Next Article in Journal
Control of Synapse Structure and Function by Actin and Its Regulators
Next Article in Special Issue
Protective Effects of Growth Differentiation Factor-6 on the Intervertebral Disc: An In Vitro and In Vivo Study
Previous Article in Journal
Correlation between Sialylation Status and Cell Susceptibility to Amyloid Toxicity
Previous Article in Special Issue
Causes of and Molecular Targets for the Treatment of Intervertebral Disc Degeneration: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomaterials and Cell-Based Regenerative Therapies for Intervertebral Disc Degeneration with a Focus on Biological and Biomechanical Functional Repair: Targeting Treatments for Disc Herniation

1
Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
2
Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
*
Author to whom correspondence should be addressed.
Cells 2022, 11(4), 602; https://doi.org/10.3390/cells11040602
Submission received: 26 December 2021 / Revised: 22 January 2022 / Accepted: 7 February 2022 / Published: 9 February 2022
(This article belongs to the Special Issue Intervertebral Disc Degeneration and Its Regeneration)

Abstract

:
Intervertebral disc (IVD) degeneration is a common cause of low back pain and most spinal disorders. As IVD degeneration is a major obstacle to the healthy life of so many individuals, it is a major issue that needs to be overcome. Currently, there is no clinical treatment for the regeneration of degenerated IVDs. However, recent advances in regenerative medicine and tissue engineering suggest the potential of cell-based and/or biomaterial-based IVD regeneration therapies. These treatments may be indicated for patients with IVDs in the intermediate degenerative stage, a point where the number of viable cells decreases, and the structural integrity of the disc begins to collapse. However, there are many biological, biomechanical, and clinical challenges that must be overcome before the clinical application of these IVD regeneration therapies can be realized. This review summarizes the basic research and clinical trials literature on cell-based and biomaterial-based IVD regenerative therapies and outlines the important role of these strategies in regenerative treatment for IVD degenerative diseases, especially disc herniation.

1. Introduction

Intervertebral disc (IVD) degeneration is a common cause of low back pain that affects the daily life of afflicted individuals and is the cause of most spinal disorders [1,2,3,4]. Surgical treatments, such as discectomy, arthroplasty, and spinal fusion, have been widely used for the treatment of IVD diseases, including disc herniation, spinal canal stenosis, and spinal deformities. However, these methods limit spine mobility and fail to maintain the function of the treated IVD for extended periods of time [5]. Furthermore, the appearance of postoperative adjacent intervertebral disorders and reports of functional impairment are widely recognized. Meanwhile, conservative treatments for low back pain, such as the administration of analgesics, are only symptomatic therapies and do not focus on the underlying etiology [4]. In other words, there is currently no clinical treatment that can prevent or reverse IVD degeneration. Fortunately, recent tissue engineering approaches have revealed the molecular cascade involved in IVD degeneration, and treatments aimed at the regeneration of degenerated or damaged IVDs have been attempted [5]. Specifically, cell-based therapies and biomaterial-based regenerative therapies have recently attracted attention as therapeutic methods to prevent or repair IVD degeneration.
IVDs consist of an inner gel-like structure, nucleus pulposus (NP), and external annulus fibrosus (AF) [6]. The extracellular matrix (ECM) of gelatinous NP is composed of glycosaminoglycans, proteoglycans (PG), and type II collagen, which are highly hydrated [7,8]. The function of the NP is to distribute the hydraulic pressure under compressive loads [7,8]. IVD degeneration is characterized by a loss of hydration and degradation of the ECM of the NP with NP degeneration leading to overall changes in the biomechanics of the spine [5,7,8,9]. Therefore, regenerative therapy of the NP is a promising strategy to restore the function of IVDs that exhibit less advanced deformities [4,7,10,11].
Since the degree of IVD degeneration generally reflects the regenerative capacity of the disc, treatment strategies for IVD regeneration are also based on the severity and stage of IVD degeneration [12]. In the early stages of disc degeneration, the structural integrity of the disc is preserved, and there are many surviving native disc cells; therefore, biomolecular and genetic engineering interventions may have a regenerative effect [12]. In the intermediate stages of degeneration, the number of surviving native disc cells decreases, and the structural integrity of the disc begins to disintegrate [12]. Among the potential interventions, biomaterial-based therapies that maintain the disc structure and activate the remaining cells are expected to be applicable to degenerative disc diseases, such as lumbar disc herniation, which typically affects relatively young people up to the age of 40 years (Figure 1). In degenerative disc diseases, such as lumbar disc herniation combined with lumbar spinal canal stenosis, which tends to occur in people over 50 years of age, the number of remaining cells is further reduced. Therefore, cell-based therapies that repopulate the disc with healthy cells that may restore normal tissue homeostasis, in addition to the combination of biomaterials with cell therapies, are expected to be effective in these situations (Figure 1). Multiple clinical trials have been conducted to investigate cell-based and biomaterial-based therapies to treat IVD degeneration, with each trial being supported by numerous preclinical animal studies and basic science experiments [12].
In this article, we review the strategies for disc regeneration/repair therapy for IVD degeneration, with a focus on cell-based therapy and soft biomaterial-based approaches. This includes
  • cell-based IVD regeneration therapy,
  • biomaterial-based IVD regeneration therapy, and
  • disc regeneration/repair treatment for IVD herniation.
To prepare this review, 231 English-language articles were extracted from PubMed searches using the keywords “intervertebral disc degeneration” AND “regenerative therapy” without a date limitation (start of the database through October 2021). Additional keywords (biomaterials, cell transplantation, disc herniation, biomechanical, biological) were used to narrow down the papers to the topics most relevant to this review. Moreover, additional papers were obtained by analyzing papers containing excellent reviews.

2. Cell-Based IVD Regeneration Therapy: Cell Transplantation

Apoptosis of the NP cells is a characteristic phenomenon that occurs in the early stages of IVD degeneration [4,11,13,14]. NP cells play an important role in the synthesis of ECM proteins that maintain IVDs. Aging and degeneration of the IVD result in a decrease in the number of NP cells and a reduction in the production of ECM [4,13,15,16,17,18]. As the number of viable NP cells is reduced in the early to intermediate stages of disc degeneration, intradiscal cell therapy (e.g., stem cells or chondrocytes) can be used to restore normal tissue homeostasis to the disc and repopulate the disc with healthy cells (Figure 1).
There is increasing evidence that supports the use of biological and cell-based therapies for IVD degeneration. Several cell sources, including IVD-derived cells (NP-derived cells), chondrocyte-like cells, mesenchymal stem cells (MSCs), induced pluripotent stem cells, and embryonic stem cells, have been proposed and evaluated for disc regeneration therapy via cell transplantation [12,19,20] (Table 1). Several clinical trials have also been conducted using these cells for IVD degeneration [12,19,21,22,23,24] (Table 2).

2.1. Autologous IVD-Derived Cells as Therapy for IVD Regeneration

Transplantation of autologous NP-derived cells is physiologically more natural than allogenic transplantation, and autologous transplantation can avoid graft-versus-host reactions [25]. NP cells have the ability to survive in the harsh, bloodless environment of the IVD and are able to produce IVD-specific ECM [12]. However, harvesting autologous cells from a patient’s disc is invasive, and the cell yield is relatively low because the tissue source is compromised [19]. Furthermore, the ability of NP cells isolated from degenerated NPs alone is insufficient to delay further disc degeneration [25]. The co-culture of NP cells with MSCs, which allows for direct cell-to-cell contact, has been shown to significantly improve the viability of NP cells [26,27]. In a pilot clinical trial, these co-cultured cells were transplanted into a degenerated lumbar disc at a level adjacent to the fusion surgery segment. Three years post-surgery, there was no progression of disc degeneration and no back pain, suggesting the minimal efficacy needed to slow the further degeneration of human IVDs [25]. In the EuroDISC study in which percutaneous transplantation of autologous disc cells was investigated, disc chondrocytes harvested at the time of discectomy were grown in vitro and then injected into patient discs at three months postoperatively [28]. In the 2-year analysis, patients had significantly less back pain compared with that of the control patients, and NP fluid levels on magnetic resonance imaging (MRI) remained higher at the treated and adjacent discs [28].

2.2. MSC Therapy for IVD Regeneration

MSCs are the most common clinically evaluated cell type for disc regeneration therapy. MSCs are undifferentiated somatic cells that are capable of self-renewal and have the potential to differentiate into any lineage of mesenchymal origin, including chondrogenic and IVD-cell lineages, owing to their ability to differentiate into a variety of mesodermal lineages [12,81,82]. MSCs are thought to resemble perivascular and pericyte cells and are found almost everywhere in the body where vascularity is abundant [81,83]. Accordingly, autologous MSCs can be easily harvested from the bone marrow or adipose tissue [12]. Numerous studies have demonstrated that MSCs promote tissue repair and reduce inflammatory damage, while multiple preclinical animal models have been used to demonstrate the ability of MSCs to differentiate into NP cells, restore disc height and hydration, and inhibit the inflammatory cascade, leading to disc regeneration [12,26,50,67,84,85,86]. Various sources of MSCs have been identified and studied, including the bone marrow, synovial membrane, and adipose tissues [87] (Table 1). A systematic review of the safety and efficacy of MSCs for treating IVD degeneration has shown that three types of MSCs can successfully inhibit IVD degeneration, bone marrow-derived, synovial-derived, and adipose tissue-derived MSCs [87].
Both autologous and allogenic MSCs are being investigated for their ability to regenerate discs by transplanting them into degenerated discs [81]. The use of autologous MSCs as an injectable therapy has been investigated in several completed or ongoing clinical trials [12,81]. Two preliminary clinical studies have reported that transplantation of autologous bone marrow stem cells into the human lumbar disc can improve pain and other clinical outcomes, raise the level of disc stability, and increase disc hydration on MRI [51,52]. In several subsequent clinical trials, patients treated with autologous cultured bone marrow-derived MSCs for IVD degeneration showed significant improvements in pain, function, and overall subjective improvement with minimal adverse events at 4 to 6 years after treatment [58,88]. These studies have demonstrated the long-term efficacy of autologous bone-marrow-derived mesenchymal stromal cell (BM-MSC) therapy in inhibiting disease progression. However, no conclusions have been drawn regarding the optimal conditions of culturing and administrating of MSCs [24]. Although autologous cell transplantation has the lowest risk of immunogenic reactions, it has several limitations for clinical application, including a lack of shelf availability, the need to harvest tissues from patients, the time and expense of cell growth, differentiation, and selection, and the potential risk of infection [19,60,89].
On the other hand, allogeneic MSCs are typically harvested from young, healthy donors and can solve the problem of shelf availability and may produce better long-term functional outcomes as they are not subject to age-related changes and other effects based on the patient’s protoplasm that may occur with autologous cells [22,89]. Animal studies have shown that allogeneic MSCs injected into the NP region of IVDs can survive and proliferate, producing beneficial effects on IVD degeneration [48,57,59,67,89,90,91,92]. In addition, a phase I/II randomized controlled trial examining the therapeutic efficacy of allogeneic BM-MSCs for IVD degeneration confirmed the feasibility and safety of this approach for patients with IVD degeneration [59]. This trial reported rapid improvement in pain and disability for the cell therapy group compared with that for the control group, and improvement in degeneration was observed on MRI. As noted above, MSC transplantation to treat IVD degeneration is able to repair IVD degeneration in patients with low back pain, providing pain relief and functional recovery. However, MSC therapy to treat musculoskeletal disorders has not yet been approved for clinical use by the U.S. Food and Drug Administration (FDA) [78,93].

2.3. Use of Bone Marrow Aspirate Concentrate (BMAC) for IVD Regeneration Therapy

The use of BMAC has been approved by the FDA to treat musculoskeletal diseases, and its application as an IVD regeneration therapy has been reported [78,79,80]. In an animal study in which a bioresorbable alginate gel containing BMAC was implanted into rabbits of a discectomy model, the BMAC demonstrated regenerative effects on disc degeneration, comparable to that of BM-MSCs [78]. In addition, a prospective clinical trial in which BMAC was injected percutaneously into patients with lumbar disc degeneration showed that disc-derived low back pain can be reduced and disc hydration sporadically increased, demonstrating the usefulness of this treatment [79,80]. Furthermore, BMAC produced using autologous non-cultured cells can be obtained in a single step, which has the potential of cost and time-saving advantages, reduced risk of infection, and a lower chance of sample confusion compared with that of cultured autologous or allogeneic cells [78].

2.4. Problems of IVD Regeneration Therapy Using Cell-Only Transplantation

The challenges of IVD regeneration therapy using cell-only transplantation include problems of the transplanted cells adapting to the environment, cell survival, and leakage of the transplanted cells from the injection site. The environment of degenerated IVDs is unsuitable for cell viability and survival, with low oxygen levels, limited nutrition, acidic pH, and high osmolarity, which adversely affect the function and survival of the transplanted cells [65,94,95,96]. Furthermore, the risk of MSCs injected into the IVD leaking from the injection site and causing osteophyte formation has been reported in in vivo studies [45,97]. In a systematic review of MSC-based therapies for IVD degeneration, MSC-related complications were found in 2.7% of all animal models, including osteophyte formation associated with cell leakage [45,87,98].
The usefulness during cell transplantation of combining soft biomaterials as cell carriers and scaffolds, such as alginate, fibrin, atelocollagen, and hyaluronic acid, has been reported [5,26,64,68,92,99,100,101,102]. Hydrogels and other soft biomaterials are expected to not only serve as carriers to hold cells transplanted into IVDs but also act as sealants to prevent cell leakage, thereby improving biomechanical function, protecting the transplanted cells, and even activating the remaining native cells.

3. Biomaterial-Based IVD Regeneration Therapy: Soft Biomaterials Used to Regenerate Biological and Biomechanical Function

Although a number of stem cell-based therapies focusing on progenitor cell expansion and transplantation have been investigated as a means of disc regeneration therapy, there are many challenges to their clinical application, including immune rejection, pathogen infection, potential tumor formation, and host tissue engraftment [7,103,104,105,106]. On the other hand, matrix-based medicine using soft biomaterials may provide an alternative single-step process using biomaterials amenable to long-term storage, which can be used for on-demand treatment [7,107].
One goal of disc regeneration therapy is to restore the biomechanical disc function that supports the trunk and maintains mobility. In a healthy IVD, the NP compresses when an axial load is applied to the IVD and transmits the load radially to the AF [81]. Laminated AF has a high tensile strength and can expand radially in response to the added load [81]. There are few cells in the NP (4×103 cells per mm3), with NP cells accounting for only approximately 1% of the volume of IVD tissue [81,108,109,110]. The ECM produced by the NP cells is mostly composed of PG, which provides cushioning to the NP by retaining water [81]. As the IVD degradation process progresses, a decrease in PG of the NP reduces the swelling pressure of the disc, resulting in a decrease in the aggregate and instantaneous shear modulus [109,111,112]. In addition, the nucleus loses water and becomes fibrous, with the mechanical properties of the ECM being further impaired [81]. This deterioration leads to reduced flexibility of the NP and alters the loading pattern within the disc, leading to AF delamination [81,109] (Figure 2). Since the initial degeneration of IVD occurs primarily in the nucleus, gelatinous NP appears to be a promising target for therapeutic intervention (Figure 2) [109]. Therefore, tissue engineering using hydrogels and other soft biomaterials may be an alternative to current treatments.

3.1. Soft Biomaterials for NP Repair and/or Regeneration

Ideally, biomaterials for NP repair should accommodate both the biological and mechanical aspects of IVD repair and regeneration [109]. The objectives required for a soft biomaterial-based NP repair approach in terms of the biological response include the soft biomaterial
  • being biocompatible, non-toxic, and safe in vivo;
  • support cell survival;
  • promote ECM formation;
  • reduce inflammation; and
  • inhibit pathological fibrosis [113].
In terms of biomechanics, the soft biomaterial should (1) remain within the disc under in vivo loading conditions and (2) improve biomechanical disc function and spinal stability.
The concept of using biomaterials that can be injected into IVDs began in the 1960s when Nachemson et al. [114] proposed a method of injecting vulcanized silicone into degenerated discs for nucleus augmentation. Over the subsequent decades, IVD substitutes have been developed to restore disc function [115]. NP replacement using injectable, in situ curable materials can maintain immediate disc height and mechanical disc weight-bearing capacity [116,117,118,119,120] but is restricted by the risk of complications, such as implant dislocation and endplate damage, and by the limited potential for biological repair [116,117].

