Hexarelin Manufacturing Profile: Synthesis, Purification & Quality Control Standards

Hexarelin Manufacturing Profile: Technical Specifications and Quality Control Parameters

Complete Manufacturing Documentation for Hexarelin (His-D-2-Methyl-Trp-Ala-Trp-D-Phe-Lys-NH2) Production

1. Executive Manufacturing Summary

Hexarelin, a synthetic hexapeptide growth hormone secretagogue with the sequence His-D-2-Methyl-Trp-Ala-Trp-D-Phe-Lys-NH2, represents a critical challenge in peptide manufacturing due to its incorporation of non-standard amino acids and C-terminal amidation. This manufacturing profile provides comprehensive technical specifications for solid-phase peptide synthesis (SPPS), purification protocols, analytical validation methods, and quality control parameters required for pharmaceutical-grade production.

The manufacturing process outlined in this document adheres to current Good Manufacturing Practice (cGMP) guidelines as defined by the FDA and ICH Q7 standards for active pharmaceutical ingredients. Manufacturing facilities must maintain environmental controls, validated equipment, and documented quality systems to ensure consistent production of Hexarelin meeting United States Pharmacopeia (USP) monograph requirements for peptide purity, identity, and potency.

Table 1: Hexarelin Critical Manufacturing Parameters
Parameter Specification Test Method
Molecular Formula C47H58N12O6 Mass Spectrometry
Molecular Weight 887.04 g/mol ESI-MS, MALDI-TOF
Sequence His-D-2-Me-Trp-Ala-Trp-D-Phe-Lys-NH2 Amino Acid Analysis, Edman Degradation
Minimum Purity ≥98.0% (HPLC) RP-HPLC (220 nm, 280 nm)
Stereochemistry D-amino acids at positions 2 and 5 Chiral HPLC, CD Spectroscopy
Water Content ≤8.0% (w/w) Karl Fischer Titration
TFA Content ≤0.1% (w/w) Ion Chromatography
Acetate Content Report value Ion Chromatography
Endotoxin Level ≤5.0 EU/mg LAL Chromogenic Assay
Bioburden ≤10 CFU/g USP <61>, <62>

Manufacturing scale-up from research quantities to commercial batch sizes requires careful optimization of coupling efficiencies, aggregation control during synthesis, and purification yield optimization. Typical commercial batch sizes range from 100 grams to 5 kilograms, with overall process yields of 15-25% calculated from initial resin loading to final purified product.

2. Solid-Phase Peptide Synthesis Protocol

2.1 Synthesis Strategy and Resin Selection

Hexarelin synthesis is conducted using Fmoc (9-fluorenylmethyloxycarbonyl) solid-phase methodology on an automated peptide synthesizer equipped with real-time monitoring capabilities. The C-terminal amide functionality requires utilization of Rink amide resin (4-methylbenzhydrylamine, MBHA) with a loading capacity of 0.4-0.7 mmol/g. Alternative resins including PAL-PEG or ChemMatrix-Rink Amide may be employed to reduce aggregation during synthesis of the hydrophobic sequence.

The synthesis proceeds in a C-to-N terminal direction, requiring sequential coupling of six amino acid residues including two D-configured amino acids and one non-standard methylated tryptophan derivative. Critical synthesis considerations include:

  • Incorporation of D-2-methyl-tryptophan at position 2 requires pre-activated amino acid derivatives or extended coupling times
  • D-phenylalanine at position 5 necessitates chiral purity verification of starting materials
  • Multiple tryptophan residues increase oxidation susceptibility during synthesis and handling
  • Hydrophobic character of the sequence promotes aggregation requiring specialized coupling conditions

2.2 Detailed Coupling Cycle Parameters

Standard coupling cycles employ a 4-fold molar excess of protected amino acids relative to resin loading. Activation is achieved using 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) in the presence of DIEA (N,N-diisopropylethylamine) base.

Table 2: Fmoc-SPPS Coupling Cycle for Hexarelin
Step Reagent/Solvent Duration Purpose
Resin Swelling DMF 15 min Resin matrix expansion
Fmoc Deprotection 20% piperidine in DMF 2 × 5 min N-terminal deprotection
Wash DMF 5 × 1 min Reagent removal
Amino Acid Activation Fmoc-AA (4 eq), HBTU (3.9 eq), DIEA (8 eq) in DMF 2 min Active ester formation
Coupling Activated amino acid solution 45-60 min Peptide bond formation
Wash DMF 5 × 1 min Excess reagent removal
Kaiser Test Ninhydrin reagent 3 min at 100°C Coupling completion verification
Capping (if needed) Ac2O/DIEA in DMF 10 min Unreacted amine acetylation

For difficult couplings, particularly D-2-methyl-tryptophan incorporation, double coupling procedures are employed with extended reaction times up to 2 hours. Microwave-assisted coupling at controlled temperatures (50-75°C) may be utilized to enhance coupling kinetics while minimizing racemization risks for D-amino acid residues. Real-time UV monitoring at 301 nm during Fmoc deprotection provides quantitative assessment of coupling efficiency, with acceptable values greater than 99.5% for each cycle.

