Tesamorelin Manufacturing Profile: Technical Specifications and Quality Standards
1. Introduction to Tesamorelin Manufacturing
Tesamorelin represents a synthetic analog of human growth hormone-releasing hormone (GHRH) consisting of all 44 amino acids of the native sequence with the addition of a trans-3-hexenoyl group at the N-terminus. This modification confers enhanced stability and resistance to enzymatic degradation compared to endogenous GHRH, extending the peptide's biological half-life while maintaining potent growth hormone secretagogue activity. Manufacturing this complex therapeutic peptide requires specialized solid-phase synthesis capabilities, advanced purification systems, and rigorous analytical testing protocols to achieve pharmaceutical-grade specifications.
The production of tesamorelin demands strict adherence to current Good Manufacturing Practices (cGMP) as outlined in FDA 21 CFR Parts 210 and 211, with particular attention to peptide-specific quality attributes including sequence fidelity, N-terminal modification efficiency, epimerization control, and aggregation prevention. The molecular weight of 5135.9 Da (free base) and the presence of multiple sensitive amino acid residues necessitate carefully optimized synthesis conditions, protective group selection, and cleavage chemistry to minimize impurity formation while maximizing overall yield.
This comprehensive manufacturing profile provides detailed technical specifications covering all aspects of tesamorelin production from raw material qualification through final product release. Manufacturing personnel, quality control professionals, and regulatory compliance specialists will find systematic guidance on synthesis parameters, purification methodologies, analytical methods, batch documentation requirements, stability protocols, storage specifications, and certificate of analysis standards. The information presented aligns with ICH Q7 guidelines for active pharmaceutical ingredient manufacturing and represents industry best practices for therapeutic peptide production.
2. Solid-Phase Peptide Synthesis Process
Tesamorelin synthesis employs Fmoc (9-fluorenylmethyloxycarbonyl) solid-phase peptide synthesis (SPPS) methodology, which provides optimal efficiency for assembling the 44-residue sequence. The synthesis presents significant technical challenges due to peptide length, the presence of aggregation-prone sequences, and the requirement for site-specific N-terminal hexenoylation. Manufacturing facilities must utilize automated or semi-automated peptide synthesizers with precise reagent delivery, temperature control, and monitoring capabilities to ensure reproducible synthesis outcomes.
2.1 Resin Selection and Amino Acid Sequence
Tesamorelin synthesis begins with selection of appropriate solid support resin matched to the C-terminal amide requirement. Rink Amide MBHA resin (100-200 mesh, 0.3-0.5 mmol/g substitution) provides optimal performance for this 44-residue sequence. Lower substitution levels (0.3-0.4 mmol/g) are preferred for longer peptides to reduce steric crowding and aggregation during on-resin assembly.
The complete tesamorelin amino acid sequence proceeds from C-terminus to N-terminus as follows:
Sequence (C→N): Arg-Leu-Arg-Ala-Arg-Ala-Gly-Arg-Glu-Gln-Asn-Ser-Glu-Gly-Gln-Gln-Arg-Ser-Met-Ile-Asp-Gln-Leu-Leu-Arg-Lys-Ala-Arg-Ser-Leu-Gln-Gly-Leu-Val-Lys-Arg-Tyr-Ser-Gln-Thr-Phe-Ile-Ala-Asp-Ala-Tyr-[trans-3-hexenoyl]
Critical resin specifications include:
- Particle size distribution: 100-200 mesh (74-149 μm) for optimal flow properties
- Swelling volume: minimum 5 mL/g in N,N-dimethylformamide (DMF)
- Moisture content: maximum 3% by Karl Fischer titration
- Functional group loading: verified by quantitative Fmoc determination (±10% of specification)
- Heavy metal content: less than 10 ppm total by ICP-MS analysis
2.2 Amino Acid Coupling Strategy
The 44-step synthesis requires optimization of coupling chemistry to maintain high efficiency throughout the sequence assembly. Standard coupling employs HBTU (O-benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluorophosphate) and HOBt (1-hydroxybenzotriazole) activation in the presence of DIEA (N,N-diisopropylethylamine) as base. For particularly challenging couplings involving sterically hindered residues or aggregation-prone sequences, alternative activating agents such as HATU or DIC/Oxyma provide improved results.
Standard coupling parameters include:
| Parameter | Specification | Monitoring Method |
|---|---|---|
| Amino Acid Equivalents | 3-5 equivalents (5 for difficult sequences) | Gravimetric verification |
| Activator Equivalents | 3-5 equivalents HBTU/HOBt | Solution preparation verification |
| Base Equivalents | 6-10 equivalents DIEA | Volumetric measurement |
| Coupling Time | 60-120 minutes (double coupling for Arg, His) | Automated timer validation |
| Reaction Temperature | 25°C ± 3°C (elevated to 40°C for difficult couplings) | Continuous temperature recording |
| Coupling Efficiency | ≥99.5% by Kaiser or chloranil test | Colorimetric assay after each coupling |
2.3 Aggregation Prevention Strategies
Tesamorelin contains multiple sequences prone to β-sheet formation and on-resin aggregation, particularly in the central and C-terminal regions. Aggregation during synthesis leads to incomplete couplings, deletion sequences, and reduced crude purity. Manufacturing protocols incorporate several aggregation-prevention strategies:
- Chaotropic additives: 5-10% N-methylpyrrolidone (NMP) in DMF during coupling and deprotection steps
- Temperature elevation: Coupling at 40-50°C for aggregation-prone sequences (positions 15-30)
- Pseudoproline dipeptides: Incorporation at Ser-Gln and Thr-Phe positions to disrupt β-sheet formation
- Extended deprotection: Triple deprotection cycles (5 min + 10 min + 10 min) with 20% piperidine/DMF
- Alternative solvents: Addition of 10% dichloromethane (DCM) to DMF to enhance resin swelling
2.4 N-Terminal Hexenoylation
Following assembly of the complete 44-amino acid sequence, the final synthetic step involves attachment of the trans-3-hexenoic acid moiety to the N-terminal tyrosine α-amino group. This modification distinguishes tesamorelin from native GHRH and contributes to enhanced metabolic stability. The hexenoylation reaction requires careful optimization to achieve quantitative conversion while avoiding side reactions.
Hexenoylation procedure specifications:
- Reagent: Trans-3-hexenoic acid (5 equivalents, minimum 98% purity, verified by GC)
- Activation: HBTU/HOBt (5 equivalents each) or DIC/Oxyma system
- Base: DIEA (10 equivalents) to ensure complete deprotonation
- Reaction time: 4-8 hours with monitoring by test cleavage and MS analysis
- Completion criteria: ≥98% hexenoylation by analytical HPLC-MS
- Verification: Mass shift of +96 Da corresponding to hexenoyl addition
2.5 Fmoc Deprotection Monitoring
Each coupling cycle requires preceding Fmoc deprotection of the N-terminal amino group using piperidine in DMF. Deprotection employs a dual-treatment protocol: initial 5-minute treatment with 20% piperidine/DMF followed by 15-minute treatment with fresh reagent. Deprotection monitoring occurs via UV spectroscopy at 301 nm, detecting the dibenzofulvene-piperidine adduct released during Fmoc removal.
Deprotection completeness criteria include:
- UV absorbance return to baseline at 301 nm (indicating complete Fmoc removal)
- Integrated UV peak area within 90-110% of theoretical (based on resin loading)
- Negative ninhydrin test (Kaiser test) following incomplete deprotection indicates resin damage
- DMF wash cycles (minimum 5 × 2 minutes) to remove piperidine and adduct before coupling
For the extended 44-residue synthesis, automated synthesizers with real-time UV monitoring and electronic record-keeping ensure complete documentation of deprotection efficiency throughout the assembly process, supporting FDA cGMP documentation requirements.
3. Cleavage and Crude Peptide Recovery
Following completion of the solid-phase synthesis including N-terminal hexenoylation, the protected tesamorelin sequence undergoes simultaneous cleavage from the solid support and removal of amino acid side-chain protecting groups. This critical step employs strongly acidic conditions with scavenger additives to prevent side reactions, particularly oxidation of methionine and tryptophan residues and alkylation of sensitive functional groups by carbocations generated during protecting group removal.
