Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Sep 1.
Published in final edited form as: Tech Shoulder Elb Surg. 2017 Sep 1;18(3):e6–e14. doi: 10.1097/BTE.0000000000000132

Cell-based approaches for augmentation of tendon repair

Camila B Carballo 1, Amir Lebaschi 1, Scott A Rodeo 1
PMCID: PMC5737795  NIHMSID: NIHMS886748  PMID: 29276433

Abstract

Cell-based approaches are among the principal interventions in orthobiologics to improve tendon and ligament healing and to combat degenerative processes. The number of options available for investigation are expanding rapidly and investigators have an increasing number of cell types to choose from for research purposes. However, in part due to the current regulatory environment, the list of available cells at clinicians’ disposal for therapeutic purposes is still rather limited. In this review, we present an overview of the main cellular categories in current use. Notable recent developments in cell-based approaches include the introduction of diverse sources of mesenchymal stem cells, pluripotent cells of extra-embryonic origin, and the emerging popularity of fully differentiated cells such as tenocytes and endothelial cells. Delivery strategies are discussed and a succinct discussion of the current regulatory environment in the United States is presented.

Keywords: cell-based, biologics, tendon, orthopedics, augmentation, repair

Overview of cell types and classifications

Cell-based therapies, which involve injection/inoculation/implantation of living cellular elements to patients, are becoming an increasingly prominent therapeutic option in orthopaedics. Paul Niehans (1882–1971), a Swiss physician, is probably the father of cell-based therapy. In 1931 he injected calf embryonic material to a patient, although there are no data available on the efficacy of his cell-injection practice. In 1953, it was found that rejection of organ transplants in laboratory animals could be prevented or diminished by pre-inoculating them with cells from donor animals. This preliminary finding finally led to the first successful human bone marrow transplantation in 19681. Since then, orthopedic diseases have been one of the earliest targets for cellular therapy, and cartilage repair was the first indication for this therapeutic approach2. In 1994, Lars Peterson’s group designed a study to regenerate cartilage tissue with autologous cartilage cells and showed evidence of regeneration in both animals and humans3.

Although adult mesenchymal stem cells have been the most widely used cellular element in orthopedics4, a number of other cell-based approaches have been used. Cells used in orthobiologics can be of embryonic, fetal, or adult origin and in turn, can be either autologous or allogeneic. Cells can also be categorized based on differentiation potential. Embryonic cells within the first few divisions after fertilization are totipotent, i.e., they are capable of creating another embryo and differentiate into any cell type, including extra-embryonic cells, and have the highest therapeutic potential. After the first few divisions, embryonic cells become pluripotent and can differentiate to all cells types, except for extra-embryonic cells. Pluripotent cells can be procured directly from human embryos (embryonic stem cells [ESCs]) or from a process known as somatic cell nuclear transfer (SCNT)5, 6.

The work of Takahashi and Yamanaka in 2006 opened a new avenue in cell-based approaches by introducing the third method for creating pluripotent stem cells: reprogramming adult cells back to a pluripotent state (induced pluripotent stem [iPS] cells). Their group was the first to demonstrate successful de-differentiation of somatic cells into a pluripotent ESC-like status by transfection with four embryonic transcription factors7.

Because of serious ethical issues, including the possibility of human embryo creation/destruction and risk of cloning an entire organism, the use of pluripotent stem cells is heavily regulated, and therefore, is not readily accessible for research and therapeutic purposes. Accordingly, in the United States these cells can only be used in an FDA-approved trial after a stringent review process5, 8. A serious consideration is that ESC and iPS cells have oncogenic potential. If they are injected in an undifferentiated state, they can potentially cause teratomas, and mice generated from iPS cells show high rates of neoplasia. This oncogenicity may be due to the transcription factors used for de-differentiation which are known to be oncogenes, insufficient epigenetic remodeling, or the oncogenic retroviruses used for transfection9, 10.

After the first 8 weeks of the embryonic stage, there is the next level of undifferentiated state where cells are “multipotent”. These cells, commonly referred to as “stem cells,” are able to differentiate into a limited number of cell types and reside in almost all tissues even after full development and are named based on their tissue of origin, such marrow- or adipose-derived stem cells. They pose less ethical and political controversy, tend to carry less risk, and cause fewer technical challenges than their pluripotent counterparts. Mesenchymal stem cells (MSCs) are by far the most widely used adult stem cells. Although bone marrow and adipose tissues are currently the most popular sources of MSCs because of less elaborate procurement methods, these cells can be obtained from almost all tissue types11, including tendons and ligaments. Bone marrow-derived mesenchymal stem cells can differentiate into several types of connective tissue including cartilage, bone, tendon, ligament, adipose, and muscle6, 12, 13.

All the above-mentioned cells can be considered some type of stem cell, although if not otherwise specified, current common usage in the literature mainly refers to undifferentiated adult stem cells. The term “adult” essentially refers to any stem cell beyond the first 8 weeks of embryonic life, where differential potential is confined to a limited number of cell types.

Fully differentiated cells, such as tenocytes, chondrocytes, and tissue-specific endothelial cells, constitute another category of cells with growing popularity in cell-based studies.

Delivery Strategies

Selection of an effective delivery technique is a crucial step in cell-based therapeutics. Currently, there is no consensus about the ideal carrier construct as few clinical data are available for cell-based approaches in tendon repair10.

The two major delivery categories are direct injection of a cell suspension alone and implantation of cells that are placed in a matrix carrier vehicle. Generally, selection of a delivery category depends on the goal of treatment. Replacement of a lost part will most probably require matrix-based deliveries. Application of a cell suspension is more appropriate in cases where the overall integrity of the tissue is maintained or restored, e.g., degenerative conditions or surgical repairs. An important obstacle in the use of matrix-based approaches is the insufficiency of diffusion of substances into the matrix to nourish and sustain the embedded cells beyond a certain thickness of matrix and, therefore, the need for vascularization, which is still an unsolved clinical problem14.

A cell suspension can be injected either locally or systemically. Successful systemic injection relies on homing of the injected cells to the target tissue/organ15, 16. Homing of stem cells subsequent to systemic injection has been shown in a variety of highly vascular tissues, including bone marrow, myocardium17, and liver18. Systemic administration of cells entails passage through pulmonary capillaries with potential risk of entrapment in the microvasculature19. Although MSC homing to fracture site has been documented in the literature20, direct delivery of cells appear to be a more plausible approach for tendons, regardless of the delivery category.