2.3 Aggregation Prevention Strategies

The hydrophobic nature and secondary structure propensity of Hexarelin necessitate aggregation control measures throughout synthesis. Chaotropic additives are incorporated into coupling and deprotection solutions to disrupt peptide-peptide interactions:

  • 6-8 M guanidine hydrochloride in deprotection solutions
  • 0.4 M LiCl in DMF coupling solutions
  • 2% (v/v) piperazine substitution in deprotection cocktails
  • Pseudoproline dipeptide building blocks at strategic positions

Temperature control during synthesis maintains reactor temperature at 25°C ± 2°C to prevent premature aggregation while ensuring adequate coupling kinetics. Higher synthesis scales may require implementation of continuous flow SPPS methodologies to maintain consistent mixing and reagent contact with growing peptide chains.

2.4 Cleavage and Global Deprotection

Following completion of chain assembly and final Fmoc removal, the peptide-resin is subjected to simultaneous cleavage and side-chain deprotection using a TFA-based cocktail. The standard cleavage mixture consists of:

  • TFA (trifluoroacetic acid): 92.5% (v/v)
  • TIS (triisopropylsilane): 2.5% (v/v)
  • Water: 2.5% (v/v)
  • EDT (1,2-ethanedithiol): 2.5% (v/v)

The cleavage reaction proceeds for 2-3 hours at room temperature with gentle agitation. Multiple tryptophan residues require inclusion of scavengers (EDT, thioanisole) to prevent alkylation by t-butyl cations. Following cleavage, the peptide solution is filtered to remove spent resin, and crude peptide is precipitated by addition of cold diethyl ether (10-fold volume excess). The precipitate is collected by centrifugation, washed three times with cold ether, and dried under nitrogen stream to yield crude Hexarelin as an off-white to light tan powder. Typical crude purity ranges from 40-60% by HPLC analysis, necessitating extensive purification.

3. Preparative Purification Protocols

3.1 Primary Purification: Preparative RP-HPLC

Purification of crude Hexarelin to pharmaceutical-grade specifications requires multi-stage reversed-phase high-performance liquid chromatography (RP-HPLC) employing C18 stationary phases. The purification strategy typically involves an initial preparative-scale separation followed by semi-preparative polishing to achieve target purity specifications exceeding 98%.

Table 3: Preparative RP-HPLC Parameters for Hexarelin Purification
Parameter Specification Notes
Column C18, 10 μm, 300 Å pore size 50 mm × 250-500 mm dimensions
Mobile Phase A 0.1% TFA in water (v/v) HPLC-grade water, sequencing-grade TFA
Mobile Phase B 0.1% TFA in acetonitrile (v/v) HPLC-grade acetonitrile
Flow Rate 80-120 mL/min Scale dependent
Gradient 25-45% B over 30-60 min Optimize for resolution
Column Temperature 25°C ± 2°C Thermostated column compartment
Detection 220 nm, 280 nm Dual wavelength monitoring
Injection Load 500-1000 mg crude peptide Column loading 10-20 mg/mL bed volume
Sample Preparation 20-40 mg/mL in 20% ACN/water Pre-filter through 0.45 μm PVDF

Crude Hexarelin is dissolved in the minimum volume of dilute acetonitrile solution (typically 20-30% ACN with 0.1% TFA) and filtered through 0.45 μm PVDF membranes prior to injection. The preparative gradient is optimized to achieve baseline resolution between Hexarelin and primary impurities including deletion sequences, diastereomers, and oxidation products. UV detection at 220 nm provides general peptide detection, while 280 nm monitoring specifically tracks tryptophan-containing species.

Product fractions are collected based on predetermined retention time windows established during method development. Each fraction undergoes analytical RP-HPLC assessment to determine purity. Fractions meeting intermediate purity specifications (typically ≥95%) are pooled for subsequent polishing steps. Material of insufficient purity may be recycled through additional preparative runs or diverted to reprocessing streams.

3.2 Polishing and Counter-Ion Exchange

Pooled preparative fractions undergo semi-preparative HPLC polishing using shallower gradients (typically 30-40% B over 60-90 minutes) to achieve final purity specifications. This polishing step effectively removes near-neighbor impurities co-eluting during initial purification. Following polishing, the peptide exists as the TFA salt due to ion-pairing with trifluoroacetate counter-ions from mobile phase additives.

Counter-ion exchange to acetate or chloride salts may be performed to reduce TFA content, improving stability and reducing potential toxicity concerns. Counter-ion exchange is accomplished through repeated preparative HPLC runs using volatile acid modifiers (acetic acid, hydrochloric acid) or through solid-phase extraction on ion-exchange resins. The acetate salt form is commonly preferred for pharmaceutical applications due to improved stability profiles and reduced hygroscopicity compared to TFA salts.

