MedPath

Cenegermin Advanced Drug Monograph

Published:Sep 30, 2025

Generic Name

Cenegermin

Brand Names

Oxervate

Drug Type

Biotech

CAS Number

1772578-74-1

Associated Conditions

Neurotrophic Keratopathy

Report on Cenegermin (Oxervate®): A First-in-Class Recombinant Nerve Growth Factor for Neurotrophic Keratitis

Executive Summary

Cenegermin, marketed under the brand name Oxervate®, represents a paradigm shift in the management of neurotrophic keratitis (NK), a rare and degenerative corneal disease. It is a first-in-class, topical biologic therapy, distinguished as the first-ever recombinant human nerve growth factor (rhNGF) approved for ophthalmic use.[1] The therapeutic innovation of Cenegermin lies in its disease-modifying mechanism of action, which directly addresses the underlying pathophysiology of NK. By mimicking the function of endogenous nerve growth factor, it promotes the repair of damaged corneal nerves and stimulates the healing of the corneal epithelium, thereby restoring the trophic support that is lost in this condition.[3]

Pivotal, multicenter, randomized controlled trials have demonstrated the robust efficacy of Cenegermin in achieving complete corneal healing in a majority of patients with moderate to severe NK, a significant improvement over vehicle control.[6] The safety profile is well-characterized and considered manageable, with the most common adverse events being local to the eye, such as transient ocular pain, which may be indicative of the desired nerve regeneration process. The regulatory journey of Cenegermin was marked by multiple expedited pathway designations from global health authorities, including Orphan Drug, Breakthrough Therapy, Fast Track, and Priority Review status, underscoring the profound unmet medical need it addresses.[6]

Despite its clinical benefits, the practical application of Cenegermin is associated with a significant treatment burden. The therapy requires a complex and demanding administration protocol, including strict cold-chain storage, meticulous daily preparation, and a frequent dosing schedule of six times per day for eight weeks.[9] This complexity necessitates comprehensive patient education to ensure adherence and optimal outcomes. Beyond its approved indication, Cenegermin is also under investigation for its therapeutic potential in other ocular surface diseases, such as dry eye syndrome, signaling a promising future for neurotrophic-based therapies in ophthalmology.[6]

Molecular Profile and Bio-manufacturing

Biochemical Identity of Cenegermin as a Recombinant Human Nerve Growth Factor (rhNGF)

Cenegermin is the international nonproprietary name (INN) for a highly purified, recombinant form of human beta-nerve growth factor (β-NGF).[1] In the United States, it is formally designated with the nonproprietary name cenegermin-bkbj.[1] As a biotech drug, it is engineered to be structurally and functionally identical to the endogenous human NGF, a neurotrophin that plays a fundamental role in the growth, differentiation, maintenance, and survival of neurons.[6] Pharmacologically, it is classified as a protein-based therapy, a neuroprotectant, and a nerve growth factor receptor agonist, reflecting its role in stimulating specific cellular pathways to promote tissue repair.[1]

Molecular Structure, Amino Acid Sequence, and Physicochemical Properties

The biologically active form of Cenegermin is a non-covalent homodimer, composed of two identical polypeptide chains.[1] Each monomeric chain consists of 118 amino acids and has a relative molecular mass of approximately 13,267 Daltons (g/mol).[1] The empirical chemical formula for the monomer is

.[1]

The primary structure, or amino acid sequence, of the Cenegermin monomer is as follows [6]:

SSSHPIFHRGEFSVCDSVSVWVGDKTTATDIKGKEVMVLGEVNINNSVFKQYFFETKCRDPNPVDSGCRGIDSKHWNSYCTTTHTFVKALTMDGKQAAWRFIRIDTACVCVLSRKAVR

The tertiary structure of each monomer is stabilized by three intramolecular disulfide bridges. These covalent bonds form a highly conserved and stable three-dimensional conformation known as a "cysteine knot".[6] This intricate structure is essential for the molecule's biological activity, ensuring proper receptor binding and signal transduction. The key physicochemical and identifying properties of Cenegermin are summarized in Table 1.

Table 1: Physicochemical and Molecular Properties of Cenegermin

ParameterValueSource(s)
International Nonproprietary Name (INN)Cenegermin1
US Adopted Name (USAN)Cenegermin-bkbj1
DrugBank IDDB139261
CAS Number1772578-74-11
UNIIB6E7K36KT81
ATC CodeS01XA241
Molecular Formula (Monomer)1
Molar Mass (Monomer)Approx. 13267 g/mol1
Amino Acid Count (Monomer)1186
StructureNon-covalent homodimer with cysteine knot motif1

Production via Recombinant DNA Technology in Escherichia coli

Cenegermin is manufactured through a sophisticated bio-engineering process utilizing recombinant DNA technology.[1] The process begins with the gene encoding human NGF, which is isolated and then spliced into the genome of a laboratory strain of the bacterium

Escherichia coli, specifically strain HMS174.[6] This genetically modified bacterium serves as a biological factory for the protein.

