View Article

  • Recent Advances in Analytical and Bioanalytical Methods For COVID-19 and Other Critical New Therapeutics (2022-2024): A Comprehensive Review

  • Department of Pharmaceutical Sciences, SJJTU, Rajasthan, India

Abstract

The emergence of novel and repurposed therapeutics—particularly in response to global health crises such as COVID-19—has driven significant innovation in analytical and bio-analytical methodologies. These methods play a pivotal role in ensuring the specificity, sensitivity, and accuracy required for drug discovery, development, quality assurance, and regulatory approval. This review provides a comprehensive overview of recent advances in analytical and bioanalytical techniques applied to the estimation and characterization of newly developed pharmaceutical agents. Emphasis is placed on innovations in chromatographic (HPLC, UPLC), spectroscopic (UV, IR, MS), hyphenated (LC-MS/MS, GC-MS), and biosensor-based technologies. Applications in therapeutic drug monitoring, pharmacokinetics, stability testing, and clinical diagnostics are highlighted, showcasing their critical importance in modern pharmaceutical analysis.

Keywords

Analytical methods, Bioanalytical methods, COVID-19 drugs, LC-MS/MS, HPLC, Emerging therapeutics, Pharmacokinetics, Drug monitoring, Biosensors, Hyphenated techniques

Introduction

Pharmaceutical analysis is a dynamic field that plays a crucial role in drug discovery, development, quality assurance, and therapeutic monitoring. The recent surge in novel drug entities, such as antiviral agents, monoclonal antibodies, and RNA-based therapeutics, necessitates the development of precise and advanced analytical methods. Analytical techniques are essential in ensuring drug safety, efficacy, stability, and purity, while bio-analytical methods provide insights into drug metabolism, bioavailability, and pharmacokinetics [1][2]. The COVID-19 pandemic has underscored the importance of rapid and reliable analytical approaches. Drugs like remdesivir, molnupiravir, and nirmatrelvir/ritonavir (Paxlovid) have gained prominence, necessitating sophisticated analytical tools for their evaluation. Additionally, with novel complex therapeutic modalities including monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), oligonucleotides, peptides, and targeted small molecules, necessitate advanced analytical and bioanalytical methods to support R&D, PK/PD studies, QC, and therapeutic drug monitoring (TDM). This review explores the latest analytical and bio-analytical methods and advances (2022-2024) employed for these critical drugs [3][4], driven by regulatory evolution (ICH M10) [26] and technological innovation.

Analytical Techniques for Recent Small Molecule Drugs

Remdesivir

Remdesivir is a nucleotide analogue prodrug approved for the treatment of COVID-19. The analysis of remdesivir and metabolites requires sensitive and selective techniques due to its rapid metabolism. High-performance liquid chromatography (HPLC) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) are widely used [5]. A recent HPLC method utilized a C18 column with a gradient mobile phase of acetonitrile and phosphate buffer, achieving good resolution and peak symmetry. LC-MS/MS methods offer lower detection limits and better specificity, particularly for pharmacokinetic studies. These methods often use solid-phase extraction (SPE) or protein precipitation for sample preparation, followed by electrospray ionization (ESI) in positive mode [6].

HPLC and LC-MS/MS Method Parameters and Validation for Remdesivir

Figure 1. Representative HPLC chromatogram of Remdesivir using UV detection.

Parameter

HPLC Method (UV Detection)

LC-MS/MS Method (ESI Positive Mode)

Column Type

C18

C18

Mobile Phase

Acetonitrile: Phosphate Buffer

Water (0.1% FA): Acetonitrile (0.1% FA)

Retention Time

~4.2 min

~3.6 min

Detection Wavelength

245 nm

MRM Transition: m/z 603 → 200

Linearity Range

10–100 μg/mL

0.5–100 ng/mL

Limit of Detection

2 μg/mL

0.2 ng/mL

Limit of Quantification

6 μg/mL

0.5 ng/mL

Recovery (%)

95.2 ± 1.4

92.7 ± 1.2

Intra-day Precision CV

<2.5%

<3.0%

Sample Prep

Protein Precipitation

Solid Phase Extraction

Case Study: Baricitinib - Analytical Evaluation in COVID-19 Therapy

Baricitinib, a JAK inhibitor, has been repurposed as a treatment option for COVID-19 due to its anti-inflammatory properties. Quantification of baricitinib in human plasma was achieved using LC-MS/MS.

