CJC-1295 Manufacturing Profile: Technical Specifications and Quality Standards

CJC-1295 Manufacturing Profile: Technical Specifications and Quality Standards

1. Introduction to CJC-1295 Manufacturing

CJC-1295 (Drug Affinity Complex: Growth Hormone Releasing Hormone) represents a synthetic analog of growth hormone-releasing hormone (GHRH) requiring precise manufacturing protocols to ensure product consistency, purity, and stability. This modified peptide consists of 30 amino acids with a Drug Affinity Complex (DAC) component that extends its half-life through albumin binding. Manufacturing facilities must maintain stringent quality control measures throughout synthesis, purification, and final product formulation to meet pharmaceutical-grade specifications.

The manufacturing process for CJC-1295 demands specialized equipment, validated procedures, and comprehensive documentation systems compliant with current Good Manufacturing Practices (cGMP) as outlined by the FDA's guidance on cGMP regulations for peptide therapeutics. Production facilities must implement quality management systems that address all aspects of peptide synthesis, from raw material qualification through final product release testing.

This technical profile provides detailed specifications for CJC-1295 manufacturing operations, including synthesis parameters, purification methodologies, analytical testing requirements, batch documentation standards, stability protocols, and certificate of analysis criteria. Manufacturing personnel and quality control professionals will find comprehensive guidance on process controls, acceptance criteria, and regulatory compliance requirements specific to this modified GHRH analog.

2. Solid-Phase Peptide Synthesis (SPPS) Process

CJC-1295 production utilizes Fmoc (9-fluorenylmethyloxycarbonyl) solid-phase peptide synthesis methodology, which provides superior efficiency and purity profiles compared to Boc-based strategies for peptides of this length. The synthesis process initiates with resin selection and proceeds through iterative coupling and deprotection cycles to assemble the 30-amino acid sequence with the DAC modification.

2.1 Resin Selection and Loading

Manufacturing begins with selection of appropriate solid support resin. For CJC-1295, Rink Amide MBHA resin (100-200 mesh, 0.4-0.8 mmol/g substitution) provides optimal performance for C-terminal amidation. Resin loading must be verified through quantitative Fmoc determination using UV spectroscopy at 301 nm following piperidine deprotection, with acceptance criteria of 90-110% of theoretical loading capacity.

Pre-swelling procedures require resin exposure to N,N-dimethylformamide (DMF) for minimum 30 minutes prior to first coupling cycle. Resin quality specifications must include:

  • Particle size distribution: 100-200 mesh (74-149 μm)
  • Swelling volume: minimum 4 mL/g in DMF
  • Moisture content: maximum 5% by Karl Fischer titration
  • Heavy metal content: less than 10 ppm total
  • Functional group loading: 0.4-0.8 mmol/g verified by elemental analysis

2.2 Coupling Cycle Parameters

Each amino acid coupling cycle follows validated protocols using HBTU/HOBt activation chemistry. The standard coupling procedure employs 3-fold molar excess of Fmoc-protected amino acid relative to resin loading, activated with HBTU (3 equivalents) and HOBt (3 equivalents) in presence of DIEA (6 equivalents). Coupling reactions proceed for 45-60 minutes with mechanical agitation, maintaining temperature at 20-25°C.

Critical process parameters for coupling reactions include:

Parameter Specification Monitoring Method
Amino Acid Excess 3.0 equivalents ± 0.2 Gravimetric verification
Activation Time 2-5 minutes Process timing validation
Coupling Duration 45-60 minutes Automated synthesis monitoring
Temperature Control 20-25°C ± 2°C Continuous temperature monitoring
DMF Quality Peptide synthesis grade, <0.005% water Karl Fischer analysis per batch
Coupling Efficiency >99.5% by Kaiser test Colorimetric ninhydrin assay

2.3 Deprotection and Monitoring

Fmoc deprotection employs 20% piperidine in DMF (v/v) for two sequential treatments: initial 5-minute treatment followed by 15-minute treatment. Deprotection completion must be verified through UV monitoring of dibenzofulvene-piperidine adduct formation at 301 nm, with baseline return confirming complete Fmoc removal prior to subsequent coupling.

Quality control during synthesis requires Kaiser (ninhydrin) testing after each coupling cycle to verify completion. Negative Kaiser test (no blue coloration) confirms successful coupling with free amine consumption. Difficult couplings, particularly for sterically hindered residues, may require double coupling procedures with extended reaction times up to 120 minutes.

2.4 DAC Modification

The Drug Affinity Complex modification distinguishes CJC-1295 from standard GHRH analogs. This modification involves attachment of maleimidoproprionic acid (MPA) at the lysine-15 epsilon amino group during solid-phase synthesis. The DAC coupling requires specialized handling:

  • Selective deprotection of Lys-15 side chain using 2% hydrazine in DMF
  • MPA activation using EDC/NHS chemistry (5 equivalents each)
  • Extended coupling time of 4-6 hours with continuous monitoring
  • Protection of other reactive sites through orthogonal protection strategies
  • Verification of DAC attachment through mass spectrometry during synthesis

DAC modification efficiency must exceed 95% as determined by analytical HPLC and mass spectrometry analysis of test cleavage samples. This critical quality attribute directly impacts the peptide's pharmacokinetic profile and requires rigorous process validation as specified in ICH Q7 guidelines for API manufacturing.

3. Cleavage and Crude Peptide Recovery

Following completion of solid-phase assembly, the peptide undergoes cleavage from the resin support with simultaneous removal of side-chain protecting groups. This critical operation requires precise control of reagent composition, reaction conditions, and work-up procedures to maximize yield while minimizing oxidation and other degradation pathways.

