Abstract
Low back pain (LBP) is a common clinical problem and a major cause of physical disability, imposing a prominent socioeconomic burden. Intervertebral disc degeneration (IDD) has been considered the main cause of LBP. The current treatments have limited efficacy because they cannot address the underlying degeneration. With an increased understanding of the complex pathological mechanism of IDD, various medications and biological reagents have been used for intradiscal injection for the treatment of LBP. There is increasing clinical evidence showing the benefits of these therapies on symptomatic relief and their potential for disc repair and regeneration by targeting the disrupted pathways underlying the cause of the disease. A brief overview of the potential and limitations for these therapies are provided in this review, based on the recent and available data from clinical trials and systematic reviews. Finally, future perspectives are discussed.
Keywords: intervertebral disc degeneration, intradiscal injection, low back pain
Intervertebral disc degeneration (IDD) has been thought to be the main cause of low back pain (LBP). A variety of medications and biologic reagents are used for intradiscal injection for the treatment of LBP. The combination injection of stem cells and hydrogel‐based biomaterials may be the ideal candidates for LBP.
1. INTRODUCTION
Low back pain (LBP) is a common symptom that occurs below the costal margin and above the inferior gluteal fold, which refers to pain, muscle tension, or stiffness. The global prevalence of LBP in 2017 was 7.83%, and 577 million people were affected at any time. 1 In 2019, a systematic review of 13 studies from northern Europe, North America, and Israel reported that the prevalence of LBP ranged between 14% and 20%. 2 A systematic review and meta‐analysis revealed that the lifetime prevalence of LBP was 47% in low‐, lower‐middle‐, and upper‐middle‐income countries in Africa. 3 LBP is a major cause of physical disability in people of all ages and socioeconomic statuses, which places a prominent socioeconomic burden on public health. 4 In a recent study assessing the incidence, prevalence, and years lived with disability associated with 354 diseases, LBP was identified as the leading cause of worldwide productivity loss and of years lived with disability in 126 countries. 5 Moreover, the economic burden of LBP is approximately £2.8 billion in the United Kingdom and more than $100 billion in the United States per year. 6 , 7
Intervertebral disc degeneration (IDD) is considered a major cause of LBP, 8 which also causes other musculoskeletal and spine diseases, such as disc herniation, spinal stenosis, structural instability, and spondylosis. The etiology of IDD is multifactorial, including genetic predisposition, abnormal biomechanical loading, decreased nutrient transport, aging, and lifestyle factors. 9 The development of IDD is characterized by certain pathological features, including elevated inflammatory cytokine levels, progressive loss of the extracellular matrix (ECM), changes in cell phenotype, and increased cell senescence and death. These cellular and biochemical changes further lead to progressive functional and structural impairment.
Treatment for LBP can be divided into three stages. Conservative therapy should be considered as the first line of care in case of an acute episode, including the use of nonsteroidal anti‐inflammatory drugs (NSAIDs), muscle relaxants, opioids, and antitumor necrosis factor antagonists, in conjunction with nonpharmacological strategies such as traction, manipulation, and physical therapy, all of which lead to improvement in most patients. 10 If the symptoms do not subside after the conservative therapy, more invasive treatments are generally considered, including epidural injections, facet injections, and radiofrequency ablations. Finally, if patients are refractory to the above treatments or experience progressive neurological deficits, surgery is recommended, such as endoscopic lumbar discectomy, posterior lumbar interbody fusion, and disc replacement. Unfortunately, to date, all treatments have limited efficacy, and no specific therapies for IDD exist.
Hence, there is a strong clinical demand for the development of regenerative therapy to restore and maintain the native disc structure and mechanical function. Based on the ongoing investigations and understanding of the pathological mechanism of IDD, increasing biological approaches have shown their benefit for disc repair and regeneration. 11 Presently, the regenerative therapy for IDD can be divided into three categories: gene therapy, cell therapy, and tissue engineering biomaterials. 12 , 13 Especially, cell therapy and tissue engineering approach targeting the multiple disrupted pathways underlying the cause of IDD are considered to be potential therapeutic strategies. 14 There are growing clinical investigations and trials illuminating the potential for these therapies in pain control and disc regeneration.
Intradiscal injection is a minimally invasive outpatient procedure for the treatment of LBP. In this procedure, a needle is inserted in the nucleus pulposus (NP) via a percutaneous approach. Usually, this procedure is performed under imaging guidance such as real‐time multiplanar fluoroscopy and computed tomography (CT), to improve the success rate and reduce adverse events. Recently, the use of ultrasound for guiding intradiscal injection has made this procedure more convenient, safer, and decreased the physical load and radiation exposure. 15 , 16
The common medications used for intradiscal injection include oxygen‐ozone (O2‐O3) mixture, steroids, methylene blue, and thermal decompression device, to reduce the inflammatory response or remove the degenerated disc by dehydration and dissolution of the NP tissues. 17 In recent years, various biological reagents and biomaterials, including platelet‐rich plasma (PRP), stem cells, and hydrogel, have been used for intradiscal injection, drawing increased attention for the screening of the ideal injectable medications for IDD. These therapies target the multiple disrupted pathways underlying the cause of the disease and have the potential for disc repair and regeneration. 18 With the increasing variety of biological reagents used for intradiscal injection, there is an urgent need for a review to better illuminate the potential and limitations of these therapies in the treatment of LBP.
Many reports have identified the benefit of intradiscal injection using many biological reagents for disc regeneration in vitro and animal experiments, such as senolytic targeting cellular senescence, antioxidant targeting mitochondrial dysfunction, small molecule natural compound, and some ingredients extracted from the traditional Chinese medicine herb. 19 , 20 , 21 , 22 However, their efficacy and feasibility for intradiscal injection are not confirmed by clinical trials; therefore, these studies were excluded from our review.
Therefore, this article aimed to distill the most recent and available data from clinical trials and systematic reviews, combined with the targeting pathways underlying the pathological mechanism of IDD, to illustrate the effectiveness of intradiscal injection of different medications or biological reagents in the treatment of LBP, and provide more comprehensive and authentic evidence for the selection of these treatments.
2. GLUCOCORTICOIDS
Inflammatory response is considered to be an important cause of LBP. Glucocorticoids are widely used in the treatment of LBP, such as epidural injection, 23 , 24 sacroiliacs joint, 25 nerve root block, 26 injections following discectomy, 27 because of their powerful anti‐inflammatory effects.
In a randomized controlled trial (RCT) conducted by Cao et al, 28 after intradiscal glucocorticoid injection, the visual analog scale (VAS) and Oswestry Disability Index (ODI) scores improved significantly at 3 or 6 months compared to those observed with saline injection. Recently, Nguyen et al 29 revealed in a multicenter RCT that intradiscal injection of methylprednisolone acetate can effectively alleviate the symptoms of LBP at 1 month, but without long‐term efficacy. Moreover, for LBP with active discopathy, intradiscal injection of prednisolone acetate can reduce pain intensity at 1 month but not at 3 and 6 months. 30 Due to the half‐life of glucocorticoids, their anti‐inflammatory effects and efficacy are difficult to maintain for a long time. However, it is unclear whether multiple injections can maintain long‐term efficacy.
