GHRP-2 Manufacturing Profile: Synthesis, Purification & Quality Control Specifications

GHRP-2 Manufacturing Profile: Comprehensive Process Documentation and Quality Specifications

1. Manufacturing Overview and Molecular Characterization

Growth Hormone Releasing Peptide-2 (GHRP-2) represents a class of synthetic hexapeptides designed to stimulate growth hormone secretion through ghrelin receptor activation. Manufacturing this peptide requires strict adherence to current Good Manufacturing Practice (cGMP) regulations and validated process controls to ensure consistent product quality, purity, and potency across production batches.

The molecular formula C45H55N9O6 corresponds to a calculated molecular weight of 817.97 Da for the free base form. Manufacturing specifications must account for the peptide's hygroscopic nature and susceptibility to oxidative degradation during synthesis, purification, and storage phases. Process development for GHRP-2 production follows established solid-phase peptide synthesis (SPPS) protocols with specific modifications to accommodate the peptide's unique sequence: D-Ala-D-2-Nal-Ala-Trp-D-Phe-Lys-NH2.

This manufacturing profile documents standard operating procedures, critical quality attributes (CQAs), and acceptance criteria employed in commercial-scale GHRP-2 production. The documented processes align with ICH Q7 guidelines for active pharmaceutical ingredient manufacturing and incorporate risk-based quality control strategies specific to synthetic peptide production.

Table 1: GHRP-2 Molecular Specifications
Parameter Specification Method Reference
Chemical Name D-Alanyl-3-(2-naphthyl)-D-alanyl-L-alanyl-L-tryptophyl-D-phenylalanyl-L-lysinamide IUPAC Nomenclature
Sequence D-Ala-D-2-Nal-Ala-Trp-D-Phe-Lys-NH2 Standard Three-Letter Code
Molecular Formula C45H55N9O6 Elemental Analysis
Molecular Weight 817.97 Da (free base) Mass Spectrometry (ESI-MS)
CAS Number 158861-67-7 Chemical Abstracts Service
Appearance White to off-white lyophilized powder Visual Inspection
Solubility Freely soluble in water, DMSO; sparingly soluble in ethanol Ph. Eur. 2.9.40

2. Solid-Phase Peptide Synthesis (SPPS) Process

GHRP-2 production employs Fmoc (9-fluorenylmethyloxycarbonyl) solid-phase peptide synthesis methodology using automated peptide synthesizers validated for GMP manufacturing. The synthesis proceeds from C-terminus to N-terminus on a Rink amide resin substrate, which provides the required C-terminal amide functionality critical to GHRP-2's biological activity. Selection of Fmoc chemistry over Boc chemistry minimizes harsh acidic conditions during synthesis, reducing racemization risks at stereochemically sensitive D-amino acid positions.

2.1 Resin Selection and Loading

Manufacturing begins with Rink amide MBHA resin (100-200 mesh, 0.4-0.7 mmol/g loading capacity) pre-qualified through supplier certification and in-house testing. Resin batches undergo swelling tests in N,N-dimethylformamide (DMF) and loading capacity verification via UV spectrophotometry following Fmoc deprotection. Target loading density of 0.5 mmol/g represents optimal balance between synthesis efficiency and steric accessibility during coupling reactions.

The first amino acid coupling attaches Fmoc-Lys(Boc)-OH to the resin using HBTU (O-benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluorophosphate) activation in the presence of DIPEA (N,N-diisopropylethylamine). Double coupling protocols ensure >99.5% incorporation at this initial step, verified through quantitative ninhydrin testing or Fmoc quantification of wash fractions.

2.2 Coupling Cycle Parameters

Each subsequent amino acid coupling follows standardized activation and coupling protocols with parameters adjusted based on residue reactivity profiles. D-amino acids and sterically hindered residues (D-2-Nal, D-Phe) require extended coupling times and elevated temperatures to achieve acceptable incorporation rates. Process validation establishes critical coupling parameters including reagent equivalents, activation time, coupling duration, and reaction temperature for each position.

Table 2: Standard Coupling Cycle Parameters
Step Reagent/Condition Duration Temperature
Resin Swelling DMF 15 min 25°C
Fmoc Deprotection 20% piperidine in DMF 2 × 5 min 25°C
Wash DMF 5 × 1 min 25°C
Amino Acid Activation Fmoc-AA (4 eq), HBTU (3.95 eq), DIPEA (6 eq) in DMF 2 min 25°C
Coupling Activated amino acid solution 45-60 min 25-40°C
Wash DMF 5 × 1 min 25°C
Coupling Verification Ninhydrin or Kaiser test 5 min 100°C (test)

2.3 Critical Coupling Considerations

Position-specific coupling challenges require process modifications at three key residues. The D-2-naphthylalanine (D-2-Nal) incorporation at position 2 presents steric hindrance requiring double or triple coupling with 6-8 equivalents of amino acid and extended reaction times (90-120 minutes) at elevated temperature (40°C). Coupling efficiency monitoring through micro-cleavage and HPLC analysis ensures >98% incorporation before proceeding to subsequent residues.

Tryptophan coupling at position 4 requires protection against indole alkylation during subsequent synthesis steps. Standard Fmoc-Trp(Boc)-OH provides adequate protection while maintaining coupling reactivity. Critical process parameters include limiting reaction temperature to 25°C and minimizing base exposure during deprotection steps to prevent indole degradation.

The two D-phenylalanine residues necessitate careful monitoring for racemization. Process validation studies demonstrate that standard coupling conditions with HBTU activation maintain stereochemical integrity when coupling times do not exceed 60 minutes and reaction temperatures remain below 30°C. Periodic chiral HPLC analysis during process qualification verifies D-configuration retention at >99.5%.