The bacteria are then cultivated in large-scale, controlled bioreactors, a process known as fermentation. During this growth phase, the bacterial cellular machinery transcribes and translates the inserted human gene, producing large quantities of the protein.[20] The protein is initially synthesized as an inactive precursor, or pro-peptide (ProNGF).[2] This is a common outcome when expressing complex human proteins in a simpler prokaryotic system like

E. coli.

Following fermentation, the bacterial cells are harvested, and the ProNGF is extracted and subjected to a rigorous multi-step purification process. A critical final step involves enzymatic hydrolysis, where a specific enzyme is used to cleave the pro-sequence from the protein, yielding the mature, 118-amino acid rhNGF molecule.[2] This ensures that the final active pharmaceutical ingredient is structurally and functionally identical to naturally secreted human NGF.[6]

The selection of an E. coli expression system is a pivotal manufacturing decision with significant downstream consequences. While this prokaryotic system is highly efficient and cost-effective for achieving high yields of recombinant protein, it lacks the complex cellular machinery for post-translational modifications that are native to human cells. Consequently, producing a biologically active, correctly folded human protein like NGF requires a sophisticated and technically demanding downstream process, including protein refolding and precise enzymatic cleavage. This manufacturing complexity, described by the developer as a "revolutionary method," represents a major scientific achievement.[8] However, it is also a primary driver of the therapy's substantial cost and establishes a formidable technological barrier to the future development of potential biosimilars.

Pharmacological Profile

Mechanism of Action: Restoring Corneal Homeostasis via Tropomyosin Receptor Kinase A (TrkA) Agonism

Cenegermin exerts its therapeutic effects by functioning as a peripherally selective agonist for two distinct classes of nerve growth factor receptors: the high-affinity tropomyosin receptor kinase A (TrkA) and the low-affinity p75 neurotrophin receptor (p75NTR).[1] These receptors are abundantly expressed on the surfaces of various cells within the anterior segment of the eye, including the corneal epithelium and nerves, the conjunctiva, the iris, the ciliary body, and the lacrimal gland.[16]

By binding to and activating these receptors, Cenegermin precisely mimics the biological activity of endogenous NGF. This binding initiates a cascade of intracellular signaling pathways that are fundamental for the differentiation, maintenance, and survival of neurons, as well as the proliferation and integrity of epithelial cells.[5] This mechanism directly targets and counteracts the central pathophysiology of neurotrophic keratitis. NK is characterized by impaired trigeminal nerve function, which results in a loss of the critical trophic support that nerves provide to the cornea.[2] By replenishing this missing growth factor, Cenegermin helps to restore the normal physiological processes that maintain a healthy ocular surface.

Pharmacodynamics: Trophic Effects on Corneal Innervation, Epithelial Integrity, and Lacrimation

The primary pharmacodynamic effect of Cenegermin is the promotion of corneal healing through multiple, synergistic actions. It acts directly on corneal epithelial cells to stimulate their growth, differentiation, and survival, which is essential for repairing persistent epithelial defects.[2] Furthermore, it supports the regenerative potential of limbal epithelial stem cells, the source of new corneal epithelial cells, and has been experimentally shown to support the recovery of corneal nerve fiber density, a process known as re-innervation.[2]

In addition to its effects on the cornea, Cenegermin also binds to receptors located on the lacrimal glands. This interaction stimulates tear production, which provides lubrication and delivers essential protective factors to the ocular surface, further contributing to a healthy environment conducive to healing.[2] The cumulative result of these pharmacodynamic actions is the restoration of corneal integrity, the normalization of healing processes, and the re-establishment of ocular surface homeostasis.[14]

Pharmacokinetics of Ophthalmic Administration: A Locally Acting Biologic with Negligible Systemic Exposure

Absorption and Distribution

Following topical administration as an eye drop, Cenegermin acts primarily at the site of application. It is absorbed locally into the tissues of the anterior eye, mainly the conjunctiva and peri-orbital tissues, with only minor penetration through the cornea itself.[16] Consequently, its distribution is largely confined to the anterior portion of the eye.[16] A defining feature of its pharmacokinetic profile is its negligible systemic absorption. Clinical studies have consistently shown that after ophthalmic administration, plasma concentrations of Cenegermin do not rise above the natural baseline levels of endogenous NGF, and there is no evidence of systemic accumulation with repeated dosing.[16]

Metabolism and Elimination

As a protein, Cenegermin is metabolized through standard proteolytic pathways. It is expected to be broken down locally within the ocular tissues into smaller peptides and constituent amino acids by endogenous proteases.[16] The majority of the administered dose is cleared from the ocular surface through the natural process of tear secretion and drainage via the nasolacrimal duct. The portion that enters the nasolacrimal duct eventually reaches the nasal and oropharyngeal cavities, where it is further degraded by proteases.[16]

Special Populations

The large molecular size of Cenegermin (approximately 13.3 kDa for the monomer) combined with its extremely limited systemic absorption from the eye makes it highly unlikely that clinically relevant amounts would be excreted into breast milk. Therefore, exposure to a nursing infant is not expected, and adverse effects in breastfed infants are considered improbable.[2]

The pharmacokinetic profile of Cenegermin is the critical enabler of its favorable safety profile. The ability to deliver a biologically potent growth factor directly to the target tissue while avoiding meaningful systemic exposure is a key principle of its therapeutic design. NGF is a powerful mitogen and survival factor, and systemic exposure could carry theoretical risks, a concern reflected in the official precaution regarding its use in patients with pre-existing ocular cancer.[23] The localized action of Cenegermin effectively mitigates these risks. The ophthalmic route of administration, coupled with the drug's molecular properties that limit systemic absorption, creates a "pharmacokinetic firewall." This spatial restriction of its powerful biological effects to the ocular surface is central to its positive benefit-risk profile, ensuring that its therapeutic action is delivered precisely where it is needed without causing unintended effects elsewhere in the body.

