PT-141 (Bremelanotide) Manufacturing Profile: Complete Technical Specifications and Quality Control Parameters
1. Introduction to PT-141 Manufacturing
PT-141 (Bremelanotide), a synthetic peptide derivative of α-melanocyte-stimulating hormone (α-MSH), represents a significant advancement in melanocortin receptor agonist manufacturing. As a cyclic heptapeptide with the sequence Ac-Nle-cyclo[Asp-His-D-Phe-Arg-Trp-Lys]-OH, PT-141 requires precise synthesis protocols and rigorous quality control measures to achieve pharmaceutical-grade specifications.
The molecular formula C50H68N14O10 with a molecular weight of 1025.18 g/mol presents specific manufacturing challenges related to peptide cyclization, stereochemical integrity, and stability maintenance. This technical profile provides comprehensive manufacturing guidance for production facilities engaged in peptide synthesis operations, covering solid-phase peptide synthesis (SPPS) protocols, purification methodologies, analytical verification methods, and stability considerations critical for commercial-scale production.
Manufacturing PT-141 to pharmaceutical standards demands adherence to Current Good Manufacturing Practice (cGMP) guidelines, implementation of validated analytical methods, and comprehensive documentation systems. The presence of the D-Phe residue at position 4 and the cyclic structure connecting Asp-2 to Lys-7 requires specialized synthesis expertise and quality control protocols beyond standard linear peptide manufacturing processes.
2. Solid-Phase Peptide Synthesis Protocol
2.1 Resin Selection and Loading
PT-141 synthesis initiates with appropriate resin selection based on the C-terminal structure. Rink amide MBHA resin (0.4-0.7 mmol/g substitution) serves as the preferred solid support for PT-141 production, providing the required C-terminal amide functionality while maintaining adequate swelling properties in synthesis solvents. Alternative resins including Wang resin or 2-chlorotrityl chloride resin may be employed depending on specific manufacturing requirements and downstream processing considerations.
Resin loading procedures follow standardized protocols with Fmoc-Lys(Boc)-OH attachment as the first amino acid. Loading efficiency should achieve 85-95% as verified by quantitative Fmoc determination using UV spectroscopy at 301 nm following piperidine deprotection. Incomplete loading results in sequence truncations that complicate purification and reduce overall yield metrics.
2.2 Linear Sequence Assembly
Amino acid coupling proceeds using Fmoc-strategy SPPS with the following optimized sequence: Fmoc-Lys(Boc)-OH → Fmoc-Trp(Boc)-OH → Fmoc-Arg(Pbf)-OH → Fmoc-D-Phe-OH → Fmoc-His(Trt)-OH → Fmoc-Asp(OtBu)-OH → Fmoc-Nle-OH. Each coupling cycle employs 3-4 molar equivalents of protected amino acid activated with HBTU/HOBt (3-4 eq) in the presence of DIEA (6-8 eq) in DMF solvent.
| Amino Acid Position | Protected Residue | Coupling Reagent | Equivalents | Reaction Time (min) | Coupling Efficiency (%) |
|---|---|---|---|---|---|
| Position 1 (Lys) | Fmoc-Lys(Boc)-OH | HBTU/HOBt | 4.0 | 60 | ≥98.5 |
| Position 2 (Trp) | Fmoc-Trp(Boc)-OH | HBTU/HOBt | 4.0 | 90 | ≥97.5 |
| Position 3 (Arg) | Fmoc-Arg(Pbf)-OH | HBTU/HOBt | 4.0 | 90 | ≥98.0 |
| Position 4 (D-Phe) | Fmoc-D-Phe-OH | HBTU/HOBt | 3.5 | 75 | ≥98.5 |
| Position 5 (His) | Fmoc-His(Trt)-OH | HBTU/HOBt | 4.0 | 120 | ≥97.0 |
| Position 6 (Asp) | Fmoc-Asp(OtBu)-OH | HBTU/HOBt | 3.5 | 60 | ≥98.5 |
| Position 7 (Nle) | Fmoc-Nle-OH | HBTU/HOBt | 3.5 | 60 | ≥98.5 |
Critical attention must be directed toward His and Trp coupling steps, which demonstrate reduced reactivity compared to other residues. Extended coupling times (90-120 minutes) and elevated temperatures (40-50°C) may be required to achieve target coupling efficiencies above 97%. Kaiser test or chloranil test monitoring after each coupling verifies completion before proceeding to the next Fmoc deprotection cycle.
2.3 N-Terminal Acetylation
Following assembly of the linear heptapeptide sequence, N-terminal acetylation proceeds using acetic anhydride (10 eq) and DIEA (10 eq) in DMF for 30 minutes. This modification is essential for PT-141 biological activity and must achieve >99% conversion as verified by analytical HPLC of a small cleaved sample. Alternative acetylation reagents including acetyl chloride or pre-formed acetyl-imidazole may be employed depending on manufacturing preferences and equipment capabilities.
