DSIP Manufacturing Profile: Technical Specifications for Peptide Manufacturers | PeptideForge

DSIP (Delta Sleep-Inducing Peptide) Manufacturing Profile: Comprehensive Technical Specifications and Quality Control Standards

1. Introduction to DSIP Manufacturing

Delta Sleep-Inducing Peptide (DSIP) represents a nonapeptide with the amino acid sequence Trp-Ala-Gly-Gly-Asp-Ala-Ser-Gly-Glu. First isolated from the cerebral venous blood of rabbits in a state of sleep, DSIP has since become a compound of significant interest in pharmaceutical manufacturing and research applications. For manufacturers and quality control professionals, understanding the precise synthesis methodologies, purification protocols, and analytical specifications is critical to delivering pharmaceutical-grade material that meets or exceeds industry standards.

This manufacturing profile provides comprehensive technical documentation for DSIP production, encompassing solid-phase peptide synthesis (SPPS) protocols, advanced purification strategies, rigorous quality control procedures, batch specification requirements, stability parameters, storage protocols, and Certificate of Analysis (CoA) standards. Manufacturing facilities producing DSIP must adhere to Good Manufacturing Practice (GMP) guidelines to ensure batch-to-batch consistency, structural integrity, and regulatory compliance.

The molecular formula of DSIP is C35H48N10O15, with a molecular weight of 848.81 g/mol. The peptide's relatively short sequence and lack of disulfide bonds simplify manufacturing compared to more complex therapeutic peptides, yet stringent process control remains essential to achieve the purity levels demanded by pharmaceutical applications. Modern manufacturing protocols consistently achieve purity levels exceeding 98.5%, with many batches surpassing 99% purity as verified by reverse-phase high-performance liquid chromatography (RP-HPLC) and mass spectrometry (MS) analysis.

2. Solid-Phase Peptide Synthesis (SPPS) Methodology

2.1 Fmoc Strategy Overview

DSIP is synthesized using Fmoc (9-fluorenylmethoxycarbonyl) solid-phase peptide synthesis, which has become the industry standard for peptide manufacturing due to its orthogonal protection scheme, mild deprotection conditions, and compatibility with automation platforms. The Fmoc/tBu protection strategy allows for base-labile temporary N-terminal protection while maintaining acid-labile side-chain protection, facilitating efficient sequential amino acid coupling without premature deprotection of functional groups.

The synthesis proceeds on a solid support resin, typically a polystyrene-based Wang resin or Rink amide resin pre-loaded with the C-terminal amino acid (glutamic acid for DSIP). The choice of resin depends on whether the C-terminus is required as a free carboxylic acid or amide. For DSIP production, Wang resin is commonly employed to yield the free acid form upon cleavage. Resin substitution levels typically range from 0.4 to 0.7 mmol/g to optimize coupling efficiency and minimize steric hindrance during chain assembly.

2.2 Synthesis Protocol Parameters

The synthesis protocol follows a systematic cycle of deprotection, coupling, and washing steps for each amino acid addition. Prior to synthesis initiation, the resin must be swelled for a minimum of 30 minutes in dichloromethane (DCM) or N,N-dimethylformamide (DMF) to expand the polymer matrix and ensure accessibility of reactive sites. Proper swelling is critical for achieving uniform coupling across the resin bed.

Table 1: DSIP SPPS Synthesis Parameters
Process Step Reagent/Condition Concentration/Ratio Duration Temperature
Resin Swelling DCM or DMF 10-15 mL/g resin 30-60 min 20-25°C
Fmoc Deprotection Piperidine in DMF 20% (v/v) 2 × 5 min 20-25°C
Resin Washing DMF 10 mL/g resin 3-5 cycles 20-25°C
Amino Acid Coupling Fmoc-AA-OH 3-5 eq excess 15-60 min 20-25°C
Coupling Agent HATU/HOAt or HBTU/HOBt 3-5 eq Co-administered 20-25°C
Base Activator DIEA or NMM 6-10 eq Co-administered 20-25°C
Final Cleavage TFA/TIS/H₂O 95:2.5:2.5 (v/v/v) 2-4 hours 20-25°C

Fmoc deprotection is accomplished using 20% piperidine in DMF, applied in two treatments of 5 minutes each to ensure complete removal of the protecting group. The deprotection reaction releases the fluorenyl-piperidine adduct, which exhibits strong UV absorption at 301 nm, providing a convenient spectrophotometric method for monitoring deprotection efficiency and calculating coupling yields throughout the synthesis. Automated synthesizers continuously monitor this UV signal to verify successful deprotection before proceeding to the coupling step.

2.3 Coupling Reagents and Activation Chemistry

Amino acid coupling employs contemporary coupling reagents such as HATU (1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate) in combination with HOAt (1-hydroxy-7-azabenzotriazole), or alternatively HBTU (O-benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluoro-phosphate) with HOBt (1-hydroxybenzotriazole). These reagents activate the carboxyl group of incoming Fmoc-protected amino acids, forming highly reactive esters that undergo rapid aminolysis with the free N-terminal amine on the growing peptide chain.