Clinical Efficacy in Neurotrophic Keratitis

Pathophysiological Rationale for NGF Replacement Therapy in NK

Neurotrophic keratitis is a degenerative corneal disease that arises from damage to the sensory fibers of the trigeminal nerve, which innervate the cornea.[5] This nerve damage leads to a significant reduction or complete loss of corneal sensation. Beyond sensation, these nerves provide continuous trophic support to the corneal epithelium, releasing neuropeptides and growth factors that are essential for cellular metabolism, proliferation, and wound healing.[2] The loss of this trophic support disrupts the homeostasis of the ocular surface, leading to spontaneous epithelial breakdown, the development of persistent epithelial defects (PEDs), and in severe cases, corneal ulceration, stromal melting, perforation, and permanent vision loss.[2] Cenegermin therapy is founded on the direct and logical rationale of replacing the missing endogenous NGF. By topically administering a recombinant form of this essential protein, the therapy aims to restore the critical trophic signaling required to stimulate corneal nerve repair and promote the healing of the damaged epithelium.[4]

Critical Analysis of Pivotal Trials: NGF0212 (REPARO) and NGF0214

The clinical efficacy and safety of Cenegermin for the treatment of NK were rigorously established in two pivotal, multicenter, randomized, double-masked, vehicle-controlled clinical trials.[6] These studies, known as NGF0212 (conducted in Europe and often referred to as REPARO) and NGF0214 (conducted in the United States), enrolled a combined total of 151 patients. The study population consisted of adults with moderate (Stage 2, characterized by a PED) or severe (Stage 3, characterized by a corneal ulcer) NK.[2] In both trials, patients were randomized to receive either Cenegermin ophthalmic solution at a concentration of 20 mcg/mL or a matching vehicle (placebo) solution. The treatment regimen was intensive, requiring patients to instill one drop into the affected eye six times daily at two-hour intervals for a duration of eight weeks.[7] The design elements of these key trials are summarized in Table 2.

Table 2: Summary of Pivotal Clinical Trial Designs (NGF0212 & NGF0214)

ParameterStudy NGF0212 (REPARO)Study NGF0214
PhasePhase 1/2Phase 2
Geographic RegionEuropeUnited States
Number of Patients (Randomized)101 (50 Cenegermin, 51 Vehicle)48 (24 Cenegermin, 24 Vehicle)
Patient PopulationAdults with Stage 2 or 3 Neurotrophic KeratitisAdults with Stage 2 or 3 Neurotrophic Keratitis
InterventionCenegermin 20 mcg/mL ophthalmic solutionCenegermin 20 mcg/mL ophthalmic solution
ComparatorVehicle ophthalmic solutionVehicle ophthalmic solution
Dosing Regimen1 drop, 6 times daily at 2-hour intervals for 8 weeks1 drop, 6 times daily at 2-hour intervals for 8 weeks
Primary EndpointPercentage of patients with complete corneal healing at Week 8Percentage of patients with complete corneal healing at Week 8
Follow-up Duration48 weeks post-treatment24 weeks post-treatment
Data compiled from sources.7 Note: Patient numbers may vary slightly between sources based on analysis populations (e.g., intention-to-treat vs. per-protocol).

Primary and Secondary Efficacy Outcomes: Rates of Complete Corneal Healing

The primary efficacy endpoint in both pivotal trials was the percentage of patients who achieved complete corneal healing at the end of the 8-week treatment period, as assessed by masked central readers evaluating corneal fluorescein staining.[30] The results from both studies demonstrated a statistically significant and clinically meaningful superiority of Cenegermin over vehicle.

  • In study NGF0214 (the US trial), a conservative assessment defined complete healing as 0 mm of lesion staining with no other residual staining. By this measure, 65.2% (15 out of 23) of patients in the Cenegermin arm achieved complete healing, compared to only 16.7% (4 out of 24) in the vehicle arm. This yielded a substantial treatment difference of 48.6% (95% CI: 24.0%–73.1%; p < 0.001).[7]
  • In study NGF0212 (the REPARO trial in Europe), using a similar definition, 72.0% (36 out of 50) of patients treated with Cenegermin achieved complete corneal healing at week 8, compared to 33.3% (17 out of 51) of patients who received the vehicle. The treatment difference was 38.7% (95% CI: 20.7%–56.6%; p < 0.01).[7]

The durability of the healing effect was also assessed. Among patients who were healed after the 8-week treatment course, the rate of disease recurrence over a 48-week follow-up period was relatively low, reported to be between 14% and 20%.[7] The primary efficacy outcomes are detailed in Table 3.

Table 3: Primary Efficacy Outcomes (Complete Corneal Healing) in Pivotal Trials

StudyTreatment ArmN (Healed / Total)% Healed at Week 8Treatment Difference (vs. Vehicle)95% Confidence Intervalp-value
NGF0214Cenegermin 20 mcg/mL15 / 2365.2%48.6%24.0%, 73.1%< 0.001
Vehicle4 / 2416.7%---
NGF0212Cenegermin 20 mcg/mL36 / 5072.0%38.7%20.7%, 56.6%< 0.01
Vehicle17 / 5133.3%---
Data compiled from sources.7 Healing defined as 0 mm staining. Patient numbers reflect specific analysis populations reported in the sources.