The majority of studies on tendons have used some form of a scaffold for local delivery of cells. Scaffold application techniques for tendons can be divided into gel suspensions, 3D scaffolds of solid tissue, and combination methods. Gel suspensions offer a favorable 3D filling of the defect, but the lower structural integrity and consequent reduced stability in comparison to other matrix materials may result in loss of the gel at the repair site due to erosion or resorption10. Fibrin sealant is a widely used gel scaffold21, 22 with several advantages including FDA approval, viability of suspended cells, and absence of an adverse effect on tendon healing23. Other scaffolds included collagen gel24, gel-collagen sponge composites25, and de-cellularized slices of tendon26.

Other important issues are timing and frequency of delivery and the rate of release when a scaffold is used as the vehicle27. A single, one time administration of any cell-based therapeutic may not be sufficient to harness the full potential biologic effects because injected/implanted cells may have limited longevity. Also, the dynamic nature of the cellular composition in the recipient environment may lead to absence of desirable cell-cell interactions at the right micro-anatomical location in a timely fashion. On the other hand, repeated administration is challenging even in animal models and, in case of human patients, may subject the process to greater regulatory restrictions.

Current cell-based approaches

Fetal and Embryonic cells

Culture of ESCs is inherently difficult because of the very small number of cells available to initiate the culture and the complex mixture of growth factors required for induction of differentiation6. Almost all investigations regarding the use of fetal and embryonic cells in tendon have been either in vitro or animal studies.

Stepwise differentiation of human ESCs promotes tendon regeneration by secreting fetal tendon matrix and differentiation factors. Chen et al showed MSCs that were derived subsequent to differentiating human ESCs are capable of regenerating patellar tendon in a rat model without formation of teratomas.28 In another report, Watts et al reported that the use of intra-lesional injection of male, fetal derived embryonic-like stem cells in an equine flexor tendon injury model led to improved tissue architecture, tendon size, tendon lesion size, and tendon linear fiber pattern29.

In vitro differentiation of ESCs to produce tendinous structures is an active area in tissue engineering. Cohen et al30 described the efficient derivation of connective tissue progenitors (CTPs) from human ESC lines and fetal tissues. CTPs were induced to generate tendon tissues in vitro, with ultrastructural characteristics and biomechanical properties typical of mature tendons. They also interposed rolled sheets of cultured CTPs in nude rat Achilles tendon full-thickness defects. The group found restoration of plantar flexion compared to control rats.

Amniotic and placenta-derived cells

MSCs derived from extra-embryonic tissues such as umbilical cord and placental tissues are emerging as an attractive source because of relatively easy availability as these tissues are normally discarded at birth, eliminating many ethical concerns31,32, 33. Accordingly, and also as an alternative to conventional pluripotent cells, amnion-derived stem cells, especially amniotic epithelial cells (AECs), are promising sources for cell-based therapy and have been a focus of active investigation34.

The amnion derives from the epiblast before gastrulation, the event prior to which cells are still pluripotent. Therefore, amnion-derived cells can potentially differentiate into all cell types35,36. Interestingly, MSCs isolated from amniotic fluid, umbilical cord blood37 and Wharton’s jelly in the horse show similar biological characteristics: all cell lines expand rapidly in culture, exhibit multi-differentiation potential, are positive for CD90, CD44, CD105, and negative for CD34, CD14 and CD4538.

Amniotic fluid is another source of AECs. These cells demonstrate a typical epithelial appearance39 and their pluripotency has been shown by the expression of molecular markers of pluripotent stem cells35, 36. Other very interesting characteristics displayed by amniotic cells are absence of tumorigenicty35, low immunogenicity, and the ability to induce immune-tolerance40. The latter two features make these cells promising candidates for allogeneic scenarios. Liu et al have also shown that ovine AECs can differentiate into bone and tendon tissue, both in vitro and when implanted into live animals4042,43. Furthermore, stemness and a favorable effect of ovine AECs in a large animal model of Achilles tendon injury have been reported by Muttini44 and Barboni40.

An alternative to amnion-derived cells, human amniotic membrane (HAM) is a more conveniently obtainable source which has been used in burn patients and to prevent peritendinous adhesions45. Dogramaci et al have recently demonstrated favorable results following application of HAM in an ovine flexor tendon reinforced tendon repair.

More recently, using extra-embryonic tissue, Park et al46 demonstrated the effects of human umbilical cord blood-derived MSC injection to a full-thickness subscapularis tendon tear in a rabbit model without surgical repair, which were evaluated by gross morphology, histology, and motion analysis of the rabbit activity. The group reported partial healing of tendon tears with histologic evidence of regenerated tendon tissue predominantly composed of type I collagens. Of note, the group did not detect any teratoma formation in study samples.

Mesenchymal stem cells

Mesenchymal stem cells (MSCs) appear to be excellent candidates in cell-based orthobiologics due to their long-term proliferation, high self-renewal rates, and ability of differentiation toward specific cell lineages. Although the exact origin of MSCs is still unclear, it is being increasingly recognized that many tissues harbor an intrinsic stem cell “niche” that could potentially be exploited or stimulated in different ways47. One of the more recent hypotheses states that their origin is associated with a unique population of cells lining blood vessel walls, namely endothelial and/or perivascular cells48.

MSCs have been a focus of intense in vitro and preclinical/animal research. Their ability to differentiate into specific lineages, including a tenogenic lineage, makes them a promising cell source for tendon and tendon-to-bone repair. This section will review several pre-clinical and some recent clinical studies that used MSCs for rotator cuff repair and elbow conditions.

Bone-marrow derived cells

Bone-marrow was first described as a viable source of MSCs (BM-MSCs) in 1970 by Friedenstein et al49. Bone marrow represents the standard and the most common source of autologous MSCs with the ability to biologically augment various tendon healing sites.

There are several reports on different methods of obtaining BM-MSCs safely and efficiently. McLain et al50 were able to isolate autologous BM-MSCs from iliac crest aspirate. Later, several groups showed that concentration of autologous bone marrow aspirate enhances the numbers of progenitor cells51, 52. Although the techniques of obtaining and concentrating bone marrow aspirate are evolving53, fewer than 0.01% of isolated cells are true multipotent stem cells based on the standard criteria described by Dominici et al54. In addition to iliac crest, BM-MSCs have also been successfully harvested from vertebrae50, femur, tibia, and humerus5557, obviating the need for iliac crest harvesting.

Animal studies that used MSCs to improve tendon-bone healing in rotator cuff repair have shown encouraging results (Table 1). Gulotta et al demonstrated a positive effect using allogeneic BM-MSCs transduced with the gene for matrix type-I matrix metalloproteinase (MT1-MMP) and scleraxis (SCX) in a rat rotator cuff model58,59. They showed that SCX led to a significance increase in the strength of repair and the amount of fibrocartilage at 4 weeks21. These effects were not seen when BM-MSCs transduced with BMP-13 were used60.

Table 1.

Preclinical studies of MSC therapy in rotator cuff repair.

