| Citation: | Yangrui Zhang, Yizhen Liu, Yangyang Chen, Chen Guo, Fengting Ou, Junhuan Lin, Hanmeng Guo, Tao Ke, Lushan Yu. Quantifying protein residues in APIs: Bradford assay mechanism and limitations, outperformed by HILIC-MS/MS[J]. Journal of Pharmaceutical Analysis. doi: 10.1016/j.jpha.2026.101552 |
| [1] |
U.S. Food and Drug Administration (FDA), Code of Federal Regulations, Title 21, Section 210.3: Definitions, (2025)
|
| [2] |
K. Rosenthal, S. Lutz, Recent developments and challenges of biocatalytic processes in the pharmaceutical industry, Curr. Opin. Green Sustain. Chem. 11 (2018) 58-64.
|
| [3] |
J. Krueger, A.P. Dieskau, J. Hassfeld, et al., Chemical process development in the pharmaceutical industry in Europe: Insights and perspectives from industry scientists, Angew. Chem. Int. Ed. 64 (2025), e202420719.
|
| [4] |
S.S. Choi, Y. Katsuyama, L. Bai, et al., Genome engineering for microbial natural product discovery, Curr. Opin. Microbiol. 45 (2018) 53-60.
|
| [5] |
X. Yan, X. Liu, C. Zhao, et al., Applications of synthetic biology in medical and pharmaceutical fields, Signal Transduct. Target. Ther. 8 (2023), 199.
|
| [6] |
A.S. Wells, G.L. Finch, P.C. Michels, et al., Use of enzymes in the manufacture of active pharmaceutical ingredients: A science and safety-based approach to ensure patient safety and drug quality, Org. Process Res. Dev. 16 (2012) 1986-1993.
|
| [7] |
D.J. Newman, G.M. Cragg, Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019, J. Nat. Prod. 83 (2020) 770-803.
|
| [8] |
J.A. van Santen, E.F. Poynton, D. Iskakova, et al., The Natural Products Atlas 2.0: A database of microbially-derived natural products, Nucleic Acids Res. 50 (2022) D1317-D1323.
|
| [9] |
X. Huang, P. Men, S. Tang, et al., Aspergillus terreus as an industrial filamentous fungus for pharmaceutical biotechnology, Curr. Opin. Biotechnol. 69 (2021) 273-280.
|
| [10] |
I. Tzani, M. Castro-Rivadeneyra, P. Kelly, et al., Detection of host cell microprotein impurities in antibody drug products, Nat. Commun. 15 (2024), 8605.
|
| [11] |
S. Ahuja, Assuring quality of drugs by monitoring impurities, Adv. Drug Deliv. Rev. 59 (2007) 3-11.
|
| [12] |
M. Jones, N. Palackal, F. Wang, et al., “High-risk” host cell proteins (HCPs): A multi-company collaborative view, Biotechnol. Bioeng. 118 (2021) 2870-2885.
|
| [13] |
Rosenberg, A. S. & Worobec, A. A risk-based approach to immunogenicity concerns of therapeutic protein products: part 1 considering consequences of the immune response to a protein. BioPharm Int 17, 22 (2004).
|
| [14] |
S. Panikulam, H. Morgan, M. Gutknecht, et al., Host cell protein-mediated adjuvanticity and immunogenicity risks of biotherapeutics, Biotechnol. Adv. 81 (2025), 108575.
|
| [15] |
S. Michalik, F. Siegerist, R. Palankar, et al., Comparative analysis of ChAdOx1 nCoV-19 and Ad26.COV2.S SARS-CoV-2 vector vaccines, Haematologica 107 (2022) 947-957.
|
| [16] |
N.A. Hanania, M. Noonan, J. Corren, et al., Lebrikizumab in moderate-to-severe asthma: Pooled data from two randomised placebo-controlled studies, Thorax 70 (2015) 748-756.
