) or fetal bovine serum (FBS) during culture,61-63 or during cryopreservation.64 To avoid this issue, human feeder cells have been used, including fetal muscle cells, fetal fibroblasts, adult fallopian tubal epithelial cells, and dermal fibroblasts.65,66 To reduce additional contamination from animal-based products, serum- and xeno-free reagents are available for cell expansion and cryopreservation.67,68 Matrigel, although widely used as a substrate for ESC culture and differentiation, is not suitable for clinical use because it is derived from mouse sarcoma cells.
As alternatives to Matrigel, several cell-free and xeno-free substrates have become available. These include extracellular glycoproteins such as vitronectin, laminin, fibronectin, and a number of synthetic substrates such as APMAAm, PMVE-alt-MA, PMEDSAH, and PAM6co-PSS2.69,70
To mitigate concerns of contamination with animal products, ESC lines have been successfully derived, cultured, and preserved in xeno-free conditions.66,71-74 The generation of more clinically relevant cell lines has largely focused on key areas of ESC derivation, expansion, and storage that were traditionally dependent on animal products: 1) the separation of the inner cell mass (ICM) from the trophectoderm and zona pellicuda, 2) establishment of human feeder sources, 3) ESC expansion protocols, and 4) cryopreservation methods and reagents. Bypassing the use of animal products during ICM harvest has been accomplished by simple mechanical dissection,74 chemical dissection of the zona pellicuda using acid solutions,72 or laser drilling.66,71 Traditional MEF layers have effectively been replaced with human feeder cells such as human foreskin and placental stromal fibroblasts, and fetal and umbilical cord tissue.66,72,74 Various xeno-free media and reagents are now commercially available for hESC culture on human feeder cells, most being supplemented with human serum albumin and basic fibroblast growth factor (bFGF). GMP-grade human serum albumin66 and many FDA-approved xeno-free reagents are available for slow cooling and vitrification, such as Cryostor10 used for HAD-C 100, 102, and 106 cell lines.66
Clinical Trials
The majority of hESC clinical trials to date have been conducted with earlier non-xeno free cell lines (H1, MA09, and I6).75 More recently, completely xeno-free hESC have been used for Phase I/II clinical trials such as the Hadassah hESC cell lines (HAD-C 100, 102, 106) for retinal degeneration and amyotrophic lateral sclerosis (73; clinicaltrials.gov). Clinical trials specific to ES-MSC are limited. A trial of hES-MSC for meniscus injury has been initiated as well as trials for interstitial cystitis and intrauterine adhesions (clinicaltrials.gov). To our knowledge, no clinical trials have been initiated using hESC-derived cells for cartilage tissue regeneration. Xeno-free clinical grade hESC may eliminate previous concerns associated with the use of animal products and expedite the use of hESC in clinical trials for cartilage regeneration.
Compared to the speed of development from discovery to animal studies, the last stage of clinical translation remains relatively sluggish. The major obstacles are technical challenges in cell manufacturing, regulatory hurdles in establishing safety, lack of translation to preclinical models, and lack of validation of preclinical models. Despite several reports of apparent success in animals, these potential therapies have not progressed to clinical trials. This lack of progress emphasizes the need to revisit the true value of these models. There is an unmet need for a validated translational pipeline that progresses predictably from in vitro, ex vivo, and small animal proof of concept to definitive preclinical studies in large animals.
Conclusions and Future Perspectives
Despite the potential advantages and numerous reports of ESC for chondrogenesis, no treatment has yet been approved for clinical studies. Ethical concerns regarding the source and derivation of embryonic cells have not been fully resolved. Technical issues with maintaining stemness during culture and storage, and safety risks such as teratogenesis and immune rejection require validation. A variety of approaches can differentiate ESC into MSC with potential for cartilage tissue formation and repair in vitro, ex vivo, and in animal models. To satisfy the requirements for scalable manufacturing, a clinical cell therapy candidate needs to be relatively simple, efficient, robust, and reproducible. Methods relying on spontaneous differentiation are simpler but not as efficient as more targeted approaches. Methods replicating developmental biology are more efficient and reproducible, but involve many steps in a complicated process. The small-molecule approach, arguably, combines the advantages of the above two methods because of the relative efficiency, reproducibility, and simplicity. Another important development in this field is the derivation of xeno-free cell lines and application of xeno-free conditions that are critical for clinical translation.