3.1 Cleavage Cocktail Formulation
Tesamorelin cleavage employs Reagent K or modified Reagent K formulations optimized for peptides containing methionine, tryptophan, arginine, glutamine, and asparagine residues. The standard cleavage cocktail composition consists of:
- Trifluoroacetic acid (TFA): 82.5% v/v (peptide synthesis grade, minimum 99.5% purity)
- Phenol: 5% w/v (crystalline, freshly liquefied or with BHT stabilizer)
- Water: 5% v/v (Type I ultrapure, 18.2 MΩ·cm resistivity)
- Thioanisole: 5% v/v (anhydrous, minimum 99% purity)
- 1,2-Ethanedithiol (EDT): 2.5% v/v (minimum 98% purity)
For enhanced methionine protection in tesamorelin (Met-27), some protocols incorporate additional scavengers:
- Ammonium iodide: 0.5-1.0% w/v to reduce Met sulfoxide formation
- 2,2'-(Ethylenedioxy)diethanethiol (DODT): Substitute for EDT at 2.5% for superior Trp protection
3.2 Cleavage Procedure and Parameters
The cleavage reaction proceeds at controlled temperature with specific timing to balance complete deprotection against acid-catalyzed side reactions. Process parameters include:
| Parameter | Specification | Quality Impact |
|---|---|---|
| Cleavage Cocktail Volume | 15-20 mL per gram of resin | Adequate scavenger capacity and mixing |
| Reaction Temperature | 20-25°C (room temperature) | Minimize oxidation and sequence deletion |
| Reaction Duration | 2.5-4.0 hours with periodic mixing | Complete deprotection without excessive degradation |
| Mixing Frequency | Every 30 minutes by manual agitation | Homogeneous exposure to cleavage reagents |
| Vessel Type | Glass reaction vessel with cap (pressure release) | Containment of CO₂ from tBu/Pbf deprotection |
3.3 Peptide Precipitation and Washing
Following cleavage completion, the resin undergoes filtration through a fritted glass funnel or filter paper, and the TFA filtrate containing dissolved peptide flows directly into a 10-fold excess volume of cold diethyl ether (-20 to 0°C) maintained in an ice bath. The crude tesamorelin precipitates immediately as a white to off-white solid. Collection occurs through centrifugation at 3000-5000 × g for 10 minutes at 4°C.
The crude pellet undergoes multiple ether wash cycles to remove:
- Residual TFA and scavengers (phenol, thioanisole, EDT)
- Cleaved protecting groups (Pbf, tBu, Trt derivatives)
- Truncated or deletion peptide sequences with altered solubility
Washing protocol specifications:
- Minimum 3 wash cycles with fresh cold diethyl ether (10 volumes each)
- Each wash includes pellet resuspension by vortexing and re-centrifugation
- Final wash supernatant verified as colorless (no yellow phenol coloration)
- Pellet drying under vacuum at room temperature (4-12 hours) or nitrogen stream (2-4 hours)
- Alternative: Direct lyophilization from 10% acetic acid solution after ether extraction
3.4 Crude Peptide Characterization
Following precipitation, washing, and drying, the crude tesamorelin undergoes analytical characterization prior to purification. Quality attributes assessed include:
- Crude purity: 30-55% by analytical RP-HPLC at 214 nm (acceptable range varies with synthesis optimization)
- Mass confirmation: ESI-MS or MALDI-TOF showing expected molecular ion at 5135.9 Da (±1 Da)
- Hexenoylation verification: MS confirmation of N-terminal modification (presence of 5135.9 Da peak, absence of significant des-hexenoyl at 5039.9 Da)
- Major impurities: Deletion sequences, truncations, incomplete deprotection products identified by LC-MS
- Crude yield: 25-50% based on initial resin loading (calculated as mg crude peptide per mmol resin loading)
Crude peptide storage prior to purification occurs at -20°C in sealed containers with desiccant. Stability data supports storage up to 6 months under these conditions, though immediate processing to purification optimizes final product quality by minimizing oxidation and aggregation during storage.
4. Preparative HPLC Purification
Tesamorelin purification employs preparative reversed-phase high-performance liquid chromatography (RP-HPLC) as the primary method for achieving pharmaceutical-grade purity. The 5.1 kDa molecular weight and moderate hydrophobicity of tesamorelin provide favorable chromatographic behavior on C18 or C8 stationary phases. The purification strategy must efficiently separate the target peptide from closely related impurities including deletion sequences, oxidation products, deamidated variants, and residual des-hexenoyl peptide while maintaining product integrity and maximizing recovery yield.
4.1 Preparative Column Selection
Column selection for tesamorelin purification balances resolution requirements against loading capacity and throughput considerations. Recommended specifications include:
- Column chemistry: C18 (preferred) or C8 reversed-phase silica
- Dimensions (small scale): 250 × 50 mm or 250 × 100 mm
- Dimensions (production scale): 250 × 150 mm or 300 × 250 mm
- Particle size: 10-15 μm for preparative applications (5 μm for analytical)
- Pore size: 300 Å to accommodate 5.1 kDa molecular weight
- Carbon load: 12-18% for C18 chemistry
- Endcapping: Complete endcapping to minimize silanol interactions
- pH stability: 2.0-8.0 operational range
4.2 Mobile Phase System Design
Binary gradient elution with acidified aqueous and organic phases provides optimal separation of tesamorelin from impurities. The mobile phase system utilizes:
Mobile Phase A (Aqueous Phase):
- Water: HPLC-grade, filtered through 0.22 μm membrane, conductivity <5 μS/cm
- Trifluoroacetic acid (TFA): 0.05-0.1% v/v for ion-pairing
- pH: Approximately 2.0-2.2 (not adjusted, inherent from TFA)
Mobile Phase B (Organic Phase):
- Acetonitrile: HPLC-gradient grade, minimum 99.9% purity
- Trifluoroacetic acid (TFA): 0.05-0.1% v/v (matched to Phase A)
- Water content: <0.05% verified by Karl Fischer titration
Mobile phase preparation requires daily fresh preparation, degassing via helium sparging or vacuum degassing, and storage at controlled room temperature (15-25°C). TFA concentration optimization balances peak shape (higher TFA improves tailing) against potential product modification and difficulty in subsequent TFA removal.
4.3 Gradient Method Development
Method development for tesamorelin purification employs analytical HPLC screening to identify optimal gradient conditions providing baseline resolution between the target peptide and critical impurities. A typical optimized preparative gradient includes:
| Time (min) | %B | Flow Rate (mL/min) | Phase Description |
|---|---|---|---|
| 0-5 | 30 | 50-100 | Initial equilibration, sample loading |
| 5-10 | 30-35 | 50-100 | Wash highly polar impurities, truncations |
| 10-50 | 35-45 | 50-100 | Shallow gradient for critical separation region |
| 50-60 | 45-95 | 50-100 | Steep gradient to elute hydrophobic impurities |
| 60-65 | 95 | 50-100 | Column wash, removal of strongly retained material |
| 65-75 | 95-30 | 50-100 | Re-equilibration for next injection |
Detection wavelengths include 214 nm (primary, peptide bond absorption) and 280 nm (secondary, aromatic residue monitoring for Tyr, Phe, Trp). Column temperature control at 25-30°C ensures reproducible retention times across multiple purification runs.
4.4 Sample Loading and Fraction Collection
Crude tesamorelin dissolution for loading onto preparative columns requires optimization to maximize solubility while preventing pre-column aggregation. Sample preparation protocols include:
- Dissolve crude peptide in minimal volume of 20-30% acetonitrile with 0.1% TFA
- Target loading concentration: 20-50 mg/mL (verify solubility for each crude batch)
- Centrifuge dissolved sample at 10,000 × g for 10 minutes to remove particulates
- Filter through 0.45 μm PVDF syringe filter immediately before injection
- Column loading: 5-20 mg crude peptide per mL of column volume
Automated fraction collection targets the tesamorelin peak with defined threshold settings:
- Collection initiation: 10-12% of peak maximum on ascending slope
- Collection termination: 10-12% of peak maximum on descending slope
- Fraction volume: 50-200 mL depending on peak width and scale
- Peak tracking: Automated retention time adjustment (±0.5 min) across runs
4.5 Fraction Analysis and Pooling
Each collected fraction undergoes analytical RP-HPLC testing to determine purity profile before pooling decisions. Analytical method employs matched gradient conditions on analytical-scale column (4.6 × 250 mm, 5 μm C18) with detection at 214 nm.