Study Year Animal Repair Model Cell Type N MSCs Delivery Method
Gulotta et al 20 2009 Rat SST Allogenic BM-MSCs 106/animal Fibrin Glue Carrier
Gulotta et al 54 2010 Rat SST Allogenic BM-MSCs transduced with MT1-MMP 106/animal Fibrin Glue Carrier
Gulotta et al 56 2011 Rat SST Allogenic BM-MSCs transduced with BMP-13 106/animal Fibrin Glue Carrier
Gulotta et al 55 2011 Rat SST Allogenic BM-MSCs transduced with SCX 106/animal Fibrin Glue Carrier
Shen et al 84 2012 Rabbit SST Allogenic TDSCs 6 × 105/animal Seeded in Silk Collagen scaffold
Yokoya et al 59 2012 Rabbit IST Autologous BM-MSCs n/a Seeded in polyglycolic acid (PGA) sheet
Kida et al 57 2013 Rat SST Autologous BM-MSCs n/a Transosseous drilling
Kim et al 60 2013 Rabbit SST Autologous BM-MSCs n/a Seeded in open-cell polylactic acid (OPLA) scaffold
Levy et al 58 2013 Rat SST Autologous BM-MSCs n/a Transosseous drilling + cannulated nitinol implant
Oh et al 73 2014 Rabbit SSCT Allogenic ADSCs 107/animal Injection into SSC muscle
Mora et al 74 2014 Rat SST Allogenic ADSCs 2 × 106/animal Collagen Carrier
Tao et al 83 2015 Rabbit SST Allogenic TDSCs 4 × 106/animal Fibrin Glue Carrier
Park et al 44 2015 Rabbit SSCT Human UCB-MSCs 0.1mL/animal Ultrasound-guided injection
Degen et al 61 2016 Athymic Rat SST Human BM-MSCs 106/animal Fibrin Glue Carrier
Zong et al 62 2016 Athymic Rat SST Human BM-MSCs 106/animal Fibrin Glue Carrier

ADSCs: Adipose Derived stem/stromal cells, BMP-13: bone morphogenetic protein-13, IST: Infraspinatus tendon, MSCs: Mesenchymal stem cells, MT1-MMP: membrane type 1 matrix metalloproteinase, N: Number, SCX: Scleraxis, SSCT: subscapularis tendon, SST: Supraspinatus tendon, TDSCs: Tendon Derived stem/stromal cells, UCB: Umbilical Cord Blood.

Another approach to access bone marrow is the creation of multiple transosseous channels in the greater tuberosity, a technique known as microfracture. This has been evaluated in rats to stimulate autologous BM-MSCs into rotator cuff defects61, 62. In rabbits, the cells from bone marrow aspirate were either seeded on a polyglycolic acid (PGA) sheet63 or in an open-cell polylactic acid (OPLA) scaffold64. The outcomes of these cell-based scaffold studies have shown improved histological and biomechanical tendon properties but incomplete repair of the tendon-bone insertion.

More recently, human BM-MSCs have been implanted in an athymic rat supraspinatus tendon detachment and repair model. The results showed improved fibrocartilage formation, collagen orientation, and biomechanical strength 2 weeks following repair65. Further analysis of the data demonstrate that Indian hedgehog (Ihh) and Sox9 signaling play an important role in the tendon-to-bone healing mechanism66.

Recently, autologous bone marrow has been used to augment rotator cuff repair in the clinical setting, both through multiple bone channels to promote local infiltration of MSCs67,68,69,70 and application of non-concentrated71 and concentrated72, 73 aspirate obtained from the iliac crest (Table 2). Although the results have shown significantly reduced re-tear rates based on tendon imaging, even when BM-MSCs were recruited rather than implanted, the clinical outcomes have shown only minor improvement. To further elucidate the influence of BM-MSCs, it is critical to identify the optimal number, concentration, and characteristics of multipotent stem cells that can be isolated and transplanted to the patient. A fundamental limitation is the fact that the number of defined stem cells by formal molecular criteria in either bone marrow or adipose tissue is very small.

Table 2.

Clinical studies of MSC therapy in rotator cuff repair.

Study Year N Repair Model Cell Type N MSCs Harvested Site Delivery Method Time Points
Ellera Gomes et al 67 2012 14 Complete RC tears Autologous BM-MSCs n/a Dorsal IC Injection at the repaired tendon insertion site and bony footprint 12 m
Jo et al 63 2013 124 SST Autologous BM-MSCs n/a Humerus Multiple channels in the greater tuberosity 24 m
Milano et al 64 2013 80 Full-thickness RC tears BMS n/a Humerus Microfractures of the greater tuberosity 28 m
Hernigou et al 68 2014 90 SST Autogenous BM-MSC 51,000/12mL Anterior IC Injection at the repaired tendon insertion site and bony footprint 6 m and 10 years
Taniguchi et al 65 2015 111 All-sized + massive RC tears BMS n/a Humerus Drilling of multiple holes at the footprint 12-14 m
Skoff et al 69 2015 10 Revision RCr Autogenous BM-MSC n/a Dorsal IC Graft Incubation and injection of remaining marrow to the RC construct 24 m
Yoon et al 66 2016 75 Massive RC tears BMS n/a Humerus Drilling of multiple holes at the footprint + Patch (human dermis) graft 12 m

BMS: bone marrow stimulation, BM-MSCs: Bone marrow mesenchymal stem cells, IC: Iliac Crest, m: Month, MSCs: Mesenchymal stem cells, RC: Rotator cuff, RCr: Rotator cuff repair, SST: Supraspinatus tendon.

The application of MSCs in lateral epicondylitis has shown encouraging results. Singh et al have showed that a single injection of bone marrow aspirate from iliac crest improved the Patient-rated Tennis Elbow Evaluation (PRTEE) score after a short term follow up (maximum 12 weeks)74. More recently, Lee et al injected allogeneic adipose-derived MSCs mixed with fibrin sealant under ultrasound guidance. They demonstrated that the procedure was safe and effective in improving elbow pain, performance, and structural defects after approximately 1 year follow up75.

Adipose-derived stem cells

Another important source of multipotent stem cells (ADSCs) is the adipose tissue. Zuk et al have shown a favorable potential for augmenting rotator cuff repair with ADSCs76. Oh et al were among the first to report the use of injected ADSCs in a rabbit subscapularis rotator cuff model and found better healing properties and histologically decreased fatty infiltration of the muscle77. Mora et al. used ADSCs with a collagen carrier in a rat supraspinatus repair model and demonstrated no improvement in the biomechanical properties of the tendon-to-bone healing, but the ADSC group showed less inflammation based on histologic analysis of the healing tissue78. These results suggest that ADSCs could be a promising source. However, more studies are necessary to clarify the roles of these cells in the tendon-bone healing and their effect on muscle degeneration in rotator cuff tears.

