Vasopressin Manufacturing Profile: Synthesis, Purification & Quality Control Standards
Target Audience: Contract manufacturing organizations, pharmaceutical quality control professionals, peptide synthesis facilities, regulatory affairs specialists
Vasopressin (arginine vasopressin, AVP, antidiuretic hormone) represents a critical nonapeptide hormone requiring stringent manufacturing controls and comprehensive quality assurance protocols. This manufacturing profile details the complete production workflow from solid-phase peptide synthesis through final release testing, addressing the technical challenges inherent in producing this cyclic peptide with its disulfide bridge and multiple functional residues.
1. Peptide Structure and Manufacturing Considerations
Vasopressin (CAS 113-79-1) contains nine amino acid residues with the sequence Cys-Tyr-Phe-Gln-Asn-Cys-Pro-Arg-Gly-NH₂, featuring a critical disulfide bridge between Cys1 and Cys6 that creates the cyclic structure essential for biological activity. The molecular formula C₄₆H₆₅N₁₅O₁₂S₂ yields a molecular weight of 1084.23 g/mol (monoisotopic mass 1083.4364 Da). Manufacturing facilities must address several technical challenges specific to this peptide structure.
The presence of two cysteine residues requires careful oxidation control during cyclization to ensure correct disulfide bond formation. Improper oxidation conditions can lead to intermolecular disulfide formation, linear peptide retention, or incorrect pairing, all of which compromise product quality. The C-terminal glycine amide requires specialized resin selection and cleavage strategies to prevent C-terminal carboxylic acid formation. The arginine residue presents deprotection challenges, while the asparagine residue is susceptible to deamidation under alkaline or elevated temperature conditions.
Manufacturing operations must implement validated synthesis protocols that address these structural vulnerabilities while maintaining production efficiency and batch-to-batch consistency. The GHRP-2 manufacturing profile provides additional context on handling arginine-containing peptides in production environments.
2. Solid-Phase Peptide Synthesis Protocol
Vasopressin synthesis utilizes Fmoc (9-fluorenylmethoxycarbonyl) solid-phase peptide synthesis on Rink amide MBHA resin (0.4-0.7 mmol/g substitution) to ensure C-terminal amide formation. Production-scale synthesis typically employs 50-500g resin batches depending on facility capacity and market demand. The synthesis proceeds from C-terminus to N-terminus with each coupling cycle requiring careful monitoring and validation.
2.1 Resin Loading and Synthesis Parameters
Initial resin swelling in DMF (dimethylformamide) for 30 minutes precedes the synthesis cycles. Fmoc-protected amino acids are activated using standard coupling reagents in 3-5 molar excess relative to resin substitution. Manufacturing facilities commonly employ HBTU (O-benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluoro-phosphate) or HATU (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) with DIEA (N,N-diisopropylethylamine) base in DMF solvent.
| Position | Amino Acid | Protected Form | Coupling Time (min) | Critical Parameters |
|---|---|---|---|---|
| 9 (C-term) | Gly-NH₂ | Fmoc-Gly-OH on Rink Amide | 45-60 | Resin pre-swelling, complete loading |
| 8 | Arg | Fmoc-Arg(Pbf)-OH | 60-90 | Extended coupling, guanidine protection |
| 7 | Pro | Fmoc-Pro-OH | 90-120 | Secondary amine, double coupling |
| 6 | Cys | Fmoc-Cys(Trt)-OH | 60-90 | Trityl protection, disulfide precursor |
| 5 | Asn | Fmoc-Asn(Trt)-OH | 60-90 | Prevent deamidation, side-chain protection |
| 4 | Gln | Fmoc-Gln(Trt)-OH | 60-90 | Prevent deamidation, side-chain protection |
| 3 | Phe | Fmoc-Phe-OH | 45-60 | Hydrophobic residue aggregation risk |
| 2 | Tyr | Fmoc-Tyr(tBu)-OH | 60-90 | Phenol protection, oxidation sensitivity |
| 1 (N-term) | Cys | Fmoc-Cys(Trt)-OH | 60-90 | Disulfide precursor, final coupling |
Each coupling cycle includes Fmoc deprotection using 20% piperidine in DMF (2 × 5 minutes), amino acid activation and coupling (60-120 minutes depending on residue), washing with DMF (5 × resin volume), and Kaiser test or TNBS assay confirmation of complete coupling. Manufacturing protocols mandate double coupling for Pro, Arg, and both Cys residues to ensure >99.5% coupling efficiency at each step, as recommended by established peptide synthesis guidelines.1
2.2 Cleavage and Deprotection
Upon synthesis completion, the protected peptide undergoes simultaneous cleavage from the resin and side-chain deprotection using a TFA (trifluoroacetic acid) cocktail. The standard cleavage mixture contains TFA (94%), water (2.5%), triisopropylsilane (2.5%), and ethanedithiol (1%) to prevent cysteine oxidation during cleavage. This composition provides effective scavenging of reactive carbocations released during protecting group removal while maintaining cysteine thiols in reduced form.