3.3 Lyophilization Process

Purified Hexarelin solutions undergo controlled lyophilization to produce stable, free-flowing powder suitable for long-term storage and subsequent formulation. The lyophilization process follows a validated cycle designed to maintain peptide integrity while achieving target residual moisture specifications:

Table 4: Hexarelin Lyophilization Cycle Parameters
Phase Temperature Pressure Duration
Loading 5°C Atmospheric -
Freezing -45°C Atmospheric 4 hours
Primary Drying -30°C shelf, -45°C condenser 50-100 mTorr 48-72 hours
Secondary Drying 20°C ramp to 25°C 50 mTorr 12-24 hours
Backfill 25°C Nitrogen to atmospheric -

Prior to lyophilization, purified peptide solutions are adjusted to optimal pH (4.0-5.0) and may be supplemented with lyoprotectants including mannitol (2-5% w/v) or trehalose (1-3% w/v) to stabilize peptide conformation during freeze-drying. Solutions are filled into pre-sterilized glass vials under aseptic conditions at controlled fill volumes (typically 2-10 mL per vial). Residual moisture content is verified by Karl Fischer titration and must not exceed 8.0% (w/w) to ensure product stability.

4. Analytical Method Validation and Quality Control Testing

4.1 Identity Testing Methods

Comprehensive identity confirmation of Hexarelin requires multiple orthogonal analytical techniques to verify primary structure, molecular weight, amino acid composition, and stereochemical configuration. Identity testing is performed on each manufacturing lot and documented within the Certificate of Analysis.

Mass Spectrometry: Electrospray ionization mass spectrometry (ESI-MS) or matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) provides accurate molecular weight determination. The observed molecular ion [M+H]+ at m/z 887.04 ± 0.5 must be consistent with the theoretical mass. High-resolution mass spectrometry confirms elemental composition matching the molecular formula C47H58N12O6.

Amino Acid Analysis: Following complete acid hydrolysis (6 N HCl, 110°C, 24 hours under nitrogen), amino acid composition is determined by ion-exchange chromatography with post-column ninhydrin derivatization or pre-column derivatization followed by RP-HPLC analysis. The molar ratios of detected amino acids must correspond to the theoretical composition: His(1), Trp(2), Ala(1), Phe(1), Lys(1). Note that acid hydrolysis destroys the methyl modification on tryptophan and may cause partial tryptophan degradation, requiring correction factors.

Peptide Sequencing: Edman degradation sequencing or tandem mass spectrometry (MS/MS) fragmentation analysis confirms the amino acid sequence. MS/MS generates characteristic b-ion and y-ion series enabling sequence verification and detection of sequence variants. The presence of D-amino acids at positions 2 and 5 requires chiral analysis methods for complete structural confirmation.

4.2 Purity Determination by RP-HPLC

Reversed-phase HPLC serves as the primary method for purity assessment and is validated according to ICH Q2(R1) guidelines for specificity, linearity, accuracy, precision, detection limit, quantitation limit, and robustness. The analytical method must demonstrate capability to separate Hexarelin from potential impurities including deletion sequences, stereoisomers, oxidation products, and synthesis-related impurities.

Table 5: Validated Analytical RP-HPLC Method for Hexarelin Purity
Parameter Specification
Column C18, 5 μm, 300 Å, 4.6 mm × 250 mm
Mobile Phase A 0.1% TFA in water
Mobile Phase B 0.1% TFA in acetonitrile
Flow Rate 1.0 mL/min
Column Temperature 40°C ± 1°C
Gradient 20-50% B over 30 minutes
Detection Wavelength 220 nm (primary), 280 nm (secondary)
Injection Volume 20 μL
Sample Concentration 1.0 mg/mL in mobile phase A
Run Time 40 minutes (including re-equilibration)
System Suitability Tailing factor ≤2.0, theoretical plates ≥3000

Purity is calculated by area normalization at 220 nm, with the main peak area expressed as a percentage of total integrated area. The acceptance criterion for pharmaceutical-grade Hexarelin is ≥98.0% purity, with individual impurities not exceeding 0.5% and total impurities not exceeding 2.0%. Forced degradation studies during method validation demonstrate the stability-indicating nature of the method by resolving degradation products from the main peak.

4.3 Chiral Purity Assessment

The presence of D-amino acids at positions 2 (D-2-methyl-tryptophan) and 5 (D-phenylalanine) necessitates stereospecific analysis to detect racemization or use of incorrect stereoisomers during synthesis. Chiral purity is assessed through specialized analytical techniques:

  • Chiral RP-HPLC: Using columns containing chiral selectors (e.g., β-cyclodextrin bonded phases) or chiral mobile phase additives
  • Chiral Derivatization: Pre-column derivatization with chiral reagents (e.g., Marfey's reagent) followed by standard RP-HPLC
  • Circular Dichroism Spectroscopy: CD spectra comparison to reference standards confirms overall stereochemical configuration

Specifications require that L-amino acid epimers (resulting from racemization of D-residues) be present at levels not exceeding 0.5% of the total peptide content. This stringent specification ensures maintenance of biological activity, as stereochemical configuration directly impacts receptor binding and pharmacological properties.