Parameter

Result

Column Type

C18

Mobile Phase

Acetonitrile: Formic Acid (0.1%)

Retention Time

4.5 min

Detection

MRM Transition: m/z 372 → 251

LOQ

0.2 ng/mL

Recovery (%)

96.8 ± 1.7

Linearity Range

0.2–500 ng/mL

Precision (%RSD)

≤ 5%

Matrix Effect

<7%

Molnupiravir

Molnupiravir, another antiviral agent for COVID-19, is a prodrug of the ribonucleoside analogue N4-hydroxycytidine (NHC). Analytical challenges stem from its rapid conversion to NHC in vivo. LC-MS/MS remains the gold standard for quantifying both molnupiravir and NHC in plasma and tissues [7]. Recent studies utilized a reverse-phase C18 column and gradient elution with formic acid in water and acetonitrile. Mass spectrometric detection in positive ion mode enabled detection limits as low as 0.5 ng/mL. Sample preparation involved SPE to improve recovery and minimize matrix effects [8].

Comparative Pharmacokinetic and Analytical Profile: Molnupiravir vs. NHC

Figure 2. Overlay of LC-MS/MS chromatograms for Molnupiravir and NHC.

Parameter

Molnupiravir

NHC (Active Metabolite)

Structure

Prodrug of N4-hydroxycytidine

Nucleoside analogue

Tmax (plasma)

~1.5 hours

~3 hours

Cmax (mean)

~1.2 μg/mL

~3.5 μg/mL

Half-life

0.9 hours

3.3 hours

LOD (LC-MS/MS)

0.5 ng/mL

0.2 ng/mL

LOQ

1.5 ng/mL

0.5 ng/mL

Sample Preparation

Protein Precipitation

SPE

Matrix Effect

Moderate

Low

Lenacapavir

Lenacapavir is a long-acting capsid inhibitor approved for HIV-1 infection. Its low dosing frequency and lipophilic nature require specific analytical methods. Reverse-phase HPLC with UV detection has been developed for formulation analysis, while LC-MS/MS is used for bioanalytical applications [9].

Optimized methods include the use of acidic mobile phases to enhance retention and resolution. LC-MS/MS methods employ ESI and multiple reaction monitoring (MRM) for high specificity. These methods are validated for linearity, accuracy, precision, and robustness according to ICH guidelines [10].

Lenacapavir LC-MS/MS Method Validation Summary

Parameter

Result

Column

C18

Mobile Phase

Acetonitrile: Formic Acid in Water

Retention Time

~5.1 minutes

LOD

0.1 ng/mL

LOQ

0.3 ng/mL

Linearity Range

0.5–500 ng/mL

Recovery (%)

89.4 ± 2.3%

Precision (RSD)

<5%

Accuracy

97.2% to 102.4%

Matrix Effect

Negligible (<8%)

Nirmatrelvir-Ritonavir (Paxlovid)

Paxlovid, a combination of nirmatrelvir and ritonavir, has been used to treat mild-to-moderate COVID-19. Nirmatrelvir is a protease inhibitor, and ritonavir boosts its plasma concentration. Analytical methods focus on simultaneous quantification of both components in biological matrices [11]. UHPLC-MS/MS methods have been developed with short run times (<5 min) and high sensitivity (LLOQ ~1 ng/mL). Sample preparation involves protein precipitation, and analysis is conducted using C18 columns with mobile phases comprising ammonium formate and acetonitrile. These methods are validated for bioequivalence and clinical studies [12].

