Analytical Method Development and Validation of Ademetionine:
A Detailed Review
Mayur Bhamare, Rushikesh Bachhav, Mayur Chavan, Ganesh Sonawane, Sunil Mahajan, Vijayraj Sonawane
Department of Pharmaceutical Quality Assurance, Divine College of Pharmacy, Satana.
*Corresponding Author E-mail: mayurbhamare0404@gmail.com
ABSTRACT:
Ademetionine, commonly known as S-Adenosyl-L-methionine (SAMe), is a pivotal biological compound involved in methylation, neurotransmitter synthesis, glutathione production, and liver detoxification. This compound is widely studied for its therapeutic potential in depression, osteoarthritis, and liver diseases. The analysis of SAMe in pharmaceutical formulations and biological samples requires robust analytical methodologies due to its sensitivity to environmental factors like pH, light, and temperature. Key analytical techniques include High-Performance Liquid Chromatography (HPLC), often coupled with fluorescence or mass spectrometry detection, which provides high specificity and sensitivity. Other methods, such as Capillary Electrophoresis (CE), UV detection, and Nuclear Magnetic Resonance (NMR), offer complementary insights into its quantification, structural elucidation, and purity. Method development involves optimizing parameters like mobile phase composition, pH control, and sample preparation to ensure stability and accurate detection. Validation processes adhere to ICH guidelines, emphasizing specificity, linearity, accuracy, precision, and robustness. Therapeutically, SAMe is utilized for its hepatoprotective properties, mood-enhancing effects, and potential to alleviate osteoarthritis symptoms. Despite its benefits, careful administration is advised due to side effects like gastrointestinal disturbances and the risk of mania in individuals with bipolar disorder. This review underscores the importance of precise analytical methods for quality control and pharmacokinetic studies, facilitating the clinical and pharmaceutical applications of SAMe.
KEYWORDS: Ademetionine, Methylation, Neurotransmitter synthesis, Glutathione production, Liver detoxification.
INTRODUCTION
Ademetionine1,2
Ademetionine, also known as S-Adenosyl-L-methionine (SAMe), is a naturally occurring compound found in nearly all tissues and fluids in the body. It plays a vital role in various biochemical processes, particularly those related to methylation, a critical metabolic process that regulates the expression of genes and the function of proteins. Ademetionine is a crucial molecule for several bodily processes, including methylation, liver health, and mood regulation, with growing interest in its therapeutic potential for various conditions.
Analyzing ademetionine (SAMe) in pharmaceutical formulations, biological fluids, and tissues is essential for quality control, pharmacokinetic studies, and clinical monitoring. The analytical methods used for the detection and quantification of ademetionine are highly specialized due to the compound’s sensitivity to pH, light, and temperature. Various techniques have been developed and optimized for precise measurement, including. chromatographic, spectroscopic, and electrophoretic methods.
Functions of ademetionine:3
· Methylation reactions: Ademetionine is the principal methyl donor in the human body, involved in transferring methyl groups to DNA, proteins, lipids, and other molecules. This helps in regulating genetic expression and metabolic pathways.
· Synthesis of neurotransmitters: It assists in the synthesis of serotonin, dopamine, and norepinephrine, which are essential for mood regulation. Due to this, SAMe is sometimes linked with mental health, particularly in the treatment of depression.
· Glutathione production: Ademetionine contributes to the production of glutathione, an important antioxidant in the liver that protects cells from oxidative damage.
· Detoxification and liver function: It plays a significant role in liver health, where it assists in detoxification processes, particularly in those with liver disorders like cirrhosis or fatty liver disease.
Therapeutic Uses:
· Ademetionine has gained attention in medical and therapeutic contexts for its potential benefits.
· Depression: SAMe supplements are sometimes used as an alternative or adjunct treatment for depression due to their involvement in the production of mood-regulating neurotransmitters.
· Osteoarthritis: It has shown potential in relieving pain and improving function in patients with osteoarthritis by promoting cartilage health.
· Liver disease: SAMe has hepatoprotective properties, and it is sometimes used in the management of chronic liver conditions to improve liver function and prevent damage.
Forms and Administration:
· Ademetionine is available in supplement form, often referred to as SAMe.
· It can be taken orally or administered via injection in some therapeutic contexts.
· Supplements are widely available over-the-counter in various doses and formulations.
