Determining Part 1: Methodology

Determining protein Nadine Ritter and John McEntire concentration is one of the most important analytical methods hether a product consists of crude nucleic acids, cell culture media containing dyes used in the discovery, development, and tissue homogenates, semipurified or surfactants, and formulations with high con- manufacturing of biological fluids, or highly purified centrations of reducing agents, detergents, or salts protein products. recombinant , the amount may each require different types of protein con- Part 1 discusses a of protein in a given amount of centration analysis to obtain accurate results in number of methods for sample is valuable information. the presence of interfering matrix components. analyzing samples Protein concentration is analyzed to The amount of sample available for testing ranging from complex Wmonitor the amount of material recovered at each (both volume and concentration) is also a driving mixtures to highly step in a purification process and to determine the purified proteins. factor in selecting the appropriate method. Pro- appropriate quantity of material to use in subse- tein assays can vary significantly in their mini- quent process steps. In biopharmaceutical pro- mum required volume, their linear working duction, determining protein concentration is one range, and their ability to measure highly diluted way to measure the manufacturing consistency of samples accurately. There is often a trade-off be- each batch or the stability of a protein solution tween sensitivity and accuracy, although vendors over time. Protein concentration values are also of commercial protein kits are working to needed when preparing test samples for analytical improve both. methods to assess product purity, identity, and Matrix components can significantly affect potency. Because of its wide-ranging use in oper- protein concentration determination. Therefore, ations from discovery and development through with most protein concentration assays you commercial manufacturing, the measurement of should analyze the sample buffer alone to evalu- protein concentration is one of the most useful ate and correct for background signals. If the and significant test methods performed on interference is too great, then either remove or di- biotechnology products. In Part 1 of this discus- lute the matrix or select another assay. Even sion, we look at a number of methods for deter- when no individual components interfere, the mining protein concentration. synergistic effect of some substance combina- The nature of the protein preparation to be tions can create assay artifacts. When in doubt, it tested is a critical factor in the selection of an ap- is always wise to confirm the effects of the spe- propriate concentration determination method. cific buffer component or components at the con- Some methods, such as colorimetric assays, can centrations expected for the product with the pro- be used with both crude and purified protein solu- tein concentration assay chosen. tions to determine a total protein concentration. Others, such as UV absorbance, require solutions COLORIMETRIC METHODS of relatively pure protein for accurate results. Colorimetric assays are commonly used in bio- Unless methods are chosen that can isolate the molecular laboratories for determining protein signal specific for a single protein, heterogeneous concentration because the procedures and their in- protein mixtures will generate a combined re- strumentation requirements are simple. In such as- sponse from the proteins present. Solutions says, the protein is reacted with one or more chro- containing host cell elements such as lipids or mogenic substances to yield a detectable signal.

12 BioPharm APRIL 2002 Two basic forms of colorimetric assays are used. in data from a standard curve. Generally, a stock The first form involves reactions between the pro- solution of the standard protein is prepared and tein and chemicals in solution that yield colored, its concentration accurately determined (by fluorescent, or chemiluminescent products. In the amino acid analysis or microanalytical total second form, a colored dye is bound to the protein nitrogen, for example). and an absorbance shift is generated. Within the For long-term use (and when validating a col- linear working range of both types of assays, the orimetric assay), it is important to consider the intensity of the color quantitatively correlates to stability of the standards under their intended the amount of protein present. storage conditions. If a stock solution of the stan- These methods are well established, proven dard shows precipitation or microbial growth with many different protein preparations, and rel- upon storage, its protein concentration will no atively easy to use. When optimized for