Pooling criteria specifications:
- Primary pool: Fractions with ≥95% purity by area at 214 nm qualify for primary pool
- Secondary pool: Fractions with 85-95% purity collected separately for re-purification
- Rejected fractions: Fractions with <85% purity or containing unidentified impurities >2%
- Mass spectrometry verification: Representative fractions analyzed by ESI-MS or MALDI-TOF to confirm molecular weight
4.6 Desalting and TFA Removal
Purified fractions contain TFA as the ion-pairing agent and acetonitrile as organic modifier, both requiring removal prior to lyophilization. Two primary desalting approaches are employed:
Method 1: Rotary Evaporation
- Remove bulk acetonitrile under vacuum at 30-35°C
- Dilute with water (3-4 volumes) and repeat concentration
- Perform minimum 3 water dilution/concentration cycles
- Adjust pH to 4.5-5.5 with dilute ammonium hydroxide if needed
- Final concentration to appropriate volume for lyophilization
Method 2: Size-Exclusion Chromatography Desalting
- Column: Sephadex G-25 or G-10, dimensions scaled to peptide quantity
- Equilibration: 0.1% acetic acid in water (3-5 column volumes)
- Sample loading: Concentrated peptide solution (≤5% column volume)
- Elution monitoring: UV detection at 214 nm, collect peptide peak
- TFA removal efficiency: >99% as verified by ion chromatography
The desalted peptide solution undergoes sterile filtration through 0.22 μm PES membrane prior to lyophilization, with bioburden testing confirming <10 CFU/100 mL before processing per USP sterility requirements for parenteral products.
5. Lyophilization and Final Product Formulation
Lyophilization (freeze-drying) converts purified tesamorelin solution into a stable solid dosage form with extended shelf-life and convenient reconstitution properties. The lyophilization cycle must remove water and residual volatile solvents while preserving peptide structure, preventing aggregation, and producing an elegant cake appearance with rapid reconstitution characteristics. Formulation development incorporates appropriate excipients to provide structural support during freezing and drying, enhance long-term stability, and facilitate accurate dosing after reconstitution.
5.1 Pre-Lyophilization Formulation Development
Tesamorelin formulation for lyophilization incorporates excipients serving multiple functions including bulking, cryoprotection, pH buffering, and cake structure support. Based on the FDA-approved formulation for commercial tesamorelin products, the standard composition includes:
- Tesamorelin (active): 2 mg per vial (target dose)
- Mannitol: 20 mg (10:1 ratio to peptide, crystalline bulking agent)
- Sucrose: 10 mg (amorphous component, cryoprotectant)
- Histidine: 0.78 mg (pH buffer and antioxidant)
- Water for injection: q.s. to target fill volume (typically 2-3 mL)
Formulation preparation specifications:
- Excipient grade: All excipients must meet USP/NF or Ph. Eur. specifications
- Water quality: Water for Injection (WFI), endotoxin level <0.25 EU/mL
- Solution preparation: Dissolve excipients in WFI, adjust pH to 6.0-7.5 with dilute HCl or NaOH
- Peptide addition: Add purified tesamorelin and mix gently to dissolve
- pH verification: Final pH 6.0-7.5 (critical for stability and reconstitution)
- Sterile filtration: 0.22 μm PES membrane filtration into sterile fill vessel
- Bioburden: <10 CFU/100 mL prior to filling (for aseptic processing)
5.2 Vial Filling and Stopper Placement
Aseptic vial filling occurs in ISO Class 5 (Grade A) environment with appropriate background environment (Grade B for aseptic processing). Process parameters include:
| Parameter | Specification | Quality Control |
|---|---|---|
| Vial Type | Type I borosilicate glass, 5-10 mL capacity | Certificate of conformance from supplier |
| Vial Preparation | Depyrogenated at 250°C for 30 minutes | LAL testing of rinse water <0.25 EU/mL |
| Stopper Type | Bromobutyl or chlorobutyl rubber, coated | Extractables/leachables qualification |
| Fill Volume | 2.0-3.0 mL (overfill to account for lyophilization loss) | Gravimetric verification every 10-20 vials |
| Fill Volume Accuracy | ±5% of target volume | In-process weight checks with statistical control |
| Stopper Placement | Partially seated (2-3 mm above full seal position) | Visual verification, allows vapor escape |
| Particulate Matter | Zero visible particles per USP <790> | 100% visual inspection under controlled lighting |
5.3 Lyophilization Cycle Development
Lyophilization cycle development utilizes differential scanning calorimetry (DSC) and freeze-dry microscopy to determine the glass transition temperature (Tg') and collapse temperature (Tc) of the formulation. For the mannitol/sucrose/histidine formulation, Tg' typically occurs at -40 to -35°C, establishing the maximum product temperature during primary drying at -38 to -33°C (typically 2-5°C below Tg').
A validated three-phase lyophilization cycle includes:
Phase 1: Freezing
- Initial shelf temperature: +5°C (ambient loading temperature)
- Ramp rate: -1.0°C per minute to -5°C (controlled ice nucleation region)
- Hold at -5°C: 60 minutes (allows uniform ice nucleation across all vials)
- Ramp rate: -0.5°C per minute to -45°C
- Hold at -45°C: Minimum 3 hours (ensures complete crystallization)
Phase 2: Primary Drying
- Vacuum initiation: Reduce chamber pressure to 80-120 mTorr (10.7-16.0 Pa)
- Shelf temperature ramp: +0.1 to +0.2°C per minute to -35°C
- Hold at -35°C: 30-48 hours (duration depends on fill volume and vial configuration)
- Product temperature monitoring: Thermocouples in center and edge vials
- Endpoint determination: Pirani gauge pressure converges with capacitance manometer
- Alternative endpoint: Product temperature equals shelf temperature (±2°C)
Phase 3: Secondary Drying
- Shelf temperature ramp: +0.15°C per minute to +25°C
- Chamber pressure: Maintain 80-120 mTorr
- Hold at +25°C: 6-10 hours (removes residual bound water)
- Target residual moisture: <2% by Karl Fischer titration
5.4 Stoppering, Crimping, and Seal Integrity
Following completion of secondary drying, automated stoppering occurs within the lyophilizer chamber under controlled atmosphere. Chamber backfilling with nitrogen or argon (to 950 ± 50 mbar) precedes stopper insertion to minimize oxygen exposure and moisture re-absorption. Hydraulic ram advances stoppers to fully sealed position, and vials undergo immediate unloading for aluminum crimp seal application.
Critical quality attributes of lyophilized tesamorelin include:
- Appearance: Uniform white to off-white cake or powder, no collapse or melt-back
- Cake integrity: Intact cake structure, no cracking or shrinkage from vial walls
- Residual moisture: ≤2.0% by Karl Fischer titration (specification <3%)
- Reconstitution time: Complete dissolution within 30-60 seconds with gentle swirling
- Reconstitution appearance: Clear, colorless solution free of visible particles
- Headspace composition: <3% oxygen for nitrogen-backfilled vials (measured by GC)
- Container closure integrity: Passes vacuum decay test and dye ingress testing per USP <1207>
Seal integrity testing employs non-destructive methods (100% testing via vacuum decay) and destructive methods (statistical sampling for dye ingress), ensuring product protection throughout shelf-life per FDA guidance for lyophilized parenteral products.
6. Quality Control Testing and Release Specifications
Comprehensive analytical testing confirms each tesamorelin batch meets pharmaceutical-grade specifications prior to commercial release. The quality control program employs orthogonal analytical methods to assess identity, purity, potency, safety, and stability-indicating attributes. All testing occurs in accordance with validated analytical procedures following ICH Q2(R1) guidelines and USP general chapters for analytical method validation.
