Abstract
Cochlear implants are highly successful in restoring speech perception but variability in outcomes exists. Post-surgical fibrosis and neo-ossification are thought to play a significant role, being linked to increased impedance and loss of residual hearing and posing challenges for re-implantation. Hence, there is growing interest in pharmacological interventions to limit intracochlear fibrosis and neo-ossification. While current approaches focus on steroids, studies in other organs have identified many candidate drugs. However, selection is hindered by a limited understanding of the molecular and cellular mechanisms driving fibrosis after implantation. This review introduces potential drug candidates for cochlear implant-induced fibrosis, with many targeting core fibrotic pathways such as TGF-β/SMAD, PDGF, and Wnt/β-catenin or inhibiting pro-inflammatory signalling. By drawing on lessons from other tissues, this review identifies mechanisms and therapeutic approaches adaptable to the cochlea. Understanding fibrosis across organs will guide strategies to prevent or reverse cochlear fibrosis. Their translation requires careful evaluation of local delivery, minimal ototoxicity, and effects on the electrode–tissue interface.
1. Introduction
Hearing restoration surgery has advanced considerably since the first cochlear implant in 1961 [1], but variability in speech perception outcomes has been reported, with the underlying causes not yet fully understood [2]. Current cochlear implant candidacy includes both individuals with profound deafness and those with residual natural hearing [3]. Maintenance of residual hearing and hearing quality in these individuals has been a major research focus which has led to significant changes in cochlear implant electrode design, composition, and surgical approach. A key factor thought to influence cochlear implant performance is the formation of fibrotic scar tissue and neo-ossification around the implanted electrode. Histopathological analyses of post-mortem temporal bones from cochlear implant recipients consistently reveal varying degrees of fibrosis and ossification surrounding the electrode [4,5,6,7,8,9,10]. Although the effect of implant-induced fibrosis on hearing outcomes remains unclear, these studies indicate that most, if not all, cochlear implant patients develop some degree of intracochlear fibrosis.
Tissue damage from physical trauma, infection, metabolic insults, or ageing disrupts cellular homeostasis and, depending on the type, location and duration of injury, can markedly impact tissue function. Furthermore, the capacity for tissue repair depends on the intrinsic capacity of the cells within that tissue to proliferate or self-renew. Tissues and cells without this capacity heal through a process of scar formation. The cellular and molecular events driving wound healing are tightly regulated to ensure resolution of inflammation, effective repair and to minimise the formation of fibrotic scars. Pathological fibrosis is thought to occur as a consequence of repetitive injury that drives the chronicity of the response through the perpetuation of an exaggerated, overexuberant inflammatory and deregulated wound healing response, which ultimately results in excessive and disorganised extracellular matrix (ECM) deposition at sites of injury [11].
Fibrosis and scarring occur in many clinical settings and are a significant and growing clinical problem. In a global burden of disease analysis, liver cirrhosis and other chronic liver diseases, interstitial lung disease (ILD) and pulmonary sarcoidosis, chronic kidney disease and ischaemic heart disease, were reported to be responsible for 46.2, 3.77, 41.5 and 182 million DALYs (disability adjusted life years), respectively, worldwide [12]. Furthermore, it is estimated that the annual combined global incidence of fibrotic diseases, including heart, liver, kidney, lung, skin and systemic fibrotic disorders is ~4968/100,000 people years [13]. Although the pathogenesis of cochlear fibrosis is not well understood, detailed mechanistic studies on scarring and fibrosis in other organs, along with transcriptome and proteome analyses during fibrosis initiation, have revealed common pathways driving fibrogenesis across tissues. This raises the potential for repurposing existing therapies to treat cochlear fibrosis.
2. Fibrosis in the Inner Ear
In the inner ear, fibrotic scarring is known to occur as a consequence of infections, such as bacterial meningitis [14,15]. Infection is a leading cause of moderate to severe acquired sensorineural hearing loss, which is often permanent. For example, bacterial meningitis can lead to haring loss in up to 32% of children and 69% of adults [16]. Hearing loss may result from the spread of bacterial products and inflammatory mediators through the meninges and cerebrospinal fluid via the cochlear aqueduct [17] or as a consequence of the development of fibrosis. For example, cytomegalovirus infection-induced hearing loss in a mouse model is linked to the development of fibrosis and aberrant macrophage function in the cochlea [18,19]. Furthermore, a clinical study performed using the National Temporal Bone, Hearing and Balance Pathology Resource Registry Database, identified new bone formation or fibrosis in all cases of autoimmune associated hearing loss and in 20–30% of cases of labyrinthitis or meningitis [20]. Sensorineural hearing loss was also identified in 40% of patients with systemic sclerosis (SSc), an autoimmune-mediated fibrotic connective tissue disease with multi-organ involvement [21]. Data from a histological case study also suggested that both the middle and inner ear can be affected by SSc, with evidence of reduced numbers of outer hair cells, dendrites and ganglion cells in the cochlea and perivascular fibrosis linked to vasculitis [22]. One case study also suggested that auditory neuropathy, the functional loss of synapses and/or auditory nerve fibres, occurred as a result of SSc [23]. However, a recent systematic review on the relationship between SSc and hearing loss concluded that although most papers suggest a higher incidence of hearing impairment in patients with SSc, there is a lack of sufficient evidence to provide a firm estimate of prevalence and insight in underlying mechanisms [24].
Significant intracochlear fibrosis and ossification also occurs following cochlear implant surgery, with inflammation driving the formation of dense fibrotic tissue around the electrode array, a process driven by foreign body and wound healing responses initiated by trauma and damage to inner ear tissue at the time of surgery [25] (summarised in Figure 1). A recent clinical study reported cochlear fibrosis/ossification in the basal turn of the cochlea in 5 of 17 (29.4%) cochlear implant revision surgeries [26]. Post-mortem temporal bone studies place this number much higher with fibrosis and ossification observed in almost all individuals who have undergone cochlear implant surgery in their lifetime [4,5,25,27,28]. A recent 3-dimensional imaging study performed on post-mortem temporal bones from 20 patients showed evidence of fibrosis and ossification in all cochleae, and on average, 33% of the cochlear volume was taken up with fibrous and osseous tissue [6].
Figure 1.
The development of fibrosis and neo-ossification after cochlear implantation. The response to cochlear implantation begins at the time of implant insertion (T0), where surgical trauma and cochlear implantation trigger an acute inflammatory response. Blood proteins and platelets accumulate at the insertion site and also adsorb to the surface of the implant forming a provisional matrix. Phagocytic cells including neutrophils and macrophages migrate to the site of injury to clear any cellular debris and adhere to the provisional matrix surrounding the electrode tract where they produce and secrete cytokines which promote wound healing and the foreign body response (FBR). Specifically, these cytokines trigger the activation of macrophages, which fuse to form foreign body giant cells (FBGCs) as well as the migration, proliferation and differentiation of fibroblasts/fibrocytes, cells which are responsible for the deposition of extracellular matrix (ECM) leading to the fibrous encapsulation and subsequent neo-ossification around the electrode. SV: scala vestibuli, SM: scala media, ST: scala tympani, SL: spiral ligament, StV: stria vascularis, CN: cochlear nerve. Created with Biorender.
The specific impact that fibrosis or ossification has on the function of the cochlear implant remains largely unknown. Interestingly, in individuals with autoimmune or immune-mediated sensorineural hearing loss, fibrosis can occur because of systemic disease, and this has been linked to higher variability in cochlear implant outcomes in this population [29]. Several studies have suggested that fibrosis and neo-ossification after cochlear implantation have a detrimental effect on hearing measures in both patient and animal studies. Indeed, animal studies demonstrate a negative relationship between the extent of new tissue growth after cochlear implantation and hearing measures, including auditory brainstem response thresholds and spiral ganglion cell counts [30,31,32]. Several human studies corroborate these data, showing a correlation between fibrosis and neo-ossification with a worsening of hearing measures, including word recognition scores and auditory thresholds [27,33,34]. The potential relationship between fibrosis and hearing loss after cochlear implantation is particularly significant in patients with partial hearing loss receiving hybrid cochlear implants. Within this patient demographic, all studies show an immediate deterioration of residual hearing post-surgery and further hearing loss at various degrees and rates in the long-term [35,36,37,38]. This poses a significant challenge, as electric-acoustic stimulation in cochlear implant users with residual hearing offers substantial benefits over electrical stimulation alone, including improvements in speech [39] and music perception [40,41]. While residual hearing loss may have several causes, including excitotoxicity in sensory cells after inflammation and mechanical trauma to the microstructures of the cochlea, fibrosis also seems to be a significant factor. A case study of post-mortem temporal bone of a cochlear implant user who lost residual hearing after implantation found that fibrosis within the cochlea was the most likely explanation for this residual hearing loss [33]. This is likely due to the effect of fibrosis on cochlear mechanics. Indeed, using mathematical modelling, Choi et al. demonstrated that efforts to preserve residual hearing clinically may be limited by the changes in mechanotransduction caused by intracochlear fibrosis, which dampens the wave response of the basilar membrane [42]. Several animal model studies in different species have confirmed implant-induced tissue reaction in the cochlea [43,44,45,46,47]. O’Leary and colleagues previously demonstrated in an animal model that progressive hearing loss after cochlear implant surgery is linked to the extent of the implant-induced tissue reaction [32] and that low frequency hearing loss can be delayed by altering the surgical approach to implantation [48].
Given evidence that fibrosis in the inner ear increases variability in hearing outcomes following cochlear implantation and may adversely affect residual hearing, a longstanding challenge has been to identify surgical and pharmacological strategies to minimise fibrosis and preserve residual hearing. However, clinical studies have been hampered by the lack of audiological markers for fibrosis. Retrospective analysis of post-mortem temporal bone tissue and linked clinical data suggest electrode impedance and word recognition score as surrogate measures of fibrosis. For example, Kamakura et al. showed a negative correlation between consonant-nucleus-consonant word score and the percentage of intracochlear new bone formation, and this was related to the amount of trauma to the basilar membrane at the time of surgery [27]. A recent study found that higher four-point impedances, reflecting the local environment at the neural-tissue interface, along the electrode array correlate with poorer hearing outcomes. The author suggested that these changes represent physiological alterations in basilar membrane mechanics, likely resulting from fibrotic changes within the cochlea [49]. This aligns with data from a guinea pig model, which demonstrated a correlation between increased impedance and the extent of fibrosis and neo-ossification [50]. While higher impedances have been linked to increased fibrosis, clinical quantitative data correlating electrode impedance and the extent of fibrosis and/or hearing loss remain limited. Heutink et al. performed one of the first studies to correlate the extent of implant-induced neo-ossification with residual hearing. Using ultra-high-resolution CT, they showed that new bone formation led to long-term loss of residual hearing in the low-frequency range [51]. Overall, these studies further support the negative impact intracochlear tissue reaction has on hearing and highlights the need to limit the extent of fibrosis and bone formation post implant.
3. Neo-Ossification
The relationship between neo-ossification and fibrosis after cochlear implantation remains unclear. Neo-ossification is classified as metaplastic, arising from surgical disruption of the inner ear blood supply, or osteoplastic, involving osteoblasts and developing after endosteal damage [52]. Post-implantation neo-ossification is likely osteoplastic. Although it occurs in several inner ear pathologies, including meningitis, otosclerosis, otitis media and trauma, it is unknown whether its development after implantation is directly or indirectly linked to fibrosis [53]. Geerardyn et al. [6] observed a correlation between implant duration and cochlear neo-ossification, but not with fibrosis, suggesting it may represent progressive ECM deposition with fibrosis transitioning to bone [4]. Although some mechanisms driving fibrosis and neo-ossification after cochlear implantation are known, further research is required to clarify these pathways and support development of targeted strategies to reduce both complications.
Like fibrosis, post-implantation neo-ossification may reflect dysregulated wound healing, particularly when the cochlear lateral wall is damaged [28,54]. Mesothelial cells can also transdifferentiate into osteoblastic cells [55], potentially contributing to both fibrosis and neo-ossification. In a mouse model, Bas et al. reported increased expression of bone-associated genes, including bone morphogenic protein 4 (BMP4), connective tissue growth factor (CTGF), and fibroblast growth factor (FGF)7 [56], with higher CTGF levels where fibrosis bordered neo-ossification or bone [57].
Although some mechanisms driving fibrosis and neo-ossification after cochlear implantation are known, further research is required to clarify these pathways and support development of targeted strategies to reduce both complications.
4. Fibrosis in Other Tissues
In most tissues, fibrosis is part of normal wound healing and, in skin, helps maintain barrier integrity. However, excessive dermal scarring can impair thermoregulation, limit tissue movement, cause pain and cosmetic distress, and lead to psychological effects [58]. While most scars remodel, some individuals develop hypertrophic scars with excessive, disordered collagens, or keloids that grow beyond the wound boundary [59].
In organs such as the heart and lung, excessive ECM deposition severely impairs function. For example, after myocardial infarction, ischaemic reperfusion injury leads to functional cardiomyocytes being replaced with scar tissue, disrupting electrical conduction [60]. In the lung, disorganised collagen within the interstitium restricts mechanical function and gas exchange [61]. Treatments to reduce fibrosis are limited. Some interventional approaches, such as pulse field ablation, may help reduce post-infarct arrhythmias [62,63], while antifibrotic drugs like pirfenidone (PFD), have been designed to target transforming growth factor-beta (TGF-β)-driven fibroblast activation [64,65,66,67,68]. In idiopathic pulmonary fibrosis (IPF), both pirfenidone (PFD) and nintedanib (NTD) target fibrosis progression by inhibiting key profibrotic signalling pathways [69,70], although their clinical utility is often constrained by adverse side effects [71,72,73]. Chronic kidney disease is a major global issue, with tubulointerstitial fibrosis the key process leading to functional decline and kidney failure [74]. Abnormal repair involving endothelial, epithelial, mesangial inflammatory and immune cells drives TGF-β-mediated ECM deposition, capillary rarefaction and hypoxia, promoting fibrosis [74,75]. Drugs blocking the renin-angiotensin system, endothelin-1 and vasopressin signalling can slow progression, but no effective antifibrotic therapy exists despite many mechanistic targets under trial [76].
Liver fibrosis results from chronic liver diseases including non-alcoholic fatty liver disease, alcoholism and viral hepatitis [77]. Hepatocyte-derived danger-associated molecular patterns (DAMPs) activate Kupffer and other immune cells, driving hepatic stellate cell transdifferentiation into collagen-producing myofibroblasts [78]. Excess ECM disrupts liver architecture, and as with other fibrotic diseases, no drugs can reverse with transplantation the best option [79].
