Small Molecule
C13H18N4O3
6493-05-6
Intermittent Claudication, Venous Leg Ulcer (VLU), Severe alcoholic liver disease
Pentoxifylline (PTX), known also by the alternative name oxpentifylline, is a synthetic, small-molecule drug belonging to the dimethylxanthine class.[1] It is a tri-substituted xanthine derivative, sharing a structural lineage with other well-known methylxanthines such as caffeine, theophylline, and theobromine.[3] This chemical relationship is clinically relevant, as hypersensitivity to other xanthines is a contraindication for pentoxifylline use.[3]
Primarily classified as a hemorheologic agent, pentoxifylline's main therapeutic goal is to improve the flow properties (rheology) of blood.[4] It is also characterized as a blood viscosity reducer and a competitive, non-selective phosphodiesterase (PDE) inhibitor, a mechanism that underpins many of its physiological effects.[2]
The drug was originally developed and subsequently approved for the symptomatic treatment of intermittent claudication (IC), a condition characterized by muscle pain on exertion that is a hallmark of peripheral artery disease (PAD).[1] However, the scientific and clinical understanding of pentoxifylline has evolved significantly over the decades. Its recognized pleiotropic effects—which encompass anti-inflammatory, antioxidant, and immunomodulatory properties—have spurred extensive investigation into a diverse range of conditions far beyond its initial indication.[1] These investigational uses include venous leg ulcers, osteoradionecrosis, alcoholic hepatitis, and various diabetic complications, reflecting a growing appreciation for its complex pharmacological profile.[1]
Pentoxifylline is a well-characterized small molecule with defined physical and chemical properties. It exists as a white to almost white crystalline solid or powder, which has a characteristically bitter taste.[13] Its solubility profile is a key determinant of its formulation and absorption characteristics; it is soluble in water and ethanol but only sparingly soluble in toluene.[4] The fundamental properties and identifiers of pentoxifylline are summarized in Table 1.
Table 1: Key Chemical Identifiers and Properties of Pentoxifylline
Property | Value and Source(s) |
---|---|
Chemical Name | 1H-Purine-2,6-dione, 3,7-dihydro-3,7-dimethyl-1-(5-oxohexyl)- 18 |
IUPAC Name | 3,7-dimethyl-1-(5-oxohexyl)purine-2,6-dione 1 |
Synonyms/Other Names | Oxpentifylline, PTX, Trental, Pentoxil, BL 191, 1-(5-Oxohexyl)theobromine 2 |
CAS Number | 6493-05-6 1 |
DrugBank ID | DB00806 1 |
Molecular Formula | C13H18N4O3 2 |
Molar Mass | 278.31 g/mol 1 |
SMILES | CC(=O)CCCCN1C(=O)C2=C(N=CN2C)N(C1=O)C 1 |
InChI | InChI=1S/C13H18N4O3/c1-9(18)6-4-5-7-17-12(19)10-11(14-8-15(10)2)16(3)13(17)20/h8H,4-7H2,1-3H3 1 |
InChIKey | BYPFEZZEUUWMEJ-UHFFFAOYSA-N 1 |
Appearance | White to almost white powder or crystalline solid 13 |
Melting Point | 103.0 to 107.0 °C 14 |
Solubility | Water: ≥43 mg/mL; Soluble in ethanol; Sparingly soluble in toluene 4 |
pKa | 0.50 ± 0.70 (Predicted) 20 |
λmax | 273-276 nm 15 |
Pentoxifylline is a prescription-only medication available globally under a multitude of brand names, reflecting its long history and widespread generic availability.[7] Common brand names include Trental, Pentoxil, Pentopak, and Pentoxifylline SR.[23] Other names used in various markets or literature include Agapurin, Torental, and Rentylin.[19]
The standard pharmaceutical formulation is a 400 mg oral tablet designed for extended-release (ER) or sustained-release (SR).[3] This formulation is critical to its clinical use, as it modulates the drug's release profile to avoid the sharp peaks and troughs in plasma concentration associated with immediate-release versions. This blunted pharmacokinetic profile is intended to improve gastrointestinal tolerability, which is a common issue with methylxanthine derivatives.[4] Patients are instructed to swallow the tablet whole, without crushing or chewing, to preserve this extended-release mechanism.[7]