specific longer be as expected. Assays performed with the applications, most are precise and reproducible. solution will not be accurate. To increase confi- However, their disadvantages include limited dence in the stock standard solution concentration sensitivity at very low ranges (for example, accuracy over time, perform stability tests and es- below 1 µg/mL), interferences from buffer com- tablish appropriate storage conditions. ponents, unstable chromogenic reaction products, and potential inaccuracies if a protein standard does not react similarly to the test protein. Although the method Standards. The quantitation of these assays is instructions or based on the preparation of a standard curve with known concentrations of standards. Standards can literature references be nonproduct surrogate proteins (such as bovine Buffers and reagents. In addition to stable stan- may give the linear serum albumin, BSA) or a well-characterized dard solutions, accurate assay results use the reference standard of the product itself. A set of same type of buffer in preparing the standard working range of x,y data points is generated from the known stan- curve dilutions as in the test sample. If the con- dard concentrations and their corresponding centration of buffer components affects a colori- most colorimetric absorbance readings, and a best-fit regression line metric assay, adjust the standards to contain the assays, it should be is drawn through them. The test protein same concentration of buffer constituents across absorbance readings are plotted against this best- the dilution range of the standard curve. For some verified for specific fit line, and the concentration results are calcu- colorimetric assays (such as the Lowry method), applications. lated by linear regression analysis. chromogenic reactions are a function of time. Because protein standards drive the quantita- Therefore, the timing of the reaction incubations tive results, the accuracy of colorimetric assays for individual assay tubes can be critical to depends on the validity of the standard curve. obtaining accurate and precise protein concentra- Key elements in preparing accurate and precise tion results. You may have to coordinate the ad- standard curves include the protein chosen as the dition of reagents to ensure that each vial is incu- known standard (either a surrogate or a reference bated for the same length of time at the same protein), the preparation of the standard curve di- temperature before measuring its absorbance lutions, establishment of the assay’s linear work- reading. ing range, and the stability of the stock standard The effect of reagents on spectrophotometric solution. cuvettes used for holding assay solutions can af- Not all proteins react similarly with the vari- fect precision. Reagents such as colloidal gold ous chromogenic reagents used in colorimetric (Aurodye) or Coomassie blue dye can bind to assays. For the most accurate results, use a some cuvettes, causing a drift in absorbance read- protein standard that will react as closely as pos- ings from the first to the last sample. Also, the sible to the protein or proteins being tested. use of glass rather than plastic cuvettes can affect Ideally, a highly purified solution of the protein assay results. Procedures for cleaning quartz or being tested will provide the most accurate stan- glass cuvettes between samples must be adequate dard curve values because it will react in the to prevent sample carry over. When using dispos- assay exactly as will the test article. When a well- able plastic cuvettes, manufacturing quality and characterized product reference standard is consistency from lot to lot are critical for obtain- unavailable, a representative protein such as BSA ing reliable data. can be used as a surrogate standard. Procedures. Although the method instructions In either case, you must know the exact or literature references may give the linear work- concentration of the standard to have confidence ing range of most colorimetric assays, it should BioPharm APRIL 2002 13 Colorimetric Assays We describe here six colorimetric protein assay’s working range is 0.01–1 mg/mL. chloroauric acid (Aurodye), which is highly concentration assays that may be used with Color reaction development is time- sensitive to protein. The working range of biopharmaceutical products. For additional dependent and light-sensitive and can vary this assay is 20–200 ng/mL. The reaction colorimetric assays, refer to Thorne (1). Many significantly from protein to protein. In occurs in 30 minutes, and the color is stable of these assays are available as commercial addition, numerous buffer components can for up to 24 hours. This assay is rugged to a kits containing all assay reagents and, in interfere with the Lowry assay, including wide range of buffer components