Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 28.
Published in final edited form as: Semin Ophthalmol. 2021 Feb 27;36(4):224–231. doi: 10.1080/08820538.2021.1890793

Nerve Growth Factor as an Ocular Therapy: Applications, Challenges, and Future Directions

Levi N Kanu 1, Joseph B Ciolino 1
PMCID: PMC8551931  NIHMSID: NIHMS1746729  PMID: 33641595

Abstract

Nerve growth factor (NGF), the prototypical neurotrophin first discovered in the 1950s, has recently garnered increased interest as a therapeutic agent promoting neuronal health and regeneration. After gaining orphan drug status within the last decade, NGF-related research and drug development has accelerated. The purpose of this article is to review the preclinical and clinical evidence of NGF in various applications, including central and peripheral nervous system, skin, and ophthalmic disorders. We focus on the ophthalmic applications including not only the FDA-approved indication of neurotrophic keratitis but also retinal disease and glaucoma. NGF represents a promising therapy whose therapeutic profile is evolving. The challenges related to this therapy are reviewed, along with possible solutions and future directions.

Keywords: Nerve growth factor, NGF, neurotrophic keratitis, neurotrophic keratopathy, neurotrophin

INTRODUCTION

Nerve growth factor (NGF) was first identified as a protein secreted by mouse sarcomas with the ability to stimulate growth in sympathetic and sensory neurons.1,2 Since then, NGF has become prototypical of a class of proteins – neurotrophins – characterized by their involvement in neuronal protection and growth, including brain-derived nerve growth factor, neurotrophin-3, and neurotrophin-4/5.

The proenzyme of NGF, pro-NGF, is secreted and latently present in tissue until activation by cleavage. Pro-NGF consists of a complex of three subunits: alpha (α), beta (β), and gamma (γ). The active form of NGF is a dimer of β-NGF (molecular weight 26-kDa) consisting of two non-covalently bonded β subunits. Upon cleavage of pro-NGF, dimerized β-NGF is free to bind cell-surface receptors.3,4 Neurotrophic effects are primarily mediated by tropomyosin kinase receptor A (TrkANGFR),5,6 with secondary effects mediated by the p75 neurotrophin receptor (p75NTR).7 TrkA is a high-affinity receptor whose activation acts via a mitogen activated protein kinase pathway, a phosphoinositide 3-kinase pathway, and a phospholipase 3 pathway to produce downstream effects.8, 1 In addition, the activated NGF/TrkANGFR complex is internalized in an endocytic vesicle and undergoes retrograde transport from the synaptic terminal membrane to the neuronal cell body to effect transcriptional responses,911 which may have effects distinct from the cell surface signaling pathways.12 Equally as versatile as its molecular pathways are the various cell signaling mechanisms of NGF, including endocrine, paracrine, and autocrine pathways.13 Moreover, the signaling mechanisms and pathways of NGF interact with various neuronal and non-neuronal cells, including fibroblasts, glial cells, epithelial cells, immune cells, and neuroendocrine cells.1316

Considering its diversity of targets and effects, NGF has been investigated for use in a variety of clinical applications, including peripheral sensory neuropathies, skin ulcers, neurotrophic keratopathy, and central nervous system disorders such as Alzheimer’s and Parkinson’s disease.17 In this article, we review the clinical applications and challenges of using NGF as a therapy, with an emphasis on ocular indications.

NON-OPHTHALMIC APPLICATIONS

Central Nervous System

NGF has been extensively studied in the treatment of degenerative diseases of the central nervous system such as Alzheimer’s Disease and Parkinson’s Disease. Part of the pathophysiology of these neurodegenerative conditions is thought to be related to diminished production of neurotrophic factors, especially among the cholinergic neurons of the basal forebrain.18,19 Oral and parenteral routes of NGF administration have had limited success, due to poor blood-brain barrier penetration and enzymatic degradation.20,21 Therefore, NGF has been delivered to the CNS primarily through intracerebroventricular administration. Intracerebroventricular NGF was found to promote survival and regeneration of central cholinergic neurons after axonal transection in rodents.2225 However, clinical studies of NGF have been limited by poor efficacy and frequent adverse events.26

Due to the challenges associated with intracerebroventricular administration, non-invasive approaches have also been investigated, including intranasal2729 and topical ocular administration.30,31 Gene- and cell-based therapies, protein conjugates, and sustained release cerebral implants have also been investigated for CNS NGF distribution, with varying degrees of success.32,33

Peripheral Nervous System

In addition to CNS pathologies, NGF has been investigated as a treatment in peripheral nervous system (PNS) disease. While the blood-brain barrier may hinder NGF delivery to the CNS, access to the PNS is less protected and affords better drug penetration to target neurons. Early studies on rats with streptozotocin-induced diabetes and diabetic peripheral neuropathy showed that subcutaneous injection of rhNGF prevented the reduction in substance P, calcitonin gene-related peptide, and temperature sensitivity typically seen in this model.34,35 While phase I studies of systemic and subcutaneous rhNGF administration found a substantial incidence of injection site hyperalgesia and myalgias,36 subcutaneous rhNGF injections were found to significantly improve subjective and objective sensory measures in a phase II trial of diabetic polyneuropathy.37 However, similar improvements in subjective and objective sensory measures were not replicated in phase III trials of diabetic polyneuropathy,38 nor in phase II trials of HIV-associated peripheral neuropathy.39 Explanations for the discrepancy between these trials include potential confounding by unblinding as a result of prominent local hyperalgesia in the rhNGF injection, differences in rhNGF formulation, and a lower clinical dose than used in preclinical animal models.40

Skin

In addition to promoting cutaneous neuronal regeneration through regulation of complex neuroendocrine pathways, NGF and other neurotrophins exert effects on keratinocytes, fibroblasts, vascular endothelial cells, and immune cells to modulate cutaneous tissue remodeling and wound healing.41 Epithelial cells and keratinocytes adjacent to skin wound edges express higher levels of NGF. In a diabetic mouse model42 as well as various other mouse models of skin wounds,43 topical NGF application has been found to accelerate cutaneous wound healing. Clinically, topical mNGF has been applied to pressure ulcers,44 diabetic foot ulcers,45 and vasculitic ulcers in patients with rheumatoid arthritis46 in non-controlled studies. In a randomized controlled trial, topical mNGF demonstrated efficacy in the treatment of pressure ulcers.47 Despite this early success, larger and more rigorous trials establishing the utility of topical NGF have not yet been performed.