2.4 Final Deprotection and Cleavage

Following complete sequence assembly and final Fmoc removal, peptide-resin undergoes simultaneous side-chain deprotection and resin cleavage using Reagent K formulation: TFA/phenol/water/thioanisole/ethanedithiol (82.5:5:5:5:2.5, v/v/v/v/v). This cocktail composition provides comprehensive protection against tryptophan alkylation, methionine oxidation, and lysine side reactions during the acid cleavage process. Cleavage proceeds for 2.5-3.0 hours at room temperature with nitrogen overlay to minimize oxidative side reactions.

Post-cleavage processing involves filtration to remove spent resin, peptide precipitation using cold diethyl ether (10× volume), and multiple ether washes to remove scavengers and TFA. The crude peptide pellet undergoes vacuum drying to remove residual ether, followed by dissolution in 10% acetic acid solution for purification processing. Crude peptide purity typically ranges from 50-70% as determined by analytical RP-HPLC, with primary impurities consisting of deletion sequences, incomplete deprotection products, and tryptophan oxidation variants.

3. Purification Process and Method Validation

GHRP-2 purification employs multi-stage reverse-phase high-performance liquid chromatography (RP-HPLC) to achieve pharmaceutical-grade purity specifications. The purification strategy combines preparative-scale chromatography for bulk purification followed by semi-preparative polishing steps to remove closely-eluting impurities. Process development establishes validated chromatographic conditions that demonstrate robustness across multiple manufacturing batches while maintaining consistent product recovery and purity profiles.

3.1 Preparative Chromatography

Initial purification utilizes preparative RP-HPLC columns (C18, 250 × 50 mm, 10 μm particle size) with gradient elution systems employing mobile phase optimization for peptide separation. Mobile phase A consists of 0.1% TFA in water (HPLC grade), while mobile phase B contains 0.1% TFA in acetonitrile. The shallow gradient profile (15-35% B over 45 minutes) provides adequate resolution between GHRP-2 and primary synthesis-related impurities.

Crude peptide loading concentration maintains 50-100 mg/mL in 10% acetic acid, with injection volumes of 100-200 mL per run depending on column capacity. Flow rate optimization at 80-100 mL/min balances separation efficiency with processing throughput. UV detection at 220 nm and 280 nm allows simultaneous monitoring of peptide backbone absorbance and aromatic residue content. Fraction collection triggers automatically based on pre-defined UV thresholds, with target fractions collecting when absorbance exceeds 1000 mAU and terminating when absorbance falls below 200 mAU.

Table 3: Preparative Purification Parameters
Parameter Specification Rationale
Column C18, 250 × 50 mm, 10 μm Adequate loading capacity for preparative scale
Mobile Phase A 0.1% TFA in water Ion pairing and pH control
Mobile Phase B 0.1% TFA in acetonitrile Organic modifier for peptide elution
Gradient 15-35% B over 45 min Optimized resolution of target from impurities
Flow Rate 80-100 mL/min Balance of resolution and throughput
Detection UV 220 nm, 280 nm Peptide bond and aromatic monitoring
Column Temperature 25 ± 2°C Temperature control for reproducibility
Load Concentration 50-100 mg/mL Column capacity optimization

3.2 Semi-Preparative Polishing

Collected fractions from preparative purification undergo purity assessment via analytical HPLC. Fractions meeting minimum purity criteria (≥90% by HPLC) are pooled for semi-preparative polishing using narrower bore columns (C18, 250 × 20 mm, 5 μm) with refined gradient conditions. The polishing gradient (20-30% B over 40 minutes) provides enhanced resolution of remaining impurities while maintaining target peptide recovery above 85%.

Semi-preparative purification operates at 10-15 mL/min flow rate with reduced sample loading (20-30 mg per injection) to maximize resolution. Fraction collection employs tighter UV threshold windows, collecting only when absorbance exceeds 500 mAU and terminating below 100 mAU. This conservative collection strategy sacrifices moderate yield to ensure final product purity exceeds 98% by analytical HPLC.

3.3 Desalting and TFA Exchange

Purified peptide fractions contain residual TFA from mobile phase systems, requiring desalting procedures to achieve pharmaceutical-grade specifications. Size-exclusion chromatography using Sephadex G-25 columns (100 × 5 cm) eluted with 10% acetic acid effectively removes TFA and acetonitrile while maintaining peptide integrity. Alternative desalting employs C18 solid-phase extraction cartridges with sequential washes using water and dilute acetic acid, followed by peptide elution in 50% acetonitrile/0.1% acetic acid.

TFA content in final product must not exceed 0.5% w/w as determined by ion chromatography or 19F NMR quantification. Residual TFA above this threshold requires additional desalting cycles or alternative counter-ion exchange using HCl or acetic acid. Process validation demonstrates that two sequential desalting cycles consistently reduce TFA content below specification limits while maintaining peptide recovery above 90%.

3.4 Lyophilization Process

Desalted peptide solutions undergo lyophilization to produce stable, free-flowing powder suitable for long-term storage and distribution. Pre-lyophilization formulation involves dissolution in dilute acetic acid (10-20 mM) at pH 4.0-5.0, which provides optimal peptide stability during freezing and primary drying phases. Addition of lyoprotectants (mannitol 2-5% w/v) prevents aggregation and maintains peptide structural integrity during freeze-drying cycles.

Validated lyophilization cycles employ controlled freezing rates (-1°C/min to -45°C), primary drying at -25°C under 100 mTorr vacuum for 24-36 hours, and secondary drying at 25°C under 50 mTorr for 6-12 hours. Cycle development studies using thermocouples and pressure rise tests verify complete sublimation before secondary drying initiation. Final moisture content specifications (≤3.0% by Karl Fischer titration) ensure product stability during storage.

4. Quality Control Testing and Release Specifications

Comprehensive quality control testing follows ICH Q6A specifications for peptide characterization, incorporating identity, purity, potency, and safety testing methodologies. Each production batch undergoes complete analytical characterization prior to release certification, with all testing performed in qualified laboratories using validated analytical methods. Quality control strategy employs risk-based testing protocols that focus on critical quality attributes identified during process development and characterization studies.