4.4 Impurity Profiling and Characterization

Comprehensive impurity profiling identifies and quantifies synthesis-related impurities, degradation products, and process-related contaminants. Major impurity classes include:

  • Deletion Sequences: Peptides lacking one or more amino acid residues due to incomplete coupling
  • Insertion Sequences: Peptides containing additional amino acid residues from double couplings
  • Diastereomers: Peptides with incorrect stereochemistry at D-amino acid positions
  • Oxidation Products: Tryptophan oxidation to N-formylkynurenine or kynurenine
  • Truncated Sequences: Incomplete peptides from premature cleavage
  • Deamidation Products: Although Hexarelin lacks asparagine/glutamine, histidine side-chain modifications may occur

Impurities exceeding 0.1% of total peptide content require structural characterization by LC-MS/MS analysis to establish identity and source. This information guides process optimization to minimize impurity formation in subsequent batches.

5. Manufacturing Batch Specifications and Release Criteria

5.1 Comprehensive Release Testing Panel

Each manufacturing batch of Hexarelin undergoes a complete battery of release tests to verify compliance with established specifications prior to distribution. The Quality Control laboratory executes all testing according to validated standard operating procedures (SOPs) with appropriate equipment calibration and environmental monitoring. All testing must be complete and results within specification before batch release approval.

Table 6: Complete Batch Release Specifications for Hexarelin
Test Method Specification Frequency
Appearance Visual inspection White to off-white lyophilized powder Each batch
Identity (Sequence) MS/MS or Edman degradation Consistent with reference standard Each batch
Identity (MW) ESI-MS or MALDI-TOF [M+H]+ = 887.04 ± 0.5 Da Each batch
Purity (HPLC) RP-HPLC, 220 nm ≥98.0% Each batch
Single Impurity RP-HPLC, 220 nm ≤0.5% each Each batch
Total Impurities RP-HPLC, 220 nm ≤2.0% Each batch
Chiral Purity Chiral HPLC L-epimers ≤0.5% total Each batch
Water Content Karl Fischer titration ≤8.0% (w/w) Each batch
TFA Content Ion chromatography or 19F NMR ≤0.1% (w/w) for acetate salt Each batch
Acetate Content Ion chromatography Report value (typical 5-15%) Each batch
Peptide Content Amino acid analysis or UV ≥85% (as-is basis) Each batch
Endotoxin LAL chromogenic assay ≤5.0 EU/mg Each batch
Bioburden USP <61> ≤10 CFU/g Each batch
Heavy Metals ICP-MS ≤10 ppm total Annual verification
Residual Solvents GC-HS Per ICH Q3C limits Process validation

5.2 Peptide Content Determination

Accurate peptide content quantitation accounts for counter-ions, residual moisture, and other non-peptide components to express results on an as-is basis. Two complementary methods are employed:

Amino Acid Analysis Method: Following complete acid hydrolysis, stable amino acids (Ala, Phe, Lys) are quantified against external standards. Peptide content is calculated from the average recovery of these amino acids, accounting for known hydrolysis losses. This method provides the most accurate assessment but requires 24-48 hours for completion.

UV Spectroscopy Method: A rapid alternative utilizing UV absorbance at 280 nm based on tryptophan content. The extinction coefficient for Hexarelin (ε280 = 11,000 M-1cm-1 based on two tryptophan residues) allows direct concentration determination. This method provides same-day results but may be influenced by oxidative modifications to tryptophan residues.

5.3 Batch Documentation and Traceability

Complete batch records document all manufacturing steps, in-process testing, deviations, and final release testing. Required documentation includes:

  • Master Batch Record with executed step signatures and timestamps
  • Raw material certificates of analysis with vendor qualification records
  • Equipment use logs and calibration certificates
  • Environmental monitoring data (temperature, humidity, particulate counts)
  • In-process testing results (crude purity, intermediate purity)
  • Purification chromatograms and fraction collection records
  • Lyophilization cycle records with temperature/pressure profiles
  • Complete analytical testing data packages
  • Certificate of Analysis signed by QC Manager
  • Batch disposition record with QA approval

All batch records are retained for a minimum of five years beyond expiration date, or as required by applicable regulations. Electronic batch records must be maintained in 21 CFR Part 11 compliant systems with appropriate access controls and audit trails.

6. Stability Studies and Degradation Pathways

6.1 ICH Stability Testing Protocol

Formal stability studies follow ICH Q1A(R2) guidelines for stability testing of new drug substances. Both long-term and accelerated stability studies are conducted to establish shelf life and support registration requirements. Stability studies employ scientifically sound test protocols with validated analytical methods to monitor critical quality attributes over time.