The regulatory history of pentoxifylline began in Europe, where it has been marketed since 1972.[10] Its entry into the United States market occurred on August 30, 1984, when the U.S. Food and Drug Administration (FDA) approved the extended-release tablet Trental for the treatment of intermittent claudication.[10] Although the original Trental brand marketed by US Pharm Holdings has since been discontinued in the U.S., the market has been supplied by generic versions since the late 1990s from manufacturers such as Upsher-Smith Laboratories (Pentoxil), Valeant Pharmaceuticals, and Apotex.[10] In other countries, such as Canada, a sustained-release formulation from AA Pharma Inc. received authorization on July 10, 2012, underscoring its continued availability as a generic medication internationally.[29]
The therapeutic effects of pentoxifylline stem from a complex and multifaceted mechanism of action that is not yet fully defined.[3] Its actions are not limited to a single pathway but rather involve a constellation of effects on blood cells, inflammatory mediators, and cellular signaling cascades. These combined actions result in its classification as a hemorheologic, anti-inflammatory, and immunomodulatory agent.[3]
The cornerstone of pentoxifylline's activity is its ability to improve the rheological properties of blood, making it less viscous and better able to flow through the microcirculation.[2] This is achieved through several synergistic mechanisms:
At the molecular level, pentoxifylline functions as a competitive, non-selective phosphodiesterase (PDE) inhibitor, a characteristic it shares with other methylated xanthines.[2] By inhibiting PDE enzymes, particularly PDE4, it prevents the breakdown of the second messenger molecule cyclic adenosine monophosphate (cAMP).[2] The resulting increase in intracellular cAMP levels activates Protein Kinase A (PKA), a crucial enzyme that phosphorylates numerous downstream targets, mediating many of pentoxifylline's subsequent physiological effects.[2] This cAMP-PKA signaling pathway is central to the drug's vasodilatory and anti-inflammatory actions.[10]
Beyond its hemorheologic effects, pentoxifylline possesses potent anti-inflammatory and immunomodulatory properties that are increasingly recognized as central to its therapeutic potential in various diseases.[1]
Pentoxifylline contributes to the prevention of thrombosis by inhibiting platelet aggregation.[2] This effect is mediated by two distinct actions: it inhibits the synthesis of thromboxane, a potent platelet aggregator, and simultaneously increases the synthesis of prostacyclin, a platelet inhibitor and vasodilator.[3] Furthermore, pentoxifylline has been identified as an antagonist at adenosine A2 receptors, adding another layer to its complex pharmacology.[2]
The pharmacodynamic outcome of pentoxifylline's multifaceted mechanisms is a net improvement in microcirculatory blood flow and a measurable increase in tissue oxygenation, especially in areas affected by ischemia.[3] The elevation of intracellular cAMP in vascular smooth muscle cells leads to their relaxation, causing vasodilation that complements the hemorheologic effects to further enhance blood supply.[8]
An important pharmacodynamic consideration is the onset of action. The clinical benefits, such as an increase in pain-free walking distance for patients with intermittent claudication, are not immediate. Therapeutic effects typically become noticeable after 2 to 4 weeks of continuous treatment. To fully assess the drug's efficacy, a treatment course of at least 8 weeks is recommended.[6] This delayed onset suggests that the drug's effects are not merely due to acute vasodilation but rely on the gradual remodeling of hemorheologic and inflammatory parameters.