Cleavage proceeds for 2-3 hours at ambient temperature with gentle stirring. The resin is filtered, and the filtrate is precipitated into cold diethyl ether (10× volume) at -20°C. The precipitate is collected by centrifugation, washed three times with cold ether, and dried under vacuum. This yields crude linear vasopressin with free thiol groups ready for oxidative cyclization. Recovery yields at this stage typically range from 60-75% based on initial resin loading.
3. Oxidative Cyclization and Disulfide Formation
Correct disulfide bond formation between Cys1 and Cys6 is critical for vasopressin biological activity and represents one of the most technically demanding steps in the manufacturing process. The oxidation must be carefully controlled to favor intramolecular cyclization over intermolecular dimerization or polymerization. Manufacturing facilities employ validated oxidation protocols that consistently achieve >85% cyclization yields with minimal dimer formation.
3.1 Air Oxidation Method
The most common production-scale oxidation method involves air oxidation in dilute aqueous solution at controlled pH. The crude linear peptide is dissolved at 0.1-0.5 mg/mL concentration in 0.1M ammonium bicarbonate buffer, pH 8.0-8.5. The low concentration favors intramolecular cyclization, while the alkaline pH facilitates thiol ionization and disulfide formation. The solution is stirred gently while exposed to air or oxygen at 4-20°C for 24-72 hours.
Oxidation progress is monitored by analytical RP-HPLC and mass spectrometry. The linear peptide (expected mass 1085.4 Da with two free thiols) converts to the cyclic form (expected mass 1083.4 Da with loss of 2H upon disulfide formation). Complete conversion is confirmed when the linear peptide peak diminishes to <5% of total peptide content. The pH is then adjusted to 4-6 using glacial acetic acid or dilute HCl to halt oxidation, and the solution is lyophilized immediately.2
3.2 Alternative Oxidation Methods
Some manufacturing facilities employ DMSO (dimethyl sulfoxide) oxidation for faster and more controlled cyclization. The crude linear peptide is dissolved in DMSO-water mixtures (30-50% DMSO) at similar concentrations. DMSO acts as a mild oxidizing agent that promotes disulfide formation within 6-24 hours. This method offers better batch-to-batch reproducibility but requires complete DMSO removal during subsequent purification.
Iodine oxidation represents another alternative, though it requires precise stoichiometric control. A 1:1 molar ratio of I₂ to peptide in dilute acetic acid solution achieves rapid cyclization within 30-60 minutes. Excess iodine is quenched with ascorbic acid, and the mixture is immediately loaded onto purification columns. This method's speed advantage is offset by the need for additional purification steps to remove oxidation byproducts and ensure complete iodine removal.
| Oxidation Method | Reaction Time | Cyclization Yield | Dimer Formation | Production Suitability |
|---|---|---|---|---|
| Air oxidation (pH 8.0) | 24-72 hours | 75-85% | 8-15% | High (standard method) |
| DMSO oxidation | 6-24 hours | 80-90% | 5-10% | High (faster processing) |
| Iodine oxidation | 0.5-1 hour | 70-80% | 10-15% | Moderate (requires additional purification) |
| Ferricyanide oxidation | 2-4 hours | 65-75% | 15-20% | Low (metal contamination risk) |
4. Purification Strategy and Chromatographic Methods
Vasopressin purification requires multi-stage chromatography to remove synthesis impurities, deletion sequences, oxidation byproducts, dimers, and residual salts. Manufacturing facilities typically implement a two-stage purification strategy consisting of preparative reversed-phase HPLC followed by polishing ion-exchange chromatography or a second RP-HPLC step with different selectivity.
4.1 Primary Reversed-Phase HPLC Purification
The crude oxidized peptide mixture is dissolved in water containing 0.1% TFA and loaded onto preparative C18 columns (10-30 µm particle size, 300 Å pore size) designed for peptide separations. Column dimensions range from 5-30 cm diameter depending on batch scale, with 20-50 cm length providing adequate resolution. The stationary phase must tolerate acidic mobile phases and provide sufficient retention for nonapeptide separation.
The mobile phase system consists of water with 0.1% TFA (solvent A) and acetonitrile with 0.1% TFA (solvent B). A shallow gradient from 15-35% B over 60-90 minutes provides optimal resolution between vasopressin and closely related impurities. Flow rates are scaled to column dimensions, typically 50-500 mL/min for production columns. UV detection at 214 nm and 280 nm monitors peptide elution, with the 214 nm signal providing general peptide detection while 280 nm specifically tracks aromatic residues (Tyr, Phe).
Target fractions containing >95% vasopressin by analytical HPLC are pooled, and acetonitrile is removed by rotary evaporation under vacuum at temperatures not exceeding 40°C to prevent peptide degradation. The aqueous solution is then lyophilized to yield purified vasopressin as a white to off-white powder. Recovery from preparative HPLC typically ranges from 40-60% based on crude peptide loaded, with higher purity material requiring narrower fraction collection windows and lower recovery.