Analytical Approaches for Biologics and RNA-based Drugs

Monoclonal Antibodies (e.g. Tocilizumab, Casirivimab, Imdevimab)

The analysis of monoclonal antibodies (mAbs) poses unique challenges due to their size, heterogeneity, and complex structure. Traditional ligand-binding assays (e.g., ELISA) are widely used but may lack specificity and quantitative precision [13]. Hybrid LC-MS/MS methods have been developed to overcome these limitations. These methods involve enzymatic digestion (e.g., trypsin) followed by targeted peptide analysis using LC-MS. Capillary electrophoresis (CE), surface plasmon resonance (SPR), and size-exclusion chromatography (SEC) are also used for assessing purity, aggregation, and binding kinetics [14].

Case Study: Sotrovimab (Monoclonal Antibody)

Sotrovimab is a monoclonal antibody developed against SARS-CoV-2. Analytical challenges include glycosylation profiling, charge variant analysis, and intact mass measurement.

Techniques used:

  • Capillary Electrophoresis (CE-SDS): For molecular weight distribution
  • Size Exclusion Chromatography (SEC): For aggregation profiling
  • LC-MS Peptide Mapping: For sequence confirmation and post-translational modifications

Results:

  • Purity: >98% (SEC)
  • Glycoform distribution: G0F (~65%), G1F (~25%), G2F (~10%)
  • Intact Mass Accuracy: ±2 Da

mRNA Vaccines

The development of mRNA vaccines for COVID-19 introduced new analytical challenges, including the analysis of RNA integrity, encapsulation efficiency, and lipid nanoparticle characterization. RT-qPCR and digital PCR are commonly used for quantifying mRNA content and degradation [15]. Nano LC-MS/MS has been applied to detect translated proteins in vivo. Analytical ultracentrifugation (AUC), dynamic light scattering (DLS), and nanoparticle tracking analysis (NTA) are used to characterize lipid nanoparticles. High-resolution techniques ensure batch consistency and regulatory compliance [16].

Nano LC-MS/MS Method Performance for mRNA Vaccine Analysis

Parameter

Performance Value

Column Type

Nano C18 Column

Flow Rate

300 nL/min

Detection

Orbitrap MS

Resolution

>100,000

Dynamic Range

5 orders of magnitude

Sample Volume

1–5 µL

Protein ID Coverage

~80%

Quantitation Accuracy

±10%

Bioanalytical Methods for Pharmacokinetic and Clinical Studies

Bioanalytical methods are essential for evaluating drug absorption, distribution, metabolism, and excretion (ADME). LC-MS/MS remains the cornerstone for small molecule analysis, offering high sensitivity and selectivity [17]. For biologics, ligand-binding assays and hybrid LC-MS approaches provide accurate quantification in plasma and tissues. Micro-sampling techniques such as dried blood spots (DBS) and volumetric absorptive micro sampling (VAMS) are gaining popularity due to minimal invasiveness and ease of transport [18].

Emerging Technologies and Recent Advances in Pharmaceutical Analysis

Innovation Drivers

Methodological advancements respond to critical challenges:

  • Molecular complexity: mAbs, ADCs, and gene therapies demand multi-attribute characterization [36,37]
  • Ultra-low dosing: Potent oncology/CNS drugs require exceptional sensitivity (sub-ng/mL) [38]
  • Matrix interference: Analyses in CSF, tissues, and single cells [39,40]
  • Real-time monitoring: Emerging point-of-care TDM needs [41]

Technological Advancements

LC-MS Innovations

  • Microflow/nano-LC: 10-50x sensitivity gains for peptides (e.g., GLP-1 agonists) in biological matrices [42,43]
  • Trapped Ion Mobility Spectrometry (TIMS): Resolves ADC DAR heterogeneity and isobaric metabolites [44,45]
  • Hybrid immunoaffinity LC-MS: Quantifies total antibody and conjugated payload in ADCs simultaneously [46]