4.1 Identity Testing

Identity confirmation employs orthogonal analytical techniques providing complementary structural information. Primary identification uses electrospray ionization mass spectrometry (ESI-MS) confirming molecular weight within ±0.5 Da of theoretical value (817.97 Da for free base). High-resolution mass spectrometry provides additional specificity through accurate mass determination and isotope pattern analysis.

Secondary identification employs analytical RP-HPLC retention time comparison against qualified reference standard, with acceptance criteria of ±2% retention time variation. Additional identity confirmation through amino acid analysis verifies molar ratios of constituent residues within ±10% of theoretical composition. For critical batches or investigational purposes, complete sequence verification via MS/MS fragmentation analysis confirms peptide sequence and D-amino acid positioning.

4.2 Purity Analysis

Purity assessment combines multiple chromatographic and spectrometric techniques to detect and quantify potential impurities from synthesis, purification, and storage. Primary purity determination employs validated analytical RP-HPLC methods using linear gradient elution from 10-50% acetonitrile over 40 minutes on C18 columns (250 × 4.6 mm, 5 μm). UV detection at 220 nm provides comprehensive detection of peptide-related impurities with quantification limits of 0.1% for individual impurities and 0.05% reporting threshold.

Table 4: Purity Specifications and Testing Methods
Test Method Specification
Purity (HPLC) RP-HPLC, UV 220 nm ≥98.0%
Single Largest Impurity RP-HPLC, UV 220 nm ≤1.0%
Total Impurities RP-HPLC, UV 220 nm ≤2.0%
Tryptophan Oxidation RP-HPLC, fluorescence detection ≤0.5%
D/L Isomerization Chiral HPLC ≤0.5% each position
Deletion Sequences LC-MS ≤0.3% individual
Related Peptides RP-HPLC Report values ≥0.1%

Complementary purity assessment using size-exclusion chromatography (SEC) detects aggregated species and high molecular weight impurities not resolved by RP-HPLC. SEC specifications require monomer content ≥97% with individual aggregate species ≤1.5%. Capillary electrophoresis provides orthogonal separation mechanism based on charge differences, detecting ionic impurities and incomplete deprotection products.

4.3 Counter-Ion and Residual Solvent Analysis

Residual TFA content undergoes quantification via ion chromatography or 19F NMR spectroscopy with specification limits ≤0.5% w/w. Acetonitrile residual solvent analysis by gas chromatography must demonstrate levels below ICH Q3C Class 2 limits (410 ppm). Residual water content determination via Karl Fischer titration maintains specification of ≤3.0% to ensure product stability during storage.

For acetate salt formulations, acetate content verification uses ion chromatography with specifications based on stoichiometric ratios accounting for peptide charge state at physiological pH. Heavy metals testing per USP <231> ensures lead, mercury, arsenic, and cadmium levels remain below pharmacopeial limits.

4.4 Peptide Content and Potency

Peptide content determination employs amino acid analysis following acid hydrolysis (6N HCl, 110°C, 24 hours) with correction factors for destruction/incomplete hydrolysis of specific residues. Content calculation uses average of stable amino acid recoveries (Ala, Phe, Lys) compared to external standard curve, with acceptance range of 95-105% of theoretical content adjusted for moisture and TFA content.

Alternative peptide content determination uses quantitative HPLC against characterized reference standard, employing external standardization with five-point calibration curve. UV absorbance measurements at 280 nm provide supplementary content estimation based on theoretical extinction coefficient calculated from tryptophan and phenylalanine contributions (ε280 = 11,460 M-1cm-1).

4.5 Microbiological Testing

Sterility testing following USP <71> employs membrane filtration method with inoculation into thioglycollate medium (aerobic bacteria) and soybean-casein digest medium (fungi). Endotoxin testing via kinetic chromogenic LAL assay maintains specification ≤1.0 EU/mg, suitable for research applications. Total aerobic microbial count (TAMC) and total combined yeasts/molds count (TYMC) testing per USP <61> applies to non-sterile formulations with specifications derived from intended use risk assessment.

Bioburden monitoring throughout manufacturing process identifies contamination control points and verifies effectiveness of sanitization procedures. Environmental monitoring programs track viable and non-viable particulates in manufacturing suites, with alert and action limits established through baseline characterization studies.

5. Batch Manufacturing Specifications and Process Controls

Commercial-scale GHRP-2 manufacturing operates under validated batch protocols defining process parameters, in-process controls, and critical quality attributes requiring monitoring throughout production. Batch size determination balances equipment capacity, market demand, and product stability considerations, with typical manufacturing lots ranging from 10-100 grams of purified peptide. Process validation studies demonstrate consistency across three consecutive conforming batches at commercial scale before routine production implementation.

5.1 Scale-Up Considerations

Transition from development-scale to commercial-scale production requires systematic evaluation of scale-dependent parameters including mixing efficiency, heat transfer, reaction kinetics, and chromatographic loading. Synthesis scale-up maintains constant resin loading density and reagent equivalents while proportionally increasing reactor volume. Validation protocols verify that coupling efficiencies, deprotection completeness, and crude purity profiles remain consistent across scale transitions.

Chromatographic scale-up follows linear scaling principles maintaining bed height, linear flow velocity, and sample load per unit column volume. Column diameter increases proportionally to achieve target processing capacity while preserving separation resolution. Process development studies establish maximum allowable loading factors that maintain product recovery and purity specifications across column scales from analytical (4.6 mm) through preparative (50 mm) dimensions.