Cell sources need to be tested in appropriate animal models for safety, efficacy, and clinical relevance. Several small animal models have demonstrated in vivo evidence of cartilage regeneration using hESC.31,32,37,55,56,76,77 However, animal studies have not progressed to clinical trials, suggesting a need to revisit the true value of these models. It is essential to establish a validated translational pipeline that progresses reliably from in vitro, ex vivo, and small animal proof of concept to definitive preclinical studies in large animals. One disadvantage is that the response of human cells in xenogenic models is not always predictable. Another disadvantage of traditional animal models is the surgical creation of artificial cartilage lesions in young healthy animals. One needs to carefully assess the translational value of these models in appropriately predicting safety and efficacy in clinical trials. The veterinary field contains reports of clinical applications of cell therapy in animals.78,79 Animal models with clinical disease therefore may become increasingly valuable in assessing preclinical safety and efficacy to inform human clinical trials.
Contributor Information
Shawn Grogan, Shiley Center for Orthopaedic Research and Education, Scripps Health, La Jolla, CA, USA.
Joel Kopcow, Shiley Center for Orthopaedic Research and Education, Scripps Health, La Jolla, CA, USA.
Darryl D’Lima, Shiley Center for Orthopaedic Research and Education, Scripps Health, La Jolla, CA, USA.
Funding
This study was funded by the California Institute for Regenerative Medicine (PC1-08128 & TR1-01216); the Shaffer Family Foundation; and Donald and Darlene Shiley.
Author Contributions
S.G.: conceptualization, formal analysis, investigation, methodology, project administration, supervision, validation, writing. D.D.: conceptualization, funding acquisition, methodology, project administration, resources, supervision, validation, writing. J.K.: data curation, investigation, writing.
Conflict of Interest
The authors declared no potential conflicts of interest.
Data Availability
No new data were generated or analyzed in support of this research.
References
- 1. Angele P, et al. Cell-based treatment options facilitate regeneration of cartilage, ligaments and meniscus in demanding conditions of the knee by a whole joint approach. Knee Surg Sports Traumatol Arthrosc. 2021;30(4):1138-1150. 10.1007/s00167-021-06497-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Goodfriend B, Essilfie AA, Jones IA, Thomas Vangsness C. Fresh osteochondral grafting in the United States: the current status of tissue banking processing. Cell Tissue Bank. 2019;20(3):331-337. 10.1007/s10561-019-09768-5 [DOI] [PubMed] [Google Scholar]
- 3. Hangody L, et al. Mosaicplasty for the treatment of articular defects of the knee and ankle. Clin Orthop Relat Res. 2001;391(Suppl):S328-S336. 10.1097/00003086-200110001-00030 [DOI] [PubMed] [Google Scholar]
- 4. Krych AJ, Saris DBF, Stuart MJ, Hacken B. Cartilage injury in the knee: assessment and treatment options. J Am Acad Orthop Surg. 2020;28(22):914-922. 10.5435/JAAOS-D-20-00266 [DOI] [PubMed] [Google Scholar]
- 5. Gou GH, Tseng F-J, Wang S-H, et al. Autologous chondrocyte implantation versus microfracture in the knee: a meta-analysis and systematic review. Arthroscopy. 2020;36(1):289-303. 10.1016/j.arthro.2019.06.033 [DOI] [PubMed] [Google Scholar]
- 6. Kon E, Filardo G, Di Martino A, Marcacci M. ACI and MACI. J Knee Surg. 2012;25(1):17-22. 10.1055/s-0031-1299651 [DOI] [PubMed] [Google Scholar]
- 7. Cherubino P, Grassi FA, Bulgheroni P, Ronga M. Autologous chondrocyte implantation using a bilayer collagen membrane: a preliminary report. J Orthop Surg (Hong Kong). 2003;11(1):10-15. 10.1177/230949900301100104 [DOI] [PubMed] [Google Scholar]
