Enfuvirtide

Enfuvirtide (T-20)
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

Enfuvirtide disrupts HIV-1 entry into CD4+ cells by targeting the gp41 envelope glycoprotein:

  • Biomimetic peptide: Mimics the HR-2 domain of gp41, blocking conformational changes required for viral-host membrane fusion .

  • Fusion inhibition: Prevents the formation of an entry pore by binding to HR-1 in gp41, stopping viral capsid transfer .

  • Extracellular activity: Unlike reverse transcriptase or protease inhibitors, enfuvirtide acts outside host cells .

Key pharmacokinetic parameters:

ParameterValue
Plasma protein binding92% (primarily albumin)
Half-life3.8 hours
Volume of distribution5.48 L
MetabolismCatabolized by peptidases

Dose adjustments are unnecessary for age, race, or body weight, except in women <50 kg .

Pivotal Phase III Trials (TORO 1 and TORO 2):

Outcome (24 weeks)Enfuvirtide + OB*OB Alone
HIV RNA reduction (log₁₀)-1.696-0.764
CD4+ cell increase+76 cells/mm³+32 cells/mm³
Virologic failure rate41.7%64.2%

*Optimized Background regimen. At 48 weeks, 55.5% of patients achieved HIV RNA <50 copies/mL .

Synergistic effects:

  • Combined with Sifuvirtide (next-gen fusion inhibitor), potency increased by 240–300% against HIV-1 strains, including enfuvirtide-resistant variants .

Resistance and Evolution

  • Primary resistance: Rare in treatment-naïve patients .

  • Acquired resistance: Mutations in gp41 HR-1 (e.g., V38A, N42D) reduce enfuvirtide binding .

  • Cross-resistance: Low activity against HIV-2 (<i>in vitro</i>) .

Clinical Applications and Guidelines

  • Adults: Recommended for salvage therapy in treatment-experienced patients with multidrug resistance .

  • Children: FDA-approved but no longer recommended in pediatric guidelines due to limited efficacy .

Evolutionary and Therapeutic Implications

  • Viral fitness: Resistant mutants show reduced replication capacity, delaying disease progression .

  • Cost and adherence: High cost (~$20,000/year) and twice-daily injections limit accessibility .

Product Specs

Introduction
Enfuvirtide, also known as human immunodeficiency fusion inhibitor, prevents HIV from infecting healthy CD4 cells. It can decrease the viral load in the blood and boost the CD4 cell count. Enfuvirtide works by disrupting HIV-1's molecular machinery during the final stage of fusion with the target cell, thus preventing uninfected cells from becoming infected. By mimicking components of the HIV-1 fusion apparatus and displacing them, Enfuvirtide prevents normal fusion. HIV attaches to the host CD4+ cell receptor using the viral protein GP120; upon binding, GP120 undergoes conformational changes allowing the viral protein GP41 to insert itself into the host cell's plasma membrane. Entry inhibitors like Enfuvirtide bind to GP41, preventing the formation of an entry pore for the virus's capsid, effectively keeping it out of the cell.
Description
Enfuvirtide possesses the amino acid sequence Ac-Tyr-Thr-Ser-Leu-Ile-His-Ser-Leu-Ile-Glu-Glu-Ser-Gln-Asn-Gln-Gln-Glu-Lys-Asn-Glu-Gln-Glu-Leu-Leu-Glu-Leu-Asp-Lys-Trp-Ala-Ser-Leu-Trp-Asn-Trp-Phe-NH2, a molecular formula of C204H301N51O64, and a molecular weight of 4488.9 Daltons.
Physical Appearance
Sterile Filtered White Lyophilized Powder
Formulation
The protein (1 mg/ml) was lyophilized without any additives.
Solubility
Enfuvirtide lyophilized powder readily dissolves in water, achieving a concentration of 1 mg/ml.
Stability
Lyophilized Enfuvirtide, while stable at room temperature for up to 3 weeks, should be stored desiccated at a temperature below -18°C. After reconstitution, Enfuvirtide should be stored at 4°C for 2-7 days. For future use, store below -18°C. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
Greater than 98.0% as determined by RP-HPLC.

Q&A

What is the precise mechanism of action of Enfuvirtide at the molecular level?