3.1 Cleavage Cocktail Preparation

CJC-1295 cleavage employs Reagent K formulation: trifluoroacetic acid (TFA) 82.5%, phenol 5%, water 5%, thioanisole 5%, and 1,2-ethanedithiol (EDT) 2.5% by volume. This scavenger cocktail provides comprehensive protection against cationic species generated during deprotection, particularly for arginine, tryptophan, and methionine residues present in the CJC-1295 sequence.

Cleavage cocktail specifications require:

  • TFA purity: minimum 99.5%, peptide synthesis grade
  • Phenol: freshly redistilled or analytical grade with BHT stabilizer
  • Water: Type I ultrapure, 18.2 MΩ·cm resistivity
  • Thioanisole: anhydrous, 99% minimum purity
  • EDT: 98% minimum purity, used within 6 months of opening
  • Volumetric verification: ±2% of target volumes

3.2 Cleavage Process Parameters

The cleavage reaction proceeds at ambient temperature (20-25°C) for 2-4 hours with periodic mixing. Reaction scale typically employs 10-15 mL cleavage cocktail per gram of resin. The reaction vessel must be sealed to prevent moisture ingress while allowing periodic pressure release from carbon dioxide generation during Pbf and tBu deprotection.

Process controls during cleavage include:

Parameter Specification Critical Quality Impact
Cleavage Duration 2-4 hours Complete deprotection without excessive degradation
Temperature 20-25°C ± 2°C Minimize oxidation and deletion sequences
Cocktail Volume 10-15 mL/g resin Adequate scavenger capacity and mixing
Resin Separation Complete filtration within 15 minutes Minimize peptide exposure to acidic conditions
TFA Removal Rotary evaporation at <30°C Prevent thermal degradation during concentration

3.3 Precipitation and Washing

Following resin filtration, crude peptide precipitation occurs through addition of the TFA solution to cold diethyl ether (10-fold volume excess) maintained at -20 to 0°C. The precipitate forms immediately and requires collection through centrifugation at 3000-5000 × g for 10 minutes at 4°C. Multiple ether wash cycles (minimum three) remove scavengers, protecting group residues, and TFA.

Crude peptide drying proceeds under vacuum at room temperature or through lyophilization from dilute acetic acid solution. The resulting crude powder typically exhibits 30-60% purity by analytical HPLC, with the target peptide as the major component. Crude yield calculations based on resin loading typically achieve 40-70% for well-optimized synthesis protocols.

Crude peptide storage prior to purification requires protection from moisture, light, and oxidation. Storage at -20°C in sealed containers with desiccant maintains stability for up to 6 months, though immediate processing to purification minimizes potential degradation pathways.

4. High-Performance Liquid Chromatography Purification

CJC-1295 purification employs preparative reversed-phase HPLC to achieve pharmaceutical-grade purity specifications. The purification process must remove truncated sequences, deletion peptides, stereoisomers, oxidation products, and residual synthesis reagents while maintaining product integrity and maximizing recovery yield.

4.1 Column Selection and Preparation

Preparative purification utilizes C18 reversed-phase columns with the following specifications:

  • Column dimensions: 250 × 50 mm (small scale) or 250 × 100 mm (production scale)
  • Particle size: 10 μm or 15 μm for preparative applications
  • Pore size: 300 Å to accommodate 3.4 kDa molecular weight
  • Carbon load: 15-20% for optimal hydrophobic interaction
  • Endcapping: complete for reduced silanol interactions
  • pH stability: 2.0-8.0 operational range

Column conditioning requires equilibration with mobile phase for minimum 5 column volumes prior to sample injection. Column performance qualification must demonstrate theoretical plate count exceeding 5000 plates/meter and peak asymmetry factor between 0.9-1.5 for standard peptide markers.

4.2 Mobile Phase Systems

The purification employs binary gradient elution with acidified aqueous and organic phases. Standard mobile phase composition consists of:

Mobile Phase A (Aqueous):

  • Water: HPLC-grade, filtered through 0.22 μm membrane
  • TFA: 0.1% v/v (pH approximately 2.0)
  • Conductivity: <5 μS/cm prior to TFA addition

Mobile Phase B (Organic):

  • Acetonitrile: HPLC-gradient grade, minimum 99.9% purity
  • TFA: 0.1% v/v for ion-pairing consistency
  • Water content: <0.05% verified by Karl Fischer

Mobile phase preparation requires daily preparation to ensure consistency and minimize bacterial growth. Degassing through helium sparging or vacuum degassing prevents bubble formation during gradient elution. Mobile phase storage at 4°C extends usable lifetime to 48 hours maximum.

4.3 Gradient Optimization and Method Parameters

Purification method development establishes gradient conditions that provide baseline resolution between CJC-1295 and closely-related impurities. A typical optimized gradient profile includes:

Time (min) %B Flow Rate (mL/min) Purpose
0-5 20 50 Initial equilibration and sample loading
5-10 20-25 50 Wash highly polar impurities
10-40 25-35 50 Shallow gradient for critical separation
40-50 35-95 50 Steep gradient for hydrophobic impurities
50-55 95 50 Column wash
55-65 95-20 50 Re-equilibration

Detection occurs at 214 nm and 280 nm wavelengths simultaneously. The 214 nm channel monitors peptide bond absorption for quantitation, while 280 nm provides selectivity for aromatic residues. Column temperature control at 25 ± 2°C ensures reproducible retention times and separation efficiency.