- 8. Gaul F, Bugbee WD, Hoenecke HR, D'Lima DD. A review of commercially available point-of-care devices to concentrate bone marrow for the treatment of osteoarthritis and focal cartilage lesions. Cartilage. 2019;10(4):387-394. 10.1177/1947603518768080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Park YB, Ha C-W, Lee C-H, Yoon YC, Park Y-G. Cartilage regeneration in osteoarthritic patients by a composite of allogeneic umbilical cord blood-derived mesenchymal stem cells and hyaluronate hydrogel: results from a clinical trial for safety and proof-of-concept with 7 years of extended follow-up. Stem Cells Transl Med. 2017;6(2):613-621. 10.5966/sctm.2016-0157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Riboh JC, et al. Comparative efficacy of cartilage repair procedures in the knee: a network meta-analysis. Knee Surg Sports Traumatol Arthrosc. 2017;25(12):37863799. 10.1007/s00167-016-4300-1 [DOI] [PubMed] [Google Scholar]
- 11. Deng Z, Jin J, Wang S, et al. Narrative review of the choices of stem cell sources and hydrogels for cartilage tissue engineering. Ann Transl Med. 2020;8(23):1598. 10.21037/atm-20-2342 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Charlier E, Deroyer C, Ciregia F, et al. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol. 2019;165:49-65. 10.1016/j.bcp.2019.02.036 [DOI] [PubMed] [Google Scholar]
- 13. Demoor M, Ollitrault D, Gomez-Leduc T, et al. Cartilage tissue engineering: molecular control of chondrocyte differentiation for proper cartilage matrix reconstruction. Biochim Biophys Acta. 2014;1840(8):2414-2440. 10.1016/j.bbagen.2014.02.030 [DOI] [PubMed] [Google Scholar]
- 14. Matricali GA, Dereymaeker GP, Luyten FP. Donor site morbidity after articular cartilage repair procedures: a review. Acta Orthop Belg. 2010;76(5):669-674. 10.1177/1947603515607963 [DOI] [PubMed] [Google Scholar]
- 15. McCarthy HS, Richardson JB, Parker JCE, Roberts S. Evaluating joint morbidity after chondral harvest for Autologous Chondrocyte Implantation (ACI): a study of ACI-treated ankles and hips with a knee chondral harvest. Cartilage. 2016;7(1):7-15. 10.1177/1947603515607963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Secunda R, Vennila R, Mohanashankar AM, et al. Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: a comparative study. Cytotechnology. 2015;67(5):793-807. 10.1007/s10616-014-9718-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Koga H, Engebretsen L, Brinchmann JE, Muneta T, Sekiya I. Mesenchymal stem cell-based therapy for cartilage repair: a review. Knee Surg Sports Traumatol Arthrosc. 2009;17(11):1289-1297. 10.1007/s00167-009-0782-4 [DOI] [PubMed] [Google Scholar]
- 18. Savkovic V, Li H, Seon J-K, et al. Mesenchymal stem cells in cartilage regeneration. Curr Stem Cell Res Ther. 2014;9(6):469-488. 10.2174/1574888x09666140709111444 [DOI] [PubMed] [Google Scholar]
- 19. Bernardo ME, Emons JAM, Karperien M, et al. Human mesenchymal stem cells derived from bone marrow display a better chondrogenic differentiation compared with other sources. Connect Tissue Res. 2007;48(3):132-140. 10.1080/03008200701228464 [DOI] [PubMed] [Google Scholar]
- 20. Pevsner-Fischer M, Levin S, Zipori D. The origins of mesenchymal stromal cell heterogeneity. Stem Cell Rev Rep. 2011;7(3):560-568. 10.1007/s12015-011-9229-7 [DOI] [PubMed] [Google Scholar]
- 21. Studer D, Millan C, Öztürk E, Maniura-Weber K, Zenobi-Wong M. Molecular and biophysical mechanisms regulating hypertrophic differentiation in chondrocytes and mesenchymal stem cells. Eur Cell Mater. 2012;24:118-35; discussion 135. 10.22203/ecm.v024a09. discussion 135 [DOI] [PubMed] [Google Scholar]
- 22. Wagner W, Horn P, Castoldi M, et al. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One. 2008;3(5):e2213. 10.1371/journal.pone.0002213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Brown PT, Squire MW, Li WJ. Characterization and evaluation of mesenchymal stem cells derived from human embryonic stem cells and bone marrow. Cell Tissue Res. 2014;358(1):149-164. 10.1007/s00441-014-1926-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Golchin A, Chatziparasidou A, Ranjbarvan P, Niknam Z, Ardeshirylajimi A. Embryonic stem cells in clinical trials: current overview of developments and challenges. Adv Exp Med Biol. 2021;1312:19-37. 10.1007/5584_2020_592 [DOI] [PubMed] [Google Scholar]