Side Effects and Considerations:4
While SAMe is generally well-tolerated, some people may experience side effects
· Gastrointestinal symptoms (nausea, diarrhoea)
· Insomnia
· Anxiety or restlessness
It should be used cautiously in individuals with bipolar disorder as it may increase the risk of manic episodes. Always consult with a healthcare provider before starting SAMe, especially if there are pre-existing conditions or if you are on other medications.
Mechanism of Methane5,6,7:
This diagram explains the process of feed fermentation in the ruminant digestive system and the associated emission of gases such as hydrogen (H₂), carbon dioxide (CO₂), nitrous oxide (N₂O), and methane (CH₄).
Figure No. 1
1. Feed Intake (Step 1):
· Ruminants consume plant material (e.g., grass, silage) through their mouth.
· The feed enters the esophagus and travels to the stomach.
2. Bacterial Digestion/Fermentation (Step 2):
· The ruminant's stomach has four compartments: rumen, reticulum, omasum, and abomasum.
· The rumen is the primary fermentation chamber, where microbes (bacteria, protozoa, fungi) break down plant material.
· Microbial fermentation produces volatile fatty acids (VFAs), which serve as energy sources for the animal.
3. Gas Production (Step 3):
· During fermentation, microbes release hydrogen (H₂) and carbon dioxide (CO₂) as byproducts.
· These gases accumulate in the rumen and are expelled through belching (eructation).
4. Methanogenesis (Step 4):
· Methanogens, a group of archaea in the rumen, utilize hydrogen (H₂) and carbon dioxide (CO₂) to produce methane (CH₄).
· Methane is expelled into the atmosphere through belching, contributing to greenhouse gas emissions.
5. Excretion of Nitrous Oxide (N₂O):
· Nitrogen from undigested feed and microbial protein is excreted in the feces and urine.
· Nitrogenous waste can be converted into nitrous oxide (N₂O), another greenhouse gas, during manure decomposition.
Microbial Activity:
· The microbial population in the rumen consists of:
· Bacteria: Break down complex carbohydrates like cellulose and hemicellulose.
· Protozoa: Consume bacteria and assist in fiber digestion.
· Fungi: Break down lignin and fibrous material.
Environmental Impact:
· Methane (CH₄): Produced during methanogenesis, methane is a potent greenhouse gas that contributes to global warming.
· Nitrous Oxide (N₂O): Emitted from manure management, it has a high global warming potential.
Energy Utilization:
Methane represents an energy loss for the ruminant, as it contains carbon and hydrogen that could otherwise be used for growth or milk production.
Mitigation Strategies:
· Dietary changes: Adding fats, oils, or feed additives to reduce methane emissions.
· Microbial inhibitors: Substances that suppress methanogen activity.
· Improved manure management: Reducing nitrous oxide emissions from excreta.
Structure of Methionine:8,9
Figure No. 2
Molecular Structure:10
1. Functional Groups:
· Amino group (-NH₂): Attached to the central carbon.
· Carboxyl group (-COOH): Attached to the central carbon.
· Side chain (-CH₂-CH₂-S-CH₃): A sulfur-containing side chain unique to methionine.
2. Central Alpha Carbon (Cα):
The central carbon atom is bonded to the amino group, carboxyl group, a hydrogen atom, and the side chain.
3 D Description:
Methionine has a linear chain with a sulfur atom incorporated into its side chain. This sulfur group is part of a methylthio group (-S-CH₃), which gives methionine its unique properties.
Mechanism of Methane to Methionine11,12
Methane (CH₄) cannot be directly converted into methionine (an essential amino acid) in biological systems. However, there are indirect ways in which carbon from methane or other simple molecules can be incorporated into biomolecules like methionine in specialized systems or processes.
1. Methane to Methionine: Not a Direct Pathway:
Methane is a simple hydrocarbon, while methionine is a complex sulfur-containing amino acid with a methyl group. The biological pathways to synthesize methionine require more complex intermediates, not methane itself.
Methionine biosynthesis typically occurs through metabolic pathways involving carbon compounds like aspartate, sulfur sources, and methyl groups, not directly from methane.
2. Biological Context: Role of Methanotrophs:
Methanotrophs are microorganisms that metabolize methane and convert it into other carbon-containing compounds like methanol, formaldehyde, or organic acids.
These compounds can then enter central metabolic pathways, such as the citric acid cycle, to form precursors for amino acids like methionine.
For example:
Methanotrophs oxidize methane to formaldehyde (CH₂O).
Formaldehyde can be assimilated into cell biomass, forming intermediates like serine, which are precursors for amino acid biosynthesis.
