MedPath

Verteporfin Advanced Drug Monograph

Published:Aug 4, 2025

Generic Name

Verteporfin

Brand Names

Visudyne

Drug Type

Small Molecule

Chemical Formula

C41H42N4O8

CAS Number

129497-78-5

Associated Conditions

Subfoveal Choroidal Neovascularization (CNV)

A Comprehensive Monograph on Verteporfin (Visudyne®)

Section 1: Introduction and Drug Profile

1.1 Overview and Therapeutic Context

Verteporfin is a synthetic, second-generation benzoporphyrin derivative monoacid (BPD-MA), a small molecule photosensitizing agent that has carved out a significant and evolving role in modern therapeutics.[1] Marketed under the trade name Visudyne®, it is clinically supplied as a liposomal formulation designed for intravenous administration.[1] Its primary and most established application is in the field of ophthalmology as the active agent in photodynamic therapy (PDT), a modality that utilizes light to achieve targeted tissue destruction.[1] Specifically, Verteporfin PDT was developed for the selective elimination of abnormal blood vessels in the choroid, the vascular layer of the eye, which is the hallmark of conditions such as the exudative (wet) form of age-related macular degeneration (wet AMD).[1]

Beyond its foundational role as a light-activated drug, a deeper understanding of its molecular interactions has revealed a second, distinct pharmacological identity. Verteporfin has been identified as a potent, light-independent inhibitor of critical intracellular signaling pathways, most notably the Hippo-YAP pathway.[7] This discovery has catalyzed a wave of investigational research, repositioning Verteporfin as a candidate for entirely new therapeutic applications, including oncology and the burgeoning field of regenerative medicine.[4] Its journey from a specialized ophthalmic treatment to a potential multi-purpose pathway modulator illustrates a compelling narrative of scientific discovery and drug repurposing.

1.2 Identification and Chemical Properties

The nomenclature associated with Verteporfin reflects its developmental journey from a preclinical compound to a globally approved medication. Early research and internal documentation refer to it by the code CL 318,952 and the descriptive name BPD-MA (Benzoporphyrin Derivative Monoacid), signifying its origins as a specific chemical entity under investigation.[7] Upon formal characterization, it was assigned the International Nonproprietary Name (INN) "Verteporfin," and its commercial formulation was branded as Visudyne®.[1] This progression is a classic hallmark of the pharmaceutical development pipeline.

The chemical nature of Verteporfin as a benzoporphyrin derivative is fundamental to its biological activity. Its core structure is related to that of natural porphyrins, such as the heme in hemoglobin, which are characterized by a large macrocyclic ring system that is highly efficient at absorbing light. The specific modifications in Verteporfin, including the fusion of a benzene ring to the porphyrin core and the presence of various side chains, are the result of deliberate medicinal chemistry designed to optimize its therapeutic properties.[1] These modifications tune the molecule's electronic structure, shifting its maximum light absorption into the far-red spectrum (around 690 nm).[6] This is a critical feature for its ophthalmic use, as red light penetrates biological tissues more deeply and with less scatter than light of shorter wavelengths, allowing for effective activation of the drug in the choroid at the back of the eye. Furthermore, its overall structure imparts a high degree of lipophilicity, or hydrophobicity, which dictates its behavior in the body. This property necessitates its formulation within liposomes for intravenous delivery and explains its transport in the bloodstream via lipoproteins, a key factor in its preferential accumulation in the leaky, lipid-rich environment of abnormal neovasculature.[4] The fundamental identifiers and physicochemical properties of Verteporfin are consolidated in Table 1.

Table 1: Identification and Chemical Properties of Verteporfin

PropertyValueSource(s)
DrugBank IDDB00460[User Query]
TypeSmall Molecule[User Query]
CAS Number129497-78-512
Trade NameVisudyne®1
Synonyms/CodesBPD-MA, CL 318,952, Benzoporphyrin Derivative Monoacid Ring A7
Chemical FormulaC41​H42​N4​O8​2
Molecular Weight~718.8 g/mol2
IUPAC Name(4R,4aS)-rel-18-ethenyl-4,4a-dihydro-3,4-bis(methoxycarbonyl)-4a,8,14,19-tetramethyl-24H,26H-benzo[b]porphine-9,13-dipropanoic acid, monomethyl ester2
AppearanceLyophilized powder; Opaque dark green solution upon reconstitution2
SolubilitySoluble in DMSO (e.g., 14-50 mg/mL); sparingly soluble in aqueous solutions. Hydrophobic nature requires liposomal formulation for clinical IV use.2
StabilityLyophilized powder stable for ≥ 4 years at -20°C, protected from light. Reconstituted solution stable for 4 hours.2
ATC CodeS01LA01 (Antineovascularisation agents)11

Section 2: Pharmacology and Mechanisms of Action

Verteporfin exhibits a remarkable dual pharmacology, functioning both as a light-activated agent for targeted tissue destruction and as a light-independent inhibitor of fundamental cellular signaling pathways. This duality underpins its established clinical use and its expanding investigational potential.

2.1 Primary Mechanism: Light-Activated Photodynamic Action

The foundational mechanism of Verteporfin is its function as a photosensitizer in photodynamic therapy (PDT).[1] In its inactive state and at the clinically recommended dose, the drug is not cytotoxic.[1] Its therapeutic effect is contingent upon activation by a specific wavelength of light in an oxygen-rich environment.

