production_of_monoclonal_antibodies
鸡IgY
DNA免疫鸡制备IgY抗体及其应用博士生石爽概述在后基因组时代,生物医学研究面临着的挑战是将基因组学发现转换成应用基础知识、实践应用知识、临床治疗药物及其他医疗产品。
每个生命机体的遗传密码中都蕴藏着巨大的资源。
基础医学研究可以使人们进一步了解生命过程,而生物医学研究可以挖掘和开辟出疾病诊断、治疗和预防的新途径。
为了充分利用基因组学资源,有必要对生命细胞内的实际功能分子(如DNA序列编码的蛋白质)进行识别并了解其生物学特性,而自然界本身就已经设计和创造出了识别和分析千变万化的蛋白质的理想工具——抗体。
抗体作为蛋白质组研究的有力工具,广泛应用于基础研究和药物开发。
90年代初期,出现了DNA免疫动物制备抗体的技术。
既将含有目的基因的质粒DNA注射入动物肌肉、皮下或脾脏等部位,使得质粒上的遗传信息在动物体内表达,诱导动物体对DNA编码的外源蛋白产生抗体。
DNA免疫无须蛋白质纯化,故速度快,实现了高通量。
通过生物信息学和DNA免疫方法,基因特异抗体可以被设计为针对所给定蛋白的任何一个特定功能结构域,基因表达功能结构域特异性高。
通过各种DNA免疫,可于动物体内产生大量的基因特异性抗体。
这些抗体携带特定基因标识信息,是基因特异性抗体。
通过这些特定抗体与人体组织或细胞中的蛋白结合,从而进行定位和识别未知蛋白。
结合多种技术(如抗体与组织芯片技术)来识别不同生理和病理状态下、不同组织中的成千上万的未知蛋白(如蛋白表达谱)。
还可以得到基因、蛋白、细胞和疾病之间的整合及相互关联的数据。
通过本筛选过程,带有功能信息的新基因和蛋白会被发现。
其中一些基因和蛋白有可能成为有价值的药物靶点、治疗药物或诊断标记物。
此技术还可以用于治疗性抗体的制备。
DNA免疫动物制备抗体理论上,DNA免疫可制备出针对不同抗原的特异性抗体,从而引起人们广泛的兴趣。
有人通过应用DNA免疫来获得针对细胞表面分子的单体和多抗。
2-5次肌肉内注射可诱导产生抗CD54和CD157的抗体,经脾脏应用CD54编码DNA一次免疫即可诱导出抗CD54抗体,还制备出CD54单抗,所得CD54单抗可以和转染的COS细胞及其他细胞中表达的CD54蛋白反应。
英文-抗体工程
100 年前serum polyclinic antibody 1975 年2nd generation cell engineering antibody 1980 年3rd generation genes engineering antibodyhuman-mouse chimeric antibodyreshaped human antibodyantibody fusion proteinantibody libraryAntibodies are one of nature’s answers to the need for a versatile system to recognize and dispose of foreign organisms or substances.B cells, upon activation by antigen, differentiate into plasma cells, which secrete antibodies.All antibodies are categorized as immunoglobulins, B cell receptors are immunoglobulinsEach immunoglobulins molecule iscomposed of four interlinked polypeptide chain. The two long chains are call heavy chains, and the short ones, light chains.The immunoglobulins have a “stem”calledthe Fc portion andcomprising the lower halfof the two heavy chains,each “prong”containinga single antigen-bindingsite.Human monoclonal antibodies have been produced from human hybridomas for treatment The same technique can be used to clone human antibodies from transgenic miceAntibody engineeringA recombinant fragments engineered fordiagnostic and therapeutic applicationPseudomonas aeruginosa, widespread multi-drug resistant vaccine induced------production of human anti Pa flagellin antibody or Lipopolysaccharide(LPS) or toxin AHuman monoclonal antibodies for treating HCV infection, mainly to envelope protein (E2)Anti brain tumor human hybridomas antibodyused to clinical treatment over 10 yearsHuman monoclonal antibodies have beenproduced from human hybridomas for treatment The same technique can be used to clone human Antibodies from transgenic miceAntibody engineeringA recombinant fragments engineered for diagnostic and therapeutic applicationUsing the KM mouse to generated human monoclonal antibodies to carcinoembryonic antigen (CEA)40 to 65 % Ab-dependent cell-mediated cytotoxicity against the tumor cellsUsing the XenoMouse to generated monoclonal antibodies to Plasmodium falciparum42-kDa fragment of the Plasmodium falciparum merozoitesurface protein is one of the most promising vaccine candidate against the blood stage malarial parasiteHuman monoclonal anti-HCMV neutralizing antibody from KM mouse300 PFU of the virus were incubation with serial dilutions of asciteof KM mouse for 1 hr at 37°C, and used to infect the HEL cells. After adsorption for 1 hr at 37°C, the inoculum was assayed for plaques by counting, the results were shown that the plaques reductionHuman monoclonal antibodies have been produced from human hybridomas for treatment The same technique can be used to clone human Antibodies from transgenic miceAntibody engineeringA recombinant fragments engineered fordiagnostic and therapeutic applicationOwing to the broad application & wide perspective of monoclonal antibody, important advances in design, selection, and production of engineered antibodies have been madeTraditional method for recombinant monoclonal antibody construction like hybridoma technologyhas many limitation, such as duration, stabilityand class manipulationNew technology like the display of antibody fragments on the surface of filamentous phage & the subsequent selection of antibodies have been proved as an effective tool for the isolation of antigen specific antibodies, it still has its limitation小分子抗体的优点:分子小、穿透力强、廓清快、异源性低、易于大量生产可用于:靶向载体构建其他工程抗体细胞内抗体肿瘤、HIV …Practical considerationVectorPCR primersLibrary size ( titer)Efficient of ligation, the amount of DNA Screening of Fab clones (methods) AntigenRibosome display systemRibosome display has a number of advantages over phage display resulting in significantly shorter timelines for therapeutic drug developmentLarge variant libraries (more than 1012)Tailored folding conditionsElute mRNA from high affinityicovalent interactions Introduce mutation between selectionRapid cycling between selectionM 1 2 3 4 5 610 µmA B C DNative antigensRecombinant antigensTACZMES antibody technologyTaczmes are target-activated enzymes. Beta-lactamases (内酰胺酶)are the most useful enzymes identified to date. Beta-lactamases/Beta-lactamases inhibitor protein combination have been fused to human antibody libraries and target antigens These constructs, expressed in the E.coli periplasm, can facilely identify antigen binders, affinity mature selected antibodies or humanize non-human antibodiesExpression systemBacteria: glycosylated antibodies (cannot)Yeast: complement-mediated lysis (defective) Plants: carbohydrate structures (very differenet) Inscet cells: carbohydrate structures (very differenet) Mammalian cells:Applications of monoclonal antibodies to the study ofmolecules in solution ELISA, immunoprecipitationprotein epitope delineationassays of antibody-bound antigen immunoaffinity chromatographyApplications of monoclonalantibodies in microbiology viruses(as regents, choice of test formats(IFA) anti-HCMV neutralizing antibody,anti-rabies virus neutralizing antibodyanti-HBV, HCV neutralizing antibody,HIV(gp41, gp36, V3 domain of gp120)bacteria(diagnosis, antigen detection,particle based)Pseudomonas aeruginosa,anti-ICAM-1(intercellular adhesion molecule) parasitic & fungal infection (antigen detection, inhibition)Applications of monoclonal antibodies to the study of cells and tissuesantibody-based cell separationantibody-based cell destruction or agglutinationanalysis of functionanti-tumor necrosis factor-aipha monoclonal antibody to tumorFab of human autoantibodies。
单克隆抗体纯化工艺流程
单克隆抗体纯化工艺流程英文回答:Single clone antibody purification process is a crucial step in the production of monoclonal antibodies. Itinvolves several steps to ensure the isolation and purification of the desired antibody from the mixture of other proteins and contaminants.The first step in the purification process is toharvest the cells producing the monoclonal antibody. This can be done by centrifugation or filtration to separate the cells from the culture medium. Once the cells are separated, they are lysed to release the intracellular contents, including the antibody of interest.After cell lysis, the next step is to remove celldebris and insoluble components from the lysate. This canbe achieved through centrifugation or filtration methods. The clarified lysate is then subjected to a series ofchromatography steps to purify the antibody.One commonly used chromatography technique is protein A affinity chromatography. Protein A is a bacterial protein that has a high affinity for the Fc region of immunoglobulin G (IgG) antibodies. The lysate is passed through a column packed with protein A resin, and the antibody binds to the resin while other impurities are washed away. Elution of the antibody is then performedusing a low pH buffer or an IgG-specific elution buffer.Another purification step is ion exchange chromatography, which separates proteins based on their charge. The lysate is loaded onto an ion exchange column, and the antibody is selectively retained while otherproteins are washed away. Elution of the antibody is achieved by changing the pH or ionic strength of the buffer.Size exclusion chromatography is also employed to remove aggregates and further purify the antibody. This technique separates proteins based on their size, withlarger molecules eluting first. The antibody is collectedin the void volume while smaller impurities are separated.Finally, the purified antibody is subjected to a viral clearance step to ensure the removal of any potential viral contaminants. This can be achieved through viral filtration or other viral inactivation methods.Once the purification process is complete, the antibody is typically formulated into a suitable buffer for storage and further use. The purity and quality of the antibody are assessed using various analytical techniques, such as SDS-PAGE, HPLC, and ELISA.Overall, the single clone antibody purification process involves a series of steps, including cell harvesting, cell lysis, clarification, chromatography, viral clearance, and formulation. Each step is designed to isolate and purify the antibody of interest while removing impurities and contaminants.