Analysis of Functional Outcomes: Corneal Sensitivity and Visual Acuity

While Cenegermin demonstrated unequivocal success in promoting anatomical healing of the corneal surface, the pivotal trials conducted in patients with moderate to severe (Stage 2/3) NK did not show a statistically significant advantage for Cenegermin over vehicle in improving key functional outcomes. Specifically, at the 8-week primary endpoint, there were no significant differences observed in the improvement of corneal sensitivity (as measured by aesthesiometry) or best-corrected visual acuity between the treatment and control groups.[26]

Post-Marketing Evidence and Efficacy in Early-Stage (Stage 1) Disease

Following its approval, further investigation into the effects of Cenegermin was conducted. A notable post-marketing, open-label trial (known as the DEFENDO study) was designed to evaluate the efficacy and safety of Cenegermin specifically in patients with early-stage (Stage 1) NK, a population not included in the original pivotal trials.[31]

The results from this study were highly encouraging. After an 8-week course of treatment, 84.8% of patients with Stage 1 NK achieved complete corneal epithelial healing.[31] Critically, and in stark contrast to the findings in the more advanced disease stages, this study also demonstrated statistically significant improvements in functional outcomes. Patients experienced measurable gains in both corneal sensitivity and visual acuity, and these beneficial effects were sustained through the post-treatment follow-up period.[31]

The disparity in functional outcomes between the trials for advanced (Stage 2/3) and early-stage (Stage 1) NK suggests the existence of a critical "therapeutic window" for intervention. While Cenegermin is highly effective at promoting anatomical healing of the corneal surface regardless of disease stage, its capacity to restore nerve function and improve vision appears to be greatest when administered earlier in the disease continuum. In Stage 1 NK, the pathology is primarily limited to epithelial distress and neural dysfunction, which are the direct targets of NGF therapy. In the more advanced Stages 2 and 3, secondary and often irreversible pathologies, such as stromal scarring and profound nerve fiber loss, have likely already occurred. NGF therapy cannot reverse established scars. This leads to a compelling clinical conclusion: early diagnosis of NK and prompt initiation of Cenegermin therapy may be crucial not only to halt disease progression but also to maximize the potential for a full functional recovery, rather than just a structural repair. This has significant implications for clinical practice, advocating for a more aggressive approach to treating the earliest signs of the disease.

Comprehensive Safety and Tolerability Profile

Adverse Events Profile from Integrated Clinical Trial Data

The safety profile of Cenegermin has been well-characterized through its clinical development program. The therapy is generally well-tolerated, with the majority of reported adverse events being local to the eye, mild-to-moderate in severity, and transient in nature.[1]

The most frequently reported adverse reaction is eye pain following instillation, which occurred in approximately 16% of patients in clinical trials.[1] Other common adverse reactions, occurring in 1% to 10% of patients, include eye inflammation (which may encompass anterior chamber inflammation), increased lacrimation (watery eyes), eyelid pain, a sensation of a foreign body in the eye, conjunctival hyperemia (redness due to enlarged blood vessels), and photophobia (light sensitivity).[1] Systemic side effects are rare, though headache has been reported as a common adverse event.[8] A summary of the most common adverse reactions is provided in Table 4.

Table 4: Summary of Common Adverse Reactions (Frequency ≥1%)

System Organ ClassAdverse ReactionFrequency CategoryIncidence Rate (%)
Eye DisordersEye painVery Common~16%
Eye inflammationCommon1-10%
Lacrimation increasedCommon1-10%
Eyelid painCommon1-10%
Foreign body sensation in the eyeCommon1-10%
Conjunctival hyperemiaCommon1-10%
PhotophobiaCommon1-10%
Corneal depositsCommon1-10%
BlepharitisCommon1-10%
Nervous System DisordersHeadacheCommon1-10%
Data compiled from sources.1 Incidence rates are approximate based on integrated trial data.

In-Depth Review of Warnings, Precautions, and Contraindications

The prescribing information for Cenegermin outlines several key points for safe use, although it has no absolute contraindications.

Contraindications

There are no labeled contraindications for the use of Cenegermin, meaning there are no specific medical conditions in which the drug must be avoided entirely.[9]

Warnings and Precautions

  • Use with Contact Lenses: Patients must be instructed to remove contact lenses before applying Oxervate. Lenses should not be reinserted for at least 15 minutes following administration. The presence of a contact lens, whether therapeutic or corrective, could theoretically impede the distribution of Cenegermin onto the corneal lesion, potentially reducing its efficacy.[2]
  • Eye Discomfort: Patients should be counseled that mild to moderate eye discomfort, particularly eye pain, may occur during treatment. They should be advised to contact their physician if the reaction is severe or concerning.[8]
  • Eye Infections: The presence of an active eye infection is a relative contraindication. Any existing infection should be adequately treated before initiating Cenegermin therapy. If an eye infection develops during the course of treatment, Cenegermin should be suspended until the infection is resolved.[23]
  • Ocular Cancer: As a growth factor, Cenegermin may theoretically influence the growth of ocular tumors. Therefore, it should be used with caution in patients with a history of ocular cancer. It is recommended that these patients be monitored for cancer progression during and after treatment.[23]

Management of Common Side Effects, Notably Ocular Pain

The most common adverse event associated with Cenegermin, ocular pain, presents a unique clinical consideration. Neurotrophic keratitis is fundamentally a disease of corneal numbness. Therefore, the emergence of pain during treatment is often interpreted not merely as a negative side effect, but as a potential clinical sign of therapeutic success—an indication that corneal nerves are regenerating and their function is being restored.[7]

This mechanistic link between the primary adverse event and the drug's desired therapeutic action is a critical concept for patient counseling. Explaining to patients that the discomfort may be a positive signal that the nerves are "waking up" can reframe their experience, improve their understanding of the treatment process, and enhance adherence to the demanding regimen. For patients who find the pain difficult to tolerate, clinicians have suggested practical management strategies. These include temporarily reducing the dosing frequency (e.g., to three times daily) to allow for adaptation, or "chasing" the Cenegermin drop with a chilled, preservative-free artificial tear a few minutes after instillation to help soothe the eye.[40] In rare cases of severe pain, discontinuation of the medication may be necessary.[40]

Regulatory Journey and Place in Therapy

Global Regulatory Approvals and Expedited Pathway Designations

Cenegermin's path to market was characterized by rapid development and recognition from major global regulatory bodies, reflecting the significant unmet need for an effective NK treatment.