High-Resolution Mass Spectrometry

  • Intact mass analysis: Real-time monitoring of CQAs (glycosylation, oxidation) in biologics [47]
  • Multi-Attribute Methods (MAMs): Replace orthogonal assays with single LC-HRMS workflows for mAb/ADC characterization [48]

Ligand Binding Assays

  • Digital ELISA: Achieves attomolar sensitivity for biomarkers and low-dose therapeutics [49]
  • Multiplexed platforms: Simultaneously quantify 10+ analytes from ≤50μL samples (e.g., cytokine panels) [50]
  • Cell-based assays: Functional assessment of gene therapy immunogenicity [51]

Emerging Techniques

  • CE-MS: Charge-based separation for mRNA vaccines/oligonucleotides [52]
  • Wearable biosensors: Continuous monitoring of antibiotics (vancomycin) and antiepileptics [53]
  • Mass spectrometry imaging: Spatial drug distribution in tumor tissues [54]

Drug Class-Specific Advances

1. GLP-1 Agonists (Semaglutide, Tirzepatide)

  • Challenge: Peptide stability and low plasma concentrations
  • Solutions: Immunoaffinity LC-MS/MS (LLOQ 0.1 ng/mL); TIMS-enabled metabolite ID

2. ADCs (Trastuzumab Deruxtecan)

  • Challenge: DAR heterogeneity and free payload toxicity
  • Solutions: Hybrid LBA/LC-MS for conjugated payload; HRMS for in vivo DAR quantification

3. KRAS Inhibitors (Sotorasib)

  • Challenge: Reactive metabolites and tissue distribution
  • Solutions: HRMS metabolite profiling?; QWBA and MALDI imaging for tumor penetration [55,56]

4. Cell/Gene Therapies (CAR-T, AAV)

  • Challenge: Vector persistence and immunogenicity
  • Solutions: ddPCR for vector genomes [57]; cell-based NAD assays; capsid HRMS characterization [58]

Green Analytical Chemistry

Sustainability is gaining importance in pharmaceutical analysis. Green analytical chemistry (GAC) promotes the use of eco-friendly solvents, minimal reagent consumption, and energy-efficient instruments. Supercritical fluid chromatography (SFC) and miniaturized techniques align with these principles [19].

Artificial Intelligence and Machine Learning

AI and machine learning (ML) are transforming data interpretations in analytical chemistry. These technologies enable pattern recognition, predictive modeling, and optimization of chromatographic conditions. Integration with chemometrics enhances quality control and method development [20].

Microfluidics and Lab-on-a-Chip

Microfluidic platforms offer rapid, cost-effective, and portable analytical solutions. These systems integrate sample preparation, separation, and detection in a single device. They are particularly useful for point-of-care diagnostics and high-throughput screening [21].

Spectroscopic Techniques

Advanced spectroscopic methods such as Raman, near-infrared (NIR), and Fourier-transform infrared (FTIR) spectroscopy are employed for non-destructive analysis. Coupled with multivariate analysis, they provide real-time monitoring of critical quality attributes [22].

Mass Spectrometry Imaging

Mass spectrometry imaging (MSI) allows spatial localization of drugs and metabolites in tissues. Techniques like matrix-assisted laser desorption/ionization (MALDI) and desorption electrospray ionization (DESI) are used for tissue distribution studies [23].

Summary of Emerging Technologies in Pharmaceutical Analysis

Technology

Key Features

Application Areas

Sensitivity

Microfluidics

Miniaturized, rapid, portable

Point-of-care, high-throughput

Moderate

AI/ML for Chemometrics

Predictive modeling, optimization

Method development, quality control

High (with big data)

Mass Spectrometry Imaging

Spatial localization, non-destructive

Tissue distribution, oncology

High

Green Analytical Methods

Eco-friendly solvents, minimal waste

Regulatory, QC labs

Variable

FTIR/NIR Spectroscopy

Rapid, non-destructive

Identity, content uniformity

Moderate

Regulatory Considerations

Analytical method validation is guided by ICH Q2(R2), ICH M10[26], FDA, and EMA guidelines. Key parameters include accuracy, precision, linearity, and robustness with emphasis on:

  • Sensitivity: LLOQ justification for ultra-low dose drugs [27]
  • Specificity: Interference testing in complex matrices [28]
  • Analytical controls: Critical for MAMs and cell-based assays [30,31]
  • Sustainability: Green chemistry principles in method development [29].