中文回答:单克隆抗体纯化工艺流程是单克隆抗体生产过程中的关键步骤。
单抗药物发展趋势及工程设计分析
当代化工研究Modem Chemical Research167 2021・03百家争鸣单抗药物发展趋势及工程设计分析*陈凯(中国电子系统工程第二建设有限公司江苏214072)摘耍:单抗产业正进入关键的发展时期,未来随着医保对于单抗药物的覆盖加大以及国内单抗产业餉发展,市场将继续维持高速增长.本文简要介绍了单抗药物生产过程中上游工艺、下游工艺和制剂工艺的工艺流程,并对上游工艺和下游工艺的主要工艺区域进行详细介绍,最后结合人物流设计、空调系统及公用工程系统设计对单抗生产车间的设计做简要分析°关键词:单抗;生产工艺;人物流;空调系统;公用工程系统中国分类•号:T文献标识码:ADevelopment Trend and Engineering Design Analysis of Monoclonal Antibody DrugsChen Kai(China Electronic Systems Engineering Second Construction Co.,Ltd.,Jiangsu,214072) Abstract:Monoclonal antibody industry is entering a critical development period.In the Juture,with the increasing coverage of m edical insurance for monoclonal antibody drugs and the development of domestic monoclonal antibody industry,the market will continue to maintain rapid growth.This paper briefly introduces the process/low of u pstream process,downstream process and p reparation process in the production of monoclonal antibody,and introduces the main process areas of u pstream p rocess and downstream p rocess in detail.Finally,the design of m onoclonal antibody p roduction workshop is briefly analyzed in combination yvith the design of h uman logistics,air conditioning system and utility system.Key words:monoclonal antibody\production technology^human logisticsi air conditioning system^utility system单抗是单克隆抗体药物的简称,单克隆抗体药物在生物技术制药中占有重要地位,并逐渐成为生物医药领域发展的主要方向。
Fed-batchprocessdevelopment:分批补料工艺的发展
Fed-batch process developmentfor monoclonal antibody production with cellferm-pro®Authors: Anna Frison and Dr. Klaus Memmert, Novartis Pharma AG, Basel, Switzerland Published in Genetic Engineering News, Volume 22, Number 11, June 1, 20021 Introduction:Cost-effective large-scale production of monoclonal antibodies (mAb) is creating a strong demand for reliable and prompt development of highly productive, scalable processes. One of them is fed-batch, which is widely used for production of recombinant proteins, due to its operational simplicity, reliability, and flexibility in multipurpose implementation. The major advantage of fed-batch, comparing to batch, is the ability to increase maximum viable cell concentration, prolong culture lifetime, and allow product accumulation to a higher concentration.The maximisation of final product concentration in a hybridoma fed-batch process is a function of the Integral of Viable Cells Concentration (IVCC). It follows, that an increase of this variable through feeding strategy optimisation will boost the final product titer.Pre-defined feeding protocols, which are based on nutrient requirement estimations, e.g. stepwise or a sigmoid-based addition of nutrient concentrates are unlikely to meet the nutritional demands of cells growing in batch culture which vary with time and environmental conditions. It often leads to nutrient depletion or accumulation of substrates or metabolites to inhibitory levels (Zhou et al., 1996). A much better way for nutrient supply would be the online monitoring of the cell culture’s metabolic activity, and a real time control of nutrient feeding based on this parameter.The novel feeding protocol discussed here is based on the oxygen uptake rate (OUR) on-line estimation. OUR is a very important indicator for the metabolic activity of the biological system. In cell cultures it correlates with the glucose or glutamine consumption rate. In our experiments OUR was used as the control parameter for adapting the feed rate according to the cell culture’s requirements in real time. The experiments were performed using the novel cellferm-pro®system.2 cellferm-pro® system2.1 System’s assembly:The cellferm-pro® system (DASGIP AG, Juelich, Germany) consists of five modules as shown in Fig.1.Figure 1:cellferm-pro®, a parallel and fully controlled cultivation system(DASGIP AG, Juelich, Germany)The culture system comprises of a temperature controlled incubator with 4 or 8 vessels, placed on a magnetic stirrer platform, equipped with pH and pO2 electrodes, and feed and air supply/removal connections.The gassing system provides an individual mix of up to four gases to each culture vessel (e.g. compressed air, oxygen, nitrogen and carbon dioxide). O2 and CO2 gas concentration are controlled by feedback from pO2 and pH electrodes. There is an electronic mass flow control and a gas totalizer function for each vessel.The monitoring system simultaneously processes the signals from pH and pO2 electrodes, regulates pH and pO2 in the medium through adjustment of CO2 and O2 in the gas mixture to maintain the set-points, and determines the OUR online.The dosing system offers an individual and regulated delivery of a feed medium. The dosing proceeds according to user defined profiles or fully automated based on the online determined OUR (Metabolic Activity based feeding).The control system is based on Microsoft Windows. Here pH, pO2, gassing and dosing with user defined profiles are configured and the calibration of the electrodes and dosing system performed. The analyses of logged data are done using DASGIP’s ChartWizard for MS Excel®.2.2 Metabolic Activity Tool:A sophisticated algorithm computes the OUR online for each culture vessel of the cellferm-pro® system. Online OUR, supplemented with known concentration of one important feed medium component and pre-estimated ratio between substrate supply rate and oxygen uptake rate, allows an individual and automated addition of liquid media according to the actual metabolic activity of the cells.3 ExperimentalFed-batch processes were performed with the cellferm-pro® system using the Metabolic Activity tool, with an a priori determined ratio between the substrate supply rate and the oxygen uptake rate (Y S/Y O2). The criteria for the choice of an optimum Y S/Y O2 ratio were: optimum cell growth (maximum IVCC), maximum final product titer and minimum lactate produced per glucose consumed, since lactate is supposed to be one of the main inhibitors in hybridoma cell cultures.As a reference for the Metabolic Activity based fed-batch processes served a standard batch process, also performed in cellferm-pro®.3.1 Culture conditions:A recombinant mAb secreting GS-NS0 cell line has been used in the present work as a model system for suspension cell culture. All cultures were carried out in a proprietary serum-free, glutamine-free medium based on Iscove’s Modified Dulbecco’s Medium (IMDM, Amimed). For the automated Metabolic Activity based feeding a 10-fold concentrated basal medium was used. Additionally, 20-fold concentrated Iscove’s amino acids solution (IMDM/AA, Amimed) was dosed manually once a day during four subsequent days after the cell density reached 1x106 cells/mL. In the first