OPHTHALMIC APPLICATIONS

Ocular Surface Disease

Presently, NGF as an ocular therapy has been furthest advanced in the treatment of ocular surface disease. NGF is thought to regulate ocular surface homeostasis in multiple ways, including epithelial health, limbal stem cell proliferation, immune modulation, and tear production.48 TrkANGFR and p75NTR are constitutively expressed in the basal epithelial cells and the stroma of the conjunctiva, as well as the epithelial and endothelial cells of the cornea.4951 In patients with ocular surface diseases, such as vernal keratoconjunctivitis and ocular cicatricial pemphigoid, higher levels of TrkANGFR are found in the conjunctival stroma, along with inflammatory stromal infiltration,49 suggesting a potential role in ocular surface disease. Moreover, increased levels of NGF are seen in the tear film of patients with various ocular surface diseases, including contact lens-related dry eye, Sjögren’s syndrome, ocular cicatricial pemphigoid, and post-refractive dry eye.5255

Given the purported influence of NGF on the ocular surface, NGF has been investigated as a therapy in various ocular surface disease applications. Rabbits with iatrogenic corneal epithelial defects were found to have increased corneal NGF expression, and treatment with topical NGF decreased epithelial defect closure times.50 Dogs with dry eyes, induced by lacrimal gland excision, experienced significant improvement in clinical dry eye parameters – including degree of punctate keratopathy and Schirmer test – and increased conjunctival goblet cell density, when compared to controls.56 In a rabbit model, topical NGF along with docosahexaenoic acid was found to enhance the regrowth of epithelial and subbasal nerve bundles while also promoting wound healing after photorefractive keratectomy.57 Another rabbit study demonstrated significantly improved corneal sensitivity in those rabbits treated with topical NGF after LASIK when compared to vehicle.58

Based on the preponderance of promising preclinical testing, pilot clinical studies were performed in patients with neurotrophic keratopathy (NK) in the late 1990s and early 2000s.59,60 These studies were not controlled, but the safety of topical NGF was demonstrated. Decades passed before another clinical trial was performed, in part due to the relative rarity of NK as a clinical entity. In 2013, a phase I trial established the tolerability of rhNGF in escalating doses up to 180 ug/mL.61 In 2018, a phase I and a subsequent double-masked phase II clinical trial (NGF0212/REPARO phase I/II) reaffirmed the safety of topical rhNGF, demonstrated significantly decreased epithelial defect healing time, and reduced recurrence rate versus control in patients with moderate to severe NK.62,63 Despite improvements in the ocular surface, significant improvements in corneal sensitivity were not seen in this study. Nonetheless, following the overall success of these clinical trials, topical NGF has received approval from the European Medicines Agency and the United States Food and Drug Administration for the treatment of moderate to severe NK. For this use, the medication is approved as a six times per day eye drop, to be used for eight weeks.

A recently published phase II trial was the first to investigate the use of rh-NGF eye drops in dry eye syndrome, and significant improvements were seen in symptoms, ocular surface staining, and tear production.64 Further studies comparing NGF to current standards of care are needed to establish NGF as a treatment for dry eye syndrome.

Retinal Disease

In the retina, NGF is expressed by retinal pigment epithelium, Müller cells, and retinal ganglion cells, while NGF receptors are found on retinal pigment epithelium, photoreceptors, Müller cells, and retinal ganglion cells.49,65,66 Significantly reduced NGF mRNA expression and protein levels were found in the eyes of rats affected by an inherited retinal degeneration causing photoreceptor cell loss (Royal College of Surgeons rats),67 suggesting a potential role for NGF in certain forms of retinopathy. Indeed, in the same rat model, retrobulbar injections of NGF resulted in improved photoreceptor cell survival and increased retinal vascularity compared to controls.68 Similarly, a study performed on C3H mice found that the photoreceptor degeneration typically seen in this mouse model was mitigated by a single time intravitreal injection of NGF or multiple retrobulbar NGF injections.69

NGF has also been studied in diabetic animal models. Streptozotocin-induced diabetic rats are found to have increased expression of retinal NGF and NGF receptor initially, which is thought to have a protective effect.70,71 Over time, however, NGF expression decreases in this diabetic rat model, and this is followed by retinal ganglion cell and Müller cell apoptosis. These rat studies have demonstrated mitigation of retinal ganglion cell and Müller cell loss with topical NGF treatment.

Clinically, NGF has not yet been extensively studied for retinal disease. In one case report, an elderly patient with advanced non-neovascular age-related macular degeneration had improvement in visual acuity and electroretinogram amplitude after one month of topical administration of 200 μg/mL NGF, with further improvement after 3 months of treatment.72 This improvement was not robust, as acuity and ERG parameters worsened upon discontinuation. However, the testing parameters again improved after NGF was restarted. A pilot, non-controlled study has also been performed in patients with retinitis pigmentosa.73 In this study, patients received 10 days of topical NGF, which was generally well tolerated. The results suggested a possible improvement in functional ERG and visual field testing in the study patients. However, larger, controlled, and masked trials have yet to be completed.

Optic Nerve Disease

In the visual pathway, NGF receptors are found in retinal ganglion cells and Muller cells66 and in the visual cortex,74 as well as oligodendrocytes of the optic nerve.75 The possibility of cells in the optic nerve being receptive to exogenous NGF as a therapy has been tested in several experimental and clinical studies. Several early studies confirmed the neuroprotective effect of NGF on neurons in the optic nerve. Intraocular injection of NGF was found to mitigate the loss of myelinated optic nerve fibers after optic nerve transection in a rat study.76 More recently, topical administration of rh-NGF was found to prevent RGC apoptosis, promote axonal survival, and inhibit astrocyte-driven optic nerve degeneration.77 Similarly, intraocular NGF administration significantly reduced functional retinal ganglion cell damage in a rat model of retinal ischemia.78 The neuroprotective effects of NGF were corroborated in subsequent animal studies of glaucoma. In a rat model of glaucoma, achieved by injection of hypertonic saline into the episcleral vein, topical administration of NGF significantly reduced retinal ganglion cell loss.79,80 Similarly, a rabbit model of glaucoma, achieved by intracameral injection of 2% methylcellulose, resulted in increased aqueous humor NGF levels.81 The ensuing retinal and optic nerve damage was reduced in rabbits pretreated with retrobulbar injections of NGF, whereas intraocular injection of an inhibitory anti-NGF antibody resulted in worsened damage.

Limited attempts have been made to translate the animal model studies into clinical trials. In an uncontrolled study, three patients with advanced glaucoma treated with topical NGF experienced improvements in various clinical and electrophysiological parameters, including visual field mean deviation, contrast sensitivity, and pattern electroretinogram conduction delays.80 These effects appeared to persist after discontinuation. An open-label pilot study, and subsequently a double-masked clinical randomized control trial were performed investigating the use of topical NGF for the treatment of vision loss from optic pathway gliomas.82,83 Patients receiving topical NGF were found to have improved electrophysiological testing (e.g. visual evoked potential) and improved visual field testing when compared to controls. Importantly, no significant concurrent tumor growth was noted during this study, and no significant adverse effects were seen.

CHALLENGES OF OCULAR NGF THERAPY

Production

For most of its experimental history, NGF has been used in its murine form (mNGF), derived from the salivary gland of adult male mice as initially described by Bocchini and Angeletti in 196984. NGF is highly evolutionarily conserved, and high sequence homology exists across diverse species.8486 For this reason, mNGF has been used not only in benchtop research but also in human clinical trials, with success. However, the isolation process is time-intensive and technically complex, making adaptation to mass production for clinical use difficult.

Advances in genetic recombination techniques have enabled the development of recombinant human NGF (rhNGF). These recombinant forms maintain the bioactivity of mNGF while allowing scalability of NGF as a therapy for different clinical targets.87,88 Of note, recent biochemical studies of mNGF and rhNGF suggest that significant differences exist between murine and human forms, and that these two molecules should no longer be considered equivalent.89

Drug Development

Given that NK – the principle application of ocular NGF therapy – is rare, drug development has been limited by scientific and financial interest. Development interest in rh-NGF was advanced when the company currently producing rh-NGF in clinical trials (Dompé Farmaceutici S.p.A., Italy) secured orphan drug status from the European Medicine Agency (EMA) in 2013 and the United States Food and Drug Agency (FDA) in 2018. Clinical trials for rh-NGF in the treatment of NK proceeded soon after orphan drug status was granted, and the drug was approved by the EMA and the FDA soon thereafter.