The process unfolds in a precise sequence:

  1. Photosensitization and Activation: Following intravenous administration, Verteporfin circulates and accumulates in target tissues. It is then activated by a low-energy, non-thermal red light, specifically at a wavelength of 689-693 nm.[6]
  2. Cytotoxicity Cascade: The absorption of photons excites the Verteporfin molecule, initiating a photochemical reaction that transfers energy to ambient molecular oxygen (O2​). This process generates highly reactive and cytotoxic species, predominantly singlet oxygen (1O2​) along with other reactive oxygen species (ROS).[1] Singlet oxygen is an extremely potent but short-lived oxidant, meaning its destructive effects are confined to a very small radius (approximately 100 nm) from its point of generation.[21]
  3. Targeted Vaso-occlusion: The primary target of these ROS is the endothelium, the inner lining of blood vessels. The ROS inflict acute, localized damage to the endothelial cells of the choroidal neovasculature.[6] This initial injury sets off a rapid biological cascade aimed at sealing the damaged vessel:
  • The damaged endothelium releases a cocktail of procoagulant and vasoactive factors, including histamines, thromboxane, and tumor necrosis factor-alpha (TNF-α).[5]
  • These factors promote platelet aggregation and adhesion to the site of injury, initiating the formation of a fibrin clot (thrombosis).[5]
  • Simultaneously, vasoconstriction occurs, leading to a slowdown or complete cessation (stasis) of blood flow within the targeted vessel.[5]

The culmination of this process is the selective occlusion and blockage of the abnormal, leaky blood vessels. A key advantage of this therapy is the high degree of selectivity; because the activation is targeted by the laser and the ROS have a very short range of action, damage to the overlying neurosensory retina and other healthy adjacent tissues is minimized.[4]

2.2 Secondary Mechanisms: Light-Independent Pathway Inhibition

The discovery that Verteporfin possesses a second, light-independent mechanism of action represents a classic and powerful example of drug repurposing. This has transformed the molecule from a specialized ophthalmic tool into a subject of intense investigation for systemic diseases. This dual identity—acting as a physically destructive agent when activated by light and as a specific molecular inhibitor in its native state—explains its seemingly disparate applications in treating blindness, cancer, and fibrosis.

  • YAP-TEAD Inhibition: Verteporfin is now recognized as a potent inhibitor of the Hippo signaling pathway, a crucial regulatory network that controls organ size, cell proliferation, and apoptosis.[7] Its specific target is the transcriptional co-activator Yes-associated protein (YAP). Verteporfin functions by physically disrupting the protein-protein interaction between YAP and its requisite DNA-binding partners, the TEA domain (TEAD) family of transcription factors.[2] By preventing this complex from forming, Verteporfin blocks the transcription of a suite of downstream genes that drive cell growth and survival.[8] This mechanism is the molecular basis for its emerging therapeutic rationales. In oncology, where the Hippo-YAP pathway is often dysregulated and hyperactivated, Verteporfin's inhibitory action can suppress tumor growth and induce apoptosis.[8] In regenerative medicine, it can prevent the fibrotic response to injury.[9]
  • Autophagy Inhibition: In addition to its effects on the Hippo pathway, Verteporfin has been identified as an inhibitor of autophagy, the cellular process of degrading and recycling damaged components.[7] It acts at an early stage of the process, specifically by blocking the formation of the autophagosome, the double-membraned vesicle that engulfs cellular debris.[7] Research has elucidated a specific molecular mechanism for this effect: Verteporfin directly targets and modifies p62, a critical scaffold protein that acts as an adaptor, binding to both the polyubiquitinated proteins marked for degradation and the LC3 protein on the nascent autophagosomal membrane. Verteporfin induces the formation of covalently crosslinked p62 oligomers, which exhibit a reduced capacity to bind their cargo, thereby stalling the autophagic process.[12]

Section 3: Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The pharmacokinetic profile of Verteporfin is central to both its efficacy and its safety. Its rapid clearance is a key advantage for its approved photodynamic therapy application but presents a significant challenge for its investigational systemic uses.

3.1 Administration and Distribution

  • Route and Absorption: Verteporfin is administered exclusively via intravenous (IV) infusion, ensuring 100% bioavailability in the systemic circulation. The standard clinical protocol involves a 10-minute constant-rate infusion.[1]
  • Distribution: As a highly lipophilic molecule, Verteporfin does not circulate freely in the aqueous environment of the blood. Instead, it is rapidly bound to and transported by plasma lipoproteins, with a particular affinity for low-density lipoprotein (LDL).[3] This transport mechanism is a cornerstone of its targeted action. Rapidly proliferating cells, such as those found in the endothelial lining of choroidal neovasculature and in many solid tumors, upregulate their expression of the LDL receptor to meet their high metabolic demands for cholesterol. Consequently, these target tissues preferentially sequester the LDL-Verteporfin complex from the circulation, leading to a higher concentration of the drug at the site of pathology compared to surrounding healthy tissue.[4]
  • Pharmacokinetic Profile: The plasma concentration of Verteporfin over time follows a biexponential decay pattern. This indicates a two-compartment model, with an initial, rapid distribution phase where the drug moves from the central (blood) compartment into peripheral tissues, lasting approximately 1 to 3 hours. This is followed by a slower terminal elimination phase.[3] The peak plasma concentration ( Cmax​) is reliably observed at the end of the 10-minute infusion period.[3]

3.2 Metabolism and Excretion

  • Metabolism: Verteporfin undergoes very limited metabolism in the body. The primary metabolic pathway is the hydrolysis of one of its ester groups by plasma and tissue esterases, converting it to its corresponding diacid metabolite, known as benzoporphyrin derivative diacid (BPD-DA).[3] The extent of this conversion is minor, with the metabolite's total exposure (as measured by the area under the curve, or AUC) accounting for less than 10% of the parent drug's exposure.[3] A crucial metabolic feature is that Verteporfin does not appear to be a substrate for, nor does it inhibit or induce, the Cytochrome P450 (CYP450) enzyme system.[1] This significantly reduces the potential for drug-drug interactions with a wide range of other medications that are metabolized through this common pathway.
  • Excretion: The elimination of Verteporfin from the body is rapid and occurs almost entirely through the hepatobiliary system. The drug is excreted, predominantly in its unchanged form, into the bile and subsequently eliminated via the feces.[3] Renal elimination is a negligible pathway, with less than 0.01% of the administered dose being recovered in the urine.[3]
  • Elimination Half-Life (t1/2​): Consistent with its rapid excretion, Verteporfin has a short terminal elimination half-life, reported to be in the range of 5 to 6 hours.[3] This means that the majority of the drug is cleared from the systemic circulation within 24 to 48 hours.