Fibrosis can also extend beyond the originating tissue, as in keloids or post-operative adhesions, where aberrant serosal healing creates fibrous bands between organs and the body wall. Adhesions restrict abdominal and intrapleural movement, cause pain, and account for most intestinal obstructions and many infertility cases [80,81]. They are dynamic structures containing fibroblasts, blood vessels, smooth muscle and nerves [80], likely formed via cellular invasion and mesothelial cell trans-differentiation [55,82,83]. Fibrosis also forms around implanted medical devices, such as fibrotic sheaths around pacemaker leads [84] due to the foreign body response, which encapsulates foreign material to protect the host [85].
5. Mechanisms of Fibrosis
While the cellular mechanisms driving fibrosis have been thoroughly characterised in some disease settings, others, such as cochlear implant-induced fibrosis, remain comparatively understudied, with only limited investigations to date.
A defining feature of fibrosis is the accumulation of fibroblasts and myofibroblasts at sites of tissue damage or repair. These cells secrete large amounts of ECM proteins, particularly collagen. Myofibroblasts in particular are highly synthetic and play a central role in ECM production and wound contraction [86]. These cells are normally removed by apoptosis during the resolution phase of normal wound healing, however in fibrosis they persist [87,88,89]. In lung fibrosis, myofibroblasts are thought to acquire a senescent phenotype, characterised by reduced mitogenic activity but increased resistance to apoptosis [89,90,91]. The origin of myofibroblasts remains incompletely understood, but several potential sources have been proposed. These include the proliferation, recruitment, and differentiation of resident fibroblasts, as well as contributions from pericytes, endothelial cells, circulating fibrocytes drawn to the injury site, and epithelial cells undergoing epithelial-to-mesenchymal transition (EMT) [92]. EMT is driven by high levels of pro-fibrotic growth factors, particularly TGF-β1, produced at the injury site by resident and immune cells such as macrophages. There is also evidence that monocytes and macrophages can contribute to the myofibroblast pool by differentiating through a macrophage-to-myofibroblast transition (MMT) [93]. Similarly, mesothelial-to-mesenchymal transition (MesoMT) may serve as a source of myofibroblasts in serosal fibrosis, including conditions such as encapsulating peritoneal sclerosis and post-operative adhesion formation [83,94]. Within the liver, hepatic stellate cells are the predominant collagen secreting cells activated by immune cells and pro-fibrogenic mediators to differentiate into myofibroblasts [95]. Fibroblasts, myofibroblasts and macrophages all play a synergistic roll in the propagation of an abnormal tissue repair response, which can progress to fibrosis. In cochlear implantation, a driving factor for dysregulated wound healing is the implant itself, wherein the continued presence of a foreign body leads to chronic activation of the immune system, fibrosis and neo-ossification. In the inner ear, the spiral ligament contains abundant fibrocytes that play an important role in ECM deposition and fibrosis [96,97]. In addition, studies using models of Keyhole Limpet Hemocyanin-induced immune labyrinthitis and platinum wire cochlear implantation in rat showed that cochleae became filled with fibroblastic tissue derived from the mesothelial cells lining the scala tympani and vestibuli [98]. These findings suggest that mesothelial cells may contribute to local fibrosis following cochlear implantation.
Macrophage accumulation is thought to drive the early fibrotic response through secretion of profibrotic mediators which lead to the accumulation, differentiation and activation of fibroblasts/myofibroblasts within the scala [99,100], possibly through MesoMT [83]. Indeed, pro-fibrotic mediators including TGF-β, CTGF, FGF and fibronectin, have been detected as early as 3 h and up to 30 days after cochlear injury or implant insertion [56,101,102,103,104]. Notably, a recent study by Rahman and colleagues reported that macrophage depletion did not alter the extent of fibrosis following cochlear implantation in mice [105]. This suggests that while macrophages are key to the early inflammatory and immune response after implantation, they may not be the primary drivers of fibrosis. Instead, as in other tissues, the accumulation of fibroblastic cells at the cochlear implant site appears to be mediated by pro-fibrotic factors such as TGF-β, CTGF, platelet-derived growth factor (PDGF), and FGF [57,101].
6. Current Prevention and Treatments for Cochlear Implant-Mediated Fibrosis
Fibrosis and/or neo-ossification occurs in nearly all cases of cochlear implantation in both humans and animal models, typically arising at the surgical site and around the implant [5,106,107]. Increasing evidence that fibrosis impairs device function and residual hearing in hybrid (electric-acoustic) cochlear implants highlights the urgent need for strategies to prevent or reduce implantation-induced fibrosis.
6.1. Surgical
Early strategies to limit cochlear implant-associated fibrosis focused on minimising mechanical trauma to the cochlea and its microstructures during surgery and implant insertion [108]. Advances aimed at reducing surgical trauma include the use of round window (RW) insertion instead of cochleostomy, as well as softer, more flexible implant designs [109,110]. These ‘soft surgery’ techniques have been associated with reduced new tissue growth and improved residual hearing [109,111]. Despite the advancements in surgical techniques, fibrosis still limits the therapeutic benefits of implantation in some patients, suggesting that a multi-modal approach, combining surgical refinements with pharmacological interventions, may be necessary to more effectively prevent fibrosis.
6.2. Corticosteroids
Pharmacological modulation of cochlear fibrosis has largely centred around the use of steroids. Corticosteroids are anti-inflammatory synthetic analogues of steroid hormones produced in the adrenal glands [112]. Because postoperative inflammation is associated with fibrosis, neo-ossification, and elevated hearing thresholds, corticosteroids have been evaluated for their potential to limit these adverse tissue responses [113]. Corticosteroid treatment in humans and animals with cochlear implants, whether topical or systemic, short or long term, has been shown to preserve residual hearing and reduce fibrosis [114,115,116,117]. A recent systematic review of human studies reported a tendency for perioperative steroids to have a positive impact, at least in the short term, on hearing and vestibular function preservation in individuals following cochlear implantation [118]. Recent studies have also shown improved preservation of spiral ganglion neurons (SGN) after local dexamethasone elution in chronically stimulated animals [119]. A study using dexamethasone-eluting cochlear implant electrodes in guinea pigs demonstrated significant reductions in fibrotic tissue and electrode impedance compared to untreated controls [117]. Similarly, in humans, cochlear perfusion with the steroid triamcinolone resulted in significantly lower long-term electrode impedance compared to controls [120], indicating reduced fibrosis. In contrast, the systemic delivery of methylprednisolone did not reduce electrode impedance spikes [121], suggesting the anti-inflammatory action of steroids is most effective when locally administered. A more recent study using dexamethasone eluting arrays in a cochlear implant model in guinea pigs, showed inhibition of neo-ossification and reduced SGN loss; however, no effect was observed on the extent of fibrosis [122]. Similarly, a study in non-human primates demonstrated beneficial effects of a dexamethasone-eluting electrode array on tissue reaction and impedance levels [123]. In contrast, studies in a guinea pig model showed no change in monopolar electrode impedance over five weeks with dexamethasone-eluting arrays, and no significant differences in fibrotic tissue formation, new bone growth, or SGN density between array types [50]. However, four-point impedance was significantly lower in the presence of dexamethasone, and this reduction correlated with the percentage of fibrous tissue and new bone growth [50]. Furthermore, reduced electrode impedance in patients which received a dexamethasone eluting electrode as compared to conventional electrodes has been observed in two separate small cohort clinical trials [124,125]. Thus, there is currently a lack of consensus on the beneficial effect of steroids on cochlear implants; however, steroid eluting electrodes hold some promise by providing direct and sustained intracochlear drug delivery but the need for effective drug targeting approaches remains.
7. Novel Therapeutic Approaches for Cochlear Fibrosis Informed by Insights from Other Tissues
A growing body of research has delineated the cellular processes and signalling pathways driving tissue fibrosis, including inflammatory pathways, TGF-β/Mothers against decapentaplegic homologue (Smad), tyrosine kinase (TK) receptors, lysyl oxidase, interleukin (IL)-6/signal transducer and activator of transcription (STAT)3, and Wnt/β-catenin signalling, among others [126,127,128]. These pathways have since become targets for therapy, and many of the anti-fibrotic drugs currently approved for treatment target some of these pathways (summarised in Figure 2). This review will discuss four pathways that are being targeted for anti-fibrotic therapies in various tissues, with potential application in cochlear fibrosis: inflammation, TGF-β/Smad, Wnt/β-catenin and TKs.
Figure 2.
Pathogenic pathways of cochlear fibrosis and drugs in various stages of development or clinical use, focusing on different pathways that converge in fibrotic diseases such as pulmonary and cardiac fibrosis, liver cirrhosis, and systemic sclerosis. Each drug targets specific biological mechanisms contributing to the fibrotic process. Hh: Hedgehog, MSCs: mesenchymal stem cells, PMT: Pericyte to mesenchymal transition. Created with Biorender.
7.1. Inflammation
The role of inflammation and the immune system in fibrosis across various tissues has been widely debated. However, growing evidence now supports that the inflammatory and immune responses play crucial roles in both the development and progression of the fibrotic process [129,130,131]. Many inflammatory mediators and receptors have been identified as being actively involved in fibrosis. For example, the receptor for advance glycation end products (RAGE), is considered to be an important mediator, responsible for driving the inflammatory response in fibrosis. RAGE is a membrane receptor and a member of the receptor immunoglobulin superfamily. There are many variants of RAGE and notably soluble RAGE (sRAGE) is a potential serum biomarker in fibrosis [132]. RAGE activation plays an important role in EMT and DNA repair in lung fibrosis. RAGE expression is decreased in IPF patients [133] and RAGE-knock-out mice develop pulmonary fibrosis [134]. However, the effects of RAGE appear to be organ specific as opposite effects have been described in the liver. For example, RAGE activation in hepatocytes has been shown to stimulate TGF-β and monocyte chemoattractant protein 1 (MCP-1) production and induce fibroblast alpha smooth actin (αSMA) expression and collagen production, and blocking RAGE pathway activation reduced hepatitis C-induced fibrosis [135]. Similarly, SiRNA-mediated RAGE inhibition led to reduced production of profibrotic mediators IL-6, tumour necrosis factor (TNF) and TGF-β [136] and suppressed the development of hepatic fibrosis [137].
7.2. TGF-β/Smad Pathway
One of the key mediators of fibrosis is TGF-β, which is produced by various resident and immune cells, including airway epithelial cells (AECs) and macrophages, and is also present in the latent form within the ECM. TGF-β expression is upregulated in nearly all fibrotic tissues, and for many years, therapeutic approaches have focussed on targeting TGF-β-driven responses [138]. TGF-β is upregulated following cochlear implantation both in human cochlear fibrotic tissue [57] and in the cochleae of implanted mice [56]. As a central mediator of fibrosis, TGF-β exerts its pro-fibrotic effects through either the canonical Smad-dependent or non-canonical Smad-independent pathways and exacerbates fibrogenesis by inducing differentiation of cells into myofibroblasts and inducing ECM production and deposition, in particular collagen [139,140]. The Smad family comprises eight transcriptional regulators downstream of TGF-β receptors, with Smad2, Smad3 and Smad4 acting as the principal effectors. Of these, only phosphorylated Smad3 can directly bind Smad-binding elements within gene promoters to drive transcription, whereas phosphorylated Smad2 and Smad4 lack DNA-binding domains and instead modulate Smad3-dependent activation of fibrogenic genes such as type I collagen [138,141]. In contrast, Smad7 antagonises TGF-β/Smad3 signalling by competing with Smad3 for DNA-binding, making its induction a potential anti-fibrotic strategy [138,142]. TGF-β also activates several non-canonical pathways, including p38 mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K)/Akt, both implicated in EMT [143]. Additionally, Rho-like GTPases promotes cytoskeletal remodelling and mesenchymal traits [144], effects that may be amplified by TGF-β-induced reactive oxygen species (ROS) generated through nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 [145]. ROS can further enhance TGF-β signalling by augmenting Smad activity independently of inflammasome activation [146].
The Janus kinases (JAKs) are receptor-associated TKs that play a role in cytokine and growth factor signalling. Activation of JAK1 by TGF-β leads to early pro-fibrotic tyrosine-phosphorylation of STAT3, independent of Smads [147,148]. However, late phase activation of STAT3 crucially requires integration with Smad3 for proliferation, myofibroblast differentiation, and enhancing TGF-β-mediated transcription of key pro-fibrotic genes [148].
Various approaches have been trialled to block TGF-β signalling and ablate fibrosis with limited effect. The most conventional approach is to block TGF-β and TGF-β receptor interaction using receptor antagonists or monoclonal antibodies. These strategies have shown success in animal models of fibrosis, including those affecting the liver, lung, kidney and heart [141,149,150,151], but have produced limited benefits in clinical trials [152,153]. Likewise, inhibition of Smad3 or overexpression of Smad7 reduces fibrosis in animal models of liver and lung disease and in systemic conditions such as SSc [154,155,156], yet this approach remains difficult to translate clinically.
The most successful anti-fibrotic drug thought to work by inhibiting TGF-β is PFD. PFD is a non-peptide synthetic molecule with well-established anti-fibrotic and anti-inflammatory properties. Although its precise mechanism of action remains unclear, PFD is considered multimodal, inhibiting both TGF-β synthesis and TGF-β-induced collagen production. Preclinical models of fibrosis across multiple organs have shown that PFD can induce cell-cycle arrest and reduce fibroblast proliferation by down-regulating phosphorylated Smad3, α-SMA, S100A4, fibronectin, and components of the MAPK signalling pathway [157,158,159]. Its anti-inflammatory effects are linked to decreasing production of pro-inflammatory cytokines such as TNFα, IL-1β, and IL-6 [159,160]. Since receiving Food and Drug Administration (FDA)-approval for the treatment of IPF [75,161], PFD is being evaluated for its antifibrotic potential in other organ systems [159], and these properties may also be relevant within the cochlea.