|
| [17] |
M. Maier, L. Weiss, N. Zeh, et al., Illuminating a biologics development challenge: Systematic characterization of CHO cell-derived hydrolases identified in monoclonal antibody formulations, mAbs 16 (2024), 2375798.
|
| [18] |
C.H. Goey, S. Alhuthali, C. Kontoravdi, Host cell protein removal from biopharmaceutical preparations: Towards the implementation of quality by design, Biotechnol. Adv. 36 (2018) 1223-1237.
|
| [19] |
P.F. Gillespie, Y. Wang, K. Yin, et al., Automated, quantitative capillary western blots to analyze host cell proteins in COVID-19 vaccine produced in vero cell line, Vaccines 12 (2024), 1373.
|
| [20] |
D.E. Walker, F. Yang, J. Carver, et al., A modular and adaptive mass spectrometry-based platform for support of bioprocess development toward optimal host cell protein clearance, mAbs 9 (2017) 654-663.
|
| [21] |
X. Gao, B. Rawal, Y. Wang, et al., Targeted host cell protein quantification by LC-MRM enables biologics processing and product characterization, Anal. Chem. 92 (2020) 1007-1015.
|
| [22] |
S. Chutipongtanate, K. Watcharatanyatip, T. Homvises, et al., Systematic comparisons of various spectrophotometric and colorimetric methods to measure concentrations of protein, peptide and amino acid: Detectable limits, linear dynamic ranges, interferences, practicality and unit costs, Talanta 98 (2012) 123-129.
|
| [23] |
H. Wang, N. Pampati, W.M. McCormick, et al., Protein nitrogen determination by Kjeldahl digestion and ion chromatography, J. Pharm. Sci. 105 (2016) 1851-1857.
|
| [24] |
K. Reinmuth-Selzle, T. Tchipilov, A.T. Backes, et al., Determination of the protein content of complex samples by aromatic amino acid analysis, liquid chromatography-UV absorbance, and colorimetry, Anal. Bioanal. Chem. 414 (2022) 4457-4470.
|
| [25] |
M. Hayes, Measuring protein content in food: An overview of methods, Foods 9 (2020), 1340.
|
| [26] |
A. Jain, R. Jain, S. Jain, Basic Techniques in Biochemistry, Microbiology and Molecular Biology: Principles and Techniques. New York, NY: Springer US, (2020).
|
| [27] |
E. Brunelle, A.M. Le, C. Huynh, et al., Coomassie brilliant blue G-250 dye: An application for forensic fingerprint analysis, Anal. Chem. 89 (2017) 4314-4319.
|
| [28] |
M.M. Bradford, A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Anal. Biochem. 72 (1976) 248-254.
|
| [29] |
J.G. Song, K.C. Baral, G.L. Kim, et al., Quantitative analysis of therapeutic proteins in biological fluids: Recent advancement in analytical techniques, Drug Deliv. 30 (2023), 2183816.
|
| [30] |
P. Højrup, Analysis of polypeptides by amino acid analysis, . Peptide Antibodies. Springer US, (2024), pp 1–82.
|
| [31] |
D.H. Spackman, W.H. Stein, S. Moore, Automatic recording apparatus for use in chromatography of amino acids, Anal. Chem. 30 (1958) 1190-1206.
|
| [32] |
F. Duranton, U. Lundin, N. Gayrard, et al., Plasma and urinary amino acid metabolomic profiling in patients with different levels of kidney function, Clin. J. Am. Soc. Nephrol. 9 (2014) 37-45.
|
| [33] |
B. Buszewski, S. Noga, Hydrophilic interaction liquid chromatography (HILIC): A powerful separation technique, Anal. Bioanal. Chem. 402 (2012) 231-247.
|
| [34] |
M. Lioi, S. Tengattini, V. D’Atri, et al., Evaluating the potential of hydrophilic interaction liquid chromatography for collagen peptide mapping analysis, J. Chromatogr. A 1738 (2024), 465473.