4.2 Secondary Purification and Polishing
A second chromatographic step ensures final product meets pharmaceutical specifications. Ion-exchange chromatography on cation-exchange resins (sulfopropyl or carboxymethyl functional groups) effectively separates vasopressin from neutral and acidic impurities based on the arginine residue's positive charge. The peptide is loaded in 20 mM sodium phosphate buffer, pH 6.5, and eluted with a salt gradient (0-500 mM NaCl) over 30-60 minutes.
Alternatively, a second RP-HPLC step using a C8 column or different C18 stationary phase chemistry provides orthogonal selectivity. This approach effectively removes impurities that co-elute with vasopressin in the primary purification. The gradient is optimized to provide baseline resolution between the target peptide and remaining impurities while maintaining reasonable processing times. Final purity typically exceeds 98% as determined by analytical HPLC with multiple detection wavelengths.
All purification buffers and solvents must be HPLC-grade or higher quality. Water used throughout the purification process should meet USP purified water specifications (conductivity <1.1 µS/cm, total organic carbon <500 ppb). Column sanitization between batches using 0.5M NaOH or 1M acetic acid prevents cross-contamination and maintains column performance. Similar purification considerations apply to other cyclic peptides as detailed in the BPC-157 manufacturing documentation.
5. Analytical Testing and Quality Control Methods
Comprehensive analytical testing confirms vasopressin identity, purity, and quality before release. Manufacturing facilities must validate all analytical methods according to ICH Q2(R1) guidelines, demonstrating specificity, linearity, accuracy, precision, detection limit, quantitation limit, and robustness. A complete analytical testing panel includes chromatographic purity, mass spectrometry confirmation, amino acid analysis, peptide content determination, water content, and residual solvent analysis.
5.1 High-Performance Liquid Chromatography Analysis
Analytical RP-HPLC on C18 columns (4.6 mm × 150-250 mm, 5 µm particle size, 300 Å pore size) provides primary purity assessment. The peptide is dissolved at 1 mg/mL in water and injected (10-20 µL) onto the column equilibrated with water-acetonitrile-TFA mobile phase. A gradient from 10-40% acetonitrile over 30 minutes at 1 mL/min with UV detection at 214 nm resolves vasopressin from synthesis impurities, degradation products, and residual salts.
Purity is calculated as the percentage of the vasopressin peak area relative to total peak area, excluding solvent peaks and mobile phase artifacts. Specifications require ≥97.0% purity by HPLC for pharmaceutical-grade material. Key impurities that must be monitored and controlled include des-Arg vasopressin (<1.5%), des-Gly vasopressin (<1.5%), oxidized vasopressin species (<1.0%), linear vasopressin (<2.0%), and dimers/oligomers (<2.0%). The method must be validated to demonstrate resolution of at least 2.0 between vasopressin and nearest impurity peaks.
5.2 Mass Spectrometry Confirmation
Electrospray ionization mass spectrometry (ESI-MS) or matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) confirms molecular weight and assesses purity at the molecular level. For ESI-MS analysis, the peptide is dissolved at 10-100 µg/mL in water-acetonitrile (50:50) with 0.1% formic acid and infused directly into the mass spectrometer or analyzed by LC-MS.
Vasopressin exhibits characteristic multiply charged ions, typically [M+H]⁺ at m/z 1084.4, [M+2H]²⁺ at m/z 542.7, and [M+3H]³⁺ at m/z 362.1. The observed molecular weight must be within ±0.05% of the theoretical value (1084.23 Da). Mass spectrometry also detects specific impurities including linear vasopressin (theoretical mass 1086.4 Da), des-Arg vasopressin (theoretical mass 928.0 Da), and dimers (theoretical mass 2166.9 Da). The absence of significant signals at these masses provides additional purity confirmation beyond HPLC analysis.3
5.3 Amino Acid Analysis
Quantitative amino acid analysis (AAA) verifies sequence composition and provides an independent purity assessment. The peptide sample (1-5 mg) is hydrolyzed in 6M HCl at 110°C for 20-24 hours under nitrogen atmosphere. The hydrolysate is dried, reconstituted, and analyzed by ion-exchange chromatography or RP-HPLC after pre-column derivatization with phenylisothiocyanate (PITC), o-phthalaldehyde (OPA), or 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate (AQC).
| Amino Acid | Theoretical Ratio | Acceptable Range | Analytical Considerations |
|---|---|---|---|
| Cys (as cysteic acid) | 2.0 | 1.8-2.2 | Performic acid pre-oxidation required |
| Tyr | 1.0 | 0.9-1.1 | Reference residue (stable) |
| Phe | 1.0 | 0.9-1.1 | Stable under hydrolysis |
| Gln (as Glu) | 1.0 | 0.9-1.1 | Complete conversion to Glu during hydrolysis |
| Asn (as Asp) | 1.0 | 0.9-1.1 | Complete conversion to Asp during hydrolysis |
| Pro | 1.0 | 0.9-1.1 | Imino acid, special detection requirements |
| Arg | 1.0 | 0.9-1.1 | Stable under standard conditions |
| Gly | 1.0 | 0.9-1.1 | Stable, smallest amino acid |
All amino acid ratios must fall within ±10% of theoretical values when normalized to a reference residue (typically Tyr or Phe). Cysteine quantitation requires performic acid pre-oxidation to convert cysteine to cysteic acid prior to hydrolysis, as free cysteine is unstable under hydrolysis conditions. The presence of correct amino acid ratios confirms sequence identity and absence of substitution errors during synthesis.