5.2 In-Process Controls

Critical in-process controls monitor synthesis progression and identify deviation from validated process parameters requiring corrective action before proceeding to subsequent steps. Real-time monitoring includes Kaiser/ninhydrin test results at each coupling cycle (negative test required before proceeding), pH verification of deprotection and coupling solutions (pH 8-9 for coupling, pH 2-3 post-deprotection), and reaction temperature maintenance within ±2°C of target values.

Table 5: Critical In-Process Controls
Process Stage Control Parameter Acceptance Criteria Action on Failure
Resin Preparation Loading capacity 0.45-0.55 mmol/g Reject resin lot
Each Coupling Ninhydrin test Negative (blue color absent) Repeat coupling cycle
Deprotection UV absorbance (301 nm) ≥95% of theoretical Extend deprotection time
Crude Cleavage HPLC purity ≥50% Investigate synthesis failure
Post-Prep HPLC Pool purity ≥90% Re-purify or discard
Post-Polish HPLC Purity ≥98% Additional polishing step
Pre-Lyophilization TFA content ≤0.5% w/w Additional desalting
Post-Lyophilization Moisture content ≤3.0% Extended secondary drying

5.3 Environmental Monitoring

Manufacturing suite classification and environmental control maintains ISO Class 7 (Class 10,000) conditions during synthesis operations and ISO Class 8 (Class 100,000) during purification and lyophilization. Positive pressure differentials (10-15 Pa) relative to adjacent lower-classification areas prevent ingress of uncontrolled air. HEPA filtration systems provide minimum 20 air changes per hour with validated particulate removal efficiency ≥99.97% for 0.3 μm particles.

Temperature and humidity monitoring maintains environmental conditions of 20-25°C and 30-50% relative humidity during manufacturing operations. Deviations outside these ranges trigger investigation of potential impact on product quality, with corrective actions documented in batch production records. Continuous monitoring systems record environmental parameters at 5-minute intervals with automatic alert generation upon exceeding action limits.

5.4 Raw Material Qualification

All raw materials undergo supplier qualification and material testing prior to release for manufacturing use. Fmoc-protected amino acids require certificate of analysis documenting purity ≥99% by HPLC, enantiomeric purity ≥99% by chiral analysis, and identification confirmation via NMR or mass spectrometry. Coupling reagents (HBTU, DIPEA) require purity verification and water content determination, with anhydrous materials maintaining Karl Fischer results ≤0.1%.

Solvents for synthesis and purification meet ACS or HPLC grade specifications with batch-specific testing for UV absorbance, residue on evaporation, and relevant impurities. Incoming resin lots undergo swelling tests, loading capacity verification, and coupling efficiency evaluation using test sequences before release for GMP manufacturing. Vendor qualification programs include on-site audits, quality agreements, and change control notification requirements ensuring supply chain integrity.

6. Stability Studies and Degradation Pathway Characterization

Comprehensive stability programs establish product shelf-life, define storage conditions, and characterize degradation pathways that may compromise product quality during storage and handling. Stability protocol design follows ICH Q1A(R2) guidelines for stability testing of new drug substances, incorporating long-term, accelerated, and stress testing conditions. Stability-indicating analytical methods capable of separating and quantifying degradation products receive validation for specificity, accuracy, precision, and linearity across expected degradation product concentration ranges.

6.1 Long-Term Stability Studies

Long-term stability testing maintains storage at -20°C ± 5°C in sealed containers protected from light and moisture. Testing intervals at 0, 3, 6, 9, 12, 18, 24, and 36 months evaluate appearance, purity by RP-HPLC, peptide content, moisture content, and specific degradation products. Stability commitment protocols extend testing to 60 months to support extended expiration dating and confirm degradation kinetic models established during initial studies.

Stability acceptance criteria require maintenance of purity ≥97.0% throughout shelf-life with individual degradation products ≤1.5% and total impurities ≤3.0%. Peptide content must remain 90-110% of initial assay value after correction for moisture and counter-ion content. Moisture content specifications permit gradual equilibration to ≤5.0% over extended storage periods without triggering batch failure provided purity specifications remain within limits.

Table 6: Stability Study Design and Acceptance Criteria
Study Type Conditions Duration Testing Frequency
Long-Term -20°C ± 5°C 36-60 months 0, 3, 6, 9, 12, 18, 24, 36, 48, 60 months
Accelerated 5°C ± 3°C 12 months 0, 3, 6, 12 months
Intermediate 25°C/60% RH ± 2°C/5% RH 6 months 0, 3, 6 months
Stress Testing 40°C/75% RH ± 2°C/5% RH 3 months 0, 1, 2, 3 months
Freeze-Thaw -20°C to 25°C cycles 5 cycles After each cycle
Light Exposure ICH Option 2 (1.2M lux·hr visible, 200 W·hr/m² UV) Single timepoint End of exposure
pH Stability pH 2, 4, 7, 9 at 25°C 7 days 0, 1, 3, 7 days
Oxidative Stress 0.1% H₂O₂ at 25°C 24 hours 0, 6, 24 hours

6.2 Accelerated and Stress Testing

Accelerated stability studies at 5°C ± 3°C over 12 months predict degradation rates under refrigerated storage conditions commonly employed for peptide handling in research laboratories. Testing intervals at 0, 3, 6, and 12 months assess degradation kinetics and support extrapolation of shelf-life predictions. Arrhenius modeling of temperature-dependent degradation rates correlates accelerated study results with long-term storage data, validating predictive models for expiration date assignment.

Stress testing under extreme conditions (40°C/75% RH) elucidates primary degradation pathways and validates stability-indicating capability of analytical methods. Major degradation products include tryptophan oxidation variants (N-formylkynurenine, kynurenine), methionine oxidation products, deamidation products (though GHRP-2 contains no Asn/Gln), and disulfide formation between lysine side chains and oxidized tryptophan residues. Characterization via LC-MS/MS identifies degradation structures and supports degradation mechanism understanding.