2.4 On-Resin Cyclization
Peptide cyclization represents the most critical step in PT-141 manufacturing, forming the lactam bridge between the Asp side chain carboxyl group and the Lys side chain amino group. Following Fmoc removal from the N-terminal Nle and selective deprotection of Asp(OtBu) and Lys(Boc) using 2% TFA in DCM (multiple 5-minute treatments), on-resin cyclization proceeds using PyBOP or HATU activation.
Optimal cyclization conditions employ PyBOP (1.5 eq relative to resin loading), HOBt (1.5 eq), and DIEA (3 eq) in DMF at high dilution (resin concentration <0.01 M) for 4-6 hours. The extended reaction time and dilute conditions minimize intermolecular coupling (dimer/oligomer formation) while promoting intramolecular cyclization. Cyclization efficiency should exceed 85% as determined by analytical HPLC following test cleavage, with linear precursor remaining below 10% of total peptide content.
3. Cleavage and Deprotection Procedures
3.1 TFA Cleavage Cocktail
Global deprotection and resin cleavage employs a TFA-based cocktail designed to remove all side chain protecting groups while minimizing oxidation and other side reactions. The standard cleavage mixture consists of TFA (94%), water (2.5%), triisopropylsilane (TIS, 2.5%), and ethanedithiol (EDT, 1%) for 2-3 hours at room temperature.
The presence of Trp in the PT-141 sequence necessitates scavenger incorporation to prevent alkylation and oxidation. TIS serves as the primary carbocation scavenger, while EDT protects Trp from formylation and other electrophilic modifications. Water facilitates Arg(Pbf) and His(Trt) deprotection through acidolysis mechanisms. Cleavage cocktail volumes typically range from 10-20 mL per gram of resin to ensure adequate reagent excess and mixing throughout the deprotection period.
3.2 Crude Peptide Precipitation and Washing
Following cleavage completion, the resin is removed by filtration and the TFA filtrate is added dropwise to cold diethyl ether (10-15 volumes) to precipitate the crude peptide. The precipitate is collected by centrifugation (3000-4000 rpm, 10 minutes), and the ether supernatant is decanted. This precipitation/washing cycle is repeated 3-4 times using fresh cold ether to remove scavengers, protecting group fragments, and other small molecule impurities.
The final crude peptide pellet is dried under vacuum or nitrogen stream until residual ether is removed, yielding a white to off-white powder. Typical crude yields range from 40-60% based on initial resin loading, with crude purity by HPLC area percentage typically 50-75% depending on synthesis efficiency and sequence-related deletion peptides.
| Impurity Type | Description | Typical Range (%) | Source |
|---|---|---|---|
| Deletion sequences | Des-amino acid variants | 5-15 | Incomplete coupling |
| Linear precursor | Non-cyclized peptide | 3-10 | Incomplete cyclization |
| Oxidation products | Trp, Met oxidation | 2-8 | TFA cleavage conditions |
| Epimers/isomers | D-Phe racemization | 1-3 | Coupling/cyclization |
| Dimers/oligomers | Intermolecular coupling | 2-6 | Cyclization step |
| Truncations | Incomplete sequences | 3-8 | Premature termination |
4. Purification Methodology
4.1 Preparative RP-HPLC Strategy
PT-141 purification to pharmaceutical specifications requires preparative reversed-phase high-performance liquid chromatography (RP-HPLC) using C18 silica columns (particle size 10-15 μm, pore size 100-300 Å). Column dimensions for production-scale purification typically range from 5-30 cm diameter with lengths of 25-50 cm, providing theoretical plate numbers sufficient for resolution of closely eluting impurities.
The mobile phase system employs aqueous trifluoroacetic acid (0.1% TFA in water, Mobile Phase A) and acetonitrile with 0.1% TFA (Mobile Phase B). PT-141 elution occurs at approximately 25-35% acetonitrile depending on column characteristics, temperature, and flow rate parameters. A shallow gradient (0.5-1.0% acetonitrile/minute) in the 20-40% B range provides optimal separation between PT-141 and sequence-related impurities.
4.2 Purification Protocol and Fraction Collection
Crude peptide dissolution in minimal mobile phase A with addition of 10-20% mobile phase B ensures complete solubilization before loading onto the preparative column. Sample loading should not exceed 5-10% of column volume per injection to maintain resolution and avoid column overloading effects. UV detection at 214 nm and 280 nm enables peak tracking, with fraction collection triggered by UV threshold values and manual verification of peak position.
Collected fractions undergo analytical HPLC verification before pooling decisions. Only fractions meeting purity specifications (typically ≥95% by HPLC area percentage) are combined for lyophilization. Fractions containing intermediate purity (85-95%) may be subjected to additional purification cycles, while low-purity fractions (<85%) are typically reprocessed or discarded depending on economic considerations.
| Parameter | Specification | Notes |
|---|---|---|
| Column Type | C18, 10-15 μm, 100-300 Å | Reversed-phase silica |
| Column Dimensions | 5-30 cm ID × 25-50 cm L | Scale-dependent |
| Mobile Phase A | 0.1% TFA in water | HPLC-grade water |
| Mobile Phase B | 0.1% TFA in acetonitrile | HPLC-grade acetonitrile |
| Flow Rate | 50-500 mL/min | Column diameter dependent |
| Gradient | 15-45% B over 40-60 min | 0.5-1.0% B/min in critical region |
| Detection | 214 nm, 280 nm | Dual wavelength monitoring |
| Column Temperature | 25-40°C | Temperature control recommended |
| Sample Load | ≤10% column volume | Prevents overloading |
| Injection Volume | Variable by scale | Dissolved in 10-20% B |
4.3 Lyophilization Process
Purified fractions are subjected to lyophilization (freeze-drying) to remove solvents and produce stable solid product. Prior to freezing, acetonitrile concentration should be reduced to <5% through rotary evaporation or nitrogen blow-down to facilitate proper freezing and prevent product loss during the sublimation phase. Dilute aqueous fractions may be frozen directly or concentrated to reduce lyophilization time and energy consumption.