Regulatory bodies also emphasize data integrity, method lifecycle management, and risk-based approaches [24]. The concept of Analytical Quality by Design (AQbD) is being adopted to ensure method robustness and consistency. Analytical target profiles (ATP), design of experiments (DoE), and control strategies are integral components [25].

Future Perspectives

  • AI integration: Automated data processing and predictive matrix interference modeling [32]
  • Multi-omics convergence: Linking PK data with proteomics/metabolomics [33]
  • Point-of-care translation: Clinical implementation of biosensors for TDM [34]
  • Green analytics: Miniaturized LC systems reducing solvent waste.

CONCLUSION

The landscape of pharmaceutical analysis is rapidly evolving, driven by the need to support novel therapeutics and comply with stringent regulatory standards. Advancements in LC-MS (microflow, TIMS), HRMS-based MAMs, and emerging biosensors address critical needs for novel therapeutics. The integration of AI, green chemistry, multi-omics and microfluidics promises a more efficient, accurate, and sustainable future for pharmaceutical analysis. As drug molecules become more complex, analytical scientists must continue to innovate and adapt methodologies to ensure the safety and efficacy of new therapies [35]. The development and validation of robust analytical and bio-analytical methods will remain a cornerstone of modern pharmaceutical science.

REFERENCES

  1. International Conference on Harmonization (ICH) Q2(R2).
  2. FDA Guidance for Industry: Bioanalytical Method Validation.
  3. Beigel JH, et al. (2020). Remdesivir for the Treatment of Covid-19. N Engl J Med.
  4. Owen DR, et al. (2021). An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science.
  5. Avataneo V, et al. (2020). Development and validation of LC-MS/MS assay for remdesivir quantification. J Pharm Biomed Anal.
  6. Du P, et al. (2022). Quantification of remdesivir and GS-441524 by HPLC-UV. Talanta.
  7. Painter WP, et al. (2021). Human safety, tolerability, and pharmacokinetics of molnupiravir. Antimicrob Agents Chemother.
  8. Bhatia HK, et al. (2023). LC-MS/MS method for molnupiravir and NHC in plasma. Biomed Chromatogr.
  9. Margolis DA, et al. (2021). Long acting antiretrovirals: Lenacapavir development. Curr Opin HIV AIDS.
  10. Pankey GA, et al. (2022). Bioanalytical method validation for Lenacapavir. J Pharm Sci.
  11. Hammond J, et al. (2022). Clinical efficacy of Paxlovid. N Engl J Med.
  12. Sweeney ZK, et al. (2022). Analytical methods for nirmatrelvir/ritonavir. Bioanalysis.
  13. Beck A, et al. (2013). Strategies & challenges for the analytical characterization of mAbs. Nat Rev Drug Discov.
  14. Hong P, et al. (2012). Applications of capillary electrophoresis in antibody characterization. Electrophoresis.
  15. Verbeke R, et al. (2021). mRNA vaccines: challenges & analytical perspectives. J Control Release.
  16. Hassett KJ, et al. (2019). Lipid nanoparticle characterization for mRNA vaccines. Mol Ther.