Stability

NGF, and the growth factor family in general, suffers from poor protein stability and a short biological half-life.90 NGF in particular has less conformational stability than the other neurotrophins.91,92 NGF found within autologous serum tears, along with other growth factors, experiences a significant decrease in concentration after one day at room temperature.93 For this reason, drug storage is an important concern for NGF therapeutics. Current recommendations are for autologous serum tears to be prepared in single-day vials, stored frozen, and thawed but kept refrigerated for the day of use.93 The storage and handling instructions of the current FDA approved NGF eye drop, Oxervate™, reflect the logistical complexity required to maintain NGF stability, and are similar to those for autologous serum tears.94

Poor stability also affects the ability of NGF to exert effects on target tissue. rhNGF becomes unstable in plasma at physiologic temperatures after approximately four days.95 Pharmacokinetic studies of rhNGF have found that after subcutaneous injections, the molecule achieves wide distribution, preferentially to highly perfused and neuronal cell types throughout the body.96 Because of extensive protein redistribution as well as enzymatic degradation, even delivering the drug directly to the target tissue may be limited in long-term effect. For example, in a study of an intracranial polymeric rhNGF delivery system, drug concentration in brain tissue decreased exponentially with distance from the device, and tissue half-life was estimated at approximately 1.7 hours.97 NGF in topical eye drops permeates into various ocular tissues, including the cornea, retina, and optic nerve, and even reaches tissue as distant as neurons in the forebrain.30,98 However, peak levels of NGF are seen six hours after administration, necessitating frequent administration to achieve a desired local effect.99 Stability and duration of effect are reflected in the dosage instructions for Oxervate™, which require eye drop administration six times daily.

Drug stability becomes an especially important concern when considering that one of the primary target effects of NGF, neuronal regeneration, is a chronic process. Subbasal nerve fiber regeneration after keratorefractive surgery, for example, occurs over the course of years.100,101 Thus, for maximal benefit of NGF, long-term application may be necessary. The current commercially available formulation, Oxervate™, an eight-week treatment duration is recommended. Whether a second or otherwise extended treatment course is beneficial has yet to be determined.

Efficacy

Questions of efficacy arise from the first clinical trials involving NGF in NK. While preclinical studies suggest efficacy in corneal nerve regrowth after keratorefractive surgery,57 no preclinical studies have been published on an animal model of NK – the drug’s only current indication. In clinical trials, while corneal nerve regrowth was not directly assessed, patients did not experience significant changes in corneal sensitivity.63 In the case of NK, this finding may hamper the drug’s long-term efficacy. Moreover, due to the cost of the current formulation, long-term rh-NGF use may be cost-prohibitive. Still, it is relatively early in rh-NGF development, and long-term data is forthcoming.

Off-target Effects

Given the diversity of cell types responsive to NGF and its complex signaling mechanisms, off-target effects need to be minimized. In phase I studies of intravenous rhNGF in healthy patients, generalized myalgia was a common adverse effect.36 Similarly, in clinical trials of topical rhNGF, ocular pain is the most common adverse effect.63 This adverse effect may be related to the neurotrophin’s involvement in various pain pathways, or related to the sensitivity of regenerated sensory neurons. In fact, anti-NGF antibody therapy has been used to treat inflammatory and neuropathic pain.102

FUTURE DIRECTIONS

Expanded Indications

To date, NGF has been investigated as an ocular therapy in the treatment of ocular surface disease, age-related macular degeneration, retinitis pigmentosa, optic pathway glioma, and advanced glaucoma. NGF therapy in most of these applications is nascent, and further clinical studies are needed to demonstrate clinical benefit. Given the myriad cells that are responsive to NGF – namely, corneal epithelium and endothelium, epithelium of the conjunctiva, lens, iris, and ciliary body, as well as retinal pigment epithelium, Müller cells, and retinal ganglion cells49,,65,,66,,103,,104– NGF has the potential to impact numerous ocular structures. Further study into the impact of exogenous NGF on different ocular cell populations may reveal off-target effects or indeed new clinical targets.

Sustained Drug Delivery

Because neuronal regeneration is a slow process, NGF treatments require long-term application. In most experimental and clinical trials to date, NGF has been given with a saline or similar aqueous vehicle and requires frequent applications for an extended duration. In non-ocular applications, various methods have been employed to deliver a controlled amount of NGF to target tissue over an extended period of time. For example, NGF has been incorporated into microspheres of polymers like poly(L-lactide)co-glycolide105,106 for sustained release to peripheral nerves in a rodent model.107 Alginate microbeads containing NGF along with other growth factors (VEGF, IGF-1, FGF-1, PDGF, and HGF) were successfully used to deliver growth factors for over four weeks in vitro108 for a proposed treatment of stress urinary incontinence. A collagen/hydroxyapatite composite material provided sustained NGF release and was efficacious in promoting bone formation in a rodent model.109

While not yet investigated for ocular NGF, drug delivery of other therapeutics have been studied extensively in the eye.110 Current drug delivery solutions for other ocular therapeutics may be applied to NGF to address issues with NGF stability and target tissue delivery.

Gene Therapy

Some research has focused on circumventing issues with NGF delivery to target tissue by using gene therapy to allow native cells to express the therapeutic molecule. So far, this technology has primarily been investigated in CNS pathology. In one early study, autologous fibroblasts were cultivated and genetically modified to produce NGF. These cells were introduced as grafts into a region of degenerated forebrain in a monkey model, and were shown to significantly reduce degeneration of a subset of neurons in this model.111 More recently, devices have been developed which contain genetically modified cell lines growing within an implantable, encapsulated device, known as encapsulated cell biodelivery. This technology has been implemented in the treatment of Alzheimer’s disease with the NsG0202/NsG0202.1 device – a device containing cells engineered to produce NGF implanted in the basal forebrain.112,113

While ocular applications of gene therapy have so far focused on the treatment of inherited genetic conditions, such as Leber congenital amaurosis114 and retinitis pigmentosa,115 future directions may exploit this technology for the production of therapeutic molecules in native tissue.

Alternate Modalities and Targets

Udonitrectag (Recordati S.p.A.), a low molecular weight compound that mimics NGF, aims to address the issue of NGF stability. Another novel drug, NRO-1 (Neuroptika), targets an alternative neurotrophin pathway – glial cell line-derived neurotrophic factor – rather than NGF, and aims to avoid the concerns of NGF and pain.116 Both agents are currently in phase II clinical trials.

Autologous serum tears and plasma rich in growth factors are two blood-derived therapies containing NGF and other growth factors that are commonly used in various ocular surface diseases.117 The concentration of NGF in serum is orders of magnitude lower than that used in Oxervate™; however, serum tears have demonstrated efficacy in the treatment of NK.118 Rigorous, prospective clinical trials have not yet been completed.

An alternative mechanism has been targeted with substance P and insulin-like growth factor-1, which work in conjunction to promote epithelial wound healing119 and neuronal function.120 This combination therapy has been tested successfully in animals121 and in an open-label clinical trial for stages I and II NK.122 Further research of the clinical efficacy of this treatment has not yet been completed.