Enfuvirtide is a 36-amino-acid synthetic peptide that targets the HIV-1 envelope glycoprotein gp41. It functions by binding to a specific region of gp41 and disrupting the conformational changes associated with virus-host-cell fusion, thereby blocking virus entry and inhibiting viral replication . More specifically, enfuvirtide binds to gp41 at a transitional pre-fusion conformation, preventing the formation of the six-helix bundle structure required for fusion between viral and cellular membranes .

Unlike other antiretroviral agents that target viral enzymes (reverse transcriptase, protease, integrase), enfuvirtide blocks the final step in the three-step viral entry process consisting of attachment, co-receptor binding, and fusion . This unique mechanism of action makes it valuable for treating HIV strains resistant to other antiretroviral drug classes.

How does Enfuvirtide pharmacokinetics influence research study design?

When designing research protocols involving enfuvirtide, researchers must consider several pharmacokinetic factors:

  • Administration route: Enfuvirtide requires subcutaneous administration, typically twice daily . This administration route impacts study design, particularly for longitudinal investigations.

  • Absorption characteristics: As a peptide, enfuvirtide has variable absorption rates from subcutaneous tissues, with bioavailability ranging from 80-90%.

  • Distribution: Enfuvirtide has limited penetration into the central nervous system, which must be considered when studying its efficacy against HIV in neurological reservoirs.

  • Metabolism and elimination: Unlike small-molecule antiretrovirals metabolized primarily by hepatic enzymes, enfuvirtide undergoes catabolism to constituent amino acids, resulting in minimal drug-drug interactions.

  • Half-life considerations: With a relatively short half-life requiring twice-daily dosing, researchers must account for potential adherence challenges in clinical studies.

What are the molecular determinants of Enfuvirtide resistance and how can they be characterized experimentally?

Enfuvirtide resistance primarily develops through mutations in a specific 10-amino-acid motif located between residues 36 and 45 in the HIV gp41 protein . This region forms part of the binding site for enfuvirtide and is critical for viral fusion.

Experimental approaches for characterizing resistance include:

  • Genotypic analysis: Sequencing the gp41 region to identify known resistance mutations

  • Phenotypic assays: Comparing IC50 values of enfuvirtide against patient isolates versus reference strains

  • Site-directed mutagenesis: Creating specific mutations to evaluate their impact on enfuvirtide binding

  • Structural studies: Using techniques like X-ray crystallography and cryo-electron microscopy to visualize how resistance mutations alter gp41 structure

Key findings about enfuvirtide resistance include:

  • Enfuvirtide-resistant mutants typically show reduced replicative capacity compared to wild-type virus

  • Reversion to wild-type, drug-sensitive phenotype has been observed following enfuvirtide withdrawal

  • Primary resistance in treatment-naïve patients is rare, with clinical isolates generally remaining sensitive

How do mutations in gp41 affect the binding kinetics and thermodynamics of Enfuvirtide interaction?

Resistance mutations typically alter the kinetics and thermodynamics of enfuvirtide binding through several mechanisms:

  • Reduced binding affinity: Mutations can directly disrupt hydrogen bonds, electrostatic interactions, or hydrophobic contacts between enfuvirtide and gp41

  • Altered conformational dynamics: Some mutations accelerate the rate of gp41 conformational changes, reducing the window of opportunity for enfuvirtide binding

  • Steric hindrance: Certain mutations introduce bulkier side chains that physically impede enfuvirtide access to its binding site

Experimental approaches to quantify these effects include:

  • Surface plasmon resonance (SPR) to determine association and dissociation rate constants

  • Isothermal titration calorimetry (ITC) to measure binding enthalpy and entropy

  • Fluorescence-based binding assays to determine equilibrium dissociation constants

What methodological approaches should be employed when studying Enfuvirtide in treatment-experienced patients?

When designing clinical research involving enfuvirtide in treatment-experienced patients, researchers should consider:

  • Optimized background regimen: In pivotal trials, enfuvirtide was added to an individually optimized background regimen consisting of 3-5 antiretroviral drugs selected based on resistance testing and treatment history .

  • Patient selection criteria: The ideal study population consists of individuals with documented resistance to multiple antiretroviral classes and limited treatment options .

  • Adherence support strategies: Given the twice-daily subcutaneous administration requirement, studies should incorporate robust adherence support and monitoring.

  • Resistance monitoring protocols: Regular genotypic and phenotypic resistance testing of the gp41 region is essential.

  • Injection site reaction assessment: Studies should include standardized methods for documenting and grading injection site reactions, which occur in up to 98% of patients .