  • European Medicines Agency (EMA): Cenegermin received marketing authorization in the European Union under the brand name Oxervate in July 2017. The approved indication is for the treatment of moderate or severe neurotrophic keratitis in adults.[1]
  • U.S. Food and Drug Administration (FDA): The FDA approved Oxervate in August 2018 for the treatment of neurotrophic keratitis.[1]

The development and review of Cenegermin were accelerated by a remarkable collection of special regulatory designations, which are reserved for drugs that demonstrate the potential to address serious conditions. These included:

  • Orphan Drug Designation: Granted by both the FDA and EMA for the treatment of neurotrophic keratitis, a rare disease. It was also granted this status for the investigation of retinitis pigmentosa.[6]
  • Breakthrough Therapy Designation (FDA): This designation is intended to expedite the development of drugs for serious conditions where preliminary evidence indicates substantial improvement over available therapy.[8]
  • Fast Track Designation (FDA): Granted to facilitate the development and expedite the review of drugs to treat serious conditions and fill an unmet medical need.[8]
  • Priority Review (FDA): This designation shortens the FDA's review timeline and is granted to drugs that could provide significant improvements in the treatment of a serious condition.[6]

The conferral of this full suite of four major expedited pathway designations by the FDA is not merely a procedural footnote in the drug's history. It constitutes a powerful regulatory narrative that validates both the profound clinical innovation of Cenegermin and the severity of NK as a disease that previously had no approved pharmacological treatments. For a single therapeutic candidate to receive all these designations is exceptional and signifies that, from a very early stage, regulatory authorities recognized its potential to create a paradigm shift in patient care. This successful regulatory journey serves as an important case study for the development of novel therapies for other rare and neglected diseases.

Positioning as a First-in-Class, Disease-Modifying Treatment for Neurotrophic Keratitis

Cenegermin is unequivocally recognized as a first-in-class medication.[1] It holds the distinction of being the first and, to date, the only pharmacological therapy approved by the FDA specifically for the treatment of neurotrophic keratitis.[2] Furthermore, its approval marked a milestone in ophthalmology as the first-ever topical biologic to be approved in the field.[2]

Before the availability of Cenegermin, the management of NK was limited to supportive and often palliative measures, such as aggressive lubrication, bandage contact lenses, amniotic membrane transplantation, and surgical interventions like tarsorrhaphy (partially sewing the eyelids shut).[6] These approaches aimed to protect the vulnerable corneal surface but did not address the underlying neuropathic deficit. Cenegermin is therefore positioned as a true disease-modifying therapy, as it is the first treatment that directly targets the root cause of the disease—the loss of neurotrophic support—to actively promote healing and restore corneal integrity.[4]

Prescribing Protocol and Patient Management for Oxervate®

Formulation, Dosage, and the 8-Week Treatment Regimen

Oxervate is supplied as a sterile, preservative-free, clear, and colorless ophthalmic solution. The concentration of the active ingredient, cenegermin-bkbj, is 0.002%, which corresponds to 20 micrograms per milliliter (mcg/mL). It is packaged in multiple-dose vials intended for single-day use.[2]

The recommended treatment protocol is standardized and intensive. The dosage is one drop of the solution instilled into the conjunctival sac of the affected eye(s). This dose is administered six times per day, at regular two-hour intervals during waking hours. The full course of treatment is for a continuous duration of eight weeks.[2]

Detailed Administration Protocol: Storage, Handling, and Patient Education Imperatives

The use of Oxervate involves a complex and highly structured protocol that is critical for maintaining the drug's sterility and stability. Comprehensive patient education is imperative for successful therapy.

Storage (Cold Chain Management)

  • The medication requires a strict, uninterrupted cold chain. It is stored frozen at or below -20°C (-4°F) at the specialty pharmacy.[10]
  • It is dispensed to the patient in a validated insulated container. The patient must transfer the weekly cartons to a refrigerator (2°C to 8°C or 36°F to 46°F) within 5 hours of receiving the shipment.[10]
  • Unopened vials can be stored in the refrigerator for a maximum of 14 days. Vials must never be refrozen.[13]

Daily Preparation and Use

  • Each morning, the patient takes one new vial from the refrigerator. If frozen, it must be allowed to thaw at room temperature for up to 30 minutes. The vial must never be shaken, as this can denature the protein.[9]
  • Administration requires the use of a supplied Delivery System Kit, which contains single-use vial adapters, sterile single-use pipettes for drawing up each dose, and sterile disinfectant wipes.[9]
  • Once a vial adapter is attached to the vial, the clock starts. The vial is now viable for a maximum of 12 hours. During this 12-hour period, the opened vial can be stored either in the refrigerator or at room temperature (up to 25°C or 77°F).[10]
  • For each of the six daily doses, a new sterile pipette must be used to withdraw the solution from the vial adapter. The used pipette is discarded immediately after instillation, even if solution remains inside.[9]

Disposal

  • At the end of the day, or 12 hours after the vial adapter was first connected (whichever comes first), the vial and any remaining solution must be discarded.[9]

The key steps in this complex protocol are summarized in Table 5.