3.3 Influence of Patient-Specific Factors

The pharmacokinetic profile of Verteporfin, particularly its short half-life, is a critical determinant of its clinical use. For its approved application in PDT, this rapid clearance is a significant safety advantage, as it translates to a short and manageable period of skin photosensitivity for the patient.[3] However, for its investigational uses as a systemic pathway inhibitor in oncology or fibrosis, this same property becomes a major liability. Achieving sustained target engagement required for these indications is difficult with a drug that is cleared from the body within hours. This pharmacokinetic limitation directly informs the design of clinical trials for these new indications, necessitating frequent, repeated dosing schedules, such as the weekly infusions employed in the glioblastoma trial, to maintain therapeutic drug levels.[4] This challenge also drives research into novel drug delivery systems, such as nanosuspensions, designed to prolong the drug's circulation time.[4]

  • Age: Pharmacokinetic studies have shown that while patients aged 65 years and older may have a slightly higher mean drug exposure (AUC) and peak concentration (Cmax​) compared to younger patients, the ranges of these values show considerable overlap. The differences are not considered clinically significant, and therefore, no dose adjustments are recommended based on age alone.[3]
  • Hepatic Impairment: Given that Verteporfin is cleared almost exclusively by the liver, hepatic function has a notable impact on its pharmacokinetics. Patients with mild hepatic dysfunction exhibit a longer elimination half-life (approximately 19% greater) and a correspondingly higher total drug exposure (AUC increased by 34-42%) compared to subjects with normal liver function.[23] Consequently, Verteporfin is contraindicated in patients with severe hepatic impairment and should be used with caution in those with moderate impairment or biliary obstruction.[20]
  • Renal Impairment: As the kidneys play a negligible role in the drug's elimination, no dose adjustments are necessary for patients with any degree of renal impairment.[3]
  • Gender and Race: Clinical studies have found no significant differences in the pharmacokinetic parameters of Verteporfin between male and female subjects, or between subjects of Japanese and Caucasian ancestry.[3]

Section 4: Approved and Off-Label Clinical Applications in Ophthalmology

Verteporfin photodynamic therapy has been a cornerstone of ophthalmic treatment for specific neovascular conditions for over two decades. Its role has evolved significantly with the advent of newer therapies, but it remains an important tool for select patient populations.

4.1 Regulatory Status and Approved Indications

  • Regulatory Approvals: Verteporfin, formulated as Visudyne®, received its first major regulatory approvals in the year 2000. The United States Food and Drug Administration (FDA) granted approval on April 12, 2000.[27] Shortly thereafter, the European Medicines Agency (EMA) issued a marketing authorization valid throughout the European Union on July 27, 2000.[29]
  • Approved Indications: The approved indications are highly specific and consistent across major regulatory bodies. Verteporfin PDT is indicated for the treatment of patients with predominantly classic subfoveal choroidal neovascularization (CNV). This means the abnormal blood vessels are located directly beneath the fovea (the center of the macula responsible for sharpest vision) and are well-defined ("classic") on fluorescein angiography. The approved underlying causes for this type of CNV are:
  1. Age-related macular degeneration (AMD) [30]
  2. Pathologic myopia (severe nearsightedness) [30]
  3. Presumed ocular histoplasmosis syndrome (a fungal infection complication) [30]
  • Limitation of Use: A critical point in the labeling is that there is insufficient evidence to support the use of Visudyne® for CNV lesions that are predominantly occult (poorly defined or hidden beneath pigment or blood on angiography).[14]

4.2 Clinical Efficacy and Evolving Role in Wet AMD

The clinical history of Verteporfin in treating wet AMD provides a clear illustration of the dynamic lifecycle of a medical technology. It entered the market as a revolutionary, first-in-class therapy that established a new standard of care, only to be largely displaced from its first-line position by a subsequent disruptive technology. It has since been successfully repositioned as a valuable adjunctive or second-line therapy, demonstrating the ongoing evolution of clinical practice based on new evidence.

Initially, Verteporfin PDT was the "treatment of choice" for eligible patients with wet AMD.[6] Landmark clinical trials, such as the Treatment of Age-Related Macular Degeneration with Photodynamic Therapy (TAP) study and the Verteporfin in Photodynamic Therapy (VIP) study, provided robust evidence of its efficacy. These trials demonstrated that, compared to placebo, Verteporfin PDT significantly reduced the risk of moderate and severe vision loss, effectively slowing the progression of the disease.[6]

This paradigm shifted dramatically in the mid-2000s with the introduction of intravitreal anti-vascular endothelial growth factor (anti-VEGF) agents.[6] These drugs, which directly inhibit the key signaling protein responsible for neovascular growth, proved to be even more effective. The ANCHOR study provided definitive head-to-head evidence, showing that monthly injections of the anti-VEGF drug ranibizumab were superior to PDT monotherapy in preventing vision loss and, in many cases, improving vision.[21] This led to a rapid change in clinical guidelines, establishing anti-VEGF therapy as the new first-line standard of care for wet AMD.[34]

However, Verteporfin was not rendered obsolete. A more nuanced understanding of the disease and treatment responses has carved out a new role for PDT. There is a strong mechanistic rationale for combining PDT with anti-VEGF therapy. The localized hypoxia and tissue injury caused by PDT can trigger a compensatory upregulation of VEGF, which could theoretically limit the long-term efficacy of PDT alone.[21] By combining PDT with an anti-VEGF agent, this compensatory response can be directly counteracted. This creates a potentially synergistic effect where PDT provides physical closure of vessels while the anti-VEGF drug neutralizes the pro-angiogenic signals. Clinical trials like the MONT BLANC study have explored this combination, finding that combination therapy was non-inferior to anti-VEGF monotherapy, suggesting it as a viable option.[21] Today, Verteporfin PDT is often used in combination with anti-VEGFs for patients who have persistent fluid despite frequent anti-VEGF injections or for those with specific lesion types like polypoidal choroidal vasculopathy, where PDT is particularly effective.[6]