The TORO-1 and TORO-2 trials (T-20 vs. Optimized Regimen Only) provide a methodological template, demonstrating that enfuvirtide plus an optimized background regimen achieves superior virological and immunological outcomes compared to optimized background alone .

How should researchers address the challenge of injection site reactions in Enfuvirtide studies?

Injection site reactions (ISRs) represent the most common adverse effect of enfuvirtide therapy, occurring in approximately 98% of patients . For researchers studying enfuvirtide, addressing this challenge requires:

  • Standardized assessment tools: Implement validated scales for documenting ISR severity, type, and duration

  • Mitigation strategies: Evaluate techniques such as:

    • Rotation of injection sites

    • Controlled injection depth and speed

    • Massage after injection

    • Temperature modification of the solution

  • Alternative delivery approaches: Consider novel delivery systems like hydrogel-forming microneedles, which have shown promise in ex vivo studies with maximum permeation of 36.26% compared to 28.45% with conventional delivery systems .

  • Impact analysis: Assess how ISRs affect adherence, quality of life, and ultimately, virological outcomes

  • Correlation studies: Investigate patient factors that may predict ISR severity or duration

What are the optimal analytical methods for quantifying Enfuvirtide in biological samples?

Accurate quantification of enfuvirtide in biological samples is critical for pharmacokinetic studies, therapeutic drug monitoring, and research applications. Recommended analytical approaches include:

  • Liquid chromatography-mass spectrometry (LC-MS/MS):

    • Offers high sensitivity and specificity for peptide quantification

    • Typical lower limit of quantification: 5-10 ng/mL in plasma

    • Sample preparation typically involves protein precipitation followed by solid-phase extraction

  • Enzyme-linked immunosorbent assay (ELISA):

    • Useful for high-throughput screening

    • May have cross-reactivity challenges with metabolites or degradation products

    • Typically less sensitive than LC-MS/MS

  • High-performance liquid chromatography (HPLC) with UV detection:

    • Less sensitive than LC-MS/MS but more accessible

    • Typically requires larger sample volumes

    • Useful for formulation analysis and stability studies

Sample collection and handling considerations include:

  • Immediate processing or storage at -80°C to prevent peptide degradation

  • Use of protease inhibitors in collection tubes when appropriate

  • Validation of freeze-thaw stability

What in vitro models best predict the clinical efficacy of Enfuvirtide?

Several in vitro models have been developed to evaluate enfuvirtide activity, each with specific applications:

  • Cell-cell fusion assays:

    • Measure inhibition of fusion between cells expressing HIV envelope proteins and CD4/co-receptors

    • Allow high-throughput screening but may not fully recapitulate the viral entry process

  • Single-cycle infection assays:

    • Utilize pseudotyped viruses expressing HIV envelope proteins

    • Provide quantitative measure of entry inhibition

    • Useful for comparing activity against diverse HIV strains

  • Multi-cycle replication assays:

    • Measure viral replication in the presence of enfuvirtide over multiple rounds of infection

    • More physiologically relevant but lower throughput

    • Allow assessment of resistance emergence

  • Ex vivo models using primary cells:

    • Peripheral blood mononuclear cells or lymphoid tissue explants

    • Closer approximation of in vivo conditions

    • Useful for evaluating activity against primary isolates

Correlation studies between in vitro IC50/IC90 values and clinical outcomes suggest that a 10-fold increase in IC50 is associated with significantly reduced virological response.

What are the key considerations in developing alternative delivery systems for Enfuvirtide?

Research into novel delivery systems for enfuvirtide aims to overcome limitations of twice-daily subcutaneous injections. Key research considerations include:

  • Hydrogel-forming microneedles: This approach has shown significant promise, with recent Quality by Design (QbD) studies optimizing formulation parameters including:

    • PEGdiacid ratio

    • Cross-linking temperature

    • Cross-linking time

    The optimized formulation achieved a desirability index of 0.871 and demonstrated superior mechanical integrity and controlled release kinetics .

  • Critical quality attributes for delivery systems:

    • Stability of enfuvirtide within the delivery matrix

    • Controlled release kinetics

    • Maintenance of peptide structural integrity

    • Biocompatibility and local tissue tolerance

    • Manufacturing scalability and reproducibility

  • Evaluation metrics:

    • Ex vivo permeation studies

    • In vivo pharmacokinetic profiles

    • Local tolerability assessments

    • Stability studies under various storage conditions

    • Maintenance of antiviral activity

How does the potency of Enfuvirtide compare when delivered through alternative routes?