6.1 Identity Testing Methods
Multiple complementary techniques confirm tesamorelin identity and distinguish it from related peptides or impurities:
Reversed-Phase HPLC Identity
- Column: C18, 4.6 × 250 mm, 5 μm particle size, 300 Å pore
- Mobile phase: Water/acetonitrile gradient with 0.1% TFA
- Detection: UV at 214 nm with spectral acquisition (200-400 nm)
- Acceptance criteria: Retention time matches reference standard ± 2.0%
- Spectral match: UV spectrum overlay with reference (normalized) shows correlation >0.98
Mass Spectrometry Identity
- Method: Electrospray ionization mass spectrometry (ESI-MS) or MALDI-TOF MS
- Ionization mode: Positive ion mode, multiple charge states observed
- Acceptance criteria: Observed molecular weight 5135.9 ± 1.0 Da (average mass)
- Monoisotopic mass: 5130.5 Da (theoretical, for high-resolution instruments)
- N-terminal modification verification: Absence of des-hexenoyl species at 5039.9 Da (<2%)
Peptide Mapping by LC-MS/MS
- Enzymatic digestion: Trypsin or Glu-C protease (37°C, 4-18 hours)
- Analysis: RP-HPLC coupled to MS/MS with data-dependent acquisition
- Acceptance criteria: All expected peptide fragments identified with correct masses
- Sequence coverage: Minimum 95% of tesamorelin sequence confirmed
- N-terminal hexenoyl fragment: Confirmed in N-terminal peptide fragment
Amino Acid Analysis (AAA)
- Hydrolysis: 6 N HCl, 110°C, 20-24 hours under nitrogen atmosphere
- Derivatization: Pre-column or post-column derivatization (ninhydrin or OPA)
- Detection: UV or fluorescence detection following HPLC or ion-exchange separation
- Acceptance criteria: Each amino acid within ±15% of theoretical molar ratio
- Note: Tryptophan destroyed during acid hydrolysis (not quantified by this method)
6.2 Purity Assessment by Orthogonal Methods
Comprehensive purity assessment employs multiple chromatographic modes to detect different impurity classes:
Reversed-Phase HPLC Purity (Primary Method)
- Column: C18, 4.6 × 250 mm, 5 μm, 300 Å pore size
- Gradient: 25-50% acetonitrile over 40 minutes with 0.1% TFA
- Flow rate: 1.0 mL/min, column temperature 30°C
- Detection: 214 nm (peptide bond), 280 nm (aromatic residues)
- Injection volume: 20 μL of 1 mg/mL solution
- Acceptance criteria at 214 nm:
- Main peak (tesamorelin): ≥95.0% by area
- Any individual impurity: ≤2.0%
- Total impurities: ≤5.0%
Size-Exclusion Chromatography (SEC-HPLC)
- Column: Silica-based SEC, TSKgel G2000SW or equivalent, 7.8 × 300 mm
- Mobile phase: 0.1 M sodium phosphate pH 7.0 with 0.1 M sodium sulfate
- Isocratic elution: 0.5-1.0 mL/min, 30 minutes runtime
- Detection: 214 nm and 280 nm
- Acceptance criteria:
- Monomer (main peak): ≥95.0%
- High molecular weight species (aggregates): ≤3.0%
- Low molecular weight species (fragments): ≤2.0%
Ion-Exchange HPLC (Supporting Method)
- Column: Weak cation exchange (WCX) or strong cation exchange (SCX)
- Mobile phase: Phosphate or acetate buffer with salt gradient
- Detection: 214 nm
- Purpose: Detect charge variants (deamidation, oxidation affecting charge)
- Acceptance criteria: Main peak ≥90% (orthogonal to RP-HPLC)
6.3 Content and Potency Determination
Accurate determination of tesamorelin content in lyophilized vials employs validated quantitative methods:
| Method | Principle | Specification |
|---|---|---|
| Quantitative RP-HPLC | External standard calibration curve (5 points, 0.5-2.0 mg/mL) | 2.0 mg ± 0.3 mg per vial (90-110% of label claim) |
| UV Spectroscopy | Absorbance at 280 nm using calculated extinction coefficient | Confirmatory method, ±15% agreement with HPLC |
| Amino Acid Analysis | Quantitative AAA vs. calibrated amino acid standards | Supporting method for peptide content verification |
| Nitrogen Determination | Kjeldahl or combustion analysis (CHNS analyzer) | Alternative quantitation, nitrogen content 19.2-20.8% |
6.4 Impurities and Degradation Product Profiling
Comprehensive impurity characterization identifies process-related and degradation-related impurities requiring monitoring:
Process-Related Impurities
- Des-hexenoyl tesamorelin: Incomplete N-terminal modification (limit: ≤2.0%)
- Deletion sequences: Peptides missing one or more internal amino acids (individually ≤1.0%)
- Truncated peptides: Incomplete synthesis products (shorter than 44 residues, ≤1.5% total)
- Stereoisomers: D-amino acid incorporation or racemization (≤0.5%)
- Incomplete deprotection: Residual protecting groups (typically undetectable by RP-HPLC)
Degradation Products
- Met-27 oxidation: Methionine sulfoxide formation (≤1.5% at release, ≤3.0% at expiry)
- Deamidation: Asn/Gln conversion to Asp/Glu (≤2.0% at release, ≤4.0% at expiry)
- Aggregates: Covalent or non-covalent dimers/oligomers (≤3.0% by SEC-HPLC)
- Hydrolysis: Peptide bond cleavage products (≤1.0%)
Specified impurities exceeding 1.0% require identification by LC-MS/MS and qualification per ICH Q3B(R2) guidelines on impurities in new drug substances.
6.5 Physical and Chemical Properties Testing
Additional quality control tests characterize critical physical and chemical attributes:
- Appearance: White to off-white lyophilized powder or cake (visual inspection)
- Reconstitution: Dissolves completely within 60 seconds forming clear, colorless solution
- pH (reconstituted): 6.0-7.5 (1% solution in water)
- Water content: ≤3.0% by Karl Fischer titration (target <2.0%)
- Residual solvents:
- Acetonitrile: ≤410 ppm (ICH Class 2 solvent) by GC
- TFA: ≤0.5% by ion chromatography or 19F NMR
- DMF: ≤880 ppm (ICH Class 2 solvent) by GC if used in synthesis
- Other solvents: Per ICH Q3C limits
- Heavy metals: ≤10 ppm total by ICP-MS (Pd, Pt, other catalysts if applicable)
- Bacterial endotoxins: ≤5.0 EU/mg by Limulus Amebocyte Lysate (LAL) test for injectable products
- Sterility: Passes USP <71> sterility test (14-day incubation, aerobic and anaerobic media)
- Particulate matter (reconstituted): Meets USP <788> sub-visible particulate requirements
- Visible particles: Free of visible particulates per USP <790> (100% inspection)
6.6 Stability-Indicating Method Validation
Analytical methods must demonstrate stability-indicating capability through forced degradation studies exposing tesamorelin to stress conditions:
- Acid stress: 0.1 N HCl, 60°C, 24 hours (tests peptide bond hydrolysis)
- Base stress: 0.1 N NaOH, 60°C, 24 hours (tests β-elimination, deamidation)
- Oxidative stress: 3% H₂O₂, 25°C, 24 hours (tests Met, Trp oxidation)
- Thermal stress: 60°C dry heat, 7 days (tests thermal degradation pathways)
- Photostability: ICH Q1B Option 2 light exposure (1.2 million lux hours visible + 200 watt hours/m² UV)
Validated methods demonstrate separation of all major degradation products from the main peak, with peak purity confirmation via photodiode array detection or on-line mass spectrometry detection during RP-HPLC analysis.
7. Batch Documentation and Manufacturing Records
Comprehensive batch documentation provides complete traceability from raw materials through final product release, supporting regulatory compliance, customer audits, and internal quality reviews. The documentation system must comply with FDA 21 CFR Parts 210, 211, and 212 requirements and demonstrate consistent adherence to validated manufacturing procedures.