A critical examination of pentoxifylline's pharmacokinetics reveals that the parent drug may function largely as a pro-drug. There is a notable disconnect between the pharmacokinetic profile of pentoxifylline itself and its sustained clinical effects. The parent compound is cleared from the plasma very rapidly, with a half-life of less than an hour, which is seemingly too short to explain a therapeutic benefit from a medication dosed two or three times daily.[3] Furthermore, some of its key mechanisms, such as PDE inhibition, have been shown in vitro to require concentrations far exceeding those achieved in human plasma during therapy.[10]
This apparent paradox can be reconciled by understanding the central role of its pharmacologically active metabolites. Pentoxifylline is extensively metabolized into several compounds, with two major active metabolites, M1 (lisofylline) and M5, achieving plasma concentrations that are 5-fold and 8-fold greater, respectively, than the parent drug.[3] These metabolites also have longer half-lives than pentoxifylline.[3] Crucially, studies have demonstrated that these metabolites exert significant pharmacological activity—such as inhibiting neutrophil superoxide production—at clinically relevant concentrations where the parent drug itself is ineffective.[30] Therefore, the conventional view of pentoxifylline as the sole active agent is incomplete. It is more accurately conceptualized as a delivery system for its more abundant, longer-lasting, and clinically significant active metabolites, which are likely responsible for the majority of the sustained therapeutic effect.
Following oral administration, pentoxifylline is rapidly and almost completely absorbed from the gastrointestinal tract.[3] However, it is subject to extensive first-pass metabolism as it passes through the liver and is also metabolized within erythrocytes.[3] This high first-pass clearance significantly reduces the amount of unchanged drug that reaches systemic circulation, resulting in a low oral bioavailability of only 20% to 30%.[3]
The presence of food in the stomach influences its absorption kinetics. Taking pentoxifylline with meals delays its absorption and the time to reach peak plasma concentration (Tmax) but does not reduce the total amount of drug absorbed (AUC).[4] In fact, co-administration with food can increase the Cmax and AUC of the parent drug and its M1 metabolite.[4] For this reason, and to minimize the risk of gastrointestinal irritation, it is recommended that pentoxifylline be taken with meals.[3]
Pentoxifylline undergoes extensive biotransformation in the body, primarily through reduction and oxidation pathways occurring in the liver and red blood cells.[3] This process yields at least seven known metabolites, with two being of primary clinical and pharmacological importance.[10]
These active metabolites appear in the plasma very soon after administration and, as previously noted, circulate at concentrations significantly higher than the parent drug.[4] The metabolism of pentoxifylline involves the cytochrome P450 enzyme system, specifically as a substrate of CYP1A2. This creates the potential for clinically significant drug interactions with inhibitors (e.g., ciprofloxacin, fluvoxamine) or inducers of this enzyme.[6] Despite its involvement with the P450 system, there is no evidence that pentoxifylline induces its own metabolism following multiple doses.[4]
Information on the distribution of pentoxifylline is limited, but it is known that the parent drug and its M1 metabolite bind to the erythrocyte membrane, the primary site for the conversion of pentoxifylline to M1.[25]
Elimination of pentoxifylline and its metabolites is predominantly renal. Over 95% of an administered dose is excreted in the urine, with essentially no parent drug found unchanged.[2] The main excretory product is Metabolite V.[4] Less than 4% of the dose is recovered in the feces.[3]
The apparent plasma half-life of the parent drug is very short, ranging from 0.4 to 0.8 hours, while the apparent half-lives of its metabolites are longer, at 1.0 to 1.6 hours.[3] The use of an extended-release formulation results in an absorption-limited elimination rate, making the apparent half-life appear longer, in the range of 3 to 5 hours.[35]
Table 2: Summary of Key Pharmacokinetic Parameters for Pentoxifylline and its Major Metabolites
Parameter | Pentoxifylline (Parent Drug) | Metabolite I (M1) | Metabolite V (M5) |
---|---|---|---|
Oral Bioavailability | 20% to 30% 3 | - | - |
Tmax (ER form) | 2 to 4 hours 3 | 2 to 4 hours 35 | ~1.4 hours 35 |
Apparent Half-life | 0.4 to 0.8 hours 3 | 1.0 to 1.6 hours 3 | 1.0 to 1.6 hours 3 |
Relative Plasma Concentration | 1x (Reference) | ~5x greater than parent 4 | ~8x greater than parent 4 |
Pentoxifylline is officially approved by the U.S. FDA for the symptomatic treatment of patients with intermittent claudication resulting from chronic occlusive arterial disease of the limbs.[2] The drug is intended to improve function and alleviate symptoms like pain and cramping, but it is not meant to be a replacement for more definitive treatments such as surgical bypass or angioplasty.[3]
A careful analysis of the clinical evidence reveals a significant disconnect between the drug's broad and potent portfolio of mechanisms—hemorheologic, anti-inflammatory, anti-fibrotic—and its actual clinical performance in this indication. While its mechanisms would theoretically make it an ideal candidate for treating a disease of impaired blood flow and inflammation like IC, its real-world efficacy has been consistently modest and often clinically underwhelming. This suggests that the pathophysiology of walking impairment in IC is either more complex than initially theorized or less responsive to the specific pathways targeted by pentoxifylline. The magnitude of the drug's effect in this context appears insufficient to produce robust clinical benefits, especially when compared to other available therapies.