The coupling reaction is facilitated by a tertiary amine base, typically N,N-diisopropylethylamine (DIEA) or N-methylmorpholine (NMM), which neutralizes the trifluoroacetate counterion and maintains the nucleophilicity of the resin-bound amine. Standard coupling protocols utilize 3-5 molar equivalents of Fmoc-amino acid, coupling reagent, and base relative to the initial resin substitution level. For difficult couplings, particularly at sterically hindered positions such as sequential glycine residues in DSIP, extended coupling times (up to 2 hours) or double coupling procedures may be implemented to drive reactions to completion.

2.4 Quality Monitoring During Synthesis

In-process quality monitoring is essential to identify coupling failures or incomplete deprotection events before synthesis completion. The Kaiser test (ninhydrin test) or chloranil test can be employed to detect free primary amines on the resin, indicating successful deprotection and incomplete coupling respectively. A negative Kaiser test after coupling confirms successful acylation of the N-terminus. For automated synthesis platforms, quantitative UV monitoring of Fmoc deprotection provides real-time feedback on synthesis progression and allows calculation of stepwise coupling efficiencies.

Manufacturing facilities should maintain detailed batch synthesis records documenting all reagent lot numbers, coupling times, deprotection UV absorbance values, and any procedural deviations. This documentation forms an integral component of GMP compliance and enables comprehensive investigation of any quality issues that may arise during downstream purification or analytical testing. For a typical peptide synthesis workflow, synthesis efficiency directly impacts crude purity and overall manufacturing yield.

3. Cleavage and Crude Peptide Recovery

3.1 Cleavage Cocktail Composition

Upon completion of the synthesis sequence, the protected peptide-resin must undergo simultaneous cleavage from the solid support and removal of acid-labile side-chain protecting groups. For DSIP, which contains aspartic acid, serine, and glutamic acid residues with tert-butyl (tBu) ester or tert-butyl ether protection, a trifluoroacetic acid (TFA)-based cleavage cocktail is employed.

The standard cleavage cocktail consists of 95% TFA with scavengers to prevent alkylation of nucleophilic amino acid side chains by carbocations generated during protecting group removal. A typical formulation includes TFA/triisopropylsilane (TIS)/water in a ratio of 95:2.5:2.5 (v/v/v). TIS serves as a carbocation scavenger, while water scavenges tert-butyl cations specifically. For peptides containing tryptophan (present in DSIP), this scavenger combination is generally sufficient, though some protocols incorporate additional thioanisole or ethanedithiol for enhanced protection against oxidation and alkylation.

3.2 Cleavage Procedure and Precipitation

The cleavage reaction proceeds for 2-4 hours at room temperature with periodic agitation to ensure thorough mixing of the resin bed with the cleavage cocktail. The volume of cleavage cocktail should be sufficient to completely immerse the resin, typically 10-20 mL per gram of peptide-resin. Following cleavage, the resin is removed by filtration, and the filtrate containing dissolved crude peptide is concentrated under nitrogen stream to reduce the volume of TFA.

Crude peptide precipitation is achieved by addition of cold diethyl ether or methyl tert-butyl ether (MTBE) to the concentrated TFA solution, typically in a 10-fold excess volume. The peptide precipitates as a white to off-white solid, which is collected by centrifugation or filtration. The pellet is washed with additional cold ether (2-3 times) to remove residual TFA, scavengers, and cleaved protecting groups. After the final wash, the crude peptide is dried under vacuum or nitrogen stream, yielding a powder that can be dissolved for purification or stored under desiccated conditions.

3.3 Crude Purity Assessment

Prior to purification, analytical HPLC analysis of the crude peptide provides critical information regarding synthesis success and guides purification strategy selection. Crude DSIP typically exhibits purity ranging from 40-70% depending on synthesis scale, coupling efficiency, and sequence-dependent factors. The major impurities consist of deletion sequences (lacking one or more amino acids due to incomplete couplings) and truncation products. Modern SPPS optimization techniques can improve crude purity, reducing purification burden and enhancing overall process economics.

4. Preparative HPLC Purification

4.1 Reverse-Phase HPLC Methodology

Purification of crude DSIP to pharmaceutical-grade specifications requires preparative reverse-phase high-performance liquid chromatography (RP-HPLC), which separates peptides based on hydrophobicity differences between the target peptide and synthesis-related impurities. RP-HPLC utilizes a hydrophobic stationary phase, typically C18-derivatized silica with particle sizes ranging from 5-10 μm for preparative applications, packed into stainless steel columns with dimensions appropriate for the scale of purification.

The mobile phase consists of two solvents: an aqueous phase (Solvent A) and an organic phase (Solvent B), each containing a volatile acid modifier. The standard mobile phase composition employs water with 0.1% TFA as Solvent A and acetonitrile (ACN) with 0.1% TFA as Solvent B. TFA serves multiple critical functions: it suppresses ionization of silanol groups on the stationary phase (reducing peak tailing), ion-pairs with basic amino acid residues (enhancing retention and peak shape), and provides a volatile acid that can be readily removed during lyophilization.

4.2 Gradient Development and Optimization

Peptide separation is achieved by applying a gradient of increasing organic solvent concentration over time. For DSIP purification, a shallow gradient in the range of 20-40% acetonitrile typically provides optimal resolution between the target peptide and closely-related impurities. The gradient slope must be carefully optimized to balance resolution (separation quality) against run time and throughput. Typical preparative gradients employ 0.5-2% ACN increase per column volume.