5.4 Peptide Content Determination
Total peptide content is determined by quantitative amino acid analysis following hydrolysis. The concentration of multiple stable amino acids (Tyr, Phe, Arg, Pro) is measured against calibration standards, and peptide content is calculated based on the known stoichiometry. Alternatively, UV spectrophotometry at 280 nm using the theoretical extinction coefficient (ε₂₈₀ = 1,490 M⁻¹cm⁻¹ based on Tyr contribution) provides peptide quantitation.
Pharmaceutical specifications typically require ≥85% peptide content on a dry basis after correction for water and residual solvent content. The peptide content value accounts for residual TFA salts, counterions, and other non-peptide components present in the purified material. This assay is critical for accurate dosing calculations and ensures consistent biological activity across batches.
5.5 Additional Analytical Tests
Karl Fischer titration measures water content, with specifications typically requiring ≤10% w/w. Excessive water content indicates inadequate lyophilization and affects product stability. Residual solvent analysis by gas chromatography quantifies acetonitrile, DMF, and other organic solvents, ensuring compliance with ICH Q3C guidelines (acetonitrile limit 410 ppm, DMF limit 880 ppm).
Bacterial endotoxin testing by LAL (Limulus Amebocyte Lysate) assay confirms endotoxin levels <10 EU/mg for injectable-grade material. Sterility testing according to USP <71> is required for sterile products. Heavy metal analysis by ICP-MS (inductively coupled plasma mass spectrometry) ensures palladium (<10 ppm) and other metals remain below acceptable limits. These specifications align with those described in the CJC-1295 quality control protocols.
6. Batch Manufacturing Specifications and Release Criteria
Each vasopressin batch must meet comprehensive specifications before release for commercial distribution. Manufacturing facilities establish specifications based on regulatory requirements, pharmacopeial standards (USP, EP), and internal quality standards derived from batch history and process capability studies. The specification table below represents typical release criteria for pharmaceutical-grade vasopressin.
| Test Parameter | Specification | Test Method | Acceptance Criteria |
|---|---|---|---|
| Appearance | White to off-white lyophilized powder | Visual inspection | Conforms to description |
| Solubility | Freely soluble in water | Visual observation at 10 mg/mL | Clear colorless solution |
| Identity (HPLC) | Retention time matches reference | RP-HPLC, UV 214 nm | RT ± 2% of reference standard |
| Identity (MS) | Molecular weight 1084.23 ± 0.5 Da | ESI-MS or MALDI-TOF MS | [M+H]⁺ = 1084.4 ± 0.5 |
| Purity (HPLC) | ≥97.0% | RP-HPLC, UV 214 nm | Single main peak ≥97.0% area |
| Des-Arg impurity | ≤1.5% | RP-HPLC, UV 214 nm | Individual impurity ≤1.5% |
| Linear vasopressin | ≤2.0% | RP-HPLC, UV 214 nm | Individual impurity ≤2.0% |
| Dimers/oligomers | ≤2.0% | RP-HPLC, UV 214 nm | Combined total ≤2.0% |
| Any unspecified impurity | ≤0.5% | RP-HPLC, UV 214 nm | Each impurity ≤0.5% |
| Total impurities | ≤3.0% | RP-HPLC, UV 214 nm | Sum of all impurities ≤3.0% |
| Amino acid composition | Conforms to theory ± 10% | AAA after HCl hydrolysis | All residues within ±10% |
| Peptide content | ≥85.0% (dry basis) | Quantitative AAA or UV | ≥85.0% by weight |
| Water content | ≤10.0% | Karl Fischer titration | ≤10.0% w/w |
| TFA content | ≤1.0% | Ion chromatography or ¹⁹F NMR | ≤1.0% w/w |
| Acetonitrile | ≤410 ppm | GC headspace | Meets ICH Q3C limit |
| Residual solvents | Per ICH Q3C | GC-FID | All solvents within limits |
| Heavy metals (Pd) | ≤10 ppm | ICP-MS | ≤10 ppm |
| Bacterial endotoxins | ≤10.0 EU/mg | LAL assay (USP <85>) | ≤10.0 EU/mg |
| Bioburden (if non-sterile) | ≤100 CFU/g | USP <61> and <62> | Total aerobic count ≤100 CFU/g |
| Sterility (if sterile) | Sterile | USP <71> | No growth observed |
Batch release requires all specifications to be met simultaneously. Out-of-specification results trigger investigation protocols per ICH Q10 quality system requirements. Batches failing to meet specifications may undergo reprocessing (additional purification) if economically viable, but all reprocessed material must meet full specifications before release. The facility quality unit has final authority for batch disposition decisions.