  17. Xu RN, et al. (2007). LC-MS bioanalysis: overview and advances. J Pharm Biomed Anal.
  18. Spooner N, et al. (2009). Micro sampling techniques in bioanalysis. Bioanalysis.
  19. Ga?uszka A, et al. (2013). Green analytical chemistry principles. Trends Anal Chem.
  20. Agatonovic-Kustrin S, et al. (2014). Chemometrics in pharmaceutical analysis. J Pharm Biomed Anal.
  21. Mark D, et al. (2010). Microfluidic lab-on-a-chip systems. Chem Soc Rev.
  22. Blanco M, et al. (2009). Spectroscopic techniques for pharmaceutical analysis. TrAC Trends Anal Chem.
  23. Prideaux B, et al. (2015). Mass spectrometry imaging for drug distribution. Annu Rev Pharmacol Toxicol.
  24. EMA Guidelines on bioanalytical method validation.
  25. Rozet E, et al. (2013). Analytical QbD: Regulatory guidance and industry practice. J Pharm Biomed Anal.
  26. ICH M10 Guideline. Bioanalytical Method Validation. 2022.
  27. Jani D, Allinson J, Berisha F, et al. Bioanalysis. 2020; 12:255-70.
  28. ICH M10 Guideline. 2022.
  29. P?otka-Wasylka J, Mohamed HM, Kurowska-Susdorf A, et al. TrAC Trends Anal Chem.
  30. Zhang L, et al. Anal Chem. 2023;95:1525-34.
  31. Kaur S, et al. Bioanalysis. 2021; 13:1687-703.
  32. Pifferi C, Fu H, Ruiz AL, et al. Nat Mach Intell. 2023; 5:917-31.
  33. Zhang J, Hu H, Li M, et al. Nat Rev Drug Discov. 2022; 21:717-34.
  34. Aqai P, Peters J, Gerssen A, et al. Compr Anal Chem. 2021;91:257-95.
  35. Van De Merbel NC, et al. Bioanalysis. 2023; 15:5-8.
  36. Xu K, Liu L, Dere R, et al. Drug Discov Today. 2022; 27:103327.
  37. Dere R, Yi JH, Lei C, et al. Bioanalysis. 2021; 13:1115-28.
  38. Ruan Q, Ji QC, Arnold ME, et al. Bioanalysis. 2021; 13:1629-43.
  39. Yang Z, Hayes M, Fang X, et al. J Pharm Biomed Anal. 2023; 225:115207.
  40. Li H, Ortiz R, Tran L, et al. Anal Chem. 2022;94:1435-48.
  41. Zhang Z, Shah B, Richardson J. Clin Pharmacol Ther. 2023; 113:1005-19.
  42. Xu Y, Wang YM, Kaur R, et al. Anal Bioanal Chem. 2023;415:4501-13.
  43. Ruan Q, et al. Bioanalysis. 2021; 13:1629-43.
  44. Li Y, Gu C, Gruenhagen J, et al. Anal Chem. 2021;93:2066-74.
  45. Li Y, et al. Anal Chem. 2021;93:2066-74.
  46. Dere R, et al. Bioanalysis. 2021; 13:1115-28.
  47. Firth D, Langridge JI, Belford M, et al. Anal Chem. 2023;95:43-59.
  48. Saveliev S, Woodcock J. MAbs. 2023; 15:2190648.
  49. Chen B, Dodge R, Kommineni S, et al. Anal Chem. 2022;94:6148-56.
  50. Kaur S, Xu K, Saad OM, et al. Bioanalysis. 2021; 13:1687-703.
  51. Smith JG, Crist RM. Mol Ther Methods Clin Dev. 2022;24:105-18.
  52. Qu Y, Wu X, Yin Y, et al. Anal Chem. 2023;95:3595-603.
  53. Kijanka G, Dorresteijn B, Oliveira S, et al. ACS Sens. 2019;4:3308-17.
  54. Li H, et al. Anal Chem. 2022;94:1435-48.
  55. Yang Z, et al. J Pharm Biomed Anal. 2023; 225:115207.
  56. Li H, et al. Anal Chem. 2022;94:1435-48.
  57. Vugmeyster Y, DeFranco D, Szklut P, et al. Bioanalysis. 2020; 12:1279-94.
  58. Smith JG, et al. Mol Ther Methods Clin Dev. 2022;24:105-18. 2021;138:116223.