Table 2: Preparative RP-HPLC Parameters for DSIP Purification
Parameter Specification Notes
Column Type C18 reverse-phase, 5-10 μm particle size Preparative dimensions: 21.2-50 mm ID
Column Length 150-250 mm Longer columns improve resolution
Mobile Phase A H₂O + 0.1% TFA HPLC-grade water, filtered 0.22 μm
Mobile Phase B Acetonitrile + 0.1% TFA HPLC-grade ACN, filtered 0.22 μm
Gradient Range 15-45% B over 60-90 min Optimized for DSIP retention
Flow Rate 10-40 mL/min Scale-dependent; maintain linear velocity
Detection Wavelength 220 nm (primary), 280 nm (secondary) 220 nm detects peptide bonds
Injection Volume 5-20 mL Concentration: 10-50 mg/mL
Column Temperature 20-30°C Temperature control improves reproducibility

UV detection at 220 nm is employed for peak monitoring, as peptide bonds exhibit maximum absorption at this wavelength. The characteristic UV absorption enables sensitive detection of peptide elution, typically with detection limits below 0.1 mg/mL. For DSIP, which contains a tryptophan residue, secondary monitoring at 280 nm provides confirmatory identification based on the aromatic side-chain absorption, though sensitivity at this wavelength is lower than at 220 nm.

4.3 Fraction Collection and Pooling Strategy

As the peptide elutes from the column, fractions are collected based on UV detector signal threshold and retention time windows established during method development. Automated fraction collectors can be programmed to collect fractions when UV absorbance exceeds a predefined threshold, typically set to capture the main peak while excluding early-eluting and late-eluting impurities. For maximum purity, only the center-cut portions of the main peak should be collected, sacrificing some yield to ensure pharmaceutical-grade purity specifications are met.

Each collected fraction undergoes analytical HPLC analysis to verify purity before pooling. Only fractions meeting the target purity specification (typically ≥98% by RP-HPLC) are combined for lyophilization. This analytical screening prevents contamination of high-purity material with borderline fractions that could compromise final product quality. Fractions of intermediate purity (95-98%) may be re-purified in subsequent preparative runs to recover additional product, improving overall manufacturing yield and process economics.

4.4 Lyophilization Process

Pooled fractions containing purified DSIP in aqueous acetonitrile solution must be converted to a stable solid form suitable for storage and distribution. Lyophilization (freeze-drying) removes both water and volatile organic solvents, yielding a fluffy white powder with extended shelf-life stability. Prior to lyophilization, the TFA counterion content should be considered, as residual TFA can constitute 10-20% of the lyophilized mass and affect peptide stability and reconstitution properties.

The lyophilization process consists of three main phases: freezing, primary drying (sublimation of ice), and secondary drying (desorption of bound water). For peptide solutions, rapid freezing using liquid nitrogen or pre-cooled shelves (-40 to -50°C) creates a homogeneous frozen matrix that facilitates efficient sublimation. Primary drying is conducted under vacuum (50-200 mTorr) with shelf temperatures gradually ramped from -40°C to 0°C over 24-48 hours. Secondary drying at 20-25°C under high vacuum (<50 mTorr) for 6-12 hours removes residual moisture to final levels below 3% by Karl Fischer analysis.

The resulting lyophilized DSIP should appear as a white to off-white powder or cake with uniform appearance. Product vials are sealed under nitrogen or argon atmosphere to minimize oxidation during storage. For comprehensive guidance on peptide purification best practices, manufacturers should consult established industry protocols and validation studies.

5. Quality Control and Analytical Testing

5.1 Identity Confirmation

Verification of peptide identity constitutes a fundamental quality control requirement, ensuring that the synthesized product corresponds to the intended DSIP sequence. Mass spectrometry (MS) provides definitive identity confirmation through accurate measurement of the molecular mass. Electrospray ionization mass spectrometry (ESI-MS) or matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) can determine the molecular weight of DSIP with accuracy better than 0.01% (within 0.1 Da of theoretical mass).

For DSIP with molecular formula C35H48N10O15, the theoretical monoisotopic mass is 848.33 Da. High-resolution mass spectrometry should detect the protonated molecular ion [M+H]+ at m/z 849.34 or sodium adduct [M+Na]+ at m/z 871.32. The presence of the expected molecular ion with isotopic distribution matching theoretical predictions confirms correct amino acid sequence and absence of unexpected modifications. Tandem mass spectrometry (MS/MS) with peptide fragmentation can provide additional sequence confirmation through analysis of fragment ions, though this is typically not required for routine batch release if synthesis documentation is comprehensive.

5.2 Purity Determination by RP-HPLC

Purity quantification represents the most critical quality attribute for pharmaceutical-grade peptides. Analytical RP-HPLC using validated methods provides the industry-standard approach for peptide purity determination. The analytical method should employ gradient conditions capable of resolving DSIP from synthesis-related impurities, deletion sequences, and degradation products. Typical analytical methods utilize C18 columns with 3-5 μm particles, 4.6 mm internal diameter, and 150-250 mm length.

The mobile phase composition mirrors preparative conditions (water + 0.1% TFA / acetonitrile + 0.1% TFA), but the gradient is optimized for analytical resolution rather than preparative capacity. A representative analytical gradient progresses from 10% to 60% acetonitrile over 30-40 minutes at a flow rate of 1.0 mL/min. UV detection at 220 nm provides sensitive monitoring of all peptide species.

Table 3: Analytical HPLC Quality Control Specifications
Parameter Specification Acceptance Criteria
Purity (Area %) ≥98.5% by RP-HPLC Main peak ≥98.5%, single impurity <0.5%
Retention Time System suitability established RSD <2.0% for replicate injections
Peak Asymmetry Tailing factor <2.0 Symmetric peak shape indicates quality
Resolution Rs >2.0 from nearest impurity Baseline separation required
System Precision RSD <2.0% for area counts n=5 replicate injections of standard

Purity calculation follows the area normalization method, where the integrated area of the main DSIP peak is divided by the sum of all detected peak areas and multiplied by 100 to yield percentage purity. Manufacturing specifications typically require minimum purity of 98.5%, with many suppliers routinely achieving >99% purity. Individual impurity peaks should not exceed 0.5%, and total impurities should remain below 1.5%. These stringent specifications ensure pharmaceutical-grade quality suitable for research and potential therapeutic applications.

5.3 Amino Acid Analysis

Amino acid analysis (AAA) provides quantitative verification of peptide composition and serves as an orthogonal purity assessment method. The peptide is hydrolyzed under acidic conditions (6 M HCl, 110°C, 24 hours) to cleave all peptide bonds, releasing free amino acids that are subsequently quantified by ion-exchange chromatography or reverse-phase HPLC with pre- or post-column derivatization.

For DSIP, amino acid analysis should detect one residue each of tryptophan, aspartic acid, serine, and glutamic acid, two residues of alanine, and three residues of glycine. The molar ratios should conform to expected values within ±10%. AAA also enables determination of peptide content, expressed as percentage of theoretical peptide mass corrected for water content, residual TFA counterion, and other non-peptide components. Peptide content typically ranges from 70-85% for TFA salts, with the remainder consisting primarily of TFA and water.

5.4 Water Content Determination

Residual water content in lyophilized peptides affects product stability, mass calculations, and reconstitution behavior. Karl Fischer titration provides accurate quantification of water content in solid peptide samples. Pharmaceutical specifications typically require water content below 3-5% (w/w) for lyophilized peptides to ensure long-term stability and prevent hydrolytic degradation. Excessive moisture promotes peptide bond hydrolysis, deamidation of asparagine and glutamine residues, and oxidation of susceptible amino acids.

5.5 Endotoxin Testing

For peptides intended for in vivo research applications or pharmaceutical development, endotoxin testing is mandatory to ensure absence of bacterial lipopolysaccharide (LPS) contamination. The Limulus Amebocyte Lysate (LAL) assay or recombinant Factor C assay quantifies endotoxin levels, with specifications typically requiring <1.0 EU/mg for research-grade peptides or <0.5 EU/mg for GMP materials. Endotoxin contamination can arise from bacterial contamination during synthesis, purification, or lyophilization, making proper aseptic technique and equipment sanitization critical control points.

5.6 Bioburden and Sterility

Microbial contamination assessment involves bioburden determination (total aerobic microbial count) and, for sterile products, formal sterility testing according to USP <71> or EP 2.6.1. Non-sterile research peptides should demonstrate total aerobic count <10 CFU/g and absence of specified objectionable organisms. Peptides manufactured for clinical applications must be produced under aseptic conditions and demonstrate sterility through 14-day incubation testing in fluid thioglycollate medium and soybean-casein digest medium.

To understand the broader context of pharmaceutical peptide manufacturing standards, quality control professionals should reference ICH guidelines and pharmacopeial monographs that establish comprehensive testing requirements for peptide APIs.

6. Batch Specifications and Documentation

6.1 Release Specifications

Each manufactured batch of DSIP must meet comprehensive release specifications before distribution to customers or advancement to subsequent manufacturing stages. Release specifications are established based on method validation studies, stability data, and regulatory guidelines, ensuring that material consistently meets quality standards throughout its shelf life. Manufacturing facilities operating under GMP must maintain validated analytical methods and documented procedures for all specification tests.

Table 4: DSIP Batch Release Specifications
Test Parameter Method Specification
Appearance Visual Inspection White to off-white powder
Identity ESI-MS or MALDI-TOF MS 848.33 ± 0.5 Da (monoisotopic mass)
Purity RP-HPLC (220 nm) ≥98.5% (area normalization)
Single Impurity RP-HPLC (220 nm) ≤0.5% each impurity
Total Impurities RP-HPLC (220 nm) ≤1.5%
Peptide Content Amino Acid Analysis 70-85% (corrected for TFA and H₂O)
Water Content Karl Fischer Titration ≤5.0%
Amino Acid Composition AAA (hydrolysis/derivatization) Conforms to sequence ±10%
Endotoxin LAL or rFC Assay <1.0 EU/mg (research); <0.5 EU/mg (GMP)
Bioburden Total Aerobic Count <10 CFU/g
Counter Ion Ion Chromatography or NMR TFA (typical 10-20% by mass)

6.2 Batch Record Documentation

Comprehensive batch production records document all manufacturing activities from synthesis initiation through final packaging. GMP-compliant batch records include synthesis parameters (resin lot, amino acid lots, coupling reagent lots, reaction times, deprotection UV values), purification data (HPLC chromatograms, fraction collection windows, pooling decisions), lyophilization cycle parameters (temperature profiles, vacuum levels, duration), analytical testing results (all QC test data with analyst signatures), and deviation investigations (documentation of any non-conformances and corrective actions).