Table 2: Summary of Key Clinical Trials for Ophthalmic Indications

Trial NameIndication StudiedPatient Population (n)Intervention Arm(s)Comparator Arm(s)Key Efficacy Outcome(s) & ResultsSource(s)
TAP (Treatment of AMD with PDT)Wet AMD (predominantly classic subfoveal CNV)609Verteporfin PDT (6 mg/m²)Placebo infusion + sham lightAt 24 months, 53% of Verteporfin group lost <15 letters of vision vs. 37% of placebo group.6
VIP (Verteporfin in PDT)Wet AMD (subfoveal CNV, including occult with no classic)339Verteporfin PDT (6 mg/m²)Placebo infusion + sham lightSupported TAP findings for classic CNV. Insufficient evidence for occult CNV.6
VAM (Verteporfin in AMD)Wet AMD (predominantly classic subfoveal CNV)4,435Verteporfin PDT (6 mg/m²)Open-label, no comparatorLarge-scale safety and expanded access study. Confirmed low incidence of adverse events (e.g., 0.05% photosensitivity).37
ANCHORWet AMD (predominantly classic subfoveal CNV)423Ranibizumab (monthly)Verteporfin PDT (quarterly)At 24 months, 90.0% of Ranibizumab group lost <15 letters vs. 65.7% of PDT group. Ranibizumab was superior.21
MONT BLANCWet AMD (subfoveal CNV)255Verteporfin PDT + RanibizumabRanibizumab monotherapyAt 12 months, combination therapy was non-inferior to ranibizumab monotherapy.21

4.3 Compendial and Off-Label Ophthalmic Uses

Beyond its formally approved indications, Verteporfin PDT has become a standard of care for other chorioretinal conditions based on substantial clinical evidence and inclusion in medical compendia.

  • Central Serous Chorioretinopathy (CSCR): Verteporfin PDT is widely recognized as an effective off-label treatment for chronic or persistent CSCR, a condition characterized by fluid accumulation under the retina due to choroidal hyperpermeability.[1] While its use for this indication is technically unlicensed, it is supported by a large body of evidence from clinical studies and is often recommended when the condition does not resolve spontaneously.[5] To optimize the risk-benefit profile and minimize potential damage to the retinal pigment epithelium (RPE), treatment for CSCR frequently employs "safety-enhanced" protocols, which involve reducing the drug dose (e.g., half-dose) or the laser energy (e.g., half-fluence).[5]
  • Choroidal Hemangioma: This is another well-established compendial use. PDT has been shown to be effective in treating these benign vascular tumors of the choroid, leading to tumor regression, resolution of associated fluid, and improvement or stabilization of vision.[6]

Section 5: Administration, Dosing, and Clinical Practice

The administration of Verteporfin is a precise, two-step process that requires careful preparation, dosing, and light application to ensure both efficacy and safety.

5.1 Preparation and Handling

  • Reconstitution: A vial containing 15 mg of Visudyne® lyophilized powder must be reconstituted with exactly 7 mL of Sterile Water for Injection. This procedure yields a 7.5 mL volume of an opaque, dark green solution with a final Verteporfin concentration of 2 mg/mL.[16]
  • Dilution: The appropriate volume of the reconstituted drug, calculated based on the patient's body surface area, is then withdrawn from the vial. This volume is further diluted with 5% Dextrose for Injection (D5W) to a final, total infusion volume of 30 mL.[16]
  • Critical Handling Instructions: It is imperative that saline solutions (e.g., normal saline) are not used for dilution, as Verteporfin is known to precipitate in saline.[16] Additionally, Verteporfin should not be mixed in the same solution with any other drugs.[16]
  • Stability: Once reconstituted, the solution must be protected from bright light and should be used within 4 hours to ensure its potency and stability.[15]

5.2 Dosing and Infusion Protocol

  • Standard Dose: The recommended therapeutic dose of Verteporfin is 6 mg per square meter (mg/m2) of body surface area (BSA).[1] The patient's BSA is determined using their height and weight with a standard nomogram or formula.
  • Infusion: The final 30 mL diluted solution is administered intravenously as a constant-rate infusion over a period of 10 minutes. This corresponds to an infusion rate of 3 mL/minute and is best controlled using an appropriate syringe pump.[5] An in-line filter with a pore size of 1.2 microns is typically used.[16]
  • Vein Selection and Extravasation Prevention: To minimize the severe risk of extravasation (leakage of the drug into surrounding tissue), a free-flowing IV line must be established and carefully monitored. It is strongly recommended to use the largest possible vein in the arm, preferably the antecubital vein. Smaller, more fragile veins, such as those on the back of the hand, should be avoided.[16]

5.3 Laser Activation Parameters

The second step of the therapy is the precise application of laser light to activate the drug.

  • Timing: Light application must be initiated exactly 15 minutes after the start of the 10-minute intravenous infusion.[1] This timing is critical to ensure that the peak plasma concentration of the drug coincides with the light activation.
  • Laser Specifications: The procedure requires a specialized diode laser that generates non-thermal red light at a specific wavelength of 689 ± 3 nm.[5]
  • Light Dose Delivery: The standard recommended light dose is 50 Joules per square centimeter (J/cm2). This is delivered at a light intensity, or irradiance, of 600 milliwatts per square centimeter (mW/cm2). To deliver this total energy at this intensity, the laser must be applied for a duration of 83 seconds.[5]
  • Spot Size Determination: The size of the laser spot is determined based on the greatest linear dimension (GLD) of the choroidal neovascular lesion, as measured on fluorescein angiography. The treatment spot size should be set to be 1000 microns larger than the lesion's GLD. This creates a 500-micron border around the entire lesion to ensure it is fully covered by the activating light.[5] For safety, the nasal edge of the treatment spot must be positioned at least 200 microns away from the temporal edge of the optic disc, even if this means a small portion of the lesion near the nerve will not be treated.[5]

While the standard protocol is well-defined for AMD, clinical practice for off-label indications like CSCR has evolved to include modified, lower-intensity regimens. These "safety-enhanced" protocols aim to achieve a therapeutic effect while minimizing the risk of iatrogenic damage to the RPE and choroid. Table 3 contrasts the standard protocol with the most common modified regimens.