Table 7: ICH Stability Study Conditions for Hexarelin
Study Type Storage Condition Duration Testing Frequency
Long-term -20°C ± 5°C 36 months 0, 3, 6, 9, 12, 18, 24, 36 months
Long-term (refrigerated) 5°C ± 3°C 24 months 0, 3, 6, 9, 12, 18, 24 months
Accelerated 25°C ± 2°C / 60% ± 5% RH 6 months 0, 1, 2, 3, 6 months
Stress (temperature) 40°C ± 2°C / 75% ± 5% RH 3 months 0, 1, 2, 3 months
Photostability ICH Q1B conditions Per protocol End of exposure
Freeze-thaw -20°C to 25°C cycles 5 cycles After each cycle

Stability samples are stored in the intended commercial packaging configuration (typically amber glass vials with rubber stoppers and aluminum seals) to reflect real-world storage conditions. Three independent batches manufactured at commercial scale are placed on formal stability programs to capture batch-to-batch variability.

6.2 Stability-Indicating Parameters

The following quality attributes are monitored at each stability time point:

  • Appearance (color, physical state)
  • Purity by RP-HPLC with impurity profiling
  • Potency/peptide content by amino acid analysis
  • Moisture content by Karl Fischer titration
  • pH of reconstituted solution (if applicable)
  • Aggregation assessment by SEC-HPLC
  • Mass spectrometry for molecular weight verification

Acceptance criteria for stability samples are typically the same as release specifications, with allowances for expected degradation trends. A significant change is defined per ICH guidelines as a 5% decrease in purity, formation of any individual impurity exceeding 1%, or appearance of degradation products not present at release.

6.3 Primary Degradation Pathways

Understanding degradation mechanisms guides formulation development and storage recommendations. Hexarelin exhibits the following primary degradation pathways:

Oxidation: The two tryptophan residues are susceptible to oxidation, particularly at the C2 and C3 positions of the indole ring. Oxidation products include N-formylkynurenine, kynurenine, and hydroxytryptophan derivatives. Oxidation is accelerated by light exposure, elevated temperatures, trace metal contamination, and oxidizing agents. Antioxidants (ascorbic acid, sodium bisulfite) and chelating agents (EDTA) may be incorporated into formulations to minimize oxidative degradation.

Deamidation: While Hexarelin lacks asparagine and glutamine residues (common deamidation sites), the C-terminal amide group may undergo slow hydrolysis to the free carboxylic acid under aqueous conditions. This degradation pathway is pH-dependent and accelerated at alkaline pH values.

Aggregation: The hydrophobic character of Hexarelin promotes self-association and aggregate formation, particularly in concentrated solutions or at elevated temperatures. Physical instability manifests as turbidity, precipitation, or gel formation. Aggregates are monitored by size-exclusion chromatography and must not exceed 2% of total peptide content.

Hydrolysis: Peptide bond cleavage may occur under acidic or basic conditions, with preferential cleavage at Asp-Pro bonds (not present in Hexarelin) or adjacent to hydrophobic residues. The most labile bond is typically between Trp and Ala at position 2-3.

6.4 Recommended Storage Conditions

Based on stability study data, the following storage conditions are recommended for Hexarelin bulk drug substance:

  • Primary Storage: -20°C in sealed containers protected from light
  • Alternative Storage: 2-8°C for up to 24 months (if supported by data)
  • Shipping: Maintain cold chain with temperature monitoring; dry ice for frozen shipments
  • Shelf Life: 36 months at -20°C (typical); confirmed by stability data
  • Container Closure: Inert atmosphere (nitrogen or argon) recommended
  • Light Protection: Amber glass or opaque containers required

7. Handling, Storage, and Formulation Guidelines

7.1 Safe Handling Procedures

Hexarelin is classified as a research-grade biochemical requiring standard laboratory safety precautions. While not classified as a hazardous substance under OSHA criteria, peptide dust generation should be minimized to prevent respiratory exposure or skin contact sensitization. Handling procedures include:

  • Work in well-ventilated areas or under fume hoods when handling bulk quantities
  • Wear appropriate personal protective equipment (safety glasses, lab coat, nitrile gloves)
  • Avoid generating aerosols or dust clouds during weighing and transfer operations
  • Use powder containment systems for large-scale operations
  • Clean spills immediately with damp cloths to prevent dust dispersion
  • Wash hands thoroughly after handling

Safety Data Sheets (SDS) provide additional information on physical properties, hazard classifications, first aid measures, and disposal considerations. All personnel handling Hexarelin must receive appropriate training on safe handling procedures and emergency response protocols.