7.1 Master Batch Record Architecture
The Master Batch Record (MBR) defines all manufacturing operations, in-process controls, and acceptance criteria for tesamorelin production. For a peptide synthesis operation, the MBR structure includes:
Section 1: Product and Batch Identification
- Product name: Tesamorelin (free base or specified salt form)
- Product code and catalog number
- Batch size range: Minimum and maximum scale (e.g., 0.5-2.0 gram batches)
- Expected yield ranges with justification from historical data
- MBR version number with effective date and approval signatures
Section 2: Raw Materials Bill of Materials
- Complete list of Fmoc-protected amino acids with specifications and quantities
- Solid support resin specification (type, loading, particle size)
- Coupling reagents (HBTU, HOBt, HATU, DIC) with purity requirements
- Deprotection reagents (piperidine, DMF quality specifications)
- Cleavage cocktail components (TFA, scavengers with grades)
- HPLC solvents (acetonitrile, water, TFA with purity grades)
- Lyophilization excipients (mannitol, sucrose, histidine with USP/Ph.Eur. conformance)
- Each material includes: specification reference, quantity per batch unit, acceptable alternatives
Section 3: Equipment List and Qualification Status
- Peptide synthesizer (manufacturer, model, ID number, calibration status)
- Preparative HPLC system (column specifications, detector calibration)
- Lyophilizer (chamber volume, validation status, cycle qualification)
- Analytical instruments (HPLC, MS, Karl Fischer, pH meter with qualification dates)
- Verification that all equipment qualification (IQ/OQ/PQ) is current
7.2 Batch Production Record Execution
Each manufacturing batch generates a Batch Production Record (BPR) documenting actual execution of MBR procedures with real-time data capture:
Synthesis Phase Documentation
- Resin lot number, weight dispensed, loading verification results
- Each amino acid coupling: lot number, weight, activation time, coupling duration
- Deprotection monitoring: UV trace prints or electronic files, baseline return confirmation
- Kaiser test results: Pass/fail for each coupling with operator initials
- Hexenoylation reaction: reagent lots, reaction time, in-process MS confirmation
- Process deviations: Any extended coupling times, repeated couplings documented
Purification Phase Documentation
- Crude peptide weight and analytical data (purity, MS) before purification
- HPLC column lot, equilibration verification, gradient method reference
- Chromatogram for each injection with fraction collection map
- Analytical results for each fraction (purity table)
- Pooling decision with QC approval signature
- Desalting method execution with TFA removal verification data
Lyophilization Phase Documentation
- Formulation preparation: excipient lots, weights, pH measurement
- Sterile filtration: filter lot, integrity test results (bubble point)
- Fill weight verification data (sample weights throughout filling)
- Lyophilizer cycle printout: temperature and pressure profiles throughout cycle
- Product temperature data from thermocouple vials
- Cake appearance inspection results for representative vials
- Stoppering and crimping verification (torque specifications if applicable)
7.3 In-Process Control Testing Strategy
Critical quality attributes require monitoring during manufacturing to ensure process control:
| Manufacturing Stage | In-Process Test | Acceptance Criteria | Action on Failure |
|---|---|---|---|
| SPPS - Each Coupling | Kaiser (ninhydrin) test | Negative (no blue color, free amine consumed) | Repeat coupling until negative result |
| SPPS - Each Deprotection | UV monitoring at 301 nm | Return to baseline (complete Fmoc removal) | Extend deprotection time, retest |
| Hexenoylation | Test cleavage with MS analysis | ≥95% hexenoylation (MW 5135.9 Da) | Continue reaction, retest at intervals |
| Crude Peptide | Analytical HPLC and MS | Correct MW, purity ≥30% (typical range) | Proceed to purification (low purity acceptable for crude) |
| Purification Fractions | Analytical HPLC purity | ≥95% for pooling, 85-95% for repurification | Segregate fractions, reprocess or reject |
| Pre-Lyophilization Solution | Bioburden, pH, appearance | <10 CFU/100 mL, pH 6.0-7.5, clear | Re-filter (bioburden), adjust pH, clarify |
| Post-Lyophilization | Residual moisture | ≤3.0% by Karl Fischer | Extend secondary drying if possible, or reject |
7.4 Yield Calculations and Reconciliation
Yield tracking at each manufacturing stage verifies process efficiency and detects unexpected losses:
- Theoretical yield calculation: Based on resin loading × scale × molecular weight / 1000
- Crude yield: (Crude peptide weight / theoretical yield) × 100 = typically 40-60%
- Purification recovery: (Pure peptide weight / crude peptide weight) × 100 = typically 30-50%
- Lyophilization recovery: (Final vial content × vial count / pre-lyo peptide weight) × 100 = typically 85-95%
- Overall yield: (Final product weight / theoretical yield) × 100 = typically 10-25%
- Acceptance range: Process yields within ±20% of historical mean (established during validation)
Material balance reconciliation accounts for:
- Samples removed for analytical testing (documented weights)
- Rejected fractions or waste (documented and explained)
- Retained samples for stability and reference (documented)
- Discrepancies >5% require investigation as deviations
7.5 Deviation Management and CAPA System
Departures from approved MBR procedures require formal deviation documentation and investigation:
Deviation Classification
- Critical deviation: Impacts product quality, safety, or efficacy (e.g., failed sterility test, out-of-spec purity)
- Major deviation: Violates cGMP but with low quality impact (e.g., missed documentation, temperature excursion within validated range)
- Minor deviation: Administrative issues with no quality or compliance impact
Deviation Investigation Requirements
- Immediate documentation: Deviation form completed at time of discovery
- Impact assessment: QA review determines quality impact and batch disposition
- Root cause analysis: Investigation using 5-Why, fishbone, or other structured methods
- Corrective action: Immediate corrections to affected batch if feasible
- Preventive action (CAPA): Systemic changes to prevent recurrence
- Effectiveness check: Verification that CAPA successfully prevents recurrence
- Timeline: Critical deviations investigated within 5 working days, major within 30 days
Deviation trending analysis identifies patterns requiring process improvement, retraining, or revalidation activities per FDA quality systems guidance.
7.6 Batch Release Approval Workflow
Following completion of all manufacturing operations and testing, batch release requires multi-level review:
- Production review: Manufacturing supervisor verifies BPR completeness and accuracy
- QC review: Quality Control verifies all test results meet specifications
- QA review: Quality Assurance reviews entire batch record including:
- Raw material qualification and lot release documentation
- Process parameter compliance and in-process control results
- Deviation impact assessment and investigation completion
- Final product testing results vs. specifications
- Stability sample retention and storage documentation
- Final disposition: QA issues batch disposition (Release, Reject, or Hold for further investigation)
- Certificate of Analysis: Generated and signed by authorized QA personnel upon release decision
8. Stability Studies and Shelf-Life Determination
Comprehensive stability programs establish storage conditions, retest intervals, and shelf-life specifications for tesamorelin drug substance and drug product. Stability studies follow ICH Q1A(R2) guidelines for stability testing of new drug substances and drug products, generating data that supports commercial storage recommendations and expiration dating.
8.1 Stability Program Design and Study Types
Formal stability programs for tesamorelin encompass multiple study configurations:
Long-Term Stability Studies (Drug Substance)
- Storage condition: -20°C ± 5°C (primary long-term storage recommendation)
- Alternative condition: 2-8°C (refrigerated storage evaluation for distribution)
- Duration: Minimum 12 months, extending to 36 months for commercial products
- Testing frequency: 0, 3, 6, 9, 12, 18, 24, 36 months
- Sample size: Minimum three production batches from different manufacturing campaigns
- Container: Commercial packaging configuration (sealed vials with desiccant)
Accelerated Stability Studies
- Storage condition: 25°C ± 2°C / 60% RH ± 5% RH
- Duration: 6 months minimum
- Testing frequency: 0, 1, 3, 6 months
- Purpose: Predict long-term stability trends and identify degradation pathways
- Statistical analysis: Arrhenius modeling to extrapolate shelf-life from accelerated data
Intermediate Stability Studies
- Storage condition: 30°C ± 2°C / 65% RH ± 5% RH
- Duration: 12 months (required if significant change occurs at accelerated conditions)
- Testing frequency: 0, 3, 6, 9, 12 months
Stress Testing (Forced Degradation)
- Thermal stress: 40°C, 50°C, 60°C for lyophilized solid (1-4 weeks)
- Humidity stress: 75% RH at 40°C (moisture uptake and stability assessment)
- Light exposure: ICH Q1B photostability testing (Option 1 or Option 2)
- Freeze-thaw cycling: -20°C to +25°C, 5 cycles (solution stability assessment)
- Reconstituted solution: Time-course stability at 2-8°C and 25°C (in-use stability)
8.2 Stability-Indicating Analytical Test Panel
Stability samples undergo comprehensive analytical testing using validated stability-indicating methods:
| Test | Method | Release Specification | Shelf-Life Specification |
|---|---|---|---|
| Appearance | Visual inspection | White to off-white lyophilized cake | White to off-white, no significant discoloration |
| Identification (HPLC) | RP-HPLC retention time | Matches reference standard ±2% | Matches reference standard ±2% |
| Identification (MS) | ESI-MS molecular weight | 5135.9 ± 1.0 Da | 5135.9 ± 1.0 Da |
| Purity (RP-HPLC) | RP-HPLC at 214 nm | ≥95.0% | ≥92.0% |
| Assay (Content) | Quantitative HPLC | 90.0-110.0% of label claim | 85.0-115.0% of label claim |
| Water Content | Karl Fischer titration | ≤3.0% | ≤5.0% |
| Aggregates | SEC-HPLC | ≤3.0% | ≤5.0% |
| Met Oxidation | RP-HPLC peak tracking | ≤1.5% (Met-sulfoxide peak) | ≤3.0% |
| Deamidation | Ion-exchange HPLC or RP-HPLC | ≤2.0% | ≤4.0% |
| pH (reconstituted) | pH meter (1% solution) | 6.0-7.5 | 5.5-8.0 |
8.3 Degradation Pathway Characterization
Tesamorelin exhibits several well-characterized degradation pathways requiring monitoring and control:
Methionine Oxidation (Met-27)
- Primary degradation pathway: Methionine at position 27 oxidizes to methionine sulfoxide
- Mechanism: Oxygen-mediated or metal-catalyzed oxidation (Cu²⁺, Fe³⁺)
- Detection: RP-HPLC shows earlier-eluting peak (+16 Da by MS for sulfoxide)
- Temperature dependence: Accelerated at elevated temperature and in solution
- Control strategies:
- Oxygen exclusion through nitrogen/argon backfilling during lyophilization
- Storage at -20°C minimizes oxidation rate
- Antioxidant excipients (histidine provides some protection)
- Light protection (photocatalyzed oxidation prevention)
Deamidation