3. OXYGEN‐OZONE
Ozone is a strong oxidizing gas that normally exists in the atmosphere with antiseptic, analgesic, immunomodulating, and anti‐inflammatory properties. 31 The intradiscal injection of O2‐O3 mixture, with a concentration range from 10 to 40 μg O3/mL O2, has been widely used in the treatment of LBP in many countries since the 1990s, especially in Europe and Asia, as an alternative minimally invasive, safe, and cost‐effective choice. 32 , 33 Multiple studies have demonstrated its significant improvement of symptoms, because of the following properties: (a) stimulating the activity of fibroblasts to repair the damaged disc by deposition of collagen; (b) increasing the concentration of oxygen in tissues; (c) interrupting the inflammatory cascade of the arachidonic acid; (d) reducing the disc volume by breaking the glycosaminoglycan chains. 34 , 35
A previous meta‐analysis including 12 studies and almost 8000 patients ranging from 13 to 94 years showed that after treatment by oxygen/ozone, the mean improvement was 3.9 for VAS and 25.7 for ODI, the pain and function outcomes were similar to the outcomes treated by a surgical discectomy. Moreover, O2‐O3 treatment had a lower complication rate of 0.064% and a significantly shorter recovery time. 36 A recent systematic review and meta‐regression including 22 articles also highlighted the positive effects in reducing pain and improving function for patients with LBP. 37 Several studies have reported that intradiscal O2‐O3 injection can reduce the disc herniation size and improve the pain quality in the short term, 38 although its benefit can span across 10 years. 39 Furthermore, some studies and meta‐analyses reported that ozone therapy was more effective than other therapies, such as laser disc decompression and steroid injection. 40 , 41 Moreover, a recent RCT reported that intradiscal O2‐O3 injection alone was sufficient to treat LBP and radicular pain, additional periforaminal steroid injection was not beneficial. 42
Although many studies have demonstrated that intradiscal O2‐O3 injection plays a relevant role in the improvement of pain quality for patients with LBP or lumbar disc herniation (LDH), there is insufficient evidence to reinforce strong recommendations. 37 Additionally, to obtain a successful clinical outcome, the indications and selection criteria should be fully understood. The radicular pain caused by LDH is considered the best clinical indication of O2‐O3 treatment, rather than LBP; and patients with neurological motor deficit, cauda equina syndrome, or spinal infection are not recommended for O2‐O3 treatment. 35
4. METHYLENE BLUE
Nociceptive nerves grow into the NP extending from the outer layer of the annulus fibrosis or endplate, which is one of the main causes of chronic discogenic LBP. 43 , 44 Since the first synthesis in 1876, methylene blue has been used for the treatment of many painful ailments and idiopathic pruritus ani, because of its neurolytic effect by blocking nerve conduction or damage to nerve endings. 45 Meanwhile, methylene blue is known to have an anti‐inflammatory effect by inhibiting the synthesis of nitric oxide and generation of free radicals, which is beneficial for relieving pain. 46
Intradiscal methylene blue injection was first reported by Peng et al 47 and was considered an effective and minimally invasive treatment for LBP. Subsequently, its short‐term efficacy was found in other studies by Kim et al 48 and Zhang et al. 49 A meta‐analysis including five clinical studies concluded that intradiscal methylene blue injection can relieve pain symptoms and improve ODI score, which is a safe and effective procedure for the treatment of chronic LBP. 50 However, a recent multicentre RCT showed that, compared to placebo injection, intradiscal methylene blue injection did not exhibit better efficacy, and could not be recommended for patients with chronic discogenic LBP. 51 Further, an in vitro study found that a high concentration of methylene blue can inhibit proliferation and paracrine function of annulus fibrosus cells, even induce cell apoptosis, suggesting that its practical application for the treatment of discogenic LBP should be carefully considered. 52 In the past 2 years, no clinical study was conducted to confirm the efficacy of intradiscal methylene blue injection. Therefore, its safety and efficacy, especially in the long‐term, need to be further assessed.
5. MESENCHYMAL STEM CELLS
IDD usually involves a decrease in cell density, increase in inflammatory factors, and an overall reduction in the synthesis of ECM. 9 Use of cell‐based therapies, especially implantation of autologous or allogeneic mesenchymal stem cells (MSCs) may be a potential therapeutic strategy for early disc degeneration. 53 MSCs from various adult tissues, such as the bone marrow, adipose tissue, and umbilical cord, have become highly topical for disc regeneration in experimental and clinical investigations, because of their potential for differentiating into NP cells (NPCs), promoting the proliferation of NPCs, and promoting the synthesis of the ECM. Meanwhile, MSCs have powerful immunomodulatory properties and the ability to reduce the inflammatory response in the disc by promoting the production of anti‐inflammatory factors. 54
Pang et al 55 reported that intradiscal injection of umbilical cord MSCs can improve the patient's pain symptoms and functional scores. Orozco et al's study 56 reported that 26 patients suffering from degenerative disc disease, as well as candidates for surgical treatment, were selected. After intradiscal injection of bone marrow‐derived MSCs, 40% of patients improved one modified Pfirrmann grade at 12 months, the VAS and ODI scores decreased significantly at 36 months, and only six patients eventually progressed to surgery. Moreover, its long‐term efficacy was reported in a study with a follow‐up period of up to 6 years. 57
Considering that MSCs have an immune privilege or immune evasion ability, and can inhibit the immune response in a manner not restricted by the human leukocyte antigen (HLA) system, the injection of allogeneic MSCs would not cause apparent immune rejection, and would be logistically more convenient than the autologous MSC. Noriega et al 58 showed in an RCT that intradiscal injection of allogeneic bone marrow MSCs can improve disc quality quantified by Pfirrmann grading, pain symptoms, and quality of life, without causing severe adverse events, and confirmed that allogeneic MSCs therapy was a valid alternative for the treatment of IDD. A recent systematic review including seven clinical studies identified that MSCs injection was a safe and feasible option for IDD in patients at the early‐degeneration stage, evidenced by an overall clinical and radiological improvement and very low complication rate during the follow‐up. 59 However, how to maintain the viability of MSCs, and how to promote their proliferation and differentiation under the conditions of low pH, low glucose, low oxygen, and high inflammatory conditions of the degenerating intervertebral disc (IVD), remains a challenge to be further resolved in future research. 60
6. PLATELET‐RICH PLASMA
PRP is an autologous blood concentrate acquired from centrifuged whole blood, which contains a natural concentration of growth factors and cytokines, including vascular endothelial growth factor, epidermal growth factor, insulin‐like growth factor, transforming growth factor β‐1, and platelet‐derived growth factor. 61 Recently, increasing studies have demonstrated the repair and regenerative ability of PRP in many damaged or degenerated tissues, including tendons, ligaments, and cartilage, because of its potential for promoting cell proliferation, differentiation, migration, and synthesis of ECM proteins and collagen. 62 , 63 , 64 , 65 Moreover, PRP has exhibited an anti‐inflammatory effect by preventing the activation of inflammatory mediators and inhibiting metalloproteinases, making it a potential strategy for the management of LBP. 66 , 67
In a prospective observational study by Levi et al, 68 22 patients with discogenic LBP underwent intradiscal injection of PRP. If the patient's VAS improved by at least 50% and ODI decreased by at least 30%, the treatment was considered successful. Finally, the treatment success rate was 14% at 1 month, 32% at 2 months, and 47% at 6 months follow‐up. The first double‐blind RCT using PRP for discogenic LBP was reported by Tuakli‐Wosornu et al. 69 Forty‐seven patients were treated by the intradiscal injection of PRP (treatment group) or contrast agent (control group). At the 8‐week follow‐up, there were statistically significant improvements in the treatment group with numeric rating scale (NRS) best pain, functional rating index (FRI), and patient satisfaction (North American Spine Society Outcome Questionnaire) compared to the control group. No adverse events were reported, such as disc infection, neurologic injury, or progressive herniation. Furthermore, the long‐term efficacy of intradiscal PRP injection for symptomatic degenerative intervertebral discs was confirmed in an RCT with 5 to 9 years follow‐up, 70 and a positive correlation between platelet concentration of PRP and clinical outcomes was identified in a recent prospective clinical trial. 71
Recently, several meta‐analyses demonstrated that intradiscal PRP injection had shown beneficial effects in controlling pain and improving disabilities in patients with LBP, but there was a paucity of high‐quality studies to give conclusive evidence. 72 , 73 Therefore, more clinical studies, especially RCTs with multiple outcome parameters, are necessary for evaluating the true efficacy of this treatment. 74 Moreover, according to the American Society of Interventional Pain Physicians guidelines, the qualitative evidence for intradiscal PRP injection in the treatment of LBP has been assessed as level III, based on the available evidence including RCT, observational studies, meta‐analysis, and systematic review. 75
7. CONDOLIASE
During disc degeneration, lumbosacral nerve compression induced by herniated NP tissues is an important factor causing LBP and radicular pain. Chemonucleolysis was first described by Smith 76 in 1964, to dissolve the herniated NP by injection of proteolytic enzymes. Chymopapain, a nonspecific proteoglycanase derived from the papaya plant, was the main enzyme used for this procedure. Subsequently, chemonucleolysis was approved by the US Food and Drug Administration in 1982 and is widely used for the treatment of LDH in the United States and Europe, although accompanied by considerable controversy and vocal opposition. However, since the early 2000s, chymopapain was gradually withdrawn from the market and chemonucleolysis has not been available for LDH, due to safety concerns and other factors.