cultivation days NH4HCO3 was dosed to the culture to maintain NH4+ concentrations at about 0,5 mM.The culture vessels used in the cellferm-pro® system were 1 L Spinner flasks, equipped with glass ball agitator, pH and pO2 sensors and sampling ports, with a minimum start volume of 300 mL and a maximum working volume of 600 mL.3.2 Analytic:The concentration of viable cells and viability were automatically determined using the Cedex® system (Innovatis GmbH, Bielefeld, Germany). The IVCC was calculated as described by Sauer et al. (2000).The concentration of the medium’s main components such as Glucose, Glutamine, Glutamate and Lactate, four ions (Na+, K+, Ca++, NH4+), together with pH, pO2, pCO2 and osmolarity were determined using a Nova Bioprofile TM 200 Analyzer (Nova Biomedical Corp., Waltham, MA, USA)4 Results and discussion4.1 Choice of an appropriate substrate supply/oxygen uptake ratioGlucose, one of the most important energy sources for mammalian cell cultures, was chosen as the medium component on which the ratio of substrate supply rate to oxygen uptake rate (Y S/Y O2) was based. The theoretical ratio between substrate and oxygen uptake rates is 0.17 mol/mol, as 1 mole glucose requires 6 mole oxygen for complete oxidation. Since several essential substrates, except glucose, could also be limiting, four different Y S/Y O2 ratios were tested around the theoretical one in the range 0.1-0.5 mol/mol. The results are presented in Fig.2.Figure 2:Choice of the ratio between substrate supply rate and OUR (Y S/Y O2), based on the three criteria: IVCC, final product titer and minimal ratio of lactate produced perglucose consumed (Y LP/Y GC).For the YS/YO2 of 0.1 mol/mol the smallest IVCC was observed as well as a significantly lower titer. In this case cell growth ceased before the maximal working volume could be reached, assuming cell starvation (data not shown). The highest product titer of 260 mg/l was observed at YS/YO2 = 0.2. The YLP/YGC ratio increased with increasing YS/YO2. The most excessivelactate accumulation was observed for the ratio Y S/Y O2 = 0.5 mol/mol, representing in this case a theoretical maximum. Based on these observations, the Y S/Y O2 ratio of 0.2 mol/mol was chosen for the OUR-based nutrient feeding. This ratio is close to the theoretical one of 0.17 mol/mol, indicating the balanced concentration of nutrients in the feed medium.4.2 Metabolic Activity based fed-batchFig.3 shows a Metabolic Activity based feeding profile with the corresponding cell density and mAb titer over the process time. No lag phase in cell growth was observed. Exponential cell growth was observed for the first four days followed by a quasi-stationary phase of about 5 days. After the maximal working volume was reached on day 9 Metabolic Activity based feeding stopped and was replaced by stepwise feeding. In this phase the system continued evaluating the volume to be dosed, however, was only able to feed when a sample volume was taken and registered in the system as a negative value which could be replaced. In this manner, the cells obtained about 6 mL of feed once a day. Under these conditions the cells were kept alive at a cell density above 1,0 x 106 cells/mL for a period of 12 days. After day 15 the cells died and mAb accumulation stopped.Figure 3:Metabolic Activity based fed-batch cultivation: feeding profile, cell density and mAb concentration.4.3 Comparison to standard batch cultureIn a reference batch experiment, also performed with cellferm-pro® (data not shown) the process duration was only 7 days, during which no quasi-stationary phase was observed. A comparison of the standard batch process to the Metabolic Activity based fed-batch process is presented in Fig.4. As a result of improved and balanced nutrient supply the process duration could not only be increased by 196 %, more importantly the IVCC was increased by 285%, and the final product titer was increased by 209%. As the culture volume after feeding was doubled (600 mL) against the standard batch volume (300 mL) the total amount of mAb obtained was increased by 519%.Figure 4: Comparison of process duration, IVCC, titer and yield of Metabolic Activity based fed-batch with batch process.5 ConclusionsThe ideal method to adapt nutrient feeding in real time to the changing requirements of a cell culture would be online determination of a key substrate, e.g. glucose, and to use this parameter as input variable for feeding control. Online glucose determination in a sterile environment, however, is still not feasible for industrial applications. Lately a new instrument, the cellferm-pro® system, and the related Metabolic Activity software tool was introduced which implemented the OUR online determination as described by Ruffieux et al. (1998) and Ducommun et al. (2000) as a measure for glucose consumption. Using this instrument OUR can be used as the input variable for substrate feeding control. We have demonstrated that with the cellferm-pro®system and the Metabolic Activity tool a balanced and controlled nutrient supply to the cell culture is possible and that significantly improved mAb titer and productivity can be achieved. The cellferm-pro® system allowed a high experimental throughput, due to the possibility of running up to 8 experiments in parallel under the same environmental conditions. Additionally, we could use the data acquired from the cellferm-pro® process for easy scale-up of the fed-batch process to larger cell culture systems such as a 20-L Wave TM bioreactor. The cellferm-pro®system is thus a powerful tool for process development, optimization and validation.6 References:Ducommun, P., Ruffieux, P.-A., Marison, M.-P. F.I., von Stockar, U. (2000). A new method for online measurement of the volumetric oxygen uptake rate in membrane aerated animal cell cultures. Journal of Biotechnology 78, 139-147.Ruffieux, P.-A., von Stockar, U., Marison, I. W. (1998). Measurement of volumetric (OUR) and determination of specific (qO2) oxygen uptake rates in animal cell cultures. Journal of Biotechnology 63, 85-95.Sauer, P. W., Burky, J. E., Wesson, M. C., Sternard, H. D., Qu, L. (2000). A high-yielding, generic fed-batch cell culture process for production of recombinant antibodies. Biotechnol. Bioeng. 67(5), 585-597.Zhou, W., Bibila, T., Glazomitsky, K., Montalvo, J., Chan, C., DiStefano, D., Munshi, S., Robinson, D., Buckland, B., Aunins, J. (1996) Large-scale production of recombinant mouse and rat growth hormone by fed-batch CS-NS0 cell cultures. Cytotechnology 22, 239-250.。
制备多克隆抗体的流程
制备多克隆抗体的流程英文回答:The process of generating monoclonal antibodies involves several steps. Here is a general outline of the procedure:1. Immunization: The first step is to immunize an animal, typically a mouse or a rat, with the specific antigen of interest. The antigen can be a protein, peptide, or even a whole cell. The animal's immune system recognizes the antigen as foreign and mounts an immune response, producing a diverse population of antibodies.2. Cell Fusion: After a sufficient immune response is generated, the next step is to harvest immune cells, usually from the spleen, of the immunized animal. These cells, called B cells, are responsible for producing antibodies. B cells are fused with myeloma cells, a type of cancerous cell line that can divide indefinitely. Thisfusion creates hybridoma cells, which have the ability to produce antibodies and divide indefinitely.3. Selection: The fused cells are then cultured in a selective medium that allows only the hybridoma cells to survive. This medium usually contains a substance that prevents the growth of unfused cells and myeloma cells. The surviving hybridoma cells are then screened for antibody production.4. Screening: Screening involves testing the culture supernatants of the hybridoma cells for the presence