The evidence supporting pentoxifylline's efficacy in intermittent claudication is mixed and has been criticized for its quality and variability. A major Cochrane systematic review published in 2015, which included 24 studies and over 3,300 participants, found a high degree of heterogeneity among trials.[2] This variability was noted in treatment duration, dosage, and patient characteristics, making it impossible to pool the data for a conclusive meta-analysis.[38] The review concluded that while most individual studies suggested a possible improvement in pain-free and total walking distance compared to placebo, the evidence was of low certainty, and the overall role of pentoxifylline in treating IC remains uncertain.[2] Another large randomized controlled trial (RCT) found that the improvement in walking distance with pentoxifylline was not significantly different from that of placebo.[40]
When compared directly with cilostazol, the other major oral medication for IC, pentoxifylline has been shown to be demonstrably inferior. Multiple head-to-head trials and meta-analyses have solidified this conclusion.[3] A pivotal, multicenter, double-blind RCT involving nearly 700 patients provided clear evidence of this disparity. After 24 weeks of treatment, patients receiving cilostazol experienced a 54% increase in maximal walking distance (an average of 107 meters), which was significantly greater than the 30% increase (64 meters) seen in the pentoxifylline group. Critically, the improvement with pentoxifylline was not statistically superior to the placebo group, which saw a 34% increase.[40] A subsequent network meta-analysis of 29 RCTs further reinforced these findings, ranking cilostazol as the most effective treatment for improving both maximum and pain-free walking distance, with pentoxifylline ranking third, behind beraprost.[42]
Supervised exercise therapy is widely recognized as a cornerstone of management for intermittent claudication and is considered more effective than pharmacotherapy with pentoxifylline.[3] Clinical practice guidelines consistently recommend a structured, supervised exercise program as a first-line intervention for patients with IC.[46] Pharmacological agents like pentoxifylline are typically considered as adjunctive therapies or for patients who are unable to participate in an exercise program.[44]
The clinical inferiority of pentoxifylline is reflected in its standing within major national and international treatment guidelines. This has created a significant divergence between its formal regulatory approval and its recommended use in contemporary clinical practice. While the FDA label, based on data from the 1980s, still sanctions its use for IC, modern, evidence-based guidelines that incorporate comparative effectiveness data have relegated it to a minor or discouraged role.
This situation highlights a critical distinction for clinicians: regulatory approval signifies that a drug has demonstrated a favorable risk-benefit profile (often against placebo) at a specific point in time, but it does not guarantee its place as an optimal or recommended therapy. The emergence of superior treatments, such as cilostazol and well-structured exercise programs, has rendered pentoxifylline's original approval largely outdated from a best-practice standpoint.