Chondroitin sulfate ABC endolyase (condoliase), derived from the gram‐negative rod, Proteus vulgaris, is a pure mucopolysaccharidase with high substrate specificity for hyaluronic acid and chosulphaten sulfate, which are the main proteoglycans of NP tissues. Unlike chymopapain, the target of condoliase is chondroitin sulfate, which is distributed in the NP tissues but not in the nerves and vascular tissues. Therefore, condoliase can be safely and specifically used for the treatment of LDH. 77
In an RCT conducted by Matsuyama et al, 78 192 patients with LDH were included. After the intradiscal injection of different doses of condoliase, the clinical symptoms were significantly improved without causing severe adverse drug reactions. Moreover, three doses had similar efficacy, but the incidence of adverse events and decrease in disc height was dose‐dependent. Therefore, a small dose of condoliase (1.25 U) was recommended for intradiscal injection. Okada et al 79 reported that, after the intradiscal injection of condoliase, 85.4% of patients reported an improvement in pain symptoms, and no severe adverse event was observed. Furthermore, injecting condoliase into the center of the NP is recommended for obtaining better clinical effectiveness. Intradiscal injection of condoliase for the treatment of LDH has been approved by the drug regulatory authority in Japan, 80 after its efficacy and safety were confirmed in clinical phases II/III and III studies. 78 , 81 Although current studies suggest that the intradiscal injection of condoliase is a potential new, effective, and minimally invasive therapeutic strategy for patients with LDH, 82 its long‐term efficacy and side effects remain unclear, and the best clinical indications need to be further identified.
8. CYTOKINE ANTAGONIST
Cytokines, as regulatory proteins and proinflammatory biomarkers, play an important role in the occurrence and development of disc degeneration, by amplification of inflammatory response and promoting ECM degradation. 83 Especially, tumor necrosis factor‐α (TNF‐α) and interleukin‐1 (IL‐1) are presumed to be critical drivers of IDD. 84 Advances in experimental and clinical research have identified that specific cytokine antagonists may be a novel treatment strategy for LBP or early‐stage IDD, 85 , 86 , 87 including anti‐TNF: infliximab, etanercept, and adalimumab; anti‐IL‐6: tocilizumab; antinerve growth factor: tanezumab, fulranumab; anti‐Janus kinase: tofacitinib.
A previous double‐blind, placebo‐controlled pilot study reported that a single low dose (0.1‐1.5 mg) of intradiscal etanercept injection imparted no significant improvement in pain scores and function, although no severe side effects were observed. 88 However, in an RCT conducted by Sainoh et al, 89 the intradiscal injection dose of etanercept was increased to 10 mg. After 8 weeks of follow‐up, the patient's pain symptoms and function scores improved without adverse reactions such as infection and nerve damage. Additionally, the intradiscal injection of tocilizumab, a humanized anti‐IL‐6 receptor monoclonal antibody, was shown to exert a short‐term analgesic effect in patients with discogenic LBP. 90 However, there is limited evidence on cytokine antagonists for the treatment of LBP, and it cannot be currently recommended for clinical practice. Therefore, larger sample sizes and better‐designed studies are required to determine the safety and efficacy, especially, the long‐term effects.
9. HYDROGEL‐BASED BIOMATERIALS
During degeneration, the synthesis of ECM components such as type II collagen and proteoglycan decreases, which impairs the NP mechanical function, and decreases the swelling capacity and pressurization potential. Therefore, how to restore and maintaining the native disc structure and mechanical function is the key to biological‐based therapies. With the development of tissue engineering technology, various hydrogels have been made using natural or synthetic materials. 91 Natural hydrogels include hyaluronic acid, chitosan, alginate, and fibrin. 92 Generally, these materials are economical and exhibit low levels of cytotoxicity, bioactivity, and bioactive degradation.
Especially, hyaluronic acid‐based hydrogels have been drawing increasing attention as the ideal candidates for IDD, 92 because of their potential to repair and regenerate the NP through providing a three‐dimensional microenvironment for the implanted cells, 93 restoring biomechanical properties, 94 reducing inflammation response, and nociceptive behavior, 95 promoting ECM synthesis, 96 and promoting stem cell differentiation to NP‐like cells. 97
Animal experiments revealed that the injection of hyaluronic acid hydrogel not only restored the height of the intervertebral disc but also reduced inflammation and promoted the synthesis of ECM. 98 , 99 Priyadarshani et al 100 showed that crosslinking of type II collagen‐hyaluronic acid hydrogel can provide a growth‐permissive environment for NPCs and be transplanted into the disc as cell carriers. Moreover, the combined injection of hydrogel and stem cells provides a carrier for stem cells and also reduces the risk of adverse reactions such as ligament and bone hyperplasia caused by stem cell leakage. In a phase I clinical trial conducted by Kumar et al, 101 intradiscal injections of hyaluronic acid combined with autologous adipose‐derived MSCs relieved the pain symptoms of patients with LBP and also improve the functional score and quality of life, and no adverse event was observed during the 1‐year follow‐up period. In a prospective randomized, placebo‐controlled 36‐month study, 102 compared with the control groups, intradiscal injection of allogeneic mesenchymal precursor cells (MPCs) with hyaluronic acid demonstrated significant improvements in pain and function at various time points from baseline to 36 months, and there were no clinical symptoms of immune reactions to allogeneic MPCs, or other severe adverse events. However, the current research on the treatment of IDD with hydrogel‐based biomaterials is mainly conducted in vitro and animal experiments; the safety and feasibility of this option still need to be confirmed by more clinical studies.