of specific antibodies against the antigen of interest. Various techniques can be used for screening, such as enzyme-linked immunosorbent assay (ELISA) or immunofluorescence. Positive clones that produce the desired antibodies are selected for further analysis.5. Cloning: To ensure monoclonality, the selected hybridoma cells are subjected to limiting dilution, where single cells are distributed into individual wells of a culture plate. This process allows for the isolation ofindividual clones derived from a single cell. Each clone is expanded and tested for antibody production.6. Antibody Production: The selected monoclonal antibody-producing clones are grown in culture to produce a large quantity of antibodies. The antibodies can be harvested from the culture supernatant or purified using various techniques, such as protein A/G affinity chromatography.7. Characterization: The generated monoclonal antibodies are characterized for their specificity, affinity, and functionality. This involves further testing, such as Western blotting, immunohistochemistry, or flow cytometry, to determine the antibody's ability to recognize the target antigen.8. Scale-up and Production: Once the monoclonal antibodies are characterized, they can be scaled up for production. This involves large-scale culture of the selected hybridoma cells to generate a substantial amount of antibodies for research or therapeutic applications.中文回答:制备多克隆抗体的流程涉及几个步骤。
单克隆抗体
Monoclonal antibody technology allows us to produce large amounts of pure antibodies in the following way: We can obtain cells that produce antibodies naturally; we also have available a class of cells that can grow continually in cell culture. If we form a hybrid that combines the characteristic of "immortality" with the ability to produce the desired substance, we would have, in effect, a factory to produce antibodies that worked around the clock. In monoclonal antibody technology, tumor cells that can replicate endlessly are fused with mammalian cells that produce an antibody. The result of this cell fusion is a "hybridoma," which will continually produce antibodies. These antibodies are called monoclonal because they come from only one type of cell, the hybridoma cell; antibodies produced by conventional methods, on the other hand, are derived from preparations containing many kinds of cells, and hence are called polyclonal. An example of how monoclonal antibodies are derived is described below
Development of New Drugs
Development of New DrugsAimDrug development aims to produce a novel therapeutic agent which is superior in efficacy to existing remedies and which causes less frequent or less severe adverse effects.1.Evolution of a new drugThe development of a new therapeutic agent involves a multidisciplinary group in many years of work. Formerly, drugs were extracted from natural plant and animal sources. Therapeutic use was empirical and based on traditional experience. Over the last 80 years an impressive number of drugs have been synthesized chemically. With the development of genetic engineering and the production of monoclonal antibodies it is likely that even more agents will be produced artificially.Synthetic techniques have produced pure substances. This has led to increased specificity of action and, in some cases, greater efficacy and reduced toxicity. Unfortunately new drug development is expensive, and only a few substances (less than 1%) of those developed are actually marketed and used in practice.The range of novel chemical entities developed has occasionally led tounexpected toxicity. As a consequence, most governments have established bodies to regulate drug marketing, e.g. the committee on Safety of Medicines in Britain, and the Food and Drug Administration in the USA. These agencies supervise clinical research on new drugs and license new products. Although they serve to protect the public and are seen to do so, the statutory procedures that must be followed in applying for a license for a new drug add greatly to the costs and time of development.There is some evidence that the rate of introduction of entirely novel agents is slowing down. Whether this reflects economic pressures or diminished novel synthetic capacity or ability is not clear.2 Drug development strategiesSeveral strategies have been used in the development of new drugs. Over the years all have had success but no single approach has been consistently successful.Serendipity, luck an intuitionThis approach has been applied less frequently in recent years. The discovery of penicillin by Fleming was in this category.Molecular rouletteRandom chemical synthesis of new structures and pharmacological screening, this approach is wasteful and depends on the availability of sensitive animal or in-vitro models of human disease, which oftendo not exist.Minor structural changes in existing agentsOccasionally this leads to compounds of greater efficacy and rarely to drugs with novel actions detected in pharmacological screening or clinical practice.Programmed basic research with synthesis of specific chemical Intellectually this approach is the most satisfying. There have been spectacular results, e.g. levodopa and dopamine agonists in the treatment of Parkinsonism; beta-receptor blockers for angina; histamine (H2) antagonists in peptic ulcer disease; converting enzyme inhibitors in hypertension. However, this approach is expensive and this is no guarantee of success.Clinical observation of drug action in practiceThis is the traditional means of drug assessment. New applications arise from measurement of drug action in man in disease states. The antihypertensive effects of thiazide diuretics and beta-blockers were not predicted from animal screening tests. They were only identified after the drugs were available and were being used in practice.3. Experimental pharmacologyThese studies determine whether the drug has the desired profile of action in model systems. The models are selected to provide as reliable an index of efficacy in man as possible.Several models are usually employed. The models may be simple or complex and include:1)cell cultures or bacteria2)partially purified enzymes or sub-cellular particles3)isolated tissues4)perfused organs5)intact animals from mice to primates4. T oxicological assessmentIn parallel with pharmacological experiments on efficacy, the toxic effects of acute and chronic dosing are determined. Acute toxicity is less important as long as LD50 (the dose that kills 50% of animals) is not close to the ED50 (the dose causing 50% of maximal pharmacological response).Chronic toxicity testing is more relevant to clinical applications and should take place along the following lines:1)the route of administration, dose range, dose frequency andplasma levels should be appropriate to likely clinical indications.If possible, methods should be available to measure plasma concentrations and to determine patterns of metabolism.2)At least two species should be studied, usually dog and rat ormouse. If possible, a species should be selected with a similar profile of metabolism to man.3)The duration of treatment should be consistent with the likelyduration of use in man and the relative life expectancy of the animal species. Usually toxicity studies are undertaken over a period of 4 weeks to at least 1 year.4)Haematological and biochemical measurements should be madeserially. All tissues should be examined histologically at death or on sacrifice of the experimental group. An untreated control group of littermates should be maintained for comparison.Depending on the proposed patient group and disease indication, attention must be paid to:a.Effects on fertility in both males and females.b.Teratogenic effects on development of the embryo. The vulnerableperiod is very early in