but is some cases, surrogate standard proteins. In salts (10,11), Tris (11), detergents slightly sensitive to pH, performing optimally each section, we compare their general (8,12,13), sucrose (8,13), and EDTA (13). at pH 3 (17). As with the colorimetric assays operational ranges (combining their original Bicinchoninic acid assay. The bicinchoninic above, reactivity differs among proteins, so and microscaled versions). acid assay (BCA) is also a modification of it is best to analyze standards by a Check current vendor literature for the latest the biuret reaction (13). This assay uses the noncolorimetric method to verify their kits available because improvements are reduction of Cu (I) by protein to Cu (II) in an suitability for use with the test protein(s). continuously being made to enhance the alkaline solution, then bicinchoninic acid O-phthalaldehyde (OPA) assay. When limit of detection, limit of quantitation, and detects the cuprous ion to form a colored o-phthalaldehyde reacts with amines, it assay robustness to matrix components. product absorbing at 562 nm. The working generates an adduct that fluoresces on Not all proteins react the same in every range is 0.001–1 mg/mL. The BCA assay excitation at 340 nm and can be detected at assay, so it may be useful to evaluate has many advantages over the biuret and 450 nm (18). The OPA assay procedure is several methods to determine the Lowry assays: The color reaction is stable significantly more involved than the other appropriate one to use. Regardless of for hours, the assay is simple (one step), colorimetric assays. It requires which method is chosen, confirm the actual and it is rugged to many common buffer extensive sample preparation and a working ranges when first testing an assay. components (13). However, like the Lowry fluorometer rather than a spectrophotometer. Biuret assay. The biuret assay is among the assay, it exhibits some protein-to-protein However, in some applications where you oldest of the colorimetric assays, with variation in reactivity (13). Use an cannot avoid interfering matrix elements, this elements originating in the early 1900s orthogonal concentration method to may be the most suitable assay. (2–4). Protein reacts with (II) sulfate evaluate the selected standard against the You may get more accurate results with that, when reduced by the nitrogen atoms in test protein for reactivity. some protein samples if you first hydrolyze the peptide bonds to Cu1+, undergoes a Bradford dye-binding assay. The Bradford them with NaOH at high temperature. The color shift to 540 nm. The assay is assay (also known as the dye binding or working range using unhydrolyzed samples performed at room temperature with 30 Coomassie blue assay) uses Coomassie is 1–25 µg/mL. Hydrolysis can increase minutes of incubation. Its working range of brilliant blue G-250 dye, which undergoes a sensitivity 10-fold. However, you may protein detection is 0.5–10 mg/mL. Hence it color shift from 465 nm to 595 nm when introduce significant assay imprecision is unsuitable for highly dilute protein bound to protein (14–16). The binding during hydrolysis and subsequent preparations. Buffer components such as reaction is rapid and is stable for up to an neutralization and dilution. Because the Tris (5) and glycerol (6) can interfere with hour. This assay’s working range is fluorescent adducts are stable only for up to accurate concentration determination in the 0.001–0.100 mg/mL, so it can be used with 15 minutes, this assay requires timed assay. dilute protein solutions. Like the BCA assay, it coordination of active reagent additions. It is Lowry assay. The Lowry assay and its is a simple, one-step protocol, rugged to most less affected than the Lowry or Bradford modifications are enhancements of the common buffer components (14,15). But as methods by many common buffer biuret reaction (7–9). Intensified color with the Lowry and BCA, protein to protein components such as detergents, EDTA, results from the reaction of Folin– differences in reactivity can occur (15,16). thimerosal, cesium chloride, and guanidine Ciocalteau reagent: Alkaline copper reacts Colloidal gold assay. Colloidal gold has a hydrochloride. But Tris, sucrose, thiol with a protein (as in the biuret test), then the maximum absorbance at 530 nm (17). reagents, and urea do affect OPA assay phosphomolybdate-phosphotungstate salts When mixed with protein, the absorption results, and as with other colorimetric in the reagent are reduced to produce a maximum shifts and can be optimally assays, reactivity differences among maximum absorbance at 750 nm. This detected at 595 nm. The reaction involves proteins have been seen (18). be verified for specific applications — especially curve within a narrower concentration range, or if the buffer contains components not shown in a