Synovium-derived stem cells

Synovium-derived MSCs are reported to exhibit the greatest chondrogenic potential among mesenchymal tissue-derived cells79, 80 and thus could be a desirable source for enthesis restoration.MSCs from human subacromial bursa were recently characterized to be a potential synovial tissue for biological augmentation of rotator cuff repair81, 82. In a cell-based tendon tissue engineering approach, Song et al83 isolated bursa-MSCs (B-MSCs) from patients undergoing rotator cuff repair and demonstrated that when these cells are pretreated with BMP-12 and seeded in a ceramic scaffold, they expressed tenocyte markers and formed extensive bone, tendon-like tissue, as well as fibrocartilagenous tissue, confirming their substantial potential for application in tendon-to-bone repair.

Tendon derived stem cells (TDSCs)

TDSCs have been identified as an additional cell population in tendons84 and could be considered one of the newest types of MSCs. The multipotency of TDSCs were also characterized in torn human rotator cuff tendons85. TDSCs can be isolated from supraspinatus tendon and long head of biceps tendon during arthroscopic rotator cuff repair86.

Tao et al demonstrated that early growth response 1 (EGR1) transcription factor plays a key role in TDSC tenogenic differentiation and tendon formation and healing through the BMP12/Smad1/5/8 signaling pathway87. Shen et al have shown that allogeneic TDSCs seeded in silk-collagen scaffold enhanced the histological and biomechanical parameters of the rotator cuff tendon. They also demonstrated increased secretion of anti-inflammatory cytokines that prevent immunological rejection88.

Interestingly, a rare CD146+ tendon-resident stem cell population was identified in a rat patellar tendon. Subsequent to enrichment by connective tissue growth factor (CTGF), these cells demonstrated tenogenic differentiation. Application of these cells in a patellar tendon repair model successfully led to tendon regeneration and functional restoration. These data support the concept of stimulating endogenous progenitor cells, which could potentially overcome the limitations associated with transplantation of exogenous cells89.

Endothelial cells

It has been demonstrated that local endothelial cells (ECs) are a source of developmental cues for hepatic90 and pancreatic91 tissues. It was later realized that local ECs are also a source of regenerative signals in fully developed tissues. Subsequent investigations further elucidated that EC-derived growth factors play critical roles in repair and regeneration of adult bone marrow92, lung93, and liver47 in a tissue-specific fashion. These discoveries were results of transplantation of tissue-specific ECs in transgenic/mutant animals incapable of visceral tissue healing. Despite promising results from these studies on visceral tissues, hypovascularity and avascularity (in case of cartilage and the avascular zone of the meniscus) of orthopedic soft tissues renders EC-based approaches a challenging endeavor and call for special experimental designs to elucidate the potential role of ECs in these tissues.

Regulatory aspects

In the United States, the growing enthusiasm for using adult stem cell therapies in sports medicine is coupled with significant legal and regulatory obstacles. It is therefore important for the clinician to understand how adult stem cells are regulated in the United States, and how these complex rules are likely to affect what can and cannot be done in clinical practice94. In fact, a principal reason why cellular therapies have not been implemented more rapidly in the clinical setting is because of the complex and evolving regulatory requirements that have surrounded cellular products in recent years.8

Cell and cell products are regulated under both Public Health Service Act (PHSA) and Food, Drug, and Cosmetics Act (FDCA)95. Stem cells can also be considered a medical device, and therefore can be considered as combination products96. The definition of cell-based therapies used by regulatory agencies is that the essential feature of these products is the intention for use in diagnosis, treatment, or prevention of disease or affecting the structure or function of the body. The FDA oversees cell therapies through its Center for Biologics Evaluation and Research (CBER) and Center for Devices and Radiological Health (CDRH)9698.

FDA categorizes cell-based interventions as human cells, tissues, and cellular and tissue-based products (HCT/Ps) and uses a three-tiered structure to regulate their application. Current good tissue practices (cGTPs) applies throughout99. cGTPa are the requirements in subparts C and D of 21 CFR part 1271 that govern the methods used in, and the facilities and controls used for, the manufacture of HCT/Ps, including but not limited to all steps in recovery, donor screening, donor testing, processing, storage, labeling, packaging, and distribution. Table 3 outlines this regulatory structure100.

Table 3.

FDA regulatory categories.

Category 1 Category 2: Section 361 Category 3: Section 351
Oversight Level No HCT/P oversight Minimal Extensive Regulation
Product Risk Level Low Greater risk with regard to safety Greatest risk
Focus of the Regulations N/A Safety (preventing disease transmission) Safety and effectiveness.
Product Example Vascularized human organs for transplantation, whole blood and blood-derived products, and extracted human products such as collagen and bone marrow 99. Bone, cartilage, ligament, tendon, and skin 99. All other products
Establishment and/or Product Description An establishment that removes HCT/Ps from an individual and implants such HCT/Ps into the same individual during the same surgical procedure.
Products must be minimally manipulated, for homologous use and not combined with another article 99.
The product must be:
  1. No more than minimally manipulated. For cells or nonstructural tissues, it means that there must be no change in the “relevant biological characteristics of cells or tissues” during processing, storage, etc.

  2. Used for a homologous purpose.

  3. Combined with no other cells, tissues, or articles except for water, crystalloids, or a sterilizing, preserving, or storage agent, provided that their addition poses no additional concerns regarding clinical safety.

  4. These products must also have either
    1. No systemic effect or otherwise depend on the metabolic activity of living cells for their primary function or
    2. A systemic effect or depend on the metabolic activity of living cells for their primary function. and for
      • Autologous use
      • Allogeneic use in a first- or second-degree blood relative. or
      • Reproductive use.
The product is one or more of the following:
  1. More than minimally manipulated, which for cells and nonstructural tissue means to present a risk of change in cell morphology, function, expression, or other relevant biological characteristics during processing, storage, etc.

  2. Used for a nonhomologous purpose.

  3. Combined with other articles that may pose additional concerns regarding clinical safety.

  4. Have a systemic effect or otherwise rely on the metabolic activity of living cells for its primary function, and be used in a context other than autologous use, allogeneic use in a first or second degree relative, or reproductive use 98.