- Susceptible residues: Asparagine (Asn-8) and glutamine residues (Gln-9, Gln-12, Gln-16, Gln-24, Gln-25)
- Mechanism: pH and temperature-dependent hydrolysis or cyclic imide intermediate formation
- Products: Conversion to aspartic acid (Asn→Asp) or glutamic acid (Gln→Glu)
- Detection: Ion-exchange HPLC (charge change) or RP-HPLC (polarity change)
- Control strategies:
- Formulation pH optimization (6.0-7.5 minimizes deamidation rate)
- Low temperature storage (-20°C significantly reduces rate)
- Lyophilization to remove water (deamidation requires water)
Aggregation
- Types: Non-covalent aggregates (hydrophobic interaction, hydrogen bonding) and covalent aggregates (disulfide crosslinking if oxidized Met forms reactive species)
- Factors promoting aggregation: Elevated temperature, agitation, freeze-thaw cycling, reconstitution concentration
- Detection: SEC-HPLC shows high molecular weight species, dynamic light scattering (DLS)
- Control strategies:
- Bulking agents (mannitol, sucrose) in lyophilized formulation
- Single-use vial configuration (avoid repeated freeze-thaw)
- Storage at -20°C (minimizes aggregation kinetics)
- Gentle reconstitution instructions (swirl, do not shake vigorously)
Hydrolysis
- Susceptible bonds: Asp-X peptide bonds (particularly Asp-Ala at position 3-4)
- Mechanism: Acid-catalyzed hydrolysis in low pH or cyclic imide-mediated cleavage
- Detection: RP-HPLC shows fragment peptides, confirmed by MS
- Control: pH maintenance in neutral range, low temperature, dry storage
8.4 Shelf-Life Determination and Statistical Analysis
Shelf-life calculation employs linear regression analysis of stability data for each critical quality attribute. The statistical approach follows ICH Q1E guidelines for evaluation of stability data:
- Data transformation: Plot attribute values vs. time for each storage condition
- Regression analysis: Linear regression with 95% confidence intervals
- Shelf-life determination: Time point where lower 95% CI intersects acceptance criterion
- Pooling assessment: Evaluate whether data from multiple batches can be pooled (homogeneity of regression slopes and intercepts)
- Specification setting: Release specifications tighter than shelf-life to ensure product meets specifications throughout dating period
Typical tesamorelin shelf-life determinations based on stability data:
- Lyophilized powder at -20°C: 24-36 months (limited by purity drift, primarily oxidation and deamidation)
- Lyophilized powder at 2-8°C: 12-18 months (accelerated degradation vs. -20°C)
- Reconstituted solution at 2-8°C: 7-14 days (limited by oxidation and aggregation)
- Room temperature (post-reconstitution): 24 hours maximum (significant degradation risk)
8.5 Container-Closure System Qualification
Container-closure integrity testing ensures product protection throughout shelf-life:
- Moisture permeation testing: Gravimetric water vapor transmission rate at storage conditions (target: <0.5% moisture gain over shelf-life)
- Oxygen permeability: Headspace oxygen monitoring over time for nitrogen/argon backfilled vials (target: <5% O₂ at end of shelf-life)
- Extractables and leachables: Studies per USP <1663> and <1664> identifying potential rubber stopper or glass container leachables
- Seal integrity: Vacuum decay testing, helium leak testing, or dye ingress methods validating hermetic seal
- Mechanical testing: Shipping simulation (vibration, drop testing) followed by integrity verification
8.6 Post-Approval Stability Commitment and Ongoing Monitoring
Ongoing stability programs continue throughout commercial lifecycle:
- Annual commitment batches: Minimum 1 batch per year on long-term stability (first 3 years post-approval)
- Testing through expiry: Extend testing through expiration date plus 3-6 months
- Out-of-specification investigation: Any OOS result triggers formal investigation and assessment of need for field action
- Annual stability reporting: Compilation of all ongoing stability data with trending analysis
- Comparability protocols: Stability requirements for manufacturing changes, scale-up, or site transfers
- Specification revision: Stability data may support tightening or loosening of specifications based on accumulating knowledge
Stability data submission to regulatory authorities follows regional requirements, with periodic stability updates included in annual reports and variations/supplements per ICH Q1A(R2) stability testing guidelines.
9. Storage, Handling, and Distribution Requirements
Proper storage, handling, and distribution practices maintain tesamorelin quality from manufacturing release through end-user administration. Temperature control represents the most critical factor, with secondary considerations including light protection, moisture exclusion, and prevention of mechanical stress during shipping and handling operations.
9.1 Storage Conditions and Environmental Requirements
Tesamorelin storage specifications vary depending on physical form and intended use timeline:
Lyophilized Drug Substance/Product Storage
- Long-term storage (preferred): -20°C ± 5°C in sealed containers
- Alternative refrigerated storage: 2-8°C (for distribution and intermediate-term storage up to 12-18 months)
- Humidity control: Product protected by hermetically sealed vials; external storage area RH not critical but controlled humidity (30-60% RH) prevents condensation during temperature cycling
- Light protection: Store in original cartons to protect from light exposure; avoid direct sunlight or strong UV sources
- Orientation: Upright vial storage prevents stopper contact with lyophilized cake during storage
Bulk Intermediate Storage
- Purified peptide (pre-lyophilization solution): -20°C or -80°C in sealed containers, maximum 30 days (proceeding immediately to lyophilization preferred)
- Crude peptide (post-cleavage): -20°C in sealed containers with desiccant, maximum 6 months (shorter storage minimizes oxidation)
- Resin-bound peptide: -20°C under nitrogen or argon atmosphere, maximum 7 days (cleavage immediately after synthesis completion preferred)
Reconstituted Solution Storage (In-Use Conditions)
- Temperature: 2-8°C immediately after reconstitution with sterile water or specified diluent
- Stability period: Use within 7-14 days (specific timeline based on validation data and labeled instructions)
- Container: Original vial with sterile rubber stopper replaced after withdrawal
- Freezing: Do NOT freeze reconstituted solution (causes aggregation and potential loss of activity)
- Light protection: Protect from light during refrigerated storage
- Multi-dose considerations: If multi-dose vial, use preservative-containing diluent; single-dose vials preferred for sterility assurance
9.2 Environmental Monitoring and Temperature Control
Storage facilities require continuous environmental monitoring with automated alarm systems:
| Storage Area | Monitoring Parameters | Recording Frequency | Alarm Limits |
|---|---|---|---|
| Freezer (-20°C) | Temperature (validated probe) | Continuous recording, 10-15 min intervals | Alert: -23°C or -17°C Action: -25°C or -15°C |
| Refrigerator (2-8°C) | Temperature (validated probe) | Continuous recording, 10-15 min intervals | Alert: <1°C or >9°C Action: <0°C or >10°C |
| Controlled Room Temp | Temperature and RH | Continuous recording, 30 min intervals | Alert: <18°C or >27°C Action: <15°C or >30°C |
| All storage areas | Light exposure levels | Annual qualification | No direct sunlight, <200 lux ambient light |
Temperature Excursion Management
- Immediate investigation: Document excursion duration, temperature range, affected product lots
- Impact assessment: Utilize stability data and Arrhenius kinetic modeling to estimate degradation during excursion
- Testing protocol: Consider analytical testing of excursion-exposed material if borderline impact
- Product disposition: Release (if within validated limits), quarantine (pending investigation), or reject
- Customer notification: Inform customers if distributed product experienced temperature excursion exceeding validated limits
- Root cause analysis: Identify equipment failure, human error, or process gaps contributing to excursion
- CAPA implementation: Corrective action to prevent recurrence (equipment repair, alarm adjustment, training)
9.3 Shipping and Distribution Cold-Chain Management
Distribution operations require validated shipping containers and procedures maintaining temperature throughout transit:
Shipping Container Qualification
- Thermal performance qualification: Testing under summer (high ambient), winter (low ambient), and standard conditions
- Duration testing: Demonstrate temperature maintenance for 2× maximum expected transit time (e.g., 96 hours for 48-hour maximum shipping)
- Temperature mapping: Multiple data loggers throughout container volume during qualification
- Acceptance criteria: All monitored locations maintain 2-8°C (or -20°C for frozen shipment) throughout entire test duration
- Requalification frequency: Annual requalification or following significant design changes to packaging
- Seasonal configurations: Different cold pack quantities or insulation for summer vs. winter shipments
Shipping Procedure Elements
- Pre-conditioning: Verify product at proper storage temperature minimum 24 hours before shipment
- Cold packs or dry ice: Sufficient quantity based on qualified packaging and transit time plus 24-hour contingency
- Temperature monitoring: Data logger included with each shipment or representative validation approach
- Insulated containers: Validated thermal performance with certificate on file
- Package labeling: Clear "Keep Refrigerated" or "Keep Frozen" labels on all surfaces
- Orientation marking: "This Side Up" indicators preventing inversion during transit
- Transit time limits: Maximum allowable time in transit before temperature maintenance cannot be assured
- Courier selection: Validated carriers with cold-chain experience and tracking capabilities
Documentation and Record Keeping
- Shipping log: Date, time, destination, tracking number, data logger ID
- Temperature logger data: Review upon return of logger or customer return of data
- Exception reporting: Any shipments with temperature excursions documented and investigated
- Customer notification: Proactive communication if shipping delay or temperature excursion suspected
9.4 Handling Procedures and Personnel Training
All personnel handling tesamorelin require training on proper procedures:
- Minimize ambient exposure: Return product to proper storage within 15 minutes of removal
- Avoid freeze-thaw cycles: For reconstituted solution, aliquot into smaller volumes if multiple uses needed
- Aseptic technique: Use sterile needles and syringes when reconstituting for injection
- Gentle mixing: Swirl gently to dissolve; do NOT shake vigorously (causes foaming and potential aggregation)
- Personal protective equipment: Lab coat, gloves, safety glasses when handling (prevent product contamination and personal exposure)
- Spill cleanup: Procedure for peptide spill cleanup with decontamination and waste disposal
- Equipment cleaning: Validated cleaning procedures for equipment contacting tesamorelin
Standard operating procedures include photographic illustrations, step-by-step instructions, and competency assessment requirements before personnel authorization for independent handling.