development, during organogenesis.c.Mutagenicity or an increased rate of mutation in germ cell lines ornonreproductive cells, e.g. bone marrow.d.Carcinogenicity or the induction or promotion or malignanttumours.There is disagreement over the relevance of some animal carcinogenicity studies to man.Extensive formal toxicological tests are now required in most countries before drugs can be used on patients. There is considerable controversy as to the value of routine toxicology testing, as manydifferences between species, especially between man and rat, mouse, and dog, have been reported.Paradoxically, thalidomide, which was the cause of the tragedy that led to stricter drug regulation and toxicology tests, is not teratogenic in mice or rats, but has a teratogenic effect in humans, causing gross limb deformities.5 clinical evaluationOnly after animal studies have proved efficacy, and toxicological studies have provided a measure of the possible risk, can new drugs be given to humans. At this stage a further requirement is analytical evidence of chemical purity and pharmaceutical stability. Evaluation in man can be considered in four phases. The relevance and extent of studies at these stages depends on the drug and its indications. Drugs for use in rare diseases, or in life-threatening and yet untreatable states, may be evaluated in patient groups at an earlier stage than those with readily measurable effects on common diseases.Phase I involves small scale studies in normal volunteers. These studies should determine whether the drug can be given to man without serious symptoms or toxicity, and whether it has the desired pharmacological effects. These studies often begin with a dose ranging study, using 1/50 to 1/100 the effective dose in animals andincreasing until the desired effect, or adverse effects, are seen. These studies should only be performed on volunteers who are informed about the implications of the tests, and who give their consent freely. Studies should include careful assessment of clinical, haematological actions and adverse effects. Phase I studies should only be performed by experienced staff, under medical supervision, and in premises with appropriate resuscitative facilities and support.Phase II studies determine whether the new drug has the desired effect on patients with the appropriate disease. In Britain these investigations can be performed only after submission of preclinical and phase I study results to the Committee on Safety of Medicines. This body either issues a clinical trial certificate (CTC) or authorizes limited clinical trials under an exemption procedure (CTE). Phase II studies initially may be open, uncontrolled, dose-ranging experiments but should include controlled studies under single or double-blind conditions. They may involve comparisons with inactive placebo or known active agents.Phase III If results of therapeutic efficacy and safety justify it, the next step is progression to large scale clinical trials to determine how the new drug compares in clinical practice with existing remedies, and to establish its profile of action and frequency if adverse effects. After Phase III studies the evidence from all stages of development isassembled and if the conclusions indicate a useful action, the drug may be submitted to the regulatory authorities with a request for a product license.The object is to identify therapeutically useful pharmacological activity and to characterize these actions using established models and drugs of known action.When drugs with specific actions on enzymes or receptors are being studied, relatively simple cell, free systems or isolated tissue preparations can be used. When poorly characterized subjective actions are sought, particularly involving behavioral effects, it may be necessary to perform tests in conscious intact animals.Phase IV A new drug is usually marketed after only a few hundred, or at the most a few thousand, patients have been exposed to it for a relatively short period (weeks or months). Post-marketing surveillance is increasingly undertaken to assess efficacy and toxicity of new drugs on a larger scale. No uniform scheme for Phase IV supervision has yet been established, but few doubt the necessity of collecting this information on low-frequency adverse effects.6. Marketing and promotionThe rationale for the development of new drugs should be to provide better drugs; better in the sense of being either more effective, safer or cheaper.Drug development is expensive. This is borne by the pharmaceutical industry, which justifiably expects to recoup the cost of development when the product is finally marketed. In some therapeutic areas, where drugs are widely used, e.g. antibiotics, nonsteroidal anti-inflammatory drugs, analgesics, antihypertensives, heavy investment in marketing and promotion has led to the use of undistinguished new drugs in place of equally effective, cheaper and established alternatives whose side effect profile is well known. Therapeutic fads and fashions should be avoided and prescribing practices change only when good evidence of improved efficacy or reduced toxicity is available.The physician needs guidance on critical assessment of what represents an important advance. Unfortunately, his most accessible source of information is the representative of the pharmaceutical manufacturer who has been specially trained and briefed to promote his particular new products; indeed his livelihood depends on the ability to do so.Practitioners must seek out alternative sources of information from district or regional information pharmacists, specialists, clinical colleagues, postgraduate meetings and publications in the scientific literature. Publications, in themselves, can be misleading. Evidence from a few controlled studies published in well-established journalssubject to peer review are more reliable than bulky obscure proceedings of sponsored meetings to promote a particular drug. Physicians should make an active attempt to determine in what way a new drug represents an improvement over existing therapy, and what is the price in terms of adverse effects and actual cost of the drug.As new drugs may be marketed after studies in only a few hundred or thousand patients, special vigilance is required in the first few years of use to determine low-frequency, but potentially serious adverse effects.CommentsNew drug developments should be examined critically; objective evidence from several sources should be sought to highlight improved therapeutic efficacy and reduced toxicity in controlled comparison with established remedies.V alue through Innovation is our Corporate VisionChronic obstructive pulmonary disease (COPD) is a major cause of death and disability throughout the world.COPD is a progressive respiratory illness characterised by chronic airflow limitation, shortness of breath (or dyspnoea),cough, wheezing and increased sputum (mucus or phlegm) production. These symptoms, in particular breathlessness, can restrict a patient's ability to perform normal daily activities. Smoking is the predominant cause of COPD, accounting for 80-90 % of the risk of developing chronic obstructive pulmonary disease. The disease is increasingly observed in women.Early diagnosis and treatment of COPD is essentialCOPD has a significant physical and emotional impact on those who suffer from the disease. As chronic obstructive pulmonary disease progresses, lung function declines over time and physical activity becomes severely limited, disrupting the patient's ability to lead a full life, interfering with everyday tasks, like participating in family routines and social interactions. Early diagnosis and treatment is important to help patients remain independent, prevent complications and exacerbations, and improve quality of