3.2. Biological NP Repair and/or Regeneration Using Soft Biomaterials

Many polymeric materials have been experimentally investigated for use as NP-regenerative soft biomaterials. Biomaterials are hydrogels or solid scaffolds and can be divided into synthetic biopolymers and natural biomaterials [121] (Table 3). Synthetic materials include poly (D,L-lactide) (PLA) and its derivatives, polyethylene glycol (PEG), polycarbonate urethane (PU), and poly (ε-caprolactone) (PCL), some of which can function as both hydrogels and solid scaffolds [38,63,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155]. Synthetic hydrogels consist of polymer networks that can absorb a large amount of water, are easy to modify, and can be consistently and highly tunable [121]. However, most production processes of synthetic hydrogels involve the use of reactive reagents and require the complete removal of contaminants and unreacted reagents [115,156]. In comparison, natural polymer-based biomaterials mainly include hydrogels, such as alginate, agarose, fibrin, hyaluronic, collagen, chitosan, and carboxymethylcellulose [4,5,7,39,41,49,57,64,65,66,78,90,92,100,101,116,121,122,128,132,140,142,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229,230,231,232,233] (Table 3). These natural hydrogels have been extensively studied for NP tissue engineering due to their excellent biocompatibility and biological activity and their participation in the physiological turnover process [116,122,234]. A number of in vitro studies have shown that these hydrogels support cell survival and induce differentiation of residual NP disc cells and stem cells [132,142,158,159,160,163,169,188,197,203,207,208,232,235,236,237,238].
To achieve intrinsic and sustainable disc regeneration, soft biomaterials are required to support cell survival and induce in vivo differentiation of the transplanted stem cells and remaining disc cells. Hydrogels, such as collagen gel (atelocollagen), hyaluronic acid, fibrin, peptide hydrogel, polysaccharide hydrogel, and alginate, have been reported in in vivo studies to be useful as cell carriers for cell transplantation and disc regeneration therapy [26,57,60,65,68,78,92,196,239]. Degenerated discs present harsh microenvironments characterized by hypoxia, hypotrophy, acidic pH, high mechanical loading, high osmotic pressure, and a complex network of various proteases and cytokines [95,113,240,241,242,243,244]. Meanwhile, biomaterials incorporate cells into the scaffold to increase their viability, act as protective carriers to prevent the leakage of the cells from the site, and also support the transmission of mechanical loading [157]. In addition, several in vivo studies of IVD regeneration with cell-free biomaterials using hydrogels alone have reported the regenerative potential of fibrin sealant, polyglycolic acid (PGA)-hyaluronic acid scaffold, and collagen-based scaffold through hydrolysis with actinidin protease and ultra-purified alginate (UPAL) gel [7,101,107,125,192,204,220].

3.3. Mechanism of IVD Regeneration Therapy Using Cell-Free Soft Biomaterials Alone

Considering the various issues related to the clinical application of cell transplantation therapy, biological disc regeneration using cell-free soft biomaterials alone may be a new alternative to the current treatment for disc degeneration disease and ideally involve a single-step process [7,107]. For instance, there has been much interest in bioengineering approaches in recent years that exploit endogenous cell populations to restore the structure and function of IVDs, with the potential for IVD repair using cell-free soft biomaterials being promising [99,220]. Several in vivo studies have shown that various soft biomaterials have the potential to regenerate IVD tissue by supporting the survival and activation of remaining disc cells in damaged or degenerated IVDs and by promoting ECM production. However, details regarding their repair mechanisms have not yet been fully elucidated.
Several biomaterials have been analyzed in in vivo experiments with respect to their mechanisms in inducing and activating residual disc cells (Table 4). For instance, an in vivo rabbit study of IVD aspiration followed by alginate-based hydrogel called UPAL gel implantation revealed a significant increase in the percentage of GD2Tie2 cells [7,92], which are NP progenitor cells [245]. This indicated that the implanted biomaterial was able to induce endogenous NP cells and NP progenitor cells, leading to endogenous IVD repair [7]. Similar to the UPAL gel results, a collagen type 1-based scaffold called low adhesive scaffold collagen (LASCol) promotes internal migration of the remaining disc NP cells when implanted after discectomy of rat caudal IVDs [220]. Furthermore, it has been shown that LASCol promotes the formation of cell aggregative spheroids that facilitate the maintenance of the original disc NP phenotype, upregulates the expression of chondrogenic genes, and promotes disc tissue repair [220].
Biomaterials affecting the expression of various cytokines in damaged discs have also been reported as a mechanism of biomaterial-induced disc repair. For instance, fibrin injection (fibrin sealant) after discectomy of porcine IVD has been shown to suppress acute production of proinflammatory cytokines TNF-α, IL-1β, and IL-6, increase the expression of pro-resolution cytokines IL-4 and TGF-β, and inhibit discectomy-induced progressive fibrosis of NP [192]. Furthermore, hyaluronan treatment after rat tail disc injury regulates inflammation by downregulating IFNα, reduces cell death by suppressing the expression of IGFBP3 and caspase-3 fragment p17, and induces the production of ECM [205].

3.4. Effects of Biomaterials on Reduction in Pain Related to Damaged IVDs

The goal of biomaterial-based IVD therapy is to not only inhibit tissue degeneration but also to control the pain caused by disc injury and degeneration. Inflammation within the lumbar IVD is often a key factor in acute low back pain [168,247,248]. Intradiscal inflammation and sensory nerve ingrowth into the deep inner layers of the AF cause discogenic pain during the chronic phase of IVD damage and degeneration [168,249]. Several types of soft biomaterials proposed as candidates for IVD repair have been shown to inhibit inflammatory cytokines in IVDs and are expected to reduce pain. Recently, it was reported in an in vivo rat IVD injury model for which methods evaluating pain-related behavior were established that hydrogels suppress pain [168,209]. Meanwhile, implantation of a hydrogel (hyaluronic acid hydrogel and UPAL gel) in a rat caudal NP punch model inhibited nociceptive behavior in Hargreaves, von Fley, and tail-flick tests [168,209]. The following possible mechanism of the hydrogel effect in the IVD injury-induced pain model has been reported. First, hydrogels implanted into injured discs of rats have been shown to regulate inflammation by inhibiting the downstream signaling cascade that activates nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) by downregulating IL-6 and IL-β and by inhibiting their binding to receptors [209]. Second, discogenic pain in the chronic phase is caused by an increased expression of nerve growth factor (NGF) that is induced by proinflammatory cytokines and the binding of NGF to its high-affinity receptor, tyrosine kinase A (TrkA), which promotes neoinnervation of the IVD and local inflammation [168,250,251,252]. Third, hydrogel treatment of injured discs has been shown to suppress neurotrophic factors, such as NGF, and reduce NGF-TrkA binding, which mediates inhibition of neurite outgrowth of sensory nerves in the discs, resulting in reduced pain-related behavior in rats [168,209]. Finally, hydrogel treatment of damaged IVDs is expected to have a palliative effect on acute IVD pain after discectomy, as well as a preventive effect on discogenic pain [168].

3.5. Biomechanical Evaluation of Soft Biomaterials for NP Repair and/or Regeneration

From a mechanical perspective, soft biomaterials for use in NP treatment should ideally mimic the material properties of NP and withstand physiological loading conditions in order to restore their biomechanical properties [109]. The water content of NP is >85% by weight in juveniles, decreasing to approximately 70–75% in adults, and further decreases with additional aging and degeneration [253,254,255]. The swelling stress and effective, cohesive modulus of non-denatured human NPs in constrained compression tests are 0.138 MPa and 1.01 MPa, respectively [112], and the complex modulus of NP is 5.82 kPa at 1 rad per second, 10% compressive strain in torsional shear tests of the viscoelastic shear properties of NP [109,111]. There have been many in vitro studies on soft biomaterials that mimic the mechanical properties of native NP tissues, including alginate hydrogel, collagen gel, hyaluronic acid hydrogel, and polyethylene glycol hydrogel, among others [115,173,211,256]. It has been shown that these materials exhibit biomechanical properties comparable to those of NPs, such as water content, stiffness, and viscoelastic properties, making them candidate materials for use in NP therapy. These candidate soft biomaterials for NP treatment were first evaluated in vitro and subsequently in situ using ex vivo or in vivo preclinical animal models; however, no consensus has yet been established regarding their biomechanical evaluation as functional spinal units [109,257]. This may change as biomechanical evaluation methods have been proposed to establish best practices for screening the performance of newly developed hydrogel formulations and ensure that these materials meet minimum feasibility benchmarks for translation [257].
In general, biomechanical analysis should include (1) evaluation of the effect of hydrogel on disc function repair for axial, torsional, and viscoelastic motion segment responses and (2) evaluation of durability, mechanical feasibility, and the associated herniation risk [257]. An ex vivo approach using cadaveric animal/human motion segments can be used to investigate the biomechanical suitability of the material(s) under study [109]. Motion segments have been tested under uniaxial compression, lateral bending, and flexion/extension as a biomechanical evaluation of IVD [242]. Meanwhile, the axial compressive properties of IVDs are usually investigated in vertebra-disc-vertebra specimens of the lumbar spine, with the load-displacement curve showing a nonlinear viscoelastic response [242]. In other experiments using uniaxial compression, creep, stress relaxation, vibration/dynamic compression, and high load factor properties have been evaluated [242]. As IVDs are subjected to complex three-dimensional loading in vivo, they should be evaluated on a mechanical spine tester that can apply various combinations of cyclic compression, bending, and torsion to spinal segments ex vivo [242]. Typical moment–rotation graphs reveal marked nonlinearity and hysteresis and can be used to evaluate stiffness, the neutral zone, and range of motion. A setup using six degrees of freedom provides insight into the resulting range of motion and its restoration to previous values [109,242]. Currently, several ex vivo and in vivo studies have reported that soft biomaterials, including alginate, hyaluronic acid, chitosan-based hydrogels, and fibrin, are able to restore biomechanical disc functions, such as stiffness and range of motion, after disc implantation [7,192,215,258].
In contrast, it has been reported that hydrogel injected into IVDs may extrude out of the disc in vivo, with no improvement in biomechanical evaluation [206]. Therefore, to apply hydrogel candidates to preclinical animal models and clinical trials, it is very important to determine in situ IVD repair, a configuration for which there is currently no document to guide the evaluation of the development of new hydrogel systems for IVD treatment, including the evaluation of functional outcomes, such as implant herniation risk and structural durability [257].
Herniation risk following IVD repair has been assessed using a cyclic axial loading test and displacement-controlled ramp-to-failure test [7,230,233,257,259,260,261,262,263]. In particular, the ramp-to-failure test is designed to evaluate the worst-case IVD motion segment failure characteristics and hydrogel sealing properties as the motion segment is compressed with five degrees of side-bending and NP displacement is induced in the radial direction of the hydrogel [257]. Fatigue endurance testing has also been performed using a fatigue loading protocol established by Wilke et al. [264]. In this test, called the hula hoop test, the IVD motion segment is subjected to cyclic eccentric compression at an offset that induces a physiological bending moment until failure is reached, with NP extrusion being examined and flexibility testing being performed [233,257,264]. When assessing the risk of implant herniation and structural durability, the degree of biomechanical recovery should be assessed by comparing the motion segment of the repaired IVD with that of the intact IVD, and the biomechanical non-inferiority or superiority relative to standard treatment should be demonstrated by comparison with an IVD injury model that simulates discectomy [257].

3.6. Clinical Trial of Soft Biomaterials for Treating IVD Degeneration

As noted above, numerous in vitro and in vivo experiments on NP regeneration therapy based on using soft biomaterials have been performed, but very few human clinical trials have investigated the use of biomaterials alone or as a cell scaffold or delivery system for IVD regeneration. [24,113]. One of the clinical trials in which fibrin sealant was injected into the IVDs of patients with discogenic low back pain reported on the safety of the treatment with significant improvement in pain and function at a 24-month follow-up [195]. Another preliminary study showed that collagen sponges containing autologous BM-MSCs that were percutaneously transplanted into the discs of two patients resulted in improved hydration and motion segment instability of the degenerated discs and improved low back pain at two years postoperative [51]. Using a fibrin carrier in a clinical trial of 15 patients with lumbar spondylolisthesis associated with mechanical low back pain, allogeneic juvenile chondrocytes were percutaneously injected into degenerated IVDs, resulting in no apparent side effects at 12-month follow-up and significant improvement in disability and pain scores, with 77% of the patients showing improvement on MRI [39]. In a phase I study in which a combination of hyaluronic acid derivatives and autologous adipose tissue MSCs was percutaneously injected into the discs of 10 patients with chronic discogenic low back pain, there were no serious adverse events during a one-year follow-up period, and the patients showed significant improvement in visualized analog scale (VAS) and Oswestry Disability Index (ODI) scores for pain, as well as improved disc hydration on diffusion MRI [64].

4. Disc Regeneration and/or Repair Treatment for IVD Herniation

One of the aforementioned targets for NP regeneration therapy is lumbar disc herniation, which is one of the main causes of back pain and is a psychological burden [168,265,266]. A discectomy for a herniated disc relieves pain by removing the NP through fissures in the AF, which relieves nerve compression. However, this procedure does not aim to repair defects in the NP or AF, and the defect within the IVD produced by discectomy can lead to undesirable postoperative outcomes, including further disc degeneration, chronic low back pain, and recurrent herniation [168,257]. To compensate for this defect, disc reparative therapy using soft biomaterials may be useful, as patients with lumbar disc herniation are typically relatively young (<45 years old), and disc cells are expected to remain in these individuals [7,267,268] (Figure 1). However, no suitable biomaterial has been developed to date to replace NP tissue removed after discectomy [168].
Biomaterials for NP regeneration and/or repair after discectomy should not only function to biologically protect cells and promote tissue repair but should also have the following biomechanical functions to address the NP and AF defects. First, the biomaterial should fill the defective area of the NP and AF, have adhesive properties, and not extrude under in vivo loading conditions (Figure 3). Second, the biomaterial should provide structural support in order to restore biological and biomechanical function to the damaged tissues.

4.1. Adhesive Function of Soft Biomaterials after Discectomy or AF Injury

Ex vivo and in vivo studies using AF injury models have demonstrated the usefulness of soft biomaterials, such as fibrin, collagen, hyaluronic acid, and alginate-based hydrogels, for IVD regenerative therapy [7,129,140,186,190,192,193,194,205,223,257,260,262,263,269,270,271,272,273,274,275,276]. These hydrogels have the ability to adhere to tissues and allow for in situ repair of disc defects. The adhesion mechanism of hydrogels to tissues can be explained through three modalities, mechanical interlocking, electrostatic interactions, and chemical interactions [257,277,278].
Mechanical interlocking occurs via the biophysical phenomenon in which the roughness of an adherend surface causes the hydrogel to “mate” with the surface irregularities and adhere to the tissue surface [257,278,279]. Meanwhile, electrostatic interaction is a force at the molecular level in which the asymmetrical distribution of attachment surfaces due to differences in the electronegativity of atoms creates partial positive and negative charges between the attachment surfaces, which attract each other making it difficult to functionally separate them [257,279]. Finally, chemical interactions arise from biochemical phenomena acting at the atomic or molecular level and are characterized by diffusion, physisorption, and chemisorption [257,280]. For instance, constituent polymers of the hydrogel network and the biopolymers of the tissue interpenetrate each other at the interface, and diffusion occurs between the polymer adhesive and the adherend [257,281]. In comparison, physisorption is the adhesion of a biomaterial to a tissue substrate by non-covalent intermolecular interactions caused by hydrogen bonding and van der Waals forces at the interface [257,282,283]. Chemisorption is the adhesion of a hydrogel to a tissue substrate by means of multiple types of covalent bonds, such as imine bonds, amide bonds, urea bonds, N-N bonds of hydrazine derivatives, and disulfide bridges, which exist between the adhesive hydrogel and the tissue [257,278,282].
Each type of hydrogel exhibits its own physical and chemical interactions with biopolymers on the tissue surface, and the mechanism of adhesion to IVD tissue differs depending on the hydrogel formulation [257]. The main mechanisms of hydrogel adhesion for IVD repair are proposed to be chemisorption for fibrin-based and collagen-based hydrogels [192,222,257,275], and electrostatic interaction, physisorption, and diffusion for hyaluronic acid-based and alginate-based hydrogels [7,140,205,257].

4.2. Evaluation of Biomechanical and Biological Regeneration by Soft Biomaterials in IVD after Discectomy or AF Injury

Candidate biomaterials for IVD repair should be evaluated in ex vivo studies for mechanical feasibility and the associated risk of herniation after in situ application (Figure 3). As noted previously, the risk of herniation after IVD repair can be assessed using cyclic axial loading tests and displacement-controlled ramp-to-failure tests [257]. Fibrin (genipin cross-linked fibrin), collagen hydrogel (riboflavin cross-linked collagen, Rose Bengal cross-linked collagen), alginate (UPAL), chitosan, and cellulose have been shown to improve IVD failure properties and to retain hydrogels after biomechanical loading in ex vivo biomechanical studies, suggesting their clinical usefulness for IVD repair [7,186,192,223,230,233,257,262,269,270,271,272,273] (Table 5).
Histological evaluation in an in vivo AF injury/discectomy model has demonstrated the effect of several soft biomaterials on IVD tissue repair. Meanwhile, treatment with hydrogels, such as fibrin, collagen-based hydrogels (riboflavin-cross-linked collagen and citric acid-1-ethyl-3-(3-dimethylaminopropyl)carbodiimide/N-hydroxysuccinimide collagen gel), hyaluronic acid, and UPAL, has been reported to maintain the IVD structure, retain NP tissue, and cause less degeneration compared with that in disc-injury control groups [7,140,192,222,257,274,275,276,284]. Among the candidate hydrogels, hyaluronic acid-based gels have not been proven to have biomechanical functions in an AF injury model. However, it has been reported for NP augmentation and AF repair that the combination of hyaluronic acid gel with photo-crosslinked collagen gel or fibrin gel prevents disc degeneration in vivo after discectomy [204,223]. These findings indicate a potential need for a combined strategy when it comes to using biomaterials to inhibit disc degeneration after IVD herniation surgery.

4.3. Clinical Application of Soft Biomaterial Therapy for IVD Herniation

As previously noted, clinical trials of soft biomaterials for the treatment of disc degeneration are currently limited. In addition, all the clinical trials have been performed using intradiscal injection therapy for patients with no AF defects, and there have been no clinical applications of soft biomaterials for disc repair therapy after discectomy. That noted, a first human pilot study using an acellular bioresorbable UPAL gel in patients with lumbar disc herniation has been conducted [246]. In a preclinical study using a large-animal discectomy model, the UPAL gel not only exhibited sufficient biomechanical properties without protrusion but also histologically promoted disc repair [7]. UPAL gel can be suitable for various shapes of post-discectomy defects while reducing the risk of gel extrusion because the alginate gel can be rapidly cured by covering the AF surface with CaCl2 [7,78,92,168]. Based on current findings, UPAL gel is expected to be clinically applied as a soft biomaterial to safely promote disc repair after disc herniation surgery.