7.3. Wnt Pathway
Significant crosstalk between the TGF-β and Wnt pathways amplifies fibrosis by promoting fibroblast activation, myofibroblast differentiation, ECM production, and reduced ECM degradation, creating a self-sustaining feedback loop [162,163,164]. TGF-β upregulates Wnt ligands (e.g., Wnt1, Wnt3a, Wnt5a, Wnt10) and receptors (Frizzled and low-density lipoprotein receptor-related protein (LRP)5/6), stabilises β-catenin through Smad-dependent and independent mechanisms, and suppresses Wnt antagonists such as Dickkopf and Secreted Frizzled-Related Proteins. Wnt signalling, in turn, increases TGF-β receptor expression and promotes TGF-β production, enhancing Smad activation. Both pathways converge on shared transcription factors (T-cell factor/lymphoid enhancer factor for Wnt and Smads for TGF-β) to co-regulate pro-fibrotic genes including ACTA2, COL1A1, FN1 and tissue inhibitor of metalloproteinase 1 [162,163,164].
Evidence for the antifibrotic potential of Wnt inhibition comes from multiple preclinical and early clinical studies. Wnt/β-catenin signalling is consistently elevated in IPF lungs and fibrosis models [165,166,167], and its inhibition reduces myofibroblast differentiation and bleomycin-induced pulmonary fibrosis in mice [168,169]. Wnt10A is overexpressed in IPF, bleomycin models and predicts acute exacerbations [170]. The β-catenin inhibitor PRI-724 decreases fibrosis in bleomycin-treated lungs [171] and reduces hepatic stellate cell activation and ECM deposition in liver fibrosis models [172,173].
Porcupine inhibitors, which block secretion of all Wnt ligands, show broad antifibrotic activity. Wnt-C59 reduces kidney fibrosis in unilateral ureteral obstruction and cardiac fibrosis models [174,175], while GNF6231 mitigates skin fibrosis and pulmonary fibrosis in graft-versus-host disease, with evidence of reversing established fibrosis [176]. Although antibodies such as vantictumab successfully inhibit Wnt signalling via Frizzled receptors in cancer studies, toxicity has limited their use in fibrosis [177].
In the cochlea, Bas et al. identified Wnt signalling as a driver of trauma-induced fibrosis following implantation, contributing to myofibroblast differentiation and ECM production [56]. Targeting Wnt signalling may therefore help reduce cochlear implant-associated fibrosis and improve device performance, although further work is needed to enable clinical translation.
7.4. Tyrosine Kinases
TKs are enzymes that function by transferring a phosphate from an ATP molecule to a tyrosine residue on target proteins, activating them via phosphorylation. They are classified as receptor TKs, membrane receptors activated by growth factor binding (e.g., PDGF, FGF, vascular endothelial growth factor [VEGF]), and non-receptor TKs, which function within the cytoplasm [178]. Most fibrosis-related TKs are receptor kinases regulating cell division, metabolism, migration, adhesion, survival, and apoptosis [179,180]. Their central role in disease has driven the development of TK inhibitors (TKI). Because many TKs share structural similarity, single TKIs often inhibit multiple kinases with varying selectivity. Initially developed for cancer, TKIs are increasingly being explored for fibrotic diseases [180].
NTD is a triple TKI targeting VEGF, PDGF and FGF, approved by the FDA for fibrotic lung diseases [161,181]. It also inhibits phosphorylation of Smad3, STAT3, NF-κB, and Src family kinases [182], thereby blocking fibroblast-myofibroblast differentiation and reducing collagen production [183]. NTD exerts anti-inflammatory effects by shifting macrophage phenotype through indirect inhibition of PI3K/AKT and MAPK/ERK signalling [184] and suppresses Wnt3a-induced myofibroblast activation via the Src/β-catenin pathway [185].
As with PFD, interest has grown in NTD’s broader antifibrotic potential. Beyond lung disease, studies are exploring its efficacy in liver, kidney, skin, heart, and eye fibrosis [182,186], with possible relevance to the cochlea.
Imatinib is a small-molecule inhibitor that targets several TKs, including colony-stimulating factor 1 receptor, ABL, c-KIT, FLT3 (CD135), and PDGF receptor-β. It was first recognised for its antifibrotic potential when patients with chronic myeloid leukaemia treated with imatinib showed regression of bone marrow fibrosis [187]. It was later shown to inhibit fibrosis across multiple animal models, prompting clinical trials in human fibrotic diseases, particularly those affecting the lung. However, results from these trials were highly variable, with an improvement in lung function and reduction in skin fibrosis in some patients with SSc [188,189] but no significant impact in IPF [190]. Imatinib can also improve renal function in patients with nephrogenic systemic fibrosis [191].
Dasatinib is a TKI primarily used to treat chronic myelogenous leukaemia and acute lymphoblastic leukaemia. Dasatinib has also shown antifibrotic potential in preclinical studies. It inhibits TGF-β-mediated EMT in lung epithelial cells and reduces lung fibrosis in mouse models by suppressing TGF-β-induced Smad signalling [192]. Dasatinib also reduces fibrosis in other organs such as in uterine fibrosis [193]. This effect is often enhanced when used in combination with quercetin, as both act as senolytics to selectively eliminate senescent cells, which are thought to play a critical role in the development and progression of fibrosis [89]. Despite promising preclinical findings, a recent phase 1 clinical trial evaluating the combination of dasatinib and quercetin in patients with IPF did not show improvements in lung function. However, the study was underpowered, limiting the strength of any clinical conclusions [194].
There are many other TKIs being investigated as antifibrotics including gefitinib, erlotinib, sunitpinib, adavosertib, lapatinib and nilotinib. Many are being used in clinical trials alone or in combination with other treatments. However, the question remains whether these TKIs have therapeutic effects on cochlear fibrosis following cochlear implantation.
8. Therapeutic Strategies and Challenges: Implications for the Ear
Treating or preventing fibrosis in the inner ear faces two main challenges. Firstly, effective delivery of pharmacological agents to the inner ear is challenging due to its rigid bony capsule, which isolates the fluids of the membranous labyrinths from the middle ear and surrounding skull, including the brain, thereby limiting drug access. In addition, the inner ear fluids are separated from the systemic circulation by the blood-labyrinth barrier which is largely impermeable to most drug classes [195]. The second challenge is the potential for toxicity to the neural elements of the inner ear [196]. Ototoxicity effecting residual hair cells and functioning spiral ganglion cells is not desirable even in profoundly deafened patients, as the loss of these elements may worsen hearing outcomes and even prevent the potential for future use of regenerative therapies [197]. In cochlear implant recipients with partial deafness, preserving residual low-frequency hair cell function and hearing is essential [198]. Therefore, any treatment with potential ototoxic effects would significantly limit its clinical applicability.
Pharmacological Delivery to the Inner Ear
Several methods of drug administration could be used to deliver adjuvant therapies at the time of cochlear implantation; however, each method presents unique benefits and challenges. When delivering therapies to inner ear targets, systemic administration, although straightforward, results in relatively low inner ear bioavailability because of the restrictive blood-labyrinth barrier [195]. Therefore, this approach requires higher dosing, which increased risk of off-target effects. Local delivery of pharmacological agents would be preferable, although also not without challenges. Multiple strategies are available for delivering pharmacological agents to the inner ear, including bulk delivery to the middle ear for diffusion across to the round and oval windows, direct application to the RW membrane, and intracochlear delivery via either the RW or a cochleostomy.
Delivery of pharmacological agents into the middle ear generally involves trans-tympanic injections which perforate the tympanic membrane [199,200,201,202]. Although tympanic membrane healing can take several weeks in some individuals, clinical studies indicate that even repeated injections are generally well tolerated [203]. However, the presence of a substance in the middle ear does not guarantee its effective entry into the inner ear. Firstly, the eustachian tube may present an unintentional exit route from the middle ear [201]. Indeed, when intratympanic injections are used on patients with eustachian tube dysfunction, the result is often better as the drug is present in the middle ear for longer [204]. Secondly, agents must penetrate the oval or RW to enter the inner ear, but this can be impeded by false RW membranes, which occur in up to 20% of patients [205]. Moreover, application to the middle ear does not guarantee that the compound will contact these membranes. Achieving stable membrane contact requires either filling the middle ear cavity with large volumes or using novel delivery systems designed to maintain sustained contact with the RW or oval window [206]. Some of these drug delivery systems, such as gels, aerosols, osmotic pumps or sponges, also reduce eustachian tube clearance [126,203,207,208,209,210]. In addition, many studies have investigated the feasibility of direct application to the RW membrane. This can be achieved via intratympanic injection with delivery to the RW niche [202,211], though most studies use a bullostomy to access the RW [209,212,213,214,215]. Direct application of a drug to the RW allows the use of smaller volumes than bulk middle ear delivery, increases the likelihood of inner ear entry, and reduces clearance via the eustachian tube. Studies have confirmed that direct application to the RW can enhance bioavailability in the inner ear [202,216,217] and result in higher levels and faster entry compared to systemic delivery [218,219]. Third, not all pharmacological agents readily cross the RW membrane. Their ability to do so depends on hydrophobicity, molecular size, concentration gradient, and contact duration. Various strategies have been explored to enhance drug transfer across the RW, including encapsulating hydrophilic therapeutics in nanoparticles, using adjuvants or ultrasound-guided microbubbles to increase RW permeability, and employing hydrogels, sponges, Gelfoam™ (Pharmacia & Upjohn Co., Kalamazoo, MI, USA), or osmotic mini-pumps to prolong contact duration [157,220,221,222,223]. These methods have successfully increased drug delivery to the inner ear [207,224,225,226,227,228,229], although most drugs have limited bioavailability within the cochlea [227,230,231,232]. Any method applying drugs directly to the RW carries an increased risk of damage to the RW compared with generic middle ear delivery. Finally, the most efficient way to deliver therapeutic agents to the inner ear is direct intracochlear application, either via the RW or a cochleostomy [233]. Cochleostomy ensures precise delivery into the perilymph with controlled concentrations. However, direct application can compromise cochlear integrity due to increased intracochlear pressure, alterations in perilymph ionic composition, and inflammation. In addition, intracochlear fluid dynamics limit dispersion [234], concentrating the pharmacological agent at the site of administration, which is generally near the base of the cochlea. Strategies to optimise delivery throughout the cochlea include apical delivery via a pipette sealed into a fenestra and adjusting the osmotic pump rate using an algorithm [234]. Another approach to address fibrosis around cochlear implants is the use of drug-eluting electrodes. This method provides sustained, targeted delivery over time, allowing intervention in both early and late stages of fibrosis.
9. Conclusions
This review highlights numerous therapeutic targets, drawing on extensive research from other organs and tissues. Any candidate must be non-ototoxic and deliverable in a sustained manner without causing mechanical damage to the cochlea. Additionally, accurate quantification of fibrosis is essential to assess effect size relative to the only clinically used drug, corticosteroids, alongside evaluation of cochlear function outcomes.
Funding
C.M.P., W.H.A.M.M., J.K. and M.A. hold National Health and Medical Research Council Grant #2011143. K.J.B. is the recipient of an Australian Government Research Training Programme Scholarship.