shift the range higher or lower. Once you have published method. Common practice is to prepare established the linear working range of an assay, a minimum of five standard points in duplicate or the absorbance readings of test samples should triplicate across a five- to tenfold range of con- fall within the range of those generated by the centration. Using linear regression, determine the standard curve. If they are too high, it may be best-fit line through the data points. An accept- possible to dilute and reassay the protein solution. able linear fit would yield a coefficient of deter- If they are too low, the protein solutions should mination (r2) of 0.98 or greater. The precision be quantitatively concentrated and if possible, re- among replicates should be within 5%. assayed. If the precision among replicates is acceptable, but the coefficient of determination is less than UV ABSORBANCE MEASUREMENTS 0.98, the standard curve points might not reflect Determining protein concentration by measuring the linear range of the assay. Repeat the standard a solution’s absorbance at ultraviolet (UV) wave-

14 BioPharm APRIL 2002 lengths can be rapid and precise. Apart from a A protein solution’s concentration can be blank or placebo solution, no additional assay calculated from a standard curve measured reagents are needed. Protein concentration is directly by UV absorbance, similar to the colori- calculated using the extinction coefficient of the metric methods. Using a reference solution with a protein. Again, concentration accuracy depends known concentration of the same protein to be on linear regression from a standard curve. Mea- analyzed, you can generate a standard curve. suring concentration by UV absorbance is hypo- Measure the absorbances at 280 nm of each stan- thetically nondestructive to the sample. Once dard point and plot those against its known con- measurement is completed, the sample can be re- centration, then draw a best-fit line. Measure the covered and reused (although this practice is not test sample at A280, then evaluate against the recommended in typical biopharmacuetical appli- standard curve using linear regression. The accu- cations). racy of this technique depends on both the refer- UV absorbance methods have certain limita- ence protein solution and the test sample protein tions. They can detect only those proteins that solution having the same absorbance maxima and contain aromatic residues (predominantly tyro- containing the same quality and quantity of impu- sine and tryptophan). Because calculation of con- rities. centration uses the extinction coefficient of a spe- When first using the UV absorbance method cific protein, this method is more accurate with on any test sample, you should perform spectral solutions of purified proteins than with complex scans be performed from 200 to 600 nm on a solutions. It is critical that a buffer blank be ana- buffer blank or placebo, and then scan the test so- lyzed in parallel without protein because lution. If the test solution is too concentrated, it certain solvents and buffer components exhibit can be diluted in buffer or placebo to read in the UV absorption. Other biomolecules present in the linear range of the spectrophotometer (typically sample, such as nucleic acids that also absorb in 0–2 AU). By comparing the absorbance profiles the UV region, can confound the results. among blanks and samples, you should discover Calculating the results. The following equation any potential interfering signals present in the can be used to calculate protein concentration matrix. If you find overlapping absorbance peaks, from UV absorbance values: concentration you may be able to adjust the protein’s (mg/mL) = absorbance/molar extinction coeffi- absorbance values by subtracting the interference cient (19). The extinction coefficients of numer- values (if they are at low levels). This is particu- ous proteins have been published; however, those larly important in solutions containing nucleic of most new biopharmaceutical products have acids, where the A260 value is subtracted from the not. Therefore it may be necessary to determine it A280 value to correct for the presence of DNA or empirically for a given protein if it is highly puri- RNA (22). Use the corrected absorbance value to fied and quantities are sufficient to analyze accu- calculate protein concentration. rately (20). Highly purified preparations can be estimated by gravimetric (dry weight) analysis if AMINO ACID ANALYSIS the protein preparation is free of exogenous sub- Amino acid analysis is one of the most accurate stances such as salts or detergents (21). ways to determine protein concentration and a For the greatest accuracy, amino acid analysis protein’s extinction coefficient (23). This tech- can determine a protein’s composition and weight nique