5. Conclusions

IVD degenerative disease, which can cause back pain and neurological disorders, is a major obstacle to independent and healthy living by afflicted individuals and is an important issue that needs to be addressed and overcome. However, there is still no effective treatment that can reduce or restore IVD degenerative changes or even alter the course of disease progression. A number of studies have been performed on cell-based IVD regeneration therapies, mainly those using stem cells and biomaterial-based IVD regeneration therapies using tissue engineering techniques. Fortunately, there have been reports of their usefulness as IVD tissue regeneration therapies. In addition, multiple clinical trials evaluating cell therapies, biomaterial therapies, and combinations of these therapies are currently being conducted. In other words, these therapies have the opportunity to become a new and unprecedented treatment for IVD degeneration.
However, many challenges need to be overcome for these regenerative therapies to become clinically practical. With regard to cell-based therapy, there are issues such as the selection of cell types suitable for IVD regeneration therapy, securing quality cells with high proliferative and differentiation potential, problems with cell aging, and securing a sufficient number of cells. Biomaterial-based therapy requires further improvements in biocompatibility and biomechanical functions described in this review, in addition to in vivo stability, biodegradability, and non-immunogenicity. Cost and clinical safety are also important issues for both treatments. Furthermore, the proper indications and patients for these new therapies, as well as the timing of their introduction, need to be fully discussed. Further basic research and preclinical and clinical trials are needed in the future to resolve these issues. With the recent technological innovations in regenerative medicine and tissue engineering, it is expected that IVD regeneration therapy will overcome the current biological, biomechanical, and clinical limitations and ultimately achieve significant improvements in daily activities and quality of life for patients suffering from IVD degenerative diseases.