|
| [35] |
A. Periat, J. Boccard, J.L. Veuthey, et al., Systematic comparison of sensitivity between hydrophilic interaction liquid chromatography and reversed phase liquid chromatography coupled with mass spectrometry, J. Chromatogr. A 1312 (2013) 49-57.
|
| [36] |
R. Armin, J. Wachendorf, M. Weber, et al., Enhanced industrial wastewater monitoring: Method development for non-target screening of highly polar substances using ZIC-HILIC-HRMS, Anal. Bioanal. Chem. 417 (2025) 167-181.
|
| [37] |
Chinese Pharmacopoeia Commission, Pharmacopoeia of the People’s Republic of China (2025 edition, Vol. 4, Section 0731), (2025).
|
| [38] |
International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), Q2(R2) Validation of Analytical Procedures: Text and Methodology, (2023).
|
| [39] |
J. Lin, Y. Song, Y. Zhang, et al., A reliable LC-MS/MS method for the quantification of natural amino acids in human plasma and its application in clinic, J. Pharm. Biomed. Anal. 256 (2025), 116672.
|
| [40] |
Y. Ge, Y. Tang, S. Guo, et al., Simultaneous quantitation of free amino acids, nucleosides and nucleobases in Sipunculus nudus by ultra-high performance liquid chromatography with triple quadrupole mass spectrometry, Molecules 21 (2016), 408.
|
| [41] |
B. Yuan, W. Lyu, F.F. Dinssa, et al., Free amino acids in African indigenous vegetables: Analysis with improved hydrophilic interaction ultra-high performance liquid chromatography tandem mass spectrometry and interactive machine learning, J. Chromatogr. A 1637 (2021), 461733.
|
| [42] |
O. Wahl, U. Holzgrabe, Amino acid analysis for pharmacopoeial purposes, Talanta 154 (2016) 150-163.
|
| [43] |
E. Rogatsky, Pandora box of BCA assay. Investigation of the accuracy and linearity of the microplate bicinchoninic protein assay: Analytical challenges and method modifications to minimize systematic errors, Anal. Biochem. 631 (2021), 114321.
|
| [44] |
J. Cortes-Rios, A.M. Zarate, J.D. Figueroa, et al., Protein quantification by bicinchoninic acid (BCA) assay follows complex kinetics and can be performed at short incubation times, Anal. Biochem. 608 (2020), 113904.
|
| [45] |
Y. Yan, M. Zhang, J. Gao, et al., Comparison of methods for detecting protein extracted from excess activated sludge, Environ. Sci. Pollut. Res. 30 (2023) 60967-60975.
|
| [46] |
H.K. Ku, H.M. Lim, K.H. Oh, et al., Interpretation of protein quantitation using the Bradford assay: Comparison with two calculation models, Anal. Biochem. 434 (2013) 178-180.
|
| [47] |
E. Tsochatzis, M. Papageorgiou, S. Kalogiannis, Validation of a HILIC UHPLC-MS/MS method for amino acid profiling in Triticum Species wheat flours, Foods 8 (2019), 514.
|
| [48] |
J.A. Custodio-Mendoza, P. Pokorski, H. Aktas, et al., Rapid and efficient high-performance liquid chromatography-ultraviolet determination of total amino acids in protein isolates by ultrasound-assisted acid hydrolysis, Ultrason. Sonochem. 111 (2024), 107082.
|
| [49] |
K.S. Ojha, D. Granato, G. Rajuria, et al., Application of chemometrics to assess the influence of ultrasound frequency, Lactobacillus sakei culture and drying on beef jerky manufacture: Impact on amino acid profile, organic acids, texture and colour, Food Chem. 239 (2018) 544-550.
|
| [50] |
N.K.N. Bui, S. Selberg, K. Herodes, et al., Coumarin-based derivatization reagent for LC-MS analysis of amino acids, Talanta 252 (2023), 123730.
|
| [51] |
C. Le, C. Kunacheva, D.C. Stuckey, “Protein” measurement in biological wastewater treatment systems: A critical evaluation, Environ. Sci. Technol. 50 (2016) 3074-3081.
|