also yields the amino acid composition of (see below). If there is a significant discrepancy purified protein samples. When analyzing bio- between the dry weight and the weight derived pharmaceutical products, amino acid composition from amino acid analysis, your protein prepara- can be used in conjunction with other techniques tion is likely to contain additional, nonproteina- (such as ) to establish protein ceous material. If the amino acid composition of identity (24). Determining concentration by your protein is known, you can estimate the theo- amino acid analysis is an important product retical extinction coefficient based on the number characterization method and provides an orthogo- of tyrosine and tryptophan residues present (20) nal technology to colorimetric assays or UV and using a simple formula: extinction coefficient absorbance. Although it requires hydrolysis and = (number of tyrosines x 1,280) + (number of HPLC instrumentation and an experienced ana- tryptophans x 5,690)/protein molecular weight. lyst, it is sensitive enough to be used with very Although it may be useful to estimate this value low-level samples (200–500 pmol) including early in product development, the actual value eluates from electrophoretic gels or PVDF blots should be determined experimentally for use in (25,26,27). We give a brief description of the product characterization. 16 BioPharm APRIL 2002 method and some of its limitations here. A com- precolumn methods offer greater assay sensitiv- prehensive review of amino acid analysis tech- ity, but postcolumn methods are more robust nology by Anders begins on page 32 of this issue. (25). The benefits and limitations of different hy- Other detailed reviews of amino acid analysis de- drolysis procedures, derivatization reagents, and rivatization chemistries can be found elsewhere chromatographic resolution systems are detailed (20,28). elsewhere (20,28). Hydrolysis. In amino acid analysis, you Certain buffer components can interfere with hydrolytically destroy the peptide bonds between the accuracy of amino acid analysis. Samples the individual amino acids in the protein, and containing amines (such as Tris or glycine) may then measure the quantity of recovered amino yield peaks that coelute with amino acid residues acid residues. Hydrolysis is typically performed during HPLC, making accurate quantitation diffi- with strong acid and high temperature. This cult. Detergents or high amounts of salts can in- procedure adequately liberates most of a protein’s terfere with the solubilization and recovery of amino acids, with certain limitations. Asparta- hydrolyzed residues and may negatively affect mine and glutamine are converted to aspartic and chromatography. Protein properties can also glutamic acids, respectively; tryptophan, cys- interfere with analysis. Glycoproteins and teine, serine, and threonine are partially or com- lipoproteins may be incompletely hydrolyzed or pletely destroyed; peptide bonds between some incompletely solubilized after hydrolysis. Envi- residues (isoleucine, leucine, and valine) are ronmental conditions also contribute to contami- more resistant to hydrolysis than others; many nating substances: Residue on glassware, impure modified amino acids are not reliably recovered. reagents, even powder from an analyst’s gloves By modifying hydrolysis conditions or deriva- can contribute to artifacts during amino acid tization chemistry, one or the other of those analysis. issues can be addressed, but (to date) no single Calculating the results. Protein concentration can hydrolysis procedure can quantitatively yield be calculated from amino acid results in different every amino acid simultaneously. In fact, hydrol- ways (35). In each case, determine or estimate the ysis is the greatest source of error in amino acid amount of protein present in the tested aliquot. analysis (29). Compositional accuracy averages Calculate concentration using the amount of 15% error overall; individual error associated protein per volume of sample, adjusting for all with difficult residues can approach 30% with dilution steps used for sample preparation in the some derivatization chemistries (25). For maxi- method. Most analysts omit cysteine and trypto- mum accuracy, perform multiple hydrolysis time phan from consideration because of the impreci- points (24, 48, and 72 hours, for example), and sion associated with those residues (36). The sim- extrapolate the data back to estimate time-zero plest method is single residue normalization, in yields of each amino acid (30). To improve assay which the data from one stable amino acid (reli- accuracy and precision, compounds such as ably recovered by hydrolysis) is used to represent -amino-N-butyric acid, norvaline, or norleucine all amino acids in the protein (37). Phenylalanine, spiked into