Author Contributions

Conceptualization, K.Y. and H.S.; Original draft preparation, K.Y. and H.S.; writing-review and editing, H.S. and N.I. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by financial support as follows: “Project for Regenerative/Cellular Medicine and Gene Therapies” at the Japan Agency for Medical Research and Development, AMED (JP21bm0404068h0002) and Grants-in-aid from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (21H03313). The sponsor of the study had no role in report preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marras, W.S.; Ferguson, S.A.; Burr, D.; Schabo, P.; Maronitis, A. Low back pain recurrence in occupational environments. Spine 2007, 32, 2387–2397. [Google Scholar] [CrossRef]
  2. Di Iorio, A.; Abate, M.; Guralnik, J.M.; Bandinelli, S.; Cecchi, F.; Cherubini, A.; Corsonello, A.; Foschini, N.; Guglielmi, M.; Lauretani, F.; et al. From chronic low back pain to disability, a multifactorial mediated pathway: The InCHIANTI study. Spine 2007, 32, E809–E815. [Google Scholar] [CrossRef]
  3. Mielenz, T.J.; Garrett, J.M.; Carey, T.S. Association of psychosocial work characteristics with low back pain outcomes. Spine 2008, 33, 1270–1275. [Google Scholar] [CrossRef]
  4. Yamada, K.; Sudo, H.; Iwasaki, K.; Sasaki, N.; Higashi, H.; Kameda, Y.; Ito, M.; Takahata, M.; Abumi, K.; Minami, A.; et al. Caspase 3 silencing inhibits biomechanical overload-induced intervertebral disk degeneration. Am. J. Pathol. 2014, 184, 753–764. [Google Scholar] [CrossRef]
  5. Priyadarshani, P.; Li, Y.; Yao, L. Advances in biological therapy for nucleus pulposus regeneration. Osteoarthr. Cartil. 2016, 24, 206–212. [Google Scholar] [CrossRef] [Green Version]
  6. Urban, J.P.; Roberts, S. Degeneration of the intervertebral disc. Arthritis Res. Ther. 2003, 5, 120–130. [Google Scholar] [CrossRef] [Green Version]
  7. Tsujimoto, T.; Sudo, H.; Todoh, M.; Yamada, K.; Iwasaki, K.; Ohnishi, T.; Hirohama, N.; Nonoyama, T.; Ukeba, D.; Ura, K.; et al. An acellular bioresorbable ultra-purified alginate gel promotes intervertebral disc repair: A preclinical proof-of-concept study. eBioMedicine 2018, 37, 521–534. [Google Scholar] [CrossRef] [Green Version]
  8. Frith, J.E.; Cameron, A.R.; Menzies, D.J.; Ghosh, P.; Whitehead, D.L.; Gronthos, S.; Zannettino, A.C.; Cooper-White, J.J. An injectable hydrogel incorporating mesenchymal precursor cells and pentosan polysulphate for intervertebral disc regeneration. Biomaterials 2013, 34, 9430–9440. [Google Scholar] [CrossRef]
  9. Hunter, C.J.; Matyas, J.R.; Duncan, N.A. The notochordal cell in the nucleus pulposus: A review in the context of tissue engineering. Tissue Eng. 2003, 9, 667–677. [Google Scholar] [CrossRef] [Green Version]
  10. Sudo, H.; Minami, A. Caspase 3 as a therapeutic target for regulation of intervertebral disc degeneration in rabbits. Arthritis Rheum. 2011, 63, 1648–1657. [Google Scholar] [CrossRef]
  11. Sudo, H.; Minami, A. Regulation of apoptosis in nucleus pulposus cells by optimized exogenous Bcl-2 overexpression. J. Orthop. Res. 2010, 28, 1608–1613. [Google Scholar] [CrossRef]
  12. Ju, D.G.; Kanim, L.E.; Bae, H.W. Intervertebral Disc Repair: Current Concepts. Glob. Spine J. 2020, 10, 130–136. [Google Scholar] [CrossRef]
  13. Park, J.B.; Lee, J.K.; Park, S.J.; Kim, K.W.; Riew, K.D. Mitochondrial involvement in fas-mediated apoptosis of human lumbar disc cells. J. Bone Jt. Surg. Am. 2005, 87, 1338–1342. [Google Scholar]
  14. Gruber, H.E.; Hanley, E.N., Jr. Analysis of aging and degeneration of the human intervertebral disc. Comparison of surgical specimens with normal controls. Spine 1998, 23, 751–757. [Google Scholar] [CrossRef]
  15. Kim, K.W.; Ha, K.Y.; Lee, J.S.; Rhyu, K.W.; An, H.S.; Woo, Y.K. The apoptotic effects of oxidative stress and antiapoptotic effects of caspase inhibitors on rat notochordal cells. Spine 2007, 32, 2443–2448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Lotz, J.C.; Chin, J.R. Intervertebral disc cell death is dependent on the magnitude and duration of spinal loading. Spine 2000, 25, 1477–1483. [Google Scholar] [CrossRef] [PubMed]
  17. Buckwalter, J.A. Aging and degeneration of the human intervertebral disc. Spine 1995, 20, 1307–1314. [Google Scholar] [CrossRef]
  18. Rannou, F.; Lee, T.S.; Zhou, R.H.; Chin, J.; Lotz, J.C.; Mayoux-Benhamou, M.A.; Barbet, J.P.; Chevrot, A.; Shyy, J.Y. Intervertebral disc degeneration: The role of the mitochondrial pathway in annulus fibrosus cell apoptosis induced by overload. Am. J. Pathol. 2004, 164, 915–924. [Google Scholar] [CrossRef]
  19. Sakai, D.; Schol, J. Cell therapy for intervertebral disc repair: Clinical perspective. J. Orthop. Translat. 2017, 9, 8–18. [Google Scholar] [CrossRef]
  20. Tessier, S.; Risbud, M.V. Understanding embryonic development for cell-based therapies of intervertebral disc degeneration: Toward an effort to treat disc degeneration subphenotypes. Dev. Dyn. 2021, 250, 302–317. [Google Scholar] [CrossRef]
  21. Thorpe, A.A.; Bach, F.C.; Tryfonidou, M.A.; Le Maitre, C.L.; Mwale, F.; Diwan, A.D.; Ito, K. Leaping the hurdles in developing regenerative treatments for the intervertebral disc from preclinical to clinical. JOR Spine 2018, 1, e1027. [Google Scholar] [CrossRef] [PubMed]
  22. Oehme, D.; Goldschlager, T.; Ghosh, P.; Rosenfeld, J.V.; Jenkin, G. Cell-Based Therapies Used to Treat Lumbar Degenerative Disc Disease: A Systematic Review of Animal Studies and Human Clinical Trials. Stem Cells Int. 2015, 2015, 946031. [Google Scholar] [CrossRef] [PubMed]
  23. Sakai, D.; Andersson, G.B. Stem cell therapy for intervertebral disc regeneration: Obstacles and solutions. Nat. Rev. Rheumatol. 2015, 11, 243–256. [Google Scholar] [CrossRef]
  24. Binch, A.L.A.; Fitzgerald, J.C.; Growney, E.A.; Barry, F. Cell-based strategies for IVD repair: Clinical progress and translational obstacles. Nat. Rev. Rheumatol. 2021, 17, 158–175. [Google Scholar] [CrossRef]
  25. Mochida, J.; Sakai, D.; Nakamura, Y.; Watanabe, T.; Yamamoto, Y.; Kato, S. Intervertebral disc repair with activated nucleus pulposus cell transplantation: A three-year, prospective clinical study of its safety. Eur. Cell Mater. 2015, 29, 202–212; discussion 212. [Google Scholar] [CrossRef] [PubMed]
  26. Sakai, D.; Mochida, J.; Iwashina, T.; Hiyama, A.; Omi, H.; Imai, M.; Nakai, T.; Ando, K.; Hotta, T. Regenerative effects of transplanting mesenchymal stem cells embedded in atelocollagen to the degenerated intervertebral disc. Biomaterials 2006, 27, 335–345. [Google Scholar] [CrossRef]
  27. Yamamoto, Y.; Mochida, J.; Sakai, D.; Nakai, T.; Nishimura, K.; Kawada, H.; Hotta, T. Upregulation of the viability of nucleus pulposus cells by bone marrow-derived stromal cells: Significance of direct cell-to-cell contact in coculture system. Spine 2004, 29, 1508–1514. [Google Scholar] [CrossRef]
  28. Meisel, H.J.; Ganey, T.; Hutton, W.C.; Libera, J.; Minkus, Y.; Alasevic, O. Clinical experience in cell-based therapeutics: Intervention and outcome. Eur. Spine J. 2006, 15 (Suppl. 3), 397–405. [Google Scholar] [CrossRef] [Green Version]
  29. Okuma, M.; Mochida, J.; Nishimura, K.; Sakabe, K.; Seiki, K. Reinsertion of stimulated nucleus pulposus cells retards intervertebral disc degeneration: An in vitro and in vivo experimental study. J. Orthop. Res. 2000, 18, 988–997. [Google Scholar] [CrossRef]
  30. Watanabe, K.; Mochida, J.; Nomura, T.; Okuma, M.; Sakabe, K.; Seiki, K. Effect of reinsertion of activated nucleus pulposus on disc degeneration: An experimental study on various types of collagen in degenerative discs. Connect. Tissue Res. 2003, 44, 104–108. [Google Scholar] [CrossRef]
  31. Ganey, T.; Libera, J.; Moos, V.; Alasevic, O.; Fritsch, K.G.; Meisel, H.J.; Hutton, W.C. Disc chondrocyte transplantation in a canine model: A treatment for degenerated or damaged intervertebral disc. Spine 2003, 28, 2609–2620. [Google Scholar] [CrossRef] [PubMed]
  32. Gruber, H.E.; Johnson, T.L.; Leslie, K.; Ingram, J.A.; Martin, D.; Hoelscher, G.; Banks, D.; Phieffer, L.; Coldham, G.; Hanley, E.N., Jr. Autologous intervertebral disc cell implantation: A model using Psammomys obesus, the sand rat. Spine 2002, 27, 1626–1633. [Google Scholar] [CrossRef] [PubMed]
  33. Huang, B.; Zhuang, Y.; Li, C.Q.; Liu, L.T.; Zhou, Y. Regeneration of the intervertebral disc with nucleus pulposus cell-seeded collagen II/hyaluronan/chondroitin-6-sulfate tri-copolymer constructs in a rabbit disc degeneration model. Spine 2011, 36, 2252–2259. [Google Scholar] [CrossRef]
  34. Hohaus, C.; Ganey, T.M.; Minkus, Y.; Meisel, H.J. Cell transplantation in lumbar spine disc degeneration disease. Eur. Spine J. 2008, 17 (Suppl. 4), 492–503. [Google Scholar] [CrossRef] [Green Version]
  35. Iwashina, T.; Mochida, J.; Sakai, D.; Yamamoto, Y.; Miyazaki, T.; Ando, K.; Hotta, T. Feasibility of using a human nucleus pulposus cell line as a cell source in cell transplantation therapy for intervertebral disc degeneration. Spine 2006, 31, 1177–1186. [Google Scholar] [CrossRef] [PubMed]
  36. Meisel, H.J.; Siodla, V.; Ganey, T.; Minkus, Y.; Hutton, W.C.; Alasevic, O.J. Clinical experience in cell-based therapeutics: Disc chondrocyte transplantation A treatment for degenerated or damaged intervertebral disc. Biomol. Eng. 2007, 24, 5–21. [Google Scholar] [CrossRef] [PubMed]
  37. Nomura, T.; Mochida, J.; Okuma, M.; Nishimura, K.; Sakabe, K. Nucleus pulposus allograft retards intervertebral disc degeneration. Clin. Orthop. Relat. Res. 2001, 389, 94–101. [Google Scholar] [CrossRef]
  38. Ruan, D.K.; Xin, H.; Zhang, C.; Wang, C.; Xu, C.; Li, C.; He, Q. Experimental intervertebral disc regeneration with tissue-engineered composite in a canine model. Tissue Eng. Part. A 2010, 16, 2381–2389. [Google Scholar] [CrossRef]
  39. Coric, D.; Pettine, K.; Sumich, A.; Boltes, M.O. Prospective study of disc repair with allogeneic chondrocytes presented at the 2012 Joint Spine Section Meeting. J. Neurosurg. Spine 2013, 18, 85–95. [Google Scholar] [CrossRef]
  40. Gorensek, M.; Jaksimovic, C.; Kregar-Velikonja, N.; Gorensek, M.; Knezevic, M.; Jeras, M.; Pavlovcic, V.; Cor, A. Nucleus pulposus repair with cultured autologous elastic cartilage derived chondrocytes. Cell Mol. Biol. Lett. 2004, 9, 363–373. [Google Scholar]
  41. Acosta, F.L., Jr.; Metz, L.; Adkisson, H.D.; Liu, J.; Carruthers-Liebenberg, E.; Milliman, C.; Maloney, M.; Lotz, J.C. Porcine intervertebral disc repair using allogeneic juvenile articular chondrocytes or mesenchymal stem cells. Tissue Eng. Part. A 2011, 17, 3045–3055. [Google Scholar] [CrossRef] [Green Version]
  42. Omlor, G.W.; Bertram, H.; Kleinschmidt, K.; Fischer, J.; Brohm, K.; Guehring, T.; Anton, M.; Richter, W. Methods to monitor distribution and metabolic activity of mesenchymal stem cells following in vivo injection into nucleotomized porcine intervertebral discs. Eur. Spine J. 2010, 19, 601–612. [Google Scholar] [CrossRef] [Green Version]
  43. Sakai, D.; Mochida, J.; Yamamoto, Y.; Nomura, T.; Okuma, M.; Nishimura, K.; Nakai, T.; Ando, K.; Hotta, T. Transplantation of mesenchymal stem cells embedded in Atelocollagen gel to the intervertebral disc: A potential therapeutic model for disc degeneration. Biomaterials 2003, 24, 3531–3541. [Google Scholar] [CrossRef]
  44. Serigano, K.; Sakai, D.; Hiyama, A.; Tamura, F.; Tanaka, M.; Mochida, J. Effect of cell number on mesenchymal stem cell transplantation in a canine disc degeneration model. J. Orthop. Res. 2010, 28, 1267–1275. [Google Scholar] [CrossRef]
  45. Vadala, G.; Sowa, G.; Hubert, M.; Gilbertson, L.G.; Denaro, V.; Kang, J.D. Mesenchymal stem cells injection in degenerated intervertebral disc: Cell leakage may induce osteophyte formation. J. Tissue Eng. Regen. Med. 2012, 6, 348–355. [Google Scholar] [CrossRef]
  46. Wei, A.; Tao, H.; Chung, S.A.; Brisby, H.; Ma, D.D.; Diwan, A.D. The fate of transplanted xenogeneic bone marrow-derived stem cells in rat intervertebral discs. J. Orthop. Res. 2009, 27, 374–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Yang, F.; Leung, V.Y.; Luk, K.D.; Chan, D.; Cheung, K.M. Mesenchymal stem cells arrest intervertebral disc degeneration through chondrocytic differentiation and stimulation of endogenous cells. Mol. Ther. 2009, 17, 1959–1966. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, Y.G.; Guo, X.; Xu, P.; Kang, L.L.; Li, J. Bone mesenchymal stem cells transplanted into rabbit intervertebral discs can increase proteoglycans. Clin. Orthop. Relat. Res. 2005, 430, 219–226. [Google Scholar] [CrossRef]
  49. Allon, A.A.; Aurouer, N.; Yoo, B.B.; Liebenberg, E.C.; Buser, Z.; Lotz, J.C. Structured coculture of stem cells and disc cells prevent disc degeneration in a rat model. Spine J. 2010, 10, 1089–1097. [Google Scholar] [CrossRef] [Green Version]
  50. Feng, G.; Zhao, X.; Liu, H.; Zhang, H.; Chen, X.; Shi, R.; Liu, X.; Zhao, X.; Zhang, W.; Wang, B. Transplantation of mesenchymal stem cells and nucleus pulposus cells in a degenerative disc model in rabbits: A comparison of 2 cell types as potential candidates for disc regeneration. J. Neurosurg. Spine 2011, 14, 322–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Yoshikawa, T.; Ueda, Y.; Miyazaki, K.; Koizumi, M.; Takakura, Y. Disc regeneration therapy using marrow mesenchymal cell transplantation: A report of two case studies. Spine 2010, 35, 475–480. [Google Scholar] [CrossRef] [PubMed]
  52. Orozco, L.; Soler, R.; Morera, C.; Alberca, M.; Sanchez, A.; Garcia-Sancho, J. Intervertebral disc repair by autologous mesenchymal bone marrow cells: A pilot study. Transplantation 2011, 92, 822–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yang, H.; Wu, J.; Liu, J.; Ebraheim, M.; Castillo, S.; Liu, X.; Tang, T.; Ebraheim, N.A. Transplanted mesenchymal stem cells with pure fibrinous gelatin-transforming growth factor-beta1 decrease rabbit intervertebral disc degeneration. Spine J. 2010, 10, 802–810. [Google Scholar] [CrossRef] [PubMed]
  54. Bendtsen, M.; Bunger, C.E.; Zou, X.; Foldager, C.; Jorgensen, H.S. Autologous stem cell therapy maintains vertebral blood flow and contrast diffusion through the endplate in experimental intervertebral disc degeneration. Spine 2011, 36, 373–379. [Google Scholar] [CrossRef] [PubMed]
  55. Barczewska, M.; Wojtkiewicz, J.; Habich, A.; Janowski, M.; Adamiak, Z.; Holak, P.; Matyjasik, H.; Bulte, J.W.; Maksymowicz, W.; Walczak, P. MR monitoring of minimally invasive delivery of mesenchymal stem cells into the porcine intervertebral disc. PLoS ONE 2013, 8, e74658. [Google Scholar] [CrossRef]
  56. Yuan, M.; Yeung, C.W.; Li, Y.Y.; Diao, H.; Cheung, K.M.C.; Chan, D.; Cheah, K.; Chan, P.B. Effects of nucleus pulposus cell-derived acellular matrix on the differentiation of mesenchymal stem cells. Biomaterials 2013, 34, 3948–3961. [Google Scholar] [CrossRef]
  57. Crevensten, G.; Walsh, A.J.; Ananthakrishnan, D.; Page, P.; Wahba, G.M.; Lotz, J.C.; Berven, S. Intervertebral disc cell therapy for regeneration: Mesenchymal stem cell implantation in rat intervertebral discs. Ann. Biomed. Eng. 2004, 32, 430–434. [Google Scholar] [CrossRef]
  58. Elabd, C.; Centeno, C.J.; Schultz, J.R.; Lutz, G.; Ichim, T.; Silva, F.J. Intra-discal injection of autologous, hypoxic cultured bone marrow-derived mesenchymal stem cells in five patients with chronic lower back pain: A long-term safety and feasibility study. J. Transl. Med. 2016, 14, 253. [Google Scholar] [CrossRef] [Green Version]
  59. Noriega, D.C.; Ardura, F.; Hernandez-Ramajo, R.; Martin-Ferrero, M.A.; Sanchez-Lite, I.; Toribio, B.; Alberca, M.; Garcia, V.; Moraleda, J.M.; Sanchez, A.; et al. Intervertebral Disc Repair by Allogeneic Mesenchymal Bone Marrow Cells: A Randomized Controlled Trial. Transplantation 2017, 101, 1945–1951. [Google Scholar] [CrossRef]
  60. Ganey, T.; Hutton, W.C.; Moseley, T.; Hedrick, M.; Meisel, H.J. Intervertebral disc repair using adipose tissue-derived stem and regenerative cells: Experiments in a canine model. Spine 2009, 34, 2297–2304. [Google Scholar] [CrossRef] [Green Version]
  61. Jeong, J.H.; Lee, J.H.; Jin, E.S.; Min, J.K.; Jeon, S.R.; Choi, K.H. Regeneration of intervertebral discs in a rat disc degeneration model by implanted adipose-tissue-derived stromal cells. Acta Neurochir. 2010, 152, 1771–1777. [Google Scholar] [CrossRef] [PubMed]
  62. Chun, H.J.; Kim, Y.S.; Kim, B.K.; Kim, E.H.; Kim, J.H.; Do, B.R.; Hwang, S.J.; Hwang, J.Y.; Lee, Y.K. Transplantation of human adipose-derived stem cells in a rabbit model of traumatic degeneration of lumbar discs. World Neurosurg. 2012, 78, 364–371. [Google Scholar] [CrossRef]
  63. Liang, C.Z.; Li, H.; Tao, Y.Q.; Peng, L.H.; Gao, J.Q.; Wu, J.J.; Li, F.C.; Hua, J.M.; Chen, Q.X. Dual release of dexamethasone and TGF-beta3 from polymeric microspheres for stem cell matrix accumulation in a rat disc degeneration model. Acta Biomater. 2013, 9, 9423–9433. [Google Scholar] [CrossRef]