Synthetic hydrogels are made from synthetic polymers such as polyamides and polyethylene glycol. Compared to natural hydrogels, synthetic hydrogels have better reproducibility, controllability, and customizable properties. However, potential biocompatibility and cytotoxicity are big concerns of synthetic hydrogels. While these materials displayed promising clinical application potential in cell tests and animal experiments, only a few synthetic hydrogel devices have been studied in clinical research. 92 For example, GelStix Nucleus Augmentation Device is a modified polyacrylonitrile hydrogel that reconstructs the disc function by increasing hydration and IVD height. Ceylan et al 103 implanted GelStix in 29 patients with IDD and found that the VAS scores were decreased from 7.14 to 2.48 and the ODI scores were decreased from 28.14 to 17.35 after 12 months following treatment. Moreover, another randomized, double‐blind, placebo‐controlled, multicentre study has been conducted to evaluate the efficacy of treatment with the GelStix device in patients with chronic discogenic LBP, which is expected to conclude in August 2021. 104
10. SIDE EFFECTS
Generally, minimal intradiscal injection may offer good results with patient compliance and low cost, but some side effects have been reported, including discitis, 105 disc collapse, 106 and impairment of sensitivity in the lower ipsilateral limb. 107 With the application of imaging guidance, this procedure became more feasible and safer, with an overall complication rate of ~0.47%. 32 Meanwhile, IDD is considered a major concern in intradiscal injection. Theoretically, a needle puncture injury can cause increased cell death, metabolic dysfunction, annulus fibrosus integrity impairment, and NP depressurisation. 108 , 109 Therefore, some authors have highlighted small‐diameter puncture needle and minimum dose of agent to avoid disc degeneration. 110 , 111 Moreover, disc degeneration caused by intradiscal injection was rarely reported. A recent narrative review on the techniques of intradiscal injection including 6843 patients reported that only two discs showed a collapse after injection of corticosteroid. 32
11. CONCLUSION AND FUTURE PERSPECTIVES
Intradiscal injection is a minimally invasive technique widely used in the management of patients with LBP, and its safety, efficacy, and feasibility are identified by growing clinical trials. Although there are various medications or biological agents used for intradiscal injection to treat LBP, their efficacy and safety are not easily comparable because of differences in the study designs and their limited number of cases, and there is insufficient evidence to support strong recommendations for their clinical application.
Increasing reports are revealing the benefits of these medications or biological agents in relieving the clinical symptoms and their potential for disc regeneration (Table 1). However, they all have some limitations. For example, the injection of glucocorticoids, methylene blue, and cytokine inhibitors can reduce inflammation and relieve symptoms in the short‐term, but their efficacy is difficult to maintain for a long period due to their half‐lives. Although previous studies suggested that the intradiscal injection of PRP and condoliase can alleviate pain symptoms, larger‐sample and high‐quality clinical trials are still needed to confirm their efficacy. MSCs have the potential to differentiate into NPCs and promote the synthesis of the ECM; however, it is difficult for the implanted MSCs to maintain their viability in the hypoxic and acidic environment of the degenerated disc.
TABLE 1.
Medications/biological agents | Mechanism and effects | Clinical outcomes | Study type |
---|---|---|---|
Glucocorticoids | Anti‐inflammation effect 23 , 24 , 25 , 26 , 27 |
Improving VAS, NRS, ODI scores, and LBP‐related limitations in activities (Quebec Back Pain Disability Scale) in the short term Reducing the HADS depression scores in the long term |
Prospective trial, prospective randomized controlled trial 28 , 29 , 30 |
O2‐O3 | (1) Stimulating the activity of fibroblasts to repair the damaged disc by deposition of collagen; (2) Increasing the concentration of oxygen in tissues; (3) Interrupting the inflammatory cascade of the arachidonic acid; (4) Reducing the disc volume by breaking the glycosaminoglycan chains. 31 , 32 , 33 , 34 , 35 |
Improving VAS, ODI scores in the short and long term Reducing the size of the disc herniation in the long term. |
Prospective trial, prospective randomized controlled trial, systematic review, meta‐analysis. 36 , 37 , 38 , 39 , 40 , 41 |
Methylene blue |
(1) Blocking nerve conduction or damage to nerve endings 45 ; (2) Anti‐inflammation effect. 46 |
Reducing the NRS, ODI scores and improving patient satisfaction rates in the short term Improving disc degeneration condition assessed by apparent diffusion coefficient and T2 values on MRI in the long term Decreasing the usage of NSAIDs or opioid medications in the long term. |
Prospective trial, prospective randomized controlled trial, meta‐analysis. 47 , 48 , 49 , 50 |
MSCs |
(1) Differentiating into NP cells; (2) Promoting the synthesis of ECM; |
Improving VAS, ODI, FRI, SF‐36 scores in the short and long term Improving disc quality quantified by Pfirrmann grading in the long term. |
Prospective trial, prospective randomized controlled trial, systematic review. 55 , 56 , 57 , 58 , 59 |
PRP |
(1) Promoting cell proliferation, differentiation, migration; (2) Promoting the synthesis of ECM; (3) Preventing the activation of inflammatory mediators and inhibiting metalloproteinases. 62 , 63 , 64 , 65 , 66 , 67 |
Improving VAS, ODI, NRS best pain, FRI scores, and patient satisfaction (North American Spine Society Outcome Questionnaire) in the short and long term. | Prospective trial, prospective randomized controlled trial, systematic review; meta‐analysis. 68 , 69 , 70 , 71 , 72 , 73 , 74 |
Condoliase | Specifically dissolve the chondroitin sulfate in NP tissue and relieve the compression on nerve roots.77 | Improving VAS (worst leg pain), ODI, and SF‐36 scores in the short term. | Prospective trial, prospective randomized controlled trial. 78 , 79 , 80 , 81 , 82 |
Cytokine inhibitor | Anti‐inflammation effect. 85 , 86 , 87 | Improving NRS and ODI scores in the short term. | Prospective randomized controlled trial. 89 , 90 |
Hydrogel‐based biomaterials combined with stem cells |
(1) Providing a three‐dimensional microenvironment for the implanted cells (2) Restoring biomechanical properties; (3) Reducing inflammation response and nociceptive behavior; (4) Promoting the synthesis of ECM; (5) Promoting stem cell differentiation to NP‐like cells. 92 , 93 , 94 , 95 , 96 , 97 |
Improving VAS, ODI, SF‐36 scores, and disc quality quantified by Pfirrmann grading and apparent diffusion coefficient on diffusion MRI in the short and long term. | Prospective trial, prospective randomized controlled trial. 101 , 102 , 103 |
Abbreviations: ECM, extracellular matrix; FRI, functional rating index; HADS, Hospital Anxiety and Depression Scale; LBP, low back pain; MRI, Magnetic Resonance Imaging; MSC, mesenchymal stem cell; NP, nucleus pulposus; NRS, numerical rating scale; NSAID, nonsteroidal anti‐inflammatory drugs; O2‐O3, oxygen‐ozone; ODI, Oswestry Disability Index; PRP, platelet‐rich plasma; SF‐36, 36‐Item Short Form Survey; VAS, visual analog scale.