Table 5: Oxervate® Dosing, Administration, and Storage Summary

PhaseInstruction
Long-Term Storage (Pharmacy)Store frozen at or below -20°C.
Patient Storage (Unopened Vials)Transfer to refrigerator (2°C to 8°C) within 5 hours of receipt. Store for up to 14 days. Do not refreeze.
Daily PreparationRemove one vial from refrigerator. Thaw at room temperature for up to 30 minutes. Do not shake the vial. Attach a new vial adapter.
DosingEvery 2 hours (6 times per day), use a new sterile pipette to withdraw one dose. Instill one drop in the affected eye.
Post-DosingWait at least 15 minutes before instilling any other eye drops or reinserting contact lenses. Discard the used pipette immediately.
Opened Vial StorageThe opened vial (with adapter attached) is viable for a maximum of 12 hours. It can be stored in the refrigerator or at room temperature during this time.
End-of-DayDiscard the vial and any remaining solution 12 hours after opening, even if not empty.
Protocol compiled from sources.9

The extreme complexity of this administration protocol is not arbitrary; it is a direct and necessary trade-off for delivering a preservative-free biologic ophthalmic solution. Biologic proteins are inherently less stable than small-molecule drugs. While preservatives could enhance stability and allow for a simpler, traditional multi-dose eye drop bottle, they are contraindicated in this context. Many common ophthalmic preservatives, such as benzalkonium chloride, are known to be toxic to the corneal epithelium and can inhibit the very healing process the drug is intended to promote.[23] To ensure both the sterility and the stability of the Cenegermin protein without using harmful preservatives, the manufacturer had to engineer this intricate system of cold-chain logistics, single-day vials, and sterile, single-use components. This directly connects the drug's advanced molecular nature to the lived daily experience of the patient, highlighting a crucial point: the ultimate success of this high-tech therapy is heavily dependent on decidedly low-tech factors such as patient dexterity, meticulous adherence, access to refrigeration, and comprehensive education.

Practical Challenges: Adherence, Cost, and Access Considerations

The demanding nature of the Oxervate regimen presents significant practical challenges that can impact real-world effectiveness. The frequent dosing schedule, complex multi-step preparation for each dose, and the absolute requirement for cold-chain storage create a substantial treatment burden that can be difficult for some patients, particularly the elderly or those with comorbidities, to manage, potentially compromising adherence.[40]

Furthermore, access to the therapy can be a major hurdle. As a novel, complex biologic, Cenegermin has a high acquisition cost, cited in one report as potentially $12,000 out of pocket for an 8-week course if not covered by insurance.[40] The process of obtaining insurance approval can be cumbersome, often requiring documentation of failure with multiple other therapies before authorization is granted.[40] These financial and administrative barriers can prevent or delay access for eligible patients.

Future Horizons: Investigational and Off-Label Applications

Review of Clinical Data for Dry Eye Syndrome and Other Ocular Pathologies

Leveraging the known neurotrophic, anti-inflammatory, and healing properties of NGF, Cenegermin has been formally investigated for indications beyond neurotrophic keratitis.

Dry Eye Syndrome (DES)

Cenegermin has completed Phase 2 clinical trials for the treatment of moderate to severe dry eye syndrome.[6] The results of one multicenter, randomized, vehicle-controlled trial (NCT03982368) were mixed. The study did not meet its primary endpoint, which was a statistically significant improvement in an objective sign of dry eye (tear production as measured by the Schirmer I test) after 4 weeks of treatment. However, the trial did demonstrate success on a key secondary endpoint: patients treated with Cenegermin (particularly in the twice-daily and three-times-daily arms) reported statistically significant and sustained improvements in their symptoms, as measured by the SANDE (Symptom Assessment in Dry Eye) questionnaire, compared to vehicle.[44]

This divergence in outcomes—subjective symptom improvement in the absence of objective sign improvement—points toward a more nuanced therapeutic role for NGF in a complex, multifactorial disease like DES compared to its clear-cut efficacy in NK. The data suggest that Cenegermin's primary benefit in this population may not be a direct secretagogue effect on the lacrimal glands to increase tear volume. Instead, its mechanism may be more related to modulating corneal neuro-sensory pathways, reducing nerve hypersensitivity, and alleviating the discomfort and neuropathic pain that are characteristic features of a significant subset of DES patients. This repositions the drug's potential from a broad-based therapy for all dry eye to a more targeted treatment for a specific, neuro-sensory phenotype of the disease, a promising but more challenging path toward a potential future approval.

Other Ocular Pathologies

Cenegermin has also been granted orphan drug designation for retinitis pigmentosa, and its potential in glaucoma has been considered, though clinical development in these areas is less advanced.[6] Additionally, a Phase 4 clinical trial exploring its use in neurotrophic and ulcerative keratitis was registered but later withdrawn before enrollment, for reasons not specified in the available materials.[45]

The Potential of NGF Mimetics and Next-Generation Neurotrophic Therapies

The clinical and commercial success of Cenegermin has provided powerful validation for the neurotrophic pathway as a viable and potent therapeutic target in ophthalmology. This pioneering work paves the way for the development of next-generation therapies. Future research is likely to focus on molecules such as NGF mimetics—smaller, synthetic molecules engineered to bind and activate NGF receptors with high affinity.[46] These agents could potentially offer significant advantages over a recombinant protein, such as improved chemical stability (potentially eliminating the need for a cold chain), lower manufacturing costs, and simpler formulation and delivery methods. The exploration of other neurotrophic factors and related molecules, such as tavilermide or glial cell line-derived neurotrophic factor (GDNF), also represents a promising avenue for future innovation in treating degenerative ocular diseases.[12]

Conclusions

Cenegermin (Oxervate®) is a landmark achievement in ophthalmic therapeutics, representing the successful translation of decades of research on nerve growth factor into a targeted, first-in-class treatment for neurotrophic keratitis. As a recombinant human nerve growth factor, its mechanism of action is uniquely tailored to address the core pathophysiology of the disease by restoring the trophic support essential for corneal nerve function and epithelial integrity.