life. Anticholinergics are a recommended first line recommendation for COPDAnticholinergics, a traditional core competence of Boehringer Ingelheim, are a first line recommendation for the management ofCOPD in many guidelines. Their qualities beyond bronchodilatation, i.e. improvement of dyspnoea, exercise tolerance, decreasing COPD exacerbations and improving health related quality of life for patients is the focus of ongoing modern clinical research. These improvements were to some extent already seen with short-acting anticholinergics that are marketed for a longer time, but have become greater with the more recent development of long-acting anticholinergics. In addition to these important benefits the long-acting anticholinergic tiotropium also demonstrated its ability to reduce the risk of death in the 4-year UPLIFT® trial. Further analyses from this long-term trial have shown how the clinical course of the disease can be positively affected when maintenance treatment with tiotropium is initiated as early as recommended in the GOLD Guidelines (Stage II) and have also shown the importance of initiating this treatment in patients previously naïve of maintenance therapy.The most effective intervention in COPD remains giving up smoking. Consequently smoking cessation should be standard treatment for COPD, accompanied by the usage of bronchodilators.ProductsSpiriva® (tiotropium)Maintenance treatment of patients with COPD (chronic obstructive pulmonary disease, including chronic bronchitis and emphysema); the maintenance treatment of associated dyspnoea and for prevention of exacerbations.Atrovent® (ipratropium bromide)Indicated as a bronchodilator for maintenance treatment of bronchospasm associated with chronic obstructive pulmonary disease, including chronic bronchitis, emphysema and asthma. Combivent® (ipratropium bromide/salbutamol)Indicated for the management of bronchospasms associated with reversible obstructive airway diseases in patients who require more than one bronchodilator.Berodual®Bronchodual® Duovent® (fenoterol/ipratropium bromide)For prevention and treatment of symptoms in chronic obstructive airway disorders with reversible bronchospasm, such as bronchial asthma, and especially chronic bronchitis, with or without emphysema.Devices/CFC TransitionThe Montreal Protocol and several supplementary legally binding international agreements lay down the gradual elimination of all production and use of ozone depleting substances, particularly chlorofluorocarbons (CFCs) which have been widely used as aerosol propellants. Of particular relevance to Boehringer Ingelheim is the switch from CFC-driven Metered Dose Inhalers to those propelled by the more environmentally-friendly hydrofluoroalkanes (HFAs). Boehringer Ingelheim has already launched HFA-products for their established bronchodilators Be rotec®, Berodual® and Atrovent® in many countries, and has also given a lot of attention to the development of an alternative inhaler device that is propellant-free.Respima t® Soft Mist™ InhalerThe result is the Respimat® Soft Mist™ Inhaler, a highly innovative approach to inhaler technology that is designed to meet patients' needs and is also environmentally friendly. To learn more about Respimat® Soft Mist™ Inhaler we invite you to visit the global Respimat® website.Cardiovascular disease is the leading cause of mortality and morbidity worldwideCardiovascular disease (CVD) is responsible for nearly one in three deaths worldwide and is the number one cause of death. Advances in CVD research during the past two decades have resulted in an improved understanding of the chain of events that lead to end-stage heart disease.Information on Cardiovascular DiseaseIn Europe, CVD causes nearly half (48%) of all deaths, and in the US, two Americans die every minute from CVD. About half of all deaths from CVD are from coronary heart disease and nearly one-third are from stroke. Each year, 15 million people worldwide suffer strokes and 5 million are left permanently disabled (sources: WHO, AHA, World Health Report).Risk reduction in patients at high cardiovascular riskAdvances in CVD research during the past two decades have resulted in an improved understanding of the chain of events that lead to end-stage heart disease. The progression of CVD can be regarded as a continuum: from the onset of hypertension, increased low-density lipoproteins and type 2 diabetes,through atherosclerosis and cardiac remodelling, to end-organ failure with resultant myocardial infarction, stroke, heart failure and end-stage renal disease. The risk of a cardiovascular (CV) event increases with the number of CV risk factors present. Activation of the renin-angiotensin system (RAS), via angiotensin II, is implicated at all stages of this cardiovascular continuum. Blockade of the RAS, with angiotensin-converting enzyme (ACE) inhibitors or angiotensin II receptor blockers (ARBs), reduce blood pressure and also interrupt the progressive vascular damage associated with hypertension and other cardiovascular and metabolic diseases. ProductsMicardis® (telmisartan)Indicated by the US Food and Drug Administration (FDA) for the reduction of the risk of myocardial infarction (heart attack), stroke, or death from CV causes in patients 55 years of age or older at high risk of developing major CV events who are unable to take ACE inhibitors.Indicated by the European Commission (EMEA) for the reduction of CV morbidity in patients with manifest atherothrombotic CVdisease (history of coronary heart disease, stroke, or peripheral arterial disease) or type 2 diabetes mellitus with documented target organ damage.Cardiovascular diseases remain the leading cause of premature death in the developed world.Acute Myocardial Infarction (AMI) is still the main cause of death in the industrial nations, with around three million people suffering from it every year. During the last decade, the treatment of patients with AMI has been revolutionised. Time is an important factor for successful treatmentThe most important factor for a successful treatment of an acute myocardial infarction is time to treatment. Reality shows, however, that from onset of symptoms until treatment a lot of precious time is wasted. Delays are caused by the patient's hesitance to call for help as well as by ambulance response and journey times and slow in-hospital procedures. Thrombolytic therapy has established itself as one of the most successful modern treatment options of an acute myocardial infarction. It is easy to apply, is available in all hospitalsand is considered safe in view of the serious nature of the disease.ProductsActilyse® (alteplase)∙Fibrinolytic treatment of acute myocardial infarction∙Thrombolytic treatment of acute massive pulmonary embolism ∙Catheter clearance due to thrombotic occlusionActilyse® is also registered for the indication acute ischaemic stroke.Metalyse® (tenecteplase)∙Fibrinolytic treatment in acute myocardial infarctionHerba Commelinae; dayflower herbOseltamivirphosphate达菲T amiflu在罗氏提交美国联邦食品和药品管理局的申报材料中指出,奥司他韦(达菲)主要的不良反应显示为消化道的不适,包括恶心、呕吐、腹泻、腹痛等,其次是呼吸系统的不良反应[3-4],包括支气管炎、咳嗽等,此外还有中枢神经系统的不良反应,如眩晕、头痛、失眠、疲劳等。
宿主的特异性防御-免疫反应Specific Defenses of the Host-The Immune Response
• Lock and Key Concept
• Non-covalent Bonds
– Hydrogen bonds – Electrostatic forces – Van der Waal forces – Hydrophobic forces
:85/chime2/lyso-abfr.htm
Keq =
104 Affinity
106 Avidity
1010 Avidity
The ability of an individual antibody combining site to react with only one antigenic determinant. The ability of a population of antibody molecules to react with only one antigen.
ANTIGEN ( Intact cell/ Whole cell membrane/ micro-organisms ) + ADJUVANT (emulsification)
Ab titre reached in Serum Spleen removed
(source of cells)
PRODUCTION OF MONOCLONAL ANTIBODY
• Multiple Bonds • Reversible
Recommendationsf...