4.4 Fraction Collection and Pooling Strategy

Automated fraction collection targets the CJC-1295 peak with defined collection windows based on retention time and UV threshold criteria. Collection parameters must balance purity requirements against yield considerations:

  • Collection threshold: 10-15% of peak maximum on ascending edge
  • Collection termination: 10-15% of peak maximum on descending edge
  • Fraction volume: 50-100 mL per fraction tube
  • Peak tracking: automated retention time adjustment ±0.5 minutes

Individual fractions undergo analytical HPLC testing to determine purity profiles. Fractions meeting the minimum purity specification (typically 95% by area at 214 nm) qualify for pooling. Edge fractions with 85-95% purity may undergo re-purification to maximize overall yield while maintaining final product specifications.

4.5 Desalting and Final Processing

Purified fractions contain TFA and acetonitrile that require removal prior to lyophilization. Desalting options include:

Option 1: Rotary Evaporation

  • Remove acetonitrile under vacuum at 30-35°C
  • Dilute with water and repeat concentration (3 cycles minimum)
  • Adjust pH to 4.0-5.0 with dilute ammonia solution
  • Proceed directly to lyophilization

Option 2: Size-Exclusion Chromatography

  • Load concentrated peptide solution onto Sephadex G-10 or G-25 column
  • Elute with 0.1% acetic acid in water
  • Monitor elution by UV absorbance at 214 nm
  • Collect peptide-containing fractions for lyophilization

Both desalting approaches provide TFA removal exceeding 99% as verified by ion chromatography. The selection between methods depends on scale, equipment availability, and downstream processing requirements as detailed in USP harmonization standards for peptide purification.

5. Lyophilization and Final Product Formulation

Lyophilization (freeze-drying) converts purified CJC-1295 solution into stable solid form suitable for long-term storage and distribution. The lyophilization cycle must remove water while maintaining peptide structural integrity, ensuring consistent reconstitution properties and stability profiles.

5.1 Pre-Lyophilization Formulation

Prior to lyophilization, purified CJC-1295 solution undergoes formulation with appropriate excipients to ensure optimal cake structure and stability. Standard formulation includes:

  • CJC-1295 concentration: 1-5 mg/mL in final solution
  • Mannitol: 2-5% w/v as bulking agent and cryoprotectant
  • Acetic acid: 0.1% to maintain pH 4.0-5.0
  • Glycine: Optional, 1-2% w/v for additional cake stabilization
  • Fill volume: Calculated to achieve target final peptide content per vial

Solution preparation requires sterile-filtered water (0.22 μm filtration) and pharmaceutical-grade excipients. Solution pH verification and adjustment precedes sterile filtration through 0.22 μm PES membranes into pre-sterilized fill vessels. Bioburden control throughout formulation operations maintains counts below 10 CFU/100 mL prior to terminal sterilization or aseptic processing.

5.2 Vial Filling Operations

Aseptic filling procedures in ISO Class 5 (Grade A) environments prevent microbial contamination during vial filling. Process parameters include:

Parameter Specification Quality Assurance
Fill Volume Accuracy ±5% of target volume Gravimetric verification every 10 vials
Vial Type Type I borosilicate glass, 5-10 mL Certificate of analysis from vendor
Stopper Type Fluoropolymer-coated butyl rubber, 20 mm Extractables/leachables testing
Seal Integrity Vacuum hold test: <100 μm Hg loss in 60 seconds 100% inspection post-lyophilization
Particulate Matter Zero visible particles per USP <790> Visual inspection under controlled lighting

5.3 Lyophilization Cycle Parameters

The lyophilization cycle consists of three phases: freezing, primary drying, and secondary drying. Cycle development uses differential scanning calorimetry (DSC) to determine the glass transition temperature (Tg') of the formulation, establishing the maximum product temperature during primary drying. For CJC-1295 with mannitol formulations, Tg' typically occurs at -40 to -35°C.

Phase 1: Freezing

  • Initial shelf temperature: +5°C
  • Ramp rate: -1°C per minute to -45°C
  • Hold at -45°C for minimum 3 hours
  • Ensures complete ice crystallization before primary drying

Phase 2: Primary Drying

  • Chamber pressure: 50-100 mTorr (6.7-13.3 Pa)
  • Shelf temperature: -35 to -30°C (5-10°C below Tg')
  • Duration: 24-48 hours depending on fill volume
  • Endpoint determination: Pirani gauge pressure equals capacitance manometer
  • Product temperature monitoring: Thermocouples in representative vials

Phase 3: Secondary Drying

  • Shelf temperature ramp: +0.2°C per minute to +25°C
  • Chamber pressure: 50-100 mTorr maintained
  • Hold at +25°C for 4-8 hours
  • Final residual moisture target: <3% by Karl Fischer titration

5.4 Stoppering and Crimping

Following completion of secondary drying, vials undergo automated stoppering under vacuum or controlled atmosphere (nitrogen or argon backfill to 750-950 mbar). Stopper insertion occurs within the lyophilizer chamber to maintain sterility and prevent moisture re-absorption. Aluminum crimp seals secure stoppers immediately following removal from the lyophilizer.

Critical quality attributes of the lyophilized cake include:

  • Appearance: Uniform white to off-white cake or powder
  • Cake integrity: No collapse, melt-back, or shrinkage
  • Reconstitution time: Complete dissolution within 60 seconds with gentle swirling
  • Clarity after reconstitution: Clear, colorless solution free of visible particulates
  • Residual moisture: <3% by Karl Fischer, target <2%
  • Oxygen content: <5% in headspace for nitrogen-backfilled vials

Post-lyophilization cake appearance directly correlates with stability performance and requires careful process optimization according to FDA guidance on lyophilization of parenteral products.