- 25. Olee T, et al. Repair of cartilage defects in arthritic tissue with differentiated human embryonic stem cells. Tissue Eng Part A. 2014;20:3683-3492. 10.1089/ten.tea.2012.0751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Nakagawa T, Lee SY, Reddi AH. Induction of chondrogenesis from human embryonic stem cells without embryoid body formation by bone morphogenetic protein 7 and transforming growth factor beta1. Arthritis Rheum. 2009;60(12):3686-3692. 10.1002/art.27229 [DOI] [PubMed] [Google Scholar]
- 27. Nejadnik H, Diecke S, Lenkov OD, et al. Improved approach for chondrogenic differentiation of human induced pluripotent stem cells. Stem Cell Rev Rep. 2015;11(2):242-253. 10.1007/s12015-014-9581-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Jakob M, et al. Pluripotent stem cell-derived mesenchymal stem cells show comparable functionality to their autologous origin. Cells 2020;10(1):33. 10.3390/cells10010033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Xing D, et al. Clinical-grade human embryonic stem cell-derived mesenchymal stromal cells ameliorate the progression of osteoarthritis in a rat model. Molecules. 2021;26(3):604. 10.3390/molecules26030604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lee PT, Li WJ. Chondrogenesis of embryonic stem cell-derived mesenchymal stem cells induced by TGFbeta1 and BMP7 through increased TGFbeta receptor expression and endogenous TGFbeta1 production. J Cell Biochem. 2017;118(1):172-181. 10.1002/jcb.25623 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Gibson JD, O'Sullivan MB, Alaee F, et al. Regeneration of articular cartilage by human ESC-derived mesenchymal progenitors treated sequentially with BMP-2 and Wnt5a. Stem Cells Transl Med. 2017;6(1):40-50. 10.5966/sctm.2016-0020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Cheng A, Kapacee Z, Peng J, et al. Cartilage repair using human embryonic stem cell-derived chondroprogenitors. Stem Cells Transl Med. 2014;3(11):1287-1294. 10.5966/sctm.2014-0101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Craft AM, Ahmed N, Rockel JS, et al. Specification of chondrocytes and cartilage tissues from embryonic stem cells. Development. 2013;140(12):2597-2610. 10.1242/dev.087890 [DOI] [PubMed] [Google Scholar]
- 34. Evseenko D, Zhu Y, Schenke-Layland K, et al. Mapping the first stages of mesoderm commitment during differentiation of human embryonic stem cells. Proc Natl Acad Sci USA. 2010;107(31):13742-13747. 10.1073/pnas.1002077107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ferguson GB, Van Handel B, Bay M, et al. Mapping molecular landmarks of human skeletal ontogeny and pluripotent stem cell-derived articular chondrocytes. Nat Commun. 2018;9(1):3634. 10.1038/s41467-018-05573-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Oldershaw RA, Baxter MA, Lowe ET, et al. Directed differentiation of human embryonic stem cells toward chondrocytes. Nat Biotechnol. 2010;28(11):1187-1194. 10.1038/nbt.1683 [DOI] [PubMed] [Google Scholar]
- 37. Wang T, Nimkingratana P, Smith CA, et al. Enhanced chondrogenesis from human embryonic stem cells. Stem Cell Res. 2019;39:101497. 10.1016/j.scr.2019.101497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Craft AM, Rockel JS, Nartiss Y, et al. Generation of articular chondrocytes from human pluripotent stem cells. Nat Biotechnol. 2015;33(6):638-645. 10.1038/nbt.3210 [DOI] [PubMed] [Google Scholar]