The lyophilization cycle consists of freezing (typically -40 to -50°C for 2-4 hours), primary drying (temperature ramped from -40°C to 0°C over 24-48 hours under vacuum 50-200 mTorr), and secondary drying (0-25°C for 12-24 hours). Residual moisture content in the final lyophilized cake should not exceed 3-5% by Karl Fischer titration. Proper lyophilization yields a white to off-white powder or cake that readily dissolves upon reconstitution.
4.4 Desalting Procedures
TFA counter-ions from the purification mobile phase result in PT-141 trifluoroacetate salt form, which may comprise 20-40% of the lyophilized mass depending on the number of basic residues and TFA concentration. For applications requiring reduced TFA content, desalting procedures may be implemented through ion exchange chromatography or HCl salt conversion.
HCl salt conversion involves dissolving the TFA salt in water, passing through a strong anion exchange resin in chloride form, and lyophilizing the eluent. This process exchanges TFA anions for chloride, reducing overall salt mass and providing a defined chloride salt form. Alternatively, additional RP-HPLC purification using low TFA concentrations (0.01-0.05%) or alternative ion-pairing agents (HCl, acetic acid) can reduce TFA content in the final product.
5. Analytical Quality Control Methods
5.1 Identity Verification
PT-141 identity confirmation employs multiple orthogonal analytical techniques to verify molecular structure, sequence, and cyclization. Mass spectrometry serves as the primary identity test, with electrospray ionization mass spectrometry (ESI-MS) or matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) confirming the expected molecular weight of 1025.18 Da (free base) or adjusted for salt forms.
High-resolution mass spectrometry (HRMS) provides elemental composition verification through accurate mass measurement within 5 ppm tolerance. The observed m/z values for [M+H]+ at 1026.5, [M+2H]2+ at 513.8, and [M+3H]3+ at 342.9 confirm PT-141 identity with high confidence. Fragmentation patterns from MS/MS experiments provide sequence confirmation through identification of b- and y-ion series corresponding to the expected heptapeptide structure.
5.2 Purity Assessment by HPLC
Analytical RP-HPLC serves as the primary purity determination method for PT-141 release testing and stability studies. The method employs a C18 column (4.6 × 150-250 mm, 5 μm particle size) with gradient elution using 0.1% TFA in water (Mobile Phase A) and 0.1% TFA in acetonitrile (Mobile Phase B). A linear gradient from 10-60% B over 30-40 minutes at 1.0-1.5 mL/min flow rate provides adequate resolution of PT-141 from related impurities.
UV detection at 214 nm measures total peptide content through peptide bond absorption, while detection at 280 nm provides complementary information based on Trp and Phe aromatic absorption. Purity calculation employs area percentage normalization, with acceptance criteria typically set at ≥95.0% for pharmaceutical-grade material and ≥98.0% for reference standard applications.
| Parameter | Specification |
|---|---|
| Column | C18, 4.6 × 250 mm, 5 μm, 100 Å |
| Mobile Phase A | 0.1% TFA in water |
| Mobile Phase B | 0.1% TFA in acetonitrile |
| Flow Rate | 1.0 mL/min |
| Column Temperature | 30°C |
| Detection Wavelength | 214 nm (primary), 280 nm (secondary) |
| Injection Volume | 10-20 μL |
| Sample Concentration | 0.5-1.0 mg/mL |
| Gradient Program | 10-60% B over 40 minutes |
| Run Time | 50 minutes (including re-equilibration) |
5.3 Amino Acid Analysis
Quantitative amino acid analysis (AAA) provides composition verification and content determination for PT-141. The peptide undergoes hydrolysis in 6 N HCl at 110°C for 20-24 hours under nitrogen or vacuum to prevent oxidative degradation. Following hydrolysis, amino acids are derivatized using phenylisothiocyanate (PITC), o-phthalaldehyde (OPA), or ninhydrin reagents before RP-HPLC or ion exchange chromatography separation.
Expected amino acid ratios for PT-141 are Asp (1.0), His (1.0), Arg (1.0), Trp (1.0), Phe (1.0), Lys (1.0), with norleucine (Nle) requiring specialized detection since it co-elutes with leucine in standard methods. Trp quantification presents challenges due to partial destruction during acid hydrolysis, necessitating alternative hydrolysis conditions (4 N methanesulfonic acid or base hydrolysis) for accurate Trp determination.