The clinical evidence supporting its use is robust, with pivotal trials demonstrating a statistically significant and clinically profound ability to achieve complete corneal healing in patients with moderate to severe disease, for whom previous options were largely palliative. Its safety profile is well-defined and predominantly characterized by local, manageable side effects, with the most common event, eye pain, potentially serving as a paradoxical indicator of successful nerve re-innervation.

However, the profound innovation of Cenegermin is accompanied by significant practical challenges. The treatment is defined by a highly complex and burdensome administration protocol, dictated by the biologic nature of the drug and the need to avoid preservatives. This regimen, combined with the therapy's high cost and potential access hurdles, creates a substantial barrier to its widespread use and places a heavy emphasis on patient selection and education.

Looking forward, the therapeutic principle established by Cenegermin has opened new frontiers. While its path in other indications like dry eye syndrome appears more nuanced, its success has validated the neurotrophic pathway as a critical target. This will undoubtedly spur the development of next-generation therapies, such as NGF mimetics, that may one day offer similar or enhanced efficacy with a more favorable administration profile. In conclusion, Cenegermin stands as a transformative therapy that has fundamentally altered the treatment landscape for a rare and debilitating eye disease, while simultaneously illuminating a promising path for future innovation in regenerative ophthalmology.

Works cited

  1. Cenegermin - Wikipedia, accessed September 30, 2025, https://en.wikipedia.org/wiki/Cenegermin
  2. Cenegermin - StatPearls - NCBI Bookshelf, accessed September 30, 2025, https://www.ncbi.nlm.nih.gov/books/NBK573069/
  3. pmc.ncbi.nlm.nih.gov, accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6789413/#:~:text=Mechanism%20Of%20Action%20Of%20Cenegermin&text=NGF%20acts%20directly%20on%20corneal,to%20support%20corneal%20re%2Dinnervation.
  4. Clinical Review - Cenegermin (Oxervate) - NCBI Bookshelf, accessed September 30, 2025, https://www.ncbi.nlm.nih.gov/books/NBK603342/
  5. What is Cenegermin-BKBJ used for? - Patsnap Synapse, accessed September 30, 2025, https://synapse.patsnap.com/article/what-is-cenegermin-bkbj-used-for
  6. Cenegermin | New Drug Approvals, accessed September 30, 2025, https://newdrugapprovals.org/2018/08/23/cenegermin/
  7. Efficacy & Safety - Oxervate, accessed September 30, 2025, https://oxervate.com/hcp/efficacy-safety/
  8. NEUROTROPHIC KERATITIS - Dompé, accessed September 30, 2025, https://www.dompe.com/us/therapeutic-areas/ophthalmics/neurotrophic-keratitis/
  9. OXERVATE® (cenegermin-bkbj) ophthalmic solution, for topical ophthalm, accessed September 30, 2025, https://oxervate.com/wp-content/uploads/2025/01/OXERVATE-PI-Rev.-12-2024.pdf
  10. BLA 761094 Page 8 This label may not be the ... - accessdata.fda.gov, accessed September 30, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761094s000lbl.pdf
  11. Cenegermin Completed Phase 2 Trials for Dry Eye Syndrome (DES) Treatment - DrugBank, accessed September 30, 2025, https://go.drugbank.com/drugs/DB13926/clinical_trials?conditions=DBCOND0074986&phase=2&purpose=treatment&status=completed
  12. Cenegermin, accessed September 30, 2025, http://medbox.iiab.me/kiwix/wikipedia_en_medicine_2019-12/A/Cenegermin
  13. 761094Orig1s000 - accessdata.fda.gov, accessed September 30, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2018/761094Orig1s000ChemR.pdf
  14. Oxervate (cenegermin) C16015-A - Molina Marketplace, accessed September 30, 2025, https://www.molinamarketplace.com/~/media/Molina/PublicWebsite/PDF/providers/common/pa-criteria/Oxervate%20cenegermin.pdf
  15. Topical cenegermin associated ocular surface and contact lens drug precipitate deposit formation - PMC - PubMed Central, accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9133754/
  16. Cenegermin: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed September 30, 2025, https://go.drugbank.com/drugs/DB13926
  17. pmc.ncbi.nlm.nih.gov, accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6789413/#:~:text=Cenegermin%20is%20an%20rhNGF%20produced,forms%20a%20cysteine%20knot%20structure.
  18. APPLICATION FOR PATENT TERM EXTENSION UNDER 35 U.S.C. § 156 - Regulations.gov, accessed September 30, 2025, https://downloads.regulations.gov/FDA-2019-E-1945-0002/attachment_1.pdf
  19. Cenegermin - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed September 30, 2025, https://www.ncbi.nlm.nih.gov/books/NBK535611/
  20. Recombinant Human Nerve Growth Factors to Treat Disease - Unlocking Life's Code, accessed September 30, 2025, https://www.unlockinglifescode.org/genomics-insights/recombinant-human-nerve-growth-factors-treat-disease