Table 3: Head-to-Head Comparison of Pentoxifylline and Cilostazol for Intermittent Claudication
Parameter | Pentoxifylline | Cilostazol |
---|---|---|
Mechanism Class | Hemorheologic Agent, PDE Inhibitor 3 | PDE-3 Inhibitor, Antiplatelet, Vasodilator 52 |
Typical Dosing | 400 mg three times daily with meals 3 | 100 mg twice daily on an empty stomach 41 |
Efficacy (Maximal Walking Distance) | Inferior to cilostazol; often not superior to placebo 3 | Superior to pentoxifylline and placebo 40 |
Efficacy (Pain-Free Walking Distance) | Inferior to cilostazol 42 | Superior to pentoxifylline 42 |
Effect on Ankle-Brachial Index (ABI) | No significant effect [-0.01]42 | Modest improvement [+0.06]42 |
Key Adverse Events | Nausea, dyspepsia, dizziness, headache 7 | Headache, diarrhea, palpitations, dizziness 41 |
User Satisfaction Rating (Drugs.com) | 8.4 / 10 (n=5) 54 | 6.0 / 10 (n=18) 54 |
Contraindications | Recent cerebral/retinal hemorrhage, xanthine allergy 3 | Congestive heart failure of any severity 53 |
Pentoxifylline presents a fascinating case of pharmaceutical repositioning. As its clinical utility in its primary indication has diminished in the face of superior alternatives, its unique pleiotropic effects—particularly its anti-inflammatory and anti-fibrotic properties—have made it an attractive candidate for a wide and diverse array of off-label and investigational applications. This evolution suggests that the drug's future therapeutic value may lie far from its original purpose as a simple hemorheologic agent. The common mechanistic thread linking many of these disparate conditions is not poor blood rheology, but rather chronic inflammation, fibrosis, and ischemia, processes directly targeted by pentoxifylline's ability to inhibit TNF-α and modulate cytokine signaling.
The Scottish Intercollegiate Guidelines Network (SIGN) recommends pentoxifylline as an adjunctive therapy to compression bandaging for the treatment of chronic venous leg ulcers, as evidence has shown it can improve healing rates.[2] It has also been explored for transdermal use in the treatment of cellulite.[2]
One of the most promising off-label applications for pentoxifylline is in the treatment of radiation- and medication-induced bone necrosis. The combination of pentoxifylline with tocopherol (vitamin E), often referred to as the "PENTO" protocol, has demonstrated significant success in healing refractory cases of ORN and MRONJ.[1] These conditions are characterized by severe tissue fibrosis and ischemia. The protocol leverages the synergistic anti-fibrotic, antioxidant, and vasodilatory properties of the two drugs to promote healing in these challenging clinical scenarios.[55]
Pentoxifylline has shown benefit in patients with alcoholic hepatitis, with some studies demonstrating a reduction in the risk of developing the life-threatening complication of hepatorenal syndrome.[2] Its mechanism in this context is thought to be related to its inhibition of TNF-
α and its direct inhibitory actions on hepatic fibrogenesis.[14] While there is some evidence that it can lower biomarkers of inflammation in non-alcoholic steatohepatitis (NASH), its efficacy for this condition is not yet established and requires further study.[2]
The broad mechanistic profile of pentoxifylline has led to its investigation in numerous other fields:
Pentoxifylline is generally considered to be a well-tolerated medication, particularly when administered in its extended-release formulation, which mitigates sharp peaks in plasma concentration.[5] Adverse effects are most commonly gastrointestinal or related to the central nervous system and are often dose-dependent. If these side effects occur, a dose reduction to 400 mg twice daily is often sufficient to improve tolerability; if they persist, discontinuation of the drug may be necessary.[3]
Table 4: Incidence of Common and Notable Adverse Effects of Pentoxifylline
System Organ Class | Adverse Effect | Reported Incidence | Source(s) |
---|---|---|---|
Gastrointestinal | Dyspepsia / Indigestion | ~3% | 9 |
Nausea | ~2-3% | 6 | |
Vomiting | ~1% | 6 | |
Belching / Flatus / Bloating | >1% | 34 | |
Central Nervous System | Dizziness | ~2% | 7 |
Headache | ~1% | 7 | |
Aseptic Meningitis | <1% / Rare | 2 | |
Seizures | Rare / Overdose | 25 | |
Cardiovascular | Flushing | Common | 3 |
Angina / Chest Pain | <1% / Rare | 3 | |
Arrhythmia / Palpitations | <1% / Rare | 2 | |
Hypotension | <1% / Rare | 3 | |
Dermatologic / Hypersensitivity | Rash / Hives / Itching | <0.1% / Rare | 2 |
Anaphylaxis / Angioedema | <1% / Postmarketing | 6 | |
Hematologic | Bleeding / Bruising | Rare | 7 |
Aplastic Anemia / Pancytopenia | <1% / Postmarketing | 6 |
The use of pentoxifylline is contraindicated in specific patient populations where the risks are deemed to outweigh the potential benefits.