CONCLUSIONS

NGF is a bioactive molecule that influences various systems throughout the body via complex molecular pathways. In the eye, NGF exerts beneficial effects on degenerative conditions such as NK and glaucoma. NGF represents the first FDA approved treatment directed at the underlying pathophysiology of NK. Treatment of other ocular conditions may follow the success in NK. Understanding the challenges associated with NGF as an ocular therapy may help hasten the development and more widespread use of this treatment modality.

References

  • 1.Levi-Montalcini R, Booker B. Excessive growth of the sympathetic ganglia evoked by a protein isolated from mouse salivary glands. Proc Natl Acad Sci U S A. 1960;46(3):373–384. doi: 10.1073/pnas.46.3.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Levi-Montalcini R, Angeletti PU. Nerve growth factor. Physiol Rev. 1968;48(3):534–569. doi: 10.1152/physrev.1968.48.3.534. [DOI] [PubMed] [Google Scholar]
  • 3.Bradshaw RA, Murray-Rust J, Ibáñez CF, McDonald NQ, Lapatto R, Blundell TL. Nerve growth factor: structure/function relationships. Protein Sci Publ Protein Soc. 1994;3(11):1901–1913. doi: 10.1002/pro.5560031102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Murphy RA, Saide JD, Blanchard MH, Young M. Molecular properties of the nerve growth factor secreted in mouse saliva. Proc Natl Acad Sci U S A. 1977;74(7):2672–2676. doi: 10.1073/pnas.74.7.2672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kaplan DR, Hempstead BL, Martin-Zanca D, Chao MV, Parada LF. The trk proto-oncogene product: a signal transducing receptor for nerve growth factor. Science. 1991;252(5005):554–558. doi: 10.1126/science.1850549. [DOI] [PubMed] [Google Scholar]
  • 6.Klein R, Jing SQ, Nanduri V, O’Rourke E, Barbacid M. The trk proto-oncogene encodes a receptor for nerve growth factor. Cell. 1991;65(1):189–197. doi: 10.1016/0092-8674(91)90419-y. [DOI] [PubMed] [Google Scholar]
  • 7.Johnson D, Lanahan A, Buck CR, et al. Expression and structure of the human NGF receptor. Cell. 1986;47(4):545–554.doi: 10.1016/0092-8674(86)90619-7. [DOI] [PubMed] [Google Scholar]
  • 8.Segal RA. Selectivity in neurotrophin signaling: theme and variations. Annu Rev Neurosci. 2003;26(1):299–330. doi: 10.1146/annurev.neuro.26.041002.131421. [DOI] [PubMed] [Google Scholar]
  • 9.Beattie EC, Zhou J, Grimes ML, et al. Hypothesis to explain ngf actions: potential implications for neurodegeneration. Cold Spring Harb Symp Quant Biol. 1996;61:389–406. doi: 10.1101/SQB.1996.061.01.042. [DOI] [PubMed] [Google Scholar]
  • 10.Grimes ML, Beattie E, Mobley WC. A signaling organelle containing the nerve growth factor-activated receptor tyrosine kinase, TrkA. Proc Natl Acad Sci. 1997;94(18):9909–9914. doi: 10.1073/pnas.94.18.9909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Grimes ML, Zhou J, Beattie EC, et al. Endocytosis of activated trka: evidence that nerve growth factor induces formation of signaling endosomes. J Neurosci. 1996;16(24):7950–7964.doi: 10.1523/JNEUROSCI.16-24-07950.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang Y, Moheban DB, Conway BR, Bhattacharyya A, Segal RA. Cell surface trk receptors mediate ngf-induced survival while internalized receptors regulate ngf-induced differentiation. J Neurosci. 2000;20(15):5671–5678. doi: 10.1523/JNEUROSCI.20-15-05671.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sofroniew MV, Howe CL, Mobley WC. Nerve growth factor signaling, neuroprotection, and neural repair. Annu Rev Neurosci. 2001;24(1):1217–1281. doi: 10.1146/annurev.neuro.24.1.1217. [DOI] [PubMed] [Google Scholar]
  • 14.Torcia M, Bracci-Laudiero L, Lucibello M, et al. Nerve growth factor is an autocrine survival factor for memory B lymphocytes. Cell. 1996;85(3):345–356.doi: 10.1016/s0092-8674(00)81113-7. [DOI] [PubMed] [Google Scholar]
  • 15.Aloe L, Rocco ML, Omar Balzamino B, Micera A. Nerve growth factor: a focus on neuroscience and therapy. Curr Neuropharmacol. 2015;13(3):294–303. doi: 10.2174/1570159X13666150403231920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Matsuoka I, Meyer M, Thoenen H. Cell-type-specific regulation of nerve growth factor (NGF) synthesis in non-neuronal cells: comparison of Schwann cells with other cell types. J Neurosci. 1991;11(10):3165–3177. doi: 10.1523/JNEUROSCI.11-10-03165.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10(1):239. doi: 10.1186/1479-5876-10-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Appel SH. A unifying hypothesis for the cause of amyotrophic lateral sclerosis, parkinsonism, and alzheimer disease. Ann Neurol. 1981;10(6):499–505. doi: 10.1002/ana.410100602. [DOI] [PubMed] [Google Scholar]
  • 19.Is HF. Alzheimer disease caused by lack of nerve growth factor? Ann Neurol. 1983;13(1):109–110. doi: 10.1002/ana.410130127. [DOI] [PubMed] [Google Scholar]
  • 20.Poduslo JF, Curran GL, Berg CT. Macromolecular permeability across the blood-nerve and blood-brain barriers. Proc Natl Acad Sci. 1994;91(12):5705–5709. doi: 10.1073/pnas.91.12.5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Thorne RG, Frey WH. Delivery of neurotrophic factors to the central nervous system. Clin Pharmacokinet. 2001;40(12):907–946. doi: 10.2165/00003088-200140120-00003. [DOI] [PubMed] [Google Scholar]
  • 22.Hefti F Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. J Neurosci. 1986;6(8):2155–2162. doi: 10.1523/JNEUROSCI.06-08-02155.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Williams LR, Varon S, Peterson GM, et al. Continuous infusion of nerve growth factor prevents basal forebrain neuronal death after fimbria fornix transection. Proc Natl Acad Sci. 1986;83(23):9231–9235.doi: 10.1073/pnas.83.23.9231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kromer LF. Nerve growth factor treatment after brain injury prevents neuronal death. Science. 1987;235(4785):214–216. doi: 10.1126/science.3798108. [DOI] [PubMed] [Google Scholar]
  • 25.Fischer W, Wictorin K, Björklund A, Williams LR, Varon S, Gage FH. Amelioration of cholinergic neuron atrophy and spatial memory impairment in aged rats by nerve growth factor. Nature. 1987;329(6134):65–68. doi: 10.1038/329065a0. [DOI] [PubMed] [Google Scholar]
  • 26.Eriksdotter Jönhagen M, Nordberg A, Amberla K, et al. Intracerebroventricular infusion of nerve growth factor in three patients with Alzheimer’s disease. Dement Geriatr Cogn Disord. 1998;9(5):246–257.doi: 10.1159/000017069. [DOI] [PubMed] [Google Scholar]