When evaluating alternative delivery routes for enfuvirtide, researchers must consider several factors that affect potency:

  • Bioavailability comparison:

    • Subcutaneous (reference): ~80-90% bioavailability

    • Transdermal (microneedle): Varies based on formulation, with promising results showing 36.26% permeation using optimized 11×11 microneedle arrays

    • Oral delivery attempts: Generally poor bioavailability due to peptide degradation in the GI tract

    • Buccal/sublingual approaches: Intermediate bioavailability, requires permeation enhancers

  • Pharmacokinetic profile differences:

    • Subcutaneous: Relatively rapid absorption with Tmax of 4-8 hours

    • Transdermal systems: Typically extended release with lower Cmax but sustained concentrations

    • Implantable systems: Potential for zero-order release kinetics

  • Local tissue factors affecting delivery:

    • Subcutaneous tissue composition and blood flow

    • Skin barrier function for transdermal delivery

    • Mucosal permeability for alternative routes

What are the methodological challenges in evaluating Enfuvirtide against diverse HIV-1 strains?

Researchers evaluating enfuvirtide against diverse HIV-1 strains face several methodological challenges:

  • Standardization of susceptibility testing:

    • Different assay formats yield varying IC50 values

    • Phenotypic assays may use different cell lines and readouts

    • Need for appropriate reference strains for normalization

  • Viral diversity considerations:

    • Natural polymorphisms in gp41 across HIV-1 clades affect baseline susceptibility

    • Patient-derived isolates may contain mixtures of variants

    • Tropism (R5 vs. X4) may influence enfuvirtide susceptibility

  • Correlation between genotype and phenotype:

    • Complex patterns of primary and compensatory mutations

    • Need for algorithms to predict phenotypic resistance from genotypic data

    • Threshold determination for clinically significant resistance

  • Technical approaches:

    • Clonal versus population-based analysis

    • Deep sequencing to detect minor resistant variants

    • Standardization of cutoffs for resistance interpretation

Researchers should implement quality control measures including reference strains with known enfuvirtide susceptibility and inter-laboratory standardization.

How can researchers effectively model the kinetics of Enfuvirtide resistance emergence?

Modeling the kinetics of enfuvirtide resistance emergence requires sophisticated experimental and computational approaches:

  • In vitro selection experiments:

    • Serial passage in increasing enfuvirtide concentrations

    • Monitoring of resistance emergence through regular genotypic and phenotypic testing

    • Comparison of resistance pathways in different viral backgrounds

  • Mathematical modeling approaches:

    • Stochastic models of mutation accumulation

    • Population dynamics models incorporating fitness costs

    • Bayesian evolutionary models to infer resistance pathways

  • Clinical data integration:

    • Longitudinal sampling from patients on enfuvirtide therapy

    • Correlation of pharmacokinetic parameters with resistance emergence

    • Analysis of adherence patterns and their impact on resistance

  • Predictive factors to measure:

    • Baseline gp41 sequence polymorphisms

    • Pre-existing minority variants

    • Viral load at treatment initiation

    • Background regimen potency

These approaches can help predict the probability and timing of resistance emergence, guiding clinical decision-making and treatment optimization.

What are the optimal experimental designs for studying Enfuvirtide in combination with other antiretrovirals?

When designing studies of enfuvirtide in combination with other antiretrovirals, researchers should consider:

  • Interaction analysis methods:

    • Checkerboard assays to determine combination indices

    • Isobologram analysis to characterize additive, synergistic, or antagonistic effects

    • Concentration-response surface modeling

  • Endpoint selection:

    • Short-term: viral inhibition in single-cycle or multi-cycle assays

    • Medium-term: emergence of resistance

    • Long-term: durability of response and evolution of viral populations

  • Specific combination contexts to evaluate:

    • With entry inhibitors targeting different steps (attachment inhibitors, CCR5 antagonists)

    • With agents from traditional antiretroviral classes (NRTIs, NNRTIs, PIs, INSTIs)

    • With novel agents in development

  • Resistance evaluations:

    • Single versus dual resistance selection

    • Cross-resistance patterns

    • Genetic barrier to resistance for the combination

Pivotal clinical trials demonstrated that enfuvirtide added to an optimized background regimen provided superior virological and immunological activity compared to the optimized background alone, with durable responses confirmed at 48 weeks .