6.3 Solution Stability

Reconstituted peptide solution stability assessment guides end-user handling recommendations and supports formulation development for liquid presentations. Solutions prepared at 1 mg/mL in common diluents (sterile water, PBS pH 7.4, 10 mM acetic acid) undergo stability evaluation at 2-8°C and 25°C over 7-14 days. Purity analysis at 0, 24, 48, 72 hours, and 7 days identifies acceptable reconstituted storage duration before significant degradation occurs.

Results demonstrate optimal solution stability in slightly acidic conditions (pH 4-5) where tryptophan oxidation rates minimize and peptide remains fully soluble. Neutral pH solutions show accelerated degradation with 2-3% purity loss within 48 hours at 25°C, while acidic solutions maintain ≥98% purity for 72 hours refrigerated. Recommendations specify reconstituted solution use within 48 hours when stored at 2-8°C, or immediate use when stored at room temperature.

6.4 Photostability Testing

Photostability evaluation following ICH Q1B guidelines exposes samples to UV and visible light according to Option 2 requirements (≥1.2 million lux·hours visible light, ≥200 watt·hours/m² UV). Dark controls stored under identical conditions except light exposure provide baseline comparison. Testing employs both solid (lyophilized powder in clear glass vials) and solution presentations (1 mg/mL in water, clear glass vials).

Results indicate light-sensitive nature of GHRP-2, with tryptophan residue undergoing rapid photooxidation under UV exposure. Purity decreases 5-8% following ICH photostability exposure protocols, with primary degradants identified as tryptophan oxidation products. Recommendations require amber glass vials or opaque secondary packaging for commercial distribution, with storage instructions specifying protection from light throughout product shelf-life.

7. Storage Recommendations and Handling Protocols

Proper storage conditions maintain GHRP-2 stability throughout its shelf-life and preserve product quality during distribution and end-user handling. Storage recommendations derive from stability study results and consider practical constraints of cold-chain distribution, laboratory refrigeration capabilities, and typical usage patterns in research environments. Comprehensive handling protocols address reconstitution procedures, aliquoting strategies, and precautions to minimize degradation risks during routine use.

7.1 Primary Storage Conditions

Lyophilized GHRP-2 requires storage at -20°C ± 5°C in sealed containers with desiccant providing protection from moisture uptake. Individual vials employ USP Type I borosilicate glass with siliconized butyl rubber stoppers and aluminum flip-off seals ensuring container closure integrity throughout shelf-life. Secondary packaging incorporates opaque materials blocking light transmission and provides cushioning protection during shipping and storage.

Temperature excursions during shipping and temporary storage may occur within acceptable ranges provided cumulative time above -10°C remains below 72 hours and temperature never exceeds 25°C. Data loggers accompanying shipments record temperature profiles throughout transit, with alert conditions triggering product quality review before release to customer. Extended excursions or exposure above 25°C require quarantine and stability testing before distribution authorization.

7.2 Reconstitution Protocols

Recommended reconstitution employs sterile water for injection, 10 mM acetic acid, or sterile saline at concentrations ranging from 0.1-5 mg/mL depending on application requirements. Reconstitution procedure involves adding diluent slowly down vial wall to minimize foaming, followed by gentle swirling (not vortexing) until complete dissolution occurs. Typical dissolution time ranges from 30 seconds to 2 minutes at room temperature for standard vial sizes (2-10 mg fill).

Stock solution preparation at higher concentrations (≥2 mg/mL) provides convenience for multiple dosing experiments while minimizing solution volume requirements. However, solubility limits at neutral pH may restrict maximum achievable concentration to approximately 5 mg/mL. Acidic diluents (10 mM acetic acid, pH 4-5) support higher solubility and improved solution stability compared to neutral pH buffers. Alkaline conditions (pH >8) should be avoided due to accelerated deamidation and oxidation reactions.

Table 7: Storage and Handling Specifications
Parameter Specification/Recommendation
Long-Term Storage (Lyophilized) -20°C ± 5°C, protect from light and moisture
Short-Term Storage (Lyophilized) 2-8°C for up to 30 days
Shelf-Life (Lyophilized, -20°C) 24-36 months from date of manufacture
Reconstituted Solution Storage 2-8°C, use within 48 hours
Recommended Diluents Sterile water, 10 mM acetic acid, sterile saline
Reconstituted Concentration Range 0.1-5 mg/mL
Solution pH Range (Optimal Stability) pH 4.0-5.5
Freeze-Thaw Cycles (Reconstituted) Not recommended; aliquot before freezing
Light Protection Required at all times (amber vials or foil wrap)
Container Type USP Type I borosilicate glass
Desiccant Requirement Included in secondary packaging

7.3 Aliquoting and Freeze-Thaw Considerations

Best practices recommend preparing single-use aliquots of reconstituted solution to eliminate repeated freeze-thaw cycles that accelerate degradation. Aliquoting involves distributing reconstituted solution into sterile microcentrifuge tubes at volumes appropriate for single experimental use, followed by immediate freezing at -20°C or -80°C. Frozen aliquots maintain acceptable stability for 1-2 weeks at -20°C or up to 1 month at -80°C when protected from light.

If freeze-thaw cycles cannot be avoided, stability data support up to 3 freeze-thaw cycles with ≤2% purity loss, provided freezing occurs rapidly and thawing proceeds on ice or at refrigerated temperature. Room temperature thawing accelerates degradation and should be avoided. Vortexing or vigorous agitation following thawing may cause aggregation and should be replaced with gentle inversion mixing.

7.4 Shipping and Transportation

Distribution employs validated cold-chain logistics maintaining temperature control throughout transit from manufacturing facility to end user. Standard shipping procedures utilize insulated containers with frozen gel packs or dry ice providing thermal mass sufficient to maintain temperatures below -10°C for 48-72 hours depending on shipping duration and ambient conditions. Summer months or tropical destinations may require dry ice shipping to ensure adequate temperature control.