  64. Kumar, H.; Ha, D.H.; Lee, E.J.; Park, J.H.; Shim, J.H.; Ahn, T.K.; Kim, K.T.; Ropper, A.E.; Sohn, S.; Kim, C.H.; et al. Safety and tolerability of intradiscal implantation of combined autologous adipose-derived mesenchymal stem cells and hyaluronic acid in patients with chronic discogenic low back pain: 1-year follow-up of a phase I study. Stem Cell Res. Ther. 2017, 8, 262. [Google Scholar] [CrossRef] [Green Version]
  65. Zhou, X.; Wang, J.; Fang, W.; Tao, Y.; Zhao, T.; Xia, K.; Liang, C.; Hua, J.; Li, F.; Chen, Q. Genipin cross-linked type II collagen/chondroitin sulfate composite hydrogel-like cell delivery system induces differentiation of adipose-derived stem cells and regenerates degenerated nucleus pulposus. Acta Biomater. 2018, 71, 496–509. [Google Scholar] [CrossRef]
  66. Zhang, Z.; Li, F.; Tian, H.; Guan, K.; Zhao, G.; Shan, J.; Ren, D. Differentiation of adipose-derived stem cells toward nucleus pulposus-like cells induced by hypoxia and a three-dimensional chitosan-alginate gel scaffold in vitro. Chin. Med. J. 2014, 127, 314–321. [Google Scholar] [PubMed]
  67. Miyamoto, T.; Muneta, T.; Tabuchi, T.; Matsumoto, K.; Saito, H.; Tsuji, K.; Sekiya, I. Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits. Arthritis Res. Ther. 2010, 12, 206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wang, F.; Nan, L.P.; Zhou, S.F.; Liu, Y.; Wang, Z.Y.; Wang, J.C.; Feng, X.M.; Zhang, L. Injectable Hydrogel Combined with Nucleus Pulposus-Derived Mesenchymal Stem Cells for the Treatment of Degenerative Intervertebral Disc in Rats. Stem Cells Int. 2019, 2019, 8496025. [Google Scholar] [CrossRef]
  69. Chen, J.; Lee, E.J.; Jing, L.; Christoforou, N.; Leong, K.W.; Setton, L.A. Differentiation of mouse induced pluripotent stem cells (iPSCs) into nucleus pulposus-like cells in vitro. PLoS ONE 2013, 8, 75548. [Google Scholar] [CrossRef] [Green Version]
  70. Liu, Y.; Rahaman, M.N.; Bal, B.S. Modulating notochordal differentiation of human induced pluripotent stem cells using natural nucleus pulposus tissue matrix. PLoS ONE 2014, 9, 100885. [Google Scholar] [CrossRef] [Green Version]
  71. Liu, Y.; Fu, S.; Rahaman, M.N.; Mao, J.J.; Bal, B.S. Native nucleus pulposus tissue matrix promotes notochordal differentiation of human induced pluripotent stem cells with potential for treating intervertebral disc degeneration. J. Biomed. Mater. Res. A 2015, 103, 1053–1059. [Google Scholar] [CrossRef]
  72. Tang, R.; Jing, L.; Willard, V.P.; Wu, C.L.; Guilak, F.; Chen, J.; Setton, L.A. Differentiation of human induced pluripotent stem cells into nucleus pulposus-like cells. Stem Cell Res. Ther. 2018, 9, 61. [Google Scholar] [CrossRef] [Green Version]
  73. Zhang, Y.; Zhang, Z.; Chen, P.; Ma, C.Y.; Li, C.; Au, T.Y.K.; Tam, V.; Peng, Y.; Wu, R.; Cheung, K.M.C.; et al. Directed Differentiation of Notochord-like and Nucleus Pulposus-like Cells Using Human Pluripotent Stem Cells. Cell Rep. 2020, 30, 2791–2806 e2795. [Google Scholar] [CrossRef] [Green Version]
  74. Xia, K.; Zhu, J.; Hua, J.; Gong, Z.; Yu, C.; Zhou, X.; Wang, J.; Huang, X.; Yu, W.; Li, L.; et al. Intradiscal Injection of Induced Pluripotent Stem Cell-Derived Nucleus Pulposus-Like Cell-Seeded Polymeric Microspheres Promotes Rat Disc Regeneration. Stem Cells Int. 2019, 2019, 6806540. [Google Scholar] [CrossRef]
  75. Oldershaw, R.A.; Baxter, M.A.; Lowe, E.T.; Bates, N.; Grady, L.M.; Soncin, F.; Brison, D.R.; Hardingham, T.E.; Kimber, S.J. Directed differentiation of human embryonic stem cells toward chondrocytes. Nat. Biotechnol. 2010, 28, 1187–1194. [Google Scholar] [CrossRef] [PubMed]
  76. Diaz-Hernandez, M.E.; Khan, N.M.; Trochez, C.M.; Yoon, T.; Maye, P.; Presciutti, S.M.; Gibson, G.; Drissi, H. Derivation of notochordal cells from human embryonic stem cells reveals unique regulatory networks by single cell-transcriptomics. J. Cell Physiol. 2020, 235, 5241–5255. [Google Scholar] [CrossRef] [PubMed]
  77. Winzi, M.K.; Hyttel, P.; Dale, J.K.; Serup, P. Isolation and characterization of node/notochord-like cells from mouse embryonic stem cells. Stem Cells Dev. 2011, 20, 1817–1827. [Google Scholar] [CrossRef] [Green Version]
  78. Ukeba, D.; Yamada, K.; Tsujimoto, T.; Ura, K.; Nonoyama, T.; Iwasaki, N.; Sudo, H. Bone Marrow Aspirate Concentrate Combined with in Situ Forming Bioresorbable Gel Enhances Intervertebral Disc Regeneration in Rabbits. J. Bone Jt. Surg. Am. 2021, 103, e31. [Google Scholar] [CrossRef]
  79. Pettine, K.A.; Murphy, M.B.; Suzuki, R.K.; Sand, T.T. Percutaneous injection of autologous bone marrow concentrate cells significantly reduces lumbar discogenic pain through 12 months. Stem Cells 2015, 33, 146–156. [Google Scholar] [CrossRef]
  80. Pettine, K.A.; Suzuki, R.K.; Sand, T.T.; Murphy, M.B. Autologous bone marrow concentrate intradiscal injection for the treatment of degenerative disc disease with three-year follow-up. Int. Orthop. 2017, 41, 2097–2103. [Google Scholar] [CrossRef] [PubMed]
  81. Serhan, H. Advancements in the Treatment of Degenerative Disc Disease. Hamdan Med. J. 2018, 11, 175–183. [Google Scholar] [CrossRef]
  82. Otsuru, S.; Gordon, P.L.; Shimono, K.; Jethva, R.; Marino, R.; Phillips, C.L.; Hofmann, T.J.; Veronesi, E.; Dominici, M.; Iwamoto, M.; et al. Transplanted bone marrow mononuclear cells and MSCs impart clinical benefit to children with osteogenesis imperfecta through different mechanisms. Blood 2012, 120, 1933–1941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Murphy, M.B.; Moncivais, K.; Caplan, A.I. Mesenchymal stem cells: Environmentally responsive therapeutics for regenerative medicine. Exp. Mol. Med. 2013, 45, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mwale, F.; Wang, H.T.; Roughley, P.; Antoniou, J.; Haglund, L. Link N and mesenchymal stem cells can induce regeneration of the early degenerate intervertebral disc. Tissue Eng. Part. A 2014, 20, 2942–2949. [Google Scholar] [CrossRef] [Green Version]
  85. Sakai, D.; Mochida, J.; Iwashina, T.; Watanabe, T.; Nakai, T.; Ando, K.; Hotta, T. Differentiation of mesenchymal stem cells transplanted to a rabbit degenerative disc model: Potential and limitations for stem cell therapy in disc regeneration. Spine 2005, 30, 2379–2387. [Google Scholar] [CrossRef] [PubMed]
  86. Hiyama, A.; Mochida, J.; Iwashina, T.; Omi, H.; Watanabe, T.; Serigano, K.; Tamura, F.; Sakai, D. Transplantation of mesenchymal stem cells in a canine disc degeneration model. J. Orthop. Res. 2008, 26, 589–600. [Google Scholar] [CrossRef] [PubMed]
  87. Yim, R.L.; Lee, J.T.; Bow, C.H.; Meij, B.; Leung, V.; Cheung, K.M.; Vavken, P.; Samartzis, D. A systematic review of the safety and efficacy of mesenchymal stem cells for disc degeneration: Insights and future directions for regenerative therapeutics. Stem Cells Dev. 2014, 23, 2553–2567. [Google Scholar] [CrossRef]
  88. Centeno, C.; Markle, J.; Dodson, E.; Stemper, I.; Williams, C.J.; Hyzy, M.; Ichim, T.; Freeman, M. Treatment of lumbar degenerative disc disease-associated radicular pain with culture-expanded autologous mesenchymal stem cells: A pilot study on safety and efficacy. J. Transl. Med. 2017, 15, 197. [Google Scholar] [CrossRef] [Green Version]
  89. Leung, V.Y.; Aladin, D.M.; Lv, F.; Tam, V.; Sun, Y.; Lau, R.Y.; Hung, S.C.; Ngan, A.H.; Tang, B.; Lim, C.T.; et al. Mesenchymal stem cells reduce intervertebral disc fibrosis and facilitate repair. Stem Cells 2014, 32, 2164–2177. [Google Scholar] [CrossRef]
  90. Oehme, D.; Ghosh, P.; Shimmon, S.; Wu, J.; McDonald, C.; Troupis, J.M.; Goldschlager, T.; Rosenfeld, J.V.; Jenkin, G. Mesenchymal progenitor cells combined with pentosan polysulfate mediating disc regeneration at the time of microdiscectomy: A preliminary study in an ovine model. J. Neurosurg. Spine 2014, 20, 657–669. [Google Scholar] [CrossRef] [Green Version]
  91. Hee, H.T.; Ismail, H.D.; Lim, C.T.; Goh, J.C.; Wong, H.K. Effects of implantation of bone marrow mesenchymal stem cells, disc distraction and combined therapy on reversing degeneration of the intervertebral disc. J. Bone Jt. Surg. Br. 2010, 92, 726–736. [Google Scholar] [CrossRef]
  92. Ukeba, D.; Sudo, H.; Tsujimoto, T.; Ura, K.; Yamada, K.; Iwasaki, N. Bone marrow mesenchymal stem cells combined with ultra-purified alginate gel as a regenerative therapeutic strategy after discectomy for degenerated intervertebral discs. eBioMedicine 2020, 53, 102698. [Google Scholar] [CrossRef] [PubMed]
  93. Chu, C.R.; Rodeo, S.; Bhutani, N.; Goodrich, L.R.; Huard, J.; Irrgang, J.; LaPrade, R.F.; Lattermann, C.; Lu, Y.; Mandelbaum, B.; et al. Optimizing Clinical Use of Biologics in Orthopaedic Surgery: Consensus Recommendations From the 2018 AAOS/NIH U-13 Conference. J. Am. Acad. Orthop. Surg. 2019, 27, 50–63. [Google Scholar] [CrossRef] [PubMed]
  94. Kregar Velikonja, N.; Urban, J.; Frohlich, M.; Neidlinger-Wilke, C.; Kletsas, D.; Potocar, U.; Turner, S.; Roberts, S. Cell sources for nucleus pulposus regeneration. Eur. Spine J. 2014, 23 (Suppl. 3), 364–374. [Google Scholar] [CrossRef]
  95. Huang, Y.C.; Leung, V.Y.; Lu, W.W.; Luk, K.D. The effects of microenvironment in mesenchymal stem cell-based regeneration of intervertebral disc. Spine J. 2013, 13, 352–362. [Google Scholar] [CrossRef] [PubMed]
  96. Li, H.; Tao, Y.; Liang, C.; Han, B.; Li, F.; Chen, G.; Chen, Q. Influence of hypoxia in the intervertebral disc on the biological behaviors of rat adipose- and nucleus pulposus-derived mesenchymal stem cells. Cells Tissues Organs 2013, 198, 266–277. [Google Scholar] [CrossRef]
  97. Li, Y.Y.; Diao, H.J.; Chik, T.K.; Chow, C.T.; An, X.M.; Leung, V.; Cheung, K.M.; Chan, B.P. Delivering mesenchymal stem cells in collagen microsphere carriers to rabbit degenerative disc: Reduced risk of osteophyte formation. Tissue Eng. Part. A 2014, 20, 1379–1391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Sobajima, S.; Vadala, G.; Shimer, A.; Kim, J.S.; Gilbertson, L.G.; Kang, J.D. Feasibility of a stem cell therapy for intervertebral disc degeneration. Spine J. 2008, 8, 888–896. [Google Scholar] [CrossRef]
  99. Bowles, R.D.; Setton, L.A. Biomaterials for intervertebral disc regeneration and repair. Biomaterials 2017, 129, 54–67. [Google Scholar] [CrossRef]
  100. Wang, H.; Zhou, Y.; Huang, B.; Liu, L.T.; Liu, M.H.; Wang, J.; Li, C.Q.; Zhang, Z.F.; Chu, T.W.; Xiong, C.J. Utilization of stem cells in alginate for nucleus pulposus tissue engineering. Tissue Eng. Part. A 2014, 20, 908–920. [Google Scholar] [CrossRef]
  101. Leckie, S.K.; Sowa, G.A.; Bechara, B.P.; Hartman, R.A.; Coelho, J.P.; Witt, W.T.; Dong, Q.D.; Bowman, B.W.; Bell, K.M.; Vo, N.V.; et al. Injection of human umbilical tissue-derived cells into the nucleus pulposus alters the course of intervertebral disc degeneration in vivo. Spine J. 2013, 13, 263–272. [Google Scholar] [CrossRef] [Green Version]
  102. Bertram, H.; Kroeber, M.; Wang, H.; Unglaub, F.; Guehring, T.; Carstens, C.; Richter, W. Matrix-assisted cell transfer for intervertebral disc cell therapy. Biochem. Biophys. Res. Commun. 2005, 331, 1185–1192. [Google Scholar] [CrossRef]
  103. Embree, M.C.; Chen, M.; Pylawka, S.; Kong, D.; Iwaoka, G.M.; Kalajzic, I.; Yao, H.; Shi, C.; Sun, D.; Sheu, T.J.; et al. Exploiting endogenous fibrocartilage stem cells to regenerate cartilage and repair joint injury. Nat. Commun. 2016, 7, 13073. [Google Scholar] [CrossRef] [Green Version]
  104. Huey, D.J.; Hu, J.C.; Athanasiou, K.A. Unlike bone, cartilage regeneration remains elusive. Science 2012, 338, 917–921. [Google Scholar] [CrossRef] [Green Version]
  105. Waskow, C. Maintaining What Is Already There: Strategies to Rectify HSC Transplantation Dilemmas. Cell Stem Cell 2015, 17, 258–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Sicari, B.M.; Rubin, J.P.; Dearth, C.L.; Wolf, M.T.; Ambrosio, F.; Boninger, M.; Turner, N.J.; Weber, D.J.; Simpson, T.W.; Wyse, A.; et al. An acellular biologic scaffold promotes skeletal muscle formation in mice and humans with volumetric muscle loss. Sci. Transl. Med. 2014, 6, 234ra258. [Google Scholar] [CrossRef] [Green Version]
  107. Abbushi, A.; Endres, M.; Cabraja, M.; Kroppenstedt, S.N.; Thomale, U.W.; Sittinger, M.; Hegewald, A.A.; Morawietz, L.; Lemke, A.J.; Bansemer, V.G.; et al. Regeneration of intervertebral disc tissue by resorbable cell-free polyglycolic acid-based implants in a rabbit model of disc degeneration. Spine 2008, 33, 1527–1532. [Google Scholar] [CrossRef] [PubMed]
  108. Pattappa, G.; Li, Z.; Peroglio, M.; Wismer, N.; Alini, M.; Grad, S. Diversity of intervertebral disc cells: Phenotype and function. J. Anat. 2012, 221, 480–496. [Google Scholar] [CrossRef]
  109. Schmitz, T.C.; Salzer, E.; Crispim, J.F.; Fabra, G.T.; LeVisage, C.; Pandit, A.; Tryfonidou, M.; Maitre, C.L.; Ito, K. Characterization of biomaterials intended for use in the nucleus pulposus of degenerated intervertebral discs. Acta Biomater. 2020, 114, 1–15. [Google Scholar] [CrossRef]
  110. Maroudas, A.; Stockwell, R.A.; Nachemson, A.; Urban, J. Factors involved in the nutrition of the human lumbar intervertebral disc: Cellularity and diffusion of glucose in vitro. J. Anat. 1975, 120, 113–130. [Google Scholar] [PubMed]
  111. Iatridis, J.C.; Setton, L.A.; Weidenbaum, M.; Mow, V.C. Alterations in the mechanical behavior of the human lumbar nucleus pulposus with degeneration and aging. J. Orthop. Res. 1997, 15, 318–322. [Google Scholar] [CrossRef] [PubMed]
  112. Johannessen, W.; Elliott, D.M. Effects of degeneration on the biphasic material properties of human nucleus pulposus in confined compression. Spine 2005, 30, 724–729. [Google Scholar] [CrossRef]
  113. Huang, Y.C.; Hu, Y.; Li, Z.; Luk, K.D.K. Biomaterials for intervertebral disc regeneration: Current status and looming challenges. J. Tissue Eng. Regen. Med. 2018, 12, 2188–2202. [Google Scholar] [CrossRef]
  114. Nachemson, A.L.; Evans, J.H. Some mechanical properties of the third human lumbar interlaminar ligament (ligamentum flavum). J. Biomech 1968, 1, 211–220. [Google Scholar] [CrossRef]
  115. Pereira, D.R.; Silva-Correia, J.; Oliveira, J.M.; Reis, R.L. Hydrogels in acellular and cellular strategies for intervertebral disc regeneration. J. Tissue Eng. Regen. Med. 2013, 7, 85–98. [Google Scholar] [CrossRef] [Green Version]
  116. Iatridis, J.C.; Nicoll, S.B.; Michalek, A.J.; Walter, B.A.; Gupta, M.S. Role of biomechanics in intervertebral disc degeneration and regenerative therapies: What needs repairing in the disc and what are promising biomaterials for its repair? Spine J. 2013, 13, 243–262. [Google Scholar] [CrossRef] [Green Version]
  117. Carl, A.; Ledet, E.; Yuan, H.; Sharan, A. New developments in nucleus pulposus replacement technology. Spine J. 2004, 4, 325S–329S. [Google Scholar] [CrossRef] [PubMed]
  118. Coric, D.; Mummaneni, P.V. Nucleus replacement technologies. J. Neurosurg. Spine 2008, 8, 115–120. [Google Scholar] [CrossRef]
  119. Goins, M.L.; Wimberley, D.W.; Yuan, P.S.; Fitzhenry, L.N.; Vaccaro, A.R. Nucleus pulposus replacement: An emerging technology. Spine J. 2005, 5, 317S–324S. [Google Scholar] [CrossRef]
  120. Pelletier, M.H.; Cohen, C.S.; Ducheyne, P.; Walsh, W.R. Restoring Segmental Biomechanics Through Nucleus Augmentation: An In Vitro Study. Clin. Spine Surg. 2016, 29, 461–467. [Google Scholar] [CrossRef] [PubMed]
  121. Schutgens, E.M.; Tryfonidou, M.A.; Smit, T.H.; Oner, F.C.; Krouwels, A.; Ito, K.; Creemers, L.B. Biomaterials for intervertebral disc regeneration: Past performance and possible future strategies. Eur. Cell Mater. 2015, 30, 210–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Buckley, C.T.; Hoyland, J.A.; Fujii, K.; Pandit, A.; Iatridis, J.C.; Grad, S. Critical aspects and challenges for intervertebral disc repair and regeneration-Harnessing advances in tissue engineering. JOR Spine 2018, 1, e1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Richardson, S.M.; Curran, J.M.; Chen, R.; Vaughan-Thomas, A.; Hunt, J.A.; Freemont, A.J.; Hoyland, J.A. The differentiation of bone marrow mesenchymal stem cells into chondrocyte-like cells on poly-L-lactic acid (PLLA) scaffolds. Biomaterials 2006, 27, 4069–4078. [Google Scholar] [CrossRef]
  124. El-Amin, S.F.; Lu, H.H.; Khan, Y.; Burems, J.; Mitchell, J.; Tuan, R.S.; Laurencin, C.T. Extracellular matrix production by human osteoblasts cultured on biodegradable polymers applicable for tissue engineering. Biomaterials 2003, 24, 1213–1221. [Google Scholar] [CrossRef]
  125. Endres, M.; Abbushi, A.; Thomale, U.W.; Cabraja, M.; Kroppenstedt, S.N.; Morawietz, L.; Casalis, P.A.; Zenclussen, M.L.; Lemke, A.J.; Horn, P.; et al. Intervertebral disc regeneration after implantation of a cell-free bioresorbable implant in a rabbit disc degeneration model. Biomaterials 2010, 31, 5836–5841. [Google Scholar] [CrossRef]