9.5 Inventory Management and Stock Rotation
Inventory control systems prevent use of expired or out-of-specification material:
- Electronic tracking system: Real-time inventory database with automatic expiry date alerts (30-day and 7-day warnings)
- FEFO system: First Expired, First Out selection ensures oldest material used first
- Physical segregation: Clear separation of released, quarantined, and rejected material with appropriate labeling
- Expiration date verification: Check at receipt, storage assignment, and issuance to production or customers
- Periodic audits: Monthly physical inventory count verified against electronic records
- Expiry reconciliation: Expired material promptly removed from inventory and destroyed per documented procedures
- Lot traceability: Complete forward and backward traceability for recall capability
9.6 Customer Receiving Instructions and Acceptance Criteria
Customers receive written guidance on receipt inspection and acceptance:
- Immediate inspection: Examine package immediately upon delivery, note any shipping damage
- Temperature verification:
- If data logger included: Review temperature history, verify maintenance of 2-8°C throughout shipping
- If cold packs used: Verify packs still cold/frozen upon receipt
- If dry ice used: Verify dry ice still present (some sublimation acceptable)
- Product inspection: Visual examination of vials through carton window (if available) or upon opening
- Check for vial breakage or cracks
- Verify lyophilized cake appearance (white to off-white, intact cake)
- Look for signs of reconstitution or moisture ingress (wetness, cake collapse)
- Verify labels match Certificate of Analysis and purchase order
- Documentation review:
- Certificate of Analysis batch number matches vial labels
- Expiration date provides adequate working time for intended use
- Packing slip matches order and received goods
- Prompt storage: Transfer to appropriate storage (-20°C or 2-8°C per labeling) within 30 minutes of opening shipping container
- Damage or excursion reporting: Contact supplier immediately (within 24 hours) if temperature excursion or product damage observed
- Retention of shipping materials: Retain shipping container, temperature logger, and documentation if problems observed (required for investigation)
Receipt inspection procedures support quality systems and regulatory compliance per FDA Drug Supply Chain Security Act requirements and customer internal quality programs.
10. Certificate of Analysis and Release Documentation
The Certificate of Analysis (CoA) serves as the primary quality documentation supporting each tesamorelin batch release, certifying conformance to established specifications and enabling customer quality verification. The CoA must be accurate, complete, and generated through validated electronic systems or controlled manual procedures with appropriate review and approval workflows.
10.1 Certificate of Analysis Structure and Required Elements
A comprehensive tesamorelin Certificate of Analysis includes the following components:
Header Section
- Manufacturer/supplier name, complete address, and contact information (phone, email, website)
- Product name: Tesamorelin or trade name (e.g., Tesamorelin Acetate if salt form specified)
- Catalog/product code: Internal product identification number
- Batch/Lot number: Unique identifier with full traceability to manufacturing records
- Manufacturing date: Date of final lyophilization and sealing
- Expiration date or retest date: Based on validated stability data
- CoA issue date and document version number
- Storage conditions: Store at -20°C or 2-8°C as specified
- Quantity: Number of vials, vial size, and amount per vial (e.g., 10 vials, 2 mg per vial)
Product Identification and Structure
- Molecular formula: C₂₂₁H₃₆₆N₇₂O₆₇S
- Molecular weight: 5135.9 Da (average mass, free base)
- CAS number: 218949-48-5 (tesamorelin free base)
- Sequence: Hexenoyl-Tyr-Ala-Asp-Ala-Ile-Phe-Thr-Gln-Ser-Tyr-Arg-Lys-Val-Leu-Ala-Gln-Leu-Ser-Ala-Arg-Lys-Leu-Leu-Gln-Asp-Ile-Met-Ser-Arg-Gln-Gln-Gly-Glu-Ser-Asn-Gln-Glu-Arg-Gly-Ala-Arg-Ala-Arg-Leu-NH₂
- Modification: Trans-3-hexenoyl group attached to N-terminal tyrosine
10.2 Analytical Test Results Presentation
Test results appear in tabular format showing test name, analytical method, specification limits, and actual results for the specific batch:
| Test | Method | Specification | Result (Example Batch) |
|---|---|---|---|
| Appearance | Visual inspection | White to off-white lyophilized powder or cake | White lyophilized cake |
| Identification (HPLC) | RP-HPLC retention time | Conforms to reference standard ±2% | Conforms (RT: 28.7 min) |
| Identification (MS) | ESI-MS | MW: 5135.9 ± 1.0 Da | 5136.1 Da (conforms) |
| Purity by HPLC | RP-HPLC at 214 nm | ≥95.0% | 96.8% |
| Assay (Content) | Quantitative HPLC | 90.0-110.0% of label claim | 98.3% |
| Water Content | Karl Fischer titration | ≤3.0% | 1.8% |
| Aggregates (SEC) | SEC-HPLC | ≤3.0% | 1.2% |
| Acetonitrile | GC-FID | ≤410 ppm | <50 ppm |
| TFA Content | Ion chromatography | ≤0.5% | 0.12% |
| Heavy Metals | ICP-MS | ≤10 ppm | <2 ppm |
| Bacterial Endotoxins | LAL (kinetic chromogenic) | ≤5.0 EU/mg | <0.5 EU/mg |
| Sterility | USP <71> (membrane filtration) | No growth in 14 days | Passes (no growth detected) |
| pH (reconstituted) | pH meter (1% solution) | 6.0-7.5 | 6.8 |
10.3 Supporting Analytical Data Appendices
Comprehensive CoAs may include representative analytical data as appendices or referenced attachments:
- RP-HPLC chromatogram: Annotated trace at 214 nm showing main peak with retention time, integration results, and identified impurity peaks if present
- Mass spectrum: ESI-MS or MALDI-TOF spectrum showing molecular ion envelope with charge state deconvolution to average mass
- SEC-HPLC chromatogram: Size-exclusion profile demonstrating monomeric state with minimal aggregates or fragments
- Amino acid analysis report: Table showing each amino acid with theoretical vs. observed molar ratios
- Peptide map: LC-MS/MS analysis of tryptic digest confirming sequence
Electronic CoAs may embed chromatograms as images or link to electronic raw data files. Printed CoAs reference archived data locations with unique file identifiers.
10.4 Storage, Handling, and Reconstitution Instructions
The CoA specifies proper storage and handling to maintain product quality:
Storage Instructions
- Store at -20°C upon receipt for long-term storage (up to expiration date)
- Alternative: Store at 2-8°C for up to 12-18 months (verify specific product labeling)
- Protect from light: Keep vials in original carton until use
- Do not freeze reconstituted solution
- Avoid repeated freeze-thaw cycles of lyophilized product
Reconstitution Instructions (for Injectable Formulations)
- Reconstitute with sterile water for injection, USP or bacteriostatic water (if multi-dose vial)
- Add 2.0 mL sterile diluent per 2 mg vial (example; verify specific product instructions)
- Swirl gently to dissolve; do NOT shake vigorously
- Allow 30-60 seconds for complete dissolution
- Inspect visually: Solution should be clear and colorless, free of particles
- Use immediately after reconstitution or store at 2-8°C for maximum 7-14 days (verify specific stability data)
- Discard any unused portion after specified in-use period
Handling Precautions
- For research use only (if non-pharmaceutical grade) or For clinical/therapeutic use (if pharmaceutical grade)
- Handle using aseptic technique if intended for injection
- Wear appropriate personal protective equipment
- Avoid skin contact and inhalation of powder
- Consult Safety Data Sheet (SDS) for additional safety information
10.5 Quality Assurance Certification and Approvals
The CoA footer contains quality assurance certification and approval elements:
- Conformance statement: "This batch has been manufactured and tested in accordance with current Good Manufacturing Practices (cGMP) and conforms to all established specifications."