3. Synthetic Biology and Biotechnology:
In synthetic biology, engineered microbes or biotechnological systems can be designed to use methane as a carbon source to produce methionine.
Methanotrophs or other microorganisms could be genetically engineered to enhance their metabolic pathways for converting methane-derived intermediates into methionine.
4. Sulfur and Methylation in Methionine Synthesis:
Methionine synthesis in plants, fungi, and bacteria involves:
· A carbon skeleton derived from aspartate.
· A sulfur donor, such as hydrogen sulfide (H₂S) or cysteine.
· A methyl group donor, usually S-adenosylmethionine (SAMe).
5. Potential Applications:
· Converting methane into methionine is of great interest for reducing greenhouse gas emissions and producing valuable amino acids.
· Industrial processes or biotechnological innovations could achieve this goal by using methane as a feedstock for microbial or enzymatic production of methionine.
Challenges:
Methane is a stable molecule, making its conversion into complex biomolecules energy-intensive. Biological systems capable of this conversion need to balance energy efficiency, carbon utilization, and yield.Methane cannot directly convert into methionine, but with the help of methanotrophic microorganisms or synthetic biology approaches, methane can serve as a carbon source for methionine production in controlled environments.
Structure of Ademetionine:13,14
The structure of SAMe consists of:
1. Adenosine: A nucleoside formed by the combination of adenine and ribose.
2. Methionine: An essential amino acid with a sulfur-containing methyl group.
3. Linkage: A sulfonium ion (S+) connects the methyl group of methionine to the ribose moiety of adenosine.
The full structural formula:
· A purine ring (adenine).
· A ribose sugar.
· A methionine group attached via a sulfonium bond.
Figure No. 3
Mechanism of Ademetionine:15,16
Ademetionine participates in three major types of reactions:
1. Methylation (Methyl Donor):
· SAMe donates its methyl group to substrates like DNA, RNA, proteins, lipids, and small molecules.
· This process is catalyzed by methyltransferase enzymes.
· Example: DNA methylation, which regulates gene expression.
· SAMe (donor) → SAH (product) + Methylated substrate
· Structural transition involves the loss of a methyl group from the sulfonium ion.
2. Transsulfuration:
· After donating the methyl group, SAMe is converted to S-adenosylhomocysteine (SAH).
· SAH is hydrolyzed to form homocysteine, which enters the transsulfuration pathway to produce glutathione (a major antioxidant).
· SAMe → SAH → Homocysteine → Cysteine → Glutathione
· The sulfur and amino groups in methionine are conserved and redirected to biosynthesis.
3. Aminopropylation:
· SAMe is a precursor for polyamine synthesis, critical for cell growth and differentiation.
· It contributes aminopropyl groups to form spermidine and spermine.
· SAMe → Decarboxylated SAM → Aminopropyl groups for spermidine and spermine.
Figure No. 4
Analytical Method Development of Ademetionine: 17,18,19
Developing an analytical method for Ademetionine (also known as S-Adenosyl-L-methionine or SAMe), an important cofactor involved in methyl group transfers, involves several key steps to ensure accurate identification, quantification, and purity assessment of the compound. Below is a general outline of the process for developing an analytical method for Ademetionine:
1. Understanding the Compound:
a) Chemical Structure: SAMe is a derivative of the amino acid methionine and ATP, consisting of a sulfur atom, a methyl group, and a ribose triphosphate moiety. It is a zwitterion, making it challenging in terms of solubility and stability.
b) Physicochemical Properties:
SAMe is hygroscopic and sensitive to light and heat. This necessitates careful handling and storage conditions.
c) Biological Relevance:
SAMe plays a role in methylation reactions and is used as a supplement or medication for various conditions, including depression, osteoarthritis, and liver diseases.
2. Selection of Analytical Techniques:
Several techniques are typically used to develop and validate an analytical method for Ademetionine. The choice depends on the purpose (e.g., purity analysis, degradation studies, quantification, etc).
a) High-Performance Liquid Chromatography (HPLC):
HPLC is the most widely used method for the analysis of ademetionine due to its high sensitivity, specificity, and reproducibility.
HPLC with fluorescence detection (FLD):
In this method, a derivatization step is typically performed to increase sensitivity. The use of fluorescent labels can enhance detection in biological matrices.