Shipping validation studies demonstrate package performance under various temperature profiles simulating extreme seasonal conditions and shipping delays. Validation protocols include heat penetration studies, distribution simulation testing, and monitoring of actual shipments to verify temperature maintenance throughout transit. Seasonal shipping procedures may restrict certain destinations during extreme weather periods or mandate upgraded packaging configurations.

8. Certificate of Analysis (CoA) Documentation

Each manufacturing batch releases with a comprehensive Certificate of Analysis documenting all quality control test results and confirming conformance to established specifications. The CoA serves as the primary quality document provided to customers and provides complete analytical characterization supporting product use in research applications. CoA format follows industry standards incorporating company identification, product description, batch information, testing results, and authorized approver signatures.

8.1 CoA Content Requirements

Standard CoA documentation includes product name (GHRP-2), catalog number, batch/lot number, manufacturing date, retest/expiration date, and quantity manufactured. Storage condition statements reference validated stability data supporting recommended storage temperature and handling precautions. Chemical structure representation includes amino acid sequence in both three-letter and single-letter code notation along with molecular formula and molecular weight.

Analytical testing section presents all release testing results in tabular format showing test name, method reference, specification, and actual result for the specific batch. Results include identity confirmation (retention time, mass spectrum), purity analysis (RP-HPLC percentage), peptide content (mg per vial), water content, TFA content, and appearance description. Additional testing such as amino acid analysis ratios, specific rotation, or endotoxin levels appear when performed as part of extended characterization protocols.

Table 8: Representative Certificate of Analysis Data
Test Method Specification Result (Batch Example)
Appearance Visual White to off-white powder White powder
Identity (MS) ESI-MS 817.97 ± 0.5 Da 818.2 Da (M+H)⁺
Identity (HPLC) RP-HPLC RT = 18.5 ± 0.4 min RT = 18.3 min
Purity (HPLC) RP-HPLC, 220 nm ≥98.0% 98.7%
Water Content Karl Fischer ≤3.0% 2.1%
TFA Content Ion Chromatography ≤0.5% 0.3%
Peptide Content Amino Acid Analysis ≥95% 97.2%
Single Impurity RP-HPLC, 220 nm ≤1.0% 0.6%
Total Impurities RP-HPLC, 220 nm ≤2.0% 1.3%

8.2 Quality Statement and Intended Use

CoA footer contains quality statement certifying that the batch has been manufactured following validated procedures, tested according to approved specifications, and found to conform to all release criteria. Statement includes date of analysis, lot release date, and printed name and signature of Quality Assurance manager or authorized delegate responsible for batch disposition decision. Electronic signatures with audit trail may replace handwritten signatures in validated electronic quality management systems.

Intended use statement clarifies product grade and recommended applications, typically specifying "for research use only" or "for laboratory research purposes" with explicit disclaimer that product is not intended for human or veterinary use. Regulatory status statement may reference compliance with relevant quality standards (ISO 9001, GMP) or disclaimer regarding FDA approval status. Additional disclaimers address liability limitations and reference detailed terms and conditions provided separately.

8.3 Analytical Method References

CoA includes abbreviated method references with sufficient detail for customer interpretation while maintaining proprietary method details confidential. Standard reference format includes technique abbreviation (RP-HPLC, ESI-MS), detection method (UV 220 nm, positive ion mode), and key method parameters (gradient conditions, mass range). Full method validation documentation remains on file at manufacturing facility and may be provided to customers under confidentiality agreement for regulatory filing support.

Method references cite applicable pharmacopeial monographs when testing follows compendial procedures (USP, Ph. Eur., JP). For non-compendial methods developed in-house, references indicate internal method codes and validation completion dates. External customers requiring detailed method transfer information for independent testing receive complete method protocols including equipment specifications, reagent preparation procedures, system suitability requirements, and calculation formulas.

8.4 Digital CoA Distribution

Modern quality management systems support electronic CoA generation and digital distribution through secure customer portals. PDF format with embedded digital signatures provides tamper-evident documentation suitable for regulatory submissions and audit requirements. QR codes or unique verification codes allow customers to authenticate CoA validity through manufacturer website lookups preventing fraudulent documentation.

Digital archiving systems maintain CoA records according to regulatory retention requirements (typically 5-10 years minimum) with backup procedures ensuring document availability throughout retention period. Customer portal access allows retrieval of historical CoAs for previously purchased lots supporting research reproducibility and regulatory queries. Automated notification systems alert customers when retest dates approach requiring new batch purchases or reanalysis of remaining material.

9. Regulatory Considerations and Quality System Compliance

GHRP-2 manufacturing operates within comprehensive quality management systems aligned with ICH quality guidelines and applicable regulatory requirements for pharmaceutical ingredient production. While research-grade peptide synthesis may not require full FDA oversight, quality-conscious manufacturers implement GMP principles to ensure product consistency, traceability, and customer confidence. Quality system infrastructure encompasses document control, change control, deviation management, CAPA systems, and internal/external audit programs.

9.1 Quality Management System Structure

Quality assurance functions maintain independence from production operations with organizational separation ensuring objective product release decisions. Quality unit responsibilities include batch disposition authority, deviation investigation oversight, validation protocol approval, specification establishment, and stability program management. Standard operating procedures (SOPs) document all quality-critical operations with regular review cycles (minimum annually) ensuring procedure currency and incorporation of process improvements.

Document control systems employ version tracking, change history documentation, and controlled distribution procedures preventing use of obsolete procedures. Electronic document management systems with electronic signature capability streamline document workflows while maintaining 21 CFR Part 11 compliance where applicable. Training programs ensure personnel competency with documented verification of training effectiveness through written examinations or practical demonstrations before independent task performance authorization.