6. Quality Control Testing and Release Specifications

Comprehensive analytical testing validates that each CJC-1295 batch meets predetermined quality specifications prior to commercial release. The quality control testing program encompasses identity confirmation, purity assessment, potency determination, and safety testing in accordance with ICH guidelines and pharmacopeial requirements.

6.1 Identity Testing

Multiple orthogonal methods confirm CJC-1295 identity:

Amino Acid Analysis (AAA)

  • Method: Acid hydrolysis (6N HCl, 110°C, 24 hours) followed by HPLC or ion-exchange chromatography
  • Acceptance criteria: Each amino acid within ±15% of theoretical molar ratio
  • Critical residues (Tyr, Ala, Asp, Ser, Gly, Leu, Arg, Gln) must be present
  • Sample size: 50-100 μg peptide per analysis

Mass Spectrometry

  • Method: ESI-MS or MALDI-TOF MS
  • Acceptance criteria: Observed molecular weight within ±0.5 Da of theoretical (3647.3 Da)
  • MS/MS sequencing confirms N-terminal and C-terminal sequences
  • DAC modification verification through characteristic mass shift

Reversed-Phase HPLC

  • Method: Analytical HPLC with gradient elution on C18 column
  • Acceptance criteria: Retention time matches reference standard ±2%
  • Co-injection with reference standard shows single peak
  • UV spectrum (210-300 nm) matches reference standard

6.2 Purity Determination

Multi-dimensional purity assessment employs complementary analytical techniques:

Reversed-Phase HPLC Purity

  • Column: C18, 4.6 × 250 mm, 5 μm particle size, 300 Å pore
  • Mobile phase: Water/acetonitrile with 0.1% TFA gradient
  • Detection: 214 nm (quantitation), 280 nm (aromatic profile)
  • Acceptance criteria: ≥95% main peak by area at 214 nm
  • Individual impurity limit: ≤2.0%
  • Total impurities: ≤5.0%

Ion-Exchange HPLC

  • Orthogonal purity assessment based on charge differences
  • Detects deamidation products (Asn, Gln residues)
  • Column: Strong cation exchange or weak cation exchange
  • Acceptance criteria: ≥90% main peak, complements RP-HPLC data

Size-Exclusion HPLC

  • Detects aggregates, fragments, and high molecular weight impurities
  • Column: Silica-based SEC, appropriate molecular weight range
  • Acceptance criteria: Monomeric form ≥95%, aggregates ≤3%

6.3 Content and Potency Assays

Peptide content determination employs multiple validated methodologies:

Method Principle Specification
Quantitative HPLC External standard calibration at 214 nm 90-110% of label claim
UV Spectroscopy Absorbance at 280 nm using extinction coefficient Supporting data for HPLC results
Amino Acid Analysis Quantitation of hydrolyzed amino acids vs. standards Confirmatory method, 85-115% agreement
Nitrogen Determination Kjeldahl or combustion analysis for peptide nitrogen Alternative quantitation, ±15% of expected

Biological potency assessment through in-vitro bioassays measures growth hormone release activity. While not typically required for reference standard material, bioassay development provides critical data for API characterization and comparability studies during process changes.

6.4 Impurities and Degradation Products

Comprehensive impurity profiling identifies and quantifies process-related and degradation-related impurities:

Process-Related Impurities

  • Deletion sequences (missing amino acid residues)
  • Truncated peptides (incomplete synthesis)
  • Stereoisomers (D-amino acid incorporation, racemization)
  • Incomplete DAC modification (des-DAC variants)
  • Side reactions (aspartimide formation, β-elimination)

Degradation Products

  • Oxidation products (Met, Trp, Tyr residues)
  • Deamidation products (Asn, Gln conversion to Asp, Glu)
  • Hydrolysis products (peptide bond cleavage)
  • Aggregates and oligomers (non-covalent and covalent)

Individual specified impurities exceeding 0.5% require identification and qualification per ICH Q3B guidance on impurities in new drug substances. Mass spectrometry and tandem MS/MS provide structural characterization of unknown impurities above reporting thresholds.

6.5 Physical and Chemical Properties

Additional quality control tests characterize physical and chemical properties:

  • Appearance: White to off-white lyophilized powder
  • Solubility: Freely soluble in water, pH 4-6 solution
  • pH (1% solution): 4.0-6.0
  • Water content: ≤5.0% by Karl Fischer titration
  • Residual solvents: TFA ≤0.5%, acetonitrile ≤410 ppm, DMF ≤880 ppm per ICH Q3C
  • Heavy metals: ≤10 ppm by ICP-MS
  • Bacterial endotoxins: ≤5.0 EU/mg by LAL assay (if intended for injection)
  • Sterility: Passes USP <71> sterility test (if labeled as sterile)
  • Particulate matter: Meets USP <788> requirements after reconstitution

6.6 Stability-Indicating Methods

Analytical methods must demonstrate stability-indicating capability through forced degradation studies. CJC-1295 samples undergo stress conditions including:

  • Acid hydrolysis: 0.1N HCl, 60°C, 24 hours
  • Base hydrolysis: 0.1N NaOH, 60°C, 24 hours
  • Oxidation: 3% hydrogen peroxide, 25°C, 24 hours
  • Thermal stress: 60°C, dry heat, 7 days
  • Photolytic degradation: ICH Option 2 light exposure

Validated analytical methods demonstrate resolution of degradation products from the main peak, with method specificity confirmed through peak purity assessment using photodiode array detection or mass spectrometry. Method validation follows USP <1225> validation of compendial procedures and ICH Q2(R1) guidelines for analytical method validation.