Table 3: Standard and Modified Dosing Protocols for Verteporfin PDT

ParameterStandard Protocol (for AMD)Half-Dose Protocol (for CSCR)Half-Fluence Protocol (for CSCR)Half-Time Protocol (for CSCR)
Verteporfin Dose6 mg/m²3 mg/m²6 mg/m²6 mg/m²
Fluence (Total Light Dose)50 J/cm²50 J/cm²25 J/cm²25 J/cm²
Irradiance (Light Intensity)600 mW/cm²600 mW/cm²600 mW/cm²600 mW/cm²
Time (Laser Duration)83 seconds83 seconds42 seconds42 seconds
Primary RationaleMaximize vaso-occlusion of robust CNVReduce drug load to decrease systemic and ocular side effectsReduce total energy delivery to minimize RPE/choroidal damageReduce total energy delivery (equivalent to half-fluence)
Source(s)5555

Section 6: Safety and Tolerability Profile

Verteporfin therapy is generally well-tolerated, with most adverse events being mild to moderate and transient.[47] However, its unique mechanism gives rise to a specific and critical safety concern—photosensitivity—and a risk of serious adverse events that require careful patient management and counseling.

6.1 Adverse Events and Side Effects

An analysis of the drug's documentation reveals an evolution in the official guidance regarding photosensitivity. Early pharmacokinetic data, which showed a rapid drug clearance with a half-life of 5-6 hours, suggested that a 24 to 48-hour period of photosensitivity precautions would be sufficient.[1] Indeed, the large VAM safety study reported a very low incidence of photosensitivity reactions (0.05%) with a 24-hour protection period.[37] However, current prescribing information and patient-facing materials from regulatory agencies and the manufacturer uniformly mandate a much longer 5-day period of precautions.[31] This discrepancy does not reflect a change in the drug's properties but rather a conservative, risk-averse regulatory evolution. The severe nature of potential photosensitivity reactions (severe burns and blistering) and the need to protect all patients, including those who might clear the drug more slowly (e.g., due to mild, undiagnosed hepatic impairment), likely prompted the adoption of a longer, more robust safety buffer to minimize risk.[16]

  • Most Common Adverse Events (incidence 10-30%):
  • Injection Site Reactions: Pain, edema, inflammation, discoloration, and extravasation at the infusion site are frequently reported.[5]
  • Visual Disturbances: Transient changes in vision are common, including blurred vision, decreased visual acuity, seeing flashes of light (photopsia), and visual field defects such as scotomas or floaters.[1]
  • Common Adverse Events (incidence 1-10%):
  • Infusion-Related Pain: A characteristic back pain, sometimes radiating to the chest or pelvis, can occur during the infusion but typically resolves upon its completion.[26]
  • Photosensitivity Reactions: Sunburn-like reactions on exposed skin can occur, usually within 24 hours of treatment if precautions are not followed.[1]
  • Systemic Symptoms: Asthenia (weakness or lack of energy), nausea, and flu-like symptoms (fever, chills) may occur.[48]
  • Serious Adverse Events (Rare):
  • Severe Vision Decrease: An acute and severe decrease in vision, defined as a loss of 4 or more lines on an eye chart, has been reported in 1-4% of patients within one week of treatment.[26] This risk appears to be higher in patients with occult-only CNV lesions.[26] Patients experiencing this should not be retreated until their vision has fully recovered to pre-treatment levels.[25]
  • Extravasation with Necrosis: If the drug leaks out of the vein during infusion, it can cause severe pain, inflammation, and, if the area is exposed to light, severe burns that can lead to localized skin necrosis (tissue death).[20]
  • Anaphylaxis and Hypersensitivity: Severe, life-threatening allergic reactions can occur during or immediately following the infusion. Symptoms may include hives, dyspnea (difficulty breathing), flushing, chest pain, syncope (fainting), and, on rare occasions, convulsions.[25] Medical supervision during infusion is essential.

Table 4: Summary of Adverse Events Associated with Verteporfin

System Organ ClassAdverse EventFrequencyDescription/NotesSource(s)
Eye DisordersVisual disturbances (blurred vision, photopsia, visual field defects), Dry eyes, Eyelid edemaCommonUsually transient.1
Severe vision decrease (≥4 lines)UncommonOccurs within 1 week post-treatment. Higher risk with occult CNV.26
General Disorders and Administration Site ConditionsInjection site reactions (pain, edema, inflammation, extravasation)CommonManaged with cold compresses and analgesics if needed.5
Infusion-related pain (back, chest)CommonOccurs during infusion and is transient.26
Asthenia, Fever, Flu-like symptomsCommonGeneral systemic side effects.48
Injection site necrosisRareA serious complication of extravasation with light exposure.16
Skin and Subcutaneous Tissue DisordersPhotosensitivity reaction (sunburn)CommonPreventable by strict adherence to light avoidance precautions.1
Rash, Pruritus, UrticariaUncommonCan be signs of a hypersensitivity reaction.26
Immune System DisordersHypersensitivity, Anaphylactic reactionRarePotentially life-threatening; requires immediate medical attention.25
Cardiac DisordersMyocardial infarction, Chest painRareReported in patients, sometimes with pre-existing cardiovascular history.26

6.2 Contraindications, Warnings, and Precautions

Strict adherence to contraindications and precautions is essential for the safe use of Verteporfin.