Data Availability Statement
No new data were created or analyzed in this study.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
The following abbreviations are used in this manuscript:
| AEC | Airway epithelial cell |
| αSMA | Alpha smooth muscle actin |
| BMP4 | Bone morphogenic protein 4 |
| CN | Cochlear nerve |
| CTGF | Connective tissue growth factor |
| DAMPs | Danger-associated molecular patterns |
| ECM | Extracellular matrix |
| EMT | Epithelial-to-mesenchymal transition |
| FBGCs | Foreign body giant cells |
| FBR | Foreign body response |
| FDA | Food and Drug Administration |
| FGF | Fibroblast growth factor |
| IL | Interleukin |
| IPF | Idiopathic pulmonary fibrosis |
| JAK | Janus kinase |
| LRP | Low-density lipoprotein receptor-related protein |
| MAPK | Mitogen-activated protein kinase |
| MCP-1 | Monocyte chemoattractant protein 1 |
| MesoMT | Mesothelial-to-mesenchymal transition |
| MMT | Macrophage-to-myofibroblast transition |
| NADPH | Nicotinamide adenine dinucleotide phosphate |
| NTD | Nintedanib |
| PFD | Pirfenidone |
| PI3K | Phosphatidylinositol-3-kinase |
| RAGE | Receptor for advance glycation end products |
| RW | Round window |
| SGN | Spiral ganglion neurons |
| SL | Piral ligament |
| SM | Scala media |
| Smad | Mothers against decapentaplegic homologue |
| sRAGE | Soluble receptor for advance glycation end products |
| SSc | Systemic sclerosis |
| ST | Scala tympani |
| Stat | Signal transducer and activator of transcription |
| StV | Stria vascularis |
| SV | Scala vestibuli |
| TGF-β | Transforming growth factor beta |
| TGF-βR | Transforming growth factor beta receptor |
| TK | Tyrosine kinase |
| TKI | Tyrosine kinase inhibitor |
| TNF | Tumour necrosis factor |
| VEGF | Vascular endothelial growth factor |
References
- Mudry, A.; Mills, M. The early history of the cochlear implant: A retrospective. JAMA Otolaryngol. Head Neck Surg. 2013, 139, 446–453. [Google Scholar] [CrossRef] [PubMed]
- Walia, A.; Shew, M.A.; Kallogjeri, D.; Wick, C.C.; Durakovic, N.; Lefler, S.M.; Ortmann, A.J.; Herzog, J.A.; Buchman, C.A. Electrocochleography and cognition are important predictors of speech perception outcomes in noise for cochlear implant recipients. Sci. Rep. 2022, 12, 3083. [Google Scholar] [CrossRef] [PubMed]
- Messersmith, J.J.; Entwisle, L.; Warren, S.; Scott, M. Clinical Practice Guidelines: Cochlear Implants. J. Am. Acad. Audiol. 2019, 30, 827–844. [Google Scholar] [CrossRef] [PubMed]
- Danielian, A.; Ishiyama, G.; Lopez, I.A.; Ishiyama, A. Predictors of Fibrotic and Bone Tissue Formation With 3-D Reconstructions of Post-implantation Human Temporal Bones. Otol. Neurotol. 2021, 42, e942–e948. [Google Scholar] [CrossRef]
- Fayad, J.N.; Makarem, A.O.; Linthicum, F.H., Jr. Histopathologic assessment of fibrosis and new bone formation in implanted human temporal bones using 3D reconstruction. Otolaryngol. Head Neck Surg. 2009, 141, 247–252. [Google Scholar] [CrossRef]
- Geerardyn, A.; Zhu, M.; Wu, P.; O’MAlley, J.T.; Nadol, J.B.; Liberman, M.C.; Nakajima, H.H.; Verhaert, N.; Quesnel, A.M. Three-dimensional quantification of fibrosis and ossification after cochlear implantation via virtual re-sectioning: Potential implications for residual hearing. Hear. Res. 2023, 428, 108681. [Google Scholar] [CrossRef]
- Ishiyama, A.; Ishiyama, G.; Lopez, I.A.; Linthicum, F.H., Jr. Temporal Bone Histopathology of First-Generation Cochlear Implant Electrode Translocation. Otol. Neurotol. 2019, 40, e581–e591. [Google Scholar] [CrossRef]
- Linthicum, F.H., Jr.; Doherty, J.K.; Lopez, I.A.; Ishiyama, A. Cochlear implant histopathology. World J. Otorhinolaryngol. Head Neck Surg. 2017, 3, 211–213. [Google Scholar] [CrossRef]
- Linthicum, F.H., Jr.; Fayad, J.; Otto, S.R.; Galey, F.R.; House, W.F. Cochlear implant histopathology. Am. J. Otol. 1991, 12, 245–311. [Google Scholar] [CrossRef]
- Seyyedi, M.; Nadol, J.B., Jr. Intracochlear inflammatory response to cochlear implant electrodes in humans. Otol. Neurotol. 2014, 35, 1545–1551. [Google Scholar] [CrossRef]
- Mutsaers, S.E.; Bishop, J.E.; McGrouther, G.; Laurent, G.J. Mechanisms of tissue repair: From wound healing to fibrosis. Int. J. Biochem. Cell Biol. 1997, 29, 5–17. [Google Scholar] [CrossRef]
- Diseases, G.B.D.; Injuries, C. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar]
- Zhao, X.; Kwan, J.Y.Y.; Yip, K.; Liu, P.P.; Liu, F.F. Targeting metabolic dysregulation for fibrosis therapy. Nat. Rev. Drug Discov. 2020, 19, 57–75. [Google Scholar] [CrossRef] [PubMed]
- Ge, N.N.; Brodie, H.A.; Tinling, S.P. Long-term hearing loss in gerbils with bacterial meningitis treated with superoxide dismutase. Otol. Neurotol. 2008, 29, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
- Orzan, E.; Muzzi, E.; Caregnato, I.; Cossu, P.; Marchi, R.; Ghiselli, S. Uncommon Post-Meningitis Hearing Threshold Improvement: A Case Report. J. Int. Adv. Otol. 2018, 14, 484–487. [Google Scholar] [CrossRef]
- Lucas, M.J.; Brouwer, M.C.; van de Beek, D. Neurological sequelae of bacterial meningitis. J. Infect. 2016, 73, 18–27. [Google Scholar] [CrossRef]
- Mook-Kanamori, B.B.; Geldhoff, M.; van der Poll, T.; van de Beek, D. Pathogenesis and pathophysiology of pneumococcal meningitis. Clin. Microbiol. Rev. 2011, 24, 557–591. [Google Scholar] [CrossRef]
- Li, X.; Shi, X.; Wang, C.; Niu, H.; Zeng, L.; Qiao, Y.; Xu, K. Pathological changes of the inner ear cochlea in different time windows of murine cytomegalovirus-induced hearing loss in a mouse model. Acta Otolaryngol. 2015, 135, 536–541. [Google Scholar] [CrossRef]
- Schraff, S.A.; Schleiss, M.R.; Brown, D.K.; Meinzen-Derr, J.; Choi, K.Y.; Greinwald, J.H.; Choo, D.I. Macrophage inflammatory proteins in cytomegalovirus-related inner ear injury. Otolaryngol. Head Neck Surg. 2007, 137, 612–618. [Google Scholar] [CrossRef]
- Keithley, E.M.; Chen, M.C.; Linthicum, F. Clinical diagnoses associated with histologic findings of fibrotic tissue and new bone in the inner ear. Laryngoscope 1998, 108, 87–91. [Google Scholar] [CrossRef]
- Maciaszczyk, K.; Waszczykowska, E.; Pajor, A.; Bartkowiak-Dziankowska, B.; Durko, T. Hearing organ disorders in patients with systemic sclerosis. Rheumatol. Int. 2011, 31, 1423–1428. [Google Scholar] [CrossRef]
- Abou-Taleb, A.; Linthicum, F.H., Jr. Scleroderma and hearing loss: (histopathology of a case). J. Laryngol. Otol. 1987, 101, 656–662. [Google Scholar] [CrossRef]
- Santarelli, R.; Scimemi, P.; Dal Monte, E.; Genovese, E.; Arslan, E. Auditory neuropathy in systemic sclerosis: A speech perception and evoked potential study before and after cochlear implantation. Eur. Arch. Otorhinolaryngol. 2006, 263, 809–815. [Google Scholar] [CrossRef]
- Sieskiewicz, M.; Rebacz, D.; Sieskiewicz, A. Hearing impairment in systemic sclerosis patients-what do we really know? Front. Med. 2024, 11, 1322170. [Google Scholar] [CrossRef]
- Foggia, M.J.; Quevedo, R.V.; Hansen, M.R. Intracochlear fibrosis and the foreign body response to cochlear implant biomaterials. Laryngoscope Investig. Otolaryngol. 2019, 4, 678–683. [Google Scholar] [CrossRef] [PubMed]
- Canzano, F.; Di Lella, F.; Guida, M.; Pasanisi, E.; Govoni, M.; Falcioni, M. Revision cochlear implant surgery for clinical reasons. Acta Otorhinolaryngol. Ital. 2023, 43, 65–73. [Google Scholar] [CrossRef] [PubMed]
- Kamakura, T.; Nadol, J.B., Jr. Correlation between word recognition score and intracochlear new bone and fibrous tissue after cochlear implantation in the human. Hear. Res. 2016, 339, 132–141. [Google Scholar] [CrossRef] [PubMed]
- Li, P.M.; Somdas, M.A.; Eddington, D.K.; Nadol, J.B., Jr. Analysis of intracochlear new bone and fibrous tissue formation in human subjects with cochlear implants. Ann. Otol. Rhinol. Laryngol. 2007, 116, 731–738. [Google Scholar] [CrossRef]
- Mancini, P.; Atturo, F.; Di Mario, A.; Portanova, G.; Ralli, M.; De Virgilio, A.; de Vincentiis, M.; Greco, A. Hearing loss in autoimmune disorders: Prevalence and therapeutic options. Autoimmun. Rev. 2018, 17, 644–652. [Google Scholar] [CrossRef]
- Araki, S.; Kawano, A.; Seldon, H.L.; Shepherd, R.K.; Funasaka, S.; Clark, G.M. Effects of intracochlear factors on spiral ganglion cells and auditory brain stem response after long-term electrical stimulation in deafened kittens. Otolaryngol. Head Neck Surg. 2000, 122, 425–433. [Google Scholar] [CrossRef]
- Bas, E.; Bohorquez, J.; Goncalves, S.; Perez, E.; Dinh, C.T.; Garnham, C.; Hessler, R.; Eshraghi, A.A.; Van De Water, T.R. Electrode array-eluted dexamethasone protects against electrode insertion trauma induced hearing and hair cell losses, damage to neural elements, increases in impedance and fibrosis: A dose response study. Hear. Res. 2016, 337, 12–24. [Google Scholar] [CrossRef]
- O’Leary, S.J.; Monksfield, P.; Kel, G.; Connolly, T.; Souter, M.; Chang, A.; Marovic, P.; O’LEary, J.; Richardson, R.; Eastwood, H. Relations between cochlear histopathology and hearing loss in experimental cochlear implantation. Hear. Res. 2013, 298, 27–35. [Google Scholar] [CrossRef] [PubMed]
- Quesnel, A.M.; Nakajima, H.H.; Rosowski, J.J.; Hansen, M.R.; Gantz, B.J.; Nadol, J.B. Delayed loss of hearing after hearing preservation cochlear implantation: Human temporal bone pathology and implications for etiology. Hear. Res. 2016, 333, 225–234. [Google Scholar] [CrossRef] [PubMed]
- Scheperle, R.A.; Tejani, V.D.; Omtvedt, J.K.; Brown, C.J.; Abbas, P.J.; Hansen, M.R.; Gantz, B.J.; Oleson, J.J.; Ozanne, M.V. Delayed changes in auditory status in cochlear implant users with preserved acoustic hearing. Hear. Res. 2017, 350, 45–57. [Google Scholar] [CrossRef] [PubMed]
- Barbara, M.; Mattioni, A.; Monini, S.; Chiappini, I.; Ronchetti, F.; Ballantyne, D.; Mancini, P.; Filipo, R. Delayed loss of residual hearing in Clarion cochlear implant users. J. Laryngol. Otol. 2003, 117, 850–853. [Google Scholar] [CrossRef]
- Gerbert, M.; Ernst, A.; Seidl, R.; Decker, L.; Scholz, S.; Lauer, G.; Mittmann, P. Preservation of Residual Hearing: Long-Term Results With a Mid-Scala Electrode. J. Otolaryngol. Head Neck Surg. 2024, 53, 19160216241250351. [Google Scholar] [CrossRef]
- Kim, J.S. Electrocochleography in Cochlear Implant Users with Residual Acoustic Hearing: A Systematic Review. Int. J. Environ. Res. Public Health 2020, 17, 7043. [Google Scholar] [CrossRef]
- Salmon, M.K.; Quimby, A.E.; Bartellas, M.; Kaufman, H.S.; Bigelow, D.C.; Brant, J.A.; Ruckenstein, M.J. Long-Term Hearing Outcomes After Hybrid Cochlear Implantation. Otol. Neurotol. 2023, 44, 679–683. [Google Scholar] [CrossRef]
- Incerti, P.V.; Ching, T.Y.; Cowan, R. A systematic review of electric-acoustic stimulation: Device fitting ranges, outcomes, and clinical fitting practices. Trends Amplif. 2013, 17, 3–26. [Google Scholar] [CrossRef]
- Miranda, P.C.; Sampaio, A.L.; Lopes, R.A.; Ramos Venosa, A.; de Oliveira, C.A. Hearing preservation in cochlear implant surgery. Int. J. Otolaryngol. 2014, 2014, 468515. [Google Scholar] [CrossRef]
- Parkinson, A.J.; Rubinstein, J.T.; Drennan, W.R.; Dodson, C.; Nie, K. Hybrid Music Perception Outcomes: Implications for Melody and Timbre Recognition in Cochlear Implant Recipients. Otol. Neurotol. 2019, 40, e283–e289. [Google Scholar] [CrossRef]
- Choi, C.H.; Oghalai, J.S. Predicting the effect of post-implant cochlear fibrosis on residual hearing. Hear. Res. 2005, 205, 193–200. [Google Scholar] [CrossRef] [PubMed]
- Braack, K.J.; Miles, T.; Amat, F.; Brown, D.J.; Atlas, M.D.; Kuthubutheen, J.; Mulders, W.H.; Prêle, C.M. Using x-ray micro computed tomography to quantify intracochlear fibrosis after cochlear implantation in a Guinea pig model. Heliyon 2023, 9, e19343. [Google Scholar] [CrossRef] [PubMed]