  126. Feng, G.; Jin, X.; Hu, J.; Ma, H.; Gupte, M.J.; Liu, H.; Ma, P.X. Effects of hypoxias and scaffold architecture on rabbit mesenchymal stem cell differentiation towards a nucleus pulposus-like phenotype. Biomaterials 2011, 32, 8182–8189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Kim, H.Y.; Kim, H.N.; Lee, S.J.; Song, J.E.; Kwon, S.Y.; Chung, J.W.; Lee, D.; Khang, G. Effect of pore sizes of PLGA scaffolds on mechanical properties and cell behaviour for nucleus pulposus regeneration in vivo. J. Tissue Eng. Regen. Med. 2017, 11, 44–57. [Google Scholar] [CrossRef]
  128. Mizuno, H.; Roy, A.K.; Vacanti, C.A.; Kojima, K.; Ueda, M.; Bonassar, L.J. Tissue-engineered composites of anulus fibrosus and nucleus pulposus for intervertebral disc replacement. Spine 2004, 29, 1290–1297; discussion 1297–1298. [Google Scholar] [CrossRef]
  129. Xin, L.; Xu, W.; Yu, L.; Fan, S.; Wang, W.; Yu, F.; Wang, Z. Effects of annulus defects and implantation of poly(lactic-co-glycolic acid) (PLGA)/fibrin gel scaffolds on nerves ingrowth in a rabbit model of annular injury disc degeneration. J. Orthop. Surg. Res. 2017, 12, 73. [Google Scholar] [CrossRef]
  130. Xin, L.; Zhang, C.; Zhong, F.; Fan, S.; Wang, W.; Wang, Z. Minimal invasive annulotomy for induction of disc degeneration and implantation of poly (lactic-co-glycolic acid) (PLGA) plugs for annular repair in a rabbit model. Eur. J. Med. Res. 2016, 21, 7. [Google Scholar] [CrossRef] [Green Version]
  131. Danhier, F.; Ansorena, E.; Silva, J.M.; Coco, R.; Le Breton, A.; Preat, V. PLGA-based nanoparticles: An overview of biomedical applications. J. Control. Release 2012, 161, 505–522. [Google Scholar] [CrossRef] [PubMed]
  132. Collin, E.C.; Grad, S.; Zeugolis, D.I.; Vinatier, C.S.; Clouet, J.R.; Guicheux, J.J.; Weiss, P.; Alini, M.; Pandit, A.S. An injectable vehicle for nucleus pulposus cell-based therapy. Biomaterials 2011, 32, 2862–2870. [Google Scholar] [CrossRef] [PubMed]
  133. Francisco, A.T.; Hwang, P.Y.; Jeong, C.G.; Jing, L.; Chen, J.; Setton, L.A. Photocrosslinkable laminin-functionalized polyethylene glycol hydrogel for intervertebral disc regeneration. Acta Biomater. 2014, 10, 1102–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lin, C.C.; Anseth, K.S. PEG hydrogels for the controlled release of biomolecules in regenerative medicine. Pharm. Res. 2009, 26, 631–643. [Google Scholar] [CrossRef] [Green Version]
  135. Nguyen, Q.T.; Hwang, Y.; Chen, A.C.; Varghese, S.; Sah, R.L. Cartilage-like mechanical properties of poly (ethylene glycol)-diacrylate hydrogels. Biomaterials 2012, 33, 6682–6690. [Google Scholar] [CrossRef] [Green Version]
  136. Raeber, G.P.; Lutolf, M.P.; Hubbell, J.A. Molecularly engineered PEG hydrogels: A novel model system for proteolytically mediated cell migration. Biophys. J. 2005, 89, 1374–1388. [Google Scholar] [CrossRef] [Green Version]
  137. Zhu, J. Bioactive modification of poly(ethylene glycol) hydrogels for tissue engineering. Biomaterials 2010, 31, 4639–4656. [Google Scholar] [CrossRef] [Green Version]
  138. Scholz, B.; Kinzelmann, C.; Benz, K.; Mollenhauer, J.; Wurst, H.; Schlosshauer, B. Suppression of adverse angiogenesis in an albumin-based hydrogel for articular cartilage and intervertebral disc regeneration. Eur. Cell Mater. 2010, 20, 24–36; discussion 27–36. [Google Scholar] [CrossRef]
  139. Benz, K.; Stippich, C.; Osswald, C.; Gaissmaier, C.; Lembert, N.; Badke, A.; Steck, E.; Aicher, W.K.; Mollenhauer, J.A. Rheological and biological properties of a hydrogel support for cells intended for intervertebral disc repair. BMC Musculoskelet. Disord. 2012, 13, 54. [Google Scholar] [CrossRef] [Green Version]
  140. Benz, K.; Stippich, C.; Fischer, L.; Mohl, K.; Weber, K.; Lang, J.; Steffen, F.; Beintner, B.; Gaissmaier, C.; Mollenhauer, J.A. Intervertebral disc cell- and hydrogel-supported and spontaneous intervertebral disc repair in nucleotomized sheep. Eur. Spine J. 2012, 21, 1758–1768. [Google Scholar] [CrossRef] [Green Version]
  141. Thomas, J.D.; Fussell, G.; Sarkar, S.; Lowman, A.M.; Marcolongo, M. Synthesis and recovery characteristics of branched and grafted PNIPAAm-PEG hydrogels for the development of an injectable load-bearing nucleus pulposus replacement. Acta Biomater. 2010, 6, 1319–1328. [Google Scholar] [CrossRef]
  142. Halloran, D.O.; Grad, S.; Stoddart, M.; Dockery, P.; Alini, M.; Pandit, A.S. An injectable cross-linked scaffold for nucleus pulposus regeneration. Biomaterials 2008, 29, 438–447. [Google Scholar] [CrossRef]
  143. Attia, M.; Santerre, J.P.; Kandel, R.A. The response of annulus fibrosus cell to fibronectin-coated nanofibrous polyurethane-anionic dihydroxyoligomer scaffolds. Biomaterials 2011, 32, 450–460. [Google Scholar] [CrossRef] [PubMed]
  144. Dahl, M.C.; Ahrens, M.; Sherman, J.E.; Martz, E.O. The restoration of lumbar intervertebral disc load distribution: A comparison of three nucleus replacement technologies. Spine 2010, 35, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
  145. Hu, J.; Chen, B.; Guo, F.; Du, J.; Gu, P.; Lin, X.; Yang, W.; Zhang, H.; Lu, M.; Huang, Y.; et al. Injectable silk fibroin/polyurethane composite hydrogel for nucleus pulposus replacement. J. Mater. Sci Mater. Med. 2012, 23, 711–722. [Google Scholar] [CrossRef] [PubMed]
  146. Mauth, C.; Bono, E.; Haas, S.; Paesold, G.; Wiese, H.; Maier, G.; Boos, N.; Graf-Hausner, U. Cell-seeded polyurethane-fibrin structures—a possible system for intervertebral disc regeneration. Eur. Cell Mater. 2009, 18, 27–38; discussion 29–38. [Google Scholar] [CrossRef] [PubMed]
  147. Park, H.S.; Gong, M.S.; Park, J.H.; Moon, S.I.; Wall, I.B.; Kim, H.W.; Lee, J.H.; Knowles, J.C. Silk fibroin-polyurethane blends: Physical properties and effect of silk fibroin content on viscoelasticity, biocompatibility and myoblast differentiation. Acta Biomater. 2013, 9, 8962–8971. [Google Scholar] [CrossRef]
  148. Yang, X.; Li, X. Nucleus pulposus tissue engineering: A brief review. Eur. Spine J. 2009, 18, 1564–1572. [Google Scholar] [CrossRef] [Green Version]
  149. Agrawal, C.M.; Ray, R.B. Biodegradable polymeric scaffolds for musculoskeletal tissue engineering. J. Biomed. Mater. Res. 2001, 55, 141–150. [Google Scholar] [CrossRef]
  150. Dash, T.K.; Konkimalla, V.B. Poly-small je, Ukrainian-caprolactone based formulations for drug delivery and tissue engineering: A review. J. Control. Release 2012, 158, 15–33. [Google Scholar] [CrossRef]
  151. Li, W.J.; Tuli, R.; Okafor, C.; Derfoul, A.; Danielson, K.G.; Hall, D.J.; Tuan, R.S. A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials 2005, 26, 599–609. [Google Scholar] [CrossRef] [PubMed]
  152. Lopez, A.; Persson, C.; Hilborn, J.; Engqvist, H. Synthesis and characterization of injectable composites of poly[D,L-lactide-co-(epsilon-caprolactone)] reinforced with beta-TCP and CaCO3 for intervertebral disk augmentation. J. Biomed. Mater. Res. B Appl. Biomater. 2010, 95, 75–83. [Google Scholar] [CrossRef] [PubMed]
  153. Koepsell, L.; Remund, T.; Bao, J.; Neufeld, D.; Fong, H.; Deng, Y. Tissue engineering of annulus fibrosus using electrospun fibrous scaffolds with aligned polycaprolactone fibers. J. Biomed. Mater. Res. A 2011, 99, 564–575. [Google Scholar] [CrossRef]
  154. Koepsell, L.; Zhang, L.; Neufeld, D.; Fong, H.; Deng, Y. Electrospun nanofibrous polycaprolactone scaffolds for tissue engineering of annulus fibrosus. Macromol. Biosci. 2011, 11, 391–399. [Google Scholar] [CrossRef]
  155. Martin, J.T.; Milby, A.H.; Chiaro, J.A.; Kim, D.H.; Hebela, N.M.; Smith, L.J.; Elliott, D.M.; Mauck, R.L. Translation of an engineered nanofibrous disc-like angle-ply structure for intervertebral disc replacement in a small animal model. Acta Biomater. 2014, 10, 2473–2481. [Google Scholar] [CrossRef] [Green Version]
  156. Slaughter, B.V.; Khurshid, S.S.; Fisher, O.Z.; Khademhosseini, A.; Peppas, N.A. Hydrogels in regenerative medicine. Adv. Mater. 2009, 21, 3307–3329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Wei, A.; Shen, B.; Williams, L.; Diwan, A. Mesenchymal stem cells: Potential application in intervertebral disc regeneration. Transl. Pediatr. 2014, 3, 71–90. [Google Scholar] [CrossRef]
  158. Bron, J.L.; Vonk, L.A.; Smit, T.H.; Koenderink, G.H. Engineering alginate for intervertebral disc repair. J. Mech. Behav. Biomed. Mater. 2011, 4, 1196–1205. [Google Scholar] [CrossRef] [PubMed]
  159. Chou, A.I.; Akintoye, S.O.; Nicoll, S.B. Photo-crosslinked alginate hydrogels support enhanced matrix accumulation by nucleus pulposus cells in vivo. Osteoarthr. Cartil. 2009, 17, 1377–1384. [Google Scholar] [CrossRef] [Green Version]
  160. Chou, A.I.; Nicoll, S.B. Characterization of photocrosslinked alginate hydrogels for nucleus pulposus cell encapsulation. J. Biomed. Mater. Res. A 2009, 91, 187–194. [Google Scholar] [CrossRef]
  161. Guo, J.F.; Jourdian, G.W.; MacCallum, D.K. Culture and growth characteristics of chondrocytes encapsulated in alginate beads. Connect. Tissue Res. 1989, 19, 277–297. [Google Scholar] [CrossRef] [PubMed]
  162. Larsen, B.; Haug, A. Biosynthesis of alginate. 1. Composition and structure of alginate produced by Azotobacter vinelandii (Lipman). Carbohydr. Res. 1971, 17, 287–296. [Google Scholar] [CrossRef]
  163. Leone, G.; Torricelli, P.; Chiumiento, A.; Facchini, A.; Barbucci, R. Amidic alginate hydrogel for nucleus pulposus replacement. J. Biomed. Mater. Res. A 2008, 84, 391–401. [Google Scholar] [CrossRef] [PubMed]
  164. Li, Z.; Gunn, J.; Chen, M.H.; Cooper, A.; Zhang, M. On-site alginate gelation for enhanced cell proliferation and uniform distribution in porous scaffolds. J. Biomed. Mater. Res. A 2008, 86, 552–559. [Google Scholar] [CrossRef] [PubMed]
  165. Lee, K.Y.; Mooney, D.J. Alginate: Properties and biomedical applications. Prog. Polym. Sci. 2012, 37, 106–126. [Google Scholar] [CrossRef] [Green Version]
  166. Melrose, J.; Smith, S.; Ghosh, P.; Taylor, T.K. Differential expression of proteoglycan epitopes and growth characteristics of intervertebral disc cells grown in alginate bead culture. Cells Tissues Organs 2001, 168, 137–146. [Google Scholar] [CrossRef]
  167. Nunamaker, E.A.; Purcell, E.K.; Kipke, D.R. In vivo stability and biocompatibility of implanted calcium alginate disks. J. Biomed. Mater. Res. A 2007, 83, 1128–1137. [Google Scholar] [CrossRef] [Green Version]
  168. Ura, K.; Yamada, K.; Tsujimoto, T.; Ukeba, D.; Iwasaki, N.; Sudo, H. Ultra-purified alginate gel implantation decreases inflammatory cytokine levels, prevents intervertebral disc degeneration, and reduces acute pain after discectomy. Sci. Rep. 2021, 11, 638. [Google Scholar] [CrossRef]
  169. Naqvi, S.M.; Buckley, C.T. Differential response of encapsulated nucleus pulposus and bone marrow stem cells in isolation and coculture in alginate and chitosan hydrogels. Tissue Eng Part. A 2015, 21, 288–299. [Google Scholar] [CrossRef] [Green Version]
  170. Zeng, Y.; Chen, C.; Liu, W.; Fu, Q.; Han, Z.; Li, Y.; Feng, S.; Li, X.; Qi, C.; Wu, J.; et al. Injectable microcryogels reinforced alginate encapsulation of mesenchymal stromal cells for leak-proof delivery and alleviation of canine disc degeneration. Biomaterials 2015, 59, 53–65. [Google Scholar] [CrossRef]
  171. Bidarra, S.J.; Barrias, C.C.; Granja, P.L. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta Biomater. 2014, 10, 1646–1662. [Google Scholar] [CrossRef] [PubMed]
  172. Shao, X.; Hunter, C.J. Developing an alginate/chitosan hybrid fiber scaffold for annulus fibrosus cells. J. Biomed. Mater. Res. A 2007, 82, 701–710. [Google Scholar] [CrossRef]
  173. Cloyd, J.M.; Malhotra, N.R.; Weng, L.; Chen, W.; Mauck, R.L.; Elliott, D.M. Material properties in unconfined compression of human nucleus pulposus, injectable hyaluronic acid-based hydrogels and tissue engineering scaffolds. Eur. Spine J. 2007, 16, 1892–1898. [Google Scholar] [CrossRef] [Green Version]
  174. Gruber, H.E.; Hoelscher, G.L.; Leslie, K.; Ingram, J.A.; Hanley, E.N., Jr. Three-dimensional culture of human disc cells within agarose or a collagen sponge: Assessment of proteoglycan production. Biomaterials 2006, 27, 371–376. [Google Scholar] [CrossRef]
  175. Gupta, A.; Bhat, S.; Jagdale, P.R.; Chaudhari, B.P.; Lidgren, L.; Gupta, K.C.; Kumar, A. Evaluation of three-dimensional chitosan-agarose-gelatin cryogel scaffold for the repair of subchondral cartilage defects: An in vivo study in a rabbit model. Tissue Eng. Part. A 2014, 20, 3101–3111. [Google Scholar] [CrossRef] [Green Version]
  176. Hunt, N.C.; Grover, L.M. Cell encapsulation using biopolymer gels for regenerative medicine. Biotechnol. Lett. 2010, 32, 733–742. [Google Scholar] [CrossRef]
  177. Lazebnik, M.; Singh, M.; Glatt, P.; Friis, L.A.; Berkland, C.J.; Detamore, M.S. Biomimetic method for combining the nucleus pulposus and annulus fibrosus for intervertebral disc tissue engineering. J. Tissue Eng. Regen. Med. 2011, 5, 179–187. [Google Scholar] [CrossRef] [PubMed]
  178. Nerurkar, N.L.; Sen, S.; Huang, A.H.; Elliott, D.M.; Mauck, R.L. Engineered disc-like angle-ply structures for intervertebral disc replacement. Spine 2010, 35, 867–873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Tilwani, R.K.; Bader, D.L.; Chowdhury, T.T. Biomechanical Conditioning Enhanced Matrix Synthesis in Nucleus Pulposus Cells Cultured in Agarose Constructs with TGFbeta. J. Funct. Biomater. 2012, 3, 23–36. [Google Scholar] [CrossRef] [Green Version]
  180. Ahmed, T.A.; Dare, E.V.; Hincke, M. Fibrin: A versatile scaffold for tissue engineering applications. Tissue Eng. Part. B Rev. 2008, 14, 199–215. [Google Scholar] [CrossRef]
  181. Colombini, A.; Ceriani, C.; Banfi, G.; Brayda-Bruno, M.; Moretti, M. Fibrin in intervertebral disc tissue engineering. Tissue Eng. Part. B Rev. 2014, 20, 713–721. [Google Scholar] [CrossRef] [PubMed]
  182. Diederichs, S.; Baral, K.; Tanner, M.; Richter, W. Interplay between local versus soluble transforming growth factor-beta and fibrin scaffolds: Role of cells and impact on human mesenchymal stem cell chondrogenesis. Tissue Eng. Part. A 2012, 18, 1140–1150. [Google Scholar] [CrossRef] [PubMed]
  183. Eyrich, D.; Brandl, F.; Appel, B.; Wiese, H.; Maier, G.; Wenzel, M.; Staudenmaier, R.; Goepferich, A.; Blunk, T. Long-term stable fibrin gels for cartilage engineering. Biomaterials 2007, 28, 55–65. [Google Scholar] [CrossRef]
  184. Ho, S.T.; Cool, S.M.; Hui, J.H.; Hutmacher, D.W. The influence of fibrin based hydrogels on the chondrogenic differentiation of human bone marrow stromal cells. Biomaterials 2010, 31, 38–47. [Google Scholar] [CrossRef] [PubMed]
  185. Li, Z.; Kaplan, K.M.; Wertzel, A.; Peroglio, M.; Amit, B.; Alini, M.; Grad, S.; Yayon, A. Biomimetic fibrin-hyaluronan hydrogels for nucleus pulposus regeneration. Regen. Med. 2014, 9, 309–326. [Google Scholar] [CrossRef]
  186. Likhitpanichkul, M.; Dreischarf, M.; Illien-Junger, S.; Walter, B.A.; Nukaga, T.; Long, R.G.; Sakai, D.; Hecht, A.C.; Iatridis, J.C. Fibrin-genipin adhesive hydrogel for annulus fibrosus repair: Performance evaluation with large animal organ culture, in situ biomechanics, and in vivo degradation tests. Eur. Cell Mater. 2014, 28, 25–37; discussion 28–37. [Google Scholar] [CrossRef]
  187. Ma, K.; Titan, A.L.; Stafford, M.; Zheng, C.; Levenston, M.E. Variations in chondrogenesis of human bone marrow-derived mesenchymal stem cells in fibrin/alginate blended hydrogels. Acta Biomater. 2012, 8, 3754–3764. [Google Scholar] [CrossRef] [Green Version]
  188. Park, S.H.; Cho, H.; Gil, E.S.; Mandal, B.B.; Min, B.H.; Kaplan, D.L. Silk-fibrin/hyaluronic acid composite gels for nucleus pulposus tissue regeneration. Tissue Eng Part. A 2011, 17, 2999–3009. [Google Scholar] [CrossRef] [Green Version]
  189. Pirvu, T.; Blanquer, S.B.; Benneker, L.M.; Grijpma, D.W.; Richards, R.G.; Alini, M.; Eglin, D.; Grad, S.; Li, Z. A combined biomaterial and cellular approach for annulus fibrosus rupture repair. Biomaterials 2015, 42, 11–19. [Google Scholar] [CrossRef]
  190. Schek, R.M.; Michalek, A.J.; Iatridis, J.C. Genipin-crosslinked fibrin hydrogels as a potential adhesive to augment intervertebral disc annulus repair. Eur. Cell Mater. 2011, 21, 373–383. [Google Scholar] [CrossRef]
  191. Stern, S.; Lindenhayn, K.; Schultz, O.; Perka, C. Cultivation of porcine cells from the nucleus pulposus in a fibrin/hyaluronic acid matrix. Acta Orthop. Scand. 2000, 71, 496–502. [Google Scholar] [CrossRef] [PubMed]
  192. Buser, Z.; Kuelling, F.; Liu, J.; Liebenberg, E.; Thorne, K.J.; Coughlin, D.; Lotz, J.C. Biological and biomechanical effects of fibrin injection into porcine intervertebral discs. Spine 2011, 36, E1201–E1209. [Google Scholar] [CrossRef] [Green Version]
  193. Long, R.G.; Ferguson, S.J.; Benneker, L.M.; Sakai, D.; Li, Z.; Pandit, A.; Grijpma, D.W.; Eglin, D.; Zeiter, S.; Schmid, T.; et al. Morphological and biomechanical effects of annulus fibrosus injury and repair in an ovine cervical model. JOR Spine 2020, 3, e1074. [Google Scholar] [CrossRef] [PubMed]
  194. Zhou, Z.; Zeiter, S.; Schmid, T.; Sakai, D.; Iatridis, J.C.; Zhou, G.; Richards, R.G.; Alini, M.; Grad, S.; Li, Z. Effect of the CCL5-Releasing Fibrin Gel for Intervertebral Disc Regeneration. Cartilage 2020, 11, 169–180. [Google Scholar] [CrossRef]