Recommendations for Monoclonal Antibody ProductionMonoclonal antibodies, following the development of this technology in 1975, have become an extraordinarily important resource for medical research, diagnosis, therapy, and basic science. In recognizing the overwhelming importance of this technology to modern medical science, it is also important to recognize that monoclonal antibody production is still largely dependent on the use of experimental animals.In November 1997, The Office of Laboratory Animal Welfare (OLAW) at the National Institutes of Health, forwarded a letter to all Public Health Service (PHS) awardee institutions and Institutional Animal Care and Use Committees (IACUCs) on avoiding or minimizing discomfort, distress, and pain in the care and use of animals for the production of monoclonal antibodies using mouse ascites method since there is evidence that the ascites method of monoclonal antibody production causes discomfort, distress, or pain. Accordingly, the IACUCs at all Public Health Service awardee institutions must critically evaluate the proposed use of the ascites method. The Committee must determine that a) the proposed use is scientifically justified, b) methods that avoid or minimize discomfort, distress, and pain (including in vitro methods) have been considered, and c) the latter have been found unsuitable. Fulfillment of this three-part IACUC responsibility, with appropriate documentation, is considered central to an institution's compliance with its Animal Welfare Assurance and the PHS Policy.In keeping with the need to minimize both the use and the discomfort and pain inflicted on experimental animals, the following considerations and recommendations in the application of monoclonal antibody technology are proposed.Monoclonal antibody technology relies on experimental animals for two basic steps. The first step involves immunizing mice. Although there are alternatives to this step (either by in vitro immunization steps or using recombinant libraries), these alternatives have not proven to provide the same level of efficiency, specificity, or affinity of antibody available from traditional immunization protocols. In addition, with the advent of alternatives to Freund's adjuvant, less than minimal or slight pain or discomfort accompanies this step and therefore there is no compelling reason to consider alternatives at this time. Careful consideration of the total number of required animals should be the investigators main concern in justification of this step.A second common step that uses animals comes in antibody production. In order to produce large quantities of monoclonal antibody, it has been traditional to grow hybridoma cell lines as an ascites tumor in vivo. In general, this procedure produces antibody titers approximately 1000-10,000-fold higher than those obtained in tissue culture. Since 1-10 ml of ascites fluid containing 1-5 mg/ml specific antibody can be obtained per animal while hybridoma cells in culture produce only 0.5-5µg/ml, one mouse can produce antibody equivalent to between 1 and 100 liters of tissue culture fluid. The main advantages of ascites are the extremely high yield of antibody and that the method is not excessively labor-intensive. However, its main disadvantage is the potential pain and discomfort caused to animals, due to painful peritonitis, abdominal tension, and infiltratively-growing tumors. It should also be noted that the monoclonal antibody produced by this method is contaminated by endogenous immunoglobulin and has the potential for contamination by viruses or bioreactive cytokines that may interfere with later use.There are in vitro alternatives to monoclonal antibody production by ascites. These include standard static or agitated suspension cell cultures, membrane-based and matrix-based culture systems, and high cell-density bioreactors. The disadvantages to these systems include their substantially greater effort and higher labor costs, increased costs due to the components of tissue culture media, and the poor growth and/or antibody secretion of some hybridoma lines in vitro. In particular, high cell-density bioreactors are probably beyond the capability of most laboratories due to the high initial and ongoing material and labor costs and the specialized expertise required.In light of the above considerations, the New York University IACUC requests that investigators consider the following recommendations in designing monoclonal antibody production.In general, in vitro methods for monoclonal antibody production are considered standard and accepted practice. The use of the ascites method requires rigorous and well- documented justification. Justifications based solely on cost or convenience will not be considered adequate.In addition, the NYU IACUC strongly urges the Medical Center to establish a monoclonal antibody core facility with high-density cell culture bioreactor capability that would obviate any need for continued use of the ascites method.1. Most applications require only small quantities and low concentrations of antibodies. Examples of common applications requiring only small quantities of antibody include immunoblots, immunoprecipitations, immunocytochemistry, flow cytometry, and small-scale unity columns. Such applications are easily accommodated by use of unpurified tissue culture supernatants or by monoclonal antibodies purified from tissue culture supernatants. For purposes requiring up to approximately 10-50 mg of antibody, standard tissue culture methods involving growth of up to 50 liters of hybridoma cells should be considered the method of choice. Proposed use of ascites for applications of this kind would require specific justification (e.g., use of a hybridoma with unfavorable growth characteristics in vitro) in addition to the considerations described in 3 (below).2. Small-scale membrane-based culture systems are available that facilitate production of monoclonal antibodies in the range of 10-100 mg per culture in 5-30 days. These systems are relatively inexpensive and do not require specialized facilities or expertise. Investigators with applications that require up to 1 g of monoclonal antibody should consider the use of this in vitro alternative for production. Use of ascites production for such applications would require specific justification demonstrating that special circumstances warrant use of the in vivo method. In addition to the considerations described in 3 (below), justification must demonstrate that the disadvantages of ascites production (including pain and discomfort to the animals and potential in vivo contamination) are outweighed by the specific requirements of the individual project.3. Production of greater than 1 g of monoclonal antibody by in vitro methods is probably outside the current capabilities of most laboratories. For these applications, ascites production is presently the only alternative for in-house production. Approval of such projects will require adequate justification for the required amounts of antibody. In addition, the investigator must demonstrate the required expertise for working with tumor-bearing mice, including an adequate daily monitoring system to insure that animals do not experience unnecessary pain or discomfort Assurance must be made that no animal will be allowed to develop tumors larger than 20% of host body weight. Ascites fluid must be harvested on a single occasion only, either under terminal anesthesia or post mortem. Animals must be killed without delay if they show more than mild distress, overt tumor deposits or spread, or significant dehydration or cachexia.4. The specific guidelines for consideration by Principal Investigators when developing animal study proposals and for Animal Care and Use Committees when reviewing proposals involving the mouse ascites method are:a. The volume of the priming agent should be reduced to as small a volume as necessary to elicit the growth of ascitic tumors and at the same time reduce the potential for distress caused by the irritant properties of the priming agent. Although 0.5 ml Pristane has been standard for adult mice, 0.1-0.2 ml has been found to be as effective for many hybridomas.b. The time interval between priming and inoculation of hybridoma cells as well as the number of cells in the inoculum are determined empirically. Inocula range from 105 -107 cells in volumes of 0.1 - 0.5 ml and are usually administered 10 -14 days after priming. Generally, very high concentrations are associated with greater mortality and concentrations < 1 x 105 cells elicit fewer ascitic tumors and these tend to have a smaller volume yield. Cell suspensions should be prepared under sterile conditions in physiological solutions.c. Hybridomas should be MAP (mouse antibody production) or PCR tested before introduction into the animal host to prevent potential transmission of infectious agents from contaminated cell lines into facility mouse colonies and possibly to humans handling the animals.d. Animals should be monitored at least once daily, seven days a week by personnel familiar with clinical signs associated with ascites production and circulatory shock.e. Ascites pressure should be relieved before abdominal distension is great enough to cause discomfort or interfere with normal activity. Manual restraint or anesthesia may be used for tapping. Aseptic technique should be used in withdrawing ascitic fluid. The smallest needle possible that allows for good flowIn accordance with the Animal Welfare Assurance and the Public Health Service Policy, and to appropriately document that investigators proposing the use of monoclonal antibodies have considered alternatives to minimize discomfort, distress, and pain, in future please refer to these recommendations before submitting protocols to the IACUC. Additionally, subsequent to the November 1997 OPRR Letter, the IACUC expects that investigators will incorporate these recommendations into all new applications submitted for funding.。
Enzyme-linkedImmunosorbentAssay(ELISA)_395108327
Solid-phase assays for antibodies employing ligands labelled with radioisotopes or enzymes(enzyme-linked immunosorbent assay; ELISA) are most widely used of all immunological assays.
Procedure of ELISA
Incubate microtitre plate well with antigen; Wash off unbound antigen; Incubate with antibody; Wash off unbound antibody; Incubate with labelled anti-immunoglobulin; Wash off unbound labelled antibody; Count/incubate with enzyme substrate solution.
Immunochemical techniques
The study of antibodies(and some other immunologically important molecules such as complement components) is known as immunochemistry.Such methods are known as immunochemical techniques.
Polyclonal antibody against HumanIgG-HRP (horseradish peroxidase) Substrate solution, OD490nm.