Reference

  1. International Conference on Harmonization (ICH) Q2(R2).
  2. FDA Guidance for Industry: Bioanalytical Method Validation.
  3. Beigel JH, et al. (2020). Remdesivir for the Treatment of Covid-19. N Engl J Med.
  4. Owen DR, et al. (2021). An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science.
  5. Avataneo V, et al. (2020). Development and validation of LC-MS/MS assay for remdesivir quantification. J Pharm Biomed Anal.
  6. Du P, et al. (2022). Quantification of remdesivir and GS-441524 by HPLC-UV. Talanta.
  7. Painter WP, et al. (2021). Human safety, tolerability, and pharmacokinetics of molnupiravir. Antimicrob Agents Chemother.
  8. Bhatia HK, et al. (2023). LC-MS/MS method for molnupiravir and NHC in plasma. Biomed Chromatogr.
  9. Margolis DA, et al. (2021). Long acting antiretrovirals: Lenacapavir development. Curr Opin HIV AIDS.
  10. Pankey GA, et al. (2022). Bioanalytical method validation for Lenacapavir. J Pharm Sci.
  11. Hammond J, et al. (2022). Clinical efficacy of Paxlovid. N Engl J Med.
  12. Sweeney ZK, et al. (2022). Analytical methods for nirmatrelvir/ritonavir. Bioanalysis.
  13. Beck A, et al. (2013). Strategies & challenges for the analytical characterization of mAbs. Nat Rev Drug Discov.
  14. Hong P, et al. (2012). Applications of capillary electrophoresis in antibody characterization. Electrophoresis.
  15. Verbeke R, et al. (2021). mRNA vaccines: challenges & analytical perspectives. J Control Release.
  16. Hassett KJ, et al. (2019). Lipid nanoparticle characterization for mRNA vaccines. Mol Ther.
  17. Xu RN, et al. (2007). LC-MS bioanalysis: overview and advances. J Pharm Biomed Anal.
  18. Spooner N, et al. (2009). Micro sampling techniques in bioanalysis. Bioanalysis.
  19. Ga?uszka A, et al. (2013). Green analytical chemistry principles. Trends Anal Chem.
  20. Agatonovic-Kustrin S, et al. (2014). Chemometrics in pharmaceutical analysis. J Pharm Biomed Anal.
  21. Mark D, et al. (2010). Microfluidic lab-on-a-chip systems. Chem Soc Rev.
  22. Blanco M, et al. (2009). Spectroscopic techniques for pharmaceutical analysis. TrAC Trends Anal Chem.
  23. Prideaux B, et al. (2015). Mass spectrometry imaging for drug distribution. Annu Rev Pharmacol Toxicol.
  24. EMA Guidelines on bioanalytical method validation.
  25. Rozet E, et al. (2013). Analytical QbD: Regulatory guidance and industry practice. J Pharm Biomed Anal.
  26. ICH M10 Guideline. Bioanalytical Method Validation. 2022.
  27. Jani D, Allinson J, Berisha F, et al. Bioanalysis. 2020; 12:255-70.
  28. ICH M10 Guideline. 2022.
  29. P?otka-Wasylka J, Mohamed HM, Kurowska-Susdorf A, et al. TrAC Trends Anal Chem.
  30. Zhang L, et al. Anal Chem. 2023;95:1525-34.
  31. Kaur S, et al. Bioanalysis. 2021; 13:1687-703.
  32. Pifferi C, Fu H, Ruiz AL, et al. Nat Mach Intell. 2023; 5:917-31.
  33. Zhang J, Hu H, Li M, et al. Nat Rev Drug Discov. 2022; 21:717-34.
  34. Aqai P, Peters J, Gerssen A, et al. Compr Anal Chem. 2021;91:257-95.
  35. Van De Merbel NC, et al. Bioanalysis. 2023; 15:5-8.
  36. Xu K, Liu L, Dere R, et al. Drug Discov Today. 2022; 27:103327.
  37. Dere R, Yi JH, Lei C, et al. Bioanalysis. 2021; 13:1115-28.
  38. Ruan Q, Ji QC, Arnold ME, et al. Bioanalysis. 2021; 13:1629-43.
  39. Yang Z, Hayes M, Fang X, et al. J Pharm Biomed Anal. 2023; 225:115207.
  40. Li H, Ortiz R, Tran L, et al. Anal Chem. 2022;94:1435-48.
  41. Zhang Z, Shah B, Richardson J. Clin Pharmacol Ther. 2023; 113:1005-19.
  42. Xu Y, Wang YM, Kaur R, et al. Anal Bioanal Chem. 2023;415:4501-13.
  43. Ruan Q, et al. Bioanalysis. 2021; 13:1629-43.
  44. Li Y, Gu C, Gruenhagen J, et al. Anal Chem. 2021;93:2066-74.
  45. Li Y, et al. Anal Chem. 2021;93:2066-74.
  46. Dere R, et al. Bioanalysis. 2021; 13:1115-28.
  47. Firth D, Langridge JI, Belford M, et al. Anal Chem. 2023;95:43-59.
  48. Saveliev S, Woodcock J. MAbs. 2023; 15:2190648.
  49. Chen B, Dodge R, Kommineni S, et al. Anal Chem. 2022;94:6148-56.
  50. Kaur S, Xu K, Saad OM, et al. Bioanalysis. 2021; 13:1687-703.
  51. Smith JG, Crist RM. Mol Ther Methods Clin Dev. 2022;24:105-18.
  52. Qu Y, Wu X, Yin Y, et al. Anal Chem. 2023;95:3595-603.
  53. Kijanka G, Dorresteijn B, Oliveira S, et al. ACS Sens. 2019;4:3308-17.
  54. Li H, et al. Anal Chem. 2022;94:1435-48.
  55. Yang Z, et al. J Pharm Biomed Anal. 2023; 225:115207.
  56. Li H, et al. Anal Chem. 2022;94:1435-48.
  57. Vugmeyster Y, DeFranco D, Szklut P, et al. Bioanalysis. 2020; 12:1279-94.
  58. Smith JG, et al. Mol Ther Methods Clin Dev. 2022;24:105-18. 2021;138:116223.