  195. Yin, W.; Pauza, K.; Olan, W.J.; Doerzbacher, J.F.; Thorne, K.J. Intradiscal injection of fibrin sealant for the treatment of symptomatic lumbar internal disc disruption: Results of a prospective multicenter pilot study with 24-month follow-up. Pain Med. 2014, 15, 16–31. [Google Scholar] [CrossRef] [Green Version]
  196. Omlor, G.W.; Lorenz, S.; Nerlich, A.G.; Guehring, T.; Richter, W. Disc cell therapy with bone-marrow-derived autologous mesenchymal stromal cells in a large porcine disc degeneration model. Eur. Spine J. 2018, 27, 2639–2649. [Google Scholar] [CrossRef]
  197. Chen, Y.C.; Su, W.Y.; Yang, S.H.; Gefen, A.; Lin, F.H. In situ forming hydrogels composed of oxidized high molecular weight hyaluronic acid and gelatin for nucleus pulposus regeneration. Acta Biomater. 2013, 9, 5181–5193. [Google Scholar] [CrossRef]
  198. Chung, C.; Beecham, M.; Mauck, R.L.; Burdick, J.A. The influence of degradation characteristics of hyaluronic acid hydrogels on in vitro neocartilage formation by mesenchymal stem cells. Biomaterials 2009, 30, 4287–4296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Erickson, I.E.; Huang, A.H.; Sengupta, S.; Kestle, S.; Burdick, J.A.; Mauck, R.L. Macromer density influences mesenchymal stem cell chondrogenesis and maturation in photocrosslinked hyaluronic acid hydrogels. Osteoarthr. Cartil. 2009, 17, 1639–1648. [Google Scholar] [CrossRef] [Green Version]
  200. Kenne, L.; Gohil, S.; Nilsson, E.M.; Karlsson, A.; Ericsson, D.; Helander Kenne, A.; Nord, L.I. Modification and cross-linking parameters in hyaluronic acid hydrogels--definitions and analytical methods. Carbohydr. Polym. 2013, 91, 410–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Kogan, G.; Soltes, L.; Stern, R.; Gemeiner, P. Hyaluronic acid: A natural biopolymer with a broad range of biomedical and industrial applications. Biotechnol. Lett. 2007, 29, 17–25. [Google Scholar] [CrossRef]
  202. Su, W.Y.; Chen, Y.C.; Lin, F.H. Injectable oxidized hyaluronic acid/adipic acid dihydrazide hydrogel for nucleus pulposus regeneration. Acta Biomater. 2010, 6, 3044–3055. [Google Scholar] [CrossRef]
  203. Peroglio, M.; Grad, S.; Mortisen, D.; Sprecher, C.M.; Illien-Junger, S.; Alini, M.; Eglin, D. Injectable thermoreversible hyaluronan-based hydrogels for nucleus pulposus cell encapsulation. Eur. Spine J. 2012, 21 (Suppl. 6), 839–849. [Google Scholar] [CrossRef]
  204. Woiciechowsky, C.; Abbushi, A.; Zenclussen, M.L.; Casalis, P.; Kruger, J.P.; Freymann, U.; Endres, M.; Kaps, C. Regeneration of nucleus pulposus tissue in an ovine intervertebral disc degeneration model by cell-free resorbable polymer scaffolds. J. Tissue Eng. Regen. Med. 2014, 8, 811–820. [Google Scholar] [CrossRef] [PubMed]
  205. Kazezian, Z.; Sakai, D.; Pandit, A. Hyaluronic Acid Microgels Modulate Inflammation and Key Matrix Molecules toward a Regenerative Signature in the Injured Annulus Fibrosus. Adv. Biosyst. 2017, 1, e1700077. [Google Scholar] [CrossRef] [PubMed]
  206. Reitmaier, S.; Kreja, L.; Gruchenberg, K.; Kanter, B.; Silva-Correia, J.; Oliveira, J.M.; Reis, R.L.; Perugini, V.; Santin, M.; Ignatius, A.; et al. In vivo biofunctional evaluation of hydrogels for disc regeneration. Eur. Spine J. 2014, 23, 19–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Kim, D.H.; Martin, J.T.; Elliott, D.M.; Smith, L.J.; Mauck, R.L. Phenotypic stability, matrix elaboration and functional maturation of nucleus pulposus cells encapsulated in photocrosslinkable hyaluronic acid hydrogels. Acta Biomater. 2015, 12, 21–29. [Google Scholar] [CrossRef] [Green Version]
  208. Tsaryk, R.; Gloria, A.; Russo, T.; Anspach, L.; De Santis, R.; Ghanaati, S.; Unger, R.E.; Ambrosio, L.; Kirkpatrick, C.J. Collagen-low molecular weight hyaluronic acid semi-interpenetrating network loaded with gelatin microspheres for cell and growth factor delivery for nucleus pulposus regeneration. Acta Biomater. 2015, 20, 10–21. [Google Scholar] [CrossRef] [PubMed]
  209. Mohd Isa, I.L.; Abbah, S.A.; Kilcoyne, M.; Sakai, D.; Dockery, P.; Finn, D.P.; Pandit, A. Implantation of hyaluronic acid hydrogel prevents the pain phenotype in a rat model of intervertebral disc injury. Sci. Adv. 2018, 4, eaaq0597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Omlor, G.W.; Nerlich, A.G.; Lorenz, H.; Bruckner, T.; Richter, W.; Pfeiffer, M.; Guhring, T. Injection of a polymerized hyaluronic acid/collagen hydrogel matrix in an in vivo porcine disc degeneration model. Eur. Spine J. 2012, 21, 1700–1708. [Google Scholar] [CrossRef] [Green Version]
  211. Bron, J.L.; Koenderink, G.H.; Everts, V.; Smit, T.H. Rheological characterization of the nucleus pulposus and dense collagen scaffolds intended for functional replacement. J. Orthop. Res. 2009, 27, 620–626. [Google Scholar] [CrossRef] [PubMed]
  212. Bron, J.L.; Mulder, H.W.; Vonk, L.A.; Doulabi, B.Z.; Oudhoff, M.J.; Smit, T.H. Migration of intervertebral disc cells into dense collagen scaffolds intended for functional replacement. J. Mater. Sci. Mater. Med. 2012, 23, 813–821. [Google Scholar] [CrossRef] [Green Version]
  213. Cen, L.; Liu, W.; Cui, L.; Zhang, W.; Cao, Y. Collagen tissue engineering: Development of novel biomaterials and applications. Pediatr. Res. 2008, 63, 492–496. [Google Scholar] [CrossRef] [PubMed]
  214. Cheng, Y.H.; Yang, S.H.; Su, W.Y.; Chen, Y.C.; Yang, K.C.; Cheng, W.T.; Wu, S.C.; Lin, F.H. Thermosensitive chitosan-gelatin-glycerol phosphate hydrogels as a cell carrier for nucleus pulposus regeneration: An in vitro study. Tissue Eng. Part. A 2010, 16, 695–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Malhotra, N.R.; Han, W.M.; Beckstein, J.; Cloyd, J.; Chen, W.; Elliott, D.M. An injectable nucleus pulposus implant restores compressive range of motion in the ovine disc. Spine 2012, 37, E1099–E1105. [Google Scholar] [CrossRef] [Green Version]
  216. Nicodemus, G.D.; Bryant, S.J. Cell encapsulation in biodegradable hydrogels for tissue engineering applications. Tissue Eng Part. B Rev. 2008, 14, 149–165. [Google Scholar] [CrossRef]
  217. Strange, D.G.; Oyen, M.L. Composite hydrogels for nucleus pulposus tissue engineering. J. Mech Behav. Biomed. Mater. 2012, 11, 16–26. [Google Scholar] [CrossRef]
  218. Wang, L.; Stegemann, J.P. Thermogelling chitosan and collagen composite hydrogels initiated with beta-glycerophosphate for bone tissue engineering. Biomaterials 2010, 31, 3976–3985. [Google Scholar] [CrossRef] [Green Version]
  219. Wilke, H.J.; Heuer, F.; Neidlinger-Wilke, C.; Claes, L. Is a collagen scaffold for a tissue engineered nucleus replacement capable of restoring disc height and stability in an animal model? Eur. Spine J. 2006, 15 (Suppl. 3), 433–438. [Google Scholar] [CrossRef] [Green Version]
  220. Takeoka, Y.; Yurube, T.; Morimoto, K.; Kunii, S.; Kanda, Y.; Tsujimoto, R.; Kawakami, Y.; Fukase, N.; Takemori, T.; Omae, K.; et al. Reduced nucleotomy-induced intervertebral disc disruption through spontaneous spheroid formation by the Low Adhesive Scaffold Collagen (LASCol). Biomaterials 2020, 235, 119781. [Google Scholar] [CrossRef]
  221. Friedmann, A.; Baertel, A.; Schmitt, C.; Ludtka, C.; Milosevic, J.; Meisel, H.J.; Goehre, F.; Schwan, S. Intervertebral Disc Regeneration Injection of a Cell-Loaded Collagen Hydrogel in a Sheep Model. Int. J. Mol. Sci. 2021, 22, 4248. [Google Scholar] [CrossRef]
  222. Wang, Y.; Wang, X.; Shang, J.; Liu, H.; Yuan, Y.; Guo, Y.; Huang, B.; Zhou, Y. Repairing the ruptured annular fibrosus by using type I collagen combined with citric acid, EDC and NHS: An in vivo study. Eur. Spine J. 2017, 26, 884–893. [Google Scholar] [CrossRef] [PubMed]
  223. Sloan, S.R., Jr.; Wipplinger, C.; Kirnaz, S.; Navarro-Ramirez, R.; Schmidt, F.; McCloskey, D.; Pannellini, T.; Schiavinato, A.; Hartl, R.; Bonassar, L.J. Combined nucleus pulposus augmentation and annulus fibrosus repair prevents acute intervertebral disc degeneration after discectomy. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  224. Bertolo, A.; Mehr, M.; Aebli, N.; Baur, M.; Ferguson, S.J.; Stoyanov, J.V. Influence of different commercial scaffolds on the in vitro differentiation of human mesenchymal stem cells to nucleus pulposus-like cells. Eur. Spine J. 2012, 21 (Suppl. 6), 826–838. [Google Scholar] [CrossRef] [Green Version]
  225. Cheng, Y.H.; Yang, S.H.; Liu, C.C.; Gefen, A.; Lin, F.H. Thermosensitive hydrogel made of ferulic acid-gelatin and chitosan glycerophosphate. Carbohydr. Polym. 2013, 92, 1512–1519. [Google Scholar] [CrossRef] [PubMed]
  226. Iwasaki, N.; Yamane, S.T.; Majima, T.; Kasahara, Y.; Minami, A.; Harada, K.; Nonaka, S.; Maekawa, N.; Tamura, H.; Tokura, S.; et al. Feasibility of polysaccharide hybrid materials for scaffolds in cartilage tissue engineering: Evaluation of chondrocyte adhesion to polyion complex fibers prepared from alginate and chitosan. Biomacromolecules 2004, 5, 828–833. [Google Scholar] [CrossRef]
  227. Di Martino, A.; Sittinger, M.; Risbud, M.V. Chitosan: A versatile biopolymer for orthopaedic tissue-engineering. Biomaterials 2005, 26, 5983–5990. [Google Scholar] [CrossRef] [PubMed]
  228. Richardson, S.M.; Hughes, N.; Hunt, J.A.; Freemont, A.J.; Hoyland, J.A. Human mesenchymal stem cell differentiation to NP-like cells in chitosan-glycerophosphate hydrogels. Biomaterials 2008, 29, 85–93. [Google Scholar] [CrossRef]
  229. Roughley, P.; Hoemann, C.; DesRosiers, E.; Mwale, F.; Antoniou, J.; Alini, M. The potential of chitosan-based gels containing intervertebral disc cells for nucleus pulposus supplementation. Biomaterials 2006, 27, 388–396. [Google Scholar] [CrossRef]
  230. Smith, L.J.; Gorth, D.J.; Showalter, B.L.; Chiaro, J.A.; Beattie, E.E.; Elliott, D.M.; Mauck, R.L.; Chen, W.; Malhotra, N.R. In vitro characterization of a stem-cell-seeded triple-interpenetrating-network hydrogel for functional regeneration of the nucleus pulposus. Tissue Eng. Part A 2014, 20, 1841–1849. [Google Scholar] [CrossRef] [Green Version]
  231. Sun, W.; Zhang, K.; Liu, G.; Ding, W.; Zhao, C.; Xie, Y.; Yuan, J.; Sun, X.; Li, H.; Liu, C.; et al. Sox9 gene transfer enhanced regenerative effect of bone marrow mesenchymal stem cells on the degenerated intervertebral disc in a rabbit model. PLoS ONE 2014, 9, e93570. [Google Scholar] [CrossRef] [Green Version]
  232. Reza, A.T.; Nicoll, S.B. Characterization of novel photocrosslinked carboxymethylcellulose hydrogels for encapsulation of nucleus pulposus cells. Acta Biomater. 2010, 6, 179–186. [Google Scholar] [CrossRef]
  233. Lin, H.A.; Varma, D.M.; Hom, W.W.; Cruz, M.A.; Nasser, P.R.; Phelps, R.G.; Iatridis, J.C.; Nicoll, S.B. Injectable cellulose-based hydrogels as nucleus pulposus replacements: Assessment of in vitro structural stability, ex vivo herniation risk, and in vivo biocompatibility. J. Mech. Behav. Biomed. Mater. 2019, 96, 204–213. [Google Scholar] [CrossRef]
  234. Malafaya, P.B.; Silva, G.A.; Reis, R.L. Natural-origin polymers as carriers and scaffolds for biomolecules and cell delivery in tissue engineering applications. Adv. Drug Deliv. Rev. 2007, 59, 207–233. [Google Scholar] [CrossRef] [Green Version]
  235. Thorpe, A.A.; Boyes, V.L.; Sammon, C.; Le Maitre, C.L. Thermally triggered injectable hydrogel, which induces mesenchymal stem cell differentiation to nucleus pulposus cells: Potential for regeneration of the intervertebral disc. Acta Biomater. 2016, 36, 99–111. [Google Scholar] [CrossRef] [PubMed]
  236. Vadala, G.; Russo, F.; Musumeci, M.; D’Este, M.; Cattani, C.; Catanzaro, G.; Tirindelli, M.C.; Lazzari, L.; Alini, M.; Giordano, R.; et al. Clinically relevant hydrogel-based on hyaluronic acid and platelet rich plasma as a carrier for mesenchymal stem cells: Rheological and biological characterization. J. Orthop. Res. 2017, 35, 2109–2116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Lin, H.A.; Gupta, M.S.; Varma, D.M.; Gilchrist, M.L.; Nicoll, S.B. Lower crosslinking density enhances functional nucleus pulposus-like matrix elaboration by human mesenchymal stem cells in carboxymethylcellulose hydrogels. J. Biomed. Mater. Res. A 2016, 104, 165–177. [Google Scholar] [CrossRef] [PubMed]
  238. Wachs, R.A.; Hoogenboezem, E.N.; Huda, H.I.; Xin, S.; Porvasnik, S.L.; Schmidt, C.E. Creation of an injectable in situ gelling native extracellular matrix for nucleus pulposus tissue engineering. Spine J. 2017, 17, 435–444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Zhang, Y.; Drapeau, S.; Howard, S.A.; Thonar, E.J.; Anderson, D.G. Transplantation of goat bone marrow stromal cells to the degenerating intervertebral disc in a goat disc injury model. Spine 2011, 36, 372–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  240. Gilbert, H.T.J.; Hodson, N.; Baird, P.; Richardson, S.M.; Hoyland, J.A. Acidic pH promotes intervertebral disc degeneration: Acid-sensing ion channel -3 as a potential therapeutic target. Sci. Rep. 2016, 6, 37360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Huang, Y.C.; Urban, J.P.; Luk, K.D. Intervertebral disc regeneration: Do nutrients lead the way? Nat. Rev. Rheumatol. 2014, 10, 561–566. [Google Scholar] [CrossRef] [PubMed]
  242. Newell, N.; Little, J.P.; Christou, A.; Adams, M.A.; Adam, C.J.; Masouros, S.D. Biomechanics of the human intervertebral disc: A review of testing techniques and results. J. Mech. Behav. Biomed. Mater. 2017, 69, 420–434. [Google Scholar] [CrossRef]
  243. Risbud, M.V.; Shapiro, I.M. Role of cytokines in intervertebral disc degeneration: Pain and disc content. Nat. Rev. Rheumatol. 2014, 10, 44–56. [Google Scholar] [CrossRef]
  244. Schmidt, H.; Reitmaier, S.; Graichen, F.; Shirazi-Adl, A. Review of the fluid flow within intervertebral discs - How could in vitro measurements replicate in vivo? J. Biomech. 2016, 49, 3133–3146. [Google Scholar] [CrossRef]
  245. Sakai, D.; Nakamura, Y.; Nakai, T.; Mishima, T.; Kato, S.; Grad, S.; Alini, M.; Risbud, M.V.; Chan, D.; Cheah, K.S.; et al. Exhaustion of nucleus pulposus progenitor cells with ageing and degeneration of the intervertebral disc. Nat. Commun. 2012, 3, 1264. [Google Scholar] [CrossRef] [Green Version]
  246. Yamada, K.; Kenichiro, M.; Ito, Y.M.; Inage, F.; Isoe, T.; Yokota, N.; Sugita, O.; Sato, N.; Tha, K.K.; Iwasaki, N.; et al. Exploratory clinical trial on the safety and capability of dMD-001. in lumbar disc herniation: Study protocol for a first-in-human pilot study. Contemp. Clin. Trials Commun. 2021, 23, 100805. [Google Scholar] [CrossRef] [PubMed]
  247. Miyagi, M.; Ishikawa, T.; Kamoda, H.; Suzuki, M.; Murakami, K.; Shibayama, M.; Orita, S.; Eguchi, Y.; Arai, G.; Sakuma, Y.; et al. ISSLS prize winner: Disc dynamic compression in rats produces long-lasting increases in inflammatory mediators in discs and induces long-lasting nerve injury and regeneration of the afferent fibers innervating discs: A pathomechanism for chronic discogenic low back pain. Spine 2012, 37, 1810–1818. [Google Scholar] [CrossRef]
  248. Burke, J.G.; Watson, R.W.; McCormack, D.; Dowling, F.E.; Walsh, M.G.; Fitzpatrick, J.M. Intervertebral discs which cause low back pain secrete high levels of proinflammatory mediators. J. Bone Joint. Surg. Br. 2002, 84, 196–201. [Google Scholar] [CrossRef]
  249. Freemont, A.J.; Watkins, A.; Le Maitre, C.; Baird, P.; Jeziorska, M.; Knight, M.T.; Ross, E.R.; O’Brien, J.P.; Hoyland, J.A. Nerve growth factor expression and innervation of the painful intervertebral disc. J. Pathol. 2002, 197, 286–292. [Google Scholar] [CrossRef] [PubMed]
  250. Lotz, J.C.; Ulrich, J.A. Innervation, inflammation, and hypermobility may characterize pathologic disc degeneration: Review of animal model data. J. Bone Jt. Surg. Am. 2006, 88 (Suppl. 2), 76–82. [Google Scholar] [CrossRef]
  251. Aoki, Y.; Takahashi, Y.; Ohtori, S.; Moriya, H.; Takahashi, K. Distribution and immunocytochemical characterization of dorsal root ganglion neurons innervating the lumbar intervertebral disc in rats: A review. Life Sci. 2004, 74, 2627–2642. [Google Scholar] [CrossRef] [PubMed]
  252. Woolf, C.J.; Allchorne, A.; Safieh-Garabedian, B.; Poole, S. Cytokines, nerve growth factor and inflammatory hyperalgesia: The contribution of tumour necrosis factor alpha. Br. J. Pharmacol. 1997, 121, 417–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Sobajima, S.; Kim, J.S.; Gilbertson, L.G.; Kang, J.D. Gene therapy for degenerative disc disease. Gene Ther. 2004, 11, 390–401. [Google Scholar] [CrossRef] [PubMed]
  254. Hallen, A. Hexosamine and ester sulfate content of human nucleus pulposus at different ages. Acta Chem. Scand. 1958, 12, 1869–1872. [Google Scholar] [CrossRef] [Green Version]
  255. Hallen, A. The collagen and ground substance of the human nucleus pulposus at different ages. Acta Chem. Scand. 1962, 16, 705–709. [Google Scholar] [CrossRef] [Green Version]