Main Regulatory Requirements Physicians must follow current Good Tissue Practices (cGTPs), but otherwise need not register as an establishment with the FDA’s CBER or submit a list of the HCT/Ps used. Physicians must employ current Good Tissue Practices (cGTPs), register their office or clinic as an “establishment” and submit an annually updated list of each HCT/P manufactured to CBER. They need not obtain premarketing approval before using the product or follow current Good Manufacturing Practices (cGMPs) in preparing them 99. Establishments must register and file a list of their HCT/Ps with CBER each year. Physician or clinic must complete the expensive process of obtaining formal premarket approval from the FDA (this can involve submitting a New Drug Application, an Investigational New Drug Application, Biologics License Application, or, when dealing with a Section 501k medical device, a premarket approval application or premarket notification). Physicians must follow the FDA-prescribed current Good Manufacturing Practices (cGMPs) and prescription drug labeling requirements that govern commercial pharmaceutical manufacturers.

The critical term to define in practice is “minimal manipulation.” For cells or nonstructural tissues, minimal manipulation is “processing of the HCT/P [that] does not alter the relevant biological characteristics of cells or tissues.” The degree of cell manipulation is critical in determining where an HCT/P will fall in the following three-tiered framework. Certain methods have been expressly regarded as minimal manipulation101:

  1. Centrifugation

  2. Cutting, grinding, or shaping

  3. Soaking in antibiotic solution

  4. Sterilization by ethylene oxide treatment or irradiation

  5. Cell separation

  6. Lyophilization

  7. Cryopreservation or freezing

Combining HCT/Ps with other “articles” can increase safety concerns. Therefore, the regulations exempt combining the HCT/P with water, crystalloids, or a sterilizing, preserving, or storage agent, provided that their addition poses no additional concerns regarding clinical safety. Other key criteria for classification as low risk are summarized in Table 3.

To comply with FDA regulations, it is of utmost importance that a physician understand what regulatory category a particular procedure involves. Specifically, review of previous classifications of similar products as well as an understanding of the FDA regulatory framework surrounding orthobiologics is important. A dialogue with the FDA is important during the planning of trials and prior to the initiation of new therapeutics. Failing to satisfy any of these requirements will expose the physician and clinic to increasingly stiff sanctions, ranging from site inspections and warning letters to a permanent injunction of the procedure or a shutdown of the entire establishment102. It is important to note that in determining the regulatory category and occurrence of non-compliance, it is FDA’s interpretation that counts.

Off-shore establishments have been increasingly used in an attempt to circumvent FDA regulations. Physicians should be mindful that FDA’s oversight holds true even if a single seemingly minor or irrelevant step (e.g., obtaining blood or marrow sample) of the overall application of HCT/P takes place in the U.S., with the rest of steps carried out offshore. It is possible to perform the entire process of harvesting, culturing, expanding, and injecting the patient’s own stem cells overseas, which is apparently outside FDA jurisdiction. However, through its formal Global Initiative, the FDA is currently forging collaborations with countries around the world to harmonize regulations. The central goal of this effort is to build regulatory capacity and develop international standards so that all the countries will employ similar approaches when regulating medical drugs and devices, including adult stem cell therapies94.

Acknowledgments

Source of Funding

This work has been partially funded by NIH T32 Training Grant.

Footnotes

Conflict of Interest

Authors have nothing to disclose. Authors have no conflict of interest.