5.4 Peptide Content Determination
Absolute peptide content quantification accounts for moisture, salt, and residual solvent content to report actual peptide mass per total product mass. This multi-step process combines UV spectrophotometry or amino acid analysis for peptide quantification with Karl Fischer titration for moisture determination and ion chromatography or titration for counter-ion quantification.
UV spectrophotometry at 280 nm utilizes Trp absorption (ε280 = 5,500 M⁻¹cm⁻¹ per Trp residue) to calculate peptide concentration in solution. This value, combined with moisture content (typically 2-5%), TFA content (5-15% for TFA salt), and other impurities, enables calculation of peptide content on an anhydrous, salt-free basis. Pharmaceutical specifications typically require ≥85% peptide content on an as-is basis or ≥95% on a dried, salt-free basis.
5.5 Stereochemical Purity
The presence of D-Phe at position 4 in PT-141 requires verification of stereochemical integrity, as racemization during synthesis or purification would generate diastereomeric impurities affecting biological activity. Chiral HPLC methods employing β-cyclodextrin-modified stationary phases or specialized chiral columns can resolve D-Phe-containing PT-141 from L-Phe epimers.
Alternatively, amino acid analysis following peptide hydrolysis using chiral derivatization reagents (Marfey's reagent, FDAA) enables detection of L-Phe at position 4, which would indicate racemization. Acceptance criteria typically limit epimeric impurities to <1.0% to ensure biological activity specifications are met.
5.6 Residual Solvent Analysis
Residual solvents from synthesis and purification must be quantified and controlled according to ICH Q3C guidelines. Gas chromatography with flame ionization detection (GC-FID) or headspace GC-MS methods detect and quantify residual DMF, DCM, TFA, acetonitrile, diethyl ether, and other solvents potentially present in the final product.
Class 2 solvents (acetonitrile, DCM, DMF) are limited to concentrations defined by Permitted Daily Exposure (PDE) values, typically 410 ppm for acetonitrile, 600 ppm for DCM, and 880 ppm for DMF. Class 3 solvents (TFA, acetic acid, ethanol) have limits of 5000 ppm. Properly executed lyophilization should reduce all residual solvents to well below these thresholds, with typical values <100 ppm for most solvents.
5.7 Bacterial Endotoxin Testing
For parenteral applications, PT-141 must meet bacterial endotoxin specifications as determined by Limulus Amebocyte Lysate (LAL) testing according to USP <85> Bacterial Endotoxins Test. The endotoxin limit is calculated based on the maximum dose and route of administration, typically specified as <0.5 EU/mg for peptide pharmaceuticals intended for injection.
The kinetic chromogenic LAL method provides quantitative endotoxin determination with detection limits below 0.005 EU/mL. Sample preparation involves dissolution in endotoxin-free water followed by appropriate dilution to fall within the assay's valid range while avoiding interference from the peptide or formulation components. Manufacturing under cGMP conditions with proper environmental controls and use of depyrogenated equipment and endotoxin-free materials ensures endotoxin levels remain below specification limits.
6. Batch Release Specifications
Pharmaceutical-grade PT-141 batch release requires comprehensive testing against established specifications covering identity, purity, potency, and safety parameters. These specifications are derived from compendial standards, regulatory guidance, and manufacturing capability studies demonstrating process consistency and control.
| Test | Method | Acceptance Criteria |
|---|---|---|
| Appearance | Visual inspection | White to off-white lyophilized powder |
| Identity | ESI-MS or MALDI-TOF MS | [M+H]+ = 1026.5 ± 1.0 Da |
| Identity | Analytical HPLC (retention time) | Retention time matches reference standard ± 2% |
| Purity (HPLC) | RP-HPLC (214 nm) | ≥95.0% (area percentage) |
| Single Impurity | RP-HPLC (214 nm) | ≤3.0% (individual impurity) |
| Amino Acid Composition | AAA (post-hydrolysis) | Asp, His, Arg, Trp, Phe, Lys = 1.0 ± 0.15 |
| Peptide Content | UV spectrophotometry or AAA | ≥85.0% (as-is basis) |
| Water Content | Karl Fischer titration | ≤5.0% |
| TFA Content | Ion chromatography or NMR | Report results (typically 5-15%) |
| Residual Solvents | GC-FID or GC-MS | Acetonitrile ≤410 ppm, DMF ≤880 ppm, DCM ≤600 ppm |
| Bacterial Endotoxin | LAL (kinetic chromogenic) | ≤0.5 EU/mg |
| Sterility | USP <71> Sterility Test | Meets USP requirements (if applicable) |
| pH (0.1% solution) | pH meter | 3.5-5.5 |
| Specific Rotation | Polarimetry | Report results (chiral verification) |
6.1 Process Validation Requirements
Manufacturing process validation for PT-141 production demonstrates that the established process consistently produces material meeting all predetermined specifications. Validation protocols encompass three consecutive conformance batches manufactured under routine production conditions, with comprehensive testing beyond standard release specifications to establish process understanding and control.