Photo
Prasanth V G
Corresponding author

Research Scholar, Dept of pharmacy, SJJT University, Rajasthan, India

Photo
Ankit Singh
Co-author

Department of Pharmaceutical Sciences, SJJTU, Rajasthan, India

Prasanth V G, Ankit Singh, Recent Advances in Analytical and Bioanalytical Methods For COVID-19 and Other Critical New Therapeutics (2022-2024): A Comprehensive Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 7, 130-138. https://doi.org/10.5281/zenodo.15783646

More related articles
Method Development And Validation Of Propylthiour...
Shalu Bharti, Dev Prakash Dahiya, Palak , Chinu Kumari, ...
Migraine Headache: An Overview ...
Jasmine Dhiman, Neha, Sanjeev Duggal, ...
Related Articles
A Review On Chromatographic Separation Techniques ...
Swati Appasaheb Pachore , Anushka Anil shinde, Laware sandip Gangadhar, ...
Natural Hypertension Remedies: A Review of Medicinal Plant Efficacy...
Dr. Payal Vaja, Hiral Popaniya, Dilip Ghusar, Vivek Solanki, Siddhi Upadhyay, Harsh Madiya, ...
Method Development And Validation Of Propylthiouracil By UV Spectroscopy...
Shalu Bharti, Dev Prakash Dahiya, Palak , Chinu Kumari, ...
More related articles
Method Development And Validation Of Propylthiouracil By UV Spectroscopy...
Shalu Bharti, Dev Prakash Dahiya, Palak , Chinu Kumari, ...
Migraine Headache: An Overview ...
Jasmine Dhiman, Neha, Sanjeev Duggal, ...
Method Development And Validation Of Propylthiouracil By UV Spectroscopy...
Shalu Bharti, Dev Prakash Dahiya, Palak , Chinu Kumari, ...
Migraine Headache: An Overview ...
Jasmine Dhiman, Neha, Sanjeev Duggal, ...