7. Batch Documentation and Manufacturing Records

Comprehensive batch documentation provides complete traceability from raw materials through final product release. The documentation system must comply with FDA 21 CFR Part 211 requirements and support regulatory inspections, customer audits, and internal quality reviews.

7.1 Master Batch Record Structure

The Master Batch Record (MBR) defines all manufacturing operations, in-process controls, and specifications for CJC-1295 production. MBR components include:

  • Product identification and batch size range
  • Complete list of raw materials with specifications and quantities
  • Equipment identification and qualification status
  • Step-by-step manufacturing instructions with process parameters
  • In-process control tests with acceptance criteria
  • Sampling procedures and sampling plans
  • Yield calculations and acceptance ranges (expected yield ± limits)
  • Packaging and labeling instructions
  • Storage conditions and retest/expiration dating

MBR version control requires formal change control procedures for any revisions. Changes undergo quality assurance review and approval prior to implementation, with historical versions archived for regulatory reference.

7.2 Batch Production Record Execution

Each manufacturing batch generates a Batch Production Record (BPR) that documents actual execution of MBR procedures. The BPR captures:

Raw Material Documentation

  • Identity and lot number of each raw material
  • Actual quantities dispensed with verification signatures
  • Certificate of analysis review and approval
  • Retest or expiration date verification
  • Storage condition compliance documentation

Process Execution Records

  • Date and time stamps for each manufacturing operation
  • Equipment identification and calibration status
  • Actual process parameters (temperature, time, pH, volumes)
  • In-process test results with pass/fail determination
  • Deviation documentation and corrective actions
  • Operator identification and qualification status
  • Line clearance verification between operations

7.3 In-Process Control Strategy

Critical process parameters require real-time monitoring and documentation throughout manufacturing:

Manufacturing Stage In-Process Control Frequency Action on OOS Result
Solid-Phase Synthesis Kaiser test after coupling Each cycle Repeat coupling until negative
Solid-Phase Synthesis UV monitoring at 301 nm Each deprotection Extend deprotection time
Cleavage Reaction time verification Continuous Extend within validated range
Purification Fraction purity by HPLC Each fraction Exclude from pool or re-purify
Lyophilization Pressure/temperature monitoring Continuous recording Investigate cycle failure, may reprocess
Fill/Finish Fill weight verification Every 10th vial Adjust fill volume, recheck

7.4 Batch Numbering and Traceability

Unique batch numbering systems provide complete traceability throughout the product lifecycle. Standard batch number formats include manufacturing date codes, product identifiers, and sequential counters. The batch number appears on all documentation, labels, and certificates of analysis.

Traceability systems link:

  • Forward traceability: Raw material lots to intermediate lots to final product batches to customers
  • Backward traceability: Customer complaints or deviations back through manufacturing to raw material sources
  • Electronic batch record systems with database integration for rapid recall capability
  • Raw material vendor qualification and approved supplier lists
  • Equipment usage logs correlated with batch production records

7.5 Yield Calculations and Material Balance

Yield calculations at each manufacturing stage verify process efficiency and detect losses requiring investigation:

  • Crude yield: (Crude peptide weight / theoretical yield from resin loading) × 100
  • Purification yield: (Pure peptide weight / crude peptide weight) × 100
  • Overall yield: (Final product weight / theoretical yield) × 100
  • Acceptance ranges: Typically ±15% of expected historical yields

Material balance accounting reconciles all inputs, outputs, samples, and waste. Significant discrepancies (typically >5% unexplained material loss) require investigation and documentation as deviations with corrective action implementation.

7.6 Deviation and CAPA Management

Any departure from approved procedures requires formal deviation reporting, investigation, and corrective action. The deviation management process includes:

  • Immediate deviation identification and documentation
  • Impact assessment on product quality (critical/major/minor classification)
  • Investigation of root cause using appropriate tools (5-Why, fishbone diagrams)
  • Corrective action plan with implementation timeline
  • Preventive actions to eliminate recurrence
  • Effectiveness verification of implemented actions
  • Batch disposition decision (release, reject, reprocess)

Trending analysis of deviations identifies systemic issues requiring process improvements or revalidation activities. CAPA systems comply with FDA quality systems guidance for pharmaceutical manufacturers.

8. Stability Studies and Shelf-Life Determination

Comprehensive stability programs establish storage conditions, retest dating, and shelf-life specifications for CJC-1295. Stability studies follow ICH Q1A(R2) guidelines for stability testing of new drug substances and provide data supporting commercial storage recommendations.

8.1 Stability Study Design

Formal stability programs encompass multiple study types:

Long-Term Stability Studies

  • Storage condition: -20°C ± 5°C (recommended long-term storage)
  • Alternative condition: 2-8°C (refrigerated storage evaluation)
  • Duration: Minimum 12 months, extending to proposed shelf-life + 3 months
  • Testing frequency: 0, 3, 6, 9, 12, 18, 24, 36 months
  • Sample configuration: Final product in commercial packaging
  • Batch selection: Minimum 3 batches from different manufacturing campaigns

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 shelf-life and identify degradation pathways

Stress Testing

  • Temperature stress: 40°C, 50°C for solid; elevated temperatures for solution
  • Humidity stress: 75% RH for moisture sensitivity assessment
  • Light exposure: ICH Q1B photostability testing
  • Freeze-thaw cycling: -20°C to +25°C, multiple cycles
  • Reconstituted solution stability: Time to use studies

8.2 Stability-Indicating Test Methods

Stability samples undergo comprehensive analytical testing using validated stability-indicating methods:

Test Method Acceptance Criteria at Release Shelf-Life Specification
Appearance Visual inspection White to off-white powder No significant color change
Identification HPLC retention time, MS Matches reference standard Matches reference standard
Purity (HPLC) RP-HPLC at 214 nm ≥95.0% ≥92.0%
Assay Quantitative HPLC 95.0-105.0% 90.0-110.0%
Aggregates SEC-HPLC ≤3.0% ≤5.0%
Water Content Karl Fischer ≤3.0% ≤5.0%
Oxidation Products RP-HPLC peak tracking ≤1.0% individual ≤2.0% individual
Deamidation Products Ion-exchange HPLC ≤2.0% ≤4.0%

8.3 Degradation Pathway Characterization

CJC-1295 exhibits several primary degradation pathways requiring monitoring:

Oxidation

  • Primary site: Methionine residues (if present in formulation)
  • Secondary sites: Tryptophan, tyrosine residues
  • Mechanism: Free radical or metal-catalyzed oxidation
  • Control strategy: Oxygen exclusion, EDTA addition, light protection

Deamidation

  • Sites: Asparagine and glutamine residues
  • Mechanism: pH-dependent hydrolysis or cyclic imide formation
  • Temperature dependence: Accelerated at elevated temperatures
  • Control strategy: pH optimization (4.0-5.0), low temperature storage

Aggregation

  • Types: Covalent (disulfide bonds, cross-linking) and non-covalent
  • Factors: Concentration, temperature, agitation, freeze-thaw
  • Detection: SEC-HPLC, dynamic light scattering, turbidity
  • Control strategy: Bulking agents, minimal agitation, single-use vials

8.4 Shelf-Life Determination

Statistical analysis of stability data establishes retest dates and expiration dating. The analysis employs linear regression of time-zero normalized data for each quality attribute. Shelf-life represents the time point where the 95% confidence interval intersects the acceptance criterion.

Typical CJC-1295 shelf-life specifications:

  • Lyophilized powder at -20°C: 24-36 months
  • Lyophilized powder at 2-8°C: 12-18 months
  • Reconstituted solution at 2-8°C: 7-14 days
  • Room temperature (post-reconstitution): 24 hours maximum

8.5 Container-Closure Qualification

Container-closure systems undergo qualification studies demonstrating product protection throughout shelf-life. Qualification includes:

  • Moisture vapor transmission rate testing at storage conditions
  • Oxygen permeability testing for inert atmosphere packaging
  • Extractables and leachables studies per USP <1663> and <1664>
  • Seal integrity testing including vacuum decay and dye ingress
  • Drop testing and shipping qualification studies
  • Light transmission testing for amber glass or opaque packaging

8.6 Post-Approval Stability Commitment

Ongoing stability programs continue post-approval to monitor commercial batches and detect trends requiring investigation. The commitment protocol specifies:

  • Annual batch testing: Minimum 1 batch per year for first 3 years
  • Long-term storage: Testing through expiration date + 3 months
  • Out-of-specification investigations: Formal investigation and reporting
  • Annual stability reports: Trending analysis and compilation
  • Comparability protocols: Stability requirements for manufacturing changes

9. Storage, Handling, and Distribution Requirements

Proper storage, handling, and distribution practices maintain CJC-1295 quality from manufacturing release through end-user receipt. Temperature control, environmental monitoring, and shipping qualification ensure product stability throughout the distribution chain.

9.1 Storage Conditions and Requirements

CJC-1295 storage specifications depend on physical form and packaging configuration:

Lyophilized Product Storage

  • Temperature: -20°C ± 5°C for long-term storage (preferred)
  • Alternative: 2-8°C for intermediate-term storage (up to 12 months)
  • Humidity: Ambient (product protected by primary packaging)
  • Light: Protect from direct light exposure; store in cartons
  • Orientation: Upright storage prevents stopper contact with product

Bulk Intermediate Storage

  • Purified peptide (pre-lyophilization): -20°C in sealed containers, maximum 30 days
  • Crude peptide: -20°C in sealed containers with desiccant, maximum 180 days
  • Resin-bound peptide: -20°C under nitrogen or argon, maximum 7 days

Reconstituted Solution Storage

  • Temperature: 2-8°C immediately after reconstitution
  • Stability: Maximum 14 days when protected from light
  • Container: Original vial with sterile reconstitution
  • Freezing: Not recommended; may cause aggregation

9.2 Environmental Monitoring Programs

Storage areas require continuous environmental monitoring to verify condition maintenance:

Storage Area Monitoring Parameters Frequency Action Limits
Freezer (-20°C) Temperature Continuous recording, 15-min intervals Alert: -23°C or -17°C; Action: -25°C or -15°C
Refrigerator (2-8°C) Temperature Continuous recording, 15-min intervals Alert: <1°C or >9°C; Action: <0°C or >10°C
Controlled Room Temperature Temperature, RH Continuous recording, 30-min intervals Alert: <18°C or >27°C; Action: <15°C or >30°C
All storage areas Light exposure Annual verification No direct sunlight, <200 lux ambient

Temperature excursion investigations follow formal protocols including:

  • Documentation of excursion duration and magnitude
  • Assessment of impact using stability data and kinetic modeling
  • Product disposition decision (release, quarantine, or reject)
  • Customer notification for distributed material if applicable
  • Root cause analysis and corrective action implementation

9.3 Shipping and Distribution Qualification

Distribution operations maintain cold-chain integrity through qualified shipping containers and procedures. Shipping qualification includes:

Shipping Container Qualification

  • Thermal qualification testing: Summer, winter, and ambient conditions
  • Duration testing: Minimum 2× expected transit time
  • Temperature mapping: Data loggers throughout container volume
  • Acceptance criteria: Maintain 2-8°C (or -20°C) throughout qualification duration
  • Requalification: Annual or after significant design changes

Shipping Procedure Components

  • Product pre-conditioning: Verify proper storage temperature before shipment
  • Cold packs or dry ice: Sufficient quantity for transit duration plus contingency
  • Temperature monitoring devices: Data loggers with each shipment or representative validation
  • Packaging materials: Insulated containers with validated thermal performance
  • Package orientation: Clear labeling preventing inversion
  • Transit time limits: Maximum allowable transit time before requalification required

9.4 Handling Procedures and Training

Personnel handling CJC-1295 require training on proper procedures to prevent product degradation:

  • Minimize exposure to room temperature: Return to storage within 15 minutes
  • Avoid repeated freeze-thaw cycles: Aliquot bulk solutions if multiple uses planned
  • Use clean, dry equipment: Contamination and moisture compromise stability
  • Wear appropriate PPE: Gloves to prevent product contamination and personal exposure
  • Gentle mixing only: Avoid vigorous shaking that causes foaming/aggregation
  • Sterile technique: When reconstituting for in-vivo use
  • Immediate return to storage: After sample removal or reconstitution

Standard operating procedures document all handling steps with photographic illustrations and include competency assessment requirements for personnel authorization.

9.5 Inventory Management and Stock Rotation

Inventory control systems prevent use of expired or compromised material:

  • Electronic inventory tracking: Real-time inventory with automatic expiry alerts
  • FEFO (First Expired, First Out) system: Automated selection of shortest-dated material
  • Segregation protocols: Physical separation of released, quarantined, and rejected material
  • Expiry date verification: Check at receipt, storage, and issuance
  • Periodic inventory audits: Monthly physical verification against electronic records
  • Destruction procedures: Documented disposal of expired or rejected material

9.6 Customer Receiving and Acceptance

Customer receiving procedures verify product integrity upon receipt:

  • Immediate inspection: Check package condition upon delivery
  • Temperature verification: Review data logger if included, verify cold pack status
  • Product inspection: Examine vials for damage, discoloration, or reconstitution
  • Documentation review: Verify Certificate of Analysis matches product received
  • Prompt storage: Transfer to appropriate storage within 30 minutes of receipt
  • Damage reporting: Contact supplier immediately if temperature excursion or damage noted

Distributors and end-users should maintain receiving logs documenting condition at delivery as part of quality systems per FDA Drug Supply Chain Security Act requirements.

10. Certificate of Analysis and Release Documentation

The Certificate of Analysis (CoA) provides comprehensive quality documentation supporting each CJC-1295 batch release. This critical document certifies product conformance to specifications and enables customer quality assessment and regulatory compliance verification.

10.1 Certificate of Analysis Components

A complete CJC-1295 Certificate of Analysis includes the following sections:

Header Information

  • Manufacturer name, address, and contact information
  • Product name: CJC-1295 (with DAC) or specified trade name
  • Catalog or product code number
  • Batch or lot number with full traceability
  • Manufacturing date and expiration/retest date
  • CoA issue date and document version number
  • Storage conditions and handling recommendations

Physical and Chemical Characteristics

  • Appearance description (visual assessment result)
  • Molecular formula: C165H269N47O46
  • Molecular weight: 3647.3 Da (theoretical, average mass)
  • CAS number: 863288-34-0
  • Sequence: Tyr-D-Ala-Asp-Ala-Ile-Phe-Thr-Gln-Ser-Tyr-Arg-Lys(DAC)-Val-Leu-Ala-Gln-Leu-Ser-Ala-Arg-Lys-Leu-Leu-Gln-Asp-Ile-Leu-Ser-Arg-NH2

10.2 Analytical Test Results Table

Test results appear in tabular format with specification ranges and actual results:

Test Method Specification Result
Appearance Visual White to off-white powder White lyophilized powder
Identity (HPLC) RP-HPLC RT matches standard ±2% Conforms (RT: 24.3 min)
Identity (MS) ESI-MS MW: 3647.3 ±0.5 Da 3647.4 Da
Purity (HPLC) RP-HPLC at 214 nm ≥95.0% 97.2%
Assay (Content) Quantitative HPLC 95.0-105.0% 98.7%
Water Content Karl Fischer ≤5.0% 2.1%
Acetonitrile GC ≤410 ppm <50 ppm
TFA Ion chromatography ≤0.5% 0.08%
Heavy Metals ICP-MS ≤10 ppm <2 ppm
Bacterial Endotoxins LAL ≤5.0 EU/mg <0.5 EU/mg
Sterility USP <71> Passes No growth detected

10.3 Chromatogram and Spectral Data

Representative analytical data appendices strengthen CoA documentation:

  • HPLC chromatogram: Annotated trace showing main peak and identified impurities
  • Mass spectrum: ESI-MS or MALDI-TOF spectrum with molecular ion labeled
  • Amino acid analysis: Tabular results showing theoretical vs. observed ratios
  • SEC chromatogram: Size-exclusion profile demonstrating monomeric state

Electronic CoAs may include embedded chromatograms, while printed versions reference archived raw data files with unique identifiers.