Therefore, combined injections are expected to compensate for their individual components' limitations. Especially, the combination of cell therapy and tissue engineering technology may be the ideal medications for IDD, because of their potential for restoring the native function of IVD and disc regeneration. For example, stem cell‐embedded hydrogel injection maintains the mechanical properties of IVD and provides a carrier for stem cells, which is beneficial for their survival, proliferation, and differentiation. Although the current evidence is mainly derived from animal experiments, preclinical experiments, and limited clinical trials, there are certain challenges for its clinical application. With the growing clinical data and future systematic reviews, it is not unrealistic to hypothesize that this option can provide longer relief and greater clinical benefits to this patient population, and play an important role in the management of LBP.
CONFLICT OF INTEREST
The authors declare no potential conflict of interest.
AUTHOR CONTRIBUTIONS
Fu Zhang and Songjuan Wang contributed equally to the concept of the paper and the writing of the article. Baoliang Li contributed to the article revision. Zhiyu Zhou and Shaoyu Liu provided substantial contributions to reviewing the format, revising the article critically, and its final approval. All authors have read and approved the final submitted article.
Zhang, F. , Wang, S. , Li, B. , Tian, W. , Zhou, Z. , & Liu, S. (2022). Intradiscal injection for the management of low back pain. JOR Spine, 5(1), e1186. 10.1002/jsp2.1186
Fu Zhang and Songjuan Wang (shared first authorship) and Zhiyu Zhou and Shaoyu Liu (shared last authorship) contributed equally to this study.
Funding information foundation of Shenzhen Committee for Science and Technology Innovation, Grant/Award Numbers: JCYJ20190809142211354, GJHZ20180929160004704; Fundamental Research Funds for the Central Universities, Grant/Award Number: 19ykzd05; Sanming Project of Medicine in Shenzhen, Grant/Award Number: SZSM201911002; the Beijing Municipal Health Commission, Grant/Award Number: BMHC2021‐6;BMHC‐2019‐9;BMHC‐2018‐4;PXM2020; the National Natural Science Foundation of China, Grant/Award Numbers: 81772400, 31900583, 32071351; the Natural Science Foundation of Guangzhou City, Grant/Award Numbers: 201704030082, 201807010031, 201604046028
Contributor Information
Zhiyu Zhou, Email: zhouzhy23@mail.sysu.edu.cn.
Shaoyu Liu, Email: liushaoy@mail.sysu.edu.cn.
REFERENCES
- 1. Chang J, Wang Y, Shao L, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016;22(1):78‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Fatoye F, Gebrye T, Odeyemi I. Real‐world incidence and prevalence of low back pain using routinely collected data. Rheumatol Int. 2019;39(4):619‐626. [DOI] [PubMed] [Google Scholar]
- 3. Morris LD, Daniels KJ, Ganguli B, Louw QA. An update on the prevalence of low back pain in Africa: a systematic review and meta‐analyses. BMC Musculoskelet Disord. 2018;19(1):196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Knezevic NN, Candido KD, Vlaeyen J, Van Zundert J, Cohen SP. Low back pain. Lancet. 2021;398(10294):78‐92. [DOI] [PubMed] [Google Scholar]
- 5. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990‐2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2018;392(10159):1789‐1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Hong J, Reed C, Novick D, Happich M. Costs associated with treatment of chronic low back pain: an analysis of the UK General Practice Research Database. Spine. 2013;38(1):75‐82. [DOI] [PubMed] [Google Scholar]
- 7. Katz JN. Lumbar disc disorders and low‐back pain: socioeconomic factors and consequences. J Bone Joint Surg Am. 2006;88(Suppl 2):21‐24. [DOI] [PubMed] [Google Scholar]
- 8. Kadow T, Sowa G, Vo N, Kang JD. Molecular basis of intervertebral disc degeneration and herniations: what are the important translational questions? Clin Orthop Relat Res. 2015;473(6):1903‐1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Oichi T, Taniguchi Y, Oshima Y, Tanaka S, Saito T. Pathomechanism of intervertebral disc degeneration. JOR Spine. 2020;3(1):e1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Gianola S, Bargeri S, Del Castillo G, et al. Effectiveness of treatments for acute and subacute mechanical non‐specific low back pain: a systematic review with network meta‐analysis. Br J Sport Med. 2021;56(1):41‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Moriguchi Y, Alimi M, Khair T, et al. Biological treatment approaches for degenerative disk disease: a literature review of in vivo animal and clinical data. Global Spine J. 2016;6(5):497‐518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ju DG, Kanim LE, Bae HW. Intervertebral disc repair: current concepts. Global Spine J. 2020;10(suppl 2):130S‐136S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Roh EJ, Darai A, Kyung JW, et al. Genetic therapy for intervertebral disc degeneration. Int J Mol Sci. 2021;22(4):1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Choi Y, Park MH, Lee K. Tissue engineering strategies for intervertebral disc treatment using functional polymers. Polymers (Basel). 2019;11(5):872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wu TJ, Hung CY, Lee CW, Lam S, Clark TB, Chang KV. Ultrasound‐guided lumbar Intradiscal injection for discogenic pain: technical innovation and presentation of two cases. J Pain Res. 2020;13:1103‐1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lam K, Hung CY, Wu TJ. Ultrasound‐guided L5/S1 intradiscal needle placement using biplanar approach with the patient in the lateral decubitus position—a report of three cases. Pain Pract. 2021:1‐6. [DOI] [PubMed] [Google Scholar]
- 17. Knezevic MPNN, Mandalia DS, Raasch MJ, Knezevic MI, Candido KD. Treatment of chronic low back pain‐new approaches on the horizon. J Pain Res. 2017;10:1111‐1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Motejunas MW, Bonneval L, Carter C, Reed D, Ehrhardt K. Biologic therapy in chronic pain management: a review of the clinical data and future investigations. Curr Pain Headache Rep. 2021;25(5):30. [DOI] [PubMed] [Google Scholar]
- 19. Patil P, Dong Q, Wang D, et al. Systemic clearance of p16INK4a‐positive senescent cells mitigates age‐associated intervertebral disc degeneration. Aging Cell. 2019;18(3):e12927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kang L, Liu S, Li J, Tian Y, Xue Y, Liu X. The mitochondria‐targeted anti‐oxidant MitoQ protects against intervertebral disc degeneration by ameliorating mitochondrial dysfunction and redox imbalance. Cell Prolif. 2020;53(3):e12779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wang J, Nisar M, Huang C, et al. Small molecule natural compound agonist of SIRT3 as a therapeutic target for the treatment of intervertebral disc degeneration. Exp Mol Med. 2018;50(11):1‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Wang Y, Zuo R, Wang Z, et al. Kinsenoside ameliorates intervertebral disc degeneration through the activation of AKT‐ERK1/2‐Nrf2 signaling pathway. Aging (Albany, NY). 2019;11(18):7961‐7977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Manchikanti L, Pampati V, Hirsch JA. Retrospective cohort study of usage patterns of epidural injections for spinal pain in the US fee‐for‐service Medicare population from 2000 to 2014. BMJ Open. 2016;6(12):e13042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Helm IS, Harmon PC, Noe C, et al. Transforaminal epidural steroid injections: a systematic review and meta‐analysis of efficacy and safety. Pain Physician. 2021;24(S1):S209‐S232. [PubMed] [Google Scholar]