6.1 In-Process Controls
Manufacturing facilities implement in-process controls at critical process stages to ensure consistent quality and reduce batch failure risk. Key in-process tests include:
- Synthesis monitoring: Kaiser test or TNBS assay after each coupling to confirm >99.5% coupling efficiency
- Crude purity: HPLC analysis of crude peptide post-cleavage to assess synthesis quality (target ≥30% crude purity)
- Cyclization monitoring: HPLC and MS analysis during oxidation to optimize reaction time and minimize dimers
- Purification fractions: HPLC analysis of all collected fractions to guide pooling decisions and optimize recovery
- Intermediate purity: HPLC analysis after first purification step to determine if additional purification is required
These in-process controls provide early warning of process deviations and enable real-time adjustments to maintain product quality. Trending of in-process data over multiple batches identifies process drift and supports continuous improvement initiatives.
7. Stability Studies and Storage Recommendations
Vasopressin stability studies follow ICH Q1A(R2) guidelines for new drug substances, encompassing long-term, accelerated, and stress conditions. These studies establish shelf-life, storage conditions, and retest dates while identifying major degradation pathways. Vasopressin's primary stability concerns include oxidation of Tyr and Cys residues, deamidation of Asn and Gln residues, and disulfide bond rearrangement or reduction.
7.1 Stability Study Design
Long-term stability studies are conducted at 25°C ± 2°C and 60% RH ± 5% RH for 12-36 months, with testing at 0, 3, 6, 9, 12, 18, 24, and 36 months. Accelerated studies at 40°C ± 2°C and 75% RH ± 5% RH for 6 months include testing at 0, 1, 2, 3, and 6 months. Refrigerated storage studies at 5°C ± 3°C for 36 months provide data for cold-storage products. All studies use the same container-closure system as the commercial package configuration.
Stress testing at 60°C, high humidity (90% RH), pH extremes (pH 3 and pH 9), oxidative conditions (0.3% H₂O₂), and photostability conditions (ICH Q1B) identifies inherent stability characteristics and potential degradation pathways. These studies inform storage recommendations and guide formulation development for pharmaceutical products.
| Storage Condition | HPLC Purity After 12 Months | Major Degradation Products | Recommendations |
|---|---|---|---|
| -20°C, desiccated | >98% | Minimal degradation | Preferred long-term storage |
| 2-8°C, desiccated | 97-98% | Trace oxidation, deamidation | Acceptable for 24+ months |
| 25°C/60% RH | 95-97% | Oxidation (3-5%), deamidation (2-3%) | Maximum 12-18 months |
| 40°C/75% RH (6 months) | 90-94% | Oxidation (5-7%), deamidation (3-5%) | Not recommended |
| Aqueous solution (pH 5-7, 4°C) | 85-90% | Disulfide reduction, deamidation | Use within 7-14 days |
7.2 Degradation Pathways and Impurity Formation
The primary degradation pathway involves oxidation of the tyrosine phenol group and cysteine sulfur atoms. Exposure to oxygen, light, or elevated temperatures promotes formation of oxidized species detectable by HPLC as peaks eluting later than native vasopressin. Mass spectrometry confirms +16 Da mass increases corresponding to single oxygen addition. These oxidized forms exhibit reduced biological activity and must be controlled below 2% for pharmaceutical-grade material.4
Deamidation of Asn5 and Gln4 represents another significant degradation mechanism, particularly under alkaline or elevated temperature conditions. Asparagine deamidation proceeds through cyclic imide intermediates, yielding aspartic acid and isoaspartic acid products. This results in peptides with +1 Da mass increases and altered chromatographic behavior. The deamidation rate increases with temperature and pH, making refrigerated storage at neutral pH critical for long-term stability.
Disulfide bond reduction or scrambling can occur in aqueous solution, particularly in the presence of thiols or reducing agents. This yields linear vasopressin (observed as +2 Da mass) or potentially scrambled isomers with non-native disulfide connectivity. These degradation products demonstrate significantly reduced or absent biological activity. Storage as dry powder in sealed containers under inert atmosphere minimizes this degradation pathway.
7.3 Recommended Storage Conditions
Based on stability data, vasopressin bulk drug substance should be stored at -20°C or below in sealed containers with desiccant, protected from light. Under these conditions, the peptide maintains >98% purity for 36+ months. If refrigerated storage (2-8°C) is used, packaging should include desiccant and/or inert atmosphere backfilling, with an established shelf-life of 24 months based on stability data demonstrating purity >97% at 24 months.
Room temperature storage is not recommended for long-term stability but may be acceptable for shipping and short-term handling (maximum 30 days). All containers should be amber glass or HDPE bottles with tight-sealing caps and Parafilm sealing to minimize moisture and oxygen ingress. Similar storage considerations apply to related peptides as outlined in the ipamorelin storage protocols.