Pentoxifylline can participate in several clinically significant drug-drug interactions, primarily related to its metabolism and its pharmacodynamic effects on blood pressure, coagulation, and glucose levels.
Dosing and monitoring adjustments are necessary for certain patient populations to ensure safety and efficacy.
Overdose with pentoxifylline can lead to a range of symptoms, which may occur in a progressive order. Initial signs can include flushing and drowsiness, potentially progressing to hypotension, faintness, unusual excitement, and ultimately, convulsions (seizures), fever, and loss of consciousness.[26]
There is no specific antidote for pentoxifylline overdose. Management is supportive and symptomatic. Treatment strategies include emptying the stomach via gastric lavage and the administration of activated charcoal to limit further absorption. Vital functions, particularly respiration and blood pressure, must be supported.[26]
Pentoxifylline is a pharmacologically complex agent whose clinical profile is best understood by reconciling its in vitro mechanisms with its in vivo pharmacokinetic behavior and observed therapeutic outcomes. The drug's activity is not solely attributable to the parent compound. A deeper analysis shows that pentoxifylline functions largely as a pro-drug, rapidly metabolized into longer-lasting and more abundant active metabolites, M1 and M5, which are likely the primary mediators of its sustained clinical effects. This understanding resolves the apparent contradiction between the parent drug's short half-life and its clinical dosing schedule.
Furthermore, there is a clear mismatch between the drug's powerful and broad-spectrum mechanisms—including potent hemorheologic, anti-inflammatory, and anti-fibrotic actions—and its modest clinical performance in its sole FDA-approved indication of intermittent claudication. This suggests that while mechanistically promising, the magnitude of its effect is insufficient to produce robust benefits in this specific disease, pushing the therapeutic focus towards conditions where its anti-inflammatory and anti-fibrotic properties are more central to the pathophysiology.
In the context of modern vascular medicine, pentoxifylline's position has evolved significantly. For its approved indication of intermittent claudication, it is now largely considered a legacy drug. A significant divergence exists between its regulatory approval, granted decades ago based on superiority over placebo, and its current standing in major clinical practice guidelines. These guidelines, which incorporate modern comparative effectiveness data, consistently favor cilostazol for pharmacotherapy and supervised exercise as the primary intervention. Consequently, the routine prescription of pentoxifylline for intermittent claudication may not align with current evidence-based best practices.
However, this decline in its primary indication has been paralleled by a rise in its investigational use for other complex conditions, illustrating a classic case of pharmaceutical repositioning. The true contemporary value of pentoxifylline appears to lie in its off-label application for niche disorders driven by chronic inflammation and fibrosis. In areas such as osteoradionecrosis (as part of the PENTO protocol), venous leg ulcers, and potentially certain liver diseases, pentoxifylline offers a unique mechanistic approach that is not merely a "me-too" therapy but one that may fill an unmet clinical need. In these contexts, it is not just a "blood flow" agent but a targeted anti-inflammatory and anti-fibrotic drug.
The extensive off-label use and promising preliminary data for pentoxifylline underscore a clear and pressing need for high-quality, adequately powered, multicenter clinical trials to definitively establish its efficacy and safety in these new roles.[12] The existing evidence, while encouraging, is often derived from small studies, case series, or trials with methodological limitations.
Priority areas for future research should include:
A significant barrier to conducting this necessary research is the drug's generic status. Having been off-patent for decades, there is little financial incentive for pharmaceutical companies to fund the large, expensive trials required for regulatory approval in new indications.[12] Overcoming this challenge, perhaps through publicly funded research initiatives, will be critical to fully elucidating and realizing the untapped therapeutic potential of this multifaceted, decades-old molecule.
Published at: July 31, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.