How does Enfuvirtide interact with novel broadly neutralizing antibodies targeting HIV envelope proteins?

The interaction between enfuvirtide and broadly neutralizing antibodies (bNAbs) targeting HIV envelope proteins represents an emerging research area:

  • Mechanistic interactions:

    • Enfuvirtide targets gp41 fusion intermediates

    • bNAbs target various epitopes on gp120 and gp41

    • Potential for complementary or competitive binding depending on epitope

  • Experimental approaches:

    • Binding competition assays to determine interference or enhancement

    • Sequential versus simultaneous addition experiments

    • Resistance profiling to identify shared or distinct escape pathways

  • Synergy evaluations:

    • Combination index determination for different bNAb classes

    • Analysis of resistance barrier elevation

    • Potential for dose reduction of one or both agents

  • Translational implications:

    • Combination strategies for therapeutic applications

    • Potential for preventing viral escape

    • Applications in cure strategies targeting the latent reservoir

What are the most promising structural modifications to Enfuvirtide being explored to improve its pharmacokinetic profile?

Researchers are investigating several structural modifications to enhance enfuvirtide's properties:

  • Half-life extension strategies:

    • Lipidation: Addition of fatty acid chains to extend half-life

    • PEGylation: Attachment of polyethylene glycol to reduce renal clearance

    • Fc-fusion constructs: Fusion with immunoglobulin Fc region to enable FcRn recycling

  • Stability enhancements:

    • D-amino acid substitutions: To resist proteolytic degradation

    • Cyclization approaches: To constrain peptide conformation

    • Terminal modifications: To protect against exopeptidase activity

  • Conformational optimization:

    • Stapled peptide technology: To stabilize α-helical conformations

    • Non-natural amino acid incorporation: To enhance binding affinity

    • Backbone modifications: To improve stability while maintaining activity

These modifications aim to maintain or enhance binding to the target gp41 region while improving pharmacokinetic properties to enable less frequent dosing.

What emerging technologies might transform research on HIV fusion inhibitors like Enfuvirtide?

Emerging technologies with potential to transform HIV fusion inhibitor research include:

  • Advanced structural biology techniques:

    • Cryo-electron microscopy for visualizing fusion intermediates

    • Single-molecule FRET to track conformational dynamics in real-time

    • Hydrogen-deuterium exchange mass spectrometry to map binding interfaces

  • Novel delivery platforms:

    • Implantable long-acting delivery systems

    • Targeted nanoparticle approaches

    • Cell-based delivery systems

  • Genetic engineering approaches:

    • Vectored immunoprophylaxis for sustained production

    • CRISPR-based approaches to modify host cell susceptibility

    • Synthetic biology platforms for novel inhibitor design

  • Computational methods:

    • Machine learning for prediction of resistance pathways

    • Molecular dynamics simulations for rational design

    • Systems biology approaches to understand viral-host interactions

These technologies have the potential to address current limitations in enfuvirtide therapy and expand the application of fusion inhibitors in HIV treatment and prevention.

Product Science Overview

Development and History

Enfuvirtide was developed by researchers at Duke University, who later formed a pharmaceutical company called Trimeris. The development of enfuvirtide began in 1996, and it was initially designated as T-20. In 1999, Trimeris partnered with Hoffmann-La Roche to complete the development of the drug .

Mechanism of Action

Enfuvirtide is a 36-amino acid biomimetic peptide that mimics the structure of HIV proteins responsible for the fusion of the virus to cell membranes. The drug works by binding to the gp41 transmembrane protein of the HIV envelope, which is crucial for the fusion of the viral and host cell membranes. By interfering with the conformational changes required for membrane fusion, enfuvirtide effectively blocks the entry of the virus into the host cell .

Clinical Significance

Enfuvirtide represents the first of a new class of antiretroviral drugs known as fusion inhibitors. Its development marked a significant advancement in the treatment of HIV, particularly for patients who have developed resistance to other classes of antiretroviral drugs. The introduction of enfuvirtide provided a new option for heavily treated patients with advanced HIV disease .

Pharmacokinetics

Enfuvirtide is administered subcutaneously and has a bioavailability of approximately 84.3%. It is highly protein-bound (92%) and is metabolized in the liver. The elimination half-life of enfuvirtide is around 3.8 hours .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.