Reconstituted aqueous solutions should be prepared immediately before use. If storage of reconstituted solution is necessary, formulation with appropriate buffers (pH 5.0-6.5), antioxidants (ascorbic acid, sodium metabisulfite), and antimicrobial preservatives (benzyl alcohol, metacresol) extends stability. Even in optimized formulations, solutions should be stored at 2-8°C and used within 14 days. Freezing of aqueous solutions is not recommended due to potential aggregation upon thawing.
8. Certificate of Analysis Components
Each vasopressin batch must be accompanied by a Certificate of Analysis (CoA) documenting all quality testing results and confirming compliance with specifications. The CoA serves as the primary quality document for customer acceptance and regulatory filing purposes. Manufacturing facilities must maintain robust QC systems to ensure accurate, complete, and traceable CoA generation.
8.1 Certificate of Analysis Format and Required Information
A comprehensive vasopressin CoA includes the following sections and data elements:
Product Identification Section:
- Product name: Vasopressin (Arginine Vasopressin, AVP)
- CAS number: 113-79-1
- Molecular formula: C₄₆H₆₅N₁₅O₁₂S₂
- Molecular weight: 1084.23 g/mol
- Sequence: Cys-Tyr-Phe-Gln-Asn-Cys-Pro-Arg-Gly-NH₂ (disulfide bridge Cys1-Cys6)
- Catalog number or product code
- Batch/Lot number with manufacturing date
- Quantity manufactured and packaged
- Expiration date or retest date
Analytical Testing Results Section:
- Complete test results for all specification parameters listed in Section 6
- Test methods used with reference to validation documentation
- Specification limits and actual results for each test
- Pass/fail determination for each parameter
- Chromatograms for HPLC analysis (primary trace)
- Mass spectrum showing major ions and molecular weight confirmation
Storage and Handling Section:
- Recommended storage temperature and conditions
- Container type and closure system
- Special handling precautions
- Reconstitution instructions for powder products
Quality Assurance Section:
- CoA issue date
- QC analyst signature and date
- QA approval signature and date
- Statement of GMP compliance (if applicable)
- Regulatory status declaration
- Certificate validity statement
8.2 HPLC Chromatogram Requirements
The CoA must include a representative HPLC chromatogram showing baseline separation between vasopressin and major impurities. The chromatogram should display appropriate scale (y-axis in mAU or AU), retention times for all significant peaks, integration parameters, and peak purity assessments if available from diode array detection. The vasopressin peak should be clearly labeled with its retention time and area percentage.
Key impurity peaks, if present above 0.5%, should be labeled with retention times and area percentages. The chromatogram baseline should be stable without significant drift or noise, and integration should be appropriate with no forced or manual adjustments without justification. Peak asymmetry factors and theoretical plate numbers may be reported to demonstrate column performance and method suitability.
8.3 Quality Documentation and Traceability
The manufacturing facility must maintain complete batch records including synthesis logs, purification records, analytical raw data, instrument calibration records, reference standard traceability, and deviation reports (if any). These records support the CoA and must be available for customer audits or regulatory inspections. All data must be traceable to specific instruments, analysts, and dates of analysis.
Reference standards used for identity and quantitation must be characterized materials with established purity and traceability to USP Reference Standards or authenticated vendor materials. The CoA should reference the lot number of standards used for testing. Instrument calibration certificates should be current and demonstrate equipment qualification per ICH Q2 guidelines.
9. Regulatory Considerations and GMP Compliance
Vasopressin manufacturing for pharmaceutical applications requires compliance with current Good Manufacturing Practice (cGMP) regulations as defined by FDA 21 CFR Parts 210-211, EMA GMP guidelines, or equivalent regional regulations. Manufacturing facilities must be registered with appropriate regulatory authorities and maintain inspectional readiness at all times. For API (Active Pharmaceutical Ingredient) manufacturing, ICH Q7 guidelines provide specific GMP requirements applicable to peptide synthesis operations.5
9.1 Facility Requirements and Quality Systems
Manufacturing facilities must implement comprehensive quality management systems encompassing:
- Quality assurance unit: Independent QA/QC department with authority to approve or reject all batches, review batch records, investigate deviations, and approve changes to manufacturing processes
- Validated processes: Process validation demonstrating consistent production of material meeting specifications across minimum three consecutive batches
- Facility design: Appropriate environmental controls, segregated areas for different operations, proper material flow to prevent cross-contamination
- Equipment qualification: IQ/OQ/PQ protocols for all critical equipment (synthesizers, HPLC systems, lyophilizers)
- Analytical method validation: Complete validation per ICH Q2(R1) for all release and stability testing methods
- Change control: Formal system for evaluating and approving changes to processes, methods, specifications, or equipment
- CAPA system: Corrective and preventive action system for addressing deviations, OOS results, and customer complaints
- Training program: Documented training for all personnel in GMP requirements and their specific job functions
Environmental monitoring programs ensure manufacturing areas maintain appropriate cleanliness levels. While aseptic processing requirements depend on the final product form, bulk peptide synthesis typically occurs in classified cleanrooms (ISO 7 or ISO 8) with appropriate gowning and environmental monitoring for viable and non-viable particulates.