Reversed-phase HPLC (RP-HPLC):
This is commonly used for separating SAMe from other compounds. In RP-HPLC, SAMe is eluted using a mobile phase typically consisting of water, acetonitrile, and buffer systems. HPLC is one of the most common methods used for quantifying SAMe due to its sensitivity and precision. It can be coupled with various detectors depending on the nature of the analysis.
Advantages:
· High specificity and accuracy.
· Suitable for complex biological matrices (e.g., blood, plasma, tissue samples).
Limitations:
· SAMe is unstable under alkaline or acidic conditions, so careful sample preparation is needed.
· Derivatization may be required to improve sensitivity.
b) UV Detection: Useful for monitoring absorption at specific wavelengths. Ademetionine is detected at wavelengths around 254 nm, but since it has low natural absorbance, derivatization is often required for improved detection sensitivity.
c) Mass Spectrometry (LC-MS):
Provides specificity and sensitivity, especially for complex matrices such as biological samples.LC-MS is a highly sensitive and selective method used for the quantification of ademetionine in complex matrices like plasma, urine, and tissues. LC-MS/MS (Tandem Mass Spectrometry): This is often used to enhance sensitivity and specificity. Multiple reaction monitoring (MRM) is used for accurate quantification of SAMe in biological samples.
Advantages:
· Extremely high sensitivity and specificity.
· Capable of detecting low concentrations of ademetionine in complex biological samples.
· Can identify metabolites or degradation products.
Limitations:
· Requires advanced equipment and technical expertise.
· Expensive due to equipment and consumables.
d) Refractive Index Detector (RID):
Useful for non-chromophore compounds, but less sensitive.
e) Capillary Electrophoresis (CE):
This method is useful due to SAMe’s zwitterionic nature, providing good resolution of different charged species. Capillary electrophoresis separates charged molecules like ademetionine based on their size-to-charge ratio under an electric field.
CE with UV or electrochemical detection: SAMe can be detected using various detectors depending on the type of sample and desired sensitivity. This method has been used for both pharmaceutical formulations and biological samples.
Advantages:
· High resolution and efficiency.
· Requires smaller sample volumes and fewer reagents compared to HPLC.
Limitations:
· Lower sensitivity compared to LC-MS.
· Can be more difficult to handle with complex biological samples.
f) NMR (Nuclear Magnetic Resonance):
NMR can be used for structural elucidation and to ensure the integrity of the compound. NMR spectroscopy can be used for structural identification and quantification of ademetionine, especially in its pure form or in well-defined matrices.
Proton (¹H) NMR:
Provides detailed information on the structure and purity of SAMe.
Carbon (¹³C) NMR:
Complements proton NMR for carbon-based structural analysis.
Advantages:
· Non-destructive method.
· Provides detailed structural and conformational information.
Limitations:
· Requires a large amount of sample compared to chromatographic methods.
· Not typically used for trace-level quantification in complex samples.
g) FTIR (Fourier Transform Infrared Spectroscopy): Useful for functional group identification and confirming the presence of key functional groups like amine and methyl groups.
3. Sample Preparation of Ademetionine20,21
Solubility Considerations: SAMe is soluble in water and slightly soluble in alcohol. It’s sensitive to pH and should be handled in buffered solutions.
Proper sample preparation is crucial for accurate analysis of ademetionine. The following steps are typically involved:
a) Extraction: Ademetionine is extracted from pharmaceutical formulations or biological matrices using suitable solvents such as methanol or acetonitrile. For biological samples, protein precipitation or solid-phase extraction may be employed.
b) Dilution: The extracted sample is diluted to an appropriate concentration using a buffer or solvent compatible with the analytical method.
c) Filtration: Samples are filtered through a 0.22 µm or 0.45 µm membrane filter to remove particulates that could interfere with analysis.
d) Stability Considerations: Ademetionine is sensitive to oxidation and hydrolysis. Therefore, samples should be prepared and analysed under conditions that minimize degradation, such as low temperatures and inert atmospheres.
e) Standard Preparation: A calibration curve is constructed using known concentrations of ademetionine standards to ensure quantification accuracy.
4. Mobile Phase Selection for HPLC23,24
a) pH Control: Since SAMe is sensitive to pH, selecting an appropriate buffer is critical. Commonly used buffers include phosphate or acetate buffers.
b) Organic Modifiers: Methanol or acetonitrile can be used as organic modifiers to improve the retention and separation of SAMe.
c) Ion Pairing: SAMe can form ion pairs with counterions, improving its retention on reverse-phase columns (C18).