10.4 Storage and Handling Instructions

The CoA specifies proper storage and handling to maintain stated quality:

  • Storage temperature: Store at -20°C upon receipt
  • Protect from light: Keep in original packaging until use
  • Avoid repeated freeze-thaw: Aliquot upon first opening if multiple uses planned
  • Reconstitution instructions: Dissolve in sterile water or specified buffer
  • Reconstituted stability: Use within specified timeframe (typically 7-14 days at 2-8°C)
  • Shipping conditions: Product shipped on dry ice or cold packs as appropriate

10.5 Quality Assurance Certification

The CoA footer includes quality assurance certification:

  • Statement of compliance: "This batch has been manufactured and tested in accordance with current Good Manufacturing Practices and meets all established specifications."
  • QA approval signature: Authorized quality assurance personnel signature
  • Approval date: Date of final batch release
  • Regulatory status disclaimer: "For research use only" or "Pharmaceutical grade" as applicable
  • Technical support contact: Phone and email for questions

10.6 Supplementary Documentation

Additional documentation available upon request supports customer quality systems:

  • Material Safety Data Sheet (MSDS/SDS): Safety information per GHS requirements
  • Regulatory support file: DMF reference, CEP, or other regulatory documentation
  • Stability summary: Summary of stability data supporting expiration dating
  • Amino acid sequence certificate: Detailed sequence confirmation data
  • Residual solvents report: Complete ICH Q3C solvent panel results
  • Heavy metals report: Detailed ICP-MS results for individual elements
  • Manufacturing flow diagram: Simplified process overview
  • Reference standard comparison: Data demonstrating equivalence to reference material

10.7 Electronic Certificate Systems

Modern quality systems employ electronic CoA generation and distribution:

  • LIMS integration: Automated CoA generation from laboratory information systems
  • Electronic signatures: 21 CFR Part 11 compliant approval workflows
  • PDF/A format: Long-term archival format with embedded metadata
  • QR code inclusion: Links to verification portal and raw data access
  • Audit trails: Complete traceability of CoA generation and modifications
  • Customer portal access: Self-service CoA retrieval by batch number
  • Version control: Automated versioning for reissued or corrected CoAs

10.8 Certificate of Analysis Review and Approval

Before CoA issuance, quality assurance performs comprehensive review:

  • Verification of all test results against specifications
  • Confirmation of testing completion by qualified personnel
  • Review of any deviations, investigations, or retests
  • Verification of calculations and data transcription accuracy
  • Confirmation of expiration date calculation
  • Approval authority verification and electronic signature application
  • Final document format and accuracy check before distribution

CoA approval represents the final quality gate before product release, confirming compliance with all applicable regulations including FDA cGMP requirements and customer specifications.

Conclusion: Manufacturing Excellence for CJC-1295 Production

CJC-1295 manufacturing demands rigorous process control, comprehensive analytical testing, and systematic quality assurance throughout the production lifecycle. The integration of validated solid-phase synthesis protocols, orthogonal purification strategies, and stability-indicating analytical methods ensures consistent delivery of pharmaceutical-grade peptide meeting or exceeding industry specifications.

Manufacturing facilities implementing these technical specifications achieve several critical quality outcomes: batch-to-batch consistency through validated process parameters, impurity profiles consistently below regulatory thresholds, extended shelf-life through optimized formulation and storage conditions, and complete traceability supporting regulatory compliance and customer confidence.

Success in CJC-1295 production requires cross-functional expertise spanning synthetic chemistry, analytical method development, quality systems, and regulatory compliance. Organizations must invest in specialized equipment including automated peptide synthesizers, preparative HPLC systems, lyophilizers, and comprehensive analytical instrumentation including mass spectrometry and multi-dimensional chromatography platforms.

Personnel qualifications represent an equally critical investment. Manufacturing and quality control staff require specialized training in peptide chemistry, cGMP regulations, and statistical process control methodologies. Regular competency assessment, continuing education, and participation in industry working groups ensure teams remain current with evolving best practices and regulatory expectations.

The manufacturing profile presented herein provides the technical foundation for CJC-1295 production operations. Facilities should customize these specifications based on their specific equipment capabilities, regulatory requirements, and customer needs while maintaining alignment with fundamental quality principles. Regular process review, continuous improvement initiatives, and proactive adoption of technological advances in peptide manufacturing will drive ongoing optimization and competitive advantage.

For additional technical resources on peptide manufacturing, consult complementary profiles available through PeptideForge.com covering related topics including ipamorelin synthesis optimization, advanced purification methodologies, analytical method validation protocols, lyophilization cycle development, and comprehensive stability testing programs.

Manufacturing excellence in CJC-1295 production ultimately serves the broader objective of providing researchers, clinicians, and pharmaceutical developers with reliable, high-quality peptide tools for advancing scientific understanding and therapeutic development. The technical rigor detailed throughout this manufacturing profile reflects the industry's commitment to quality, safety, and regulatory compliance as foundational principles guiding modern peptide production operations.

References and Regulatory Resources

  1. 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
  2. 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
  3. 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
  4. United States Pharmacopeia. (2024). USP Harmonization Standards for Peptide Manufacturing. USP-NF General Chapters. Available at: https://www.usp.org/harmonization-standards/pdg
  5. International Council for Harmonisation (ICH). (2023). ICH Q3B(R2): Impurities in New Drug Substances. Available at: https://www.ich.org/page/quality-guidelines
  6. United States Pharmacopeia. (2024). USP <1225> Validation of Compendial Procedures. Available at: https://www.usp.org/validation
  7. United States Pharmacopeia. (2024). USP <1663> Assessment of Extractables Associated with Pharmaceutical Packaging/Delivery Systems and USP <1664> Assessment of Drug Product Leachables Associated with Pharmaceutical Packaging/Delivery Systems. Available at: https://www.usp.org/chemical-medicines/general-chapters
  8. 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
  9. 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
  10. 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