9.2 Validation Requirements

Critical process validations demonstrate consistency and reproducibility of manufacturing operations through prospective validation protocols executed across multiple production batches. Synthesis process validation evaluates coupling efficiency, deprotection completeness, cleavage yield, and crude purity consistency across three consecutive conforming batches at commercial scale. Purification process validation demonstrates chromatographic method robustness, reproducible separation resolution, and consistent product recovery rates.

Analytical method validation follows ICH Q2(R1) guidelines establishing specificity, linearity, accuracy, precision, detection limit, quantitation limit, range, and robustness for all release testing methods. Validation protocols define acceptance criteria for each validation parameter based on method intended use and required performance characteristics. Revalidation triggers include method modification, equipment changes, or trending results suggesting method performance degradation.

Cleaning validation establishes effective procedures for equipment cleaning between batches preventing cross-contamination and carryover. Validation protocols define acceptance criteria for visual cleanliness, residual peptide detection limits, and cleaning agent residues. Surface swab sampling combined with rinse water analysis provides comprehensive contamination assessment. Worst-case scenario selection considers hardest-to-clean equipment surfaces, maximum hold times before cleaning, and lowest solubility products as validation challenges.

9.3 Supplier Qualification and Management

Raw material suppliers undergo formal qualification processes including initial supplier assessment questionnaires, on-site audits (for critical materials), quality agreement establishment, and ongoing performance monitoring. Critical material suppliers provide batch-specific certificates of analysis, maintain change notification systems alerting to manufacturing process modifications, and support supply continuity through dual sourcing or strategic inventory maintenance.

Vendor audit programs evaluate supplier quality systems, manufacturing capabilities, testing laboratories, and regulatory compliance status. Audit frequency depends on material criticality and supplier performance history, ranging from annual audits for critical suppliers to biennial or triennial audits for commodity suppliers with established performance records. Remote audits using video conference technology may supplement or replace on-site visits while maintaining audit effectiveness.

9.4 Reference Standard Management

Primary reference standards undergo complete characterization through orthogonal analytical techniques establishing assigned values for critical quality attributes. Characterization includes high-resolution mass spectrometry, quantitative NMR, amino acid analysis, multi-dimensional chromatography, and absolute peptide content determination. Primary standard material assignment requires review and approval by multiple qualified analysts with documentation retained throughout standard lifetime.

Working reference standards prepared from primary standards or well-characterized production batches support routine quality control testing. Qualification protocols compare working standards against primary standards demonstrating equivalent performance in all analytical methods. Requalification intervals (typically annually) verify continued standard suitability through stability trending and comparative testing. Storage conditions for reference materials mirror or exceed product storage requirements ensuring standard stability throughout use period.

10. Advanced Analytical Characterization and Impurity Profiling

Comprehensive peptide characterization extends beyond routine release testing to include advanced analytical techniques providing detailed structural confirmation and impurity identification. These methods support process understanding, facilitate troubleshooting investigations, and enable regulatory submissions requiring extensive product characterization. Implementation of orthogonal analytical approaches increases confidence in product identity and provides multiple independent verification mechanisms.

10.1 High-Resolution Mass Spectrometry

High-resolution accurate mass (HRAM) analysis employing Orbitrap or time-of-flight (TOF) mass spectrometers provides molecular weight determination with ±5 ppm mass accuracy or better. This precision enables differentiation of isobaric species and confirmation of elemental composition through isotope pattern analysis. HRAM data supports structural confirmation through comparison of measured versus theoretical mass values, with agreement within instrument specifications confirming molecular formula.

Tandem mass spectrometry (MS/MS) via collision-induced dissociation (CID) generates sequence-specific fragment ions enabling de novo sequence confirmation. Fragmentation patterns produce b-ions (N-terminal fragments) and y-ions (C-terminal fragments) whose mass values correlate with expected fragments from the known sequence. Complete y-ion and b-ion series coverage provides high-confidence sequence verification and confirms D-amino acid positioning through retention of stereochemistry-specific protecting groups during synthesis.

10.2 Nuclear Magnetic Resonance Spectroscopy

Proton NMR (¹H NMR) spectroscopy provides orthogonal identity confirmation and structural characterization complementing mass spectrometry data. Characteristic chemical shifts for aromatic residues (tryptophan indole protons at 7.2-7.6 ppm, naphthylalanine aromatic signals at 7.4-7.9 ppm, phenylalanine aromatic multiplets at 7.2-7.4 ppm) confirm presence and relative quantities of aromatic amino acids. Integration ratios between aromatic and aliphatic regions verify expected amino acid composition.

Two-dimensional NMR techniques including COSY (correlation spectroscopy), TOCSY (total correlation spectroscopy), and NOESY (nuclear Overhauser effect spectroscopy) enable complete resonance assignment and secondary structure determination in solution. These methods support conformational analysis and can detect structural variants arising from sequence errors or unexpected post-translational modifications. Quantitative NMR (qNMR) using internal standards provides absolute peptide content determination independent of amino acid analysis, serving as reference method for primary standard value assignment.

10.3 Peptide Mapping and Sequence Verification

Enzymatic digestion using sequence-specific proteases (trypsin, chymotrypsin, pepsin) generates characteristic fragment patterns analyzable by LC-MS. Trypsin digestion cleaves C-terminal to lysine and arginine residues (though GHRP-2 contains only one lysine at position 6), producing predictable fragment masses. Chymotrypsin preferentially cleaves C-terminal to aromatic residues (Trp, Phe), generating multiple fragments from GHRP-2's aromatic-rich sequence. Mass analysis of digestion products confirms expected fragment masses and verifies sequence continuity.

Edman degradation sequencing provides classical N-terminal sequence verification through stepwise removal and identification of amino acids from the N-terminus. While largely superseded by MS-based methods, Edman sequencing retains value for confirming N-terminal integrity and detecting N-terminal modifications or truncations. MALDI-TOF-MS analysis of peptide ladder sequences generated through partial Edman degradation combines classical sequencing with modern mass analysis for rapid sequence confirmation.