All batch documentation must be retained for a minimum of five years beyond product expiration date, or longer as required by applicable regulatory jurisdictions. Electronic batch records should be maintained in validated, audit-trail-enabled systems that prevent unauthorized modification and ensure data integrity. For facilities producing peptides under contract manufacturing agreements, batch records may be provided to clients in redacted format to protect proprietary process information while demonstrating compliance with quality standards.

6.3 Lot Numbering and Traceability

Each DSIP batch receives a unique lot number that enables complete traceability from final product back through all raw materials, intermediates, and process steps. Lot numbering systems should incorporate manufacturing date codes and sequential batch identifiers to facilitate inventory management and investigation of quality issues. The lot number appears on all product labels, Certificates of Analysis, and shipping documentation, enabling customers to trace material provenance and access batch-specific quality data.

Raw material traceability is particularly critical for amino acids, resins, and solvents, as variations in starting material quality can impact synthesis efficiency and product purity. Manufacturers should maintain comprehensive supplier qualification programs and implement incoming raw material testing protocols to verify identity, purity, and conformance to specifications before use in production. For insights into GMP peptide manufacturing workflows, quality professionals should review industry case studies demonstrating best practices in batch documentation and traceability systems.

7. Stability Studies and Degradation Pathways

7.1 Degradation Mechanisms

Understanding peptide degradation pathways enables rational design of formulation and storage strategies to maximize product stability. DSIP is susceptible to several degradation mechanisms common to peptides: hydrolytic cleavage of peptide bonds, particularly adjacent to aspartic acid residues; deamidation of asparagine residues (though DSIP contains no Asn); oxidation of tryptophan, methionine, and cysteine residues (tryptophan present in DSIP); and aggregation through intermolecular interactions, especially at higher concentrations.

The aspartic acid residues in DSIP at positions 5 and the glutamic acid at position 9 represent potential sites for acid-catalyzed peptide bond hydrolysis, particularly under acidic conditions or elevated temperatures. Tryptophan at position 1 is prone to oxidation, forming various oxidation products including N-formylkynurenine, kynurenine, and hydroxytryptophan derivatives. These oxidative modifications can be promoted by exposure to light, oxygen, peroxides, and transition metal contaminants.

7.2 Lyophilized Peptide Stability

Properly lyophilized and stored DSIP demonstrates excellent long-term stability. Stability studies conducted under ICH-compliant conditions provide data supporting labeled shelf-life claims. Real-time stability testing at recommended storage conditions (-20°C or colder) has demonstrated that DSIP maintains >98% purity for at least 36 months when stored in sealed containers protected from light and moisture.

Table 5: DSIP Stability Data Summary
Storage Condition Time Point Purity (HPLC) Main Degradation Products
-20°C, desiccated, dark 0 months 99.2% N/A (T0)
-20°C, desiccated, dark 12 months 99.0% Trace oxidation products
-20°C, desiccated, dark 24 months 98.8% Oxidation products <0.4%
-20°C, desiccated, dark 36 months 98.6% Oxidation products <0.6%
4°C, desiccated, dark 6 months 98.2% Oxidation products <0.8%
Room temp, ambient 3 weeks 97.5% Oxidation, deamidation
Room temp, ambient 8 weeks 95.8% Multiple degradation products

Accelerated stability studies at elevated temperatures (40°C, 60% relative humidity) provide predictive data on degradation kinetics and identify primary degradation pathways. Under accelerated conditions, DSIP shows increased formation of oxidized tryptophan derivatives and peptide hydrolysis products. These studies validate the necessity of cold storage and moisture protection for maintaining long-term product quality.

7.3 Solution-State Stability

Reconstituted DSIP solutions exhibit significantly reduced stability compared to lyophilized powder, requiring careful attention to storage conditions and use timelines. Following reconstitution in sterile water, bacteriostatic water, or phosphate-buffered saline (PBS), DSIP solutions should be stored at 2-8°C and used within 7 days to maintain purity above specification limits. For extended storage of reconstituted peptide, aliquots should be prepared and stored frozen at -20°C or preferably -80°C.

Freeze-thaw cycles should be minimized, as repeated freezing and thawing can promote aggregation and loss of potency. The addition of carrier proteins such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) can enhance solution stability and prevent adsorptive losses to container surfaces, particularly at low peptide concentrations (<1 mg/mL). However, protein additives may interfere with certain analytical methods and downstream applications, requiring case-by-case evaluation.

pH significantly impacts solution stability. DSIP contains acidic residues (Asp, Glu) and is typically most stable at slightly acidic to neutral pH (5.0-7.0). Extreme pH values should be avoided, as acidic conditions (pH <3) promote peptide bond hydrolysis, while alkaline conditions (pH >9) can cause base-catalyzed degradation and racemization. For detailed recommendations on peptide formulation and stability optimization, manufacturers should consult formulation development specialists and stability-indicating method validation studies.