Critical process parameters (CPPs) requiring validation include amino acid coupling efficiency, cyclization yield, purification recovery, and final product purity distribution. Process analytical technology (PAT) implementation through in-process HPLC monitoring enables real-time verification of synthesis progression and supports batch disposition decisions. Validation documentation demonstrates that the manufacturing process operates in a state of control, with batch-to-batch variability maintained within acceptable ranges.
6.2 Reference Standards
Qualified reference standards are essential for analytical method validation and routine QC testing. Primary reference standards should be obtained from reputable suppliers with comprehensive characterization data including NMR spectroscopy, high-resolution mass spectrometry, amino acid analysis, and purity determination by multiple orthogonal methods. Reference standard purity should exceed 98.0% to serve as a reliable basis for test sample comparison.
Working reference standards derived from well-characterized production batches may be qualified against primary standards and used for routine testing. Reference standard stability monitoring under defined storage conditions (-20°C or -80°C) with periodic requalification (annually or biannually) ensures continued suitability for use in quality control applications.
7. Stability Characteristics and Testing
7.1 Degradation Pathways
PT-141 stability is governed by multiple degradation pathways common to peptide pharmaceuticals, with specific considerations related to the cyclic structure and amino acid composition. Primary degradation mechanisms include deamidation of Asn residues (though not present in PT-141), oxidation of Trp and Met residues, hydrolysis of peptide bonds, and potential disulfide bond formation between Cys residues (if present as impurities).
Oxidation of the Trp residue at position 5 represents the most significant degradation pathway for PT-141, particularly under aqueous conditions and exposure to light or oxidizing agents. Oxidized Trp generates multiple products including N-formylkynurenine, kynurenine, and hydroxytryptophan derivatives, detectable by HPLC as peaks eluting near the main product peak. Minimizing oxygen exposure, controlling pH, and including antioxidants in formulations can mitigate Trp oxidation during storage and handling.
7.2 Stability Testing Protocols
Formal stability studies follow ICH Q1A guidelines with long-term testing at 25°C/60% RH for 12-36 months and accelerated testing at 40°C/75% RH for 6 months. Additional stress testing under elevated temperature (50-60°C), high humidity (>75% RH), oxidative conditions (hydrogen peroxide exposure), and photolytic conditions (visible and UV light) provides understanding of degradation mechanisms and supports shelf life justification.
| Study Type | Conditions | Duration | Time Points |
|---|---|---|---|
| Long-term | 25°C ± 2°C / 60% ± 5% RH | 36 months | 0, 3, 6, 9, 12, 18, 24, 36 months |
| Intermediate | 30°C ± 2°C / 65% ± 5% RH | 12 months | 0, 6, 12 months |
| Accelerated | 40°C ± 2°C / 75% ± 5% RH | 6 months | 0, 1, 2, 3, 6 months |
| Stress (thermal) | 50-60°C | 1-3 months | Weekly or biweekly |
| Stress (oxidative) | 0.3% H₂O₂, 25°C | 24-72 hours | 0, 24, 48, 72 hours |
| Stress (photolytic) | ICH light conditions | Per ICH Q1B | At defined light exposure levels |
| Freeze-thaw | -20°C to 25°C cycles | 5 cycles | After each cycle |
Stability samples undergo comprehensive testing including appearance, HPLC purity, peptide content, moisture content, and pH (for solutions). Degradation product identification and characterization through LC-MS analysis supports understanding of degradation mechanisms and enables development of stability-indicating analytical methods. Out-of-specification results trigger investigation and potential revalidation of storage conditions or shelf life specifications.
7.3 Formulation Strategies for Enhanced Stability
Lyophilized PT-141 demonstrates superior stability compared to aqueous solutions, with properly stored lyophilized material maintaining >95% purity for 24-36 months at -20°C. Formulation development for reconstituted products incorporates buffers (phosphate, citrate, acetate) to maintain optimal pH range (4.0-5.5), antioxidants (ascorbic acid, sodium metabisulfite) to prevent Trp oxidation, and tonicity modifiers (mannitol, trehalose) for injectable applications.
Excipient selection considers compatibility with PT-141 and the intended route of administration. Cryoprotectants such as sucrose or trehalose (2-5% w/v) protect peptide structure during freeze-thaw cycles and lyophilization. Surfactants (polysorbate 80 at 0.01-0.05%) may be included to prevent surface adsorption and aggregation, particularly for low-concentration formulations. All excipients must meet pharmaceutical grade requirements and undergo compatibility testing with PT-141 under stressed conditions.
8. Storage and Handling Requirements
8.1 Storage Conditions
PT-141 lyophilized powder requires storage at -20°C ± 5°C in sealed containers protected from light and moisture. Desiccant inclusion in the storage container provides additional moisture protection, maintaining water content below specification limits throughout the shelf life. Foil-lined or amber glass containers prevent photodegradation of the light-sensitive Trp residue, while inert atmosphere (nitrogen or argon) headspace minimizes oxidative degradation.
Short-term storage at 2-8°C (refrigerated conditions) is acceptable for periods up to 3-6 months based on stability data, facilitating manufacturing operations and distribution without requiring frozen storage throughout the supply chain. Reconstituted solutions require refrigerated storage (2-8°C) and should be used within 7-14 days depending on formulation and preservative content, with longer-term storage requiring validation data demonstrating maintained potency and purity.