  • Absolute Contraindications:
  1. Porphyria: As a porphyrin derivative itself, Verteporfin can exacerbate this group of genetic disorders affecting heme synthesis. Its use is strictly contraindicated in any patient with porphyria.[1]
  2. Known Hypersensitivity: The drug is contraindicated in patients with a known allergy to Verteporfin or any of the excipients in the Visudyne® formulation.[1]
  3. Severe Hepatic Impairment: Due to its primary elimination through the bile, the drug is contraindicated in patients with severe liver disease, as impaired clearance would dramatically increase drug exposure and the risk of toxicity.[20]
  • Critical Precaution: Photosensitivity: This is the most important warning associated with Verteporfin therapy.
  • Duration: Patients become generally photosensitive for a period of 5 days following the infusion.[16]
  • Light Sources to Avoid: Patients must avoid exposing unprotected skin and eyes to both direct sunlight and bright indoor light sources. This includes tanning beds, bright halogen lighting, and the high-power lights used in dental offices and surgical operating rooms.[20]
  • Ineffectiveness of Sunscreen: It is crucial to counsel patients that standard UV-blocking sunscreens are not effective in preventing a photosensitivity reaction. The reaction is triggered by visible light (in the absorption range of the drug), not just ultraviolet (UV) light.[20]
  • Required Protection: For any necessary outdoor exposure during the 5-day period, patients must wear protective clothing (long sleeves, pants, gloves) and dark sunglasses.[32]
  • Role of Ambient Light: Patients should not remain in total darkness. Exposure to normal, ambient indoor light (e.g., from shaded lamps) is safe and encouraged, as it helps to inactivate the residual drug in the skin through a process called photobleaching.[20]

6.3 Drug Interactions

  • Photosensitizing Agents: Caution is advised when co-administering Verteporfin with other medications known to cause photosensitivity. These include certain antibiotics (e.g., tetracyclines, sulfonamides), diuretics, and antipsychotics (phenothiazines). The concurrent use of aminolevulinic acid, another photosensitizer, is a serious interaction.[25] Such combinations could potentiate the risk and severity of photosensitivity reactions.
  • Antagonists: The herbal remedy feverfew (Tanacetum parthenium) has been reported to act as an antagonist to Verteporfin through an unknown mechanism. Taking these two substances simultaneously is not recommended.[1]
  • Metabolic Interactions: As Verteporfin is not metabolized by the CYP450 enzyme system, the likelihood of clinically significant metabolic drug-drug interactions is low.[1]

Section 7: Investigational and Future Applications

The elucidation of Verteporfin's light-independent molecular activities has opened a new chapter in its therapeutic story, launching it into investigational use for diseases far removed from its original ophthalmic indications. These disparate applications in oncology and regenerative medicine are unified by a common mechanistic thread: the inhibition of the Hippo-YAP signaling pathway. This pathway serves as a fundamental biological "master switch" regulating tissue growth, cell proliferation, and the fibrotic response to injury. Verteporfin is now being explored as a specific tool to therapeutically modulate this master switch in different disease contexts.

7.1 Oncology

The rationale for using Verteporfin as an anti-cancer agent stems directly from its ability to inhibit the YAP-TEAD interaction and induce apoptosis, independent of light activation.[7] In many cancers, the YAP pathway is hyperactivated, driving uncontrolled proliferation and tumor growth. By disrupting this pathway, Verteporfin offers a targeted therapeutic strategy.

  • Ongoing Clinical Trials: Verteporfin is being actively investigated in several oncology settings:
  • Glioblastoma: A Phase I/II clinical trial is currently evaluating Verteporfin (as Visudyne®) for the treatment of recurrent, EGFR-mutated glioblastoma, a highly aggressive brain tumor.[4] In this trial, the drug is used systemically as a chemotherapy agent, without light activation. The protocol involves frequent (weekly) intravenous infusions, a design necessitated by the drug's short pharmacokinetic half-life.[4]
  • Pancreatic Cancer: Verteporfin is being studied in combination with the immunotherapy agent pembrolizumab for unresectable or metastatic pancreatic cancer. In this setting, PDT is used to create localized tumor destruction, which may release tumor antigens and enhance the patient's immune response to the cancer, thereby facilitating the activity of the immunotherapy drug.[53]
  • Other Cancers: Other active trials are exploring Verteporfin PDT for the treatment of localized prostate cancer and cutaneous metastases of breast cancer.[4]

7.2 Regenerative Medicine: Scar Prevention and Hair Regeneration

Perhaps the most transformative emerging application for Verteporfin is in the field of regenerative medicine, specifically for the prevention of fibrosis (scarring) and the regeneration of skin appendages.

  • Anti-Fibrotic Mechanism: Groundbreaking research has uncovered the mechanism by which Verteporfin can promote regenerative healing. Following a skin injury, mechanical tension in the wound environment activates the YAP pathway in dermal fibroblasts. This activation triggers the expression of the transcription factor Engrailed-1 (En1), converting the normal, regenerative Engrailed-1-negative fibroblasts (ENFs) into pro-fibrotic, scar-forming Engrailed-1-positive fibroblasts (EPFs).[9] These EPFs are responsible for depositing the dense, linear collagen that constitutes a scar. By locally administering Verteporfin into the wound, its YAP-inhibiting action blocks this crucial mechanotransduction signal. The fibroblasts never receive the cue to become scar-formers; they remain as ENFs and proceed to remodel the tissue in a regenerative, rather than fibrotic, manner.[9]
  • Preclinical and Early Human Evidence:
  • Animal Models: Seminal work from Stanford University has demonstrated that local injection of Verteporfin into full-thickness wounds in mice and pigs—whose skin is an excellent model for human skin—prevents scarring.[9] The treated wounds healed not with a scar, but with regenerated tissue that was nearly indistinguishable from unwounded skin. This included the regrowth of complex dermal appendages that are normally lost in scars, such as hair follicles and sweat glands, and the restoration of the skin's normal mechanical strength and elasticity.[22]
  • Human Application: This remarkable preclinical success has spurred experimental application in humans, particularly in the context of hair restoration surgery.[1] Preliminary results from early-stage experiments where Verteporfin was injected into donor site incisions during hair transplants have shown significantly reduced scarring and, critically, the growth of new hair follicles directly through the healed incision line—an outcome not seen with conventional healing.[54] This application has the potential to revolutionize the field of plastic and reconstructive surgery by minimizing surgical scars and improving cosmetic and functional outcomes.