  • 27.Capsoni S, Giannotta S, Cattaneo A. Nerve growth factor and galantamine ameliorate early signs of neurodegeneration in anti-nerve growth factor mice. Proc Natl Acad Sci U S A. 2002;99(19):12432–12437. doi: 10.1073/pnas.192442999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.De Rosa R, Garcia AA, Braschi C, et al. Intranasal administration of nerve growth factor (NGF) rescues recognition memory deficits in AD11 anti-NGF transgenic mice. Proc Natl Acad Sci U S A. 2005;102(10):3811–3816.doi: 10.1073/pnas.0500195102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Capsoni S, Marinelli S, Ceci M, et al. Intranasal “painless” human nerve growth factors slows amyloid neurodegeneration and prevents memory deficits in app x ps1 mice. PLoS One. 2012;7(5):e37555.doi: 10.1371/journal.pone.0037555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lambiase A, Pagani L, Di Fausto V, et al. Nerve growth factor eye drop administrated on the ocular surface of rodents affects the nucleus basalis and septum: biochemical and structural evidence. Brain Res. 2007;1127(1):45–51.doi: 10.1016/j.brainres.2006.09.102. [DOI] [PubMed] [Google Scholar]
  • 31.Tirassa P The nerve growth factor administrated as eye drops activates mature and precursor cells in subventricular zone of adult rats. Arch Ital Biol. 2011;149(2):205–213. doi: 10.4449/aib.v149i1.1359. [DOI] [PubMed] [Google Scholar]
  • 32.Faustino C, Rijo P, Reis CP. Nanotechnological strategies for nerve growth factor delivery: therapeutic implications in Alzheimer’s disease. Pharmacol Res. 2017;120:68–87. doi: 10.1016/j.phrs.2017.03.020. [DOI] [PubMed] [Google Scholar]
  • 33.Mitra S, Behbahani H, Eriksdotter M. Innovative therapy for alzheimer’s disease-with focus on biodelivery of NGF. Front Neurosci. 2019;13. doi: 10.3389/fnins.2019.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Apfel SC, Arezzo JC, Brownlee M, Federoff H, Kessler JA. Nerve growth factor administration protects against experimental diabetic sensory neuropathy. Brain Res. 1994;634(1):7–12. doi: 10.1016/0006-8993(94)90252-6. [DOI] [PubMed] [Google Scholar]
  • 35.Fernyhough P, Diemel LT, Hardy J, Brewster WJ, Mohiuddin L, Tomlinson DR. Human recombinant nerve growth factor replaces deficient neurotrophic support in the diabetic rat. Eur J Neurosci. 1995;7(5):1107–1110. doi: 10.1111/j.1460-9568.1995.tb01098.x. [DOI] [PubMed] [Google Scholar]
  • 36.Petty BG, Cornblath DR, Adornato BT, et al. The effect of systemically administered recombinant human nerve growth factor in healthy human subjects. Ann Neurol. 1994;36(2):244–246. doi: 10.1002/ana.410360221. [DOI] [PubMed] [Google Scholar]
  • 37.Apfel SC, Kessler JA, Adornato BT, Litchy WJ, Sanders C, Rask CA. Recombinant human nerve growth factor in the treatment of diabetic polyneuropathy. NGF study group. Neurology. 1998;51(3):695–702. doi: 10.1212/wnl.51.3.695. [DOI] [PubMed] [Google Scholar]
  • 38.Apfel SC, Schwartz S, Adornato BT, et al. Efficacy and safety of recombinant human nerve growth factor in patients with diabetic polyneuropathy: a randomized controlled trial. JAMA. 2000;284(17):2215–2221.doi: 10.1001/jama.284.17.2215. [DOI] [PubMed] [Google Scholar]
  • 39.Schifitto G, Yiannoutsos C, Simpson DM, et al. Long-term treatment with recombinant nerve growth factor for HIV-associated sensory neuropathy. Neurology. 2001;57(7):1313–1316.doi: 10.1212/wnl.57.7.1313. [DOI] [PubMed] [Google Scholar]
  • 40.Apfel SC. Nerve growth factor for the treatment of diabetic neuropathy: what went wrong, what went right, and what does the future hold? In: Elsevier, editors. International Review of Neurobiology. Vol. 50. Academic Press: Neurobiology of Diabetic Neuropathy. 2002: 393–413. doi: 10.1016/S0074-7742(02)50083-0. [DOI] [PubMed] [Google Scholar]
  • 41.Peters EMJ, Raap U, Welker P, et al. Neurotrophins act as neuroendocrine regulators of skin homeostasis in health and disease. Horm Metab Res Horm Stoffwechselforschung Horm Metab. 2007;39(2):110–124.doi: 10.1055/s-2007-961812. [DOI] [PubMed] [Google Scholar]
  • 42.Matsuda H, Koyama H, Sato H, et al. Role of nerve growth factor in cutaneous wound healing: accelerating effects in normal and healing-impaired diabetic mice. J Exp Med. 1998;187(3):297–306. doi: 10.1084/jem.187.3.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li AK, Koroly MJ, Schattenkerk ME, Malt RA, Young M. Nerve growth factor: acceleration of the rate of wound healing in mice. Proc Natl Acad Sci U S A. 1980;77(7):4379–4381. doi: 10.1073/pnas.77.7.4379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bernabei R, Landi F, Bonini S, et al. Effect of topical application of nerve-growth factor on pressure ulcers. Lancet. 1999;354(9175):307.doi: 10.1016/S0140-6736(99)02784-1. [DOI] [PubMed] [Google Scholar]
  • 45.Generini S, Tuveri MA, Matucci Cerinic M, Mastinu F, Manni L, Aloe L. Topical application of nerve growth factor in human diabetic foot ulcers. A study of three cases. Exp Clin Endocrinol Diabetes Off J Ger Soc Endocrinol Ger Diabetes Assoc. 2004;112(9):542–544. doi: 10.1055/s-2004-821313. [DOI] [PubMed] [Google Scholar]
  • 46.Tuveri M, Generini S, Matucci-Cerinic M, Aloe L. NGF, a useful tool in the treatment of chronic vasculitic ulcers in rheumatoid arthritis. Lancet. 2000;356(9243):1739–1740. doi: 10.1016/S0140-6736(00)03212-8. [DOI] [PubMed] [Google Scholar]
  • 47.Landi F, Aloe L, Russo A, et al. Topical treatment of pressure ulcers with nerve growth factor. Ann Intern Med. 2003;139(8):635–641. doi: 10.7326/0003-4819-139-8-200310210-00006. [DOI] [PubMed] [Google Scholar]
  • 48.Lambiase A, Sacchetti M, Bonini S. Nerve growth factor therapy for corneal disease. Curr Opin Ophthalmol. 2012;23(4):296–302. doi: 10.1097/ICU.0b013e3283543b61. [DOI] [PubMed] [Google Scholar]
  • 49.Lambiase A, Bonini S, Micera A, Rama P, Bonini S, Aloe L. Expression of nerve growth factor receptors on the ocular surface in healthy subjects and during manifestation of inflammatory diseases. Invest Ophthalmol Vis Sci. 1998;39:1272–1275. [PubMed] [Google Scholar]
  • 50.Lambiase A, Manni L, Bonini S, Rama P, Micera A, Aloe L. Nerve growth factor promotes corneal healing: structural, biochemical, and molecular analyses of rat and human corneas. Invest Ophthalmol Vis Sci. 2000;41:1063–1069. [PubMed] [Google Scholar]