samples can be used as quantitative alanine, and leucine are typically stable residues. internal standards to correct for potential sample This method requires knowledge of the theoreti- losses during hydrolysis. cal amino acid composition of the protein. The Quantitation. After sample hydrolysis, the percentage of a chosen residue contained in the released amino acid residues are resolved by protein is calculated. The actual amount obtained HPLC and quantitated. To be detected by UV or of the chosen residue is multiplied by the known fluorescence, the residues must be derivatized amount of that residue in the protein. The data with a chromagenic agent. That can be done be- are then normalized to represent 100% of the fore (precolumn) or after (postcolumn) HPLC. In protein’s mass, yielding an estimate of recovery precolumn analysis, the residues are derivatized, based on weight percent. chromatographically separated, then detected; in Best-fit analysis is a more accurate estimation postcolumn analysis the residues are separated, method. It requires data from all the amino acids derivatized, then detected. Currently, the four present (except cysteine and tryptophan) (36). predominant derivatization chemistries are Like normalization, this method requires theoreti- phenylisothiocyanate (PITC) (31), 6-amino- cal compositional values for the protein. Calculate quinolyl-N-hydroxysuccinimidyl carbamate an initial average yield by dividing the amount (AQC) (32) (both precolumn), OPA (33), and obtained by the known (theoretical) residue ninhydrin (34) (both postcolumn). In general, amount, then calculate the mean of those data. Compare the amount obtained of each residue to 20 BioPharm APRIL 2002 Nitrogen Assays colorimetric methods. However, you will get in- accurate results using samples that contain non- Kjeldahl assay. The Kjeldahl assay Nessler assay. The Nessler assay is proteinaceous sources of nitrogen or ammonia involves digestion of the protein sample similar to the Kjeldahl assay. Protein by a combination of acid, catalysts, and samples are digested with acid, but unless you can remove the exogenous material high temperature, which converts the hydrogen peroxide is used to accelerate before analysis. Incomplete digestions can result nitrogen to ammonium sulfate (39). Then, the oxidation of nitrogen to ammonium in imprecision, so you may need to optimize di- steam-distill the ammonium sulfate to sulfate (40). After digestion, add Nessler gestion conditions for individual applications. liberate ammonia gas. Quantitate the reagent (mercury and potassium iodide Also, precision can be affected by the loss of ma- ammonia by indirect titration against an in sodium hydroxide) to the reaction to terial to the sides of tubes and flasks. Take care to acid or direct titration against a base. Use produce a colored complex with the amount of ammonia to calculate the ammonia. Use an ammonium sulfate completely recover liquid droplets adhering to nitrogen in the sample, and then standard reference solution to quantitate glass vessels, especially after digestion. The Kjel- mathematically convert that to protein the yield of ammonia, then evaluate the dahl method requires specific apparati for the mass. To account for potential test sample data against that. Calculate quantitative distillation of ammonium sulfate. environmental contaminants, measure total nitrogen from the ammonia The Nessler reactions do not require a distillation the reagent blanks (without protein), and recovered, which indicates the mass of subtract that from the test sample data. protein present in the samples. As step. Test an ammonium sulfate reference above, use a reagent blank as an preparation of known concentration as a environmental control for ammonia. APPLICATIONS AND VALIDATION check solution to ensure accuracy of the Early in the product development cycle, simple assay. and rapid methods for estimating protein concen- tration can be used to establish purification the initial average yield value and process parameters. As product development discard any residue deviating more than 15%. Use progresses, more sophisticated concentration the remaining residues to recalculate a mean methods may be employed for protein characteri- value, which is considered the best-fit (most accu- zation. When used in release and stability rate) data indicating the protein amount recovered. programs, protein concentration methods should If the theoretical composition for a protein is be selected that are as accurate and precise, sim- unknown, you can use relative weight percent to ple and robust as possible. These features will as- calculate concentration (20). First, multiply the sure the method can be validated for its intended obtained amount of each residue by the molecular use, and can be conducted successfully by trained