References

  • 1.Starzl TE. History of clinical transplantation. World journal of surgery. 2000;24:759–782. doi: 10.1007/s002680010124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Noh MJ, Lee KH. Orthopedic cellular therapy: An overview with focus on clinical trials. World journal of orthopedics. 2015;6:754–761. doi: 10.5312/wjo.v6.i10.754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brittberg M, et al. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. The New England journal of medicine. 1994;331:889–895. doi: 10.1056/NEJM199410063311401. [DOI] [PubMed] [Google Scholar]
  • 4.Wang YK, Chen CS. Cell adhesion and mechanical stimulation in the regulation of mesenchymal stem cell differentiation. Journal of cellular and molecular medicine. 2013;17:823–832. doi: 10.1111/jcmm.12061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bongso A, Lee E. Stem Cells: From Bench to Bedside. World Scientific Publishing Company; Singapore: 2010. [Google Scholar]
  • 6.Petrou IG, et al. Cell therapies for tendons: old cell choice for modern innovation. Swiss medical weekly. 2014;144:w13989. doi: 10.4414/smw.2014.13989. [DOI] [PubMed] [Google Scholar]
  • 7.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 8.Anz AW, et al. Application of biologics in the treatment of the rotator cuff, meniscus, cartilage, and osteoarthritis. The Journal of the American Academy of Orthopaedic Surgeons. 2014;22:68–79. doi: 10.5435/JAAOS-22-02-68. [DOI] [PubMed] [Google Scholar]
  • 9.Rodolfa K, Di Giorgio FP, Sullivan S. Defined reprogramming: a vehicle for changing the differentiated state. Differentiation; research in biological diversity. 2007;75:577–579. doi: 10.1111/j.1432-0436.2007.00213.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Schmitt A, et al. Application of stem cells in orthopedics. Stem cells international. 2012;2012:394962. doi: 10.1155/2012/394962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. Journal of cell science. 2006;119:2204–2213. doi: 10.1242/jcs.02932. [DOI] [PubMed] [Google Scholar]
  • 12.Caplan AI. New era of cell-based orthopedic therapies. Tissue engineering Part B, Reviews. 2009;15:195–200. doi: 10.1089/ten.teb.2008.0515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Murray IR, et al. Recent insights into the identity of mesenchymal stem cells: Implications for orthopaedic applications. The bone & joint journal. 2014;96-B:291–298. doi: 10.1302/0301-620X.96B3.32789. [DOI] [PubMed] [Google Scholar]
  • 14.Rouwkema J, Rivron NC, van Blitterswijk CA. Vascularization in tissue engineering. Trends in biotechnology. 2008;26:434–441. doi: 10.1016/j.tibtech.2008.04.009. [DOI] [PubMed] [Google Scholar]
  • 15.Levesque JP, et al. Granulocyte colony-stimulating factor induces the release in the bone marrow of proteases that cleave c-KIT receptor (CD117) from the surface of hematopoietic progenitor cells. Experimental hematology. 2003;31:109–117. doi: 10.1016/s0301-472x(02)01028-7. [DOI] [PubMed] [Google Scholar]
  • 16.Petit I, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nature immunology. 2002;3:687–694. doi: 10.1038/ni813. [DOI] [PubMed] [Google Scholar]
  • 17.Barbash IM, et al. Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution. Circulation. 2003;108:863–868. doi: 10.1161/01.CIR.0000084828.50310.6A. [DOI] [PubMed] [Google Scholar]
  • 18.Glanemann M, et al. Transplantation of monocyte-derived hepatocyte-like cells (NeoHeps) improves survival in a model of acute liver failure. Annals of surgery. 2009;249:149–154. doi: 10.1097/SLA.0b013e31818a1543. [DOI] [PubMed] [Google Scholar]
  • 19.Jones E, McGonagle D. Human bone marrow mesenchymal stem cells in vivo. Rheumatology. 2008;47:126–131. doi: 10.1093/rheumatology/kem206. [DOI] [PubMed] [Google Scholar]
  • 20.Granero-Molto F, et al. Regenerative effects of transplanted mesenchymal stem cells in fracture healing. Stem cells. 2009;27:1887–1898. doi: 10.1002/stem.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gulotta LV, et al. Application of bone marrow-derived mesenchymal stem cells in a rotator cuff repair model. The American journal of sports medicine. 2009;37:2126–2133. doi: 10.1177/0363546509339582. [DOI] [PubMed] [Google Scholar]
  • 22.Chong AK, et al. Bone marrow-derived mesenchymal stem cells influence early tendon-healing in a rabbit achilles tendon model. The Journal of bone and joint surgery American volume. 2007;89:74–81. doi: 10.2106/JBJS.E.01396. [DOI] [PubMed] [Google Scholar]
  • 23.Lusardi DA, Cain JE., Jr The effect of fibrin sealant on the strength of tendon repair of full thickness tendon lacerations in the rabbit Achilles tendon. The Journal of foot and ankle surgery : official publication of the American College of Foot and Ankle Surgeons. 1994;33:443–447. [PubMed] [Google Scholar]
  • 24.Awad HA, et al. Repair of patellar tendon injuries using a cell-collagen composite. Journal of orthopaedic research : official publication of the Orthopaedic Research Society. 2003;21:420–431. doi: 10.1016/S0736-0266(02)00163-8. [DOI] [PubMed] [Google Scholar]
  • 25.Juncosa-Melvin N, et al. The effect of autologous mesenchymal stem cells on the biomechanics and histology of gel-collagen sponge constructs used for rabbit patellar tendon repair. Tissue engineering. 2006;12:369–379. doi: 10.1089/ten.2006.12.369. [DOI] [PubMed] [Google Scholar]
  • 26.Omae H, et al. Engineered tendon with decellularized xenotendon slices and bone marrow stromal cells: an in vivo animal study. Journal of tissue engineering and regenerative medicine. 2012;6:238–244. doi: 10.1002/term.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kueckelhaus M, et al. Sustained release of amnion-derived cellular cytokine solution facilitates achilles tendon healing in rats. Eplasty. 2014;14:e29. [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen X, et al. Stepwise differentiation of human embryonic stem cells promotes tendon regeneration by secreting fetal tendon matrix and differentiation factors. Stem cells. 2009;27:1276–1287. doi: 10.1002/stem.61. [DOI] [PubMed] [Google Scholar]
  • 29.Watts AE, et al. Fetal derived embryonic-like stem cells improve healing in a large animal flexor tendonitis model. Stem cell research & therapy. 2011;2:4. doi: 10.1186/scrt45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cohen S, et al. Repair of full-thickness tendon injury using connective tissue progenitors efficiently derived from human embryonic stem cells and fetal tissues. Tissue engineering Part A. 2010;16:3119–3137. doi: 10.1089/ten.TEA.2009.0716. [DOI] [PubMed] [Google Scholar]
  • 31.Longo UG, et al. Mesenchymal stem cell for prevention and management of intervertebral disc degeneration. Stem cells international. 2012;2012:921053. doi: 10.1155/2012/921053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Marcus AJ, Woodbury D. Fetal stem cells from extra-embryonic tissues: do not discard. Journal of cellular and molecular medicine. 2008;12:730–742. doi: 10.1111/j.1582-4934.2008.00221.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Veryasov VN, et al. Isolation of mesenchymal stromal cells from extraembryonic tissues and their characteristics. Bulletin of experimental biology and medicine. 2014;157:119–124. doi: 10.1007/s10517-014-2506-0. [DOI] [PubMed] [Google Scholar]
  • 34.Tahan AC, Tahan V. Placental amniotic epithelial cells and their therapeutic potential in liver diseases. Frontiers in medicine. 2014;1:48. doi: 10.3389/fmed.2014.00048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Miki T, et al. Stem cell characteristics of amniotic epithelial cells. Stem cells. 2005;23:1549–1559. doi: 10.1634/stemcells.2004-0357. [DOI] [PubMed] [Google Scholar]