- 25. Krishnan R, Kurup V, Vadivelu N, Dai F, Zhou B, Rajput K. Does choice of steroid matter for treatment of chronic low back pain with sacroiliac joint injections: a retrospective study. Curr Pain Headache Rep. 2021;25(5):34. [DOI] [PubMed] [Google Scholar]
- 26. Dhakal GR, Hamal PK, Dhungana S, Kawaguchi Y. Clinical efficacy of selective nerve root block in lumbar radiculopathy due to disc prolapse. J Nepal Health Res Counc. 2019;17(2):242‐246. [DOI] [PubMed] [Google Scholar]
- 27. Budisulistyo T, Atmaja F. Percutaneous discectomy followed by CESI might improve neurological disorder of drop foot patients due to chronic LDH. Brain Sci. 2020;10(8):539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cao P, Jiang L, Zhuang C, et al. Intradiscal injection therapy for degenerative chronic discogenic low back pain with end plate Modic changes. Spine J. 2011;11(2):100‐106. [DOI] [PubMed] [Google Scholar]
- 29. Nguyen C, Boutron I, Baron G, et al. Intradiscal glucocorticoid injection for patients with chronic low back pain associated with active discopathy: a randomized trial. Ann Intern Med. 2017;166(8):547‐556. [DOI] [PubMed] [Google Scholar]
- 30. Tavares I, Thomas E, Cyteval C, et al. Intradiscal glucocorticoids injection in chronic low back pain with active discopathy: a randomized controlled study. Ann Phys Rehabil Med. 2020;64(2):101396. [DOI] [PubMed] [Google Scholar]
- 31. de Sire A, Agostini F, Lippi L, et al. Oxygen‐ozone therapy in the rehabilitation field: state of the art on mechanisms of action, safety and effectiveness in patients with musculoskeletal disorders. Biomolecules. 2021;11(3):356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Migliore A, Sorbino A, Bacciu S, et al. The technique of intradiscal injection: a narrative review. Ther Clin Risk Manag. 2020;16:953‐968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Magalhaes FN, Dotta L, Sasse A, Teixera MJ, Fonoff ET. Ozone therapy as a treatment for low back pain secondary to herniated disc: a systematic review and meta‐analysis of randomized controlled trials. Pain Physician. 2012;15(2):E115‐E129. [PubMed] [Google Scholar]
- 34. Murphy K, Elias G, Steppan J, et al. Percutaneous treatment of herniated lumbar discs with ozone: investigation of the mechanisms of action. J Vasc Interv Radiol. 2016;27(8):1242‐1250. [DOI] [PubMed] [Google Scholar]
- 35. Muto M, Giurazza F, Silva RP, Guarnieri G. Rational approach, technique and selection criteria treating lumbar disk herniations by oxygen‐ozone therapy. Interv Neuroradiol. 2016;22(6):736‐740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Steppan J, Meaders T, Muto M, Murphy KJ. A metaanalysis of the effectiveness and safety of ozone treatments for herniated lumbar discs. J Vasc Interv Radiol. 2010;21(4):534‐548. [DOI] [PubMed] [Google Scholar]
- 37. Migliorini F, Maffulli N, Eschweiler J, Bestch M, Tingart M, Baroncini A. Ozone injection therapy for intervertebral disc herniation. Br Med Bull. 2020;136(1):88‐106. [DOI] [PubMed] [Google Scholar]
- 38. Elawamy A, Kamel EZ, Hassanien M, Wahba OM, Amin SE. Implication of two different doses of intradiscal ozone‐oxygen injection upon the pain alleviation in patients with low back pain: a randomized, single‐blind study. Pain Physician. 2018;21(1):E25‐E31. [PubMed] [Google Scholar]
- 39. Buric J, Rigobello L, Hooper D. Five and ten year follow‐up on intradiscal ozone injection for disc herniation. Int J Spine Surg. 2014;8:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Rahimzadeh P, Imani F, Ghahremani M, Faiz S. Comparison of percutaneous intradiscal ozone injection with laser disc decompression in discogenic low back pain. J Pain Res. 2018;11:1405‐1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Andrade RR, Oliveira‐Neto OB, Barbosa LT, Santos IO, Sousa‐Rodrigues CF, Barbosa FT. Effectiveness of ozone therapy compared to other therapies for low back pain: a systematic review with meta‐analysis of randomized clinical trials. Rev Bras Anestesiol. 2019;69(5):493‐501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Ercalik T, Kilic M. Efficacy of intradiscal ozone therapy with or without periforaminal steroid injection on lumbar disc herniation: a double‐blinded controlled study. Pain Physician. 2020;23(5):477‐484. [PubMed] [Google Scholar]
- 43. Ni S, Ling Z, Wang X, et al. Sensory innervation in porous endplates by Netrin‐1 from osteoclasts mediates PGE2‐induced spinal hypersensitivity in mice. Nat Commun. 2019;10(1):5643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Freemont AJ, Peacock TE, Goupille P, Hoyland JA, O'Brien J, Jayson MI. Nerve ingrowth into diseased intervertebral disc in chronic back pain. Lancet. 1997;350(9072):178‐181. [DOI] [PubMed] [Google Scholar]
- 45. Alda M. Methylene blue in the treatment of neuropsychiatric disorders. CNS Drugs. 2019;33(8):719‐725. [DOI] [PubMed] [Google Scholar]
- 46. Li JW, Wang RL, Xu J, et al. Methylene blue prevents osteoarthritis progression and relieves pain in rats via upregulation of Nrf2/PRDX1. Acta Pharmacol Sin. 2021:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Peng B, Pang X, Wu Y, Zhao C, Song X. A randomized placebo‐controlled trial of intradiscal methylene blue injection for the treatment of chronic discogenic low back pain. Pain. 2010;149(1):124‐129. [DOI] [PubMed] [Google Scholar]
- 48. Kim SH, Ahn SH, Cho YW, Lee DG. Effect of intradiscal methylene blue injection for the chronic discogenic low back pain: one year prospective follow‐up study. Ann Rehabil Med. 2012;36(5):657‐664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Zhang X, Hao J, Hu Z, Yang H. Clinical evaluation and magnetic resonance imaging assessment of intradiscal methylene blue injection for the treatment of discogenic low back pain. Pain Physician. 2016;19(8):E1189‐E1195. [PubMed] [Google Scholar]
- 50. Guo X, Ding W, Liu L, Yang S. Intradiscal methylene blue injection for discogenic low back pain: a meta‐analysis. Pain Pract. 2019;19(1):118‐129. [DOI] [PubMed] [Google Scholar]
- 51. Kallewaard JW, Wintraecken VM, Geurts JW, et al. A multicenter randomized controlled trial on the efficacy of intradiscal methylene blue injection for chronic discogenic low back pain: the IMBI study. Pain. 2019;160(4):945‐953. [DOI] [PubMed] [Google Scholar]