5. Method Development Process:
a) Choosing the Column: For HPLC, C18 reverse-phase columns are commonly used. Other options include ion-exchange columns due to the ionic nature of SAMe.
b) Optimization of Parameters: Optimize parameters such as flow rate, injection volume, temperature, and detection wavelength.
c) System Suitability Testing (SST): Perform system suitability tests (e.g., peak symmetry, resolution, repeatability, etc.) to ensure the method's robustness.
6. Quantification and Purity Assessment:
The developed method should allow for the quantification of SAMe in various matrices such as pharmaceuticals, dietary supplements, and biological samples. Purity is assessed to ensure no impurities or degradation products interfere with the results.
7. Regulatory Considerations:
Ensure that the developed method complies with regulatory guidelines (e.g., ICH, USP) for pharmaceuticals. This includes validation according to parameters such as specificity, linearity, accuracy, precision, LOD/LOQ, and robustness.
Analytical Method Validation of Ademetionine:24,25
Analytical method validation for Ademetionine (SAMe) is a critical process to ensure that the method used for its analysis is reliable, accurate, and consistent. This process is typically guided by regulatory frameworks such as the International Council for Harmonisation (ICH) guidelines (Q2(R1)), which outline key validation parameters.
Below is an overview of the validation process for Ademetionine, focusing on the standard parameters that need to be assessed.
1. Specificity:
Procedure: Test the method by spiking samples with potential impurities and degradation products. Assess whether the method can distinguish between SAMe and these substances.
Outcome: The method should show that the peaks of SAMe and any impurities are well separated with no significant overlap.
2. Linearity:
Procedure: Prepare a series of standard solutions of SAMe at different concentrations (typically 50-150% of the expected concentration). Plot the peak area (or signal) against the concentration and calculate the correlation coefficient (R²).
Acceptance Criteria:
The correlation coefficient (R²) should typically be ≥ 0.99 for acceptable linearity.
3. Accuracy (Recovery):
Procedure: Add known amounts of SAMe (usually in three levels: low, medium, and high concentrations) to the matrix (e.g., placebo or biological fluid) and compare the measured amount to the actual added amount. Perform the analysis in triplicate.
Outcome:
The percentage recovery should typically be within 98-102% for pharmaceutical formulations.
4. Precision:
Types of Precision:
a) Repeatability (Intra-day Precision): The precision under the same operating conditions over a short interval of time (e.g., within a single day).
b) Intermediate Precision (Inter-day Precision): The precision within a laboratory over different days, analysts, and equipment.
Procedure:
Analyze multiple replicates (usually 6) of a sample at a specified concentration on the same day (repeatability) and on different days (intermediate precision).
Outcome:
The relative standard deviation (RSD) should typically be ≤ 2%.
5. Limit of Detection (LOD) and Limit of Quantification (LOQ):
LOD: The lowest concentration of SAMe that can be detected but not necessarily quantified.
LOQ: The lowest concentration of SAMe that can be quantified with acceptable accuracy and precision.
Procedure: These parameters are often determined based on the signal-to-noise ratio. Typically, LOD is defined as a signal-to-noise ratio of 3:1 and LOQ as 10:1.
Outcome: The LOD and LOQ should be low enough to detect and quantify SAMe at the required levels, especially in purity and stability tests.
6. Robustness:
Procedure: Small variations in parameters such as pH, temperature, mobile phase composition, flow rate, and detection wavelength should be introduced to assess the impact on the method's performance.
Outcome: The method should show minimal variation in results under slightly altered conditions, indicating that it is robust.
7. Ruggedness:
Procedure: The method is tested by different operators, on different days, and using different instruments.
Outcome: Consistent results under varied conditions.
8. System Suitability Testing (SST):
Parameters Assessed:
a) Resolution: Should be assessed for separation between SAMe and impurities.
b) Theoretical Plates: A measure of column efficiency.
c) Tailing Factor: Indicates the symmetry of the SAMe peak.
d) Repeatability: Check the repeatability of injections.
Outcome: All system suitability parameters should meet predefined acceptance criteria before sample analysis can begin.
9. Range
Procedure: The range is typically assessed during the linearity, accuracy, and precision studies. The working range for SAMe analysis might be 50-150% of the expected concentration, depending on the dosage form or application.
Outcome: The method should perform well across this range, ensuring reliable results for both low and high concentrations of SAMe.