Section 8: Conclusion and Expert Synthesis

Verteporfin represents a medication with a remarkable and evolving therapeutic narrative. It was introduced as a pioneering, first-in-class photosensitizer for photodynamic therapy, fundamentally changing the treatment landscape for patients with neovascular ocular diseases like wet AMD. Its ability to selectively occlude abnormal choroidal vessels with minimal collateral damage established a new therapeutic paradigm at a time when options were severely limited. Although the subsequent arrival of highly effective anti-VEGF agents displaced it as the primary first-line therapy for wet AMD, Verteporfin has adeptly maintained a crucial niche role. It remains an indispensable tool in combination with anti-VEGFs for treatment-refractory cases, for patients with specific lesion subtypes, and as a primary therapy for other conditions like chronic central serous chorioretinopathy and choroidal hemangioma.

The most compelling chapter in Verteporfin's story, however, is its recent renaissance, driven by the power of mechanistic discovery. The elucidation of its light-independent function as a potent inhibitor of the fundamental Hippo-YAP signaling pathway has effectively given the drug a "second life." This discovery has propelled it from the specialized realm of ophthalmology to the forefront of investigational research in two of the most challenging areas of medicine: oncology and regenerative medicine. The fact that its potential to treat aggressive brain cancer and its ability to enable scar-free wound healing stem from the same core mechanism—the modulation of a single biological master switch—underscores the profound impact of basic science on clinical innovation.

Looking forward, the trajectory of Verteporfin will be defined by both challenges and immense opportunities. For its systemic applications in oncology and fibrosis, the primary hurdle is its unfavorable pharmacokinetic profile. Overcoming the limitation of its short half-life will be critical for success and will likely depend on the development of novel, long-acting formulations or drug delivery systems. Conversely, its potential to become the first clinically approved therapeutic that can prevent scarring and promote true tissue regeneration represents a paradigm shift of monumental proportions. Such an advance would have transformative implications for surgery, trauma care, and the treatment of fibrotic diseases. The ongoing journey of Verteporfin serves as a powerful case study in the lifecycle of a drug, demonstrating that even a mature therapeutic can find new and profound purpose through the relentless pursuit of scientific understanding.