  21. Oxervate (cenegermin) dosing, indications, interactions, adverse effects, and more, accessed September 30, 2025, https://reference.medscape.com/drug/oxervate-cenegermin-1000288
  22. Mechanism of Action - Oxervate, accessed September 30, 2025, https://oxervate.com/hcp/mechanism-of-action/
  23. Oxervate, INN-cenegermin - European Medicines Agency, accessed September 30, 2025, https://www.ema.europa.eu/en/documents/product-information/oxervate-epar-product-information_en.pdf
  24. PRODUCT MONOGRAPH INCLUDING PATIENT MEDICATION INFORMATION PrOXERVATE cenegermin Ophthalmic Solution, 0.002% (20 mcg/mL) Topica, accessed September 30, 2025, https://pdf.hres.ca/dpd_pm/00049622.PDF
  25. Product Information Cenegermin (rbe) - Therapeutic Goods Administration (TGA), accessed September 30, 2025, https://www.tga.gov.au/sites/default/files/auspar-cenegermin-rbe-191126-pi.docx
  26. cenegermin, ophthalmic solution - OHP Preferred Drug List : Oregon Health Plan, accessed September 30, 2025, https://www.orpdl.org/durm/meetings/meetingdocs/2020_12_03/archives/2020_12_03_CenegerminNDE.pdf
  27. Cenegermin (Oxervate) use while Breastfeeding - Drugs.com, accessed September 30, 2025, https://www.drugs.com/breastfeeding/cenegermin.html
  28. Oxervate | European Medicines Agency (EMA), accessed September 30, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/oxervate
  29. Topical Cenegermin-Bkbj Dramatically Resolves Neurotrophic Keratitis Associated With Pars Plana Vitrectomy | Ophthalmic Surgery, Lasers and Imaging Retina - Slack Journals, accessed September 30, 2025, https://journals.healio.com/doi/10.3928/23258160-20250124-01
  30. Topical Recombinant Human Nerve Growth Factor (Cenegermin) for ..., accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/31585826/
  31. Phase IV Multicenter, Prospective, Open-Label Clinical Trial of Cenegermin (rhNGF) for Stage 1 Neurotrophic Keratopathy (DEFENDO) - PMC, accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10787719/
  32. Topical Recombinant Human Nerve Growth Factor (Cenegermin) for Neurotrophic Keratopathy: A Multicenter Randomized Vehicle-Controlled Pivotal Trial - Loma Linda University, accessed September 30, 2025, https://experts.llu.edu/en/publications/topical-recombinant-human-nerve-growth-factor-cenegermin-for-neur-2
  33. Navigating the Neurotrophic Keratitis Treatment Landscape - PRIME Education, accessed September 30, 2025, https://primeinc.org/go/related_materials_download/media.primeinc.org/upload/programs/109WB242/Navigating_the_Neurotrophic_Keratitis_Treatment_Landscape.pdf
  34. Cenegermin: A Review in Neurotrophic Keratitis - PubMed, accessed September 30, 2025, https://pubmed.ncbi.nlm.nih.gov/32185680/
  35. Cenegermin Completed Phase 4 Trials for Neurotrophic Keratopathy Treatment - DrugBank, accessed September 30, 2025, https://go.drugbank.com/drugs/DB13926/clinical_trials?conditions=DBCOND0077055&phase=4&purpose=treatment&status=completed
  36. Neurotrophic Keratitis Treatment - OXERVATE (cenegermin-bkbj), accessed September 30, 2025, https://oxervate.com/
  37. Dosing & Administration - Oxervate, accessed September 30, 2025, https://oxervate.com/hcp/dosing-administration/
  38. What Is OXERVATE? - Download A Doctor Discussion Guide, accessed September 30, 2025, https://oxervate.com/about-oxervate/
  39. What to expect with OXERVATE®, accessed September 30, 2025, https://oxervate.com/wp-content/uploads/2024/01/72956_OXERVATE_QuickstartGuide-Digital_122023.pdf
  40. Neurotrophic Keratitis: Two Treatments in Practice - American Academy of Ophthalmology, accessed September 30, 2025, https://www.aao.org/eyenet/article/neurotrophic-keratitis-two-treatments-in-practice
  41. www.ncbi.nlm.nih.gov, accessed September 30, 2025, https://www.ncbi.nlm.nih.gov/books/NBK573069/#:~:text=Cenegermin%20is%20a%20medication%20belonging,in%20the%20field%20of%20ophthalmology.
  42. Neurotrophic Keratitis (NK) - Cornea Research Foundation of America, accessed September 30, 2025, https://www.cornea.org/Learning-Center/Conditions-Research-Areas/Neurotrophic-Keratitis-(NK).aspx
  43. How To Use OXERVATE - Dosing & Application Information, accessed September 30, 2025, https://oxervate.com/how-to-use-oxervate/
  44. Recombinant human nerve growth factor (cenegermin) for moderate-to-severe dry eye: phase II, randomized, vehicle-controlled, dose-ranging trial - PubMed Central, accessed September 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11253442/
  45. Cenegermin Withdrawn Phase 4 Trials for Neurotrophic Keratopathy / Keratitis, Ulcerative Treatment | DrugBank Online, accessed September 30, 2025, https://go.drugbank.com/drugs/DB13926/clinical_trials?conditions=DBCOND0145587%2CDBCOND0077055&phase=4&purpose=treatment&status=withdrawn
  46. New and Emerging Treatments for Neurotrophic Keratitis, accessed September 30, 2025, https://www.aao.org/eyenet/article/new-emerging-treatments-neurotrophic-keratitis

Published at: September 30, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.