Table No.1
Summary of Key Parameters and Acceptance Criteria |
||
Parameter |
Procedure |
Acceptance Criteria |
Specificity |
Analyse SAMe in presence of impurities and degradation products. |
Clear separation of SAMe from other substances. |
Linearity |
Plot peak area vs. concentration. |
R² ≥ 0.99. |
Accuracy |
Recovery studies at different concentrations. |
98-102% recovery. |
Precision |
Analyse replicates (intra-day and inter-day). |
RSD ≤ 2%. |
LOD |
Signal-to-noise ratio method. |
Typically, 3:1 for detection. |
LOQ |
Signal-to-noise ratio method. |
Typically, 10:1 for quantification. |
Robustness |
Small changes in method conditions. |
Minimal variation in results. |
Ruggedness |
Testing by different operators, instruments, and labs. |
Consistent results across variations. |
System Suitability |
Evaluate key parameters (e.g., resolution, tailing). |
Parameters meet criteria before analysis. |
Range |
Assessed during linearity, precision, accuracy. |
Effective in the working range (e.g., 50-150%). |
Analytical method validation for Ademetionine ensures the reliability and reproducibility of the method across various parameters. Following ICH guidelines ensures compliance with industry standards and regulatory requirements for pharmaceuticals.
IR Spectroscopy of Ademetionone:25,26
FTIR (Fourier Transform Infrared) spectrum, which shows the transmittance of infrared light through a sample as a function of wavenumber (in cm⁻¹).
1. X-axis: Wavenumber (cm⁻¹)
· The x-axis represents the wavenumber, which is inversely proportional to the wavelength of infrared light.
· Higher wavenumbers (e.g., 4000-3000 cm⁻¹) correspond to high-energy vibrations, such as O-H or N-H bonds.
· Lower wavenumbers (e.g., 1500-600 cm⁻¹) correspond to lower-energy vibrations, such as C-H, C-C, or fingerprint regions.
2. Y-axis: Transmittance (%)
· The y-axis shows the transmittance, indicating how much infrared light passes through the sample without being absorbed.
· Peaks (dips in the curve) correspond to absorption bands, where specific functional groups in the sample absorb infrared light at characteristic wavenumbers.
3. Major Peaks and Their Possible Assignments:27,28
· ~3900-3775 cm⁻¹: Broad absorption in this region often indicates O-H stretching (e.g., water or hydroxyl groups).
· 3364-2930 cm⁻¹: Typically associated with N-H stretching (amines or amides) and C-H stretching (alkanes or aromatics).
· 1688-1612 cm⁻¹: These are likely C=O stretching (carbonyl groups in ketones, aldehydes, or carboxylic acids).
· 1419-1325 cm⁻¹: These peaks are associated with C-H bending or O-H deformation in phenols or alcohols.
· 1142-1025 cm⁻¹: This region often corresponds to C-O stretching (esters, ethers, or alcohols).
· 833-610 cm⁻¹: Peaks here are in the fingerprint region, which is unique to the specific molecular structure of the compound.
Figure No. 5
CONCLUSION:
Ademetionine (S-Adenosyl-L-methionine, SAMe) is a vital biological compound with significant roles in methylation reactions, neurotransmitter synthesis, antioxidant production, and liver detoxification. Its therapeutic applications in managing depression, osteoarthritis, and liver diseases highlight its clinical importance. However, due to its sensitivity to environmental factors, developing robust and reliable analytical methods is crucial for its accurate detection and quantification in pharmaceutical and biological matrices.
Techniques like HPLC, LC-MS, and CE provide high specificity and sensitivity, while UV detection, NMR, and FTIR offer complementary insights into its structural and functional properties. Method development and validation, adhering to ICH guidelines, ensure accuracy, precision, and reproducibility, supporting its application in quality control and pharmacokinetic studies.
Overall, SAMe therapeutic potential and analytical challenges necessitate continued research to enhance its stability, optimize its use in clinical settings, and develop advanced methodologies for its analysis, ultimately contributing to improved patient outcomes and pharmaceutical innovations.
REFERANCE:
1. ICH Harmonised Tripartite Guideline. Validation of Analytical Procedures: Text and Methodology Q2(R1). International Conference on Harmonisation; 2005.
2. FDA. Guidance for Industry: Analytical Procedures and Methods Validation for Drugs and Biologics. U.S. Department of Health and Human Services, Food and Drug Administration; 2015.
3. European Medicines Agency (EMA). Guideline on Bioanalytical Method Validation. London: EMA; 2011.
4. USP General Chapter <1225>: Validation of Compendial Procedures. United States Pharmacopeia and National Formulary (USP 40-NF 35). Rockville, MD: USP Convention; 2017.