8.2 Packaging Considerations
Primary packaging materials must be qualified for compatibility with PT-141 through extractables and leachables testing according to USP <1663> and <1664> guidelines. Type I borosilicate glass vials provide the highest level of protection against moisture transmission and chemical interaction, with closure systems employing butyl rubber stoppers and aluminum seals ensuring hermetic sealing.
Secondary packaging includes outer cartons providing light protection and physical protection during shipping and handling. Temperature monitoring devices (temperature indicators or data loggers) accompany shipments requiring cold chain maintenance, with deviation reporting protocols ensuring product quality is not compromised during distribution. Stability data supporting temperature excursion scenarios (e.g., 24-48 hours at room temperature) provides flexibility for handling minor deviations without automatic product rejection.
8.3 Reconstitution Protocols
Reconstitution of lyophilized PT-141 requires sterile, pyrogen-free diluent appropriate for the intended application. Bacteriostatic water for injection (containing 0.9% benzyl alcohol), sterile water for injection, or normal saline (0.9% sodium chloride) serve as common reconstitution vehicles. The diluent volume is selected to achieve the target concentration, typically 0.5-10 mg/mL depending on dosing requirements and delivery method.
Reconstitution procedure involves adding the specified diluent volume to the lyophilized vial, allowing gentle dissolution without vigorous shaking which could denature the peptide or generate foam. Complete dissolution typically occurs within 1-3 minutes at room temperature, yielding a clear to slightly opalescent solution. The reconstituted solution should be inspected visually for particulate matter and discoloration before use, with any abnormalities warranting rejection of the vial.
| Product Form | Storage Condition | Stability Duration | Container Type |
|---|---|---|---|
| Lyophilized powder | -20°C, protected from light | 24-36 months | Type I glass vial, hermetically sealed |
| Lyophilized powder | 2-8°C, protected from light | 3-6 months | Type I glass vial, hermetically sealed |
| Reconstituted solution | 2-8°C, protected from light | 7-14 days | Original vial or sterile syringe |
| Bulk solution (pre-lyophilization) | -80°C | 6-12 months | Polypropylene or glass bottles |
9. Certificate of Analysis Documentation
9.1 CoA Components and Format
The Certificate of Analysis (CoA) serves as the official quality documentation accompanying each PT-141 batch, certifying that the material meets all established specifications. A comprehensive CoA includes batch identification (batch number, manufacturing date, expiration date), storage conditions, product description, detailed test results with acceptance criteria, and authorized signatures from QC and QA personnel.
Test results presented in the CoA must reference the specific analytical methods employed, including method validation status and any deviations from standard procedures. For quantitative tests, results include the measured value, units, and acceptance range. For qualitative tests (appearance, identity), results indicate conformance or non-conformance to the specification. All data must be traceable to raw analytical data maintained in the batch record and available for regulatory inspection.
9.2 Batch Traceability
Complete batch traceability documentation connects the final PT-141 product to all raw materials, intermediates, and processing steps involved in its manufacture. The batch record includes resin lot numbers, amino acid lot numbers, reagent lot numbers, solvent lot numbers, column identification, fraction collection records, and processing equipment identification.
Electronic batch records in modern manufacturing facilities provide enhanced traceability and data integrity compared to paper-based systems, with automatic capture of processing parameters, timestamps, and operator identification. Deviations from standard procedures are documented with investigation and disposition, ensuring that all material released meets quality standards despite any processing variations encountered during manufacture.
9.3 Regulatory Compliance Documentation
For PT-141 marketed as a pharmaceutical product or active pharmaceutical ingredient (API), documentation must support cGMP compliance according to 21 CFR Parts 210 and 211 (US FDA) or equivalent international standards (ICH, EU GMP). This includes equipment qualification (IQ/OQ/PQ), analytical method validation reports, cleaning validation, process validation, environmental monitoring, personnel training records, and change control documentation.
Stability data supporting the assigned expiration date or retest date must be available in the annual stability report, with ongoing stability monitoring ensuring that marketed material continues to meet specifications throughout its shelf life. Regular audits by QA and regulatory inspections verify compliance with cGMP requirements and proper implementation of quality systems.
| Section | Information Required |
|---|---|
| Product Information | Product name, CAS number, molecular formula, molecular weight, batch number |
| Manufacturing Information | Manufacturing date, expiration/retest date, batch size, storage conditions |
| Physical Description | Appearance, color, form (lyophilized powder) |
| Identity Tests | Mass spectrometry results, HPLC retention time, amino acid composition |
| Purity Tests | HPLC purity (%), single impurity limits, peptide content |
| Water Content | Karl Fischer result (%), specification |
| Residual Solvents | GC results for acetonitrile, DMF, DCM, others (ppm) |
| Microbial Quality | Endotoxin level (EU/mg), sterility (if applicable) |
| Additional Tests | pH, specific rotation, other relevant parameters |
| Conclusion | Statement of conformance to specifications |
| Approvals | QC analyst signature/date, QA manager signature/date |
10. Manufacturing Troubleshooting and Optimization
10.1 Common Synthesis Issues
Incomplete cyclization represents the most frequent challenge in PT-141 manufacturing, resulting in linear peptide impurities that are difficult to separate during purification. Optimization strategies include extended cyclization reaction times (6-12 hours), increased temperature (30-40°C), higher dilution to minimize intermolecular reactions, and alternative coupling reagents (HATU, PyAOP) that may provide superior activation efficiency for hindered lactam formation.