  • 51.Micera A, Lambiase A, Aloe L, Bonini S, Levi-Schaffer F, Bonini S. Nerve growth factor involvement in the visual system: implications in allergic and neurodegenerative diseases. Cytokine Growth Factor Rev. 2004;15(6):411–417. doi: 10.1016/j.cytogfr.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 52.Liu Q, McDermott AM, Miller WL. Elevated nerve growth factor in dry eye associated with established contact lens wear. Eye Contact Lens. 2009;35(5):232–237. doi: 10.1097/ICL.0b013e3181b3e87f. [DOI] [PubMed] [Google Scholar]
  • 53.Lambiase A, Micera A, Sacchetti M, Cortes M, Mantelli F, Bonini S. Alterations of tear neuromediators in dry eye disease. Arch Ophthalmol. 2011;129(8):981–986. doi: 10.1001/archophthalmol.2011.200. [DOI] [PubMed] [Google Scholar]
  • 54.Lee HK, Lee KS, Kim HC, Lee SH, Kim EK. Nerve growth factor concentration and implications in photorefractive keratectomy vs laser in situ keratomileusis. Am J Ophthalmol. 2005;139(6):965–971. doi: 10.1016/j.ajo.2004.12.051. [DOI] [PubMed] [Google Scholar]
  • 55.Lee HK, Ryu IH, Seo KY, Hong S, Kim HC, Kim EK. Topical 0.1% prednisolone lowers nerve growth factor expression in keratoconjunctivitis sicca patients. Ophthalmology. 2006;113(2):198–205. doi: 10.1016/j.ophtha.2005.09.033. [DOI] [PubMed] [Google Scholar]
  • 56.Coassin M, Lambiase A, Costa N, et al. Efficacy of topical nerve growth factor treatment in dogs affected by dry eye. Graefes Arch Clin Exp Ophthalmol Albrecht Von Graefes Arch Klin Exp Ophthalmol. 2005;243(2):151–155.doi: 10.1007/s00417-004-0955-2. [DOI] [PubMed] [Google Scholar]
  • 57.Esquenazi S, Bazan HEP, Bui V, He J, Kim DB, Bazan NG. Topical Combination of NGF and DHA increases rabbit corneal nerve regeneration after photorefractive keratectomy. Invest Ophthalmol Vis Sci. 2005;46(9):3121–3127. doi: 10.1167/iovs.05-0241. [DOI] [PubMed] [Google Scholar]
  • 58.Joo M-J, Yuhan KR, Hyon JY, et al. The effect of nerve growth factor on corneal sensitivity after laserin situ keratomileusis. Arch Ophthalmol. 2004;122(9):1338–1341.doi: 10.1001/archopht.122.9.1338. [DOI] [PubMed] [Google Scholar]
  • 59.Lambiase A, Rama P, Bonini S, Caprioglio G, Aloe L. Topical treatment with nerve growth factor for corneal neurotrophic ulcers. N Engl J Med. 1998;338(17):1174–1180. doi: 10.1056/NEJM199804233381702. [DOI] [PubMed] [Google Scholar]
  • 60.Bonini S, Lambiase A, Rama P, Caprioglio G, Aloe L. Topical treatment with nerve growth factor for neurotrophic keratitis. Ophthalmology. 2000;107(7):1347–1351. doi: 10.1016/S0161-6420(00)00163-9. [DOI] [PubMed] [Google Scholar]
  • 61.Ferrari MP, Mantelli F, Sacchetti M, et al. Safety and pharmacokinetics of escalating doses of human recombinant nerve growth factor eye drops in a double-masked, randomized clinical trial. Biodrugs. 2014;28(3):275–283.doi: 10.1007/s40259-013-0079-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bonini S, Lambiase A, Rama P, et al. Phase I trial of recombinant human nerve growth factor for neurotrophic keratitis. Ophthalmology. 2018;125(9):1468–1471.doi: 10.1016/j.ophtha.2018.03.004. [DOI] [PubMed] [Google Scholar]
  • 63.Bonini S, Lambiase A, Rama P, et al. Phase II randomized, double-masked, vehicle-controlled trial of recombinant human nerve growth factor for neurotrophic keratitis. Ophthalmology. 2018;125(9):1332–1343.doi: 10.1016/j.ophtha.2018.02.022. [DOI] [PubMed] [Google Scholar]
  • 64.Sacchetti M, Lambiase A, Schmidl D, et al. Effect of recombinant human nerve growth factor eye drops in patients with dry eye: a phase IIa, open label, multiple-dose study. Br J Ophthalmol. 2020;104(1):127–135.doi: 10.1136/bjophthalmol-2018-312470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chakrabarti S, Sima AAF, Lee J, Brachet P, Dicou E. Nerve growth factor (NGF), proNGF and NGF receptor-like immunoreactivity in BB rat retina. Brain Res. 1990;523(1):11–15. doi: 10.1016/0006-8993(90)91630-Y. [DOI] [PubMed] [Google Scholar]
  • 66.Carmignoto G, Comelli MC, Candeo P, et al. Expression of NGF receptor and NGF receptor mRNA in the developing and adult rat retina. Exp Neurol. 1991;111(3):302–311.doi: 10.1016/0014-4886(91)90097-V. [DOI] [PubMed] [Google Scholar]
  • 67.Amendola T, Aloe L. Developmental expression of nerve growth factor in the eye of rats affected by inherited retinopathy: correlative aspects with retinal structural degeneration. Arch Ital Biol. 2002;140(2):81–90. doi: 10.4449/aib.v140i2.458. [DOI] [PubMed] [Google Scholar]
  • 68.Lenzi L, Coassin M, Lambiase A, Bonini S, Amendola T, Aloe L. Effect of exogenous administration of nerve growth factor in the retina of rats with inherited retinitis pigmentosa. Vision Res. 2005;45(12):1491–1500. doi: 10.1016/j.visres.2004.12.020. [DOI] [PubMed] [Google Scholar]
  • 69.Lambiase A, Aloe L. Nerve growth factor delays retinal degeneration in C3H mice. Graefes Arch Clin Exp Ophthalmol. 1996;234(1):S96–S100. doi: 10.1007/BF02343055. [DOI] [PubMed] [Google Scholar]
  • 70.Hammes HP, Federoff HJ, Brownlee M. Nerve growth factor prevents both neuroretinal programmed cell death and capillary pathology in experimental diabetes. Mol Med. 1995;1(5):527–534. doi: 10.1007/BF03401589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mantelli DF, Lambiase A, Colafrancesco V, et al. Effects on retinal ganglion cell fate: new evidence from an animal model of diabetes. Eur J Ophthalmol Published Online May. 2013;9. doi: 10.5301/ejo.5000359. [DOI] [PubMed] [Google Scholar]
  • 72.Lambiase A, Coassin M, Tirassa P, Mantelli F, Aloe L. Nerve growth factor eye drops improve visual acuity and electrofunctional activity in age-related macular degeneration: a case report. Ann Ist Super Sanita. 2009;45(4):439–442. doi: 10.1590/s0021-25712009000400014. [DOI] [PubMed] [Google Scholar]
  • 73.Falsini B, Iarossi G, Chiaretti A, et al. NGF eye-drops topical administration in patients with retinitis pigmentosa, a pilot study. J Transl Med. 2016;14;229(3):746–753.doi: 10.1186/s12967-015-0750-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rossi FM, Sala R, Maffei L. Expression of the nerve growth factor receptors trka and p75ntr in the visual cortex of the rat: development and regulation by the cholinergic input. J Neurosci. 2002;22(3):912–919. doi: 10.1523/JNEUROSCI.22-03-00912.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cohen RI, Marmur R, Norton WT, Mehler MF, Kessler JA. Nerve growth factor and neurotrophin-3 differentially regulate the proliferation and survival of developing rat brain oligodendrocytes. J Neurosci. 1996;16(20):6433–6442. doi: 10.1523/JNEUROSCI.16-20-06433.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Carmignoto G, Maffei L, Candeo P, Canella R, Comelli C. Effect of NGF on the survival of rat retinal ganglion cells following optic nerve section. J Neurosci Off J Soc Neurosci. 1989;9(4):1263–1272. doi: 10.1523/JNEUROSCI.09-04-01263.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Guo L, Davis BM, Ravindran N, et al. Topical recombinant human Nerve growth factor (rh-NGF) is neuroprotective to retinal ganglion cells by targeting secondary degeneration. Sci Rep. 2020;10(1):3375.doi: 10.1038/s41598-020-60427-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Siliprandi R, Canella R, Carmignoto G. Nerve growth factor promotes functional recovery of retinal ganglion cells after ischemia. Invest Ophthalmol Vis Sci. 1993;34:3232–3245. [PubMed] [Google Scholar]
  • 79.Colafrancesco V, Parisi V, Sposato V, et al. Ocular application of nerve growth factor protects degenerating retinal ganglion cells in a rat model of glaucoma. J Glaucoma. 2011;20(2):100–108. doi: 10.1097/IJG.0b013e3181d787e5. [DOI] [PubMed] [Google Scholar]
  • 80.Lambiase A, Aloe L, Centofanti M, et al. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: implications for glaucoma. Proc Natl Acad Sci U S A. 2009;106(32):13469–13474.doi: 10.1073/pnas.0906678106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lambiase A, Centofanti M, Micera A, et al. Nerve growth factor (NGF) reduces and NGF antibody exacerbates retinal damage induced in rabbit by experimental ocular hypertension. Graefes Arch Clin Exp Ophthalmol Albrecht Von Graefes Arch Klin Exp Ophthalmol. 1997;235(12):780–785.doi: 10.1007/BF02332863. [DOI] [PubMed] [Google Scholar]
  • 82.Falsini B, Chiaretti A, Barone G, et al. Topical nerve growth factor as a visual rescue strategy in pediatric optic gliomas: a pilot study including electrophysiology. Neurorehabil Neural Repair. 2011;25(6):512–520.doi: 10.1177/1545968310397201. [DOI] [PubMed] [Google Scholar]
  • 83.Falsini B, Chiaretti A, Rizzo D, et al. Nerve growth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. Brain J Neurol. 2016;139(Pt 2):404–414. doi: 10.1093/brain/awv366. [DOI] [PubMed] [Google Scholar]
  • 84.Bocchini V, Angeletti PU. The nerve growth factor: purification as a 30,000-molecular-weight protein. Proc Natl Acad Sci U S A. 1969;64(2):787–794. doi: 10.1073/pnas.64.2.787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Schwarz MA, Fisher D, Bradshaw RA, Isackson PJ. Isolation and sequence of a cDNA clone of beta-nerve growth factor from the guinea pig prostate gland. J Neurochem. 1989;52(4):1203–1209. doi: 10.1111/j.1471-4159.1989.tb01867.x. [DOI] [PubMed] [Google Scholar]
  • 86.Suter U, Heymach JV, Shooter EM. Two conserved domains in the NGF propeptide are necessary and sufficient for the biosynthesis of correctly processed and biologically active NGF. Embo J. 1991;10(9):2395–2400. doi: 10.1002/j.1460-2075.1991.tb07778.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Colangelo AM, Finotti N, Ceriani M, et al. Recombinant human nerve growth factor with a marked activity in vitro and in vivo. Proc Natl Acad Sci. 2005;102(51):18658–18663.doi: 10.1073/pnas.0508734102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Burton LE, Schmelzer CH, Szonyi E, Yedinak C, Gorrell GA. Activity and biospecificity of proteolyzed forms and dimeric combinations of recombinant human and murine nerve growth factor. J Neurochem. 1992;59(5):1937–1945. doi: 10.1111/j.1471-4159.1992.tb11030.x. [DOI] [PubMed] [Google Scholar]
  • 89.Paoletti F, Malerba F, Bruni Ercole B, Lamba D, Cattaneo A. A comparative analysis of the structural, functional and biological differences between mouse and human nerve growth factor. Biochim Biophys Acta BBA - Proteins Proteomics. 2015;1854(3):187–197. doi: 10.1016/j.bbapap.2014.12.005. [DOI] [PubMed] [Google Scholar]
  • 90.Mitchell AC, Briquez PS, Hubbell JA, Cochran JR. Engineering growth factors for regenerative medicine applications. Acta Biomater. 2016;30:1–12. doi: 10.1016/j.actbio.2015.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Timm D, de Haseth P, Neet K. Comparative equilibrium denaturation studies of the neurotrophins: nerve growth factor, brain-derived neurotrophic factor, neurotrophin 3, and neurotrophin 4/5. Biochemistry. 1994;33(15):4667–4676. doi: 10.1021/bi00181a602. [DOI] [PubMed] [Google Scholar]
  • 92.Bothwell MA, Shooter EM. Dissociation equilibrium constant of beta nerve growth factor. J Biol Chem. 1977;252(23):8532–8536. doi: 10.1016/S0021-9258(19)75252-1. [DOI] [PubMed] [Google Scholar]
  • 93.Bradley JC, Simoni J, Bradley RH, McCartney DL, Brown SM. Time- and temperature-dependent stability of growth factor peptides in human autologous serum eye drops. Cornea. 2009;28(2):200–205. doi: 10.1097/ICO.0b013e318186321e. [DOI] [PubMed] [Google Scholar]
  • 94.Dompé Farmaceutici S.p.A. OXERVATE CENEGERMIN-BKBJ [Package Insert].; 2018.
  • 95.Nguyen CB, É S, Sadick MD, Hotaling TE, Mendoza-Ramirez J-LEE. Stability and interactions of recombinant human nerve growth factor in different biological matrices: in vitro and in vivo studies. Drug Metab Dispos. 2000;28:590–597. [PubMed] [Google Scholar]
  • 96.Nguyen CB, Harris L, É S, et al. Tissue disposition and pharmacokinetics of recombinant human nerve growth factor after acute and chronic subcutaneous administration in monkeys. Drug Metab Dispos. 2000;28(5):598–607. [PubMed] [Google Scholar]
  • 97.Saltzman WM, Mak MW, Mahoney MJ, Duenas ET, Cleland JL. Intracranial delivery of recombinant nerve growth factor: release kinetics and protein distribution for three delivery systems. Pharm Res. 1999;16(2):232–240. doi: 10.1023/a:1018824324275. [DOI] [PubMed] [Google Scholar]
  • 98.Fausto VD, Fiore M, Tirassa P, Lambiase A, Aloe L. Eye drop NGF administration promotes the recovery of chemically injured cholinergic neurons of adult mouse forebrain. Eur J Neurosci. 2007;26(9):2473–2480. doi: 10.1111/j.1460-9568.2007.05883.x. [DOI] [PubMed] [Google Scholar]