  256. Vernengo, J.; Fussell, G.W.; Smith, N.G.; Lowman, A.M. Evaluation of novel injectable hydrogels for nucleus pulposus replacement. J. Biomed. Mater. Res. B. Appl. Biomater. 2008, 84, 64–69. [Google Scholar] [CrossRef]
  257. DiStefano, T.J.; Shmukler, J.O.; Danias, G.; Iatridis, J.C. The Functional Role of Interface Tissue Engineering in Annulus Fibrosus Repair: Bridging Mechanisms of Hydrogel Integration with Regenerative Outcomes. ACS Biomater. Sci. Eng. 2020, 6, 6556–6586. [Google Scholar] [CrossRef]
  258. Gullbrand, S.E.; Schaer, T.P.; Agarwal, P.; Bendigo, J.R.; Dodge, G.R.; Chen, W.; Elliott, D.M.; Mauck, R.L.; Malhotra, N.R.; Smith, L.J. Translation of an injectable triple-interpenetrating-network hydrogel for intervertebral disc regeneration in a goat model. Acta Biomater. 2017, 60, 201–209. [Google Scholar] [CrossRef]
  259. Vergroesen, P.P.; Bochyn Ska, A.I.; Emanuel, K.S.; Sharifi, S.; Kingma, I.; Grijpma, D.W.; Smit, T.H. A biodegradable glue for annulus closure: Evaluation of strength and endurance. Spine 2015, 40, 622–628. [Google Scholar] [CrossRef]
  260. Cruz, M.A.; Hom, W.W.; DiStefano, T.J.; Merrill, R.; Torre, O.M.; Lin, H.A.; Hecht, A.C.; Illien-Junger, S.; Iatridis, J.C. Cell-Seeded Adhesive Biomaterial for Repair of Annulus Fibrosus Defects in Intervertebral Discs. Tissue Eng. Part. A. 2018, 24, 187–198. [Google Scholar] [CrossRef]
  261. Long, R.G.; Rotman, S.G.; Hom, W.W.; Assael, D.J.; Illien-Junger, S.; Grijpma, D.W.; Iatridis, J.C. In vitro and biomechanical screening of polyethylene glycol and poly(trimethylene carbonate) block copolymers for annulus fibrosus repair. J. Tissue Eng. Regen. Med. 2018, 12, 727–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Chik, T.K.; Ma, X.Y.; Choy, T.H.; Li, Y.Y.; Diao, H.J.; Teng, W.K.; Han, S.J.; Cheung, K.M.; Chan, B.P. Photochemically crosslinked collagen annulus plug: A potential solution solving the leakage problem of cell-based therapies for disc degeneration. Acta Biomater. 2013, 9, 8128–8139. [Google Scholar] [CrossRef] [PubMed]
  263. DiStefano, T.J.; Shmukler, J.O.; Danias, G.; Di Pauli von Treuheim, T.; Hom, W.W.; Goldberg, D.A.; Laudier, D.M.; Nasser, P.R.; Hecht, A.C.; Nicoll, S.B.; et al. Development of a two-part biomaterial adhesive strategy for annulus fibrosus repair and ex vivo evaluation of implant herniation risk. Biomaterials 2020, 258, 120309. [Google Scholar] [CrossRef] [PubMed]
  264. Wilke, H.J.; Ressel, L.; Heuer, F.; Graf, N.; Rath, S. Can prevention of a reherniation be investigated? Establishment of a herniation model and experiments with an anular closure device. Spine 2013, 38, 587–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Amin, R.M.; Andrade, N.S.; Neuman, B.J. Lumbar Disc Herniation. Curr. Rev. Musculoskelet. Med. 2017, 10, 507–516. [Google Scholar] [CrossRef]
  266. Engel-Yeger, B.; Keren, A.; Berkovich, Y.; Sarfaty, E.; Merom, L. The role of physical status versus mental status in predicting the quality of life of patients with lumbar disk herniation. Disabil. Rehabil. 2018, 40, 302–308. [Google Scholar] [CrossRef]
  267. Andersson, G.B. Epidemiological features of chronic low-back pain. Lancet 1999, 354, 581–585. [Google Scholar] [CrossRef]
  268. Seki, S.; Kawaguchi, Y.; Chiba, K.; Mikami, Y.; Kizawa, H.; Oya, T.; Mio, F.; Mori, M.; Miyamoto, Y.; Masuda, I.; et al. A functional SNP in CILP, encoding cartilage intermediate layer protein, is associated with susceptibility to lumbar disc disease. Nat. Genet. 2005, 37, 607–612. [Google Scholar] [CrossRef]
  269. Long, R.G.; Zderic, I.; Gueorguiev, B.; Ferguson, S.J.; Alini, M.; Grad, S.; Iatridis, J.C. Effects of Level, Loading Rate, Injury and Repair on Biomechanical Response of Ovine Cervical Intervertebral Discs. Ann. Biomed. Eng. 2018, 46, 1911–1920. [Google Scholar] [CrossRef]
  270. Long, R.G.; Burki, A.; Zysset, P.; Eglin, D.; Grijpma, D.W.; Blanquer, S.B.G.; Hecht, A.C.; Iatridis, J.C. Mechanical restoration and failure analyses of a hydrogel and scaffold composite strategy for annulus fibrosus repair. Acta Biomater. 2016, 30, 116–125. [Google Scholar] [CrossRef] [Green Version]
  271. Scheibler, A.G.; Gotschi, T.; Widmer, J.; Holenstein, C.; Steffen, T.; Camenzind, R.S.; Snedeker, J.G.; Farshad, M. Feasibility of the annulus fibrosus repair with in situ gelating hydrogels - A biomechanical study. PLoS ONE 2018, 13, e0208460. [Google Scholar] [CrossRef] [Green Version]
  272. Borde, B.; Grunert, P.; Hartl, R.; Bonassar, L.J. Injectable, high-density collagen gels for annulus fibrosus repair: An in vitro rat tail model. J. Biomed. Mater. Res. A 2015, 103, 2571–2581. [Google Scholar] [CrossRef] [Green Version]
  273. Sloan, S.R., Jr.; Galesso, D.; Secchieri, C.; Berlin, C.; Hartl, R.; Bonassar, L.J. Initial investigation of individual and combined annulus fibrosus and nucleus pulposus repair ex vivo. Acta Biomater. 2017, 59, 192–199. [Google Scholar] [CrossRef]
  274. Grunert, P.; Borde, B.H.; Towne, S.B.; Moriguchi, Y.; Hudson, K.D.; Bonassar, L.J.; Hartl, R. Riboflavin crosslinked high-density collagen gel for the repair of annular defects in intervertebral discs: An in vivo study. Acta Biomater. 2015, 26, 215–224. [Google Scholar] [CrossRef]
  275. Pennicooke, B.; Hussain, I.; Berlin, C.; Sloan, S.R.; Borde, B.; Moriguchi, Y.; Lang, G.; Navarro-Ramirez, R.; Cheetham, J.; Bonassar, L.J.; et al. Annulus Fibrosus Repair Using High-Density Collagen Gel: An In Vivo Ovine Model. Spine 2018, 43, 208–215. [Google Scholar] [CrossRef]
  276. Moriguchi, Y.; Borde, B.; Berlin, C.; Wipplinger, C.; Sloan, S.R.; Kirnaz, S.; Pennicooke, B.; Navarro-Ramirez, R.; Khair, T.; Grunert, P.; et al. In vivo annular repair using high-density collagen gel seeded with annulus fibrosus cells. Acta Biomater. 2018, 79, 230–238. [Google Scholar] [CrossRef]
  277. Brockmann, W.; Geiß, P.L.; Klingen, J.; Schrder, B. Adhesion. In Adhesive Bonding; Wiley-VCH Verlag GmbH & Co. KGaA: Weinheim, Germany, 2008; pp. 11–28. [Google Scholar]
  278. Ghobril, C.; Grinstaff, M.W. The chemistry and engineering of polymeric hydrogel adhesives for wound closure: A tutorial. Chem. Soc. Rev. 2015, 44, 1820–1835. [Google Scholar] [CrossRef]
  279. Ebnesajjad, S.L. Introduction and adhesion theories. In Adhesives Technology Handbook; Elsevier: Amsterdam, The Netherlands, 2009; pp. 1–19. [Google Scholar]
  280. Ebnesajjad, S. Theories of Adhesion. In Surface Treatment of Materials for Adhesive Bonding; Elsevier: Amsterdam, The Netherlands, 2014; pp. 77–91. [Google Scholar]
  281. Jabbari, E.P.; Peppas, N.A. Polymer-Polymer Interdiffusion and Adhesion. J. Macromol. Sci. Polym. Rev. 1994, 34, 205–241. [Google Scholar] [CrossRef]
  282. Epherre, R.; Goglio, G.; Mornet, S.; Duguet, E. Hybrid magnetic nanoparticles for targeted delivery. In Biocompatibility, Surface Engineering, and Delivery of Drugs, Genes and Other Molecules; Elsevier: Amsterdam, The Netherlands, 2017; Volume 4, pp. 750–771. [Google Scholar]
  283. Ngo, B.K.D.; Grunlan, M.A. Protein Resistant Polymeric Biomaterials. ACS Macro. Lett. 2017, 6, 992–1000. [Google Scholar] [CrossRef] [Green Version]
  284. Grunert, P.; Hudson, K.D.; Macielak, M.R.; Aronowitz, E.; Borde, B.H.; Alimi, M.; Njoku, I.; Ballon, D.; Tsiouris, A.J.; Bonassar, L.J.; et al. Assessment of intervertebral disc degeneration based on quantitative magnetic resonance imaging analysis: An in vivo study. Spine 2014, 39, 369–378. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Biomaterial-based and cell-based intervertebral disc regeneration treatment strategies based on the stage of disc degeneration.
Figure 1. Biomaterial-based and cell-based intervertebral disc regeneration treatment strategies based on the stage of disc degeneration.
Cells 11 00602 g001
Figure 2. Normal to degenerated lumbar intervertebral discs in rabbits. The initial degeneration of intervertebral discs occurs primarily in the nucleus, making the gelatinous nucleus pulposus a promising target for therapeutic approaches using soft biomaterials.
Figure 2. Normal to degenerated lumbar intervertebral discs in rabbits. The initial degeneration of intervertebral discs occurs primarily in the nucleus, making the gelatinous nucleus pulposus a promising target for therapeutic approaches using soft biomaterials.
Cells 11 00602 g002
Figure 3. Herniated disc model with a 4.5 mm diameter hole in a pig lumbar disc injected with hydrogel. The hydrogel, which has little adhesive ability, easily extrudes from the disc under compressive load.
Figure 3. Herniated disc model with a 4.5 mm diameter hole in a pig lumbar disc injected with hydrogel. The hydrogel, which has little adhesive ability, easily extrudes from the disc under compressive load.
Cells 11 00602 g003
Table 1. Candidate cell sources for intervertebral disc (IVD) regeneration therapy.
Table 1. Candidate cell sources for intervertebral disc (IVD) regeneration therapy.
Cell SourcesReferences
Differentiated CellsIVD-derived cells (nucleus pulposus (NP)-derived cells)[25,26,27,28,29,30,31,32,33,34,35,36,37,38]
Chondrocyte-like cells (including chondrocytes derived articular cartilage)[39,40,41]
Stem CellsMesenchymal stem cells (MSCs)
Bone marrow-derived MSCs[26,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59]
Adipose-derived MSCs[34,60,61,62,63,64,65,66]
Synovial-derived MSCs[67]
Nucleus pulposus-derived MSCs[68]
Induced pluripotent stem (iPS) cells[69,70,71,72,73,74]
Embryonic stem (ES) cells[75,76,77]
Bone marrow aspirate concentrate (BMAC)[78,79,80]
Table 2. Clinical trials on cell-based intervertebral disc (IVD) regeneration therapy.
Table 2. Clinical trials on cell-based intervertebral disc (IVD) regeneration therapy.
Cell TypeModeCarrierAdministration MethodIndicationnOutcomeReferences
Differentiated CellsIntervertebral disc cellsAutologousNonePercutaneous injectionLumbar disc herniation at 12 weeks postoperatively112Improvement in pain, disc hydration improved on MRI[28,36]
Activated nucleus pulposus cellsAutologousNonePercutaneous injectionDisc degeneration adjacent to fused disc9No progression of disc degeneration[25]
Juvenile articular chondrocytesAllogenicFibrinPercutaneous injectionDegenerative disc disease with low back pain15Improvement in pain and clinical indices, and on MRI[39]
Stem CellsBone marrow MSCsAutologousCollagen spongePercutaneous injectionLumbar spinal canal stenosis2Vacuum phenomenon and motion segment instability improved on radiograph, hydration improved on MRI [51]
Bone marrow MSCsAutologousNonePercutaneous injectionChronic low back pain10Rapid improvement in pain and disability, hydration improved on MRI[52]
Bone marrow MSCsAutologousNonePercutaneous injectionDegenerative disc disease with low back pain5Self-reported overall improvement, improvement in strength and mobility[58]
Bone marrow MSCsAllogenicNonePercutaneous injectionDegenerative disc disease with low back pain24Improvement in pain and disability, and on quantitative MRI[59]
Adipose-derived MSCsAutologousHyaluronic acidPercutaneous injectionChronic discogenic low back pain10Improvement in pain and clinical indices[64]
Bone marrow concentrateAutologousNonePercutaneous injectionDiscogenic low back pain26Improvement in pain and clinical indices[79,80]
Table 3. Candidate biomaterials for intervertebral disc regeneration therapy.
Table 3. Candidate biomaterials for intervertebral disc regeneration therapy.
BiomaterialsReferences
Synthetic BiomaterialsPolylactic acid (PLA), Polyglycolic acid (PGA), Polylactic-co-glycolic acid (PLGA)[38,63,123,124,125,126,127,128,129,130,131]
Polyethylene glycol (PEG)[132,133,134,135,136,137,138,139,140,141,142]
Polycarbonate urethane (PU)[143,144,145,146,147,148]
Poly epsilon-caprolactone (PCL)[149,150,151,152,153,154,155]
Natural BiomaterialsAlginate[7,66,78,92,100,128,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172]
Agarose[4,173,174,175,176,177,178,179]
Fibrin[39,41,49,90,101,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196]
Hyaluronic acid[57,64,132,140,142,173,185,188,197,198,199,200,201,202,203,204,205,206,207,208,209,210]
Collagen[65,132,142,197,210,211,212,213,214,215,216,217,218,219,220,221,222,223]
Chitosan[66,172,214,218,224,225,226,227,228,229,230,231]
Carboxymethylcellulose[232,233]
Table 4. Soft biomaterials as candidates for cell-free intervertebral disc (IVD) regeneration therapy.
Table 4. Soft biomaterials as candidates for cell-free intervertebral disc (IVD) regeneration therapy.
Composition of Soft BiomaterialsAbbreviationClinical Trials/PreclinicalMechanism of RegenerationRef.
AlginateUPAL (ultra-purified alginate)Clinical (in progress)/Preclinical (in vivo, rabbit, sheep)Induction of endogenous NP cells and NP progenitor cells (GD2Tie2 cells), leading to endogenous IVD repair[7,246]
CollagenLASCOL (low adhesive scaffold collagen)Preclinical (in vivo, rat)Promotion of the formation of cell aggregative spheroids that facilitate the maintenance of the original disc NP phenotype, upregulation of the expression of chondrogenic genes[220]
FibrinFibrin sealantClinical/Preclinical (in vivo, rat)Suppression of the acute proinflammatory cytokine (TNF-α, IL-1β, IL-6) production, increasing expression of pro-resolution cytokines (IL-4, TGF-β), inhibiting nucleotomy-induced progressive fibrosis of the NP[192,195]
Hyaluronic acidHMW HA (high molecular weight hyaluronic acid microgel)Preclinical (in vivo, rat)Regulation of inflammation by downregulating IFNα, reduction in cell death by suppressing expression of IGFBP3 and caspase-3 fragment p17, induction of the production of extracellular matrix[205]
Table 5. Biomechanical evaluation of soft biomaterials for intervertebral disc (IVD) repair after a discectomy or annulus fibrosus injury.
Table 5. Biomechanical evaluation of soft biomaterials for intervertebral disc (IVD) repair after a discectomy or annulus fibrosus injury.
Composition of Soft BiomaterialsIVD Model (Ex Vivo)Biomechanical Evaluation MethodOutcomeReferences
FibrinGenipin cross-linked fibrinBovineCyclic axial tension–compression, torsionFull restoration of compressive stiffness, partial restoration of tensile and torsional stiffness[186]
OvineCyclic axial tension–compression, torsionRestoration of axial range of motion and torque range[269]
BovineCyclic flexion–extension, torsion, bendingRestoration of torsional stiffness, bending range of motion, low risk of herniation in bending and compression[270]
BovineRamp-to-failure testLow risk of herniation[271]
Collagen hydrogelRiboflavin cross-linked collagenOvineCyclic axial tension–compression, torsionRestoration of torsional stiffness and torque range (combined with nucleus pulposus augmentation using hyaluronic acid) [223]
RatAxial compression (uniaxial stress-relaxation)Improvement in effective equilibrium and instantaneous
moduli (combined with nucleus pulposus augmentation using hyaluronic acid)
[273]
RatAxial compression (uniaxial stress-relaxation)Improvement in effective equilibrium and instantaneous
moduli
[272]
Rose Bengal cross-linked collagenRabbitCyclic axial compression, torsion push-out testNo extrusion after loading (40,320 cycles with 0.4 to 0.8 MPa compressive loading, 0–25 degree torsion)[262]
AlginateUltra-purified alginate (UPAL)OvineStatic axial compression, rotation, flexion–extension, bending,
cyclic axial compression
No extrusion after loading (compression loading test up to 1000 N, or 1000 cycles with −300 N to 300 N of axial loading). Partially restored compression stiffness [7]
ChitosanTriple-interpenetrating-network hydrogel comprised of dextran, chitosan, and teleosteanHumanCyclic axial compressionNo extrusion after loading (10,000 cycles with 0.12 and 0.96 MPa compressive loading) [230]
CelluloseCarboxymethylcelluloseBovineRamp-to-failure test, fatigue endurance testReduction in herniation risk compared to injury group, restoration of failure strength, maximum stiffness, and subsidence to failure.
Restoration of fatigue endurance compared to injury group.
[233]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamada, K.; Iwasaki, N.; Sudo, H. Biomaterials and Cell-Based Regenerative Therapies for Intervertebral Disc Degeneration with a Focus on Biological and Biomechanical Functional Repair: Targeting Treatments for Disc Herniation. Cells 2022, 11, 602. https://doi.org/10.3390/cells11040602

AMA Style

Yamada K, Iwasaki N, Sudo H. Biomaterials and Cell-Based Regenerative Therapies for Intervertebral Disc Degeneration with a Focus on Biological and Biomechanical Functional Repair: Targeting Treatments for Disc Herniation. Cells. 2022; 11(4):602. https://doi.org/10.3390/cells11040602

Chicago/Turabian Style

Yamada, Katsuhisa, Norimasa Iwasaki, and Hideki Sudo. 2022. "Biomaterials and Cell-Based Regenerative Therapies for Intervertebral Disc Degeneration with a Focus on Biological and Biomechanical Functional Repair: Targeting Treatments for Disc Herniation" Cells 11, no. 4: 602. https://doi.org/10.3390/cells11040602

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop