1132 Residual Host Cell Protein Measurement in Biopharmaceuticals

1132 Residual Host Cell Protein Measurement in Biopharmaceuticals
1132 Residual Host Cell Protein Measurement in Biopharmaceuticals

Although safety concerns regarding residual DNA impurities are not as prominent as they once were, the levels of residual DNA in any bioprocess remain a key quality attribute and help define the process.

á1132? Residual Host Cell Protein Measurement in Biopharmaceuticals

1. INTRODUCTION AND SCOPE

Many medicinal products are produced through recombinant technology via a host cell (e.g., bacteria, yeast, or mammali-an, insect, or plant cell lines). During the manufacture of such products, some amount of non-product, host cell-derived mate-rial will inevitably be introduced into the process stream. This process results in a mixture of the desired product and host cell-derived impurities, including host cell proteins (HCPs), and other process-related impurities that will be targeted for clearance through bioprocessing.

Residual HCPs have the potential to affect product quality, safety, and efficacy; therefore, the quantity of HCPs should be low. The product purification processes must be optimized to consistently remove as many HCPs as feasible, with the goal of making the product as pure as possible.

The primary concern with HCPs in biopharmaceutical products is their potential to induce anti-HCP antibodies that could induce a clinical effect in patients. In addition, HCPs may possibly act as adjuvants, which can induce anti-drug antibodies that

can affect the safety or efficacy of the drug. A more extensive discussion of immunogenicity and its effect on preclinical and

clinical studies is described in USP general chapter Immunogenicity Assays—Design and Validation of Immunoassays to Detect An-

ti-Drug Antibodies á1106?. HCPs can also have a direct effect on the quality of the product itself. For example, proteolytic HCPs,even in minute quantities, can cleave the desired protein product over time, reducing or eliminating biological potency or al-tering stability.

This chapter focuses on HCP immunoassays for recombinant therapeutic products. It does not address products such as vac-cines or gene-, cell-, or tissue-based therapies, although the general principles discussed may apply to the measurement of

HCPs in these products. The design and validation of immunoassays for HCPs involve unique and significant challenges due to:

1) the wide variety of possible HCPs in medicinal products; 2) the general use of polyclonal antibody reagents to detect them;

3) the lack of exactly matched standards for quantitation; 4) in some cases, a considerable effect from sample dilution effects;and 5) inherent limitations to measure single HCP species.

The chapter includes assay development strategies throughout the product and process development lifecycle, and it de-scribes approaches to demonstrate that the assay is fit for use (e.g., illustrates unit operation clearance of HCPs, lot release).Because of the complexity of HCP immunoassays, careful development and characterization of critical reagents are required,particularly for the immunogen used to elicit the anti-HCP antibodies, the antibody reagent(s), and the assay HCP standard.Because HCP testing is an essential part of process development and product quality control, HCP testing is also discussed in conjunction with regulatory requirements and other considerations for guidance on an overall control strategy for HCPs. A brief outline of the general chapter follows:

1. Introduction and Scope

1.1 Considerations for Manufacturing, Characterization, and Consistency

2. Terminology

3. HCP Immunoassay Methods

3.1 The Assay Development Cycle

3.2 Development and Characterization of HCP Reagents

3.3 Immunoassay Method Development and Qualifying as Fit for Use

4. HCP Immunoassay Method Validation

4.1 Accuracy

4.2 Sensitivity and Assay Range

4.3 Sample Linearity

4.4 Specificity

5. Supporting Technologies for Residual HCP Detection, Identification, and Measurement

5.1 Considerations for Electrophoretic Methods

5.2 Considerations for Western Blot Methods

5.3 Considerations for Chromatographic and Proteomic Methods

5.4 Concluding Remarks on Supporting Technologies for HCPs

6. Use of HCP Immunoassays for Process Development, Characterization, and Validation

6.1 Assays for Individual HCPs

6.2. Control Strategy

7. Summary and Conclusions

8. Bibliography

1416 á1130? Nucleic Acid-Based Techniques / General Information USP 39

1.1 Considerations for Manufacturing, Characterization, and Consistency

Different cell-based expression systems are used to manufacture medicinal products, such as bacteria (Escherichia coli, Pseu-domonas fluorescens), yeast (Saccharomyces cerevisae, Pichia pastoris), mammalian cells (e.g., Chinese hamster ovary (CHO), mouse myeloma cell line NSO, and others), insect cells (baculovirus-infected Spodoptera frugiperda cells), and plant cells (to-bacco, Arabidopsis, rice). The particular HCP profile is unique and specific to the particular host cells under specific culture con-ditions and manufacturing processes. HCPs can vary in pI (~3–11) and hydrophobicity, and HCPs display a wide range of mo-lecular weights (from ~5kDa to at least ~250kDa), depending on the host cell and manufacturing process used. The number of HCPs in upstream samples can run anywhere from several hundred to more than one thousand proteins, depending on the host cell and culture conditions. Although many cellular hosts have been used in biopharmaceutical manufacturing historically, the most experience has been gained using E. coli and the mammalian cells CHO, NSO, SP2/0, and human embryonic kidney cell line HEK293. The guidance in this chapter draws most heavily from the experience with these expression systems; howev-er, general principles apply broadly to any host cell system.

In mammalian cells, the recombinant protein is typically secreted from the cells into the cell culture fluid (CCF), along with many of the HCPs. However, it has been observed that intracellular protein trafficking may not proceed in a normal fashion in production cultures. For example, proteins usually associated with intracellular organelles, such as lysosomes, may be found in the CCF of largely viable cell cultures, because the clones have been selected for maximum protein export. In addition, as some of the cells die, their soluble, intracellular proteins are released into the CCF. Some harvest operations also lyse cells; therefore, the resulting harvested CCF typically contains both secreted and intracellular HCPs. While this mixture of proteins incubates in the fermenter, additional changes in the HCP population may occur, for example, as the result of enzymatic activ-ity (e.g., proteinases or sialidases).

HCP assays provide important information about the composition of the material entering the downstream recovery process and how each purification step affects HCP clearance. In some cases, HCPs can even bind to, and co-purify with, certain prod-ucts. Process characterization and validation studies are needed to show which process steps remove HCPs and also to demon-strate the robustness of these steps for consistently removing HCPs. As such, HCP assays are an essential part of purification process development and help ensure manufacturing consistency. Lastly, reproducible and reliable HCP assays may be re-quired to measure residual HCPs remaining in the drug substance (DS) used to make drug product (DP) that is delivered to the patient. HCP levels should be measured in: 1) preclinical lots used in toxicology assessment, 2) all lots during clinical develop-ment, and 3) process validation samples from the final manufacturing process. After approval, HCP monitoring may be re-quired as an element of the control system. Subsequent sections of this chapter discuss in more detail the use of HCP assays in process validation and in a good manufacturing practices (GMP) control system.

2. TERMINOLOGY

To help establish a common nomenclature in the literature and with regulatory agencies, Table 1 lists common terms with their definitions (indicating how they are used in this chapter) in addition to synonyms that have been used historically. Note that the term “platform” indicates that the same set of standards and reagents is used within a company to test a variety of products made from the same type of expression system (e.g., CHO cells) grown under similar upstream conditions. In the case of platform HCP assays, the antibodies to HCP are obtained from animals immunized with HCP antigens generated from a common upstream process that is applicable to many products, even if the downstream purifications are different. This ap-proach allows the knowledge from prior products to be leveraged. Justification that an assay is suitable for a new product, using the same expression system and common upstream conditions, is therefore often relatively straightforward.

The HCP immunogen used to generate platform anti-HCP antibodies and used often as the assay calibration standard is, by design, comprised of a broad set of HCPs. In contrast, the qualifier “process-specific” indicates that the immunogen/standard has been prepared from a set of HCPs unique to a given process (either a unique upstream cell culture process or a unique downstream purification process). Process-specific assays are, therefore, limited in their utility, and each must be fully qualified for each process. Process-specific immunogens and calibration standards are, by intent, more narrow and specific to a given process. “Commercially available” assays produced by vendors are often derived from a combination of strains and harvest/ purification procedures, and these assays are intended to have a broad application; but these commercially available assays are not specifically designed for a given manufacturer’s proprietary cell line, and users do not have control over reagent availability and lot-to-lot consistency.

Table 1. HCP-associated Terminology

Term Definition Historical Synonyms Commercial-

ly available

Available to the public for commercial sale; typically a combination of upstream isolates

and corresponding antibodies made by the vendor and sold as reagents or kits.

Generic

a In some cases, both purifications are performed, typically the protein A/G first, then the HCP affinity.

b Although ppm has been used historically, it is not advised because this term is used to reflect mass per unit volume for other types of tests. It is recognized that ng is used conventionally as a value derived from interpolation from an HCP standard curve (in units of ng/mL), where the signal is reflective of antibody binding and, unlike the therapeuti

c protein concentration measurement, does not strictly reflect the mass of HCP that may be present.

USP 39General Information / á1132? Residual Host Cell Protein1417

Table 1. HCP-associated Terminology (Continued)

Term Definition Historical Synonyms

Platform The same set of an HCP standard and antibodies is developed with a company’s propriet-ary host cell strain and used broadly within a type (e.g., CHO) across several products

when the upstream conditions are similar.

Custom, in-house, proprietary

Upstream process specific An assay designed from material where the upstream culture process deviates significant-ly from the platform.This is generally before any purification and may be applied to more than one product if

these parameters are similar.

Custom, in-house, product-specific,proprietary

Downstream process specific An assay designed from materials where the downstream unit operations are used to en-rich the HCP population.This may be applied to more than one product if these parameters are similar. This is

rarely used today and is not recommended except for certain products with exceptional

downstream processing.

Custom, in-house, product-specific,proprietary

Assay for an individual HCP

An assay using a standard composed of an identified, single, known HCP and its specific antibody/antibodies.Single analyte assay or Custom,HCP-specific

Coverage Describes the assessment of how completely a population of polyclonal antibodies recog-nize the population of HCPs.

The coverage assessment may be made on the HCP population used as the HCP antigen

or from the product production culture.

Qualification Demonstration of suitability of analytical methods (including reagents used in these methods) for their intended application to a given process and in-process samples.

Protein A/G-affinity puri-

fication

Affinity purification of antibodies with immobilized Protein A or G a Affinity chromatography

HCP-affinity purification Affinity purification of antibodies using immobilized HCP (antigen)a Affinity chromatography, immunoaffinity chromatography

Null cell The cell strain used for production that does not contain the product-specific genetic ele-ments; includes untransfected parental cells and cells transfected with the expression

vector but without the product gene.

Parental, blank, or mock-transfected cell ng/mg The numerical quantity (ratio) of HCP per product, where ng represents HCP mass and mg represents the product mass.

It is calculated by dividing the HCP concentration (ng/mL) by the product protein con-

centration (mg/mL).

ppm b

a In some cases, both purifications are performed, typically the protein A/G first, then the HCP affinity.

b Although ppm has been used historically, it is not advised because this term is used to reflect mass per unit volume for other types of tests. It is recognized that ng is used conventionally as a value derived from interpolation from an HCP standard curve (in units of ng/mL), where the signal is reflective of antibody binding and, unlike the therapeuti

c protein concentration measurement, does not strictly reflect the mass of HCP that may be present.

3. HCP IMMUNOASSAY METHODS

Immunoassay methods rely on antibodies that recognize, as broadly as possible, the population of HCPs entering the down-stream purification process; therefore, the sandwich immunoassay, designed with polyclonal antibodies, is the workhorse of HCP monitoring and quantitation. This assay format offers a combination of high sensitivity, specificity, throughput, automa-tion potential, rapid turnaround, quantitative results, and low cost per assay that is unmatched by any other currently available assay technology. Other immunoassay formats (e.g., competitive immunoassays) may or may not be suitable, because they lack either the specificity or the sensitivity afforded by the sandwich format. Although these methods result in a single HCP value for a given lot, the number can give greater weight to HCPs for which high-affinity antibodies are present in the re-

agent(s)—and no or low weight to HCPs which are either not recognized or recognized by low-affinity antibodies in the assay.For these reasons, orthogonal measures of product purity are often needed. More details on these methods can be found in 5.Supporting Technologies for Residual HCP Detection, Identification, and Measurement .

The basic principles and design of immunoassays are discussed in USP chapter Immunological Test Methods—Enzyme-Linked Immunosorbent Assay (ELISA) á1103?. The format that is most commonly used for HCP testing is the sandwich immunoassay with detection systems such as colorimetric, electrochemiluminescent (ECL), chemiluminescent, radioactive, or others. Homo-geneous immunoassays, including competitive assays, where all of the reagents are combined at once and the binding occurs in a single step without washing, may be problematic due to antigen excess leading to antibody insufficiency issues (discussed later in the chapter); therefore, these formats should be used with caution. The heterogeneous sandwich immunoassay format described in chapter á1103? is generally preferred, because the dynamic range and sensitivity may be reduced in the homoge-neous format. The formats, with their advantages and disadvantages, are discussed further in chapter Immunological Test Meth-ods—General Considerations á1102?. Data analysis is typically performed with a nonlinear fit of the sigmoidal curve generated by a wide range of standard concentrations, although some analyses may focus on the low end of the curve for greater sensi-tivity.

1418 á1132? Residual Host Cell Protein / General Information USP 39

3.1 The Assay Development Cycle

Figure 1 illustrates common assay development plans, depending on the reagents available at various stages. Fewer bridging studies are required when: 1) platform reagents are available, and 2) upstream processes are historically consistent. Figure 1A illustrates a scenario where platform methods are not available, and a commercially available assay is used up to the stage of process validation with appropriate assay qualification. For phase III and beyond, either a platform or upstream, process-specif-ic method is preferred. A bridging study should be performed to support assay replacement. If the commercial assay is inten-ded for phase III and post-approval, care must be taken to fully demonstrate that the assay reagents are applicable to the proc-ess HCPs. An additional consideration is that the reagents are from an outside vendor over whom the biopharmaceutical man-

ufacturer has less control.

Figure 1A. If there is no platform assay before product development starts.

Figure 1B. If there is a platform assay before product development starts.

Figure 1B suggests that a platform assay can be used through all stages of product development if already available when product development starts. The platform assay should be qualified for each new product. Switching to an upstream, process-specific assay may be necessary for phase III or process validation and beyond, if the cell culture process is significantly changed from the platform process, and may introduce significantly different HCP populations. A bridging study should be performed to support assay replacement.

The limitations of a downstream-specific HCP assay should be considered, because it may be tempting to think that HCP assays are improved by taking the HCPs from null expression through the first column(s) and immunizing animals with a downstream column pool. Historically, the logic was that the immunogen and standard would be enriched in those HCPs most likely to enter the recovery process and be in the final product. This strategy was based on the assumption that, rather than having thousands of irrelevant proteins in the immunogen, only those most likely to be in the process will be present in the immunogen; thus, the resulting assay will be focused on those HCPs of greatest interest. Potential concerns that may arise as a result of this approach are:

1.If only HCPs from a null cell run pool from the first column are used, then later changes during process development

(e.g., changes in column run conditions) could invalidate the HCP assay. Thus, process development becomes very re-

stricted and involves the risk of needing to develop multiple HCP assays for slight process changes (or the need to man-age the uncertainty).

2.HCPs that co-purify with a product may do so because they bind directly or indirectly to the product protein. A null cell

run of a column without the product protein would miss these HCPs.

3.Nonspecific adsorption to chromatography resins is not uncommon and, often, is not a reproducible phenomenon. Com-

pared to the first passage, subsequent passage of the null cell run material over a column will likely produce a different set of HCPs after passage over a new column resin.

3.2 Development and Characterization of HCP Reagents

3.2.1PREPARATION OF ANTIGEN/HCP STANDARD

As described in 1.1 Considerations for Manufacturing, Characterization, and Consistency, the total HCP “antigen” is a complex population of proteins; therefore, when generating the HCP antigen/standard reagent, it is important to ensure that: 1) the calibration standard is representative of the cell line and manufacturing process, 2) its protein concentration is accurately quantified, and 3) the immunogen is administered in a way that generates polyclonal antibodies with reactivity to as many different HCPs as possible. The HCP antigen composition should also be comprehensive enough to tolerate normal process manufacturing changes during the life cycle of the product(s).

In addition to the uses above, the HCP antigen may also be needed to prepare the affinity column for purification of the antisera. If affinity purification is used, the quantity of HCP antigen needed should be carefully planned. The amount produced should be large enough to provide sufficient inventory for many years (often 10–20 years); ideally, for the whole life span of the product. However, the antigen preparation process should be performed and documented in a way that facilitates a po-

tential resupply.

USP 39General Information / á1132? Residual Host Cell Protein1419

Because the HCP assay is used to test DS samples that contain trace HCP impurities, any cross-reactivity of the anti-HCP anti-bodies with the product may compromise the test method and yield biased results. Therefore, any contamination of the HCP antigen with product must be avoided to prevent the generation of anti-product antibodies.

3.2.1.1 Preparation of HCP antigen from mammalian cells:The majority of biopharmaceutical products produced today are expressed in mammalian cells, e.g., CHO cells. A typical process for the preparation of CHO HCP antigen is outlined in Figure 2

.

Figure 2. Example of mammalian HCP antigen preparation.

The first step is to establish a null cell that does not express the product gene, either by using non-transfected (i.e., parental)cells with a common origin to those used to make product alone, or to transfect them with the vector used to create the pro-

duction cell line without the product coding gene (historically described as a “mock” transfection). The main advantage to the

latter is that selective markers (e.g., dihydrofolate reductase or glutamine synthetase) are expressed. Alternatively, antibodies

can be raised to selective markers independently. Another advantage is that mock-transfected null cells express selection mark-ers and may be grown in cell culture conditions more closely resembling the manufacturing process. Using pools of mock

transfected null lines which have not been cloned allows for the maximum potential genetic diversity and therefore gives the

highest potential of generating a broad host cell protein population. However, these cultures, which are seeded with cell pools

(not cloned), may demonstrate more variability from run to run. Both approaches [non-transfected (parental) and mock trans-fected] are commonly used. Both are also subject to the following issues: the cell culture process optimized for the production

clone may not lead to similar viability and cell density of the null cells. In addition, in the absence of the stresses experienced by cells producing large quantities of product protein, the null cell lines may exhibit a different HCP profile. These changes in viability, cellular metabolism, and cell density may alter the HCP profile of the antigen, making it less representative of the manufacturing process. More about choosing appropriate HCP antigen preparation conditions is discussed below.

Once the null cells have been established, the HCP antigen is prepared in a bioreactor, cell culture flask, or cell culture bag reflective of the product or process cell culture conditions. A platform cell culture process may be applied, which represents a common manufacturing process for several products (e.g., CHO-derived monoclonal antibodies). Typically, the resulting HCP antigen is minimally processed to ensure a broad HCP spectrum, making the antigen suitable for HCP monitoring of multiple products generated from the platform process. A platform HCP antigen also may be produced by combining several null cell runs with slightly different cell culture conditions (e.g., allowing the culture to remain in the bioreactor for longer periods of time to vary the cell viability). This approach is used to obtain a broader HCP spectrum, and antisera raised against this immu-nogen may be suitable for HCP detection in products generated from a variety of slightly different processes. These reagents also may increase the robustness of the HCP immunoassay toward process changes.

Alternatively, the HCP antigen may be prepared using an upstream, process-specific approach in which the cell culture proc-ess is tailored to mimic the production process for a unique product or specific process. The advantage of this approach may be the high relevance of the resulting HCP antigen to a particular upstream process. As with platform reagents, because no additional processing purification steps are included, the antigen contains a broad range of HCPs, and the antigen will often remain suitable for HCP monitoring after downstream production-process changes. The disadvantage of this approach is that the use of the reagents typically will be limited to a single (or few) product(s) or process(es).

The HCP antigen can be produced from a representative small scale, pilot scale, or production scale run. There are pros and cons for each approach. Pilot or production scale may mimic the actual process best; however, normal variation between cell culture runs may not be reflected if only one run at large scale is applied. Conversely, several small-scale preparations can be pooled and may better reflect the run-to-run variability of the cell culture process. Regardless of the scale, the presence of product should be aggressively avoided, because its presence in the HCP immunogen will compromise the quality of the re-sulting immunoassay antisera.

After performing the null cell run, the HCP antigen can be prepared from the cell lysate, the concentrated CCF, or as a mix-ture of both as shown in Figure 2. If the antigen is prepared from CCF, the cells may be harvested at a time when production line harvest is performed or some days later, to allow for some of the cells to lyse and release HCPs into CCF and thereby broaden the HCP spectrum.

To prepare immunogen from cell lysate, the cells are harvested by centrifugation, and the cell pellets are washed and lysed (e.g., by using repeated cycles of freeze/thaw, or high pressure homogenization). Conditions are typically selected to mimic

the production process (i.e., to prepare immunogen from CCF, cells and debris are removed by centrifugation, and the CCF is

1420 á1132? Residual Host Cell Protein / General Information USP 39

then treated by ultrafiltration/diafiltration). The buffer is exchanged (e.g., into PBS or HEPES), and the CCF is concentrated. The cut-off for filtration should be chosen to minimize loss of HCP, e.g., 10kDa or less.

3.2.1.2 Preparation of HCP antigen from bacterial cells:Many of the above-described principles used in the preparation of mammalian HCP antigens also apply to prokaryotic cell culture. However, some unique challenges should be considered. In particular, manufacturing processes for bacterial expression system come in more varieties, such as those yielding concentra-ted protein deposits known as inclusion bodies, which can represent up to 95% of the total cell protein. HCP impurities are also present in the inclusion bodies, including bacterial membrane proteins, ribosomal subunit proteins, and cytoplasmic pro-teins, such as small heat shock proteins or chaperones. The formation of inclusion bodies in the production strain makes it more difficult to generate a representative HCP preparation from a null cell fermentation process. The null cell may not have the same HCP profile, and it will not generate inclusion bodies with which specific HCPs may co-purify, as described above. In other cases, HCPs have been created from periplasmic secretion systems, if the production process can be replicated in the null cell.

Because there is no obvious approach for overcoming these limitations, in practice the bacterial HCP antigen is usually gen-erated from the lysates of washed cells, using null cell fermentations, that are grown and induced under conditions represent-ing the upstream manufacturing process as closely as possible. As for any HCP antigen, it is necessary to demonstrate that the HCP profile is representative of the manufacturing process before use in assay development. This approach yields the full com-plement of HCPs expressed and hence is very broad.

Because most animals have been exposed to bacterial antigens and may have pre-existing antibodies to many bacterial cell proteins, it is important to characterize the pre-existing antibody responses, and if appropriate, consider the use of specific pathogen-free (SPF) animals. The presence of significant levels of pre-existing anti-HCP antibodies may confound the analysis of the antibody responses; therefore, it is important to screen preimmune sera from animals for pre-existing antibodies prior to immunization.

Null bacterial cells transfected with the expression vector contain chaperone genes that may be expressed at high levels (e.g., 5% of the total HCPs). Such overexpression of particular HCP impurities might necessitate the development of single-analyte assays for these particular impurities (see 6.1 Assays for Individual HCPs).

3.2.1.3 Characterization of the HCP antigen:Several analyses are recommended before immunization: 1) protein content (total protein assay); 2) absence of product (as shown by Western blotting, immunoassay, and/or MS analysis); and 3) charac-terization by 1-D or 2-D-polyacrylamide gel electrophoresis (PAGE). Protein concentration is measured to establish the stand-ard concentration for future use and to determine the overall amount of the prepared antigen. The most commonly used methods are the bicinchoninic assay (BCA), the Bradford assay, and amino acid analysis (AAA), although the colorimetric methods require a standard (e.g., BSA) that is not well matched to the (HCP) analyte. Absorbance at 280 nm (A280) may also be used although it is less specific for protein (e.g., nucleic acids will also be measured). These methodologies often provide similar results, but some methods may be more significantly affected by the presence of interfering substances than others. One should consider using two orthogonal methods to exclude a gross over- or underestimation of the protein concentration by one particular method. For more information, see general chapter Biotechnology-Derived Articles—Total Protein Assay á1057?, which discusses the advantages and disadvantages of various protein analysis methods. The HCP concentration must be as-signed with a scientifically sound approach that is used consistently for its lifetime. The lack of product can be assessed with gels, anti-product Western blots, or other suitable methods. Lastly, the 1-D and 2-D gels help characterize the pattern of HCPs, show that a broad spectrum of proteins is present, and show that a reasonable match to production is present. However, this can be challenging because of the presence of large amounts of the product, which can obscure HCP detection. Comparisons may be made between the specific populations of host proteins produced in the null cultures and production cultures using the orthogonal methods discussed in 5. Supporting Technologies for Residual HCP Detection, Identification, and Measurement. 3.2.1.4 HCP standard reference reagents:Frozen HCP material is usually stable for a very long time, but stability of the HCP standard reagents should be monitored over time because the assay lifetime can be very long. HCP standard stability can be confirmed by monitoring the HCP standard performance in the HCP immunoassay as well as in orthogonal methods. Appropriate controls within the assay range may be established to monitor assay performance. Controls may be prepared from independent dilutions of the HCP standard, product samples or intermediate pools, or spiked product samples. However, there is a slight risk that control samples prepared as a dilution from the same HCP standard material can degrade at the same rate as undiluted HCP standard, and degradation of the standard will not be detected. To mitigate the risk, data from several standard curve parameters (e.g., signal, background, slope, coefficient of determination) are often assessed for each assay and used to support HCP standard stability. Using material different from the reference to prepare controls will help ensure that the degradation rate will be independent, and HCP standard degradation can be detected easily.

Assay control charts can be established and used to record reference curve parameters, control sample values, and lot-specif-ic information for the critical reagents, such as labeled HCP antibodies. Acceptable ranges for the controls should be establish-ed on the basis of multiple runs (usually 20–30) and may be used as part of routine assay acceptance criteria. In addition, the values for % coefficient of variation (CV) of standard and control replicates are often a part of the assay acceptance criteria. Differences over time in HCP standard curve performance or HCP control levels, or an increase in assay variability, may indicate

a lack of HCP standard stability.

USP 39General Information / á1132? Residual Host Cell Protein1421

3.2.2 PREPARATION AND CHARACTERIZATION OF ANTIBODIES

3.2.2.1 Preparation of antibodies to HCPs:The anti-HCP antibodies prepared are often used for many applications (West-ern blots, immunoassay) over a long period of time (e.g. the anticipated life of a product or platform). Therefore, material

needs should not be underestimated. The recommended antisera quantity will depend on many factors including the intended use, the predicted quantity purified from a given volume of antisera, the amount coated on microtiter plates, and many other considerations. The choice of animal species used to generate anti-HCP antibodies is driven by the immunogen, the require-ments for the particular method, and the preferences of the individual investigator. Rabbits, sheep, and goats are often chosen for immunization programs. In some cases, multiple animal species have been used. In such cases, using species more phylo-genetically distant from mammals (e.g., chickens) have helped to generate anti-HCP antibodies to conserved mammalian HCP proteins. The choice of animal species may also be driven by a desire to either increase the amount of antisera available from one or several animals (goats or sheep), or to use a strategy to obtain a greater diversity of responses provided by the use of many more individual animals from a single species (e.g., rabbits). The number of animals chosen to generate the antiserum pool(s) depends on the species used and the duration of the immunization protocol. If smaller animals are used to generate the antiserum, more individual animals are generally needed (e.g., 10–20 rabbits). In contrast, if larger animals (goats or sheep) are used, investigators usually will immunize 3–10 animals per protocol. Typical immunization protocols provide ap-proximately 100–150 mL of antiserum per rabbit or approximately 500 mL of antiserum per goat, and the total yield of Pro-tein A or Protein G purified antibody is approximately 1gram per rabbit or 5grams per goat. Animals should be screened for pre-existing antibodies against drug substance prior to immunization. If positive results are found, then these animals should be excluded from immunization.

A portion of the prepared HCP antigen is mixed with an appropriate adjuvant (most commonly, Freund’s adjuvant or in combination with incomplete adjuvant) and used for the immunization of animals. Each animal receives an initial priming im-munization, followed by multiple (often 4 to 8) booster immunizations, to mature the immune response and generate high-titer, high-affinity antibody responses. Antiserum is collected from each animal before the initial priming immunization (time

0) and at appropriate intervals (usually 10–14 days) after booster doses. Four to six bleeds are usually collected from each ani-

mal. However, the duration of the immunization protocol may be extended by increasing the number of booster immuniza-

tions, making it possible to collect additional high-titer antisera. Other immunization strategies may also be helpful (e.g., cas-cade immunization or size fractionation of the HCP immunogen).

Investigators should perform titer or Western blot analyses of the individual test bleeds before antiserum pooling to screen for and remove sera that 1) have low titer, 2) display an immunodominant immune response to a small group of HCPs, or 3)have nonspecific binding characteristics or anti-product reactivity. This evaluation should also be performed on the final anti-body pools to demonstrate broad coverage of HCPs and a lack of binding to the therapeutic product. In addition, some labo-ratories have found it useful to evaluate antibodies for aggregates using size exclusion chromatography as described below.Acid elution conditions may cause some antibodies to aggregate, and multimers of this type, particularly when labeled with either biotin or horseradish peroxidase, may lead to increased assay background. In addition, some laboratories include a final process-scale size-exclusion chromatography step to reduce aggregate levels to low levels (e.g., <5%).

Multiple approaches to the purification of antibodies from antiserum pools have successfully generated antibodies suitable for use in HCP immunoassays. Antibody purification is often performed by Protein A, Protein G, or HCP column affinity chro-matography. The application of coated magnetic beads can also provide benefits in some scenarios. The choice of Protein A or Protein G is driven by which animal species was used to generate the antibody. Manufacturers provide standard protocols for these routine antibody purifications. When affinity purifying anti-HCP antibodies, materials such as chromatography resins,which may have been used previously for other products, should be avoided, and additional cleaning cycles should be in place to minimize carryover. Ideally, manufacturers should use a resin that has never been exposed to a DP. Before the affinity purifi-cation step, some laboratories also include an initial 50% ammonium sulfate precipitation of the crude antiserum to enrich and concentrate the antibody fraction and thereby extend the performance and duration of use of affinity columns. The use of Protein A or Protein G purification strategies generates reagents in which the anti-HCP-specific antibodies compose a smaller proportion of the total antibody. In contrast, HCP affinity purification strategies selectively purify anti-HCP-specific antibodies from the antisera or antibody-enriched preparations. In this case, the HCP antigens are coupled to an activated resin, and the antiserum is purified over the column following established protocols or the resin manufacturer’s instructions. The purified an-tibodies may be further purified by size-exclusion chromatography to remove aggregates, then dialyzed, concentrated, divided into aliquots, and stored frozen (typically ?70° or colder). Neat antisera should also be stored frozen (see á1106? for additional information regarding storage of serum samples).

Affinity purification of specific antibodies using HCP immobilized on a column requires careful management of the column preparation and use so that it can be reproduced reliably for future purifications. In addition, if reused, appropriate resin stor-age and regeneration procedures should be evaluated and controlled to avoid degradation during storage. Done properly, it is a reliable, reproducible process that has led to consistent anti-HCP antibodies and HCP immunoassays. After loading the col-umn with antisera, an optimized wash procedure is critical. Because the preparation is a mixture of antibodies with differing affinities, extensive washing may remove low-affinity antibodies. Similarly, loading large amounts of anti-HCP antisera onto the column may result in high-affinity antibodies, displacing low-affinity antibodies if they recognize epitopes that are in close proximity. In either case, the low-affinity antibodies may be removed. In theory, higher affinity antibodies may also be more

challenging to elute. Whatever load and wash conditions are initially selected for the antigen-specific affinity purification, these

1422 á1132? Residual Host Cell Protein / General Information USP 39

conditions need to be maintained in future production lots of the antibodies. A comparison between these two purification strategies is shown in Table 2. Both approaches are commonly used.

Table 2. Antibody Purification Strategies

Type of Purification Pros Cons

Protein A or G Affinity Column Chromatography Robust and reproducible for quickly separating

IgG fraction from other serum proteins

Lower proportion of antibodies specific to HCPs

and may have a higher amount of low affinity

antibodies

HCP Affinity Column Chromatography Enriches for specific high affinity anti-HCP anti-

bodies; excludes nonspecific antibodies and se-

rum proteins

More skill and documentation required to make

and maintain resins consistently. Some high af-

finity anti-HCP antibodies may not be eluted in

usable form and may not be recovered

3.2.2.2 Characterization of antibodies to HCPs:Antibodies prepared for capture and detection are assessed independently and as part of the sandwich immunoassay pair. The concentration of the unmodified antibody is determined most commonly by absorbance at 280 nm, AAA, or BCA. Each approach is acceptable, provided it is applied consistently and (if colorimetric) is standardized similarly. It is important to determine the concentration of the capture and detection antibodies in each batch because they will be diluted to a certain concentration in the optimized immunoassay method. The detection and capture an-tibodies are characterized by their performance in the immunoassay method(s). The best label to antibody conjugation ratio should also be empirically evaluated primarily based on the best performance in the assay. This optimal ratio should be targe-ted in the future when new labeled antibodies are made. Finally, although antibodies are generally very stable when stored frozen (e.g., ?70°), investigators should ensure storage integrity and determine a defined period of use. This can be accom-plished by continuous monitoring with trending charts or by requalification experiments at specified time intervals.

The quality of the antibody pair (capture and detector) is often evaluated in two ways:

1.Show that the antibody pairs are specific and sensitive in an immunoassay format for the HCPs present in a series of sam-

ples taken from the unit operations of a given purification scheme, including the DS. This topic is also addressed in more detail in

4. HCP Immunoassay Method Validation. In concept, the ability to detect and measure HCPs (immunoreactivity) in

a series of actual process samples from a given process for a product in development is demonstrated with data showing:

1) sequential clearance as the product is purified, 2) sample linearity throughout a dilution series, and 3) lack of cross-

reactivity to product or matrix. The reduction of HCPs during the purification process is determined by clearance factors where the HCP content (in ng/mg) at each step in the process is divided by the amount in the prior step. Typically, start-ing samples from the cell culture may have HCP levels ranging from several hundred thousand to several million ng/mg, and final products may have HCP levels ranging from <1 to 100 ng/mg (showing many logs of clearance). When these trends are observed, they suggest that the process and the assay are functioning effectively, and the antibodies used in the immunoassay have suitable quality attributes. If these trends are not observed, it may not necessarily reflect a problem with the antibodies, because it is also possible that the purification process is not effective.

2.Show that a broad range of HCPs in the calibration standard is recognized (i.e., that the coverage of the HCP population

is adequate). Coverage is evaluated for at least the capture antibodies using 2-D gel Western blots or immunoaffinity frac-tionation of the total HCP population by immobilizing the anti-HCP antibodies on a column. In addition it is recommen-ded to test the coverage of the detection antibody if different coating and detection antibodies are used. The benefits and limitations of using either 2-D gels or immunoaffinity fractionation to demonstrate coverage is discussed more fully below.

Whichever method is used, the extent of coverage must be addressed in qualifying the immunochemical reagents.

HCP coverage evaluations help assess the ability of the antibodies to recognize a wide range of HCPs in the calibration standard and those present in in-process and DS samples. Two methods (2-D gels followed by Western blot analysis and im-munoaffinity purification followed by 2-D gel analysis) are in fairly common usage, and both have the limitation that they tend to underestimate the true antigen binding in immunoassay methods but may have value in comparing two preparations head-to-head (or a platform to a commercial reagent). Both procedures use reducing 2-D gel electrophoresis to separate HCPs in the HCP standard or in early process sample(s) (e.g., harvest). Numerical coverage comparisons should be used with caution because of the many method variables and the art required to reproduce results, even with the same reagents in the same laboratory. Because of this variability, the results are best evaluated qualitatively. Comparisons of batches (or sources) of anti-bodies should be done side by side as much as possible to determine if antibody lots are comparable or if one is superior to another. One approach, the SDS-PAGE/Western, is essentially a comparison of the number of spots present in the immunologi-cally stained membrane versus the number in a duplicate gel (or blot) stained for total protein (e.g., by fluorescent dye or silver; see also USP chapter Immunological Test Methods—Immunoblot Analysis á1104?). Differential staining of each antibody preparation may power the analysis, because it can be used to analyze the same 2-D gel. The second approach, immunoaffini-ty binding/SDS-PAGE, involves comparing the flow through and eluate to the load from the HCP calibration standard (or early process sample) passed over a resin to which the capture antibodies have been covalently immobilized. The resin is washed after loading and before elution to remove nonspecifically bound HCPs. For the purpose of sample comparison, difference gel electrophoresis (DIGE) technology could be helpful. Table 3 lists the advantages and disadvantages of these two coverage methods.

USP 39General Information / á1132? Residual Host Cell Protein1423

Table 3. Comparison of Coverage Methods

Pros Cons

2-D SDS-PAGE/Western blot Good separation of individual HCPs allows for individual spot counting.

HCP antigens are denatured, may not represent what is seen in immunological assays (e.g., ELISA).

High variability—numerical “percent coverage” values vary

widely with the same material tested within a single lab

and between different laboratories.

Transfer efficiency of a broad range of HCPs difficult to op-

timize, leading to underestimates due to over-transfer

through the membrane or failure to transfer from the gel,

dependent on molecular weight.

Matching spots between blots and gels are difficult and

not standardized.

Immunoaffinity binding/1- or 2-D SDS-PAGE HCPs bind to antibody resin in solution under native con-ditions similar to the sandwich immunoassay.May underrepresent some HCPs if they are bound too

tightly and do not elute, resulting in an underestimation

of coverage.

Analysis of spots in gels does not require immunoblotting and is simpler because the problems of transfer are avoi-ded.Preparation of the anti-HCP resin must be done carefully and may be difficult to reproduce. Results can be de-

pendent on resin loading and elution conditions.

Proteins at low concentration that only show up in West-

erns will not be detected.

More information about the appropriate use of SDS-PAGE/Western blot methods and ways to minimize the disadvantages

above can be found in the later section 5.2 Considerations for Western Blot Methods .

An example of a suitable use of this approach is shown in Figure 3.Figure 3. Left panel: 2-D IEF/SDS-PAGE analysis of representative CHO HCP calibration standard stained with a sensitive fluo-

rescent stain. Right panel: Western blot analysis of the same gel shown in the left panel.

3.2.3GENERATION AND QUALIFICATION OF REPLACEMENT REAGENTS

When the supply of HCP reagents are depleted or if their performance declines, new reagents may be needed. Often, new HCP antigens and new HCP antibodies are both generated at the same time. In theory, the new set of reagents should be created by following the same protocol that was used for the previous set of reagents to match the performance of the new and old assays as closely as possible and produce consistent data for a given program. In practice, however, it is not always possible (nor advisable) to generate the assay reagents exactly the same way that they were originally made. For example, if the manufacturing process has been optimized over time and the original platform assay reagents are no longer relevant, a careful assessment should be made. When such changes are required, the ideal way to confirm the quality of the replaced reagents is through characterization of antibodies (see below) and demonstrating that they can detect similar or greater HCP log reduction, from harvest to DS, using the same samples tested head to head with the original assay reagents.

When replacing an HCP antigen in which the current production process is essentially the same as the old process, the new HCP antigen should be prepared as similarly to the old as possible. It is important to compare the protein concentrations of the old and new HCP antigens side by side, using the same method to correctly assign the protein concentration for the new

1424 á1132? Residual Host Cell Protein / General Information USP 39

antigen and to ensure that the protein concentrations are comparable in the new and old HCP calibration standard. Character-ization by 1-D and 2-D SDS-PAGE should also be performed as a side-by-side comparison of the old and new HCP antigens (see above) to be sure that gross changes in protein composition have not occurred.

For antibody replacement, the same animal species should be used, if possible. In cases where a better antibody response is desired, antibodies produced by different species may be evaluated. The selection of the species should take into account all the points discussed in this chapter. A side-by-side comparison of results from process samples using old and new antibodies in a sandwich immunoassay format should be included to demonstrate suitability of the replacement antibodies. Additional char-acterization by orthogonal methods (e.g., coverage using 1-D and 2-D SDS-PAGE) is recommended, as described above. Assay qualification or validation should also be considered after changing reagents.

To fully understand the effect of replacing HCP critical reagents on assay results requires testing of process samples in bridg-ing studies using multiple samples from harvest, process intermediates from all appropriate purification steps, and the final DS. These studies can be very resource intensive, which underscores the importance of making sufficient quantities of HCP critical reagents at the outset (and minimizing replacement).

3.3 Immunoassay Method Development and Qualifying as Fit for Use

As noted in 1. Introduction and Scope, HCP immunoassays serve two important functions in the development and control of biopharmaceutical manufacturing processes. HCP immunoassays serve as a measure of product purity, and therefore are po-tentially related to patient safety. They also serve as a measure of manufacturing consistency, and therefore they are reflective of process control. To meet these requirements, HCP assays are often validated and incorporated into the cGMP control sys-tem, which includes specific reject limits (in the DS as a Certificate of Analysis (C of A) method or as an in-process control). If the final DS is a conjugated protein, the testing should be done on the protein intermediate (before conjugation). In addition, HCP assays are used to guide process development and are often used to demonstrate robust process performance in the con-text of formalized process validation activities.

HCP immunoassays are often formatted as a sandwich ELISA, and multiple antibody labeling options are available (see

á1103?). Depending on the label chosen, appropriate conjugates and substrates are selected; examples are described in

á1103?. Most proteins can be biotinylated because of their lysine content and, because biotin is small, it is less likely to affect the protein’s binding activity. Because antibodies are large and have many lysine residues, the conjugation ratio of biotin to antibody can be higher than that of smaller proteins. However, it is important to not over- or under-label the antibody (e.g., with biotin or HRP), because this might lead to either less antigen binding or lower signal/sensitivity, respectively. Controlling and defining the labeling stoichiometry (e.g., mole of biotin to mole of IgG) is useful for making future batches consistently. Multiple ratios of any antibody label should be tested and optimized for performance in the immunoassay. Excess unconjuga-ted reagents should be removed, either by dialysis, affinity purification, or by using a suitable desalting column.

Often, the standard is first screened to see whether a dose-response curve can be generated, and if so, in which concentra-tion range of analyte. The results can also reveal a starting point for initial reagent concentrations that can be optimized fur-ther. If more than one antibody pair has been prepared, then each is tested to find the best performance (see USP chapter

á1103? for more information). The signal (low calibration standard) to noise (background) ratios generated with each antibody candidate can be used to select the best pair. Typically, HCP immunoassays do not always possess a full dose-response curve with both asymptotes; therefore, for the purposes of residual HCP detection, the assay for DS release should focus on the low end of the curve.

3.3.1QUALIFYING A NEW MANUFACTURING PROCESS WITH A PLATFORM HCP ASSAY

As noted earlier, if the upstream/isolation procedure is matched, then a new product produced in that system can benefit from a platform assay approach that can decrease qualification time and effort. However, in cases where there are variations that occur in optimizing the production cultures and the cell culture design space is not completely defined, it is important to understand whether these variations are significant enough to render the platform assay unsuitable. In these cases, the plat-form assay should be qualified experimentally to demonstrate that it is suitable for measuring HCPs from the new process. Assay accuracy, precision, sensitivity, linearity, range, and specificity must be evaluated (see 4. HCP Immunoassay Method Vali-dation). In addition, the range of HCPs in the platform HCP standard should be compared to those present in the new process. The antibodies should also be characterized using samples from the new process or product using approaches discussed previ-ously.

The following approaches and concepts may be useful in determining if a platform assay is suitable for a product made with

a new process:

1.Conduct a representative, small-scale null cell run grown under conditions used in the new culture process, and deter-

mine the protein concentration of the null cell run CCF. Assay the null cell run CCF at the same nominal concentration as the assay HCP standard using the HCP sandwich immunoassay, and compare the dilution curve from the null cell run CCF to the standard used in the assay. If the standard and new null cell run CCF curves are similar (e.g., in shape and ampli-tude, and the calculated HCP concentrations are within a factor of two), then the new process is considered not different

from the platform, and a process-specific HCP assay is not usually needed.

USP 39General Information / á1132? Residual Host Cell Protein1425

2.With the CCF from the above null cell run from the new process, make a comparison to the HCP calibration standard using 2-D gel electrophoresis. This will compare the diversity of proteins produced by the two processes. However, be aware that new “spots” may appear in the 2-D gels that may not reflect immunochemical differences in the assay, for example, post-translational modifications or limited proteolysis. As such, the appearance of a few new spots or up-regula-ted or down-regulated spots in the process-specific null cell run is not a basis for invalidating the application of the plat-form assay. A qualitative assessment (e.g., versus a Western blot) is recommended.

https://www.360docs.net/doc/2613514023.html,parison of HCP levels at harvest in actual production runs (CCF samples) with the same product produced in the original versus the “new” process is also valuable. In general, immunoassay results within a factor of two are often consid-ered similar, thereby confirming the platform HCP reagents.

4.HCPs in CCF from production cultures may also be estimated on the basis of total protein measurements. The total pro-tein in the CCF is the sum of product (measured using a product-specific titer assay) and HCPs. Samples from harvests of the manufacturing run cell culture can be dialyzed, as long as they are sufficiently concentrated to make membrane losses negligible, to remove small peptides and amino acids, and then the total protein is determined (e.g., by amino acid anal-ysis). Subtraction of the product concentration from the total protein provides an estimate of the non-product protein,HCP, in mg/mL. This value may be compared with the HCP concentration from the immunoassay. Comparable values (e.g., within 2×) indicate that the HCP platform is confirmed for the new process.

3.3.2 NEED FOR NEW REAGENTS AFTER PROCESS CHANGES

Major changes, such as substantial and unambiguous changes in cell culture conditions (e.g., moving from a serum-contain-

ing process to a serum-free production process), typically require production of new reagents and development and validation

of a new HCP assay. In this example, the HCP reagents created using the old process recognize a number of serum proteins

but may not recognize many new HCP proteins in the serum-free process. Because of both the process change and the assay change, different HCP levels will be observed when the same samples are measured for HCP in the old and new assays (i.e.,

the samples from the serum-containing process may have a high level of HCP measured by the old assay and low level of HCP

measured by the new assay, whereas the samples from the serum-free process may have much lower HCP results by the old

assay but more with the new assay).If the assay was developed as a downstream, process-specific assay, changes in the purification steps can also require replac-

ing the assay with a new one that is relevant to the new downstream process. This is the biggest problem with these very specialized assays and why platform-based assays are usually preferred, because they often do not require a new assay when the downstream steps are changed.

3.3.3. SAMPLE STABILITY

It is important to demonstrate that the sample handling procedures do not affect the assay results. This is particularly true when comparing results for various intermediate process pools. Occasionally, HCPs can precipitate during freezing or when the sample pH is adjusted from acidic to neutral. In addition, storage at 2°–8° or multiple freeze-thaws may result in loss of reactivity. The analyst may need to add specific stabilizing components (e.g., divalent cations) to the samples to ensure stabili-ty.

4. HCP IMMUNOASSAY METHOD VALIDATION

HCP immunoassays for product purity assessment typically are part of the cGMP control system, either as an in-process test or on the C of A, and need to be validated appropriately. The validation approach depends in part on the types of samples that will be tested with the method. All validation parameters for a quantitative impurity test are needed when used for the final DS, whereas validation for in-process samples usually focuses on dilution linearity, interference, and precision. Regardless of whether the assay is an in-process or release assay (DS), action limits are valuable as a part of an overall control strategy.Readers are referred to ICH Q2(R1) guidelines and chapter Validation of Compendial Procedures á1225? for general expectations for assay validation, particularly those requirements for cGMP assay validation, and to chapter á1103? for sandwich immunoas-says. Although the main focus of this section is on cGMP assay validation, many of the same principles apply to the assays used to demonstrate that process purification steps are suitable for their intended purpose.

This section will focus on those aspects of HCP immunoassays that are unique or require special attention and documenta-tion. HCP immunoassays differ from more conventional immunoassays in that they are multi-analyte assays. There are poten-tially thousands of proteins in the immunogen and standard, and a correspondingly diverse set of antibodies is produced, yet only upstream CCF will contain a diversity of proteins that approximate the HCP calibration standard. Intermediate pools and final products typically contain progressively fewer of the HCP proteins that are present in the standard. As discussed previous-ly, this is the aspect of accuracy that is compromised by HCP immunoassays, and why the mass unit ratios (ng of HCP per mg of DS) are not a literal “mass” relationship. However, below are common industry approaches that address these limitations as

much as possible.

1426 á1132? Residual Host Cell Protein / General Information USP 39

The validation summary below is for the final DS or a penultimate process step, whichever is chosen for the C of A method. It should be noted that for in-process testing, the focus is on method accuracy (spike recovery), dilution linearity, and preci-sion. In the case of accuracy, some samples will have more potential interfering substances (other process impurities or addi-tives) that may need to be confirmed as non-interfering (e.g., residual DNA, leached protein A, virus inactivators, chaotropes, and low pH buffers).

4.1 Accuracy

To assess HCP assay accuracy in the DS, at a minimum the analytical assay standard should be added to appropriate samples and accurate spike recovery demonstrated over the range of the assay. Matrix interference can come from buffer components and product, and the minimum dilution required for acceptable spike recovery (typically between 70% and 130%) needs to be determined for each sample type. Typically, spike recovery experiments are conducted with spikes using at least three and potentially as many as five different levels. Because samples are assayed at multiple dilutions, the investigator may spike and then dilute the sample, or dilute the sample and then spike the diluted samples. Spikes near the quantitation limit (QL) help to evaluate the accuracy and repeatability of the assay near the QL, which is where the measurement is often the most variable. A spike recovery of 50%–200% may be acceptable for a spike at or near the QL.

Although these are minimum requirements for assay validation, they should not be interpreted as demonstrating accuracy for any one specific HCP that may co-purify with the product. To accomplish that, comparison to a standard of that particular HCP species is needed; however, because this HCP is rarely known, this may not be possible. Tips for discerning these situa-tions are provided below in the dilution linearity section and in 5. Supporting Technologies for Residual HCP Detection, Identifica-tion, and Measurement.

4.2 Sensitivity and Assay Range

HCP sandwich immunoassays often achieve standard curve sensitivity in the low single digits of ng/mL. For sample protein concentrations where the test dilution is 10 mg/mL, this implies that a sensitivity of about 1 ng of HCP per mg of product is possible. However, this sensitivity may be difficult to achieve for products with lower protein concentrations. It is important to highlight that the reported HCP ratio (ng of residual HCP relative to mg of product) is not really the ratio of masses implied by the unit ng/mg but rather “immunological equivalents” per mg of product.

The first aspect of determining QL is typically determined experimentally for each product in a given sample buffer matrix in spike recovery studies. The minimum recommended dilution (MRD) for the DS should be established by spiking the HCP standard in a sample dilution series. For example, an HCP spike of 10 ng/mL added to each of a series of solutions containing undiluted (e.g., 10 mg/mL) protein and serially diluted DS. Those dilutions in which the spike recovery is 70%–130% (50%–150% is also commonly used for levels near the QL) are considered acceptable. Typically, a proposed DS MRD is tested further to ensure that spike recovery is consistently achieved.

For the second part of the QL determination, the DS is tested at the MRD (if the DS samples have high levels of detectable HCP, a formulation buffer may be used). The QL is generally determined by the analysis of spiked concentrations of HCP and by establishing the minimum level at which the HCP can be determined with acceptable accuracy and precision. This study is typically performed at least three times, preferably by different analysts or on different days. For example, a spike of 3 ng/mL of HCP in a product protein concentration of 5 mg/mL has a QL of 0.6 ng/mg or 3 ng/mL if spike recovery is achieved in, e.g., at least four of six tests or if mean spike recovery criteria are met.

Typically, assays are set up to measure the range from a few ng/mL to >100,000 ng/mL. This range allows the analyst to test a variety of samples at multiple dilutions (see discussion of 4.3 Sample Linearity) and allows for a practical assay that can ac-commodate in-process pools with highly differing HCP levels. For example, upstream samples may contain >100,000 ng/mL of HCPs and require large dilutions to obtain results in range of the standard curve.

For routine commercial DS manufacture where the protein product concentration is known and the process impurities are well understood, testing at a single dilution may be used for release. The results are reported as the ratio of measured HCP (ng/mL) to the product concentration (mg/mL) resulting in units of ng/mg. When the DS has undetectable levels the results are reported as “less than” the assay QL (ng/mL) divided by the product concentration (mg/mL; e.g., <0.6 ng/mg in the ex-ample above). Before setting this target concentration for testing, however, the dilution linearity of the samples should be well understood and a robust manufacturing process established. In the event that the level or species of HCPs vary run-to-run it may be necessary to test each sample at multiple dilutions (see below).

4.3 Sample Linearity

HCP assay nonlinearity is not uncommon for some samples and should be assessed for multiple batches of a given sample type (e.g., from several clinical DS lots or process validation batches) to ensure consistency and proper reporting of results. Because a single HCP (or a few individual HCPs) may co-purify with product, it is possible that particular HCP(s) will be present in excess of the available antibodies in the HCP immunoassay. This is possible in multi-analyte assays where only a limited sur-

face area is available on assay microtiter plates or beads, and because thousands of different anti-HCPs are needed, antibodies USP 39General Information / á1132? Residual Host Cell Protein1427

to any one HCP will, of necessity, be limited. Furthermore, the relative amount of antibody to each separate HCP is also not controlled, so the binding capacity for each separate HCP differs. For samples exhibiting this behavior, the analyst must dilute the sample into the range of the assay, i.e., to a point where nonlinear behavior is no longer observed because the HCP has been diluted to a concentration where it no longer exceeds the available antibody. In some cases, the sensitivity of the assay becomes limiting, and one never reaches a dilution where the assay result is independent of the sample dilution. In such cases,the highest HCP ratio value (corrected for sample dilution) within the validated assay range should be reported. A detailed example of how to handle nonlinear dilutions arising as a result of antibody insufficiency follows.

Table 4 and Figure 4 show data for three different samples tested in an ELISA that has a range of 1–100 ng/mL. In this exam-ple, all samples were diluted initially to 10 mg/mL (the MRD, where spike recovery had been previously confirmed), and then serially diluted using a twofold dilution series to below the assay QL of 1 ng/mL. Whereas Sample 1 (diamonds) dilutes linearly over the full range tested, both Sample 2 (squares) and Sample 3 (triangles) HCP results plateau at higher concentrations of the sample. Saturation of the antibodies reflects antigen excess, and determination of the HCP value is made by diluting the sam-ple into the range of linear dilution; visually estimated as <2 mg/mL for the squares and <1 mg/mL for the triangles. [N OTE —If large sample dilutions are required to get into the range of the HCP assay, consider making intermediate dilutions to limit dilu-tion-related errors.]

Table 4.a Raw Data for the Graph in Figure 4a

Product Sample 1Sample 2Sample 3

Conc.(mg/mL)HCP conc.(ng/mL)HCP ratio (ng/mg)% max ra-tio value HCP conc.(ng/mL)

HCP ratio (ng/mg)% max ra-tio value HCP conc.(ng/mL)HCP ratio (ng/mg)% max ra-tio value 10.00 (neat)49 (neat) 4.983%20 (neat) 2.0<20 3.2 (neat)0.322%

5.0028.5 5.796%1

6.5 3.354% 2.50.535%2.5012 4.881%10 4.066% 1.50.642%

1.257.4 5.9100%7.4 5.997% 1.10.983%0.63 3.1 5.084% 3.3 5.387%0.9 1.4100%

0.31 1.6 5.186% 1.9 6.1100%<1<6NA 0.16<1<6NA <1<6NA <1<6NA

Reported

HCP ratio

value with guide and n Guide 1 = 5.2 (n = 6);Guide 2 = 5.2 (n = 6)Guide 1 = 5.8 (n = 3);Guide 2 = 5.3 (n = 4)Guide 1 = 1.2 (n = 2);Guide 2 = 1.4 (n = 1)a Numerical data for three samples (also graphed in Figure 4) show differing degrees of saturation of the response at higher concentrations of product (and HCP impurity) tested. Two guides on how to interpret these data (guides 1 and 2) illustrate how the “linear range” of the assay is limited to the most dilute data points shown in Table 4 and Figure 4. Note—Samples are always diluted until the HCP concentration is below the QL of the assay (in this example the QL is 1ng/mL). Guide 1: All values within 20% of maximum value are averaged. Guide 2: Values are averaged as long as the CV of the values is <20%, removing less diluted samples first. A third guide might also be used that reports the highest value measured above the QL but the suitability of this approach should be very well demonstrated by method validation.

Figure 4. Twofold serial dilution of the three samples listed in Table 4. HCP concentration results are plotted against the

product concentration in the samples, which serves as a measure of sample dilution.

Because measurements near the QL may have higher variability and less reproducibility than measurements farther from the assay limit, usually (if possible) values within the sample linear HCP concentration response range are averaged. The procedure for which data points may be averaged must be specified in advance of running the assay and is typically documented as part of the test procedure. In the example shown in Table 4, two different guidelines are illustrated. Guide 1 averages all values within 20%–25% of the maximum HCP ratio value. Guide 2 averages values only if the CV for the resulting averaged values is <20%–25%. Although their data sets are not identical, both rules are justified, based on the argument that variations of 20%–25% are within the variation seen in assay replicates and reflect the underlying reproducibility of immunoassay methods. In this example, both methods give very similar results. In contrast, the third potential guide, which reports the highest value

1428 á1132? Residual Host Cell Protein / General Information USP 39

measured above the QL, would yield results that are at least 10% higher. In addition, the validity of the third guide would depend highly on the quality of the method development.

Homogeneous (one-step incubations without washes between reagent additions) sandwich immunoassays are more prone to antigen excess and resulting dilution nonlinearity problems and may exhibit “high-dose hook effects” in which samples with large amounts of antigen give inaccurately low readings, because the antigen excess prevents formation of the complete sandwich. Suitable use of this format should be confirmed experimentally for each sample type and could be acceptable if the samples are diluted to assay QL and lack of hook effects is confirmed.

Historically, some companies have used spike-recovery data produced in the validation of accuracy to demonstrate linearity. Although this demonstrates the linear dilution of the standard, it does not demonstrate the linear dilution of the HCPs in the samples. For the reasons of antigen excess discussed above, the linear dilution of the standard is a necessary condition but not sufficient to demonstrate linearity of the assay for different sample types.

Finally, although antibody insufficiency is generally believed to be the primary cause of a lack of dilution linearity, other po-tential causes are: 1) reactivity of the antibodies with product protein, 2) the presence of non-specific antibodies (“sticky” anti-bodies), 3) reactivity of the antibodies to non-HCP proteins present in samples (such as peptones or BSA), 4) HCP aggregation, 5) HCP standard dilution profile that is not representative of the in-process or final product HCP, and 6) sample matrix interfer-ence. Cross-reactivity and matrix interference are discussed below.

4.4 Specificity

Assessment of potential matrix interference in samples is addressed in section 4.1 Accuracy and is primarily used to establish the lack of formulation interference (in the DS) and other impurities that are normally present (for upstream process samples). Another specificity issue to evaluate is the potential cross-reactivity of the anti-HCP antibodies with the product itself. Although this is possible, antibodies to HCPs that cross-react with product epitopes are rarely observed.

In those cases where cross-reactivity is observed, more false-positive assay signals are risks that must be managed. If cross-reactivity is suspected, it should be carefully and scientifically established. In the vast majority of cases, signals in HCP immuno-assays are real, indicating the presence of HCP impurities. In particular, instances where the HCP(s) are noncovalently associ-ated with product, and they co-purify, may give the appearance of cross-reactivity. However, because of the tremendous varie-ty of antibodies in a polyclonal anti-HCP pool, when measuring residual HCP in the presence of excessive levels of product, some antibodies may still bind product, yielding a false-positive signal. In this case, it may be necessary to modify the assay or the anti-HCP reagent. If a platform HCP assay is used to test multiple batches (for the same program), and different levels of HCP between batches are observed, this is strong evidence that the HCP-positive signal is real and not due to cross-reactivity. Evidence for cross-reactivity is often seen in a product that has been purified in a sequence of steps and where the HCP levels are not reduced by typical purification processes. The HCP/product ratio (ng/mg) should be used so that changes in the HCP/product ratio during purification can be compared. First, Western blots of DS are usually probed with anti-HCP antibodies to determine if HCP(s) are noncovalently associated with product. If bands are detected that are unrelated to the product, this suggests that cross-reactivity is not occurring, and further process development may be required if lower HCP levels are sought.

Alternatively, if the anti-HCP is recognizing a product-associated band, this suggests cross-reactivity. Caution should be exer-cised, however, because the product is denatured (by SDS-PAGE), and binding may be to epitopes not accessible when in the native, solution phase (i.e., in the immunoassay). In this case, a positive Western blot could be an artifact (conversely, a nega-tive Western blot could also miss cross-reactivity, but this is less probable). Regardless, if cross-reactivity is suspected, it can be confirmed and corrected by either: 1) addition of a high-purity product (or related structures, such as human IgG from a dif-ferent source, for a monoclonal antibody) to block the cross-reacting antibodies in the assay, or 2) depletion of the anti-HCP in the reagents using a product-affinity column made from highly purified product. In either case, it is important to ensure that the antigen used to block or deplete the serum is itself free of residual HCPs. This information is available from the Western blots probed with anti-HCP. If HCP bands are present, this indicates that a higher-purity product may be needed and can be prepared with orthogonal, lab-scale purifications. Sometimes reversed-phase HPLC can be effective in making this material, al-though this is not useful during product production because of its denaturing properties.

5. SUPPORTING TECHNOLOGIES FOR RESIDUAL HCP DETECTION, IDENTIFICATION, AND

MEASUREMENT

Biopharmaceutical manufacturers need an integrated control system that offers assurance of overall product purity and man-ufacturing consistency. Typically, product purity is demonstrated by using a combination of methods, and this is very much the case for HCPs. In addition to the most common sandwich immunoassay format, other assay types provide valuable com-plementary information. Tables5, 6, and 7 provide overviews for commonly used methods for HCP detection, quantitation, and characterization. Table 5 discusses electrophoretic methods; Table 6 discusses Western blot methods; and Table 7 discusses chromatographic and proteomic methods. Gel electrophoresis and immunoblots are also discussed in 3.2.2.2 Characterization of antibodies to HCPs.

Orthogonal assay methods used to provide additional assurance of purity and the lack of HCPs may be used in any of three

modes: “detection”, “identification”, or “quantitation”. In the case of detection, orthogonal assays address the question, “Is USP 39General Information / á1132? Residual Host Cell Protein1429

there any protein other than a form of the product that the immunoassay may have missed?” If this is the case, as knowledge accumulates regarding the identity of the HCP(s), it becomes possible to perform a specific protein risk assessment and, if nec-essary, generate a standard and separate test for that particular HCP impurity. The orthogonal method may then be used in a “quantitative” mode by comparing the product sample with the appropriate HCP standard for the known HCP. For example, a quantitative HPLC-MS/MS method may be developed, based on monitoring peptides that ionize well from digests of the ana-lytical standard protein and the sample.

5.1 Considerations for Electrophoretic Methods

Three electrophoretic methods that are useful in impurity analysis are 1-D and 2-D SDS-PAGE and SDS-capillary electropho-resis using non-gel sieving (CE-SDS). In all these methods, excess sample is loaded to ensure detection of trace protein impuri-ties. The drawback is that excess product obscures impurities that migrate close to the product and cause interference. Howev-er, 1-D gels or CE-SDS may already be part of the GMP control system for monitoring product-related fragments; therefore,extending these methods for HCP monitoring does not involve additional testing. 2-D gels have been used to investigate the purity of the final product and to identify minor spots in the gels associated with either product variants or HCP impurities. The presence of a new band in the gel, or a new peak in the CE-SDS electropherogram, may represent either a new product-rela-ted substance/impurity or may reflect the presence of an HCP impurity (“process related”). In either case, an investigation is warranted. If not part of the routine control system, a 1-D gel is still an excellent method for extended characterization of im-portant lots, such as registration lots, and for manufacturing investigations. Gels and CE-SDS are useful for confirming the ab-sence of gross levels of HCPs (should they be present and missed in the immunoassay). Gels are typically stained with a high

sensitivity stain, such as a fluorophore or silver. Ideally, the stain should be compatible with in-gel proteolytic digestion and

subsequent analysis by mass spectrometry to identify the protein.

Proteins differ in their staining intensity with silver or fluorescent dyes; therefore, only semiquantitative estimates of the amount of protein in a band or spot are possible (e.g., glycosylated forms may not stain as intensely). Nonetheless, because

these methods do not rely on anti-HCP antibodies, they provide an important orthogonal measure of purity.

Some proteins may appear in gels as multiple bands or spots that result from post-translational modifications or proteolytic

clipping. To the extent that a single gene product is distributed to multiple locations in the gel, this further reduces the sensi-tivity of gels to detect impurities.

As discussed following Table 7 on proteomic methods, it is often possible to excise bands or spots from the gels. These ex-cised gel fragments may then be incubated with proteases to provide in situ digestion of the protein into peptides. These pep-tides may then be fractionated and sequenced using LC-MS/MS and compared with databases to identify the origin of the peptides as either product-related or HCPs.Table 5. Analytical and Characterization Electrophoretic Methods for HCP Testing

Method Use Advantages Disadvantages Approximate

Sensitivity for HCPs

and Ability to Quantify 1-D gel, either reduced or nonreduced, stained with silver or high-sensitivity flu-orescent dye.Screening a large number of samples for consistency and presence of nonprod-uct protein bands. May be used as part of GMP con-trol system.High resolution separation;provides approximate mo-lecular weight (MW) infor-mation; relatively simple to run. Side-by-side analyses of samples provide power-ful comparisons for manu-facturing https://www.360docs.net/doc/2613514023.html,rge excess of product pro-tein may obscure HCP bands. Different proteins show differences in stain-ing intensities so only

semi-quantitative. Limited

capacity of gel lanes for to-

tal protein load.

100 ng/mg.

Semiquantitative unless used with an analytical standard for a known HCP.

Large-format 2-D gel (e.g.,20 × 25 cm) stained with silver or a high-sensitivity fluorescent dye.Screening a small number of samples for consistency and presence of unknown protein spots. Not suitable for routine use in lot re-lease but may be used for characterization of specific lots or HCP standards.High-resolution separation of trace HCP impurities from product. Provides ap-proximate MW and pI in-formation on protein https://www.360docs.net/doc/2613514023.html,rge excess of product pro-tein may obscure HCP spots. Different proteins show differences in stain-ing intensities so only sem-iquantitative. Highly tech-

nique-dependent results

requiring skilled staff and

sophisticated equipment.

100 ng/mg of total protein load).

Semiquantitative unless used with an analytical

standard for a known HCP.

CE-SDS with laser-induced fluorescence (LIF) detec-tion. Fluorescently labeled samples are separated as either reduced or nonre-duced samples.Screening a large number of samples for consistency and presence of unknown protein peaks. May be used as part of GMP con-trol system.Rapid, high-resolution method to separate fluo-rescently labeled proteins.Sensitivity may not be as high as a 1-D gel with a highly sensitive stain. Fluo-rescent labels are not spe-cific for HCP proteins, so there is no distinction be-

tween HCP and product-

related peptides. Difficult

to extend to other meas-

ures, such as Western

blots, or to identify protein

peaks.

1000 ng/mg (0.1% of total protein load).

Semiquantitative unless used with an analytical standard for a known HCP.1430 á1132? Residual Host Cell Protein / General Information USP 39

5.2 Considerations for Western Blot Methods

The Western blot technique is complementary to the sandwich immunoassay for demonstrating product purity and for char-acterizing the immunochemical reagents used in the immunoassay (see 3.2.2.2 Characterization of antibodies to HCPs). When the anti-HCP antibody is used as a probe, it may be useful to: 1) detect HCPs that are noncovalently associated with the prod-uct and (potentially) missed in immunoassay, or 2) monitor an individual HCP. In other cases, a protein that reacts well in the immunoassay may not be detected in Western blot because a conformational epitope was lost following denaturation. Immu-noblots require only one antibody to a given protein, whereas sandwich immunoassays require two. Previous work has shown that high-affinity antibodies that react with a given HCP can be more sensitive than silver or fluorescent dye staining of gels.

1-D Western blots of final product provide a simple reliable orthogonal method for demonstrating product purity. Although the same antibody preparation may be used in both the immunoassay and Western blot, for the reasons noted above, the blot may provide additional information on trace proteins (and miss others), hence its orthogonal nature. Table 6 presents the ad-vantages and disadvantages of 1-D and 2-D Western blot methods for these purposes.

Below is a list of the major considerations for optimization of SDS-PAGE/Western methods:

?2-D-gel format and size: The larger formats (18-cm length or longer) resolve proteins better and come in a variety of iso-electric focusing (IEF) application modes and molecular weight dimensions (e.g., gradients). Total protein staining of vari-ous formats can be studied to optimize the separation in both dimensions and thereby find the balance that separates most (or the maximum number) of HCPs.

?The load on 2-D gels should be optimized to balance sensitivity needed (higher load) versus resolution desired (lower load). The protein load, ionic content of the sample, and speed of separation need to be balanced to avoid over-heating and to optimize resolution. The stains or substrates to be used will influence this choice. Typically, more is loaded for the immunoblot (e.g., 200 m g) than for the total protein-stained gel or blot (e.g., 50 m g for silver stains). The difference in load may confound the separation to some extent; therefore, finding the right balance will require experimentation.

?Signal development for both the total protein (gel or blot) and the Western blot requires significant experimentation

(e.g., dye types, exposure times, buffer conditions) to optimize. For example, because it is important to detect as many

bands as possible, sometimes the signal is amplified so much that the most abundant species obscure the others, or the background increases and contrast is lost. A related issue is standardization and calibration of the densitometer or imager, which must be fine-tuned to achieve consistency under experimentally determined conditions. Both reagent and instru-ment optimization can require significant time to reproducibly establish the optimized conditions where signal is maxi-mized and background is minimized (see also á1104?).

?The transfer and blocking reagents require experimentation to identify the best conditions (such as buffers, instrument power/time, temperature, and blockers).

Table 6. Analytical and Characterization Western Blot Methods for HCP Testing

Method Use Advantages Disadvantages

Approximate

Sensitivity and Ability

to Quantify HCPs

1-D gel, Western blot Screening a large number of

samples for consistency

and presence of unknown

protein bands that react

with anti-HCP antibodies.

May be used as part of

GMP control system.

High-resolution separation;

provides approximate MW

information; relatively sim-

ple to run. Side-by-side

analyses of samples pro-

vide powerful comparisons

of lots.

Requires high-quality anti-

HCP antibody source.

Large excess of product in

the gel can lead to nonspe-

cific staining of product

bands. SDS-induced dena-

turation of proteins leads

to loss of conformational

epitopes.

Not quantitative for HCPs

unless a specific protein

standard is used. Sensitivity

depends entirely on the

quality of the antibody.

Large-format 2-D gel (e.g.,

20 × 25 cm) Western Blot

with chemiluminescence

or similar high-sensitivity

detection.

Screening a small number

of samples for consistency

and presence of unknown

protein spots reacting with

HCP antibodies. Not suita-

ble for routine use but may

be used for characteriza-

tion of specific lots of

product or characterization

of antibody reagents.

High-resolution separation

of trace HCP impurities

from product. Provides ap-

proximate MW and pI in-

formation on protein

spots.

Requires high-quality anti-

HCP antibody source.

Large excess of product

protein may obscure HCP

spots. SDS-denaturation of

proteins leads to loss of

conformational epitopes.

Produces highly technique-

dependent results requir-

ing skilled staff and sophis-

ticated equipment.

Not quantitative for HCPs

unless a specific protein

standard is used.

Sensitivity depends entirely

on the quality of the anti-

body, but 100 ng/mg is

possible.

5.3 Considerations for Chromatographic and Proteomic Methods

Mass spectrometric techniques for the detection, identification, and quantitation of individual HCPs are rapidly evolving and leverage much of the technology developed for proteomics. Furthermore, as new genomic databases become available (e.g., the CHO genome), mass spectrometric approaches are more useful. These methods (see also Applications of Mass Spectrometry á1736? for additional information) often combine sample preparation such as reduction, alkylation, and proteolytic digestion, followed by separation (e.g., with reversed-phase chromatography RP-HPLC) before introduction into a mass spectrometer USP 39General Information / á1132? Residual Host Cell Protein1431

that fragments all proteins thus providing an amino acid sequence for each peptide. The resulting sequence information is compared to the product sequence to identify product-related fragments and to a database related to the host (e.g., E. coli ,CHO) to identify HCPs.

A challenge for MS analysis stems from the overwhelming number of product-derived peptides relative to impurity peptides.One approach to address this issue of competitive ionization of the peptides (also called ion suppression of the HCP peptides by the product peptides), is to apply LC-MS/MS analysis on partially resolved HCP preparations (e.g., HPLC fractions). This purification reduces the product contribution to total ions in the mass spectrometer. Other approaches are in development.Recent technological improvements such as 1) chromatography resins able to resolve effectively using MS-friendly mobile pha-ses, 2) improved interfaces to front end LC and/or IEF separation systems, and 3) mass spectrometers with higher mass resolu-tion, accuracy, and faster scan rates, now make it possible to identify and quantify specific HCPs in DS with a high degree of confidence. Major challenges in terms of sensitivity and quantitation of sufficiently large sets of heterogeneous HCPs, cost, and QC-related issues remain to be met before this technology can replace immunoassays for the control of HCPs in DS.

As a characterization method orthogonal to the HCP immunoassay, LC-MS/MS data may be used in two ways. First, if the MS-based method does not find HCPs in samples that were also below QL in the immunoassay, then these orthogonal techni-ques can increase confidence that the HCP ELISA did not miss an HCP that has co-purified. The detection limit for many LC-MS/MS methods is currently in the range of about 10 to 100 ng of HCP per mg of product. It is therefore sufficiently sensitive to rule out a single HCP being present at a high level and is more sensitive than gels. Second, if an impurity is identified at a high level or one that is a safety concern, then one can assess whether the purification process needs to be improved to re-move it, whether the current control system is adequate, and whether development of a specific assay is required. Absence of signal does not prove absence of HCPs, but once an HCP is identified, the sensitivity of LC-MS/MS can be enhanced. LC-

MS/MS is complementary to other HCP technologies and is increasingly valuable as an orthogonal method. Detailed discus-

sion of all the proteomic approaches to the analysis of HCPs is beyond the scope of this chapter, in part because the field is evolving so rapidly that any review would become out of date quickly.

Intact HCPs also may be separated using HPLC with UV detection. The profile has been used for comparing different lots of

null cell runs, for example. Readers are referred to Chromatography á621? for the core science behind HPLC chromatographic methods. Because HCPs differ in abundance, and the chromatography may not provide unique resolution of all proteins, an individual HCP must constitute about 1% (or less with good resolution) of the total protein population for possible detection

(even then, a given HCP may be obscured by product, depending on its retention time). Nonetheless, investigation of un-

known peaks as a general practice and comparisons of the chromatographic profiles of null cell run HCPs may be useful in determining the similarity of HCPs produced under different culture conditions.

As with the electrophoretic methods discussed in this chapter, reversed-phase separation of proteolytically cleaved product samples with UV detection may already be included in a GMP control system as part of product testing. The presence of new or unexpected peaks in the UV chromatogram may indicate the presence of an HCP impurity, although this method is not overly sensitive (~1% impurity if not obscured by other peaks). However, because no new testing or sample preparation is re-quired, this opportunity to use an orthogonal method for determining purity should be considered.Table 7. Analytical and Characterization Chromatographic and Proteomic Methods for HCP Testing

Method Use Advantages

Disadvantages Approximate

Sensitivity and Ability

to Quantify

Mass Spectrometry –HPLC/electrospray ioniza-tion (ESI)-MS/MS or cIEF/ESI-MS/MS or MALDI-TOF of protein digests Identification of individual HCPs. May be applied to the characterization of a purified product lot, or null cell run HCP standards.Allows identification and monitoring of individual HCPs. May be combined with gels to identify the in-dividual proteins seen in gels.Low-throughput method re-quiring significant sample preparation, skilled staff,and sophisticated instru-mentation.100 ng/mg and semi-quan-titative if used in the dis-covery mode for unknown HCPs. 10 ng/mg (or better may be possible) and quantitative if used with an

analytical standard for a

known HCP.

RP-HPLC/UV or RP-UHPLC/UV. May be ap-plied to intact proteins or proteolytic digests Suitable for comparing null cell run standards if pro-teins are well https://www.360docs.net/doc/2613514023.html,rger proteins may need limited proteolysis or re-duction to improve resolu-tion.High resolution, separates HCPs based on hydropho-bicity and size; direct com-parison of null strain and product strain is possible via overlays.Difficult to find a stationary phase and mobile phase combination able to re-solve and elute all HCPs.Co-elution may hinder

quantitation of specific

proteins. High concentra-

tion of product peak(s)

may interfere with detec-

tion of trace HCPs.

10,000 ng/mg (1%).Semi-quantitative unless used with an analytical standard for a known HCP.

5.4 Concluding Remarks on Supporting Technologies for HCPs

A number of various hybrid analytical techniques have been attempted over the years. One example is fractionation of HCP standards by ion exchange, followed by RP chromatography, and then an immunoassay or MS analysis (2-D MS) on the pro-teins in the final fractions. Alternatively, “product subtraction” has been evaluated to remove the bulk of the total protein from

1432 á1132? Residual Host Cell Protein / General Information USP 39

final product samples before gel electrophoresis of LC-MS/MS analysis. These hybrid methods for coverage determination are very labor intensive, and they may introduce artifacts or biases of their own (e.g., antibodies to product do not typically re-move all the myriad of heterogeneous forms). As such, they have not found widespread application, although in individual cases they may provide insight into the nature of product impurities.

Electrophoresis, Western blotting, and immunoassay remain the most widely used methods for HCP reagent characterization and for demonstration of HCP clearance by purification processes, because they are robust, sensitive, and can cover a broad range of protein impurities. Immunoassay and (increasingly) mass spectrometry are highly complementary and the most pow-erful methods for monitoring residual HCP levels in samples and confirming their absence in final DSs.

6. USE OF HCP IMMUNOASSAYS FOR PROCESS DEVELOPMENT, CHARACTERIZATION, AND

VALIDATION

As described previously, although HCP testing has limitations as quantitative assays, if the immunoassay can rapidly and ro-bustly detect most of the HCPs, then it is extremely valuable during process development, characterization, and validation. HCP clearance at each intermediate process step under a variety of conditions is valuable data. Application to process valida-tion requires that the assay be demonstrated to be scientifically sound, suitable for its intended purpose, and appropriately documented. As stated earlier, process pools can differ widely in terms of buffer components, product concentration, and HCP composition. At a minimum, acceptable spike recovery of the HCP standard into in-process pool samples is necessary to qualify test dilutions of the various pools in the assay. In addition, testing each sample type at multiple dilutions is helpful during de-velopment to verify that: 1) results are within the range of the assay, and 2) nonlinearity in sample dilution is considered. In cases where the process is shown to clear HCPs robustly, process validation may also be used to justify not having a routine HCP test as part of the cGMP control system.

Figure 5 illustrates data gathered during process development and validation and shows the dilution dependence for a num-ber of in-process pools (Pool 1 is after the first CCF purification step, Pool 2 the next, and so on) through the process. Figure 5 shows HCP ratio (in ng/mg) on the y-axis plotted against the product dilution in the test well on the x-axis. Each sample is serially diluted through three or four twofold dilutions. The starting dilution is determined by both the need to dilute samples with high levels of HCP into the range of the assay and the need to test only at dilutions where acceptable spike recovery has already been demonstrated. In Figure 5, the CCF has slightly more than 1 million ng of HCP per mg of product, meaning that a little less than one-half of the total protein in the CCF is product. The first column pool reduces the HCP level to just over 10,000 ng/mg. The second column has minimal HCP clearance; the third column reduces the HCP ratio to 1,000 ng/mg. The final column in the process reduces the HCP level by an additional 2 logs, resulting in a final HCP ratio of 10 ng/mg. All of the

intermediate pools show HCP ratio values that are independent of the dilution of the sample.

Figure 5. Example of HCP clearance by each purification step. Within a sample set, the first value is the most dilute in its

dilution series.

Figure 6 presents a set of data for a different recovery process. As in Figure 5, the HCP ratio data for a serial twofold dilution series of each in-process pool sample are plotted. For CCF and the column 1 and column 2 pools, the HCP ratio (ng/mg) is independent of sample dilution (dilution linearity is achieved as in Figure 5). But after processing over the third column, dis-tinct dilution dependence is apparent in the assay. This nonlinear dilution is likely associated with antigen excess relative to available antibody for the particular set of HCP(s) in the sample (i.e., a single or limited number of HCP species co-purify with the product). With product purification, more product must be put in the assay well for residual HCPs to be detectable in im-munoassay. The product concentration in the assay may be increased with each purification step, and thus there is a similar increase in the concentration of the co-purifying HCP impurity. By column 3, the amount of the impurity likely exceeds the capacity of the available antibodies. This phenomenon is repeated in the final pool, which involves no HCP clearance but only buffer exchange by UF/DF. The reported value for these pools is the one obtained by diluting the sample to near the QL of the assay (the most dilute sample in the series) using a method described in Table 4, which is about 300 ng/mg in this case. Addi-tional discussion of assay nonlinearity is found in the 4.3 Sample Linearity subsection of 4. HCP Immunoassay Method Validation

. USP 39General Information / á1132? Residual Host Cell Protein1433

Figure 6. Example of HCP clearance with a purification process. Within a sample set, the first value is the most dilute in its

dilution series.

6.1 Assays for Individual HCPs

In some cases, a manufacturing process might result in production of relatively high levels of a single HCP that can result in

a bias in the reported results. As discussed above, in this situation the concentration of antibodies present in the total HCP

assay may not be sufficient to capture all of that particular HCP present. In such cases, where a single, known HCP is present,

another assay for that single HCP may be needed. For example, in the case of E. coli production cultures, co-expressing chaper-ones or other processing enzymes that aid in folding or disulfide formation can be produced in very high levels. In addition,these chaperones may be present in the immunogen used to develop the HCP assay reagents, in a conformation that is differ-

ent from the way the protein is presented in samples. Thus, antibodies produced to chaperones not bound to product may not recognize, or may poorly recognize, product-bound HCPs. In this case, separate assays using appropriate antibodies may

need to be developed. The existence of high levels of a single HCP is not unique to bacterial cell cultures and is possible with any cellular expression system. Another example is when a protein is intentionally added as part of the processing, then it is treated as a single HCP, and a specific assay will provide better data (than the total HCP immunoassay).

6.2 Control Strategy

6.2.1 TARGETS , ALERT LIMITS , AND REJECT LIMITS

In both United States and European Union regulations, and generally in other markets, HCPs are described as process-rela-ted impurities, and guidances state that their presence should be minimized by the use of appropriate, well-controlled manu-facturing processes. U.S. 21 CFR 610.13 requires biological products to be “...free of extraneous material except that which is unavoidable...”. ICH Q6B indicates that biologic product manufacturers should assess impurities that may be present and de-velop acceptance criteria for the impurities based on their preclinical and clinical experience. However, regulatory guidance also recognizes the inherent challenges involved in using assay-specific data and product- and process-specific reagents. Guid-ance states “The absolute purity of biotechnological and biological products is difficult to determine and the results are meth-od-dependent” (ICH Q6B), and, “In the same manner, standardization of the analytical methods would be problematic since the reagents used are product and production system-related” [European Agency for the Evaluation of Medicinal Products (EMEA) 1997].

Although FDA guidance describes the need to test for HCPs, it also allows, through validation, removal of the release testing requirement, once clearance has been shown to be consistent. An expectation exists that, whenever possible, impurities that were introduced by manufacturing processes and remain in products should be below detectable levels based on a highly sen-sitive analytical method. However, an appropriately conducted clearance study performed as part of process validation can be an acceptable substitute for lot-to-lot (C of A) testing (FDA 1997), even when low levels of HCPs are present in the DS. In

addition, “If impurities are qualitatively and quantitatively (i.e., relative amounts and/or concentrations) the same as in the DS,testing of the DP is not necessary” (ICH Q6B). This is the case for HCPs, the concentration of which in the DP may be inferred from the DS, because the manufacturing process does not introduce, or clear, HCPs when the DS is converted to the DP. Thus,in these instances, HCP testing of the DP is generally not required.

The overarching goal is to develop processes that minimize the quantities of residual HCPs in DSs. When using HCP immu-noassays as part of a manufacturing control system, limits for acceptable results need to be established. The “target” for HCP in final product gives the purification development group an objective for the process, which may or may not be achievable.Antibody processes with highly selective affinity chromatography steps (e.g., using Protein A) are sometimes able to achieve a sensitivity of 1 ng of HCP per mg of protein. Target HCP levels determine the design criteria that are then used to develop the

process.

1434 á1132? Residual Host Cell Protein / General Information USP 39

Preclinical toxicology lots are the first lots for which official HCP values should be determined, because at this point in prod-uct development, the investigator is building the data set that will be used to support the safety of subsequent clinical studies. As the molecule advances through clinical development, the preceding studies are used to support the projected safety (and risk) associated with subsequent studies. Ideally, changes in HCP level and thus clinical exposure should decrease as the prod-uct moves through clinical development.

As part of an overall control strategy, the “reject limits” are those levels which, if exceeded, render the product no longer fit for use. During product development, knowledge about the process and the product increases; therefore, it is expected that reject limits can be tightened as one moves from toxicology material to phase I/II material, to phase III, and finally to commer-cial material. Each drug candidate profile and its clinical program are unique; hence, no single numerical limit is applicable to all.

Reject limits are primarily concerned with patient safety. Typically, one does not have specific data documenting “unsafe”levels of HCPs. Rather, acceptable levels for early stage products should be determined through a risk assessment (including non-clinical data, available literature, previous experience with products manufactured using the same or a similar cell line, etc.). For commercial products, the acceptable levels are also based on the experience gathered during clinical trials. Because the HCP impurity profiles may be influenced by the purification process, the registration batches used in the pivotal clinical trials must be representative of the to-be-marketed process. Reject limits for commercial product should be set to exclude HCP values that are outside the range of clinical experience upon which the risk/benefit decision was made for the biopharmaceuti-cal.

Alert limits, trend limits, or action limits (different companies use different terms) reflect considerations of manufacturing consistency. In a commercial process, there should be sufficient manufacturing history to be able to define out-of-trend results. However, early in product development, where there is little or no manufacturing history, action limits may be used to set upper limits on the expected HCPs, based on experience with similar products for which there is a history, or in comparison to the target value. Exceeding these limits should trigger an investigation to determine whether the lot is releasable. Orthogonal measures of product purity are valuable in such investigations.

With the emergence of orthogonal measures of purity, any non-product related atypical signals should be evaluated. Efforts should be made to revise the purification process to remove any unwanted HCPs present at higher than desirable levels (based on, for example, clinical and non-clinical data, available literature, etc.), or a specific justification of its acceptability must be provided. This is accomplished as part of a risk analysis that considers the clinical exposure similar to that described above for immunoassays.

In cases where HCP assays are improved or changed due to unforeseen events, the reject limits may need to be modified, even increased in some cases (if the new assay has broader coverage). However, it is essential that samples be tested in bridg-ing studies to ensure that no changes in quality would be missed in the new, improved assay. In some cases, improved assays give higher values, so bridging studies (head-to-head with original versus improved assay) will be needed to justify the new specification limit.

6.2.2SAFETY CONSIDERATIONS IN SETTING LIMITS

HCP specification limits in units of ng/mg are typically set using process capabilities and also on the basis of clinical experi-ence (i.e., the levels used in trials where the clinical outcomes are known). Although there are several theoretical safety con-cerns about HCP impurities, the immunogenicity of the impurity is a primary consideration. An immune response may include formation of antibodies directed against the HCP or directed against the therapeutic protein, where the HCP is believed to have acted as an adjuvant. Readers are referred to á1106? for a more in-depth discussion of factors influencing immunogenici-ty. When patients receive more than one biotherapeutic protein with their associated impurities, immunogenicity is an even greater concern. Although high-dose products may contain a higher mass of residual HCPs per dose and this should be consid-ered during risk assessments, the potential effect is not necessarily predictable of a clinical outcome. In some cases, very low levels of certain HCPs have been shown to have clinical effects, whereas high levels of other HCPs have had none.

Different limits may be set, depending on whether the product has only one or a very limited number of co-purifying HCPs or it has a multiplicity of HCPs, each present as a small fraction of the total HCP level in the product. This could lead to a separate assay and specification for individual HCP(s) that cannot be cleared from the process and are expected to be present in the DS and may have bioactivity.

The host cell origin can also be a consideration. It is possible that HCPs from human cell lines (e.g., HEK293 cells) may pose less immunogenicity risk than HCPs from more distantly related species because of sequence homology. However, if these pro-teins are normally intracellular, then they might still be immunogenic and seen as “foreign” by the patient’s immune system. If such antibodies develop, then they could cross-react with endogenous human proteins and neutralize or render ineffective one or more necessary biological systems. As a consequence, arguments can be made on both sides; hence, the risks based on production cell line (microbial versus mammalian) are generally believed to be the same and cannot be practically informative a priori.

In addition to considering immunogenicity, the HCP may be bioactive and a risk. For example, a hamster homolog of a ther-apeutic protein expressed in CHO cells could be sufficiently similar to the product to co-purify. Alternatively, an anti-human

cytokine antibody product expressed in CHO cells could bind to the homologous hamster cytokine, and the complex could USP 39General Information / á1132? Residual Host Cell Protein1435

新旧动能转换相关概念解释

新旧动能转换相关概念解释 一、新动能、新经济 1.新动能指以知识、技术、信息、数据等新生产要素为支撑,聚焦新技术、新产业、新业态、新模式“四新”,推进产业智慧化、智慧产业化、跨界融合化、品牌高端化“四化”,由此汇聚形成的经济发展新动能。 2.新经济是一个不断发展变化的相对概念,指以新技术为基础,以新市场为主体支撑,以新产业、新业态、新模式为表现形式的经济形态,主要包括分享经济、信息经济、生物经济、绿色经济、创意经济和智能制造经济等。 二、“四新”“四化” 这是新旧动能转换的中心任务,即通过新技术、新产业、新业态、新模式,实现产业智慧化、智慧产业化、跨界融合化、品牌高端化。 1.新技术指能够直接或间接产业化的技术,这些技术可替代、可推广、可产生经济效益。主要包括:信息技术,如移动互联、云技术、大数据、物联网等;生物技术,如基因测序、基因技术等;智能制造技术,如3D打印、人工智能、机器人等;新材料技术,如生物基新材料、石墨烯材料、纳米技术等。 2.新产业指以市场需求为导向,运用新技术产生的能够引发重大变革的产业。主要包括:战略新兴产业,如节能环保、新一代信息技术、生物、高端装备制造、新材料等;新兴服务业,如现代金融、现代物流、文化创意、总部经济

等;运用新技术改造传统产业或相关产业跨界融合衍生的新产业,如工业物联网、互联网金融、农业新六产、全域旅游等。 3.新业态指顺应市场需求,依托创新技术,从现有产业和领域中派生出的新环节、新链条、新活动。主要包括:从现有流程中衍生出的新环节,如服装个性化定制;从现有产业链衍生出的新环节,如围绕某个文化品牌开发出新的相关产品或服务;从现有产业和领域中出现的新活动,如在创新创业领域中产生的众创、众包、众扶、众筹等活动。 4.新模式指新的商业模式,即以市场需求为中心,以实现用户价值和企业盈利为目标,打破原有产业链和价值链,通过要素重新组合形成具有较强竞争力的运营模式。主要包括;依托互联网创造全新的商业模式,如阿里巴巴、腾讯等互联网公司开展的电子商务;把硬件融入服务,如乐视等网络视频厂商提出的“付费内容服务+免费硬件”的新商业模式;提供一站式全链条服务。 5.产业智慧化指利用互联网、物联网、大数据等现代信息技术,对传统产业进行升级换代,把劳动密集型的产业转变为资本和技术密集型的产业,或者继续提高资本和技术的密集程度,最终实现产业经济价值提升。产业智慧化类型多样,如传统种植业实现智慧化就是精准农业、传统机械制造业实现智慧化就是智能制造、传统的物流运输业实现智慧化就是智慧物流等。 6.智慧产业化指把信息虚拟资源实现价值化,在此过

高中物理功与动能定理

功功率与动能定理 一、功 要点:条件、;公式: 1.功的正负的判断:;; 例1如图1所示,物体沿弧形轨道滑下后进入足够长的水平传送带,传送带以图示方向匀速运转,则传送带对物体做功情况可能是() A.始终不做功 B.先做负功后做正功 C.先做正功后不做功 D.先做负功后不做功 对应练习: 1.如图,拖着旧橡胶轮胎跑是身体耐力训练的一种有效方法.如果某受训者拖着轮胎在水平直道上跑了100 m,那么下列说法正确的是( ) A.轮胎受到地面的摩擦力做了负功 B.轮胎受到的重力做了正功 C.轮胎受到的拉力不做功 D.轮胎受到地面的支持力做了正功 2.如图所示,质量为m的物体置于倾角为θ的斜面上,物体 与斜面间的动摩擦因数为μ,在外力作用下,斜面以加速度 a沿水平方向向左做匀加速运动,位移为S,运动中物体m 与斜面体相对静止.则支持力做功为,斜面对 物体做的功为. 3.如图所示,木板可绕固定的水平轴O转动.木板从水平位置OA缓慢转到OB位置,木板上的物块始终相对于木板静止.在这一过程中,物块的重力势能增加了2J.用FN表示物块受到的支持力,用Ff表示物块受到的静摩擦力.在这一过程中,以下判断正确的是()A. FN和Ff对物块都不做功 B.FN对物块做功为2J,Ff对物块不做功 C.FN对物块不做功,Ff对物块做功为2J D.FN和Ff对物块所做的总功为4J 2.功的计算: 恒力做功: 多个力总功: 滑动摩擦力的功: 变力做功:; 例2一人在A点拉着绳通过一定滑轮,吊起质量m=50kg的物体,如图所示,开始绳与水平方向夹角为60°,当人匀速提起重物,由A点沿水平方向运动距离s=2m到达B点,此时绳与水平方向成30°角,求人对绳的拉力做的功? 例3如图所示,一质量m=2.0 kg的物体从半径为R=5.0 m的圆弧的A端在拉力作用下沿圆弧运动到B端.拉力F大小不变始终为15 N,方向始终与物体在该点的切线成37°角.物体与圆弧面的动摩擦因数为0.2.圆弧所对应的圆心角为60°,BO边为竖直方向.求这一过程中:

第四章 蛋白质纤维

第四章蛋白质纤维 §4.1蛋白质纤维的一般知识 蛋白质纤维:指基本组成物质为蛋白质的一类纤维。 毛:羊毛、驼毛、兔毛、马毛 天然蛋白质纤维蚕丝:桑蚕丝,柞蚕丝 蛋白质纤维再生蛋白质纤维大豆纤维,牛奶纤维 一蛋白质的组成及结构 属于高分子化合物,结构十分复杂,蛋白质又称朊,是构成生命最原始最基础的物质,羊毛的主要成分是:角朊(角质),丝的主要成分是丝朊(丝素)。 1 元素组成主要元素:碳、氢、氧、氮,还有少量硫磷、铁 2 氨基酸组成蛋白质的基本组成为氨基酸,主要为α-氨基酸, 结构通式: H2N—CH2—COOH R 3 分子结构 蛋白质分子是氨基酸彼此通过氨基和羧基脱水缩合,以酰胺键(即肽键-CO-NH-)联接而成的大分子。 酰胺键又称为肽键,由肽键相连接的缩氨酸叫做肽。 R 蛋白质大分子链为多肽链,又称为多缩氨酸链,是由基团—NH—CH—CO—重复连接而成。 分子之间的作用力:氢键、盐式键、二硫键 二蛋白质的两性性质 蛋白质分子中既含有氨基又含有羧基,因而具有酸性又具有碱性,是典型的两性高分子电解质。 等电点:调节溶液中的pH值,当蛋白质所带的正负电荷数相等时,此时的pH值即为蛋白质的等电点。 羊毛等电点:4.2~4.8 蚕丝等电点:3.5~5.2 在等电点时,具有特别重要的性质:蛋白质不发生电泳现象,溶解度、膨化度、粘度、渗透压、导电率等均显示最低值。

§4.2羊毛 羊毛主要指:绵羊身上剪下的毛。 羊毛的特性:弹性好,手感丰满、吸湿能力强、保暖性好,不易沾污,光泽柔和、染色性能优良,具有独特的缩绒性。 一羊毛的形态结构 原毛:从羊身上剪下来的羊毛 羊毛杂质:羊毛脂、羊汗、沙土、水分、草屑、草籽或其他植物性杂质。 羊毛脂:高级脂肪酸和高级一元醇组成的复杂的有机混合物 羊汗:有机酸盐和无机酸盐组成 羊毛可分为三个部分:毛尖、毛根、毛干。 外观:羊毛纤维具有天然卷曲、纵向呈鳞片覆盖的圆柱体, 从内至外分为三层:鳞片层(表皮层)、皮质层、髓质层 鳞片层(表皮层):逆鳞片方向的摩擦系数大于顺鳞片方向的摩擦系数,称为 定向摩擦系数,这使羊毛具有缩绒性和毡缩性。 皮质层:羊毛的主要组成部分,决定羊毛纤维的物理性能,存在天然色素,因而有些色毛的颜色难以除去。 髓质层:由薄膜细胞组成,髓质层使纤维的强度、卷曲、弹性、染色性较差。 二羊毛的化学组成和分子结构 羊毛的主要成分:角质(角朊),由α-氨基酸缩合而成。 组成元素:碳、氢、氧、氮,还有硫 分子结构:α-氨基酸缩合而成的链状大分子 构型:α-螺旋结构 三羊毛的超分子结构(具体见P99) 由3个螺旋结构的多缩氨酸链组成基本原纤微原纤原纤束(皮质细胞) 四羊毛的机械性能 1 羊毛的线密度 羊毛纤维的直径差异很大,最细绒毛直径为7μm,最粗可达240μm 2 羊毛的长度 由于天然卷曲的存在,其长度可分为自然长度和伸直长度,国产细羊毛的长度在5.5~9 cm, 半细毛的长度在6~40cm。 3 羊毛的卷曲

动能势能做功与能量转化的关系

动能势能做功与能量转化 的关系 Newly compiled on November 23, 2020

第2讲 动能 势能 [目标定位] 1.明确做功与能量转化的关系.2.知道动能的表达式,会用公式计算物体的动能.3.理解重力势能的概念,知道重力做功与重力势能变化的关系.4.理解弹性势能的概念,会分析决定弹性势能大小的因素. 一、功和能的关系 1.能量:一个物体能够对其他物体做功,则该物体具有能量. 2.功与能的关系:做功的过程就是能量转化的过程,做了多少功,就有多少能发生转化,所以功是能量转化的量度.功和能的单位相同,在国际单位制中,都是焦耳. 二、动能 1.定义:物体由于运动而具有的能量. 2.大小:物体的动能等于物体的质量与它的速度的平方乘积的一半,表达式:E k =12 m v 2,动能的国际单位是焦耳,简称焦,用符号J 表示. 3.动能是标量(填“标量”或“矢量”),是状态(填“过程”或“状态”)量. 三、重力势能 1.重力的功 (1)重力做功的特点: 只与物体运动的起点和终点的位置有关,而与物体所经过的路径无关. (2)表达式 W G =mg Δh =mg (h 1-h 2),其中h 1、h 2分别表示物体起点和终点的高度. 2.重力势能 (1)定义:由物体所处位置的高度决定的能量称为重力势能. (2)大小:物体的重力势能等于它所受重力的大小与所处高度的乘积,表达式为E p =mgh ,国际单位:焦耳. 3.重力做功与重力势能变化的关系 (1)表达式:W G =E p1-E p2=-ΔE p . (2)两种情况: 4.重力势能的相对性 (1)重力势能总是相对某一水平面而言的,该水平面称为参考平面,也常称为零势能面,选择不同的参考平面,同一物体在空间同一位置的重力势能不同. (2)重力势能为标量,其正负表示重力势能的大小.物体在参考平面上方时,重力势能为正值;在参考平面下方时,重力势能为负值. 想一想 在同一高度质量不同的两个物体,它们的重力势能有可能相同吗

新旧动能转换热词解释

新旧动能转换热词解释 1、什么是新旧动能转换。所谓新动能,是指推动经济运行发展的新动力,作用于经济发展过程而产生的新能量。新动能主要来源于两个方面:一是通过创新培育而产生的新动能,主要是指依靠知识、科技、信息、数据等新生生产要素的投入,产生的新技术、新产业、新业态、新模式,汇集形成推动经济发展的新动能,也就是李克强总理所说的“无中生有”;二是通过改造提升而产生的新动能,主要是指在传统产业领域,通过发展农业“新六产”、制造业“智能制造”、服务业“个性化定制服务”等,激发传统产业发展新活力,也就是总理所说的“有中出新”。所谓旧动能,即传统动能,是指依靠传统生产要素(主要包括土地、劳动力、资本等)投入而产生的推动经济运行发展的能量。 2、山东省“5+5”十强产业。壮大发展新一代信息技术、高端装备、新能源新材料、现代海洋、医养健康等5大新兴产业,提升发展高端化工、现代高效农业、文化创意、精品旅游、现代金融服务等5大优势产业。 3、山东省“三核引领区域融合互动”的动能转换总体格局。三核就是济南、青岛、烟台三个城市,率先突破,打造新旧动能转换的主引擎。其他14个城市开发区、高新区和海关特殊监管区作为补充,明确产业发展定位和方向。以三

核引领、实现多点突破、融合发展,形成新旧动能转换合力。 4、以“四新”促“四化”实现“四提”。以新技术、新产业、新业态、新模式,促进产业智慧化、智慧产业化、跨界融合化、品牌高端化。实现“四提”发展,一是传统产业提质效,二是新兴产业提规模,三是跨界融合提潜能,四是品牌高端提价值。 5、“去、增、提”三篇大文章。“去”,就是充分利用市场化、法治化手段,坚决化解钢铁、煤炭、电解铝、火电等过剩产能,淘汰落后产能,将产能利用率保持在合理区间,为动能转换拓展空间。“增”,就是推动新兴产业扩容倍增,突破核心关键技术,壮大发展5大新兴产业,培育一批引领行业发展的龙头企业,把山东打造成新兴产业发展的策源地和集聚区。“提”,就是深入实施新一轮高水平企业技改行动计划,提升发展5大优势产业,把传统优势转化成品牌优势和竞争优势。 6、从定位、重大项目、重要政策三个层面去谋划。就是我市和各县区在这次全省新旧动能转换当中的定位,提出的支撑转换的重大项目,和需要国家和省支持的重要政策。 7、把握好省、市、县区三位一体。也就是,无论是定位,还是重大项目、还是拟争取的政策,都要围绕新旧动能转换,市里要统一到省里的方案上来,县区要统一到市里的方案上来。

大豆蛋白纤维

大豆纤维的探究及应用 院系:外语系 学号:201313060124 姓名:司淼

目录 大豆纤维 大豆纤维释义 大豆纤维简介 大豆蛋白纤维 大豆纤维纱线 大豆纤维的面料 大豆纤维染整 大豆纤维服饰 大豆纤维衣服正确洗涤方法

大豆纤维释义 1. Soy Fiber 属于膳食纤维,在减肥过程中可以产生饱足感,而减少食物的摄取,但它们会干扰其他营养素的吸收,因此不建议单独食用。 2. SB=soybean SB=soybean 大豆纤维 3. soybean fibers soybean fibers大豆纤维 大豆纤维简介 大豆蛋白纤维属于再生植物蛋白纤维类,是以榨过油的大豆豆粕为原料,利用生物工程技术,提取出豆粕中的球蛋白,通过添加功能性助剂,与腈基、羟基等高聚物接枝、共聚、共混,制成一定浓度的蛋白质纺丝液,改变蛋白质空间结构,经湿法纺丝而成. 其有着羊绒般的柔软手感,蚕丝般的柔和光泽,棉的保暖性和良好的亲肤性等优良性能,还有明显的抑菌功能,被誉为“新世纪的健康舒适纤维”。 经过工业化规模生产,大豆纤维从纺纱到织造到染整的相关生产技术均已相对成熟,其价格已从初期的每吨7万多元,降至3.5万元左右,已被下游应用企业所认可,产业链结构也逐步形成. 大豆纤维是以脱去油脂的大豆豆粕作原料,提取植物球蛋白经合成后制成的新型再生植物蛋白纤维,是由我国纺织科技工作者自主开发,并在国际上率先实现了工业化生产的高新技术,也是迄今为止我国获得的唯一完全知识产权的纤维发明。 在成为纤维之前,要从大豆中提取蛋白质与高聚物为原料,采用生物工程等高新技术处理,经湿法纺丝而成。这种单丝,细度细、比重轻、强伸度高、耐酸耐碱性强、吸湿导湿性好。有着羊绒般的柔软手感,蚕丝般的柔和光泽,棉的保暖性和良好的亲肤性等优良性能,还有明显的抑菌功能,被誉为“新世纪的健康舒适纤维”。 以50%以上的大豆纤维与羊绒混纺成高支纱,用于生产春、秋、冬季的薄型绒衫,其效果与纯羊绒一样滑糯、轻盈、柔软,能保留精纺面料的光泽和细腻感,增加滑糯手感,也是生产轻薄柔软型高级西装和大衣的理想面料。 用大豆纤维与真丝交织或与绢丝混纺制成的面料,既能保持丝绸亮泽、飘逸的特点,又能改善其悬垂性,消除产生汗渍及吸湿后贴肤的特点,是制作睡衣、衬衫、晚礼服等高档服装的理想面料。 此外,大豆纤维与亚麻等麻纤维混纺,是制作功能性内衣及夏季服装的理想面料;与棉混纺的高支纱,是制造高档衬衫、高级寝卧具的理想材料;或者加入少量氨纶,手感柔软舒适,用于制作T恤、内衣、沙滩装、休闲服、运动服、时尚女装等,极具休闲风格。 大豆蛋白纤维是由华康集团董事长李官奇先生历经十年研究开发成功,获得世界发明专利金奖,李官奇先生的这项发明为纺织业带来了一场新的革命,在纤维材料发展史上和人造

功、功率与动能定理(解析版)

构建知识网络: 考情分析: 功和功率、动能和动能定理、机械能守恒定律、能量守恒定律是力学的重点,也是高考考查的重点,常以选择题、计算题的形式出现,考查常与生产生活实际联系紧密,题目的综合性较强。复习中要特别注意功和功率的计算,动能定理、机械能守恒定律的应用以及与平抛运动、圆周运动知识的综合应用 重点知识梳理: 一、功 1.做功的两个要素 (1)作用在物体上的力. (2)物体在力的方向上发生的位移. 2.功的物理意义 功是能量转化的量度. 3.公式 W =Fl cos_α (1)α是力与位移方向之间的夹角,l 为物体对地的位移. (2)该公式只适用于恒力做功. 4.功的正负 (1)当0≤α<π 2 时,W >0,力对物体做正功. (2)当π 2<α≤π时,W <0,力对物体做负功,或者说物体克服这个力做了功. (3)当α=π 2时,W =0,力对物体不做功. 通晓两类力做功特点 (1)重力、弹簧弹力和电场力都属于“保守力”,做功均与路径无关,仅由作用对象的初、末位置(即位移)决定。

(2)摩擦力属于“耗散力”,做功与路径有关。 二、功率 1.物理意义:描述力对物体做功的快慢. 2.公式: (1)P =W t ,P 为时间t 内的物体做功的快慢. (2)P =Fv ①v 为平均速度,则P 为平均功率. ②v 为瞬时速度,则P 为瞬时功率. 3.对公式P =Fv 的几点认识: (1)公式P =Fv 适用于力F 的方向与速度v 的方向在一条直线上的情况. (2)功率是标量,只有大小,没有方向;只有正值,没有负值. (3)当力F 和速度v 不在同一直线上时,可以将力F 分解或者将速度v 分解. 4.额定功率:机械正常工作时的最大功率. 5.实际功率:机械实际工作时的功率,要求不能大于额定功率. 三、动能 1.定义:物体由于运动而具有的能. 2.公式:E k =1 2 mv 2. 3.物理意义:动能是状态量,是标量(选填“矢量”或“标量”),只有正值,动能与速度方向无关. 4.单位:焦耳,1J =1N·m =1kg·m 2/s 2. 5.动能的相对性:由于速度具有相对性,所以动能也具有相对性. 6.动能的变化:物体末动能与初动能之差,即ΔE k =12mv 22-1 2mv 12. 四、动能定理 1.内容:在一个过程中合外力对物体所做的功,等于物体在这个过程中动能的变化. 2.表达式:(1)W =ΔE k . (2)W =E k2-E k1. (3)W =12mv 22-1 2mv 12. 3.物理意义:合外力做的功是物体动能变化的量度. 4.适用条件 (1)动能定理既适用于直线运动,也适用于曲线运动. (2)动能定理既适用于恒力做功,也适用于变力做功.

新型生物质纤维 Q..

新型生物质纤维 蛋白质纤维的现状与发展趋势 Suchow university 【前言】 随着石油价格的不断上涨及合成高分子材料对环境造成的污染日趋严重,基于天然高分子可降解材料的研究、开发及产业化受到了人们的广泛关注,蛋白质纤维已成为高分子科学研究的前沿领域之一。 所谓蛋白质纤维是指基本组成物质为蛋白质的一类纤维,具体来说分为天然蛋白质纤维如动物毛和蚕丝,再生蛋白质纤维包括再生动物蛋白纤维、再生植物蛋白纤维等。 【关键词】:天然蛋白质纤维;再生蛋白质纤维;现状;发展趋势 一、天然蛋白质纤维 1、羊毛 天然蛋白质纤维中应用广泛的是动物毛,其中羊毛应用最为广泛。羊毛纤维是一种天然蛋白质纤维,也是人类较早用于制作纺织品的一种原材料。人类大约在公元前4000一前3000年,即新石器时代,就已把羊毛纤维用作纺织材料了[1]。 羊毛特有的化学组分与结构形态决定了其许多优良性能如吸湿性优良、光泽柔和、悬垂性好、穿着舒适、而寸磨性好、不易玷污、保暖性好、手感丰满、富有弹性、抗皱性好等,是高档的纺织品原材料。羊毛的这些性能使得其产品具有质地丰厚、手感丰满、弹性好、光泽自然的特殊风格。此外,由羊毛制得的产品对人体友好,并且可生物降解对环境友好,这些均是合成纤维无法比拟的。 现阶段,除了人们常见的羊毛织物,如羊毛衣、羊毛围巾等,羊毛纤维已经逐渐渗入到其他各个领域中。 用于褥疮预防和护理的医用羊皮最近被成功地开发并投放使用。褥疮是因年

老、体弱、长期卧床、瘫痪以及不能自动翻身时患者身体局部长期受压迫而引起血液循环障碍,加上摩擦和潮湿而形成的溃疡。褥疮一旦发生不仅病人极端痛苦,而且如受感染会造成败血症而危及生命。羊皮上的羊毛密度高,柔软性好,具有独特的韧性和弹性回复能力。羊毛的摩擦系数小.毛纤维可在一定范围内移动和变形,因此,羊毛纤维可提供柔软、光滑的界面从而减少患者的皮肤张力和皮下组织的剪切力。作这种医用羊皮置于病人与床褥之间,毛丛接触皮肤起到减轻局部压力、减小摩擦、增加吸湿能力等作用[2]。 羊毛空气静电过滤器空气静电过滤器是一种带静电荷的过滤器。静电荷能够有效地吸附微尘.因此吸尘效果比传统的不带静电荷的过滤器要好。羊毛静电过滤器是一种高附加值产品,价格大约为1 000澳元/kg。羊毛的静电效果要比化纤持久,长久的静电效果是过滤器良好的过滤性能的保证。另外,羊毛的某些特征,如生态降解性等是合成纤维所不能比的。因此羊毛静电过滤器应该比化纤静电过滤器更有优势。为挽救失去的羊毛过滤器市场,科研人员正在开发羊毛过滤器的制造新方法。其中一种方法就是将能与毛产生静电效果的新型纤维与毛纤维混合加工,从而消除了化工树脂粉末的影响。该项研究正在进行,估计在两年内会有新发展[3]。 2012年,国际羊毛局(The Woolmark Company)发起羊毛运动,旨在将羊毛纤维独特的天然性和环保优势传达给消费者,并继续号召更多的国家参与到羊毛运动中。羊毛纤维虽然早已被开发利用,但其研究仍有广阔的发展前景空间。据悉,中国和澳大利亚有关院校参与并已立项的研究课题有:1)人民解放军军服用洗可穿羊毛混纺织物的开发;2)利用纳米和溶胶胶化技术进行毛织物防缩整理:3)运动服装用纯毛织物;4)澳毛与中国产纤维混纺产品的环保染色[4]。 中国的经济正在快速发展,消费者的购买力日趋增强,对羊毛产品的质量和功能性要求越来越高。中国羊毛资源丰富,养殖业增产节支是保证羊毛工业发展的基础。不断了解国外在发展羊毛产业中的研究动向,对促进中国羊毛产业的发展、繁荣国内市场,走向世界十分必要。 2、兔毛 动物毛中除了羊毛,兔毛的发展和研究也具有较大的空间。我国是重要的兔毛生产国,而兔毛纤维是一种高级天然动物纤维,其质地洁白、柔软、滑爽,具

功、功率和能量总结

一、功和功率 1、功的计算 (1)、恒力做功的计算:αcos Fl W =,F 为恒力,l 是F 的作用点相对于地的位移,α是F 和l 间的夹角。 (2)、变力做功的计算:○ 1把变力做功转化为恒力做功求解○2用动能定理求解○3用图像法求解。 (3)总功的计算 2、功率的计算 瞬时功率αcos Fv P = 平均功率t W P = 题型1、如图,水平传送带两端点A 、B 间的距离为L ,传送带开始时处于静止状态.把一个小物体放到右端的A 点,某人用恒定的水平力F 使小物体以速度v 1匀速滑到左端的B 点,拉力F 所做的功为W 1、功率为P 1,这一过程物体和传送带之间因摩擦而产生的热量为Q 1.随后让传送带以v 2的速度匀速运动,此人仍然用相同的水平力恒定F 拉物体,使它以相对传送带为v 1的速度匀速从A 滑行到B ,这一过程中,拉力F 所做的功为W 2、功率为P 2,物体和传送带之间因摩擦而产生的热量为 Q 2.下列关系中正确的是( ) A 、W 1=W 2,P 1<P 2,Q 1=Q 2 B .W 1=W 2,P 1<P 2,Q 1>Q 2 C .W 1>W 2,P 1=P 2,Q 1>Q 2 D .W 1>W 2,P 1=P 2,Q 1=Q 2 题型2、如图所示,质量均为m 的物体A 、B 通过一劲度系数为k 的轻弹簧相连,开始时B 放在地面上,A 、B 都处于静止状态.现用手通过细绳缓慢地将A 向上提升距离L 1时,B 刚要离开地面,此过程手做功W 1、手做功的平均功率为P 1;若将A 加速向上拉起,A 上升的距离为L 2时,B 刚要离开地面,此过程手做功W 2、手做功的平均功率为P 1.假设弹簧一直在弹 性限度范围内,则 A .k mg L L = =21 B .k mg L L 212=> C .W 2 > W 1 D .P 2< P 1 二、机车的启动问题 题型3、某列车发动机的额定功率为KW 4 102.1?,列车的质量为Kg 5 100.1?,列车在水 平轨道上行驶时,阻力是车重的0.1倍,2 /10s m g =。 (1)若列车保持额定功率从静止启动,则列车能达到的最大速度是多少? (2)若列车从静止开始以0.5m/s 2的加速度做匀加速直线运动,则这段过程能维持多长时间?6s 末列车的瞬时功率多大? (3)如果列车保持额定功率行驶,当列车在水平轨道上行驶速度为10m/s 时,列车的加速度为多少?

新旧动能转换

三、内涵与启示 (一)“新旧动能”政策体系产生背景 1.消费和服务业将取代投资、出口成为拉动经济增长的主要动力 过去30年,中国平均经济增速为10%,这主要是得益于其成本竞争力(劳动力、土地、环境、汇率等)、改革红利(1979年农村改革,1992年邓小平南巡改革,1998年国企改革和2001年加入WTO)和政府强势主导。如今这种增长模式已经达到了极限:成本竞争力消失,国企效率下滑,杠杆率飙升,最近资产市场震荡也暴露出政府政策调控的有效性不足。中国的经济增速从10%降至 5%-6%,意味着中国经济进入了“新常态”。“新常态”下经济增速放缓是大势所趋,经济结构也将随之深度调整。消费和服务业将取代投资、出口成为拉动经济增长的主要动力。如图1所示,从2013年第一季度开始,第三产业增加值同比增速开始高于第二产业增加值增速,成为拉动经济增长的主要动力。与此同时,作为拉动经济增长的三驾马车之一,消费支持对经济的贡献超过了资本形成的贡献,对外贸易对经济增长的贡献为负。 2.新经济对中国经济发展支撑力逐步提升

高盛在报告《“新常态”下的“新中国”经济投资》中提出了一套系统性的分析框架,以便更好地追踪变革中的中国经济。高盛从143个行业,3636家公司中,进行挑选了30个次级行业的671家公司,作为追踪中国新经济的参照。这些公司来自A股、H股和中概股,市值1.8万亿美元。这些产业遍布卫星光缆、网购、教育服务、影视娱乐、家用耐用品、能源服务、资产管理、生物科技、航天国防、IT咨询等30个领域。相对于传统产业,新经济产业中健康医疗、科技行业的比例大幅提升,合计占到60%以上。过去几年的数据也表明,新经济的表现确实要远远优于传统产业 3.山东省区域性经济风险事件频发,引起中央层面注意。 2017年初,曾经的中国500强企业山东天信集团陷入债务泥潭,7家关联公司天信集团、天圆铜业、天信光伏、天信进出口、天泽物资、天泽物流、澳纳纺织已进入破产重整程序,其中负债最多的山东天圆有限公司负债总额高达104.52亿元,负债率高达180.77%。这是继山东上市公司中国宏桥、魏桥纺织同时发布延迟刊发2016年全年业绩停牌,长兴集团破产、齐星集团债务危机后,又爆发的一起企业危机事件。一系列的区域性企业债务危机揭示了山东经济的“窘境”:虽然山东省在经济体量方面位居全国前三,但在产业结构、人均收入、科技支撑以及民营经济和新兴业态发展等许多方面,和广东、浙江等新经济活跃省份的差距还很大。同时山东省区域性经济风险事件也引起了中央层面的注意,李克强总理在两会期间参加山东代表团审议时,指出了山东省“新旧动能转换”问

新型纤维综述

新型纤维综述 一、常规纺织纤维的类别和特征(举例) 1.植物纤维:棉、麻 棉:吸湿性好,穿着舒适,光泽较暗,手感柔软,风格朴实,耐用耐洗,物美价廉,易折皱,服装保形性欠佳,耐碱不耐酸。 麻:吸湿散湿性好,干爽利汗,风格粗犷,有光泽,粗硬,弹性差,易折皱。耐碱不耐酸。 2.动物纤维:羊毛、蚕丝 羊毛:吸湿性强,手感丰满柔软,光泽柔和莹润,因表面有鳞片具有独特的缩绒性,保暖性能好,干燥时抗皱弹性好,湿态易皱,易虫蛀,耐酸不耐碱。 蚕丝:吸湿透气,舒适性极佳,滑爽柔软,光泽优雅悦目,风格高雅华丽,变形时弹性好,悬垂性好,湿态易皱,不耐汗,耐光性差,多晒会泛黄变脆,耐酸强于耐碱。 3.人造纤维:粘胶 粘胶:吸湿性好,穿着舒适,光滑明亮,柔软,悬垂性好,易皱,水洗易变形,缩水严重,湿强低,耐碱不耐酸。 4.化学纤维:锦纶、涤纶、腈纶 锦纶:耐用性,弹性好 涤纶:洗可穿的佼佼者 腈纶:最不怕光的合成羊毛 共性:强度高,不易起皱,悬垂性好,服装保形性好,易洗快干,不缩水,不霉不蛀,热定型性能好,可形成稳定造型,吸湿性差,易产生静电,易起毛起球。 二、新型纤维的种类和特性(举例) 1.天然纤维:(竹纤维)植物纤维、(蜘蛛丝)动物纤维、(银纤维)金属纤维及其他 竹纤维:竹原纤维具有良好的透气性、瞬间吸水性、较强的耐磨性和良好的染色性等特性,具有天然抗菌、抑菌、除螨、防臭和抗紫外线功能。竹再生纤维素纤维不具抗菌功能。 用途:竹纤维纱线用于服装面料、凉席、床单、窗帘、围巾等,如采用与维纶混纺的方法可生产轻薄服装面料。与棉、毛、麻、绢及化学纤维进行混纺,用于机织或针织,生产各种规格的机织面料和针织面料。机织面料可用于制作窗帘、夹克衫、衬衫、床单和毛巾等。针织面料适宜制作内衣、汗衫、T恤衫、袜子等。竹原纤维含量30%以下的竹棉混纺纱线更适合于内裤、袜子,还可用于医疗护理用品。 蜘蛛丝:高强度,高弹性,高断裂功,低密度,有良好的耐温及耐紫外线性能,有良好的生物相容性,理化性质优,每根蜘蛛丝的抗拉强度是钢材的五倍,弹性也比人造纤维好,开发前景广阔。

动能、势能、功和能量的变化关系

龙文教育学科教师辅导讲义

合外力做的功等于物体动能的变化。(这里的合外力指物体受到的所有外力的合力,包括重力)。表达式为 W=△E k 动能定理也可以表述为:外力对物体做的总功等于物体动能的变化。实际应用时,后一种表述比较好操作。不必求合力,特别是在全过程的各个阶段受力有变化的情况下,只要把各个力在各个阶段所做的功都按照代数和加起来,就可以得到总功。 3.重力势能 (l )定义:物体由于被举高而具有的能量. (2)重力势能的表述式 mgh E p = 物体的重力势能等于物体的重量和它的高度的乘积,重力势能是标量,也是状态量,其单位为J (3)重力做功与重力势能的关系 重力做功只跟物体的运动过程中初、末位置的高度差有关,而与运动的路径无关. 当物体下落时,重力做正功,重力势能p E 减少,减少的值等于重力所做的功. 当物体上升时,重力做负功,重力势能E 增加,增加的值等于重力所做的功 物体下落 210 p p G E E W >> 物体上升 210p p G E E W << (4)重力势能具有相对性. 定了参考平面,物体重力势能才有确定值.(通常以水平地面为零势能面) 重力势能的变化与参考平面选择无关. 4. 弹性势能 (1)发生弹性形变的物体,在恢复原状时能够对外做功,物体具有能量,这种能量叫弹性势能. (2)弹性势能与形变大小有关 巩固练习 1. A. B. C.地球上 D. 2.物体在运动过程中,克服重力做功为50 J A.重力做功为50 J B.物体的重力势能一定增加50 J C.物体的动能一定减少50 J D.重力做了50 J 3. A. B.相对于不同的参考平面,物体具有不同数值的重力势能,但这并不影响研究有关重力势能的问题. C.在同一高度将物体不论向任何方向抛出,只要抛出时的初速度大小相 D.放在地面的物体,它的重力势能一定等于零

碰撞过程中动能转化问题

龙源期刊网 https://www.360docs.net/doc/2613514023.html, 碰撞过程中动能转化问题 作者:邓先君邓艾 来源:《中学物理·高中》2013年第04期 碰撞是自然界中的一种普遍现象,也是一种基本的相互作用形式,无论在宏观世界还是粒子物理领域都是一个极具应用价值、值得深究的问题.中学阶段,碰撞模型的建立是简单的系 统概念的应用,是对相互作用矢量关系的深化.在生活上和教学工作过程中,我们把碰撞理解 为物体之间由于内力作用导致系统内部物体运动状态发生变化或者能量形式发生转化的过程. 粒子物理中这种相互作用过程中的能量转化还会导致粒子形态发生变化,形成新的粒子或者出现新的物质形态.在现代物理的前沿领域,粒子对撞在粒子物理或者高能物理领域应用颇多.故,在基础教育阶段,条件许可的话我们有必要对碰撞过程中物体之间的动能转化做细致深入的理解.这里重点从原理上建立碰撞模型来说明一些动能损失的相关问题,进而细致解释为什 么碰撞过程中物体黏到一起时系统动能损失最大. 体系的动量关系 碰撞过程中内力在相当短的δt时间里迅速变化(见图1).由于内力是一个巨大的冲击 力,为物体(此概念包括微观包括粒子,后不多做说明)之间强烈的相互作用提供了可能.在 这个过程中,相互作用时间δt极短,又相对于系统所受的内力而言,外力F只是一个小量, 故系统所受外力的冲量∫t0Fdt可认为趋近近于零,系统动量守恒.这是处理碰撞问题的一个基本原则,即:碰撞系统在碰撞前后按动量守恒来处理.两个质量分别为m1、m2速度分别为v1、v2(v1>v2)的体系一维碰撞的动量方程为 体系的动能转化关系 人教版的高级中学物理教材选修3-5中定义“如果碰撞过程中机械能守恒,这样的碰撞叫 做弹性碰撞;如果机械能不守恒,这样的碰撞叫做非弹性碰撞.”这是该版教材对碰撞从能量角度的定义.鉴于中学阶段势、场的概念并不清晰,保守力也未能多作介绍,所以对机械能的界 定并不具体,况且机械能只是物理学中一个阶段性的过渡概念不宜深化也无从深化.我们不推 荐过分从机械能角度去界定碰撞过程中系统的能量关系.而是从参与碰撞的物体动能变化角度 来说明能量转化的问题,这样就会显得更直观更全面更具有优越性. 个人认为在碰撞问题处理时,把系统动能减少的情形称之为非弹性碰撞,并引入完全非弹性碰撞的概念来对称定义动能损失最大的碰撞形式,这样,在教学过程中更易于清晰科学的说明问题.对应于完全非弹性碰撞,把弹性碰撞称之为完全弹性碰撞.于是整个碰撞部分从动能角度来解释,在理论上全面而具备对称性.下面所有的讨论都将建立在上述理解上. 碰撞,是建立在动量守恒条件下的相互作用模型.从原理上看,内力的冲量矢量和为零, 对系统的总动量不产生影响,但是物体之间的相互作用力可能会对系统做功.人们之所以对之

湖北省建设工程施工合同(标准版本)

工程编号: 合同编号: 湖北省建设工程施工合同 湖北省工商行政管理局 湖 北 省 建 设 厅 二○○七年九月

目录 第一部分协议书 一、工程概况 (1) 二、工程承包范围 (1) 三、合同工期 (1) 四、工程质量 (1) 五、合同价款 (2) 六、组成合同的文件 (2) 七、发包人承诺 (2) 八、承包人承诺 (2) 九、合同生效 (2) 十、合同备案 (2) 第二部分通用条款 一、总则 1 定义 (3) 2 合同文件及解释 (5) 3 语言及适用的法律、标准与规范 (5) 4 通讯联络 (6) 5 工程分包 (6) 6 现场考察 (7) 7 投标文件的完备性和义务 (7) 8 文物、化石和地下障碍物 (7) 9 安全事故处理 (8)

11 联合体的责任 (8) 12 保障 (9) 13 财产 (9) 二、合同主体 14 发包人 (9) 15 承包人 (10) 16 现场管理人员任命和更换 (12) 17 发包人代表 (12) 18 监理工程师 (13) 19 造价工程师 (14) 20 承包人代表 (14) 21 分包人和另行分包 (15) 22 承包人雇员 (15) 三、担保、保险与风险 23 工程担保 (16) 24 发包人风险 (18) 25 承包人风险 (18) 26 不可抗力 (18) 27 保险 (19) 四、工期 28 进度计划和报告 (20) 29 开工 (21)

31 工期延误 (22) 32 加快进度 (23) 33 竣工日期 (24) 34 误期赔偿 (24) 五、质量与安全 35 质量的管理 (24) 36 质量目标 (24) 37 已完工程及设备材料保护 (25) 38 安全防护和文明施工 (25) 39 放线 (26) 40 钻孔与勘探性开挖 (27) 41 发包人供应材料设备 (27) 42 承包人采购材料设备 (28) 43 材料设备的检验 (29) 44 检查和返工 (30) 45 隐蔽工程和中间验收 (30) 46 重新检验和额外检验 (31) 47 工程试车 (31) 48 竣工资料 (32) 49 竣工验收 (32) 50 质量保修 (33) 六、造价

湖北省建设工程施工合同(标准版本)--EF-2007-0203

工程编号: 合同编号: 湖北省建设工程施工合同 湖北省工商行政管理局 湖 北 省 建 设 二○○七年九月

目录 第一部分协议书 一、工程概况 (1) 二、工程承包范围 (1) 三、合同工期 (1) 四、工程质量 (1) 五、合同价款 (2) 六、组成合同的文件 (2) 七、发包人承诺 (2) 八、承包人承诺 (2) 九、合同生效 (2) 十、合同备案 (2) 第二部分通用条款 一、总则 1 定义 (3) 2 合同文件及解释 (5) 3 语言及适用的法律、标准与规范 (5) 4 通讯联络 (6) 5 工程分包 (6) 6 现场考察 (7) 7 投标文件的完备性和义务 (7) 8 文物、化石和地下障碍物 (7) 9 安全事故处理 (8) 10 知识产权 (8)

11 联合体的责任 (8) 12 保障 (9) 13 财产 (9) 二、合同主体 14 发包人 (9) 15 承包人 (10) 16 现场管理人员任命和更换 (12) 17 发包人代表 (12) 18 监理工程师 (13) 19 造价工程师 (14) 20 承包人代表 (14) 21 分包人和另行分包 (15) 22 承包人雇员 (15) 三、担保、保险与风险 23 工程担保 (16) 24 发包人风险 (18) 25 承包人风险 (18) 26 不可抗力 (18) 27 保险 (19) 四、工期 28 进度计划和报告 (20) 29 开工 (21) 30 暂停施工和复工 (21) 31 工期延误 (22) 32 加快进度 (23) 33 竣工日期 (24) 34 误期赔偿 (24) 五、质量与安全 35 质量的管理 (24) 36 质量目标 (24) 37 已完工程及设备材料保护 (25)

湖北省建筑工程施工统一用表

湖北省建筑工程施工统一用表 工程施工资料的分类 当前工程项目施工资料一般按工程技术管理、工程质量保证、工程质量验收三大类进行收集和整理,根据资料的性质,可将其划分为以下七小类: A 类工程管理资料 1、工程概况表 2、工程开工报告 3、工程竣工报告 4、工程停工报告 5、工程复工报告 6、施工进度计划分析 7、项目大事记 8、施工日志 9、不合格项处置记录 10、施工总结 B类工程技术资料 1、工程技术文件报审表 2、技术管理资料 3、设计变更文件 C类工程测量记录 1、工程定位测量记录 2、地基验槽记录 3、楼层放线记录 4、沉降观测记录 5、建筑物垂直度观测记录 D类工程施工记录 1、通用记录 2、土建专用施工记录 3、建筑安装专用施工记录 E类工程试验检验记录 1、施工试验记录(通用) 2、结构专用施工试验记录 3、设备试运转记录 4、电气专用施工试验记录 5、管道专用施工试验记录 6、通风空调专用施工试验记录 7、电梯专用施工试验记录

F类工程物质资料 施工物质资料统一用表(表F)见现行湖北省建筑工程材料(构件)检验(试验、检测)报告用表。 G类施工验收资料 1、工程检验批质量验收记录 2、分项工程质量验收记录 3、分部(子分部)工程质量验收记录 4、单位(子单位)工程质量竣工验收记录 5、单位(子单位)工程质量控制资料核查记录 6、单位(子单位)工程安全和功能检验资料核查及主要功能抽查记录 7、单位(子单位)工程观感质量检查记录 8、施工现场质量管理检查记录 9、工程检验批质量检查记录表(通用)

工程概况表 表A1 编号: 附:建筑总平面图、建筑立面图、建筑剖面图 本表由施工单位填写,城建档案馆和施工单位各保存一份。

动能、势能、功和能量的变化关系

龙文教育学科教师辅导讲义讲义编号

1 22 122212221212k k E E mv mv a v v ma Fs W -=-=-== 12k k E E W -= 即合力所做的功,等于物体动能的变化。 (2)动能定理的表述 合外力做的功等于物体动能的变化。(这里的合外力指物体受到的所有外力的合力,包括重力)。表达式为 W=△E k 动能定理也可以表述为:外力对物体做的总功等于物体动能的变化。实际应用时,后一种表述比较好操作。不必求合力,特别是在全过程的各个阶段受力有变化的情况下,只要把各个力在各个阶段所做的功都按照代数和加起来,就可以得到总功。 3.重力势能 (l )定义:物体由于被举高而具有的能量. (2)重力势能的表述式 mgh E p = 物体的重力势能等于物体的重量和它的高度的乘积,重力势能是标量,也是状态量,其单位为J (3)重力做功与重力势能的关系 重力做功只跟物体的运动过程中初、末位置的高度差有关,而与运动的路径无关. 21p p G E E W -= 当物体下落时,重力做正功,重力势能p E 减少,减少的值等于重力所做的功. 当物体上升时,重力做负功,重力势能E 增加,增加的值等于重力所做的功 物体下落 210p p G E E W >> 物体上升 210 p p G E E W << (4)重力势能具有相对性. 定了参考平面,物体重力势能才有确定值.(通常以水平地面为零势能面) 重力势能的变化与参考平面选择无关. 4. 弹性势能 (1)发生弹性形变的物体,在恢复原状时能够对外做功,物体具有能量,这种能量叫弹性势能. (2)弹性势能与形变大小有关 巩固练习 1.

新旧动能转换”这个词含义

新旧动能转换”这个词,导读 “新旧动能”概念从2015年提出到2016年内涵丰富再到2017年"新旧动能转换”具体工作推进。从演变趋势来看,中央政府已经对中国经济发展阶段有较为深刻的判断:“我国经济正处在新旧动能转换的艰难进程中“,即经济社会进入了“新常态”,同时已看手逐步推进经济”新旧动能转换”工作。 一、“新旧动能”政策体系演变过程 2015年10月,李克强总理在召开的政府会议中对当时中国经济进行了初步判断:“我国经济正处在新旧动能转换的艰难进程中”,“新旧动能"开始正式出现在囯家领导人讲话中。 2015年12月,国家行政学院决策咨询部副主任、研究员王小广在“新旧动能转换:挑战与应对”一文中从新旧动能转换必要性、新动能的主要来源和培育新动能的主要路径等方面对“新旧日动能”进行了系统阐述《十三个五年规划纲要》提出要拓展发展动力新空间,增强发展新动能。 2016年政府工作报告有三处提及“新日动能”,强调:做好“十三五”时期经济社会发展工作,要加快新旧发展动能接续转换;指出经济发展过程必然伴随着“新日动能迭代更替”的过程,并对国内经济形势进行了初步判断:长期积累的矛盾和风险进一步显现,经济增速换挡结构调整阵痛、新日动能转换相互交织,经济下行压力加大 2017年1月20日,国务院办公厅印发了《关于创新管理优化服务培育壮大经济发展新动能加快新旧动能接续转换的意见》,是我国培

育新动能加速新旧动能接续转换的第一份文件。 2017年3月6日“两会期间”,李克强总理两会期间参加山东代表团审议时指出,山东发展得益于动能转换,希望山东在国家发展中继续挑大梁,在新旧动能转换中继续打头阵。 2017年政府工作报告有两处“新旧动能转换“,并提出做好2017年政府工作中要依靠“依靠创新推动新旧动能转换和结构优化升级”,并提出“双创”是推动新旧动能转换和经济结构升级的重要力量。 4月18日,李克强总理在贯彻新发展理念培育发展新动能座谈会上强调,实现经济结构转型升级,须加快新旧动能转换。这种转换既来自“无中生有”的新技术、新业态、新模式,也来自“有中出新”的传统产业改造升级。两者相辅相成、有机统一。 二、内涵与启示 新日动能”自2015年开始出现在中央和地方政府主要领导讲话和文件中,在2016年在互联网出现的频率逐步提升。2016年末,特别是进入2017年以来,“新旧日动能”更是频繁出现在政府相关文件中,内涵也逐渐丰富和完善。“新旧动能”作为政府官方用语,并没有严格的概念界定,但是我们可以从一系列政府文件中和领导讲话中进行理解。 1.发展“新经济”是培育“新动能”的重要措施 在不同时期,“新经济”有不同的内涵,当前新经济主要是指创新性知识在知识中占主导、创意产业成为龙头产业的新经济形态。而"新动能”的内涵更加广泛,需求端(例如:消费能力提升)和供给端(例如:第三产业快速发展或新产业形态)都能成为经济发展的“新动能”。2016

相关文档
最新文档