5. Shabir GA. Validation of high-performance liquid chromatography methods for pharmaceutical analysis. Understanding the differences and similarities between validation requirements of the US Food and Drug Administration, the US Pharmacopeia and the International Conference on Harmonization. J Chromatogr A. 2003 987(1-2): 57-66.
6. Swartz ME, Krull IS. Analytical Method Development and Validation. Boca Raton, FL: CRC Press; 1997.
7. Snyder LR, Kirkland JJ, Glajch JL. Practical HPLC Method Development. 2nd ed. New York: Wiley-Interscience; 1997.
8. Kazakevich Y, LoBrutto R. HPLC for Pharmaceutical Scientists. Hoboken, NJ: John Wiley & Sons; 2007.
9. Beckett AH, Stenlake JB. Practical Pharmaceutical Chemistry. 4th ed. London: Continuum International Publishing Group; 2002.
10. Jenke DR. Compatibility of Pharmaceutical Products and Contact Materials. Hoboken, NJ: John Wiley & Sons; 2009.
11. Waksmundzka-Hajnos M, Sherma J. High Performance Liquid Chromatography in Phytochemical Analysis. Boca Raton, FL: CRC Press; 2019.
12. Dehouck P, Kaal E. Development and validation of a UHPLC-MS/MS method for the quantification of S-adenosylmethionine in dietary supplements. Food Chem. 2021; 345:128736.
13. Zhao J, Xu J, Chen Y, Ma S, Lin X. Determination of S-adenosylmethionine in pharmaceutical formulations by HPLC. J Pharm Biomed Anal. 2012; 57:78-82.
14. Tiwari G, Tiwari R, Bannerjee SK. Basics of analytical method validation for pharmaceutical analysis. Int J Res Pharm Biomed Sci. 2012;3(2):971-80.
15. Ermer J, Miller JHM. Method Validation in Pharmaceutical Analysis: A Guide to Best Practice. 2nd ed. Weinheim: Wiley-VCH; 2014.
16. Rao RN, Ramachandra B, Agwan VS. Analytical method development and validation for the determination of related substances in S-adenosylmethionine. J Pharm Biomed Anal. 2018; 156:149-55.
17. Chow SC, Liu JP. Design and Analysis of Bioavailability and Bioequivalence Studies. 3rd ed. Boca Raton, FL: CRC Press; 2008.
18. Anderer P. Effects of S-adenosyl-l-methionine on neural processing by ERP-mapping. Int Congr Ser. 2002; 1232:331-6.
19. Dennis M, Smith B. Liquid Chromatography-Tandem Mass Spectrometry in Drug Analysis. Anal Chem. 2015;87(1):19-36.
20. Wilson K, Walker J. Principles and Techniques of Biochemistry and Molecular Biology. 8th ed. Cambridge: Cambridge University Press; 2018.
21. Snyder LR, Dolan JW. High-Performance Gradient Elution: The Practical Application of the Linear-Solvent-Strength Model. Hoboken, NJ: Wiley-Interscience; 2007.
22. Day RA, Underwood AL. Quantitative Chemical Analysis. 7th ed. Upper Saddle River, NJ: Pearson Education; 2001.
23. McDowall RD. The Role of Chromatography in Pharmaceutical Quality Control. Anal Chem. 2010; 82(1): 64-70.
24. Perumal OP. Validation of HPLC and UV spectrophotometric methods for the quantification of S-adenosylmethionine in a pharmaceutical dosage form. J Chromatogr Sci. 2020; 58(7): 622-8.
25. Nunnally BK, McConnell EJ. Validation of Analytical Methods for Biopharmaceuticals. 2nd ed. Hoboken, NJ: John Wiley & Sons; 2017.
26. Hinckley L, Cope C. A practical guide to implementing ICH Q2(R1). Pharm Eng. 2011;31(6):88-93.
27. USP General Chapter <621>: Chromatography. United States Pharmacopeia and National Formulary (USP 40-NF 35). Rockville, MD: USP Convention; 2017.
28. Baertschi SW, Alsante KM, Reed RA. Pharmaceutical Stress Testing: Predicting Drug Degradation. 2nd ed. New York: Informa Healthcare; 2011.
29. Qu F, Fisher MB, Mauck RL. Analytical method validation strategies for complex pharmaceutical products. J Pharm Biomed Anal. 2016; 123:12-9.