10.4 Impurity Identification and Classification

Systematic impurity profiling characterizes synthesis-related impurities, degradation products, and process-related contaminants. Major impurity categories include deletion sequences (lacking one or more amino acids from incomplete coupling), addition sequences (containing extra residues from insufficient washing or premature coupling), and amino acid substitution variants (from activated amino acid cross-contamination). LC-MS analysis combined with retention time prediction software facilitates impurity structure assignment based on molecular weight and chromatographic behavior.

Table 9: Common GHRP-2 Impurity Profile
Impurity Type Structure/Description Typical Level Detection Method
Des-Lys⁶ GHRP-2 Deletion sequence missing C-terminal lysine 0.3-0.8% LC-MS (MW 689.8 Da)
Des-Ala¹ GHRP-2 Deletion sequence missing N-terminal D-alanine 0.2-0.5% LC-MS (MW 747.0 Da)
Trp-Oxidation Products Tryptophan oxidized to N-formylkynurenine (+16 Da) 0.1-0.4% LC-MS, fluorescence detection
Phe Substitution L-Phe in place of D-Phe (stereoisomer) 0.1-0.3% Chiral HPLC
Incomplete Deprotection Residual Boc group on lysine (+100 Da) 0.1-0.2% LC-MS (MW 918.0 Da)
Dimers Disulfide-linked or non-covalent aggregates 0.1-0.3% SEC, native MS
TFA Adducts TFA ester formation on hydroxyl groups 0.05-0.15% LC-MS (+96 Da)

Degradation product identification employs stress testing samples exposing peptide to oxidative conditions (hydrogen peroxide), acidic/basic pH extremes, elevated temperature, and light exposure. Comparative analysis of stressed versus unstressed samples using LC-MS identifies degradation products based on mass shifts corresponding to known degradation mechanisms. Tryptophan oxidation represents the primary degradation pathway, producing characteristic +16 Da (N-formylkynurenine) and +32 Da (kynurenine) mass increases. Minor degradation pathways include peptide bond hydrolysis (particularly at Ala-Trp and Trp-D-Phe positions) and lysine side chain modifications.

11. Conclusion and Quality Assurance Summary

GHRP-2 manufacturing represents a mature synthetic peptide production process with well-established synthesis protocols, validated purification methods, and comprehensive quality control strategies. The documented procedures provide reproducible production of pharmaceutical-grade material meeting stringent purity specifications (≥98% by HPLC) suitable for demanding research applications. Process understanding developed through extensive characterization studies enables consistent batch-to-batch product quality and supports troubleshooting when process deviations occur.

Critical quality attributes identified through risk assessment and process characterization include peptide purity, peptide content, sequence verification, stereochemical integrity, and freedom from specific degradation products (particularly tryptophan oxidation). Control strategies address these attributes through validated analytical methods, in-process controls at critical synthesis steps, and comprehensive release testing prior to batch distribution. Stability programs demonstrate product shelf-life of 24-36 months when stored at -20°C with appropriate moisture and light protection.

Continuous improvement initiatives leverage advanced analytical techniques and process analytical technology (PAT) to enhance process understanding and optimize manufacturing efficiency. Implementation of Quality by Design (QbD) principles facilitates systematic process development with clear linkage between process parameters and product quality attributes. These approaches support regulatory submissions when GHRP-2 or related molecules advance toward clinical development stages.

Manufacturing excellence requires sustained commitment to quality culture, personnel training, equipment maintenance, and adherence to validated procedures. Regular management review of quality metrics, trend analysis of stability data, and proactive identification of improvement opportunities ensure continued manufacturing capability and customer satisfaction. This comprehensive manufacturing profile serves as reference documentation for manufacturers, quality control laboratories, and research professionals requiring detailed technical information about GHRP-2 production and quality specifications.

12. References and Additional Resources

  1. International Conference on Harmonisation (ICH). Q7 Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients. Available at: https://database.ich.org/sites/default/files/Q7%20Guideline.pdf
  2. U.S. Food and Drug Administration. Current Good Manufacturing Practice (CGMP) Regulations. Available at: https://www.fda.gov/drugs/pharmaceutical-quality-resources/current-good-manufacturing-practice-cgmp-regulations
  3. Merrifield RB. Solid Phase Peptide Synthesis. I. The Synthesis of a Tetrapeptide. J Am Chem Soc. 1963;85(14):2149-2154. Available at: https://pubs.acs.org/doi/10.1021/ja00897a025
  4. Sigma-Aldrich. Fmoc Solid Phase Peptide Synthesis - A Practical Approach. Technical Article. Available at: https://www.sigmaaldrich.com/US/en/technical-documents/technical-article/chemistry-and-synthesis/peptide-synthesis/fmoc-solid-phase-peptide-synthesis
  5. Waters Corporation. HPLC Fundamentals - Principles and Practice. Available at: https://www.waters.com/waters/en_US/HPLC-Fundamentals/nav.htm?cid=10049055
  6. International Conference on Harmonisation (ICH). Q6A Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Products: Chemical Substances. Available at: https://database.ich.org/sites/default/files/Q6A_Guideline.pdf
  7. International Conference on Harmonisation (ICH). Q1A(R2) Stability Testing of New Drug Substances and Products. Available at: https://database.ich.org/sites/default/files/Q1A%28R2%29%20Guideline.pdf
  8. U.S. Food and Drug Administration. Lyophilization of Parenteral Drug Products - Guidance for Industry. Available at: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/lyophilization-parenteral-drug-products
  9. International Conference on Harmonisation (ICH). Quality Guidelines Overview. Available at: https://www.ich.org/page/quality-guidelines
  10. International Conference on Harmonisation (ICH). Q8(R2) Pharmaceutical Development. Available at: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/q8r2-pharmaceutical-development