- Chen, A.; Chen, D.; Lv, K.; Li, G.; Pan, J.; Ma, D.; Tang, J.; Zhang, H. Zwitterionic Polymer/Polydopamine Coating of Electrode Arrays Reduces Fibrosis and Residual Hearing Loss after Cochlear Implantation. Adv. Healthc. Mater. 2023, 12, e2200807. [Google Scholar] [CrossRef]
- Colesa, D.J.; Devare, J.; Swiderski, D.L.; Beyer, L.A.; Raphael, Y.; Pfingst, B.E. Development of a chronically-implanted mouse model for studies of cochlear health and implant function. Hear. Res. 2021, 404, 108216. [Google Scholar] [CrossRef] [PubMed]
- Hyzer, J.M.; Hill, J.D.; He, W.; Burwood, G.W.S.; Fettig, A.K.; Reiss, L.A.J. Effects of Cochlear Implantation and Steroids on the Aging Guinea Pig Cochlea. Otolaryngol. Head Neck Surg. 2024, 171, 530–537. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Ismail, H.; Lee, J.H.; Kel, G.; O’leary, J.; Hampson, A.; Eastwood, H.; O’leary, S.J. Effect of both local and systemically administered dexamethasone on long-term hearing and tissue response in a Guinea pig model of cochlear implantation. Audiol. Neurootol. 2013, 18, 392–405. [Google Scholar] [CrossRef]
- Rowe, D.; Chambers, S.; Hampson, A.; Eastwood, H.; Campbell, L.; O’Leary, S. Delayed low frequency hearing loss caused by cochlear implantation interventions via the round window but not cochleostomy. Hear. Res. 2016, 333, 49–57. [Google Scholar] [CrossRef]
- Bester, C.; Dalbert, A.; Collins, A.; Razmovski, T.; Gerard, J.-M.; O’Leary, S. Electrocochleographic Patterns Predicting Increased Impedances and Hearing Loss after Cochlear Implantation. Ear Hear. 2022, 44, 710–720. [Google Scholar] [CrossRef]
- Needham, K.; Stathopoulos, D.; Newbold, C.; Leavens, J.; Risi, F.; Manouchehri, S.; Durmo, I.; Cowan, R. Electrode impedance changes after implantation of a dexamethasone-eluting intracochlear array. Cochlear Implant. Int. 2020, 21, 98–109. [Google Scholar] [CrossRef]
- Heutink, F.; Klabbers, T.M.; Huinck, W.J.; Lucev, F.; van der Woude, W.J.; Mylanus, E.A.M.; Verbist, B.M. Ultra-High-Resolution CT to Detect Intracochlear New Bone Formation after Cochlear Implantation. Radiology 2022, 302, 605–612. [Google Scholar] [CrossRef]
- Kotzias, S.A.; Linthicum, F.H., Jr. Labyrinthine ossification: Differences between two types of ectopic bone. Am. J. Otol. 1985, 6, 490–494. [Google Scholar]
- Vashishth, A.; Fulcheri, A.; Prasad, S.C.; Bassi, M.; Rossi, G.; Caruso, A.; Sanna, M. Cochlear Implantation in Cochlear Ossification: Retrospective Review of Etiologies, Surgical Considerations, and Auditory Outcomes. Otol. Neurotol. 2018, 39, 17–28. [Google Scholar] [CrossRef]
- Hodge, S.E.; Ishiyama, G.; Lopez, I.A.; Ishiyama, A. Histopathologic Analysis of Temporal Bones With Otosclerosis Following Cochlear Implantation. Otol. Neurotol. 2021, 42, 1492–1498. [Google Scholar] [CrossRef] [PubMed]
- Lansley, S.M.; Searles, R.G.; Hoi, A.; Thomas, C.; Moneta, H.; Herrick, S.E.; Thompson, P.J.; Mark, N.; Sterrett, G.F.; Prêle, C.M.; et al. Mesothelial cell differentiation into osteoblast- and adipocyte-like cells. J. Cell. Mol. Med. 2011, 15, 2095–2105. [Google Scholar] [CrossRef] [PubMed]
- Bas, E.; Anwar, M.R.; Van De Water, T.R. TGF beta-1 and WNT Signaling Pathways Collaboration Associated with Cochlear Implantation Trauma-Induced Fibrosis. Anat. Rec. 2020, 303, 608–618. [Google Scholar] [CrossRef] [PubMed]
- Xiao, A.Y.; Lopez, I.A.; Ishiyama, G.; Ishiyama, A. Expression of TGFbeta-1 and CTGF in the Implanted Cochlea and its Implication on New Tissue Formation. Otol. Neurotol. 2024, 45, 810–817. [Google Scholar] [CrossRef]
- Ngaage, M.; Agius, M. The Psychology of Scars: A Mini-Review. Psychiatr. Danub. 2018, 30, 633–638. [Google Scholar]
- He, Y.; Deng, Z.; Alghamdi, M.; Lu, L.; Fear, M.W.; He, L. From genetics to epigenetics: New insights into keloid scarring. Cell Prolif. 2017, 50, e12326. [Google Scholar] [CrossRef]
- Hashimoto, H.; Olson, E.N.; Bassel-Duby, R. Therapeutic approaches for cardiac regeneration and repair. Nat. Rev. Cardiol. 2018, 15, 585–600. [Google Scholar] [CrossRef]
- Martinez, F.J.; Collard, H.R.; Pardo, A.; Raghu, G.; Richeldi, L.; Selman, M.; Swigris, J.J.; Taniguchi, H.; Wells, A.U. Idiopathic pulmonary fibrosis. Nat. Rev. Dis. Primers 2017, 3, 17074. [Google Scholar] [CrossRef]
- Sandhu, U.; Alkukhun, L.; Kheiri, B.; Hodovan, J.; Chiang, K.; Splanger, T.; Castellvi, Q.; Zhao, Y.; Nazer, B. In vivo pulsed-field ablation in healthy vs. chronically infarcted ventricular myocardium: Biophysical and histologic characterization. Europace 2023, 25, 1503–1509. [Google Scholar] [CrossRef]
- Younis, A.; Zilberman, I.; Krywanczyk, A.; Higuchi, K.; Yavin, H.D.; Sroubek, J.; Anter, E. Effect of Pulsed-Field and Radiofrequency Ablation on Heterogeneous Ventricular Scar in a Swine Model of Healed Myocardial Infarction. Circ. Arrhythm. Electrophysiol. 2022, 15, e011209. [Google Scholar] [CrossRef]
- Fu, Y.; Shi, J.; Qian, H.; Qin, C.; Liu, L.; Shen, J.; Ma, H.; Ma, L.; Liao, B.; Guo, Y. Alleviation of cardiac fibrosis using acellular peritoneal matrix-loaded pirfenidone nanodroplets after myocardial infarction in rats. Eur. J. Pharmacol. 2022, 933, 175238. [Google Scholar] [CrossRef] [PubMed]
- Lewis, G.A.; Dodd, S.; Clayton, D.; Bedson, E.; Eccleson, H.; Schelbert, E.B.; Naish, J.H.; Jimenez, B.D.; Williams, S.G.; Cunnington, C.; et al. Pirfenidone in heart failure with preserved ejection fraction: A randomized phase 2 trial. Nat. Med. 2021, 27, 1477–1482. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Hang, W.; Shu, H.; Zhou, N. Pirfenidone alleviates cardiac fibrosis induced by pressure overload via inhibiting TGF-beta1/Smad3 signalling pathway. J. Cell. Mol. Med. 2022, 26, 4548–4555. [Google Scholar] [CrossRef] [PubMed]
- Meyer, F.E.U.; Santos, G.L.; Doan, T.P.; DeGrave, A.N.; Bues, B.; Lutz, S. Pirfenidone affects human cardiac fibroblast proliferation and cell cycle activity in 2D cultures and engineered connective tissues. Naunyn Schmiedebergs Arch. Pharmacol. 2023, 396, 1687–1699. [Google Scholar] [CrossRef]
- Yamagami, K.; Oka, T.; Wang, Q.; Ishizu, T.; Lee, J.-K.; Miwa, K.; Akazawa, H.; Naito, A.T.; Sakata, Y.; Komuro, I. Pirfenidone exhibits cardioprotective effects by regulating myocardial fibrosis and vascular permeability in pressure-overloaded hearts. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H512–H522. [Google Scholar] [CrossRef]
- Aimo, A.; Spitaleri, G.; Nieri, D.; Tavanti, L.M.; Meschi, C.; Panichella, G.; Lupón, J.; Pistelli, F.; Carrozzi, L.; Bayes-Genis, A.; et al. Pirfenidone for Idiopathic Pulmonary Fibrosis and Beyond. Card. Fail. Rev. 2022, 8, e12. [Google Scholar] [CrossRef]
- Rangarajan, S.; Kurundkar, A.; Kurundkar, D.; Bernard, K.; Sanders, Y.Y.; Ding, Q.; Antony, V.B.; Zhang, J.; Zmijewski, J.; Thannickal, V.J. Novel Mechanisms for the Antifibrotic Action of Nintedanib. Am. J. Respir. Cell Mol. Biol. 2016, 54, 51–59. [Google Scholar] [CrossRef]
- Finnerty, J.P.; Ponnuswamy, A.; Dutta, P.; Abdelaziz, A.; Kamil, H. Efficacy of antifibrotic drugs, nintedanib and pirfenidone, in treatment of progressive pulmonary fibrosis in both idiopathic pulmonary fibrosis (IPF) and non-IPF: A systematic review and meta-analysis. BMC Pulm. Med. 2021, 21, 411. [Google Scholar] [CrossRef]
- He, M.; Yang, T.; Zhou, J.; Wang, R.; Li, X. A real-world study of antifibrotic drugs-related adverse events based on the United States food and drug administration adverse event reporting system and VigiAccess databases. Front. Pharmacol. 2024, 15, 1310286. [Google Scholar] [CrossRef]
- Mackintosh, J.A.; Keir, G.; Troy, L.K.; Holland, A.E.; Grainge, C.; Chambers, D.C.; Sandford, D.; Jo, H.E.; Glaspole, I.; Wilsher, M.; et al. Treatment of idiopathic pulmonary fibrosis and progressive pulmonary fibrosis: A position statement from the Thoracic Society of Australia and New Zealand 2023 revision. Respirology 2024, 29, 105–135. [Google Scholar] [CrossRef]
- Djudjaj, S.; Boor, P. Cellular and molecular mechanisms of kidney fibrosis. Mol. Asp. Med. 2019, 65, 16–36. [Google Scholar] [CrossRef]
- Duffield, J.S. Cellular and molecular mechanisms in kidney fibrosis. J. Clin. Investig. 2014, 124, 2299–2306. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Fu, P.; Ma, L. Kidney fibrosis: From mechanisms to therapeutic medicines. Signal Transduct. Target. Ther. 2023, 8, 129. [Google Scholar] [CrossRef]
- Roehlen, N.; Crouchet, E.; Baumert, T.F. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar] [CrossRef] [PubMed]
- Tsuchida, T.; Friedman, S.L. Mechanisms of hepatic stellate cell activation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 397–411. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Tuo, B. Current and Emerging Approaches for Hepatic Fibrosis Treatment. Gastroenterol. Res. Pract. 2021, 2021, 6612892. [Google Scholar] [CrossRef]
- Herrick, S.E.; Mutsaers, S.E.; Ozua, P.; Sulaiman, H.; Omer, A.; Boulos, P.; Foster, M.L.; Laurent, G.J. Human peritoneal adhesions are highly cellular, innervated, and vascularized. J. Pathol. 2000, 192, 67–72. [Google Scholar] [CrossRef]
- Sulaiman, H.; Gabella, G.; Davis, C.M.; Mutsaers, S.E.; Boulos, P.M.; Laurent, G.J.; Herrick, S.E. Presence and distribution of sensory nerve fibers in human peritoneal adhesions. Ann. Surg. 2001, 234, 256–261. [Google Scholar] [CrossRef] [PubMed]
- Herrick, S.E.; Mutsaers, S.E. The potential of mesothelial cells in tissue engineering and regenerative medicine applications. Int. J. Artif. Organs 2007, 30, 527–540. [Google Scholar] [CrossRef] [PubMed]
- Mutsaers, S.E.; Birnie, K.; Lansley, S.; Herrick, S.E.; Lim, C.B.; Prêle, C.M. Mesothelial cells in tissue repair and fibrosis. Front. Pharmacol. 2015, 6, 113. [Google Scholar] [CrossRef] [PubMed]
- Perry, L.; Karp, F.; Hauch, K.; Ratner, B.D. Explanted pacemakers: Observations of the long-term foreign body response. Interface 2007, 4, 11. [Google Scholar]
- Anderson, J.M.; Rodriguez, A.; Chang, D.T. Foreign body reaction to biomaterials. Semin. Immunol. 2008, 20, 86–100. [Google Scholar] [CrossRef]
- Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
- Hung, C.F. Origin of Myofibroblasts in Lung Fibrosis. Curr. Tissue Microenviron. Rep. 2020, 1, 155–162. [Google Scholar] [CrossRef]
- Virk, H.S.; Biddle, M.S.; Smallwood, D.T.; Weston, C.A.; Castells, E.; Bowman, V.W.; McCarthy, J.; Amrani, Y.; Duffy, S.M.; Bradding, P.; et al. TGFbeta1 induces resistance of human lung myofibroblasts to cell death via down-regulation of TRPA1 channels. Br. J. Pharmacol. 2021, 178, 2948–2962. [Google Scholar] [CrossRef]
- Waters, D.W.; Blokland, K.E.C.; Pathinayake, P.S.; Burgess, J.K.; Mutsaers, S.E.; Prele, C.M.; Schuliga, M.; Grainge, C.L.; Knight, D.A. Fibroblast senescence in the pathology of idiopathic pulmonary fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 315, L162–L172. [Google Scholar] [CrossRef]
- Hinz, B.; Lagares, D. Evasion of apoptosis by myofibroblasts: A hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 2020, 16, 11–31. [Google Scholar] [CrossRef]
- Waters, D.W.; Schuliga, M.; Pathinayake, P.S.; Wei, L.; Tan, H.-Y.; Blokland, K.E.C.; Jaffar, J.; Westall, G.P.; Burgess, J.K.; Prêle, C.M.; et al. A Senescence Bystander Effect in Human Lung Fibroblasts. Biomedicines 2021, 9, 1162. [Google Scholar] [CrossRef]
- Moustakas, A.; Heldin, C.H. Mechanisms of TGFbeta-Induced Epithelial-Mesenchymal Transition. J. Clin. Med. 2016, 5, 63. [Google Scholar] [CrossRef] [PubMed]
- Vierhout, M.; Ayoub, A.; Naiel, S.; Yazdanshenas, P.; Revill, S.D.; Reihani, A.; Dvorkin-Gheva, A.; Shi, W.; Ask, K. Monocyte and macrophage derived myofibroblasts: Is it fate? A review of the current evidence. Wound Repair. Regen. 2021, 29, 548–562. [Google Scholar] [CrossRef] [PubMed]
- Terri, M.; Trionfetti, F.; Montaldo, C.; Cordani, M.; Tripodi, M.; Lopez-Cabrera, M.; Strippoli, R. Mechanisms of Peritoneal Fibrosis: Focus on Immune Cells-Peritoneal Stroma Interactions. Front. Immunol. 2021, 12, 607204. [Google Scholar] [CrossRef] [PubMed]