- 39. Wu CL, Dicks A, Steward N, et al. Single cell transcriptomic analysis of human pluripotent stem cell chondrogenesis. Nat Commun. 2021;12(1):362. 10.1038/s41467-020-20598-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Wu L, Bluguermann C, Kyupelyan L, et al. Human developmental chondrogenesis as a basis for engineering chondrocytes from pluripotent stem cells. Stem Cell Rep. 2013;1(6):575-589. 10.1016/j.stemcr.2013.10.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Petrigliano FA, Liu NQ, Lee S, et al. Long-term repair of porcine articular cartilage using cryopreservable, clinically compatible human embryonic stem cell-derived chondrocytes. NPJ Regen Med. 2021;6(1):77. 10.1038/s41536-021-00187-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Mahmood A, Harkness L, Schrøder HD, Abdallah BM, Kassem M. Enhanced differentiation of human embryonic stem cells to mesenchymal progenitors by inhibition of TGF-beta/activin/nodal signaling using SB431542. J Bone Miner Res. 2010;25(6):1216-1233. 10.1002/jbmr.34 [DOI] [PubMed] [Google Scholar]
- 43. Sanchez L, Gutierrez-Aranda I, Ligero G, et al. Enrichment of human ESC-derived multipotent mesenchymal stem cells with immunosuppressive and anti-inflammatory properties capable to protect against experimental inflammatory bowel disease. Stem Cells. 2011;29(2):251-262. 10.1002/stem.569 [DOI] [PubMed] [Google Scholar]
- 44. Chen YS, Pelekanos RA, Ellis RL, et al. Small molecule mesengenic induction of human induced pluripotent stem cells to generate mesenchymal stem/stromal cells. Stem Cells Transl Med. 2012;1(2):83-95. 10.5966/sctm.2011-0022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Hannan NR, Jamshidi P, Pera MF, Wolvetang EJ. BMP-11 and myostatin support undifferentiated growth of human embryonic stem cells in feeder-free cultures. Cloning Stem Cells. 2009;11(3):427-435. 10.1089/clo.2009.0024 [DOI] [PubMed] [Google Scholar]
- 46. Vallier L, Pedersen RA. Human embryonic stem cells: an in vitro model to study mechanisms controlling pluripotency in early mammalian development. Stem Cell Rev. 2005;1(2):119-130. 10.1385/SCR:1:2:119 [DOI] [PubMed] [Google Scholar]
- 47. Vallier L, Touboul T, Brown S, et al. Signaling pathways controlling pluripotency and early cell fate decisions of human induced pluripotent stem cells. Stem Cells. 2009;27(11):2655-2666. 10.1002/stem.199 [DOI] [PubMed] [Google Scholar]
- 48. Du J, Wu Y, Ai Z, et al. Mechanism of SB431542 in inhibiting mouse embryonic stem cell differentiation. Cell Signal. 2014;26(10):2107-2116. 10.1016/j.cellsig.2014.06.002 [DOI] [PubMed] [Google Scholar]
- 49. Faial T, Bernardo AS, Mendjan S, et al. Brachyury and SMAD signalling collaboratively orchestrate distinct mesoderm and endoderm gene regulatory networks in differentiating human embryonic stem cells. Development. 2015;142(12):2121-2135. 10.1242/dev.117838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Xu C, Jiang J, Sottile V, et al. Immortalized fibroblast-like cells derived from human embryonic stem cells support undifferentiated cell growth. Stem Cells. 2004;22(6):972-980. 10.1634/stemcells.22-6-972 [DOI] [PubMed] [Google Scholar]
- 51. Kawata M, Mori D, Kanke K, et al. Simple and robust differentiation of human pluripotent stem cells toward chondrocytes by two small-molecule compounds. Stem Cell Rep. 2019;13(3):530-544. 10.1016/j.stemcr.2019.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Zhang L, Wei Y, Chi Y, et al. Two-step generation of mesenchymal stem/stromal cells from human pluripotent stem cells with reinforced efficacy upon osteoarthritis rabbits by HA hydrogel. Cell Biosci. 2021;11(1):6. 10.1186/s13578-020-00516-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Barberi T, Willis LM, Socci ND, Studer L. Derivation of multipotent mesenchymal precursors from human embryonic stem cells. PLoS Med. 2005;2(6):e161. 10.1371/journal.pmed.0020161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Bigdeli N, Karlsson C, Strehl R, et al. Coculture of human embryonic stem cells and human articular chondrocytes results in significantly altered phenotype and improved chondrogenic differentiation. Stem Cells. 2009;27(8):1812-1821. 10.1002/stem.114 [DOI] [PubMed] [Google Scholar]
- 55. Hwang NS, Varghese S, Elisseeff J. Derivation of chondrogenically-committed cells from human embryonic cells for cartilage tissue regeneration. PLoS One. 2008;3(6):e2498. 10.1371/journal.pone.0002498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Vats A, Bielby RC, Tolley N, et al. Chondrogenic differentiation of human embryonic stem cells: the effect of the micro-environment. Tissue Eng. 2006;12(6):1687-1697. 10.1089/ten.2006.12.1687 [DOI] [PubMed] [Google Scholar]