30. Sharma BK. Instrumental Methods of Chemical Analysis. 25th ed. Meerut: Krishna Prakashan Media; 2011.
31. Rao RN, Shinde DD. Development of a stability-indicating method for S-adenosylmethionine. J Pharm Sci. 2018;108(3):532-40.
32. Hoenicke K, Gatermann R. Analysis of acrylamide in different foodstuffs using liquid chromatography-tandem mass spectrometry. Anal Chim Acta. 2004; 519(1-2): 149-55.
33. Andrae WA, Hopfer R. A new approach to method validation in the pharmaceutical industry. Pharm Qual. 2021; 18(4):16-24.
34. McMaster MC, McMaster C. HPLC: A Practical User's Guide. 2nd ed. Hoboken, NJ: Wiley-Interscience; 2007.
35. Lavine BK, Workman J. Chemometrics for Analytical Chemistry. 2nd ed. Amsterdam: Elsevier; 2016.
36. T.B. Goh, M.M. Mordi, S.M. Mansor. Demethylation of Mitragynine Picrate with BBr3 and C12H25-SH /NaOMe: A new entry for Ester Derivatives Preparation based on Position Carbon 9. Asian J. Research Chem. 6(9): September 2013; Page 863-867.
37. Akhilesh Gupta, Swati Rawat, Mayuri Gandhi, Jaydeep Singh Yadav. Method Development and Acid Degradation Study of Doxofylline by RP-HPLC and LC-MS/MS. Asian J. Pharm. Ana. 1(1): Jan.-Mar. 2011; Page 10-13.
38. Akhilesh Gupta, Jaydeep Singh Yadav, Swati Rawat, Mayuri Gandhi. Method Development and Hydrolytic Degradation Study of Doxofylline by RP-HPLC and LC-MS/MS. Asian J. Pharm. Ana. 1(1): Jan.-Mar. 2011; Page 14-18.
39. Akhilesh Gupta, Swati Rawat, Arun Pandey. Method Development and Photolytic Degradation Study of Doxofylline by RP-HPLC and LC-MS/MS. Asian J. Pharm. Ana. 2011; 1(2): 29-33.
40. D. Sridharan, Umarani A. Thenmozhi, L. Pavan Kumar, Aswani Dutt Chintalapati, M. Venkata Ramanaiah, Yelika Phanikishore. Development and Validation of UV Spectrophotometric Method of Darifenacin Hydrobromide in Bulk and Tablet Dosage Form. Asian J. Pharm. Ana. 2011; 1(3): 43-45.
41. Santhi N, Rajendran S.S, Kumar Nallasivan P, Sam Solomon W.D, Venkata Narayanan R. Simultaneous Estimation of Cefixime and Ofloxacin in Bulk and Tablet Dosage Form. Asian J. Pharm. Ana. 2011; 1(3): 50-52.
42. Akhilesh Gupta, Vimal Yadav, Jaydeep S. Yadav, Swati Rawat. An Analytical Approach of Doxofylline: A Review. Asian J. Pharm. Ana. 2011; 1(4): 67-70.
43. Mahmoud M. Sebaiy, Abdullah A. El-Shanawany, Sobhy M. El-Adl, Lobna M. Abdel-Aziz, Hisham A. Hashem. Rapid RP-HPLC Method for Simultaneous Estimation of Norfloxacin and Tinidazole in Tablet Dosage Form. Asian J. Pharm. Ana. 2011; 1(4): 79-84.
44. S. Rawat, Akhilesh Gupta. Spectrophotometric Method for Simultaneous Estimation of Nimesulide and Diclofenac Sodium in Pharmaceutical Dosage Forms. Asian J. Pharm. Ana. 2011; 1(4): 85-87.
45. Gajanan Darwhekar, Priya Jain, Dinesh Kumar Jain, Gaurav Agrawal. Development and Optimization of Dorzolamide Hydrochloride and Timolol Maleate in Situ Gel for Glaucoma Treatment. Asian J. Pharm. Ana. 2011; 1(4): 93-97.
Received on 10.01.2025 Revised on 03.03.2025 Accepted on 05.04.2025 Published on 06.05.2025 Available online from May 10, 2025 Asian Journal of Pharmaceutical Analysis. 2025; 15(2):137-145. DOI: 10.52711/2231-5675.2025.00022 ©Asian Pharma Press All Right Reserved
|
|
This work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License. Creative Commons License. |
|