  • 99.Lambiase A, Tirassa P, Micera A, Aloe L, Bonini S. Pharmacokinetics of conjunctivally applied nerve growth factor in the retina and optic nerve of adult rats. Invest Ophthalmol Vis Sci. 2005;46(10):3800–3806. doi: 10.1167/iovs.05-0301. [DOI] [PubMed] [Google Scholar]
  • 100.Moilanen JAO, Holopainen JM, Vesaluoma MH, Tervo TMT. Corneal recovery after lasik for high myopia: a 2-year prospective confocal microscopic study. Br J Ophthalmol. 2008;92(10):1397–1402. doi: 10.1136/bjo.2007.126821. [DOI] [PubMed] [Google Scholar]
  • 101.Calvillo MP, McLaren JW, Hodge DO, Bourne WM. Corneal Reinnervation after LASIK: prospective 3-Year Longitudinal Study. Invest Ophthalmol Vis Sci. 2004;45(11):3991–3996. doi: 10.1167/iovs.04-0561. [DOI] [PubMed] [Google Scholar]
  • 102.Patel MK, Kaye AD, Urman RD. Tanezumab: therapy targeting nerve growth factor in pain pathogenesis. J Anaesthesiol Clin Pharmacol. 2018;34(1):111–116. doi: 10.4103/joacp.JOACP_389_15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lambiase A, Bonini S, Manni L, et al. Intraocular production and release of nerve growth factor after iridectomy. Invest Ophthalmol Vis Sci. 2002;43(7):2334–2340. [PubMed] [Google Scholar]
  • 104.Ghinelli E, Aloe L, Cortes M, Micera A, Lambiase A, Bonini S. Nerve growth factor (NGF) and lenses: effects of NGF in an in vitro rat model of cataract. Graefes Arch Clin Exp Ophthalmol. 2003;241(10):845–851. doi: 10.1007/s00417-003-0733-6. [DOI] [PubMed] [Google Scholar]
  • 105.Camarata PJ, Suryanarayanan R, Turner DA, Parker RG, Ebner TJ. Sustained release of nerve growth factor from biodegradable polymer microspheres. Neurosurgery. 1992;30(3):313–319. doi: 10.1227/00006123-199203000-00001. [DOI] [PubMed] [Google Scholar]
  • 106.Gu H, Song C, Long D, Mei L, Sun H. Controlled release of recombinant human nerve growth factor (rhNGF) from poly[(lactic acid)-co-(glycolic acid)] microspheres for the treatment of neurodegenerative disorders. Polym Int. 2007;56(10):1272–1280. doi: 10.1002/pi.2272. [DOI] [Google Scholar]
  • 107.Wang Z, Han N, Wang J, et al. Improved peripheral nerve regeneration with sustained release nerve growth factor microspheres in small gap tubulization. Am J Transl Res. 2014;6(4):413–421. [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu G, Pareta RA, Wu R, et al. Skeletal myogenic differentiation of urine-derived stem cells and angiogenesis using microbeads loaded with growth factors. Biomaterials. 2013;34(4):1311–1326. doi: 10.1016/j.biomaterials.2012.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Letic-Gavrilovic A, Piattelli A, Abe K. Nerve growth factor beta (NGF beta) delivery via a collagen/hydroxyapatite (Col/HAp) composite and its effects on new bone ingrowth. J Mater Sci Mater Med. 2003;14(2):95–102. doi: 10.1023/a:1022099208535. [DOI] [PubMed] [Google Scholar]
  • 110.Gote V, Sikder S, Sicotte J, Pal D. Ocular drug delivery: present innovations and future challenges. J Pharmacol Exp Ther. 2019;370(3):602–624. doi: 10.1124/jpet.119.256933. [DOI] [PubMed] [Google Scholar]
  • 111.Tuszynski MH, Roberts J, Senut MC, U Hs GFH Gene therapy in the adult primate brain: intraparenchymal grafts of cells genetically modified to produce nerve growth factor prevent cholinergic neuronal degeneration. Gene Ther. 1996;3:305–314. [PubMed] [Google Scholar]
  • 112.Wahlberg LU, Lind G, Almqvist PM, et al. Targeted delivery of nerve growth factor via encapsulated cell biodelivery in Alzheimer disease: a technology platform for restorative neurosurgery: clinical article. J Neurosurg. 2012;117(2):340–347.doi: 10.3171/2012.2.JNS11714. [DOI] [PubMed] [Google Scholar]
  • 113.Eyjolfsdottir H, Eriksdotter M, Linderoth B, et al. Targeted delivery of nerve growth factor to the cholinergic basal forebrain of Alzheimer’s disease patients: application of a second-generation encapsulated cell biodelivery device. Alzheimers Res Ther. 2016;8. 8(1). 10.1186/s13195-016-0195-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ruan G-X, Barry E, Yu D, Lukason M, Cheng SH, Scaria A. CRISPR/Cas9-mediated genome editing as a therapeutic approach for Leber congenital amaurosis 10. Mol Ther. 2017;25(2):331–341. doi: 10.1016/j.ymthe.2016.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bakondi B, Lv W, Lu B, et al. In vivo crispr/cas9 gene editing corrects retinal dystrophy in the s334ter-3 rat model of autosomal dominant retinitis pigmentosa. Mol Ther. 2016;24(3):556–563. doi: 10.1038/mt.2015.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Senju Pharmaceutical Co., Ltd. A randomized, double-masked, single-center, placebo-controlled single and multiple ascending dose study to assess safety, tolerability, pharmacokinetics, and efficacy of sjp-0132 in subjects with dry eye disease. clinicaltrials.gov; 2020. Accessed July 18, 2020. https://clinicaltrials.gov/ct2/show/NCT04139122
  • 117.Geerling G, Maclennan S, Hartwig D. Autologous serum eye drops for ocular surface disorders. Br J Ophthalmol. 2004;88(11):1467–1474. doi: 10.1136/bjo.2004.044347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Matsumoto Y, Dogru M, Goto E, et al. Autologous serum application in the treatment of neurotrophic keratopathy. Ophthalmology. 2004;111(6):1115–1120.doi: 10.1016/j.ophtha.2003.10.019. [DOI] [PubMed] [Google Scholar]
  • 119.Nishida T, Nakamura M, Ofuji K, Reid TW, Mannis MJ, Murphy CJ. Synergistic effects of substance P with insulin-like growth factor-1 on epithelial migration of the cornea. J Cell Physiol. 1996;169(1):159–166. doi:. [DOI] [PubMed] [Google Scholar]
  • 120.Barbariga M, Rabiolo A, Fonteyne P, Bignami F, Rama P, Ferrari G. The effect of aging on nerve morphology and substance p expression in mouse and human corneas. Invest Ophthalmol Vis Sci. 2018;59(13):5329–5335. doi: 10.1167/iovs.18-24707. [DOI] [PubMed] [Google Scholar]
  • 121.Ghiasi Z, Gray T, Tran P, et al. The effect of topical substance-p plus insulin-like growth factor-1 (igf-1) on epithelial healing after photorefractive keratectomy in rabbits. Transl Vis Sci Technol. 2018;7(1):1.doi: 10.1167/tvst.7.1.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yamada N, Matsuda R, Morishige N, et al. Open clinical study of eye-drops containing tetrapeptides derived from substance P and insulin-like growth factor-1 for treatment of persistent corneal epithelial defects associated with neurotrophic keratopathy. Br J Ophthalmol. 2008;92(7):896–900.doi: 10.1136/bjo.2007.130013. [DOI] [PubMed] [Google Scholar]

RESOURCES