- 57. Wei Y, Zeng W, Wan R, et al. Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur Cell Mater. 2012;23:1-12. 10.22203/ecm.v023a01 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. McKee C, Hong Y, Yao D, Chaudhry GR. Compression induced chondrogenic differentiation of embryonic stem cells in three-dimensional Polydimethylsiloxane Scaffolds. Tissue Eng Part A. 2017;23(9-10):426-435. 10.1089/ten.TEA.2016.0376 [DOI] [PubMed] [Google Scholar]
- 59. Rehakova D, Souralova T, Koutna I. Clinical-grade human pluripotent stem cells for cell therapy: characterization strategy. Int J Mol Sci . 2020;21(7):2435-2449. 10.3390/ijms21072435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Volarevic V, Markovic BS, Gazdic M, et al. Ethical and safety issues of stem cell-based therapy. Int J Med Sci. 2018;15(1):36-45. 10.7150/ijms.21666 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol. 2000;18(4):399-404. 10.1038/74447 [DOI] [PubMed] [Google Scholar]
- 62. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145-1147. 10.1126/science.282.5391.1145 [DOI] [PubMed] [Google Scholar]
- 63. van der Valk J, Brunner D, De Smet K, et al. Optimization of chemically defined cell culture media--replacing fetal bovine serum in mammalian in vitro methods. Toxicol In Vitro. 2010;24(4):1053-1063. 10.1016/j.tiv.2010.03.016 [DOI] [PubMed] [Google Scholar]
- 64. Ha SY, Jee BC, Suh CS, et al. Cryopreservation of human embryonic stem cells without the use of a programmable freezer. Hum Reprod. 2005;20(7):1779-1785. 10.1093/humrep/deh854 [DOI] [PubMed] [Google Scholar]
- 65. Llames S, García-Pérez E, Meana A, Larcher F, del Río M. Feeder layer cell actions and applications. Tissue Eng Part B Rev. 2015;21(4):345-353. 10.1089/ten.TEB.2014.0547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Tannenbaum SE, Turetsky TT, Singer O, et al. Derivation of xeno-free and GMP-grade human embryonic stem cells--platforms for future clinical applications. PLoS One. 2012;7(6):e35325. 10.1371/journal.pone.0035325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Holm F, Ström S, Inzunza J, et al. An effective serum- and xeno-free chemically defined freezing procedure for human embryonic and induced pluripotent stem cells. Hum Reprod. 2010;25(5):1271-1279. 10.1093/humrep/deq040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Zhang D, Mai Q, Li T, et al. Comparison of a xeno-free and serum-free culture system for human embryonic stem cells with conventional culture systems. Stem Cell Res Ther. 2016;7(1):101. 10.1186/s13287-016-0347-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. Nat Rev Mater. 2020;5(7):539-551. 10.1038/s41578-020-0199-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Higuchi A, Kao S-H, Ling Q-D, et al. Long-term xeno-free culture of human pluripotent stem cells on hydrogels with optimal elasticity. Sci Rep. 2015;5:18136. 10.1038/srep18136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Devito L, Jacquet L, Petrova A, et al. Generation of KCL034 clinical grade human embryonic stem cell line. Stem Cell Res. 2016;16(1):184-188. 10.1016/j.scr.2015.12.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Ellerstrom C, Strehl R, Moya K, et al. Derivation of a xeno-free human embryonic stem cell line. Stem Cells. 2006;24(10):2170-2176. 10.1634/stemcells.2006-0130 [DOI] [PubMed] [Google Scholar]
- 73. Tannenbaum SE, Singer O, Gil Y, et al. Hadassah, provider of “Regulatory-Ready” pluripotent clinicalgrade stem cell banks. Stem Cell Res. 2020;42:101670. 10.1016/j.scr.2019.101670 [DOI] [PubMed] [Google Scholar]
- 74. Ye J, Bates N, Soteriou D, et al. High quality clinical grade human embryonic stem cell lines derived from fresh discarded embryos. Stem Cell Res Ther. 2017;8(1):128. 10.1186/s13287-017-0561-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ilic D, Ogilvie C. Concise review: human embryonic stem cells-what have we done? What are we doing? Where are we going? Stem Cells. 2017;35(1):17-25. 10.1002/stem.2450 [DOI] [PubMed] [Google Scholar]