  • 36.Ilancheran S, et al. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biology of reproduction. 2007;77:577–588. doi: 10.1095/biolreprod.106.055244. [DOI] [PubMed] [Google Scholar]
  • 37.Evangelista M, Soncini M, Parolini O. Placenta-derived stem cells: new hope for cell therapy? Cytotechnology. 2008;58:33–42. doi: 10.1007/s10616-008-9162-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Iacono E, et al. Isolation, characterization and differentiation of mesenchymal stem cells from amniotic fluid, umbilical cord blood and Wharton’s jelly in the horse. Reproduction. 2012;143:455–468. doi: 10.1530/REP-10-0408. [DOI] [PubMed] [Google Scholar]
  • 39.Parolini O, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international Workshop on Placenta Derived Stem Cells. Stem cells. 2008;26:300–311. doi: 10.1634/stemcells.2007-0594. [DOI] [PubMed] [Google Scholar]
  • 40.Barboni B, et al. Achilles tendon regeneration can be improved by amniotic epithelial cell allotransplantation. Cell transplantation. 2012;21:2377–2395. doi: 10.3727/096368912X638892. [DOI] [PubMed] [Google Scholar]
  • 41.Mattioli M, et al. Stemness characteristics and osteogenic potential of sheep amniotic epithelial cells. Cell biology international. 2012;36:7–19. doi: 10.1042/CBI20100720. [DOI] [PubMed] [Google Scholar]
  • 42.Muttini A, et al. Experimental study on allografts of amniotic epithelial cells in calcaneal tendon lesions of sheep. Veterinary research communications. 2010;34(Suppl 1):S117–120. doi: 10.1007/s11259-010-9396-z. [DOI] [PubMed] [Google Scholar]
  • 43.Muttini A, et al. Stem cell therapy of tendinopathies: suggestions from veterinary medicine. Muscles, ligaments and tendons journal. 2012;2:187–192. [PMC free article] [PubMed] [Google Scholar]
  • 44.Muttini A, et al. Ovine amniotic epithelial cells: in vitro characterization and transplantation into equine superficial digital flexor tendon spontaneous defects. Research in veterinary science. 2013;94:158–169. doi: 10.1016/j.rvsc.2012.07.028. [DOI] [PubMed] [Google Scholar]
  • 45.Dogramaci Y, Duman IG. Reinforcement of the Flexor Tendon Repair Using Human Amniotic MembraneA Biomechanical Evaluation Using the Modified Kessler Method of Tendon Repair. Journal of the American Podiatric Medical Association. 2016;106:319–322. doi: 10.7547/15-036. [DOI] [PubMed] [Google Scholar]
  • 46.Park GY, Kwon DR, Lee SC. Regeneration of Full-Thickness Rotator Cuff Tendon Tear After Ultrasound-Guided Injection With Umbilical Cord Blood-Derived Mesenchymal Stem Cells in a Rabbit Model. Stem cells translational medicine. 2015;4:1344–1351. doi: 10.5966/sctm.2015-0040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ding BS, et al. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature. 2010;468:310–315. doi: 10.1038/nature09493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Crisan M, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell stem cell. 2008;3:301–313. doi: 10.1016/j.stem.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 49.Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell and tissue kinetics. 1970;3:393–403. doi: 10.1111/j.1365-2184.1970.tb00347.x. [DOI] [PubMed] [Google Scholar]
  • 50.McLain RF, et al. Aspiration of osteoprogenitor cells for augmenting spinal fusion: comparison of progenitor cell concentrations from the vertebral body and iliac crest. The Journal of bone and joint surgery. 2005;87:2655–2661. doi: 10.2106/JBJS.E.00230. American volume. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.McLain RF, et al. Transpedicular aspiration of osteoprogenitor cells from the vertebral body: progenitor cell concentrations affected by serial aspiration. The spine journal : official journal of the North American Spine Society. 2009;9:995–1002. doi: 10.1016/j.spinee.2009.08.455. [DOI] [PubMed] [Google Scholar]
  • 52.Hernigou P, et al. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. The Journal of bone and joint surgery. 2005;87:1430–1437. doi: 10.2106/JBJS.D.02215. American volume. [DOI] [PubMed] [Google Scholar]
  • 53.Cassano JM, et al. Bone marrow concentrate and platelet-rich plasma differ in cell distribution and interleukin 1 receptor antagonist protein concentration. Knee surgery, sports traumatology, arthroscopy : official journal of the ESSKA. 2016 doi: 10.1007/s00167-016-3981-9. [DOI] [PubMed] [Google Scholar]
  • 54.Dominici M, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317. doi: 10.1080/14653240600855905. [DOI] [PubMed] [Google Scholar]
  • 55.Beitzel K, et al. Comparison of mesenchymal stem cells (osteoprogenitors) harvested from proximal humerus and distal femur during arthroscopic surgery. Arthroscopy : the journal of arthroscopic & related surgery : official publication of the Arthroscopy Association of North America and the International Arthroscopy Association. 2013;29:301–308. doi: 10.1016/j.arthro.2012.08.021. [DOI] [PubMed] [Google Scholar]
  • 56.Hernigou P, et al. Reduced levels of mesenchymal stem cells at the tendon-bone interface tuberosity in patients with symptomatic rotator cuff tear. International orthopaedics. 2015;39:1219–1225. doi: 10.1007/s00264-015-2724-8. [DOI] [PubMed] [Google Scholar]
  • 57.Mazzocca AD, et al. Rapid isolation of human stem cells (connective tissue progenitor cells) from the proximal humerus during arthroscopic rotator cuff surgery. The American journal of sports medicine. 2010;38:1438–1447. doi: 10.1177/0363546509360924. [DOI] [PubMed] [Google Scholar]
  • 58.Gulotta LV, et al. Stem cells genetically modified with the developmental gene MT1-MMP improve regeneration of the supraspinatus tendon-to-bone insertion site. The American journal of sports medicine. 2010;38:1429–1437. doi: 10.1177/0363546510361235. [DOI] [PubMed] [Google Scholar]
  • 59.Gulotta LV, Rodeo SA. Emerging ideas: Evaluation of stem cells genetically modified with scleraxis to improve rotator cuff healing. Clinical orthopaedics and related research. 2011;469:2977–2980. doi: 10.1007/s11999-010-1727-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Gulotta LV, et al. Adenoviral-mediated gene transfer of human bone morphogenetic protein-13 does not improve rotator cuff healing in a rat model. The American journal of sports medicine. 2011;39:180–187. doi: 10.1177/0363546510379339. [DOI] [PubMed] [Google Scholar]
  • 61.Kida Y, et al. Bone marrow-derived cells from the footprint infiltrate into the repaired rotator cuff. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2013;22:197–205. doi: 10.1016/j.jse.2012.02.007. [DOI] [PubMed] [Google Scholar]
  • 62.Levy DM, et al. Rotator cuff repair augmentation with local autogenous bone marrow via humeral cannulation in a rat model. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2013;22:1256–1264. doi: 10.1016/j.jse.2012.11.014. [DOI] [PubMed] [Google Scholar]
  • 63.Yokoya S, et al. Rotator cuff regeneration using a bioabsorbable material with bone marrow-derived mesenchymal stem cells in a rabbit model. The American journal of sports medicine. 2012;40:1259–1268. doi: 10.1177/0363546512442343. [DOI] [PubMed] [Google Scholar]
  • 64.Kim YS, et al. Survivorship of implanted bone marrow-derived mesenchymal stem cells in acute rotator cuff tear. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2013;22:1037–1045. doi: 10.1016/j.jse.2012.11.005. [DOI] [PubMed] [Google Scholar]
  • 65.Degen RM, et al. The Effect of Purified Human Bone Marrow-Derived Mesenchymal Stem Cells on Rotator Cuff Tendon Healing in an Athymic Rat. Arthroscopy : the journal of arthroscopic & related surgery : official publication of the Arthroscopy Association of North America and the International Arthroscopy Association. 2016 doi: 10.1016/j.arthro.2016.04.019. [DOI] [PubMed] [Google Scholar]