- 52. Zhang L, Liu Y, Huang Z, et al. Toxicity effects of methylene blue on rat intervertebral disc annulus fibrosus cells. Pain Physician. 2019;22(2):155‐164. [PubMed] [Google Scholar]
- 53. Croft AS, Illien‐Junger S, Grad S, Guerrero J, Wangler S, Gantenbein B. The application of mesenchymal stromal cells and their homing capabilities to regenerate the intervertebral disc. Int J Mol Sci. 2021;22(7):3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Vadalà G, Ambrosio L, Russo F, Papalia R, Denaro V. Interaction between mesenchymal stem cells and intervertebral disc microenvironment: from cell therapy to tissue engineering. Stem Cells Int. 2019;2019:2376172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Pang X, Yang H, Peng B. Human umbilical cord mesenchymal stem cell transplantation for the treatment of chronic discogenic low back pain. Pain Physician. 2014;17(4):E525‐E530. [PubMed] [Google Scholar]
- 56. Pettine KA, Suzuki RK, Sand TT, Murphy MB. Autologous bone marrow concentrate intradiscal injection for the treatment of degenerative disc disease with three‐year follow‐up. Int Orthop. 2017;41(10):2097‐2103. [DOI] [PubMed] [Google Scholar]
- 57. Centeno C, Markle J, Dodson E, et al. 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(1):197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Noriega DC, Ardura F, Hernandez‐Ramajo R, et al. Intervertebral disc repair by allogeneic mesenchymal bone marrow cells: a randomized controlled trial. Transplantation. 2017;101(8):1945‐1951. [DOI] [PubMed] [Google Scholar]
- 59. Migliorini F, Rath B, Tingart M, Baroncini A, Quack V, Eschweiler J. Autogenic mesenchymal stem cells for intervertebral disc regeneration. Int Orthop. 2019;43(4):1027‐1036. [DOI] [PubMed] [Google Scholar]
- 60. Vadala G, Ambrosio L, Russo F, Papalia R, Denaro V. Stem cells and intervertebral disc regeneration overview‐what they can and can't do. Int J Spine Surg. 2021;15(s1):40‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Alves R, Grimalt R. A review of platelet‐rich plasma: history, biology, mechanism of action, and classification. Skin Appendage Disord. 2018;4(1):18‐24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Schneider A, Burr R, Garbis N, Salazar D. Platelet‐rich plasma and the shoulder: clinical indications and outcomes. Curr Rev Musculoskelet Med. 2018;11(4):593‐597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Dai WL, Zhou AG, Zhang H, Zhang J. Efficacy of platelet‐rich plasma in the treatment of knee osteoarthritis: a meta‐analysis of randomized controlled trials. Art Ther. 2017;33(3):659‐670. [DOI] [PubMed] [Google Scholar]
- 64. Wong CC, Ou KL, Lin YH, et al. Platelet‐rich fibrin facilitates one‐stage cartilage repair by promoting chondrocytes viability, migration, and matrix synthesis. Int J Mol Sci. 2020;21(2):577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Cao Y, Zhu X, Zhou R, He Y, Wu Z, Chen Y. A narrative review of the research progress and clinical application of platelet‐rich plasma. Ann Palliat Med. 2021;10(4):4823‐4829. [DOI] [PubMed] [Google Scholar]
- 66. Akeda K, Yamada J, Linn ET, Sudo A, Masuda K. Platelet‐rich plasma in the management of chronic low back pain: a critical review. J Pain Res. 2019;12:753‐767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Urits I, Viswanath O, Galasso AC, et al. Platelet‐rich plasma for the treatment of low back pain: a comprehensive review. Curr Pain Headache Rep. 2019;23(7):52. [DOI] [PubMed] [Google Scholar]
- 68. Levi D, Horn S, Tyszko S, Levin J, Hecht‐Leavitt C, Walko E. Intradiscal platelet‐rich plasma injection for chronic discogenic low back pain: preliminary results from a prospective trial. Pain Med. 2016;17(6):1010‐1022. [DOI] [PubMed] [Google Scholar]
- 69. Tuakli‐Wosornu YA, Terry A, Boachie‐Adjei K, et al. Lumbar intradiskal platelet‐rich plasma (PRP) injections: a prospective, double‐blind, randomized controlled study. PM R. 2016;8(1):1‐10. [DOI] [PubMed] [Google Scholar]
- 70. Cheng J, Santiago KA, Nguyen JT, Solomon JL, Lutz GE. Treatment of symptomatic degenerative intervertebral discs with autologous platelet‐rich plasma: follow‐up at 5‐9 years. Regen Med. 2019;14(9):831‐840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Jain D, Goyal T, Verma N, Paswan AK, Dubey RK. Intradiscal platelet‐rich plasma injection for discogenic low back pain and correlation with platelet concentration: a prospective clinical trial. Pain Med. 2020;21(11):2719‐2725. [DOI] [PubMed] [Google Scholar]
- 72. Hirase T, Jack IR, Sochacki KR, Harris JD, Weiner BK. Systemic review: is an intradiscal injection of platelet‐rich plasma for lumbar disc degeneration effective? Cureus. 2020;12(6):e8831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Chang MC, Park D. The effect of intradiscal platelet‐rich plasma injection for management of discogenic lower back pain: a meta‐analysis. J Pain Res. 2021;14:505‐512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Muthu S, Jeyaraman M, Chellamuthu G, Jeyaraman N, Jain R, Khanna M. Does the intradiscal injection of platelet rich plasma have any beneficial role in the management of lumbar disc disease? Global Spine J. 2021:1‐12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Navani A, Manchikanti L, Albers SL, et al. Responsible, safe, and effective use of biologics in the management of low back pain: American Society of Interventional Pain Physicians (ASIPP) guidelines. Pain Physician. 2019;22(1S):S1‐S74. [PubMed] [Google Scholar]
- 76. Smith L. Enzyme dissolution of the nucleus pulposus in humans. JAMA. 1964;187:137‐140. [DOI] [PubMed] [Google Scholar]
- 77. Ishibashi K, Fujita M, Takano Y, Iwai H, Inanami H, Koga H. Chemonucleolysis with chondroitin sulfate ABC endolyase for treating lumbar disc herniation: exploration of prognostic factors for good or poor clinical outcomes. Medicina (Kaunas). 2020;56(11);627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Matsuyama Y, Chiba K, Iwata H, Seo T, Toyama Y. A multicenter, randomized, double‐blind, dose‐finding study of condoliase in patients with lumbar disc herniation. J Neurosurg Spine. 2018;28(5):499‐511. [DOI] [PubMed] [Google Scholar]
- 79. Okada E, Suzuki S, Nori S, et al. The effectiveness of chemonucleolysis with condoliase for treatment of painful lumbar disc herniation. J Orthop Sci. 2020;26:548‐554. [DOI] [PubMed] [Google Scholar]
- 80. Matsuyama Y, Chiba K. Condoliase for treatment of lumbar disc herniation. Drugs Today (Barc). 2019;55(1):17‐23. [DOI] [PubMed] [Google Scholar]
- 81. Chiba K, Matsuyama Y, Seo T, Toyama Y. Condoliase for the treatment of lumbar disc herniation: a randomized controlled trial. Spine. 2018;43(15):E869‐E876. [DOI] [PubMed] [Google Scholar]
- 82. Banno T, Hasegawa T, Yamato Y, et al. Clinical outcome of condoliase injection treatment for lumbar disc herniation: indications for condoliase therapy. J Orthop Sci. 2020;26:79‐85. [DOI] [PubMed] [Google Scholar]