- 76. Yamashita A, Morioka M, Yahara Y, et al. Generation of scaffoldless hyaline cartilaginous tissue from human iPSCs. Stem Cell Rep. 2015;4(3):404-418. 10.1016/j.stemcr.2015.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Yamashita A, Tsumaki N. Recent progress of animal transplantation studies for treating articular cartilage damage using pluripotent stem cells. Dev Growth Differ. 2021;63(1):72-81. 10.1111/dgd.12706 [DOI] [PubMed] [Google Scholar]
- 78. Harman R, Carlson K, Gaynor J, et al. A prospective, randomized, masked, and placebo-controlled efficacy study of intraarticular allogeneic adipose stem cells for the treatment of osteoarthritis in dogs. Front Vet Sci 2016;3:81. 10.3389/fvets.2016.00081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Shah K, Drury T, Roic I, et al. Outcome of allogeneic adult stem cell therapy in dogs suffering from osteoarthritis and other joint defects. Stem Cells Int. 2018;2018:7309201. 10.1155/2018/7309201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. de Peppo GM, Svensson S, Lennerås M, et al. Human embryonic mesodermal progenitors highly resemble human mesenchymal stem cells and display high potential for tissue engineering applications. Tissue Eng Part A. 2010;16(7):2161-2182. 10.1089/ten.TEA.2009.0629 [DOI] [PubMed] [Google Scholar]
- 81. Karlsson C, Emanuelsson K, Wessberg F, et al. Human embryonic stem cell-derived mesenchymal progenitors-potential in regenerative medicine. Stem Cell Res. 2009;3(1):39-50. 10.1016/j.scr.2009.05.002 [DOI] [PubMed] [Google Scholar]
- 82. Hwang NS, Varghese S, Lee HJ, et al. In vivo commitment and functional tissue regeneration using human embryonic stem cell-derived mesenchymal cells. Proc Natl Acad Sci USA. 2008;105(52):20641-20646. 10.1073/pnas.0809680106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Rim YA, et al. Chondrogenic differentiation from induced pluripotent stem cells using non-viral minicircle vectors. Cells. 2020;9(3):582. 10.3390/cells9030582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Guzzo RM, Drissi H. Differentiation of human induced pluripotent stem cells to chondrocytes. Methods Mol Biol. 2015;1340:79-95. 10.1007/978-1-4939-2938-2_6 [DOI] [PubMed] [Google Scholar]
- 85. Trivedi P, Hematti P. Derivation and immunological characterization of mesenchymal stromal cells from human embryonic stem cells. Exp Hematol. 2008;36(3):350-359. 10.1016/j.exphem.2007.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Umeda K, Zhao J, Simmons P, et al. Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci Rep. 2012;2:455. 10.1038/srep00455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Qu C, Puttonen KA, Lindeberg H, et al. Chondrogenic differentiation of human pluripotent stem cells in chondrocyte co-culture. Int J Biochem Cell Biol. 2013;45(8):1802-1812. 10.1016/j.biocel.2013.05.029 [DOI] [PubMed] [Google Scholar]
- 88. Vodyanik MA, Yu J, Zhang X, et al. A mesoderm-derived precursor for mesenchymal stem and endothelial cells. Cell Stem Cell. 2010;7(6):718-729. 10.1016/j.stem.2010.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sui Y, Clarke T, Khillan JS. Limb bud progenitor cells induce differentiation of pluripotent embryonic stem cells into chondrogenic lineage. Differentiation. 2003;71(910):578-585. 10.1111/j.1432-0436.2003.07109001.x [DOI] [PubMed] [Google Scholar]
- 90. Lee HJ, Yu C, Chansakul T, et al. Enhanced chondrogenic differentiation of embryonic stem cells by coculture with hepatic cells. Stem Cells Dev. 2008;17(3):555-563. 10.1089/scd.2007.0177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Nguyen D, Hägg DA, Forsman A, et al. Cartilage tissue engineering by the 3D bioprinting of iPS cells in a nanocellulose/Alginate Bioink. Sci Rep. 2017;7(1):658. 10.1038/s41598-017-00690-y [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analyzed in support of this research.