Works cited

  1. Verteporfin - Wikipedia, accessed August 4, 2025, https://en.wikipedia.org/wiki/Verteporfin
  2. Verteporfin #64260 - Cell Signaling Technology, accessed August 4, 2025, https://www.cellsignal.com/products/activators-inhibitors/verteporfin/64260
  3. Clinical Pharmacokinetics of Verteporfin - ResearchGate, accessed August 4, 2025, https://www.researchgate.net/publication/11355069_Clinical_Pharmacokinetics_of_Verteporfin
  4. Mechanisms of Action of Photodynamic Therapy with Verteporfin for the Treatment of Age-Related Macular Degeneration | Request PDF - ResearchGate, accessed August 4, 2025, https://www.researchgate.net/publication/12234142_Mechanisms_of_Action_of_Photodynamic_Therapy_with_Verteporfin_for_the_Treatment_of_Age-Related_Macular_Degeneration
  5. Photodynamic Therapy (PDT) - EyeWiki, accessed August 4, 2025, https://eyewiki.org/Photodynamic_Therapy_(PDT)
  6. Verteporfin Photodynamic Therapy for the Treatment of Chorioretinal ..., accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11178081/
  7. Verteporfin (CL 318952) | YAP Inhibitor | MedChemExpress, accessed August 4, 2025, https://www.medchemexpress.com/Verteporfin.html
  8. Verteporfin | Hippo and YAP/TAZ Signaling - Tocris Bioscience, accessed August 4, 2025, https://www.tocris.com/products/verteporfin_5305
  9. Dr Barghouthi, accessed August 4, 2025, https://verteporfin.org/
  10. Study Details | Verteporfin for the Treatment of Recurrent High Grade EGFR-Mutated Glioblastoma | ClinicalTrials.gov, accessed August 4, 2025, https://clinicaltrials.gov/study/NCT04590664
  11. ADReCS, accessed August 4, 2025, https://www.bio-add.org/ADReCS/drugSummary.jsp?drug_id=BADD_D02352&uname=9569cc70-8838-45e8-9e64-2515bc49150a
  12. Verteporfin (CAS 129497-78-5) - Cayman Chemical, accessed August 4, 2025, https://www.caymanchem.com/product/17334/verteporfin
  13. Verteporfin | C41H42N4O8 - ChemSpider, accessed August 4, 2025, https://www.chemspider.com/Chemical-Structure.4515032.html
  14. Clinical Policy: Verteporfin (Visudyne) - PA Health & Wellness, accessed August 4, 2025, https://www.pahealthwellness.com/content/dam/centene/Pennsylvania/policies/pharmacy-policies/PA.CP.PHAR.187%20Verteporfin%20(Visudyne).pdf
  15. Verteporfin (#64260) Datasheet With Images - Cell Signaling Technology, accessed August 4, 2025, https://www.cellsignal.com/products/64260/datasheet?images=1&protocol=0
  16. Recommended VISUDYNE® (verteporfin for injection) Preparation Guide - Bausch and Lomb, accessed August 4, 2025, https://www.bauschretinarx.com/visudyne/SysSiteAssets/pdf/baus3920-visudyne-prepguide-p4.pdf
  17. Visudyne Dosage Guide - Drugs.com, accessed August 4, 2025, https://www.drugs.com/dosage/visudyne.html
  18. Verteporfin - PubChem, accessed August 4, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Verteporfin
  19. verteporfin - Drug Central, accessed August 4, 2025, https://drugcentral.org/drugcard/2816
  20. Visudyne, INN-Verteporfin - European Commission, accessed August 4, 2025, https://ec.europa.eu/health/documents/community-register/2005/200501269210/anx_9210_en.pdf
  21. Different Therapeutic Approaches for Dry and Wet AMD - PMC, accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11641571/
  22. Multi-omic analysis reveals divergent molecular events in scarring and regenerative wound healing - PubMed Central, accessed August 4, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8988390/
  23. Clinical pharmacokinetics of verteporfin - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/12017349/
  24. 21-119 Visudyne Clinical Pharmacology Biopharmaceutics Review ..., accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/21-119_VISUDYNE_biopharmr_P2.pdf
  25. Verteporfin: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 4, 2025, https://www.rxlist.com/verteporfin/generic-drug.htm
  26. Visudyne, INN-verteporfin - EMA, accessed August 4, 2025, https://www.ema.europa.eu/en/documents/product-information/visudyne-epar-product-information_en.pdf
  27. Generic Visudyne Availability - Drugs.com, accessed August 4, 2025, https://www.drugs.com/availability/generic-visudyne.html
  28. Drug Approval Package: Visudyne (Verteporfin) Injection NDA 21-119, accessed August 4, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/21-119_Visudyne.cfm
  29. Visudyne, INN: verteporfin - EMA - European Union, accessed August 4, 2025, https://www.ema.europa.eu/en/documents/overview/visudyne-epar-summary-public_en.pdf
  30. Verteporfin (Visudyne®), accessed August 4, 2025, https://www.bcbst.com/mpmanual/!SSL!/WebHelp/Verteporfin.htm
  31. For AMD patients with predominantly classic subfoveal choroidal ..., accessed August 4, 2025, https://www.bauschretinarx.com/visudyne/ecp/about/
  32. A patient's guide to treatment with VISUDYNE® (verteporfin for injection) - Retinal Consultants Medical Group, accessed August 4, 2025, https://www.retinalmd.com/media/pages/retina-treatment/photodynamic-therapy/86c3eb663a-1608669759/baus3920-visudyne-patientbrochure-rgb-p7.pdf
  33. VISUDYNE professional resources for health care providers - Bausch and Lomb, accessed August 4, 2025, https://www.bauschretinarx.com/visudyne/ecp/additional-resources/professional-resources/
  34. Wet macular degeneration - Diagnosis and treatment - Mayo Clinic, accessed August 4, 2025, https://www.mayoclinic.org/diseases-conditions/wet-macular-degeneration/diagnosis-treatment/drc-20351113
  35. Treatments for Wet AMD (Advanced Neovascular AMD) - National Eye Institute, accessed August 4, 2025, https://www.nei.nih.gov/learn-about-eye-health/eye-conditions-and-diseases/age-related-macular-degeneration/treatments-wet-amd-advanced-neovascular-amd
  36. VISUDYNE patient profiles - Bausch and Lomb, accessed August 4, 2025, https://www.bauschretinarx.com/visudyne/ecp/about/patient-profiles/
  37. Verteporfin therapy in age-related macular degeneration (VAM): an ..., accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/15300071/
  38. Photodynamic therapy with verteporfin to treat CSCR - Overview, accessed August 4, 2025, https://www.guysandstthomas.nhs.uk/health-information/photodynamic-therapy-verteporfin-treat-cscr
  39. June 2020 Bulletin 254: Photodynamic therapy (PDT) with verteporfin (Visudyne®) for chronic central serous retinopathy, accessed August 4, 2025, https://medicines.bedfordshirelutonandmiltonkeynes.icb.nhs.uk/wp-content/uploads/2020/06/Bulletin-254-PDT-with-Verteporfin-chronic-central-serous-retinopathy-1.pdf
  40. Photodynamic Therapy with Verteporfin for Chronic Central Serous Chorioretinopathy: A Review of Data and Efficacy - MDPI, accessed August 4, 2025, https://www.mdpi.com/1424-8247/13/11/349
  41. Verteporfin (Visudyne) Photodynamic Therapy - Medical Clinical Policy Bulletins | Aetna, accessed August 4, 2025, https://www.aetna.com/cpb/medical/data/500_599/0594.html
  42. Visudyne Criteria - CareFirst Provider Portal, accessed August 4, 2025, https://provider.carefirst.com/carefirst-resources/provider/pdf/drug/Visudyne-Criteria.pdf
  43. Verteporfin Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed August 4, 2025, https://www.drugs.com/dosage/verteporfin.html
  44. VISUDYNE Treatment and Preparation - Bausch and Lomb, accessed August 4, 2025, https://www.bauschretinarx.com/visudyne/treatment/
  45. Half-dose verteporfin photodynamic therapy for acute central serous chorioretinopathy: one-year results of a randomized controlled trial - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/18538401/
  46. Study Details | Central Serous Chorioretinopathy Treated by Modified Photodynamic Therapy | ClinicalTrials.gov, accessed August 4, 2025, https://www.clinicaltrials.gov/study/NCT01019668
  47. Verteporfin : a review of its use in the management of subfoveal choroidal neovascularisation - PubMed, accessed August 4, 2025, https://pubmed.ncbi.nlm.nih.gov/14609361/
  48. Verteporfin (intravenous route, injection route) - Side effects & dosage - Mayo Clinic, accessed August 4, 2025, https://www.mayoclinic.org/drugs-supplements/verteporfin-intravenous-route-injection-route/description/drg-20066668
  49. Visudyne (Verteporfin Injection): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed August 4, 2025, https://www.rxlist.com/visudyne-drug.htm
  50. Visudyne (verteporfin): Uses, Side Effects, Interactions, Pictures, Warnings & Dosing, accessed August 4, 2025, https://www.webmd.com/drugs/2/drug-18177/visudyne-intravenous/details
  51. Verteporfin Injection: MedlinePlus Drug Information, accessed August 4, 2025, https://medlineplus.gov/druginfo/meds/a607060.html
  52. A Patient Guide to Visudyne Therapy, accessed August 4, 2025, https://misericordia.mb.ca/files/eye-care-md-visudyne-therapy.pdf
  53. Clinical Trials Using Verteporfin - NCI, accessed August 4, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/verteporfin?pn=1
  54. Verteporfin In Hair Transplants | Next Breakthrough Treatment? - Hair Restoration Tour, accessed August 4, 2025, https://hairrestorationtour.com/verteporfin-hair/
  55. Use of Verteporfin for Prevention of Skin Scarring | Explore Technologies - Stanford, accessed August 4, 2025, https://techfinder.stanford.edu/technology/use-verteporfin-prevention-skin-scarring

Published at: August 4, 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.