- 83. van den Berg R, Jongbloed EM, de Schepper E, Bierma‐Zeinstra S, Koes BW, Luijsterburg P. The association between pro‐inflammatory biomarkers and nonspecific low back pain: a systematic review. Spine J. 2018;18(11):2140‐2151. [DOI] [PubMed] [Google Scholar]
- 84. Wang Y, Che M, Xin J, Zheng Z, Li J, Zhang S. The role of IL‐1beta and TNF‐alpha in intervertebral disc degeneration. Biomed Pharmacother. 2020;131:110660. [DOI] [PubMed] [Google Scholar]
- 85. Dimitroulas T, Lambe T, Raphael JH, Kitas GD, Duarte RV. Biologic drugs as analgesics for the management of low back pain and sciatica. Pain Med. 2019;20(9):1678‐1686. [DOI] [PubMed] [Google Scholar]
- 86. Li Z, Gehlen Y, Heizmann F, et al. Preclinical ex‐vivo testing of anti‐inflammatory drugs in a bovine intervertebral degenerative disc model. Front Bioeng Biotechnol. 2020;8:583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Pimentel DC, El AO, Benyamin RM, et al. Anti‐tumor necrosis factor antagonists in the treatment of low back pain and radiculopathy: a systematic review and meta‐analysis. Pain Physician. 2014;17(1):E27‐E44. [PubMed] [Google Scholar]
- 88. Cohen SP, Wenzell D, Hurley RW, et al. A double‐blind, placebo‐controlled, dose‐response pilot study evaluating intradiscal etanercept in patients with chronic discogenic low back pain or lumbosacral radiculopathy. Anesthesiology. 2007;107(1):99‐105. [DOI] [PubMed] [Google Scholar]
- 89. Sainoh T, Orita S, Miyagi M, et al. Single intradiscal administration of the tumor necrosis factor‐alpha inhibitor, etanercept, for patients with discogenic low back pain. Pain Med. 2016;17(1):40‐45. [DOI] [PubMed] [Google Scholar]
- 90. Sainoh T, Orita S, Miyagi M, et al. Single intradiscal injection of the interleukin‐6 receptor antibody tocilizumab provides short‐term relief of discogenic low back pain; prospective comparative cohort study. J Orthop Sci. 2016;21(1):2‐6. [DOI] [PubMed] [Google Scholar]
- 91. Galante R, Pinto T, Colaco R, Serro AP. Sterilization of hydrogels for biomedical applications: a review. J Biomed Mater Res B Appl Biomater. 2018;106(6):2472‐2492. [DOI] [PubMed] [Google Scholar]
- 92. Zheng K, Du D. Recent advances of hydrogel‐based biomaterials for intervertebral disc tissue treatment: a literature review. J Tissue Eng Regen Med. 2021;15(4):299‐321. [DOI] [PubMed] [Google Scholar]
- 93. Choi UY, Joshi HP, Payne S, et al. An injectable hyaluronan–methylcellulose (HAMC) hydrogel combined with Wharton's jelly‐derived mesenchymal stromal cells (WJ‐MSCs) promotes degenerative disc repair. Int J Mol Sci. 2020;21(19):7391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Zhou Z, Gao M, Wei F, et al. Shock absorbing function study on denucleated intervertebral disc with or without hydrogel injection through static and dynamic biomechanical tests in vitro. Biomed Res Int. 2014;2014:461724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Mohd II, Abbah SA, Kilcoyne M, et al. Implantation of hyaluronic acid hydrogel prevents the pain phenotype in a rat model of intervertebral disc injury. Sci Adv. 2018;4(4):q597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Zhang F, Liu X, Li B, et al. The effect of hyaluronic acid on nucleus pulposus extracellular matrix production through hypoxia‐inducible factor‐1alpha transcriptional activation of CD44 under hypoxia. Eur Cell Mater. 2021;41:142‐152. [DOI] [PubMed] [Google Scholar]
- 97. Chen P, Ning L, Qiu P, et al. Photo‐crosslinked gelatin‐hyaluronic acid methacrylate hydrogel‐committed nucleus pulposus‐like differentiation of adipose stromal cells for intervertebral disc repair. J Tissue Eng Regen Med. 2019;13(4):682‐693. [DOI] [PubMed] [Google Scholar]
- 98. Kazezian Z, Li Z, Alini M, Grad S, Pandit A. Injectable hyaluronic acid down‐regulates interferon signaling molecules, IGFBP3 and IFIT3 in the bovine intervertebral disc. Acta Biomater. 2017;52:118‐129. [DOI] [PubMed] [Google Scholar]
- 99. Kim DH, Martin JT, Elliott DM, Smith LJ, Mauck RL. Phenotypic stability, matrix elaboration and functional maturation of nucleus pulposus cells encapsulated in photocrosslinkable hyaluronic acid hydrogels. Acta Biomater. 2015;12:21‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Priyadarshani P, Li Y, Yang S, Yao L. Injectable hydrogel provides growth‐permissive environment for human nucleus pulposus cells. J Biomed Mater Res A. 2016;104(2):419‐426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Kumar H, Ha DH, Lee EJ, 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(1):262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Amirdelfan K, Bae H, McJunkin T, et al. Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: a prospective randomized, placebo‐controlled 36‐month study of safety and efficacy. Spine J. 2021;21(2):212‐230. [DOI] [PubMed] [Google Scholar]
- 103. Ceylan A, Asik I, Ozgencil GE, Erken B. Clinical results of intradiscal hydrogel administration (GelStix) in lumbar degenerative disc disease. Turk J Med Sci. 2019;49(6):1634‐1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. ClinicalTrials.gov . US National Library of Medicine. 2012. https://clinicaltrials.gov/ct2/show/NCT02763956. Accessed March 26, 2021.
- 105. Yin W, Pauza K, Olan WJ, Doerzbacher JF, Thorne KJ. 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(1):16‐31. [DOI] [PubMed] [Google Scholar]
- 106. Benyahya R, Lefevre‐Colau MM, Fayad F, et al. Intradiscal injection of acetate of prednisolone in severe low back pain: complications and patients' assessment of effectiveness. Ann Readapt Med Phys. 2004;47(9):621‐626. [DOI] [PubMed] [Google Scholar]
- 107. Lehnert T, Naguib NN, Wutzler S, et al. Analysis of disk volume before and after CT‐guided intradiscal and periganglionic ozone‐oxygen injection for the treatment of lumbar disk herniation. J Vasc Interv Radiol. 2012;23(11):1430‐1436. [DOI] [PubMed] [Google Scholar]
- 108. van Heeswijk VM, Thambyah A, Robertson PA, Broom ND. Does an annular puncture influence the herniation path?: an in vitro mechanical and structural investigation. Spine. 2018;43(7):467‐476. [DOI] [PubMed] [Google Scholar]
- 109. Vergari C, Mansfield JC, Chan D, Clarke A, Meakin JR, Winlove PC. The effects of needle damage on annulus fibrosus micromechanics. Acta Biomater. 2017;63:274‐282. [DOI] [PubMed] [Google Scholar]
- 110. Wang JY, Mansfield JC, Brasselet S, Vergari C, Meakin JR, Winlove CP. Micro‐mechanical damage of needle puncture on bovine annulus fibrosus fibrils studied using polarization‐resolved second harmonic generation(P‐SHG) microscopy. J Mech Behav Biomed Mater. 2021;118:104458. [DOI] [PubMed] [Google Scholar]
- 111. Ura K, Sudo H, Iwasaki K, Tsujimoto T, Ukeba D, Iwasaki N. Effects of intradiscal injection of local anesthetics on intervertebral disc degeneration in rabbit degenerated intervertebral disc. J Orthop Res. 2019;37(9):1963‐1971. [DOI] [PubMed] [Google Scholar]