8. Storage and Handling Recommendations

8.1 Lyophilized Product Storage

Lyophilized DSIP should be stored at -20°C or colder in a desiccated environment protected from light. Amber glass vials provide optimal protection against photodegradation of the tryptophan residue, which is susceptible to UV-induced oxidation. Vials should be sealed with inert stoppers (butyl rubber or PTFE-lined caps) and crimp-sealed to prevent moisture ingress during storage. For long-term archival storage, -80°C freezers provide enhanced stability, potentially extending shelf-life beyond the standard 36-month specification.

Desiccation is critical because even small amounts of absorbed moisture can accelerate degradation reactions. Storage containers should include desiccant packets (silica gel or molecular sieves) and, for GMP materials, humidity indicator cards to verify that desiccated conditions are maintained throughout the storage period. Secondary packaging in sealed aluminum pouches provides an additional moisture barrier and facilitates safe shipping at ambient temperatures for short durations (typically up to 3 weeks based on stability data).

8.2 Reconstitution Protocols

Proper reconstitution technique is essential to achieve complete dissolution and maintain peptide integrity. DSIP should be reconstituted using sterile, pyrogen-free water (WFI quality) or appropriate buffered solutions such as PBS or Tris-HCl buffer. The reconstitution solvent should be added gently along the vial wall, avoiding direct jetting onto the lyophilized cake, which can cause localized high shear and potential aggregation.

After solvent addition, the vial should be gently swirled or allowed to stand for 2-5 minutes to enable passive dissolution. Vigorous shaking or vortexing should be avoided, as mechanical agitation can promote aggregation and foam formation. If particulate matter or cloudiness is observed after reconstitution, the solution should not be used, as this indicates potential degradation, aggregation, or contamination. Properly reconstituted DSIP solutions should be clear and colorless to slightly yellow.

Table 6: Storage and Handling Guidelines
Product Form Storage Condition Shelf-Life Special Considerations
Lyophilized powder -20°C, desiccated, dark 36 months Amber vials, desiccant required
Lyophilized powder -80°C, desiccated, dark >36 months (extended) Long-term archival storage
Lyophilized powder 4°C, desiccated, dark 6 months Not recommended for long-term
Reconstituted solution 2-8°C (refrigerated) 7 days Sterile water or PBS
Reconstituted solution -20°C (frozen aliquots) 1-3 months Minimize freeze-thaw cycles
Reconstituted solution -80°C (frozen aliquots) 6-12 months Optimal for long-term solution storage
Shipping (lyophilized) Ambient with cold packs Up to 3 weeks transit Based on room temp stability data

8.3 Contamination Prevention

Aseptic technique is mandatory when handling peptides intended for in vitro or in vivo applications. All manipulations should be performed in a laminar flow hood or biosafety cabinet using sterile technique. Reconstitution solvents, pipette tips, and storage containers must be sterile and pyrogen-free. Personnel should wear appropriate personal protective equipment (gloves, lab coat, safety glasses) and follow established laboratory safety protocols.

Cross-contamination between different peptide lots or different peptides must be prevented through proper equipment cleaning, dedicated utensils, and clear labeling. Peptide solutions should be stored in clearly labeled containers indicating peptide identity, lot number, concentration, reconstitution date, and expiration date. For regulated manufacturing environments, detailed standard operating procedures (SOPs) should govern all storage and handling activities to ensure consistency and compliance with quality management systems.

To explore comprehensive best practices for peptide handling and laboratory management, quality control professionals should reference industry guidelines published by organizations such as the American Peptide Society and regulatory guidance documents from FDA, EMA, and other health authorities.

9. Certificate of Analysis (CoA) Requirements

9.1 CoA Documentation Standards

The Certificate of Analysis serves as the primary quality documentation accompanying each peptide batch, providing customers with verified analytical data demonstrating conformance to specifications. A comprehensive CoA for DSIP must include specific identifying information (product name, catalog number if applicable, lot/batch number, manufacturing date, expiration/retest date), physical and chemical properties (appearance, molecular formula, molecular weight, sequence), analytical test results (identity by MS, purity by HPLC with chromatogram, peptide content by AAA, water content, endotoxin level, bioburden if applicable), storage and handling recommendations, and quality assurance certification (signature and date by authorized QC manager).

CoAs must be generated from actual analytical testing data for the specific lot, not from generic or historical data. Each test result should include the analytical method reference, acceptance criteria, and measured value with appropriate units and significant figures. For HPLC purity, the CoA should either include a chromatogram image or reference a chromatogram file available upon request. Mass spectrometry data should report observed mass values and acceptable mass range based on theoretical molecular weight.

9.2 Example CoA Format

The following example illustrates a representative Certificate of Analysis for a GMP-grade DSIP batch, demonstrating the level of detail and documentation required for pharmaceutical-quality peptides. Manufacturing facilities should develop standardized CoA templates that ensure consistency across all product batches and facilitate customer quality review processes.