9.2 Documentation and Batch Records
Each manufacturing batch requires a complete batch production record documenting all manufacturing steps, in-process testing results, deviations, and final testing results. The batch record must provide complete traceability for all raw materials used, including amino acids, resins, solvents, and reagents, with vendor certifications and internal QC testing results. All critical process parameters must be recorded, including coupling times, temperatures, reagent volumes, and yields at each step.
The batch record should include equipment identification numbers, operator signatures, and QA review signatures at critical stages. Any deviations from standard operating procedures must be documented with investigation reports and impact assessments. The completed batch record undergoes comprehensive QA review before batch release, verifying all steps were performed correctly and all specifications were met.
9.3 Regulatory Filing Support
For vasopressin intended for use in drug products requiring regulatory approval (IND, NDA, ANDA, or equivalents), the manufacturing facility must provide comprehensive CMC (Chemistry, Manufacturing, and Controls) documentation including:
- Drug substance characterization data (structure elucidation, physicochemical properties)
- Manufacturing process description with flow diagrams and detailed procedures
- Process validation reports demonstrating consistency and capability
- Specifications with justification and comparison to pharmacopeial standards
- Analytical method validation reports for all release and stability methods
- Stability data supporting proposed storage conditions and retest dates
- Impurity qualification data for impurities above identification thresholds
- Container-closure system validation data
- Facility and equipment information including master lists and qualification status
The facility may undergo pre-approval inspection by regulatory authorities before approval of drug applications containing the manufactured vasopressin. Maintaining comprehensive documentation and robust quality systems ensures successful regulatory inspections and approvals. These regulatory considerations parallel those for other pharmaceutical peptides as described in the sermorelin regulatory documentation.
10. Process Development and Optimization Strategies
Continuous improvement of vasopressin manufacturing processes ensures optimal yield, purity, and cost-effectiveness while maintaining regulatory compliance. Process development activities focus on synthesis optimization, purification enhancement, analytical method improvement, and scale-up considerations. Manufacturing facilities should implement Quality by Design (QbD) principles per ICH Q8, systematically evaluating critical process parameters and their impact on critical quality attributes.
10.1 Synthesis Optimization
Key areas for synthesis optimization include coupling reagent selection, coupling time optimization, racemization prevention, and aggregation control during difficult couplings. For arginine coupling, extended coupling times (90-120 minutes) with HATU or PyBOP activating agents provide superior results compared to standard HBTU activation. The proline coupling benefits from double coupling with extended reaction times due to the secondary amine's reduced nucleophilicity.
Aggregation during synthesis, particularly during coupling of hydrophobic residues (Phe, Tyr), can be mitigated through:
- Addition of chaotropic salts (LiBr, LiCl) to coupling and deprotection solutions
- Use of pseudoproline dipeptides at strategic positions to disrupt β-sheet formation
- Elevated temperature coupling (40-50°C) for problematic sequences
- Microwave-assisted synthesis for reduced coupling times and improved efficiency
These optimization strategies can improve crude purity from 30-40% to 50-70%, significantly reducing purification burden and improving overall yields. Design of experiments (DoE) methodologies systematically evaluate multiple parameters simultaneously, identifying optimal conditions more efficiently than traditional one-factor-at-a-time approaches.6
10.2 Purification Method Development
Chromatographic method optimization focuses on stationary phase selection, mobile phase composition, gradient optimization, and scale-up strategies. For vasopressin purification, C18 columns with 300 Å pore size and intermediate carbon loading (10-15% carbon) provide optimal retention and peak shape. Alternative stationary phases including C8, phenyl-hexyl, or biphenyl columns offer different selectivity and may improve separation of specific impurity pairs.
Mobile phase optimization evaluates different ion-pairing agents (TFA, formic acid, acetic acid, heptafluorobutyric acid) and their concentrations. While TFA provides excellent peak shape and resolution, alternative acids may be preferred when TFA removal is challenging or when MS-compatible mobile phases are required for preparative LC-MS purification. Gradient shape optimization (linear, curved, step gradients) and gradient time adjustments balance resolution, throughput, and solvent consumption.
Scale-up from analytical to preparative chromatography requires attention to column loading capacity, injection volume optimization, flow rate scaling based on linear velocity, and fraction collection strategies. Overloading columns beyond 20-30 mg crude peptide per gram of stationary phase typically degrades resolution and reduces product purity. Multiple injection cycling with automated fraction collection maximizes throughput while maintaining quality.
10.3 Analytical Method Enhancement
Continuous improvement of analytical methods enhances product characterization and process understanding. Advanced techniques including:
- UHPLC (ultra-high performance liquid chromatography): Sub-2 µm particles provide enhanced resolution and reduced analysis time compared to conventional HPLC
- High-resolution mass spectrometry: Q-TOF or Orbitrap MS systems accurately determine molecular formulas and identify unknown impurities
- Peptide mapping: Enzymatic digestion followed by LC-MS/MS confirms sequence and identifies modification sites
- Circular dichroism spectroscopy: Assesses secondary structure and confirms native conformation
- Differential scanning calorimetry: Measures thermal stability and detects formulation-related changes
These advanced techniques provide deeper product understanding and support comparability assessments when manufacturing changes are implemented. The analytical toolkit should evolve as regulatory expectations and technology capabilities advance.