- Akkiz, H.; Gieseler, R.K.; Canbay, A. Liver Fibrosis: From Basic Science towards Clinical Progress, Focusing on the Central Role of Hepatic Stellate Cells. Int. J. Mol. Sci. 2024, 25, 7873. [Google Scholar] [CrossRef]
- Bianchetti, L.; Barczyk, M.; Cardoso, J.; Schmidt, M.; Bellini, A.; Mattoli, S. Extracellular matrix remodelling properties of human fibrocytes. J. Cell. Mol. Med. 2012, 16, 483–495. [Google Scholar] [CrossRef]
- Strieter, R.M.; Keeley, E.C.; Hughes, M.A.; Burdick, M.D.; Mehrad, B. The role of circulating mesenchymal progenitor cells (fibrocytes) in the pathogenesis of pulmonary fibrosis. J. Leukoc. Biol. 2009, 86, 1111–1118. [Google Scholar] [CrossRef]
- Jia, H.; François, F.; Bourien, J.; Eybalin, M.; Lloyd, R.; Van De Water, T.; Puel, J.-L.; Venail, F. Prevention of trauma-induced cochlear fibrosis using intracochlear application of anti-inflammatory and antiproliferative drugs. Neuroscience 2016, 316, 261–278. [Google Scholar] [CrossRef]
- Zhang, D.; Chen, D.; Wang, K.; Pan, J.; Tang, J.; Zhang, H. Electrical stimulation of cochlear implant promotes activation of macrophages and fibroblasts under inflammation. Laryngoscope Investig. Otolaryngol. 2023, 8, 1390–1400. [Google Scholar] [CrossRef]
- Bas, E.; Goncalves, S.; Adams, M.; Dinh, C.T.; Bas, J.M.; Van De Water, T.R.; Eshraghi, A.A. Spiral ganglion cells and macrophages initiate neuro-inflammation and scarring following cochlear implantation. Front. Cell. Neurosci. 2015, 9, 303. [Google Scholar] [CrossRef]
- Bas, E.; Dinh, C.T.; Ojo, R.; Eshraghi, A.A.; Van De Water, T.R. Loss of Residual Hearing Initiated by Cochlear Implantation: Role of Inflammation-Initiated Cell Death Pathways, Wound Healing and Fibrosis Pathways, and Potential Otoprotective Therapies. In Free Radicals in ENT Pathology; Miller, J., Le Prell, C., Rybak, L., Eds.; Humana Press: Totowa, NJ, USA; Springer: New York, NY, USA, 2015; pp. 395–421. [Google Scholar]
- Claussen, A.D.; Quevedo, R.V.; Kirk, J.R.; Higgins, T.; Mostaert, B.; Rahman, M.T.; Oleson, J.; Hernandez, R.; Hirose, K.; Hansen, M.R. Chronic cochlear implantation with and without electric stimulation in a mouse model induces robust cochlear influx of CX3CR1(+/GFP) macrophages. Hear. Res. 2022, 426, 108510. [Google Scholar] [CrossRef]
- Satoh, H.; Billings, P.; Firestein, G.S.; Harris, J.P.; Keithley, E.M. Transforming growth factor beta expression during an inner ear immune response. Ann. Otol. Rhinol. Laryngol. 2006, 115, 81–88. [Google Scholar] [CrossRef]
- Zhang, H.; Stark, G.; Reiss, L. Changes in Gene Expression and Hearing Thresholds After Cochlear Implantation. Otol. Neurotol. 2015, 36, 1157–1165. [Google Scholar] [CrossRef]
- Rahman, M.T.; Mostaert, B.J.; Hunger, B.; Saha, U.; Claussen, A.D.; Razu, I.; Nasrin, F.; Khan, N.A.; Eckard, P.; Coleman, S.; et al. Contribution of macrophages to neural survival and intracochlear tissue remodeling responses following cochlear implantation. J. Neuroinflamm. 2023, 20, 266. [Google Scholar] [CrossRef] [PubMed]
- Nadol, J.B., Jr.; O’Malley, J.T.; Burgess, B.J.; Galler, D. Cellular immunologic responses to cochlear implantation in the human. Hear. Res. 2014, 318, 11–17. [Google Scholar] [CrossRef] [PubMed]
- Nadol, J.B., Jr.; Burgess, B.J.; Gantz, B.J.; Coker, N.J.; Ketten, D.R.; Kos, I.; Roland, J.T.; Shiao, J.Y.; Eddington, D.K.; Montandon, P.; et al. Histopathology of cochlear implants in humans. Ann. Otol. Rhinol. Laryngol. 2001, 110, 883–891. [Google Scholar] [CrossRef] [PubMed]
- Lehnhardt, E. Intracochlear placement of cochlear implant electrodes in soft surgery technique. HNO 1993, 41, 356–359. [Google Scholar]
- Berrettini, S.; Forli, F.; Passetti, S. Preservation of residual hearing following cochlear implantation: Comparison between three surgical techniques. J. Laryngol. Otol. 2008, 122, 246–252. [Google Scholar] [CrossRef]
- Ishai, R.; Herrmann, B.S.; Nadol, J.B., Jr.; Quesnel, A.M. The pattern and degree of capsular fibrous sheaths surrounding cochlear electrode arrays. Hear. Res. 2017, 348, 44–53. [Google Scholar] [CrossRef]
- Gu, P.; Jiang, Y.; Gao, X.; Huang, S.; Yuan, Y.; Wang, G.; Li, B.; Xi, X.; Dai, P. Effects of cochlear implant surgical technique on post-operative electrode impedance. Acta Otolaryngol. 2016, 136, 677–681. [Google Scholar] [CrossRef]
- Reichardt, S.D.; Amouret, A.; Muzzi, C.; Vettorazzi, S.; Tuckermann, J.P.; Lühder, F.; Reichardt, H.M. The Role of Glucocorticoids in Inflammatory Diseases. Cells 2021, 10, 2921. [Google Scholar] [CrossRef]
- Ryu, K.A.; Lyu, A.-R.; Park, H.; Choi, J.W.; Hur, G.M.; Park, Y.-H. Intracochlear Bleeding Enhances Cochlear Fibrosis and Ossification: An Animal Study. PLoS ONE 2015, 10, e0136617. [Google Scholar] [CrossRef]
- Kuthubutheen, J.; Coates, H.; Rowsell, C.; Nedzelski, J.; Chen, J.M.; Lin, V. The role of extended preoperative steroids in hearing preservation cochlear implantation. Hear. Res. 2015, 327, 257–264. [Google Scholar] [CrossRef] [PubMed]
- Kuthubutheen, J.; Joglekar, S.; Smith, L.; Friesen, L.; Smilsky, K.; Millman, T.; Ng, A.; Shipp, D.; Coates, H.; Arnoldner, C.; et al. The Role of Preoperative Steroids for Hearing Preservation Cochlear Implantation: Results of a Randomized Controlled Trial. Audiol. Neurootol. 2017, 22, 292–302. [Google Scholar] [CrossRef] [PubMed]
- Briggs, R.; O’Leary, S.; Birman, C.; Plant, K.; English, R.; Dawson, P.; Risi, F.; Gavrilis, J.; Needham, K.; Cowan, R. Comparison of electrode impedance measures between a dexamethasone-eluting and standard Cochlear Contour Advance(R) electrode in adult cochlear implant recipients. Hear. Res. 2020, 390, 107924. [Google Scholar] [CrossRef] [PubMed]
- Wilk, M.; Hessler, R.; Mugridge, K.; Jolly, C.; Fehr, M.; Lenarz, T.; Scheper, V. Impedance Changes and Fibrous Tissue Growth after Cochlear Implantation Are Correlated and Can Be Reduced Using a Dexamethasone Eluting Electrode. PLoS ONE 2016, 11, e0147552. [Google Scholar] [CrossRef]
- Pimentel de Morais, C.; Branco, P.; Pereira, A.; Castelhano, L.; Donato, M.; Correia, F.; O’Neill, A.; Santos, R.; Escada, P. The Role of Steroids in the Preservation of Hearing and Vestibular Function in Cochlear Implantation. Laryngoscope 2024, 134, 3458–3465. [Google Scholar] [CrossRef]
- Scheper, V.; Hessler, R.; Hütten, M.; Wilk, M.; Jolly, C.; Lenarz, T.; Paasche, G. Local inner ear application of dexamethasone in cochlear implant models is safe for auditory neurons and increases the neuroprotective effect of chronic electrical stimulation. PLoS ONE 2017, 12, e0183820. [Google Scholar] [CrossRef]
- De Ceulaer, G.; Johnson, S.; Yperman, M.; Daemers, K.; Offeciers, F.E.; O’Donoghue, G.M.; Govaerts, P.J. Long-term evaluation of the effect of intracochlear steroid deposition on electrode impedance in cochlear implant patients. Otol. Neurotol. 2003, 24, 769–774. [Google Scholar] [CrossRef]
- Choi, J.; Payne, M.R.; Campbell, L.J.; Bester, C.W.; Newbold, C.; Eastwood, H.; O’Leary, S.J. Electrode Impedance Fluctuations as a Biomarker for Inner Ear Pathology After Cochlear Implantation. Otol. Neurotol. 2017, 38, 1433–1439. [Google Scholar] [CrossRef]
- Chambers, S.; Newbold, C.; Stathopoulos, D.; Needham, K.; Miller, C.; Risi, F.; Enke, Y.L.; Timbol, G.; Cowan, R. Protecting against electrode insertion trauma using dexamethasone. Cochlear Implant. Int. 2019, 20, 1–11. [Google Scholar] [CrossRef]
- Manrique-Huarte, R.; Zulueta-Santos, C.; Calavia, D.; de Linera-Alperi, M.Á.; Gallego, M.A.; Jolly, C.; Manrique, M. Cochlear Implantation With a Dexamethasone Eluting Electrode Array: Functional and Anatomical Changes in Non-Human Primates. Otol. Neurotol. 2020, 41, e812–e822. [Google Scholar] [CrossRef] [PubMed]
- Prenzler, N.; Salcher, R.; Büchner, A.; Warnecke, A.; Kley, D.; Batsoulis, C.; Vormelcher, S.; Mitterberger-Vogt, M.; Morettini, S.; Schilp, S.; et al. Cochlear implantation with a dexamethasone-eluting electrode array: First-in-human safety and performance results. Hear. Res. 2025, 461, 109255. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.T.; Mostaert, B.; Eckard, P.; Fatima, S.M.; Scheperle, R.; Razu, I.; Hunger, B.; Olszewski, R.T.; Gu, S.; Garcia, C.; et al. Cochlear implants with dexamethasone-eluting electrode arrays reduce foreign body response in a murine model of cochlear implantation and human subjects. medRxiv 2024. [Google Scholar] [CrossRef] [PubMed]
- Borthwick, L.A.; Wynn, T.A.; Fisher, A.J. Cytokine mediated tissue fibrosis. Biochim. Biophys. Acta 2013, 1832, 1049–1060. [Google Scholar] [CrossRef]
- Knight, D.; Mutsaers, S.E.; Prele, C.M. STAT3 in tissue fibrosis: Is there a role in the lung? Pulm. Pharmacol. Ther. 2011, 24, 193–198. [Google Scholar] [CrossRef]
- Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef]
- Antar, S.A.; Ashour, N.A.; Marawan, M.E.; Al-Karmalawy, A.A. Fibrosis: Types, Effects, Markers, Mechanisms for Disease Progression, and Its Relation with Oxidative Stress, Immunity, and Inflammation. Int. J. Mol. Sci. 2023, 24, 4004. [Google Scholar] [CrossRef]
- Mack, M. Inflammation and fibrosis. Matrix Biol. 2018, 68–69, 106–121. [Google Scholar] [CrossRef]
- Mutsaers, S.E.; Miles, T.; Prele, C.M.; Hoyne, G.F. Emerging role of immune cells as drivers of pulmonary fibrosis. Pharmacol. Ther. 2023, 252, 108562. [Google Scholar] [CrossRef]
- Machahua, C.; Montes-Worboys, A.; Planas-Cerezales, L.; Buendia-Flores, R.; Molina-Molina, M.; Vicens-Zygmunt, V. Serum AGE/RAGEs as potential biomarker in idiopathic pulmonary fibrosis. Respir. Res. 2018, 19, 215. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, K.; Iwamoto, H.; Mazur, W.; Miura, S.; Sakamoto, S.; Horimasu, Y.; Masuda, T.; Miyamoto, S.; Nakashima, T.; Ohshimo, S.; et al. Reduced endogenous secretory RAGE in blood and bronchoalveolar lavage fluid is associated with poor prognosis in idiopathic pulmonary fibrosis. Respir. Res. 2020, 21, 145. [Google Scholar] [CrossRef] [PubMed]
- Englert, J.M.; Hanford, L.E.; Kaminski, N.; Tobolewski, J.M.; Tan, R.J.; Fattman, C.L.; Ramsgaard, L.; Richards, T.J.; Loutaev, I.; Nawroth, P.P.; et al. A role for the receptor for advanced glycation end products in idiopathic pulmonary fibrosis. Am. J. Pathol. 2008, 172, 583–591. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Zhu, J.; Huang, Y.; Gao, H.; Zhao, Y. Advanced glycation end product (AGE)-induced hepatic stellate cell activation via autophagy contributes to hepatitis C-related fibrosis. Acta Diabetol. 2015, 52, 959–969. [Google Scholar] [CrossRef]
- Wang, X.W.; Li, W.D.; Xia, J.R.; Li, Z.; Cai, X.G. Small interfering RNA targeting receptor for advanced glycation end products suppresses the generation of proinflammatory cytokines. Exp. Ther. Med. 2015, 10, 584–590. [Google Scholar] [CrossRef]
- Cai, X.G.; Xia, J.-R.; Li, W.-D.; Lu, F.-L.; Liu, J.; Lu, Q.; Zhi, H. Anti-fibrotic effects of specific-siRNA targeting of the receptor for advanced glycation end products in a rat model of experimental hepatic fibrosis. Mol. Med. Rep. 2014, 10, 306–314. [Google Scholar] [CrossRef]
- Frangogiannis, N. Transforming growth factor-beta in tissue fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef]
- Budi, E.H.; Schaub, J.R.; Decaris, M.; Turner, S.; Derynck, R. TGF-beta as a driver of fibrosis: Physiological roles and therapeutic opportunities. J. Pathol. 2021, 254, 358–373. [Google Scholar] [CrossRef]
- Ong, C.H.; Tham, C.L.; Harith, H.H.; Firdaus, N.; Israf, D.A. TGF-beta-induced fibrosis: A review on the underlying mechanism and potential therapeutic strategies. Eur. J. Pharmacol. 2021, 911, 174510. [Google Scholar] [CrossRef]
- Aashaq, S.; Batool, A.; Mir, S.A.; Beigh, M.A.; Andrabi, K.I.; Shah, Z.A. TGF-beta signaling: A recap of SMAD-independent and SMAD-dependent pathways. J. Cell. Physiol. 2022, 237, 59–85. [Google Scholar] [CrossRef]