CERTIFICATE OF ANALYSIS

Product Name: Delta Sleep-Inducing Peptide (DSIP)

Sequence: Trp-Ala-Gly-Gly-Asp-Ala-Ser-Gly-Glu

Molecular Formula: C35H48N10O15

Molecular Weight: 848.81 g/mol (average mass)

CAS Number: 62568-57-4

Lot Number: DSIP-20251023-001

Manufacturing Date: October 23, 2025

Expiration Date: October 22, 2028 (36 months, -20°C storage)

Quantity: 100 mg


ANALYTICAL TEST RESULTS
Test Method Specification Result
Appearance Visual White to off-white powder White powder
Identity (MS) ESI-MS 848.33 ± 0.5 Da 848.35 Da (conforms)
Purity RP-HPLC (220 nm) ≥98.5% 99.3%
Single Impurity RP-HPLC (220 nm) ≤0.5% 0.3% (max observed)
Peptide Content AAA 70-85% 78.2%
Water Content Karl Fischer ≤5.0% 2.8%
Amino Acid Ratio AAA Conforms ±10% Conforms to sequence
Endotoxin LAL (gel clot) <0.5 EU/mg <0.25 EU/mg
Bioburden Total Aerobic Count <10 CFU/g <1 CFU/g

Storage Conditions: Store at -20°C or colder in a desiccated, light-protected environment. Use amber vials with desiccant.

Reconstitution: Reconstitute in sterile water or PBS. Use reconstituted solution within 7 days when stored at 2-8°C.

Quality Assurance Approval:

Signature: ___________________________ Date: October 23, 2025

Name: Dr. Jane Smith, QC Manager

This Certificate of Analysis is issued for the specific lot indicated. Analytical methods are validated and documented in the manufacturer's quality management system. Supporting analytical data (chromatograms, spectra) are available upon request.

9.3 Regulatory Compliance and Auditing

For GMP-manufactured DSIP intended for clinical development or commercial pharmaceutical applications, CoAs must meet enhanced regulatory requirements including reference to pharmacopeial monographs (if available), stability data supporting labeled shelf-life, validation status of analytical methods (in accordance with ICH Q2 guidelines), and change control documentation for any method or specification modifications. Regulatory inspections and customer audits will scrutinize CoA documentation, batch records, and analytical raw data to verify data integrity and GMP compliance.

Manufacturing facilities should implement robust quality management systems compliant with ICH Q7 (API manufacturing), FDA 21 CFR Part 211 (if applicable), or equivalent international standards. Electronic quality management systems with audit trails, electronic signatures, and data integrity controls are increasingly expected for regulated peptide manufacturing. For comprehensive guidance on regulatory compliance in peptide manufacturing, quality professionals should consult regulatory affairs specialists and stay current with evolving regulatory expectations.

10. Regulatory Considerations and Quality Management

10.1 GMP Compliance Framework

Manufacturing of DSIP for pharmaceutical applications requires adherence to current Good Manufacturing Practice (cGMP) regulations, which establish comprehensive quality standards for facilities, equipment, personnel, materials, production processes, and quality control systems. In the United States, peptide manufacturing is governed by FDA regulations under 21 CFR Parts 210-211 for finished pharmaceuticals and ICH Q7 guidelines for active pharmaceutical ingredients (APIs). European manufacturing facilities must comply with EU GMP guidelines published in EudraLex Volume 4, while other jurisdictions maintain similar regulatory frameworks harmonized through ICH.

GMP compliance requires manufacturing in controlled environments with appropriate cleanroom classification for sterile operations, validated manufacturing equipment and analytical instrumentation, qualified personnel with documented training, written procedures (Standard Operating Procedures) for all critical operations, comprehensive documentation and record-keeping systems, robust quality control laboratory with validated analytical methods, formal change control and deviation management processes, and regular internal audits and management review.

10.2 Process Validation

Process validation demonstrates that the manufacturing process consistently produces DSIP meeting predetermined quality specifications. Validation follows a lifecycle approach encompassing process design (development and optimization of synthesis, purification, and lyophilization processes), process qualification (installation qualification (IQ), operational qualification (OQ), and performance qualification (PQ) of equipment and systems), and continued process verification (ongoing monitoring and periodic evaluation of commercial production data).

For DSIP manufacturing, critical process parameters (CPPs) requiring validation include synthesis coupling times and reagent equivalents affecting crude purity, purification gradient conditions and fraction collection criteria affecting final purity, and lyophilization cycle parameters affecting water content and cake appearance. Process performance qualification typically involves manufacturing three consecutive conforming batches under routine conditions, demonstrating process reproducibility and capability to meet specifications.

10.3 Analytical Method Validation

All analytical methods used for DSIP quality control must be validated according to ICH Q2(R1) guidelines, demonstrating that methods are suitable for their intended purpose. Method validation parameters include specificity (ability to unequivocally assess DSIP in presence of impurities), linearity (proportional response across expected concentration range, typically 50-150% of target), accuracy (recovery of known amounts of analyte, typically 98-102%), precision (repeatability and intermediate precision, RSD typically <2%), detection limit (LOD) and quantitation limit (LOQ) for impurity methods, robustness (resistance to small deliberate variations in method parameters), and range (concentration range over which method is validated).

For RP-HPLC purity methods, validation must demonstrate resolution of DSIP from known synthesis impurities and degradation products. Forced degradation studies (acid, base, oxidative, thermal, photolytic stress) generate degradation products that test method specificity and stability-indicating capability. Mass spectrometry identity methods require demonstration of mass accuracy and specificity for DSIP versus closely-related sequences.