- QA approval:
- Signature (electronic or handwritten) of authorized Quality Assurance personnel
- Printed name and title
- Approval date
- Regulatory status:
- "For Research Use Only - Not for Human or Veterinary Use" (if research grade)
- "Pharmaceutical Grade - For Clinical Use" (if approved for human use)
- Reference to Drug Master File (DMF) number if applicable
- Contact information: Technical support phone number and email for questions
- Document control: Unique document ID, version number, page numbering (e.g., Page 1 of 2)
10.6 Supplementary Documentation Available
Additional supporting documentation available upon customer request:
- Safety Data Sheet (SDS): GHS-compliant safety information including hazard classification, handling precautions, first aid, disposal
- Regulatory support file (RSF) or Drug Master File (DMF): Comprehensive manufacturing and control information for regulatory submissions
- Certificate of Origin: Country of manufacture declaration for import/export compliance
- Statement of GMP compliance: Letter certifying manufacturing under cGMP with facility registration information
- Stability summary: Summary of stability data supporting expiration dating and storage recommendations
- Amino acid sequence certificate: Detailed sequence confirmation data from peptide mapping or Edman degradation
- Residual solvents complete panel: Extended ICH Q3C solvent testing results (if requested)
- Elemental impurities data: Detailed ICP-MS results for ICH Q3D elements (if requested)
- Sterility test raw data: Complete sterility testing documentation with media lot information
- TSE/BSE statement: Declaration of no materials of animal origin used in manufacturing (Transmissible Spongiform Encephalopathy/Bovine Spongiform Encephalopathy)
10.7 Electronic Certificate Systems and Data Integrity
Modern quality systems employ electronic CoA generation with robust data integrity controls:
- LIMS integration: Automated CoA generation from Laboratory Information Management System with direct data pull from analytical instruments
- Electronic signatures: 21 CFR Part 11 compliant approval workflows with electronic signature capture
- PDF/A archival format: Long-term archival PDF format with embedded metadata and searchability
- QR code verification: QR code linking to online verification portal where customers can authenticate CoA
- Blockchain verification: Emerging technology for tamper-proof CoA authentication
- Audit trails: Complete electronic audit trail of CoA generation, modifications, approvals, and distribution
- Customer portal access: Self-service portal for CoA retrieval by batch number
- Version control: Automated versioning for corrected or reissued CoAs with change documentation
- Digital watermarking: Prevention of unauthorized modification or duplication
10.8 CoA Review and Approval Process
Prior to CoA issuance, Quality Assurance performs comprehensive multi-level review:
- Data verification: Confirm all test results correctly transferred from laboratory worksheets or LIMS
- Specification compliance: Verify all results meet established acceptance criteria
- Calculation verification: Independent check of all calculations (assay, purity percentages, dilutions)
- Testing completeness: Confirm all required tests performed by qualified personnel using validated methods
- Deviation review: Assess any manufacturing or testing deviations and their resolution
- Retest review: Verify appropriateness of any retesting and compliance with retest policies
- Expiration date verification: Confirm correct expiration date calculation based on stability data
- Batch record review: Verify Batch Production Record review completion and release approval
- Format accuracy: Final check of document formatting, spelling, completeness
- Authorized approval: Signature by personnel with delegated authority for batch release
CoA approval constitutes the final quality gate enabling product distribution to customers, certifying conformance to all applicable requirements including FDA cGMP regulations and customer-specific quality agreements.
Conclusion: Manufacturing Excellence in Tesamorelin Production
Tesamorelin manufacturing represents a complex integration of peptide synthesis chemistry, analytical science, quality systems, and regulatory compliance expertise. The 44-residue sequence with N-terminal hexenoylation modification requires carefully optimized synthesis protocols, sophisticated purification strategies, and comprehensive analytical testing to consistently achieve pharmaceutical-grade quality specifications.
Successful tesamorelin production demands investment in specialized infrastructure including automated peptide synthesizers capable of handling long, aggregation-prone sequences; preparative HPLC systems with column capacity and resolution appropriate for complex peptide purification; pharmaceutical-grade lyophilization equipment with validated cycle development; and comprehensive analytical laboratories equipped with HPLC, mass spectrometry, amino acid analysis, and specialized testing capabilities for peptide characterization and impurity profiling.
Manufacturing facilities must implement robust quality systems encompassing validated manufacturing procedures, comprehensive batch documentation, stability programs supporting shelf-life determinations, environmental monitoring and control, cold-chain distribution validation, and electronic quality systems enabling complete traceability and data integrity. Personnel qualifications represent an equally critical investment, with synthesis chemists, purification specialists, analytical scientists, and quality professionals all requiring specialized training in peptide chemistry, cGMP regulations, and statistical process control methodologies.
The technical specifications presented throughout this manufacturing profile provide systematic guidance for establishing or optimizing tesamorelin production operations. Key quality outcomes achieved through implementation of these specifications include batch-to-batch consistency demonstrated through statistical process control and trending analysis; impurity profiles consistently below regulatory thresholds through optimized synthesis and purification; extended shelf-life enabled by formulation optimization and protective packaging systems; and complete regulatory compliance supporting commercial distribution or clinical trial material supply.
Ongoing process improvement initiatives drive manufacturing excellence through continuous monitoring of process capability, investigation and elimination of assignable causes of variation, adoption of emerging analytical technologies providing enhanced characterization capabilities, and implementation of quality-by-design principles incorporating process analytical technology and real-time release testing where appropriate. Regular participation in industry working groups, review of scientific literature, and collaboration with equipment vendors ensures manufacturing operations remain current with evolving best practices and technological advances.
For complementary technical resources addressing related aspects of peptide manufacturing, refer to additional PeptideForge.com technical profiles including sermorelin synthesis and purification methodologies, ipamorelin manufacturing specifications, CJC-1295 production protocols, GHRP-2 quality control testing, and DSIP stability and formulation development. These resources collectively provide comprehensive technical guidance supporting pharmaceutical-grade peptide manufacturing across diverse therapeutic applications.
The rigor and detail presented in this tesamorelin manufacturing profile reflect the pharmaceutical industry's commitment to quality, safety, and efficacy as foundational principles. By adhering to these specifications and continuously advancing manufacturing capabilities, production facilities provide researchers, clinicians, and pharmaceutical developers with reliable, high-quality tesamorelin supporting scientific discovery, clinical research, and therapeutic development in growth hormone modulation and related endocrine applications.
References and Regulatory Resources
- U.S. Food and Drug Administration. (2023). Current Good Manufacturing Practice (CGMP) Regulations for Drugs. 21 CFR Parts 210 and 211. Available at: https://www.fda.gov/drugs/pharmaceutical-quality-resources/current-good-manufacturing-practice-cgmp-regulations
- International Council for Harmonisation (ICH). (2023). ICH Q7: Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients. Available at: https://www.ich.org/page/quality-guidelines
- International Council for Harmonisation (ICH). (2023). ICH Q3B(R2): Impurities in New Drug Substances. Available at: https://www.ich.org/page/quality-guidelines
- United States Pharmacopeia. (2024). USP Harmonization Standards for Peptide Manufacturing. USP-NF General Chapters. Available at: https://www.usp.org/harmonization-standards/pdg
- U.S. Food and Drug Administration. (2022). Guidance for Industry: Lyophilization of Parenteral Products. Center for Drug Evaluation and Research. Available at: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/guidance-industry-lyophilization-parenteral-products
- International Council for Harmonisation (ICH). (2023). ICH Q1A(R2): Stability Testing of New Drug Substances and Products. Available at: https://www.ich.org/page/quality-guidelines
- U.S. Food and Drug Administration. (2006). Guidance for Industry: Quality Systems Approach to Pharmaceutical CGMP Regulations. Available at: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/guidance-industry-quality-systems-approach-pharmaceutical-cgmp-regulations
- U.S. Food and Drug Administration. (2023). Facts about the Current Good Manufacturing Practices (CGMPs). Available at: https://www.fda.gov/drugs/pharmaceutical-quality-resources/facts-about-current-good-manufacturing-practices-cgmps
- U.S. Food and Drug Administration. (2021). Drug Supply Chain Security Act (DSCSA). Available at: https://www.fda.gov/drugs/drug-approvals-and-databases/drug-supply-chain-security-act-dscsa
- Bachem AG. (2024). Solid Phase Peptide Synthesis (SPPS) Explained: Technical Guide. Available at: https://www.bachem.com/articles/peptides/solid-phase-peptide-synthesis-explained/