7.2 Reconstitution and Solution Preparation

Lyophilized Hexarelin requires reconstitution prior to use in biological assays, formulation development, or analytical testing. Optimal reconstitution practices ensure complete dissolution while minimizing degradation:

Table 8: Recommended Reconstitution Protocols
Solvent System Concentration Range Stability Applications
Sterile water 0.1-5.0 mg/mL Use within 24 hours at 4°C Immediate use applications
0.9% saline 0.5-10.0 mg/mL 48 hours at 4°C Physiological formulations
10 mM acetic acid 1.0-5.0 mg/mL 1 week at 4°C Stock solutions
PBS pH 7.4 0.1-2.0 mg/mL 24 hours at 4°C Biological assays
50% acetonitrile/water 1.0-10.0 mg/mL 1 month at -20°C Analytical standards

Reconstitution protocol: Add solvent slowly down the side of the vial to avoid generating foam. Allow peptide to dissolve naturally for 2-5 minutes without agitation. Gentle swirling may be employed if necessary, but avoid vortexing or vigorous shaking which can cause aggregation and denaturation. If cloudiness persists, brief sonication (30-60 seconds) in an ice bath may aid dissolution. Filter solutions through 0.22 μm filters if sterility is required.

7.3 Formulation Development Considerations

Development of stable liquid or lyophilized formulations for pharmaceutical applications requires optimization of multiple parameters to maintain peptide stability during manufacturing, storage, and use. Key formulation components include:

Buffer Systems: pH control between 4.0-6.0 typically provides optimal stability for Hexarelin. Acetate (10-50 mM), citrate (10-30 mM), or histidine (5-20 mM) buffers are commonly employed. Phosphate buffers should be avoided due to potential metal ion catalyzed oxidation.

Cryoprotectants/Lyoprotectants: Sugars and polyols protect peptide structure during freeze-drying and storage. Common excipients include mannitol (2-5% w/v), trehalose (1-3% w/v), or sucrose (3-7% w/v). These agents maintain protein structure during water removal and form amorphous glasses that provide long-term stabilization.

Surfactants: Non-ionic surfactants (polysorbate 20 or 80 at 0.01-0.1% w/v) reduce surface adsorption and aggregation, particularly important for low-concentration formulations or storage in plastic containers.

Antioxidants: Methionine (0.1-0.5% w/v), ascorbic acid (0.01-0.1% w/v), or sodium bisulfite (0.05-0.2% w/v) scavenge free radicals and protect tryptophan residues from oxidation.

Chelating Agents: EDTA (0.01-0.1% w/v) or citrate chelates trace metal ions that catalyze oxidative degradation pathways.

Formulation development follows a systematic approach with forced degradation studies, Design of Experiments (DoE) optimization, and accelerated stability assessment to identify robust compositions meeting target shelf life requirements.

7.4 Environmental and Waste Disposal

Hexarelin waste materials, including spent synthesis resins, purification fractions below specification, and expired product, require proper disposal according to institutional policies and local regulations. Recommended disposal procedures include:

  • Small quantities may be deactivated by autoclaving or chemical inactivation (10% bleach solution, 1-hour contact time)
  • Solid waste should be disposed in accordance with biohazard waste protocols
  • Liquid waste containing organic solvents requires disposal through chemical waste streams
  • Large quantities should be incinerated at licensed hazardous waste facilities
  • Do not discharge untreated peptide solutions into municipal wastewater systems

8. Certificate of Analysis Documentation

8.1 Required CoA Elements

Each batch of Hexarelin is accompanied by a Certificate of Analysis providing comprehensive documentation of quality attributes and regulatory compliance. The CoA serves as the official quality document certifying that the batch meets all established specifications and is suitable for its intended use. Required elements include:

Table 9: Standard Certificate of Analysis Format
Section Required Information
Product Information Product name, CAS number, catalog number, molecular formula, molecular weight
Batch Information Batch/lot number, manufacturing date, retest/expiration date, quantity manufactured
Storage Conditions Recommended storage temperature, light protection requirements, container specifications
Test Results All release tests with methods, specifications, and actual results
Chromatograms Representative HPLC chromatogram showing purity profile
Mass Spectrum ESI-MS or MALDI-TOF spectrum confirming molecular weight
Approvals QC manager signature, release date, company information
Disclaimer Intended use statement, research use only designation (if applicable)

8.2 Sample Certificate of Analysis

CERTIFICATE OF ANALYSIS


Product Name: Hexarelin
Sequence: His-D-2-Methyl-Trp-Ala-Trp-D-Phe-Lys-NH2
Molecular Formula: C47H58N12O6
Molecular Weight: 887.04 g/mol
CAS Number: 140703-51-1

Batch Number: HEX-2025-0423
Manufacturing Date: October 15, 2025
Retest Date: October 15, 2028
Net Weight: 5.00 g
Storage: Store at -20°C, protected from light

Test Method Specification Result
Appearance Visual White to off-white powder White powder
Identity (MW) ESI-MS 887.04 ± 0.5 887.2 [M+H]+
Purity (HPLC) RP-HPLC ≥98.0% 98.7%
Single Impurity RP-HPLC ≤0.5% 0.3% (max)
Water Content Karl Fischer ≤8.0% 4.2%
Peptide Content AAA ≥85% 89.3%
Endotoxin LAL ≤5.0 EU/mg <2.0 EU/mg

Conclusion: This batch meets all established specifications.
Approved by: Dr. Sarah Mitchell, QC Manager
Release Date: October 23, 2025
For research use only. Not for human or veterinary use.