Low crude purity following cleavage typically results from incomplete amino acid couplings during SPPS, generating deletion sequences that accumulate throughout the synthesis. Implementation of double coupling protocols for difficult positions (His, Trp, Arg), use of microwave-assisted synthesis for enhanced coupling kinetics, and capping procedures (acetic anhydride treatment) to terminate failure sequences reduce deletion impurity levels and improve overall process yield.
10.2 Purification Optimization
Insufficient resolution between PT-141 and closely eluting impurities necessitates purification method optimization through gradient adjustment, temperature modification, or alternative stationary phase selection. Shallow gradients (0.25-0.5% acetonitrile/minute) in the critical separation region enhance resolution at the expense of longer run times and reduced throughput. Higher column temperatures (40-50°C) often improve peak shape and resolution through reduced viscosity and enhanced mass transfer kinetics.
Alternative ion-pairing agents including heptafluorobutyric acid (HFBA) or formic acid provide different selectivity profiles compared to TFA, potentially improving separation of problematic impurity pairs. However, ion-pairing agent changes require re-optimization of gradients and may affect downstream desalting requirements. Scale-up from analytical to preparative purification should maintain similar linear velocities and gradient slopes (expressed as % B/column volume) to preserve separation quality observed at analytical scale.
10.3 Yield Optimization Strategies
Overall manufacturing yield from resin to purified product typically ranges from 10-25% depending on synthesis efficiency, crude purity, and purification recovery. Yield improvement strategies focus on maximizing coupling efficiency during SPPS (>98% per step target), optimizing cyclization conditions for maximum conversion, and implementing efficient purification protocols with high recovery of target fractions.
Process analytical technology (PAT) implementation enables real-time monitoring of critical process parameters and quality attributes, supporting adaptive purification strategies that maximize product recovery while maintaining purity specifications. Statistical process control (SPC) charting of key yield metrics (crude yield, purification recovery, final yield) identifies trends and variation sources, guiding continuous improvement initiatives.
10.4 Scale-Up Considerations
Translation from laboratory-scale synthesis (0.1-1.0 mmol) to production scale (10-100 mmol or larger) requires careful attention to mixing efficiency, heat transfer, reagent addition rates, and solvent volumes. Solid-phase synthesis at large scale may require specialized reactor designs with mechanical agitation or gas bubbling to ensure adequate resin swelling and reagent accessibility throughout the reaction bed.
Purification scale-up involves column diameter increases while maintaining similar bed height, flow velocity, and gradient slopes to preserve resolution. Linear velocity scaling (maintaining mL/min per cm² cross-sectional area) ensures similar chromatographic performance across scales. Process development studies at intermediate scales (pilot scale) validate scale-up approaches before full production implementation, identifying potential scale-dependent issues and enabling corrective action before commercial launch.
11. Regulatory and Quality Management Systems
11.1 cGMP Compliance Framework
Manufacturing PT-141 for pharmaceutical applications requires compliance with current Good Manufacturing Practice (cGMP) regulations as defined by FDA 21 CFR Parts 210, 211, and ICH Q7 (Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients). These regulations establish requirements for quality management systems, facility design, equipment qualification, personnel training, documentation practices, and quality control testing.
Quality management system implementation includes written procedures covering all aspects of manufacturing operations, from raw material receipt and testing through production, purification, testing, release, and distribution. Standard Operating Procedures (SOPs) undergo formal review and approval processes, with revision control ensuring current versions are available at points of use. Personnel training documentation demonstrates that all individuals performing manufacturing operations possess the required knowledge and competency.
11.2 Change Control and Deviation Management
Manufacturing changes potentially affecting product quality require evaluation through formal change control procedures before implementation. Changes are classified by risk level (critical, major, minor) based on potential impact to product quality, safety, or efficacy. Critical changes affecting synthesis conditions, purification parameters, or analytical methods require prospective validation before implementation, while minor changes may proceed with retrospective verification of non-impact.
Deviations from established procedures or specifications trigger investigation to determine root cause, assess impact to product quality, and implement corrective and preventive actions (CAPA). Deviation trending analysis identifies systemic issues requiring procedure revision or equipment modification. All deviations affecting released product are communicated to customers and regulatory authorities as appropriate based on quality impact assessment.
11.3 Vendor Qualification and Supply Chain Management
Raw materials for PT-141 synthesis including protected amino acids, resins, reagents, and solvents must be sourced from qualified suppliers with appropriate quality agreements and specifications. Vendor qualification processes evaluate suppliers' quality systems, manufacturing capabilities, and regulatory compliance status through questionnaires, audits, and sample testing.