- Hawinkels, L.J.; Ten Dijke, P. Exploring anti-TGF-beta therapies in cancer and fibrosis. Growth Factors 2011, 29, 140–152. [Google Scholar] [CrossRef]
- Yu, L.; Hebert, M.C.; Zhang, Y.E. TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. EMBO J. 2002, 21, 3749–3759. [Google Scholar] [CrossRef]
- Bhowmick, N.A.; Ghiassi, M.; Bakin, A.; Aakre, M.; Lundquist, C.A.; Engel, M.E.; Arteaga, C.L.; Moses, H.L. Transforming growth factor-beta1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol. Biol. Cell 2001, 12, 27–36. [Google Scholar] [CrossRef]
- Liu, R.M.; Desai, L.P. Reciprocal regulation of TGF-beta and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol. 2015, 6, 565–577. [Google Scholar] [CrossRef] [PubMed]
- Bracey, N.A.; Gershkovich, B.; Chun, J.; Vilaysane, A.; Meijndert, H.C.; Wright, J.R.; Fedak, P.W.; Beck, P.L.; Muruve, D.A.; Duff, H.J. Mitochondrial NLRP3 protein induces reactive oxygen species to promote Smad protein signaling and fibrosis independent from the inflammasome. J. Biol. Chem. 2014, 289, 19571–19584. [Google Scholar] [CrossRef] [PubMed]
- Finnson, K.W.; Almadani, Y.; Philip, A. Non-canonical (non-SMAD2/3) TGF-beta signaling in fibrosis: Mechanisms and targets. Semin. Cell Dev. Biol. 2020, 101, 115–122. [Google Scholar] [CrossRef] [PubMed]
- Tang, L.Y.; Heller, M.; Meng, Z.; Yu, L.-R.; Tang, Y.; Zhou, M.; Zhang, Y.E. Transforming Growth Factor-beta (TGF-beta) Directly Activates the JAK1-STAT3 Axis to Induce Hepatic Fibrosis in Coordination with the SMAD Pathway. J. Biol. Chem. 2017, 292, 4302–4312. [Google Scholar] [CrossRef]
- Breitkopf, K.; Haas, S.; Wiercinska, E.; Singer, M.V.; Dooley, S. Anti-TGF-beta strategies for the treatment of chronic liver disease. Alcohol. Clin. Exp. Res. 2005, 29, 121S–131S. [Google Scholar] [CrossRef]
- Denis, M. Neutralization of transforming growth factor-beta 1 in a mouse model of immune-induced lung fibrosis. Immunology 1994, 82, 584–590. [Google Scholar]
- Varga, J.; Pasche, B. Transforming growth factor beta as a therapeutic target in systemic sclerosis. Nat. Rev. Rheumatol. 2009, 5, 200–206. [Google Scholar] [CrossRef]
- Park, C.H.; Yoo, T.H. TGF-beta Inhibitors for Therapeutic Management of Kidney Fibrosis. Pharmaceuticals 2022, 15, 1485. [Google Scholar] [CrossRef] [PubMed]
- Peng, D.; Fu, M.; Wang, M.; Wei, Y.; Wei, X. Targeting TGF-beta signal transduction for fibrosis and cancer therapy. Mol. Cancer 2022, 21, 104. [Google Scholar] [CrossRef] [PubMed]
- Lan, H.Y. Smad7 as a therapeutic agent for chronic kidney diseases. Front. Biosci. 2008, 13, 4984–4992. [Google Scholar] [CrossRef] [PubMed]
- O’Donoghue, R.J.; Knight, D.A.; Richards, C.D.; Prêle, C.M.; Lau, H.L.; Jarnicki, A.G.; Jones, J.; Bozinovski, S.; Vlahos, R.; Thiem, S.; et al. Genetic partitioning of interleukin-6 signalling in mice dissociates Stat3 from Smad3-mediated lung fibrosis. EMBO Mol. Med. 2012, 4, 939–951. [Google Scholar] [CrossRef]
- Roberts, A.B.; Russo, A.; Felici, A.; Flanders, K.C. Smad3: A key player in pathogenetic mechanisms dependent on TGF-beta. Ann. N. Y. Acad. Sci. 2003, 995, 1–10. [Google Scholar] [CrossRef]
- Cai, H.; Liang, Z.; Huang, W.; Wen, L.; Chen, G. Engineering PLGA nano-based systems through understanding the influence of nanoparticle properties and cell-penetrating peptides for cochlear drug delivery. Int. J. Pharm. 2017, 532, 55–65. [Google Scholar] [CrossRef]
- Marwitz, S.; Turkowski, K.; Nitschkowski, D.; Weigert, A.; Brandenburg, J.; Reiling, N.; Thomas, M.; Reck, M.; Drömann, D.; Seeger, W.; et al. The Multi-Modal Effect of the Anti-fibrotic Drug Pirfenidone on NSCLC. Front. Oncol. 2019, 9, 1550. [Google Scholar] [CrossRef]
- Lopez-de la Mora, D.A.; Sanchez-Roque, C.; Montoya-Buelna, M.; Sanchez-Enriquez, S.; Lucano-Landeros, S.; Macias-Barragan, J.; Armendariz-Borunda, J. Role and New Insights of Pirfenidone in Fibrotic Diseases. Int. J. Med. Sci. 2015, 12, 840–847. [Google Scholar] [CrossRef]
- Schaefer, C.J.; Ruhrmund, D.W.; Pan, L.; Seiwert, S.D.; Kossen, K. Antifibrotic activities of pirfenidone in animal models. Eur. Respir. Rev. 2011, 20, 85–97. [Google Scholar] [CrossRef]
- Myllarniemi, M.; Kaarteenaho, R. Pharmacological treatment of idiopathic pulmonary fibrosis-preclinical and clinical studies of pirfenidone, nintedanib, and N-acetylcysteine. Eur. Clin. Respir. J. 2015, 2, 26385. [Google Scholar] [CrossRef]
- Bagheri, L.; Javanbakht, M.; Malekian, S.; Ghahderijani, B.H.; Taghipour, S.; Tanha, F.D.; Ranjkesh, M.; Cegolon, L.; Zhao, S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/beta-catenin and TGF-beta signal transduction pathways as potential targets. Eur. J. Pharmacol. 2025, 999, 177607. [Google Scholar] [CrossRef] [PubMed]
- Gumede, D.B.; Abrahamse, H.; Houreld, N.N. Targeting Wnt/beta-catenin signaling and its interplay with TGF-beta and Notch signaling pathways for the treatment of chronic wounds. Cell Commun. Signal. 2024, 22, 244. [Google Scholar] [CrossRef] [PubMed]
- Yousefi, F.; Shabaninejad, Z.; Vakili, S.; Derakhshan, M.; Movahedpour, A.; Dabiri, H.; Ghasemi, Y.; Mahjoubin-Tehran, M.; Nikoozadeh, A.; Savardashtaki, A.; et al. TGF-beta and WNT signaling pathways in cardiac fibrosis: Non-coding RNAs come into focus. Cell Commun. Signal. 2020, 18, 87. [Google Scholar] [CrossRef] [PubMed]
- Konigshoff, M.; Balsara, N.; Pfaff, E.-M.; Kramer, M.; Chrobak, I.; Seeger, W.; Eickelberg, O. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS ONE 2008, 3, e2142. [Google Scholar] [CrossRef]
- Liu, L.; Carron, B.; Yee, H.T.; Yie, T.-A.; Hajjou, M.; Rom, W.N. Wnt pathway in pulmonary fibrosis in the bleomycin mouse model. J. Environ. Pathol. Toxicol. Oncol. 2009, 28, 99–108. [Google Scholar] [CrossRef]
- Shi, J.; Li, F.; Luo, M.; Wei, J.; Liu, X. Distinct Roles of Wnt/beta-Catenin Signaling in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Mediat. Inflamm. 2017, 2017, 3520581. [Google Scholar] [CrossRef]
- Cao, H.; Wang, C.; Chen, X.; Hou, J.; Xiang, Z.; Shen, Y.; Han, X. Inhibition of Wnt/beta-catenin signaling suppresses myofibroblast differentiation of lung resident mesenchymal stem cells and pulmonary fibrosis. Sci. Rep. 2018, 8, 13644. [Google Scholar] [CrossRef]
- Henderson, W.R., Jr.; Chi, E.Y.; Ye, X.; Nguyen, C.; Tien, Y.-T.; Zhou, B.; Borok, Z.; Knight, D.A.; Kahn, M. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc. Natl. Acad. Sci. USA 2010, 107, 14309–14314. [Google Scholar] [CrossRef]
- Oda, K.; Yatera, K.; Izumi, H.; Ishimoto, H.; Yamada, S.; Nakao, H.; Hanaka, T.; Ogoshi, T.; Noguchi, S.; Mukae, H. Profibrotic role of WNT10A via TGF-beta signaling in idiopathic pulmonary fibrosis. Respir. Res. 2016, 17, 39. [Google Scholar] [CrossRef]
- Okazaki, H.; Sato, S.; Koyama, K.; Morizumi, S.; Abe, S.; Azuma, M.; Chen, Y.; Goto, H.; Aono, Y.; Ogawa, H.; et al. The novel inhibitor PRI-724 for Wnt/beta-catenin/CBP signaling ameliorates bleomycin-induced pulmonary fibrosis in mice. Exp. Lung Res. 2019, 45, 188–199. [Google Scholar] [CrossRef]
- Osawa, Y.; Oboki, K.; Imamura, J.; Kojika, E.; Hayashi, Y.; Hishima, T.; Saibara, T.; Shibasaki, F.; Kohara, M.; Kimura, K. Inhibition of Cyclic Adenosine Monophosphate (cAMP)-response Element-binding Protein (CREB)-binding Protein (CBP)/beta-Catenin Reduces Liver Fibrosis in Mice. EBioMedicine 2015, 2, 1751–1758. [Google Scholar] [CrossRef] [PubMed]
- Yamaji, K.; Iwabuchi, S.; Tokunaga, Y.; Hashimoto, S.; Yamane, D.; Toyama, S.; Kono, R.; Kitab, B.; Tsukiyama-Kohara, K.; Osawa, Y.; et al. Molecular insights of a CBP/beta-catenin-signaling inhibitor on nonalcoholic steatohepatitis-induced liver fibrosis and disorder. Biomed. Pharmacother. 2023, 166, 115379. [Google Scholar] [CrossRef] [PubMed]
- Madan, B.; Patel, M.B.; Zhang, J.; Bunte, R.M.; Rudemiller, N.P.; Griffiths, R.; Virshup, D.M.; Crowley, S.D. Experimental inhibition of porcupine-mediated Wnt O-acylation attenuates kidney fibrosis. Kidney Int. 2016, 89, 1062–1074. [Google Scholar] [CrossRef] [PubMed]
- Paul, M.A.; Wainwright, C.L.; Hector, E.E.; Ryberg, E.; Leslie, S.J.; Walsh, S.K. Short-Term Oral Administration of the Porcupine Inhibitor, Wnt-c59, Improves the Structural and Functional Features of Experimental HFpEF. Pharmacol. Res. Perspect. 2025, 13, e70054. [Google Scholar] [CrossRef]
- Chen, C.W.; Beyer, C.; Liu, J.; Maier, C.; Li, C.; Trinh-Minh, T.; Xu, X.; Cole, S.H.; Hsieh, M.H.; Ng, N.; et al. Pharmacological inhibition of porcupine induces regression of experimental skin fibrosis by targeting Wnt signalling. Ann. Rheum. Dis. 2017, 76, 773–778. [Google Scholar] [CrossRef]
- Flanagan, D.J.; Barker, N.; Di Costanzo, N.S.; Mason, E.A.; Gurney, A.; Meniel, V.S.; Koushyar, S.; Austin, C.R.; Ernst, M.; Pearson, H.B.; et al. Frizzled-7 Is Required for Wnt Signaling in Gastric Tumors with and Without Apc Mutations. Cancer Res. 2019, 79, 970–981. [Google Scholar] [CrossRef]
- Köstler, W.J. Targeting Receptor Tyrosine Kinases in Cancer. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease; Wheeler, D., Yarden, Y., Eds.; Springer: New York, NY, USA, 2015; pp. 225–278. [Google Scholar]
- Beyer, C.; Distler, J.H. Tyrosine kinase signaling in fibrotic disorders: Translation of basic research to human disease. Biochim. Biophys. Acta 2013, 1832, 897–904. [Google Scholar] [CrossRef]
- Mendoza, F.A.; Piera-Velazquez, S.; Jimenez, S.A. Tyrosine kinases in the pathogenesis of tissue fibrosis in systemic sclerosis and potential therapeutic role of their inhibition. Transl. Res. 2021, 231, 139–158. [Google Scholar] [CrossRef]
- Mazzei, M.E.; Richeldi, L.; Collard, H.R. Nintedanib in the treatment of idiopathic pulmonary fibrosis. Ther. Adv. Respir. Dis. 2015, 9, 121–129. [Google Scholar] [CrossRef]
- Li, Z.; Liu, X.; Wang, B.; Nie, Y.; Wen, J.; Wang, Q.; Gu, C. Pirfenidone suppresses MAPK signalling pathway to reverse epithelial-mesenchymal transition and renal fibrosis. Nephrology 2017, 22, 589–597. [Google Scholar] [CrossRef]
- Huang, J.; Beyer, C.; Palumbo-Zerr, K.; Zhang, Y.; Ramming, A.; Distler, A.; Gelse, K.; Distler, O.; Schett, G.; Wollin, L.; et al. Nintedanib inhibits fibroblast activation and ameliorates fibrosis in preclinical models of systemic sclerosis. Ann. Rheum. Dis. 2016, 75, 883–890. [Google Scholar] [CrossRef]
- Bellamri, N.; Morzadec, C.; Joannes, A.; Lecureur, V.; Wollin, L.; Jouneau, S.; Vernhet, L. Alteration of human macrophage phenotypes by the anti-fibrotic drug nintedanib. Int. Immunopharmacol. 2019, 72, 112–123. [Google Scholar] [CrossRef]
- Li, X.; Liu, X.; Deng, R.; Gao, S.; Yu, H.; Huang, K.; Jiang, Q.; Liu, R.; Li, X.; Zhang, L.; et al. Nintedanib Inhibits Wnt3a-Induced Myofibroblast Activation by Suppressing the Src/beta-Catenin Pathway. Front. Pharmacol. 2020, 11, 310. [Google Scholar]
- Liu, F.; Bayliss, G.; Zhuang, S. Application of nintedanib and other potential anti-fibrotic agents in fibrotic diseases. Clin. Sci. 2019, 133, 1309–1320. [Google Scholar] [CrossRef] [PubMed]
- Beham-Schmid, C.; Apfelbeck, U.; Sill, H.; Tsybrovsky, O.; Höfler, G.; Haas, O.A.; Linkesch, W. Treatment of chronic myelogenous leukemia with the tyrosine kinase inhibitor STI571 results in marked regression of bone marrow fibrosis. Blood 2002, 99, 381–383. [Google Scholar] [CrossRef] [PubMed]
- Fraticelli, P.; Gabrielli, B.; Pomponio, G.; Valentini, G.; Bosello, S.; Riboldi, P.; Gerosa, M.; Faggioli, P.; Giacomelli, R.; Del Papa, N.; et al. Low-dose oral imatinib in the treatment of systemic sclerosis interstitial lung disease unresponsive to cyclophosphamide: A phase II pilot study. Arthritis Res. Ther. 2014, 16, R144. [Google Scholar] [CrossRef]