10.4 Supply Chain and Vendor Qualification

GMP peptide manufacturing depends on qualified suppliers providing materials of appropriate quality. Raw material suppliers (amino acids, resins, coupling reagents, solvents) must be evaluated and approved through formal supplier qualification programs assessing quality systems, regulatory compliance history, technical capabilities, and reliability. Critical raw materials should be obtained from multiple qualified suppliers to ensure supply continuity and mitigate risk.

Incoming material testing verifies that raw materials meet specifications before use in production. For amino acids, testing typically includes identity (HPLC, MS, or NMR), purity (HPLC), optical rotation (chiral purity), water content, and heavy metals. Certificates of Analysis from suppliers are reviewed but do not substitute for independent testing of GMP materials. For additional insights into peptide manufacturing supply chain management, quality professionals should review industry benchmarking studies and supplier audit best practices.

10.5 Continuous Improvement and Technology Transfer

Leading peptide manufacturers implement continuous improvement programs to enhance process efficiency, yield, and quality. Process analytical technology (PAT) initiatives incorporate real-time monitoring of critical parameters, enabling data-driven process optimization and enhanced process understanding. For DSIP manufacturing, PAT applications might include in-line UV monitoring during purification, real-time moisture analysis during lyophilization, or automated fraction collection based on UV spectral deconvolution.

When transferring DSIP manufacturing processes between facilities or scaling production volumes, formal technology transfer protocols ensure process equivalence. Side-by-side comparisons of material produced at different sites or scales verify that quality attributes remain consistent. Successful technology transfer requires comprehensive process documentation, analytical method transfer and validation, personnel training, and comparative batch production demonstrating equivalent quality.

11. Conclusion

Manufacturing pharmaceutical-grade DSIP requires integration of advanced peptide synthesis chemistry, sophisticated analytical techniques, and rigorous quality management systems. This manufacturing profile has detailed the critical elements of DSIP production: Fmoc solid-phase synthesis protocols optimized for this nonapeptide sequence, preparative RP-HPLC purification achieving >98.5% purity, comprehensive quality control testing including MS identity confirmation and HPLC purity quantification, batch documentation and release specifications meeting GMP requirements, stability data supporting 36-month shelf-life under appropriate storage conditions, proper handling and storage protocols for both lyophilized and reconstituted forms, comprehensive Certificate of Analysis documentation, and regulatory compliance frameworks ensuring pharmaceutical quality.

For manufacturers and quality control professionals, adherence to these technical specifications and quality standards ensures consistent production of DSIP meeting the stringent requirements of pharmaceutical research and potential therapeutic applications. As analytical technologies advance and regulatory expectations evolve, continuous refinement of manufacturing processes and quality control methods will further enhance the reliability and quality of peptide products.

Successful DSIP manufacturing programs combine technical expertise in peptide chemistry and analytical science with robust quality management systems and regulatory compliance frameworks. By implementing the methodologies and specifications outlined in this profile, manufacturing facilities can establish validated, GMP-compliant production processes capable of delivering pharmaceutical-grade DSIP to support advancing research and clinical development initiatives.

References

  1. Bachem AG. "Solid Phase Peptide Synthesis (SPPS) Explained." Bachem Knowledge Center. Available at: https://www.bachem.com/articles/peptides/solid-phase-peptide-synthesis-explained/
  2. Amblard M, Fehrentz JA, Martinez J, Subra G. "Advances in Fmoc solid-phase peptide synthesis." Mol Biotechnol. 2016;33(3):239-254. doi:10.1385/MB:33:3:239. Available at: https://pmc.ncbi.nlm.nih.gov/articles/PMC4745034/
  3. Mant CT, Hodges RS. "HPLC Analysis and Purification of Peptides." Methods Mol Biol. 2008;386:1-21. Available at: https://pmc.ncbi.nlm.nih.gov/articles/PMC7119934/
  4. GenScript. "High Quality Peptide Synthesis Platform - Quality Control/Quality Assurance." Available at: https://www.genscript.com/peptide_quality.html
  5. AltaBioscience. "Peptide Synthesis, Purification and Product Analysis." Available at: https://altabioscience.com/articles/peptide-synthesis-purification-and-product-analysis/
  6. Bachem AG. "Peptide Purification Process and Methods: An Overview." Bachem Knowledge Center. Available at: https://www.bachem.com/knowledge-center/peptide-guide/peptide-purification-process-and-methods/
  7. Creative Peptides. "Peptide Stability Testing Services." Available at: https://www.pepdd.com/services/peptide-stability-testing.html
  8. International Council for Harmonisation (ICH). "Q2(R1): Validation of Analytical Procedures: Text and Methodology." Available at: https://www.ich.org/page/quality-guidelines
  9. GenScript. "Ensure Safe & Effective Peptide Drugs: Mastering GMP Compliance for Quality Control." Available at: https://www.genscript.com/peptide-news/key-considerations-for-gmp-compliance-in-peptide-drug-quality-control.html
  10. PolyPeptide Group. "Quality Control in Peptide Manufacturing: Specifications for Pharmaceutical Peptides." Available at: https://www.polypeptide.com/wp-content/uploads/2019/10/1401702924538c4a0cb5e0a.pdf