Incoming raw material testing verifies identity, purity, and other critical quality attributes before release to production. Certificate of Analysis review ensures supplier test results align with internal specifications, with periodic confirmatory testing providing verification of supplier data accuracy. Dual sourcing strategies for critical materials mitigate supply chain risks and ensure manufacturing continuity.
11.4 Continuous Improvement Initiatives
Quality by Design (QbD) principles guide continuous improvement efforts through systematic understanding of critical quality attributes (CQAs), critical process parameters (CPPs), and their relationships. Design of Experiments (DoE) studies characterize process robustness and identify optimization opportunities, while statistical analysis of batch data reveals trends and variation sources requiring attention.
Lean manufacturing principles applied to PT-141 production reduce waste, streamline workflows, and improve overall equipment effectiveness (OEE). Process capability indices (Cp, Cpk) quantify manufacturing performance relative to specification limits, with values >1.33 indicating capable processes and values <1.0 triggering improvement initiatives. Regular management review of quality metrics ensures sustained focus on quality and compliance objectives.
12. Conclusion and Future Directions
PT-141 (Bremelanotide) manufacturing represents a sophisticated application of solid-phase peptide synthesis technology, requiring integration of chemical synthesis expertise, analytical chemistry capabilities, and quality management systems to produce pharmaceutical-grade material meeting stringent specifications. The cyclic heptapeptide structure and presence of D-amino acid stereochemistry present specific technical challenges that have been addressed through optimized synthesis protocols, advanced purification methodologies, and comprehensive quality control testing.
Successful PT-141 manufacturing depends on rigorous process control at each stage from resin selection through final packaging, with continuous monitoring and improvement ensuring consistent product quality. The analytical testing program combining identity verification, purity assessment, peptide content determination, and impurity profiling provides comprehensive product characterization supporting regulatory compliance and customer confidence.
Future developments in PT-141 manufacturing may include implementation of automated peptide synthesizers for enhanced reproducibility and reduced cycle times, application of preparative SFC (supercritical fluid chromatography) for improved purification efficiency and reduced solvent consumption, and development of long-acting formulations extending dosing intervals through controlled release mechanisms. Continued advances in peptide manufacturing technology and quality systems will further enhance PT-141 production capabilities, supporting growing demand for this therapeutic peptide.
Manufacturers seeking to establish PT-141 production capabilities should prioritize investment in appropriate infrastructure including peptide synthesis equipment, preparative HPLC systems, lyophilization capacity, and analytical instrumentation. Development of internal expertise through training and recruitment of experienced peptide chemists and quality professionals ensures successful process implementation and regulatory compliance. Collaboration with regulatory consultants and contract manufacturing organizations can accelerate market entry while maintaining quality and compliance standards.
References
- Chan, W.C., White, P.D. (Eds.). (2000). Fmoc Solid Phase Peptide Synthesis: A Practical Approach. Oxford University Press. - Comprehensive guide to solid-phase peptide synthesis methodologies and optimization strategies.
- Amblard, M., Fehrentz, J.A., Martinez, J., Subra, G. (2006). Methods and protocols of modern solid phase peptide synthesis. Molecular Biotechnology, 33(3), 239-254. - Review of contemporary SPPS techniques applicable to complex peptide synthesis.
- Palomo, J.M. (2014). Solid-phase peptide synthesis: an overview focused on the preparation of biologically relevant peptides. RSC Advances, 4(62), 32658-32672. - Overview of SPPS applications for bioactive peptide production including cyclization strategies.
- Góngora-Benítez, M., Tulla-Puche, J., Albericio, F. (2014). Multifaceted roles of disulfide bonds. Peptides as therapeutics. Chemical Reviews, 114(2), 901-926. - Discussion of peptide cyclization and stability considerations relevant to therapeutic peptide manufacturing.
- United States Pharmacopeia. USP <621> Chromatography. - Compendial standards for chromatographic purity assessment methods.
- ICH Harmonised Tripartite Guideline Q2(R1). Validation of Analytical Procedures: Text and Methodology. (2005). - International standards for analytical method validation applicable to peptide testing.
- ICH Harmonised Tripartite Guideline Q1A(R2). Stability Testing of New Drug Substances and Products. (2003). - Guidance on stability study design and shelf life determination for pharmaceutical products.
- ICH Harmonised Tripartite Guideline Q7. Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients. (2000). - cGMP requirements for API manufacturing including peptide synthesis operations.
- FDA Guidance for Industry: Q3C — Tables and List. (2017). Residual solvent specifications and testing requirements for pharmaceutical products.
- Vlieghe, P., Lisowski, V., Martinez, J., Khrestchatisky, M. (2010). Synthetic therapeutic peptides: science and market. Drug Discovery Today, 15(1-2), 40-56. - Market analysis and manufacturing considerations for therapeutic peptide commercialization.
Internal Resources
- Comprehensive Guide to Solid-Phase Peptide Synthesis
- RP-HPLC Purification Methods for Complex Peptides
- Analytical Testing Protocols for Peptide Quality Control
- cGMP Compliance Overview for Peptide Manufacturing
- Peptide Stability Testing: Methods and Best Practices
- Formulation Development Strategies for Peptide Therapeutics
- Scale-Up Considerations for Commercial Peptide Production