- Spiera, R.F.; Gordon, J.K.; Mersten, J.N.; Magro, C.M.; Mehta, M.; Wildman, H.F.; Kloiber, S.; Kirou, K.A.; Lyman, S.; Crow, M.K. Imatinib mesylate (Gleevec) in the treatment of diffuse cutaneous systemic sclerosis: Results of a 1-year, phase IIa, single-arm, open-label clinical trial. Ann. Rheum. Dis. 2011, 70, 1003–1009. [Google Scholar] [CrossRef]
- Daniels, C.E.; Lasky, J.A.; Limper, A.H.; Mieras, K.; Gabor, E.; Schroeder, D.R. Imatinib treatment for idiopathic pulmonary fibrosis: Randomized placebo-controlled trial results. Am. J. Respir. Crit. Care Med. 2010, 181, 604–610. [Google Scholar] [CrossRef]
- Mansour, J.; Coleman, C.; Mendoza, F.; Lammi, M.; Saketkoo, L.A. Nephrogenic systemic fibrosis-related pulmonary restriction: An under-appreciated manifestation potentially reversible with imatinib therapy. J. Scleroderma Relat. Disord. 2022, 7, NP7–NP11. [Google Scholar] [CrossRef]
- Kanemaru, R.; Takahashi, F.; Kato, M.; Mitsuishi, Y.; Tajima, K.; Ihara, H.; Hidayat, M.; Wirawan, A.; Koinuma, Y.; Hayakawa, D.; et al. Dasatinib Suppresses TGFbeta-Mediated Epithelial-Mesenchymal Transition in Alveolar Epithelial Cells and Inhibits Pulmonary Fibrosis. Lung 2018, 196, 531–541. [Google Scholar] [CrossRef]
- Cavalcante, M.B.; Saccon, T.D.; Nunes, A.D.; Kirkland, J.L.; Tchkonia, T.; Schneider, A.; Masternak, M.M. Dasatinib plus quercetin prevents uterine age-related dysfunction and fibrosis in mice. Aging 2020, 12, 2711–2722. [Google Scholar] [CrossRef]
- Nambiar, A.; Kellogg, D.; Justice, J.; Goros, M.; Gelfond, J.; Pascual, R.; Hashmi, S.; Masternak, M.; Prata, L.; LeBrasseur, N.; et al. Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: Results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability. EBioMedicine 2023, 90, 104481. [Google Scholar] [CrossRef] [PubMed]
- Jahnke, K. The fine structure of freeze-fractured intercellular junctions in the guinea pig inner ear. Acta Otolaryngol. Suppl. 1975, 336, 1–40. [Google Scholar] [CrossRef] [PubMed]
- Coffin, A.B.; Boney, R.; Hill, J.; Tian, C.; Steyger, P.S. Detecting Novel Ototoxins and Potentiation of Ototoxicity by Disease Settings. Front. Neurol. 2021, 12, 725566. [Google Scholar] [CrossRef] [PubMed]
- Seyyedi, M.; Viana, L.M.; Nadol, J.B., Jr. Within-subject comparison of word recognition and spiral ganglion cell count in bilateral cochlear implant recipients. Otol. Neurotol. 2014, 35, 1446–1450. [Google Scholar] [CrossRef]
- Van de Heyning, P.H.; Dazert, S.; Gavilan, J.; Lassaletta, L.; Lorens, A.; Rajan, G.P.; Skarzynski, H.; Skarzynski, P.H.; Tavora-Vieira, D.; Topsakal, V.; et al. Systematic Literature Review of Hearing Preservation Rates in Cochlear Implantation Associated With Medium- and Longer-Length Flexible Lateral Wall Electrode Arrays. Front. Surg. 2022, 9, 893839. [Google Scholar] [CrossRef]
- Choe, W.T.; Chinosornvatana, N.; Chang, K.W. Prevention of cisplatin ototoxicity using transtympanic N-acetylcysteine and lactate. Otol. Neurotol. 2004, 25, 910–915. [Google Scholar] [CrossRef]
- Fetoni, A.R.; Eramo, S.L.M.; Di Pino, A.; Rolesi, R.; Paciello, F.; Grassi, C.; Troiani, D.; Paludetti, G. The Antioxidant Effect of Rosmarinic Acid by Different Delivery Routes in the Animal Model of Noise-Induced Hearing Loss. Otol. Neurotol. 2018, 39, 378–386. [Google Scholar] [CrossRef]
- Zou, J.; Yoshida, T.; Ramadan, U.A.; Pyykko, I. Dynamic enhancement of the rat inner ear after ultra-small-volume administration of Gd-DOTA to the medial wall of the middle ear cavity. ORL J. Otorhinolaryngol. Relat. Spec. 2011, 73, 275–281. [Google Scholar] [CrossRef]
- Ghossaini, S.N.; Liu, J.P.; Phillips, B. Round window membrane permeability to golimumab in guinea pigs: A pilot study. Laryngoscope 2013, 123, 2840–2844. [Google Scholar] [CrossRef]
- Staecker, H.; Morelock, M.; Kramer, T.; Chrbolka, P.; Ahn, J.H.; Meyer, T. Safety of Repeated-Dose Intratympanic Injections with AM-101 in Acute Inner Ear Tinnitus. Otolaryngol. Head Neck Surg. 2017, 157, 478–487. [Google Scholar] [CrossRef] [PubMed]
- Chavkin, U.; Pitaro, J.; Gavriel, H.; Taha, A.; Kalmovich, L.M.; Shilo, S.; Handzel, O.; Muhanna, N.; Warshavsky, A.; Horowitz, G.; et al. The impact of eustachian tube function on intra-tympanic steroid administration. Eur. Arch. Otorhinolaryngol. 2023, 280, 143–149. [Google Scholar] [CrossRef] [PubMed]
- Alzamil, K.S.; Linthicum, F.H., Jr. Extraneous round window membranes and plugs: Possible effect on intratympanic therapy. Ann. Otol. Rhinol. Laryngol. 2000, 109, 30–32. [Google Scholar] [CrossRef] [PubMed]
- Goycoolea, M.V.; Lundman, L. Round window membrane. Structure function and permeability: A review. Microsc. Res. Tech. 1997, 36, 201–211. [Google Scholar] [CrossRef]
- Liao, A.H.; Shih, C.-P.; Li, M.-W.; Lin, Y.-C.; Chuang, H.-C.; Wang, C.-H. Development of thermosensitive poloxamer 407-based microbubble gel with ultrasound mediation for inner ear drug delivery. Drug Deliv. 2021, 28, 1256–1271. [Google Scholar] [CrossRef] [PubMed]
- Gausterer, J.C.; Saidov, N.; Ahmadi, N.; Zhu, C.; Wirth, M.; Reznicek, G.; Arnoldner, C.; Gabor, F.; Honeder, C. Intratympanic application of poloxamer 407 hydrogels results in sustained N-acetylcysteine delivery to the inner ear. Eur. J. Pharm. Biopharm. 2020, 150, 143–155. [Google Scholar] [CrossRef]
- Miwa, T.; Saito, H.; Akita, H. Lipid nanoparticles-encapsulated brain-derived neurotrophic factor mRNA delivered through the round window niche in the cochleae of guinea pigs. Exp. Brain Res. 2021, 239, 425–433. [Google Scholar] [CrossRef]
- Praetorius, M.; Limberger, A.; Müller, M.; Lehner, R.; Schick, B.; Zenner, H.-P.; Plinkert, P.; Knipper, M. A novel microperfusion system for the long-term local supply of drugs to the inner ear: Implantation and function in the rat model. Audiol. Neurootol. 2001, 6, 250–258. [Google Scholar] [CrossRef]
- Jaudoin, C.; Carré, F.; Gehrke, M.; Sogaldi, A.; Steinmetz, V.; Hue, N.; Cailleau, C.; Tourrel, G.; Nguyen, Y.; Ferrary, E.; et al. Transtympanic injection of a liposomal gel loaded with N-acetyl-L-cysteine: A relevant strategy to prevent damage induced by cochlear implantation in guinea pigs? Int. J. Pharm. 2021, 604, 120757. [Google Scholar] [CrossRef]
- Rousset, F.; Kokje, V.B.C.; Coelho, M.D.C.; Mugnier, T.; Belissa, E.; Gabriel, D.; Gurny, R.; Krause, K.H.; Senn, P. Poly-Lactic Acid-Based Biopolymer Formulations Are Safe for Sustained Intratympanic Dexamethasone Delivery. Otol. Neurotol. 2019, 40, e739–e746. [Google Scholar] [CrossRef]
- Oishi, N.; Chen, F.Q.; Zheng, H.W.; Sha, S.H. Intra-tympanic delivery of short interfering RNA into the adult mouse cochlea. Hear. Res. 2013, 296, 36–41. [Google Scholar] [CrossRef]
- Lichtenhan, J.T.; Hartsock, J.; Dornhoffer, J.R.; Donovan, K.M.; Salt, A.N. Drug delivery into the cochlear apex: Improved control to sequentially affect finely spaced regions along the entire length of the cochlear spiral. J. Neurosci. Methods 2016, 273, 201–209. [Google Scholar] [CrossRef]
- Saber, A.; Strand, S.P.; Ulfendahl, M. Use of the biodegradable polymer chitosan as a vehicle for applying drugs to the inner ear. Eur. J. Pharm. Sci. 2010, 39, 110–115. [Google Scholar] [CrossRef]
- Borden, R.C.; Saunders, J.E.; Berryhill, W.E.; Krempl, G.A.; Thompson, D.M.; Queimado, L. Hyaluronic acid hydrogel sustains the delivery of dexamethasone across the round window membrane. Audiol. Neurootol. 2011, 16, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Mikulec, A.A.; Plontke, S.K.; Hartsock, J.J.; Salt, A.N. Entry of substances into perilymph through the bone of the otic capsule after intratympanic applications in guinea pigs: Implications for local drug delivery in humans. Otol. Neurotol. 2009, 30, 131–138. [Google Scholar] [CrossRef] [PubMed]
- Kanzaki, S.; Fujioka, M.; Yasuda, A.; Shibata, S.; Nakamura, M.; Okano, H.J.; Ogawa, K.; Okano, H. Novel in vivo imaging analysis of an inner ear drug delivery system in mice: Comparison of inner ear drug concentrations over time after transtympanic and systemic injections. PLoS ONE 2012, 7, e48480. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Han, L.; Diao, T.; Jing, Y.; Wang, L.; Zheng, H.; Ma, X.; Qi, J.; Yu, L. A comparison of systemic and local dexamethasone administration: From perilymph/cochlea concentration to cochlear distribution. Hear. Res. 2018, 370, 1–10. [Google Scholar] [CrossRef]
- Coleman, J.K.; Littlesunday, C.; Jackson, R.; Meyer, T. AM-111 protects against permanent hearing loss from impulse noise trauma. Hear. Res. 2007, 226, 70–78. [Google Scholar] [CrossRef]
- Jaffredo, M.; Duchamp, O.; Touya, N.; Bouleau, Y.; Dulon, D.; Devillard, R.; Bonnard, D. Proof of concept of intracochlear drug administration by laser-assisted bioprinting in mice. Hear. Res. 2023, 438, 108880. [Google Scholar] [CrossRef]
- Lundy, L.; Karatayli Ozgursoy, S.; Kleindienst, S. Intratympanic Dexamethasone via Saturated Gelfoam for Idiopathic Sudden Sensorineural Hearing Loss. Otolaryngol. Head Neck Surg. 2019, 160, 361–363. [Google Scholar] [CrossRef]
- Wang, J.; Youngblood, R.; Cassinotti, L.; Skoumal, M.; Corfas, G.; Shea, L. An injectable PEG hydrogel controlling neurotrophin-3 release by affinity peptides. J. Control Release 2021, 330, 575–586. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.H.; Nguyen, T.N.; Han, Y.-M.; Tran, P.; Rho, J.; Lee, J.-Y.; Son, H.-Y.; Park, J.-S. Local drug delivery using poly(lactic-co-glycolic acid) nanoparticles in thermosensitive gels for inner ear disease treatment. Drug Deliv. 2021, 28, 2268–2277. [Google Scholar] [CrossRef]
- Paulson, D.P.; Abuzeid, W.; Jiang, H.; Oe, T.; O’Malley, B.W.; Li, D. A novel controlled local drug delivery system for inner ear disease. Laryngoscope 2008, 118, 706–711. [Google Scholar] [CrossRef] [PubMed]
- Yoon, J.Y.; Yang, K.J.; Park, S.N.; Kim, D.K.; Kim, J.D. The effect of dexamethasone/cell-penetrating peptide nanoparticles on gene delivery for inner ear therapy. Int. J. Nanomed. 2016, 11, 6123–6134. [Google Scholar] [CrossRef] [PubMed]
- Creber, N.J.; Eastwood, H.T.; Hampson, A.J.; Tan, J.; O’Leary, S.J. Adjuvant agents enhance round window membrane permeability to dexamethasone and modulate basal to apical cochlear gradients. Eur. J. Pharm. Sci. 2019, 126, 69–81. [Google Scholar] [CrossRef]
- Eastwood, H.; Chang, A.; Kel, G.; Sly, D.; Richardson, R.; O’Leary, S.J. Round window delivery of dexamethasone ameliorates local and remote hearing loss produced by cochlear implantation into the second turn of the guinea pig cochlea. Hear. Res. 2010, 265, 25–29. [Google Scholar] [CrossRef]
- Scheper, V.; Seidel-Effenberg, I.; Lenarz, T.; Stover, T.; Paasche, G. Consecutive Treatment with Brain-Derived Neurotrophic Factor and Electrical Stimulation Has a Protective Effect on Primary Auditory Neurons. Brain Sci. 2020, 10, 559. [Google Scholar] [CrossRef]
- Salt, A.N.; Hartsock, J.; Plontke, S.; LeBel, C.; Piu, F. Distribution of dexamethasone and preservation of inner ear function following intratympanic delivery of a gel-based formulation. Audiol. Neurootol. 2011, 16, 323–335. [Google Scholar] [CrossRef]
- Wang, X.; Chen, Y.; Tao, Y.; Gao, Y.; Yu, D.; Wu, H. A666-conjugated nanoparticles target prestin of outer hair cells preventing cisplatin-induced hearing loss. Int. J. Nanomed. 2018, 13, 7517–7531. [Google Scholar] [CrossRef]
- Wang, X.; Dellamary, L.; Fernandez, R.; Harrop, A.; Keithley, E.M.; Harris, J.P.; Ye, Q.; Lichter, J.; LeBel, C.; Piu, F. Dose-dependent sustained release of dexamethasone in inner ear cochlear fluids using a novel local delivery approach. Audiol. Neurootol. 2009, 14, 393–401. [Google Scholar] [CrossRef]
- Forouzandeh, F.; Borkholder, D.A. Microtechnologies for inner ear drug delivery. Curr. Opin. Otolaryngol. Head Neck Surg. 2020, 28, 323–328. [Google Scholar] [CrossRef]
- Salt, A.N. Pharmacokinetics of Drug Entry into Cochlear Fluids. Volta. Rev. 2005, 105, 277–298. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).