8.3 CoA Retention and Traceability

Certificates of Analysis are permanently retained in both paper and electronic formats. Electronic copies are stored in quality management systems with controlled access and backup procedures. Customer copies are provided with each shipment and are also available through secure web portals for download and verification. Batch-specific CoAs enable full traceability from raw materials through final product distribution.

9. Regulatory Considerations and Compliance

9.1 Regulatory Status and Classification

Hexarelin is classified as a research chemical and growth hormone secretagogue analog. The regulatory status varies by jurisdiction and intended application. Manufacturing operations must comply with applicable regulations based on the peptide's intended use:

Research Use Materials: Hexarelin manufactured and distributed for laboratory research applications must be clearly labeled "For Research Use Only - Not for Human or Veterinary Use." Such materials are not subject to FDA approval requirements but must still be manufactured under quality systems ensuring product identity, purity, and consistency.

Clinical Trial Materials: Hexarelin intended for use in clinical investigations must be manufactured under cGMP conditions as defined in 21 CFR Parts 210 and 211. An Investigational New Drug (IND) application must be submitted to the FDA prior to clinical use, including comprehensive CMC (Chemistry, Manufacturing, and Controls) documentation.

Commercial Drug Substance: Hexarelin manufactured as an active pharmaceutical ingredient for commercial drug products requires full regulatory approval (NDA or ANDA) and must meet all applicable pharmacopeial standards. Manufacturing facilities are subject to pre-approval and routine inspections by regulatory authorities.

9.2 cGMP Compliance Requirements

Manufacturing facilities producing pharmaceutical-grade Hexarelin must implement comprehensive quality systems meeting current Good Manufacturing Practice requirements. Essential elements include:

  • Validated manufacturing processes with documented process validation protocols
  • Qualified equipment with calibration and maintenance programs
  • Trained personnel with documented qualification records
  • Controlled manufacturing environments with environmental monitoring
  • Raw material qualification and approved vendor programs
  • Robust analytical method validation per ICH Q2(R1)
  • Stability programs following ICH Q1A(R2) guidelines
  • Change control and deviation management systems
  • CAPA (Corrective and Preventive Action) programs
  • Regular internal audits and supplier audits

9.3 Documentation and Regulatory Filing Support

Comprehensive technical documentation packages support regulatory filings for clinical trial applications or commercial drug approvals. Required documentation includes:

  • Drug Substance Specification with justification for acceptance criteria
  • Manufacturing process description with process flow diagrams
  • Control of critical process parameters and in-process controls
  • Impurity profile characterization and qualification
  • Analytical method validation reports for all release and stability methods
  • Stability study protocols and stability data summaries
  • Reference standard qualification documentation
  • Container closure system qualification
  • Facility and equipment descriptions
  • Environmental monitoring programs

All documentation must be prepared according to ICH Common Technical Document (CTD) format for international regulatory submissions.

10. Supply Chain Management and Quality Assurance

10.1 Raw Material Qualification

Consistent manufacturing of high-quality Hexarelin requires rigorous qualification and control of all raw materials, including protected amino acids, coupling reagents, resins, and solvents. A formal vendor qualification program ensures suppliers meet defined quality standards:

Vendor Assessment: Potential suppliers undergo evaluation including quality system audits, review of manufacturing capabilities, assessment of quality documentation systems, and verification of regulatory compliance. Approved vendors are maintained on an Approved Supplier List with periodic re-qualification.

Raw Material Testing: Each lot of critical raw materials undergoes incoming inspection and testing prior to release for manufacturing use. Protected amino acids require verification of identity (NMR, MS), purity (HPLC), optical rotation, and absence of racemization. Coupling reagents are tested for identity, purity, and moisture content. Certificates of Analysis from suppliers are reviewed and verified through internal testing.

Change Control: Any changes to raw material suppliers, manufacturing sites, or specifications require formal change control evaluation including stability assessment and batch comparison studies to demonstrate equivalency.

10.2 Process Validation and Lifecycle Management

Process validation demonstrates that the manufacturing process consistently produces Hexarelin meeting all predetermined quality attributes. Validation follows a lifecycle approach including process design, process qualification, and continued process verification:

Stage 1 - Process Design: Development activities establish understanding of process parameters, identify critical quality attributes (CQAs) and critical process parameters (CPPs), and define a control strategy. Design of Experiments studies optimize synthesis conditions, coupling sequences, and purification parameters.

Stage 2 - Process Qualification: Three consecutive commercial-scale batches are manufactured under fully controlled conditions following approved manufacturing procedures. Each validation batch undergoes comprehensive testing including all release tests plus additional characterization to demonstrate process consistency and capability.

Stage 3 - Continued Process Verification: Ongoing statistical process control monitors process performance through trending of critical parameters and quality attributes. Annual product quality reviews assess process capability and identify opportunities for continuous improvement.