  • 66.Zong JC, et al. Involvement of Indian hedgehog signaling in mesenchymal stem cell-augmented rotator cuff tendon repair in an athymic rat model. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2016 doi: 10.1016/j.jse.2016.09.036. [DOI] [PubMed] [Google Scholar]
  • 67.Jo CH, et al. Multiple channeling improves the structural integrity of rotator cuff repair. The American journal of sports medicine. 2013;41:2650–2657. doi: 10.1177/0363546513499138. [DOI] [PubMed] [Google Scholar]
  • 68.Milano G, et al. Efficacy of marrow-stimulating technique in arthroscopic rotator cuff repair: a prospective randomized study. Arthroscopy : the journal of arthroscopic & related surgery : official publication of the Arthroscopy Association of North America and the International Arthroscopy Association. 2013;29:802–810. doi: 10.1016/j.arthro.2013.01.019. [DOI] [PubMed] [Google Scholar]
  • 69.Taniguchi N, et al. Bone marrow stimulation at the footprint of arthroscopic surface-holding repair advances cuff repair integrity. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2015;24:860–866. doi: 10.1016/j.jse.2014.09.031. [DOI] [PubMed] [Google Scholar]
  • 70.Yoon JP, et al. Outcomes of Combined Bone Marrow Stimulation and Patch Augmentation for Massive Rotator Cuff Tears. The American journal of sports medicine. 2016;44:963–971. doi: 10.1177/0363546515625044. [DOI] [PubMed] [Google Scholar]
  • 71.Ellera Gomes JL, et al. Conventional rotator cuff repair complemented by the aid of mononuclear autologous stem cells. Knee surgery, sports traumatology, arthroscopy : official journal of the ESSKA. 2012;20:373–377. doi: 10.1007/s00167-011-1607-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hernigou P, et al. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: a case-controlled study. International orthopaedics. 2014;38:1811–1818. doi: 10.1007/s00264-014-2391-1. [DOI] [PubMed] [Google Scholar]
  • 73.Skoff HD. Revision Rotator Cuff Reconstruction for Large Tears With Retraction: A Novel Technique Using Autogenous Tendon and Autologous Marrow. American journal of orthopedics. 2015;44:326–331. [PubMed] [Google Scholar]
  • 74.Singh A, Gangwar DS, Singh S. Bone marrow injection: A novel treatment for tennis elbow. Journal of natural science, biology, and medicine. 2014;5:389–391. doi: 10.4103/0976-9668.136198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lee SY, et al. Treatment of Lateral Epicondylosis by Using Allogeneic Adipose-Derived Mesenchymal Stem Cells: A Pilot Study. Stem cells. 2015;33:2995–3005. doi: 10.1002/stem.2110. [DOI] [PubMed] [Google Scholar]
  • 76.Zuk PA, et al. Human adipose tissue is a source of multipotent stem cells. Molecular biology of the cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Oh JH, et al. 2013 Neer Award: Effect of the adipose-derived stem cell for the improvement of fatty degeneration and rotator cuff healing in rabbit model. Journal of shoulder and elbow surgery / American Shoulder and Elbow Surgeons … [et al] 2014;23:445–455. doi: 10.1016/j.jse.2013.07.054. [DOI] [PubMed] [Google Scholar]
  • 78.Valencia Mora M, et al. Application of adipose tissue-derived stem cells in a rat rotator cuff repair model. Injury. 2014;45(Suppl 4):S22–27. doi: 10.1016/S0020-1383(14)70006-3. [DOI] [PubMed] [Google Scholar]
  • 79.De Bari C, et al. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis and rheumatism. 2001;44:1928–1942. doi: 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 80.Sakaguchi Y, et al. Comparison of human stem cells derived from various mesenchymal tissues: superiority of synovium as a cell source. Arthritis and rheumatism. 2005;52:2521–2529. doi: 10.1002/art.21212. [DOI] [PubMed] [Google Scholar]
  • 81.Steinert AF, et al. Characterization of bursa subacromialis-derived mesenchymal stem cells. Stem cell research & therapy. 2015;6:114. doi: 10.1186/s13287-015-0104-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Utsunomiya H, et al. Isolation and characterization of human mesenchymal stem cells derived from shoulder tissues involved in rotator cuff tears. The American journal of sports medicine. 2013;41:657–668. doi: 10.1177/0363546512473269. [DOI] [PubMed] [Google Scholar]
  • 83.Song N, et al. Multipotent mesenchymal stem cells from human subacromial bursa: potential for cell based tendon tissue engineering. Tissue engineering Part A. 2014;20:239–249. doi: 10.1089/ten.tea.2013.0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bi Y, et al. Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nature medicine. 2007;13:1219–1227. doi: 10.1038/nm1630. [DOI] [PubMed] [Google Scholar]
  • 85.Nagura I, et al. Characterization of progenitor cells derived from torn human rotator cuff tendons by gene expression patterns of chondrogenesis, osteogenesis, and adipogenesis. Journal of orthopaedic surgery and research. 2016;11:40. doi: 10.1186/s13018-016-0373-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Randelli P, et al. Isolation and characterization of 2 new human rotator cuff and long head of biceps tendon cells possessing stem cell-like self-renewal and multipotential differentiation capacity. The American journal of sports medicine. 2013;41:1653–1664. doi: 10.1177/0363546512473572. [DOI] [PubMed] [Google Scholar]
  • 87.Tao X, et al. EGR1 induces tenogenic differentiation of tendon stem cells and promotes rabbit rotator cuff repair. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology. 2015;35:699–709. doi: 10.1159/000369730. [DOI] [PubMed] [Google Scholar]
  • 88.Shen W, et al. Allogenous tendon stem/progenitor cells in silk scaffold for functional shoulder repair. Cell transplantation. 2012;21:943–958. doi: 10.3727/096368911X627453. [DOI] [PubMed] [Google Scholar]
  • 89.Lee CH, et al. Harnessing endogenous stem/progenitor cells for tendon regeneration. The Journal of clinical investigation. 2015;125:2690–2701. doi: 10.1172/JCI81589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Matsumoto K, et al. Liver organogenesis promoted by endothelial cells prior to vascular function. Science. 2001;294:559–563. doi: 10.1126/science.1063889. [DOI] [PubMed] [Google Scholar]
  • 91.Lammert E, Cleaver O, Melton D. Induction of pancreatic differentiation by signals from blood vessels. Science. 2001;294:564–567. doi: 10.1126/science.1064344. [DOI] [PubMed] [Google Scholar]
  • 92.Butler JM, et al. Endothelial cells are essential for the self-renewal and repopulation of Notch-dependent hematopoietic stem cells. Cell stem cell. 2010;6:251–264. doi: 10.1016/j.stem.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ding BS, et al. Endothelial-derived angiocrine signals induce and sustain regenerative lung alveolarization. Cell. 2011;147:539–553. doi: 10.1016/j.cell.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chirba MA, et al. FDA Regulation of Adult Stem Cell Therapies as Used in Sports Medicine. In: Stannard JP, Cook JL, Fortier LA, editors. Biologics in Orthopaedic Surgery. Thieme; New York: 2016. pp. 13–21. [DOI] [PubMed] [Google Scholar]
  • 95.Biological products regulated under Section 351 of the Public Health Services Act; implementation of biologics license; elimination of establishment license and product license; correction–FDA. Proposed rule; correction. Federal register. 1998;63:46718. [PubMed] [Google Scholar]
  • 96.. 21 U.S.C. In 21. 544
  • 97.. 42 U.S.C. In 42. 545
  • 98.. 21 U.S.C, Vol. 21. 546
  • 99.. 21 CFR. 547
  • 100.. 21 CFR. In 21, Vol. 21. 548
  • 101.. 21 CFR, Vol. 21. 549
  • 102.. 32 CFR. 550

RESOURCES