10.4 Quality by Design Implementation
QbD approaches systematically identify critical quality attributes (CQAs) including purity, peptide content, specific impurities, and biological activity. Risk assessment tools such as Failure Mode and Effects Analysis (FMEA) prioritize process parameters requiring control. Design spaces define validated operating ranges where consistent quality is assured, providing operational flexibility without requiring regulatory notification of changes within the design space.
Process analytical technology (PAT) enables real-time monitoring and control. In-line or at-line HPLC monitoring during purification allows dynamic fraction collection decisions based on actual purity rather than fixed time windows. UV spectroscopy during synthesis provides real-time coupling completion assessment. These PAT tools enhance process understanding and control while supporting continuous manufacturing concepts.
Conclusion
Vasopressin manufacturing requires sophisticated technical capabilities encompassing solid-phase peptide synthesis, oxidative cyclization chemistry, multi-stage purification, and comprehensive analytical testing. The structural complexity of this cyclic nonapeptide, with its critical disulfide bridge and multiple functional amino acids, demands validated processes and stringent quality control to ensure consistent product quality meeting pharmaceutical specifications.
Manufacturing facilities must implement cGMP quality systems, validated analytical methods, comprehensive stability programs, and robust documentation practices to support regulatory requirements. Process optimization through QbD principles and advanced analytical techniques drives continuous improvement while maintaining product quality and regulatory compliance. As vasopressin continues to serve important therapeutic roles in endocrine disorders and critical care medicine, reliable manufacturing processes ensure consistent availability of this essential peptide hormone.
The technical considerations outlined in this manufacturing profile provide a comprehensive framework for facilities producing vasopressin for pharmaceutical applications, research use, or commercial distribution. Similar manufacturing principles apply to related peptide hormones and therapeutic peptides, as detailed in the broader peptide manufacturing resources available at PeptideForge.com.
References
- Chan, W. C., & White, P. D. (Eds.). (2000). Fmoc Solid Phase Peptide Synthesis: A Practical Approach. Oxford University Press. https://doi.org/10.1093/oso/9780199637256.001.0001
- Moroder, L., & Musiol, H. J. (2017). Insulin—from its discovery to the industrial synthesis of modern insulin analogues. Angewandte Chemie International Edition, 56(36), 10656-10669. https://doi.org/10.1002/anie.201702493
- Bouchard, M., Zurdo, J., Nettleton, E. J., Dobson, C. M., & Robinson, C. V. (2000). Formation of insulin amyloid fibrils followed by FTIR simultaneously with CD and electron microscopy. Protein Science, 9(10), 1960-1967. https://doi.org/10.1110/ps.9.10.1960
- Manning, M. C., Patel, K., & Borchardt, R. T. (1989). Stability of protein pharmaceuticals. Pharmaceutical Research, 6(11), 903-918. https://doi.org/10.1023/A:1015929109894
- ICH Q7. (2000). Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. https://www.ich.org/page/quality-guidelines
- Collins, J. M., Porter, K. A., Singh, S. K., & Vanier, G. S. (2014). High-efficiency solid phase peptide synthesis (HE-SPPS). Organic Letters, 16(3), 940-943. https://doi.org/10.1021/ol4036825
- Behrendt, R., White, P., & Offer, J. (2016). Advances in Fmoc solid-phase peptide synthesis. Journal of Peptide Science, 22(1), 4-27. https://doi.org/10.1002/psc.2836
- Coin, I., Beyermann, M., & Bienert, M. (2007). Solid-phase peptide synthesis: from standard procedures to the synthesis of difficult sequences. Nature Protocols, 2(12), 3247-3256. https://doi.org/10.1038/nprot.2007.454
- 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. https://doi.org/10.1385/MB:33:3:239
- Henninot, A., Collins, J. C., & Nuss, J. M. (2018). The current state of peptide drug discovery: Back to the future? Journal of Medicinal Chemistry, 61(4), 1382-1414. https://doi.org/10.1021/acs.jmedchem.7b00318
Related Manufacturing Profiles:
- GHRP-2 Manufacturing Profile - Hexapeptide synthesis and purification
- BPC-157 Manufacturing Profile - Pentadecapeptide production protocols
- CJC-1295 Manufacturing Profile - Modified peptide synthesis with DAC conjugation
- Ipamorelin Manufacturing Profile - Pentapeptide manufacturing and quality control
- Sermorelin Manufacturing Profile - Long-chain peptide synthesis and regulatory considerations
- TB-500 Manufacturing Profile - Large peptide manufacturing challenges
- Hexarelin Manufacturing Profile - Hexapeptide production and specifications
This manufacturing profile is intended for informational purposes for qualified pharmaceutical and research professionals. All manufacturing activities must comply with applicable regulations and quality standards. For specific technical questions regarding vasopressin manufacturing capabilities, contact PeptideForge.com.