10.3 Distribution and Logistics

Maintaining Hexarelin quality during distribution requires controlled shipping conditions with temperature monitoring and validation of shipping configurations:

  • Frozen shipments utilize dry ice in validated shippers maintaining -20°C for specified durations
  • Refrigerated shipments employ gel packs or active refrigeration systems maintaining 2-8°C
  • Temperature data loggers accompany shipments documenting temperature exposure throughout transit
  • Packaging qualification studies demonstrate thermal performance under worst-case conditions
  • Temperature excursion protocols define acceptable limits and investigation procedures

Distribution partners undergo qualification to verify capability to maintain cold chain integrity and handle temperature-sensitive biological materials according to established procedures.

10.4 Quality Management Systems

Comprehensive quality management systems ensure consistent adherence to established specifications and regulatory requirements. Key system elements include:

  • Document Control: Controlled document management system for SOPs, specifications, protocols, and technical documentation with version control and approval workflows
  • Training Management: Personnel qualification programs with documented training records and competency assessment
  • Investigation Management: Formal investigation procedures for out-of-specification results, deviations, and quality complaints with root cause analysis and CAPA implementation
  • Change Control: Systematic evaluation and approval process for any changes to materials, equipment, processes, or specifications
  • Internal Audits: Scheduled self-inspection programs assessing compliance with established procedures and regulatory requirements
  • Management Review: Periodic quality management reviews evaluating system effectiveness and continuous improvement initiatives

References and Additional Resources

Scientific Literature and Technical Standards

  1. Deghenghi, R., et al. "GH-releasing peptides." Recent Progress in Hormone Research 56 (2001): 63-77. https://pubmed.ncbi.nlm.nih.gov/11237228/
  2. International Conference on Harmonisation. "ICH Q7: Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients." ICH, 2000. https://www.ich.org/page/quality-guidelines
  3. Fields, G.B., and Noble, R.L. "Solid phase peptide synthesis utilizing 9-fluorenylmethoxycarbonyl amino acids." International Journal of Peptide and Protein Research 35.3 (1990): 161-214. https://pubmed.ncbi.nlm.nih.gov/2191922/
  4. United States Pharmacopeia. "General Chapter <1045> Biotechnology-Derived Articles." USP 44-NF 39, 2021. https://www.usp.org/
  5. International Conference on Harmonisation. "ICH Q2(R1): Validation of Analytical Procedures: Text and Methodology." ICH, 2005. https://www.ich.org/page/quality-guidelines
  6. International Conference on Harmonisation. "ICH Q1A(R2): Stability Testing of New Drug Substances and Products." ICH, 2003. https://www.ich.org/page/quality-guidelines
  7. Amblard, M., et al. "Methods and protocols of modern solid phase peptide synthesis." Molecular Biotechnology 33.3 (2006): 239-254. https://pubmed.ncbi.nlm.nih.gov/16946453/
  8. Mant, C.T., and Hodges, R.S. "High-performance liquid chromatography of peptides and proteins: Separation, analysis, and conformation." Methods in Enzymology 271 (1996). https://www.sciencedirect.com/bookseries/methods-in-enzymology
  9. U.S. Food and Drug Administration. "Guidance for Industry: Q1B Photostability Testing of New Drug Substances and Products." FDA, 1996. https://www.fda.gov/regulatory-information/search-fda-guidance-documents
  10. Manning, M.C., et al. "Stability of protein pharmaceuticals: An update." Pharmaceutical Research 27.4 (2010): 544-575. https://pubmed.ncbi.nlm.nih.gov/20143256/

Internal Resources

Conclusion

This manufacturing profile provides comprehensive technical documentation for the production of pharmaceutical-grade Hexarelin through solid-phase peptide synthesis, preparative purification, and rigorous quality control testing. The protocols and specifications outlined herein ensure consistent manufacture of high-purity Hexarelin meeting international quality standards for research applications and pharmaceutical development.

Successful Hexarelin manufacturing requires strict adherence to validated procedures, maintenance of controlled manufacturing environments, use of qualified raw materials, and implementation of comprehensive analytical testing programs. The incorporation of non-standard amino acids and multiple hydrophobic residues necessitates specialized synthesis strategies and aggregation control measures throughout the production process.

Quality assurance systems supporting Hexarelin manufacture must encompass raw material qualification, process validation, environmental monitoring, stability assessment, and comprehensive batch documentation. These systems ensure product consistency, regulatory compliance, and full traceability from raw materials through final distribution.

As peptide manufacturing technology continues to advance, opportunities exist for process optimization including implementation of continuous flow synthesis, development of more selective purification strategies, and formulation innovations to enhance stability profiles. Ongoing commitment to quality improvement and regulatory compliance remains essential for meeting the evolving needs of the peptide manufacturing industry.

For additional technical support, custom manufacturing inquiries, or regulatory consultation services regarding Hexarelin production, contact our technical services team at [email protected] or visit our comprehensive manufacturing capabilities portal.

Document Revision: 1.0 | Last Updated: October 2025 | Classification: Technical Manufacturing Documentation

© 2025 PeptideForge.com - All Rights Reserved | Quality Policy | Terms of Service