weight of that residue. Then add individual QC analysts. Part 2 of this article will residue weights to yield total residue weight, examine strategies for performing validation of which indicates the mass of protein recovered. the methods discussed here. You can also use this method to estimate amino acid composition if the molecular weight of the REFERENCES protein is known. Divide the mass of protein re- (1) C.J.R. Thorne, Techniques for Protein and Enzyme covered by the molecular weight to yield the Biochemistry (Elsevier/North Holland Biomedical moles of protein obtained. Then divide the Press, Amsterdam, The Netherlands, 1978), pp 1–18. (2) A.G. Gornall, C.J. Bardawill, and M.M.J. David, amount of each amino acid residue by the moles J. Biol. Chem. 177, 751–766 (1949). of protein for the number of amino acids in the (3) A. Hiller, Proc. Soc. Exp. Biol. Med. 24, 385–390 protein. (1927). (4) L.C. Mokrasch and R.W.J. McGilvery, J. Biol. Chem. 221, 909–917 (1956). NITROGEN DETERMINATION (5) R.M. Robson, D.E. Goll, and M.J. Temple, J. Anal. Total nitrogen determination is another protein Biochem. 24, 339–341 (1968). (6) M.D. Zishka and J.S. Nishimura, Anal. Biochem. 34, concentration measurement technique. Two simi- 291–297, (1970). lar methods are used: the Kjeldahl and the (7) O.H. Lowry et al., Biol. Chem. 93, 265–275 (1951). (8) E.F. Hartree, Anal. Biochem. 48, 422–427 (1972). Nessler assays (see box above). Both exploit the (9) G.L. Petersen, Methods Enzymol. 91, 95–121 (1983). observation that nearly all proteins contain (10) C.G. Vallejo and R. Lagunas, Anal. Biochem. 36, approximately 16% nitrogen (38). Although these 207–212 (1970). (11) H. Kuno and H.K. Kihara, Nature 215, 974–975 Corresponding author Nadine Ritter, methods can be quite accurate, they are not as (1967). is an independent biotechnology sensitive as the techniques described earlier, and (12) G.L. Petersen, Anal. Biochem. 100, 201–220 (1979). consultant, 13613 Anndyke Place, relatively large (mg) amounts of sample are (13) P.K. Smith et al., Anal. Biochem. 50, 76–85 (1985). Germantown, MD 20874, (14) M.M. Bradford, Anal. Biochem. 72, 248–254 (1976). required for analysis. 240.372.4898, (15) J. Pierce and C.H. Suelter, Anal. Biochem. 81, 478–480 (1977). [email protected]. Both assays are particularly useful with crude (16) T. Spector, Anal. Biochem. 86, 142–146 (1978). John McEntire is an independent mixtures of proteins. Because these analyses are pharmaceutical development based on total nitrogen content, they do not Continued on page 58 consultant. demonstrate the protein-to-protein variations of

22 BioPharm APRIL 2002 Protein Concentration continued from page 22

(17) C.M. Stoscheck, Anal. Biochem. 160, 301–305 (1987). (18) M.A. Phelan, J.W. Hall, and R.E. Mayner, Biotechniques 4, 130–136 (1986). (19) C.M. Stoscheck, Methods Enzymol. 182, 50–68 (1990). (20) G. Allen, Sequencing of Proteins and Peptides: Laboratory Techniques in Biochemistry and Molecular Biology Vol. 9, R.H. Burdon and P.H. van Knippenberg, Eds. (Elsevier, Amsterdam, The Netherlands, 1989), pp. 35–55. (21) D.W. Kupke and T.E. Dorrier, Methods Enzymol. 48, 155–162 (1978). (22) E. Layne, Methods Enzymol. 3, 447–454 (1957). (23) J. Smith, Techniques in Protein Chemistry, T. Hugli, Ed. (Academic Press, Inc., San Diego, 1989), pp. 251–254. (24) A. Galat, F. Bouet, and S. Riviere, Electrophoresis 16, 1095–1103 (1995). (25) D. Strydom and J. Crabb, Encyclopedia of Bioprocess Technology (John Wiley and Sons, New York, in press). (26) S. Shoji et al., J. Chromatogr. 354, 463–470 (1986). (27) F. Gharahdaghi et al., Techniques in Protein Chemistry III, R. Angeletti, Ed. (Academic Press, Inc., San Diego, 1992), pp. 249–260. (28) S. Cohen and D. Strydom, Anal. Biochem. 174, 1–16 (1988). (29) K. Yuskel et al., Techniques in Protein Chemistry VI, J. Crabb, Ed. (Academic Press, Inc., San Diego, 1995), pp. 185–192. (30) R.J. Simpson, M.R. Neuberger, and T.Y. Liu, J. Biol. Chem. 251, 1936–1940 (1976). (31) I. Molnar-Perl, J. Chromatogr. A. 499, 557–569 (1994) (32) D. Strydom and S. Cohen, Techniques in Protein Chemistry IV, R. Angeletti, Ed. (Academic Press, Inc., San Diego, 1992), pp. 299–307. (33) J.R. Benson and P.E. Hare, Proc. Natl. Acad. Sci. USA 72, 619–622 (1979). (34) S. Moore and W.H. Stein, J. Biol. Chem. 176, 367–388 (1948). (35) Pharmacopeial Forum 24, 5499–5516 (1998). (36) K.A. West and J.W. Crabb, Current Research in Protein Chemistry: Techniques, Structure and Function, J.J. Villafranca, Ed. (Academic Press, San Diego, 1990), pp. 37–61. (37) S. Cohen, S. ABRF News 6, p.11 (December 1995). (38) J.S. Garvey et al., Methods Immunol., 3rd Ed, (WA Benjamin, Inc., London, 1977) pp. 73–79. (39) P.L. Kiek, Anal. Chem., 22, 354–365 (1950). (40) F. Lanni et al., Proc. Soc. Ecp. Bio. Med. 74, 4–9 (1950). BP

58 BioPharm APRIL 2002 Info #54