Guidance for Industry Monoclonal Antibodies Used as reagents in Drug Manufacturing
Guidance for Industry Monoclonal Antibodies Used as Reagents in Drug ManufacturingU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)March 2001Guidance for Industry Monoclonal Antibodies Used as Reagents in Drug ManufacturingAdditional copies are available from:Drug Information Branch, HFD-210Center for Drug Evaluation and Research (CDER)5600 Fishers LaneRockville, Maryland 20857(Tel) 301-827-4573(Internet) /cder/guidance/index.htmorOffice of CommunicationsTraining and Manufacturers Assistance, HFM-40Center for Biologics Evaluation and Research (CBER)1401 Rockville PikeRockville, Maryland 20852-1448(Fax) 888-CBERFAX or 301-827-3844(Voice Information) 800-835-4709 or 301-827-1800(Internet) /cber/guidelines.htmU.S. Department of Health and Human ServicesFood and Drug AdministrationCenter for Drug Evaluation and Research (CDER)Center for Biologics Evaluation and Research (CBER)March 2001TABLE OF CONTENTSI.INTRODUCTION (1)II.BACKGROUND (2)III.PRODUCTION OF MONOCLONAL ANTIBODY REAGENTS (3)IV.MONOCLONAL ANTIBODY REAGENTS IN DRUG MANUFACTURING (4)A.P URIFICATION OF D RUG S UBSTANCE (4)B.C OMPARABILITY (5)V.SPECIFICATIONS FOR MONOCLONAL ANTIBODY REAGENTS (5)A.T ESTING OF U NCONJUGATED M ONOCLONAL A NTIBODY R EAGENTS (6)B.T ESTING OF M ONOCLONAL A NTIBODY R EAGENTS L INKED TO S OLID S UPPORT (6)VI.STABILITY OF MONOCLONAL ANTIBODY REAGENTS (7)REFERENCES (8)GUIDANCE FOR INDUSTRY1Monoclonal Antibodies Used as Reagents in Drug ManufacturingI.INTRODUCTIONThis guidance is intended to provide recommendations to sponsors and applicants on the use of monoclonal antibodies (mAbs) as reagents in the manufacture of drug substances2 that are regulated by the Center for Drug Evaluation and Research (CDER) or the Center for Biologics Evaluation and Research (CBER). The guidance focuses on the chemistry, manufacturing, and control (CMC) issues that should be addressed in new drug applications (NDAs), abbreviated new drug applications (ANDAs), biologics license applications (BLAs), supplements to these applications, or investigational new drug applications (INDs).This document presents issues associated with and recommendations on the documentation to support the use of mAb reagents generated by hybridoma technology or production of recombinant mAb or their fragments in bacteria, including phage display technology, fungi (yeasts and molds), and nonprimate animal-derived transfected cell lines. Monoclonal antibodies or their fragments generated by other methods can present additional concerns. The recommendations provided in this document should be considered when such materials are used; however, the guidance does not address the particular method of production of the mAbs or their fragments.This document does not provide recommendations on mAbs that are used as diagnostics, radiolabeled imaging agents, or therapeutic products. For a discussion of mAb products for human therapeutic or diagnostic use please refer to the Points to Consider in the Manufacture and Testing of Monoclonal1 This guidance has been prepared by the Monoclonal Antibodies Working Group of the rDNA Reagent Technical Committee of the Complex Drug Substances Coordinating Committee (CDS CC) in the Center for Drug Evaluation and Research (CDER), with input from the Center for Biologics Evaluation and Research (CBER), at the FDA.2 The term drug substance, which is used throughout the text, is intended to include biological products as defined in 21 CFR 600.3(g).Antibody Products for Human Use (PTC 1997).3 The recommendations for characterization and testing for mAbs used as parenteral pharmaceuticals are by necessity stringent, and not all of them are applicable to mAbs that are used as reagents in drug manufacturing.II.BACKGROUNDMonoclonal antibodies are immunoglobulin molecules secreted from a population of identical cells (i.e., cloned cells). They are homogeneous in structure and binding specificity. In the context of this guidance, mAb reagents refers to monoclonal antibodies used as reagents in a drug substance manufacturing process.The issues related to mAbs used as reagents are somewhat different from those of mAbs used as parenteral therapeutic agents. For mAb reagents, the primary emphasis is on assessment of the following:C Biological safety, in particular the assessment of contamination of the mAb reagent withadventitious agents and/or process-related impurities from the cell substrate or cell line sources.C Performance characteristics of the mAb reagent during drug substance manufacture (e.g., avidityand specificity for the target molecule).C Potential presence of residual amounts of the mAb reagent in the final drug substance and/ordrug product.The recommendations in this guidance apply to the use of mAb reagents in the drug substance manufacturing process where the mAb reagent is used to purify the drug substance. The extent of characterization required for the mAb reagent depends on the nature of the steps that follow use of the mAb, and thus will vary among submissions. While many CMC concerns regarding the use of mAb reagents are unique to biotechnology-produced reagents, the general concepts expressed in the FDA Guideline for Submitting Supporting Documentation in Drug Applications for the Manufacture of Drug Substances (FDA 1987) also apply. An early and continued dialogue between the applicant and the Agency is encouraged to discuss the data that should be submitted to support the use of the mAb reagent.3 This document is available on the Internet at /cber/guidelines.htm.2III.PRODUCTION OF MONOCLONAL ANTIBODY REAGENTSThe sponsor or applicant should submit information (e.g., production process, specification) to support the use of the mAb reagent or a letter of authorization (LOA) to a drug master file (DMF) that contains this information.A description of the mAb manufacturing process should be provided. The description is used to assess the potential impact on the biological safety, quality, and purity of the drug substance and/or drug product. The mAb reagent should be adequately characterized and its identity, purity, and structural integrity should be assessed, as these factors are vital to its efficient and uninterrupted performance during production of drug substances (see section IV). Reagents that have not been fully characterized for viral safety should not be introduced into facilities where biologics and drugs from mammalian cell culture are produced because of the potential for cross-contamination. Additional recommendations relating to mAb reagents are:•For mAb reagents prepared using hybridoma propagation, serum additives in culture media should be free of contaminants and adventitious agents.•Manufacturers should use bovine-derived materials only from cattle that were born, raised, and slaughtered in countries that are free of BSE (bovine spongiform encephalopathy).4The predominant concern with the use of mAb reagents in drug substance manufacture is the introduction of adventitious agents (e.g., viruses, bacteria, fungi, mycoplasma) and/or process-related impurities (e.g., protein and DNA contaminants, column leachables, media components) into the drug substance. Of particular concern are those that are not removed during drug substance manufacture steps after the introduction of the mAb reagent. In many instances, the extent of the cell bank safety characterization and the clearance studies for adventitious agents and/or process-related impurities should follow the established standards for mAbs intended for human use (see PTC 1997, sections II.B and C). A reduced level (i.e., less than recommended in PTC 1997) of testing of cell banks and/or validation of the procedures used to remove or inactivate adventitious agents and/or process-related impurities during purification of the mAb may be appropriate under certain circumstances, with justification. Early dialogue with the Agency is encouraged when a reduced level of testing and/or validation is planned. A reduced level can be justified when, for example:4 A list of countries affected by BSE or those that have a substantial risk associated with BSE (due to a lack of implementation of an adequate surveillance program) can be found on the Internet at/NCIE/country.html.3C The drug product is terminally sterilized.C The use of the reagent is followed by adequate steps for the removal and/or inactivation of theadventitious agents and/or process-related impurities. In this instance, the overall assessment of the removal and/or inactivation process can take into account validation data from steps in the manufacture of the reagent and manufacture of the drug substance.C Processing steps downstream of the reagent include extremes of pH or organic solvents, andthere are reliable data in the scientific literature that the extremes remove and/or inactivateadventitious agents and/or process-related impurities.C The mAb reagent is produced in an expression system in which human infectious agents do notpropagate (e.g., plants, bacteria, fungi, insect cultures).IV.MONOCLONAL ANTIBODY REAGENTS IN DRUG MANUFACTURINGA major use of mAb reagents is in the purification of drug substance by mAbs attached to a solid support (e.g., immunoaffinity chromatography). Issues relating to and recommendations on the information to submit in support of the use of mAb reagents in the purification process are discussed below. The information that should be submitted to support other uses of mAb reagents in drug manufacture will depend on the use and are not discussed in this guidance. Sponsors or applicants with questions on documentation to support other uses of mAb reagents are encouraged to contact the Agency.A.Purification of Drug SubstanceThe drug substance purification processes should be described in the application. The drugsubstance manufacturer should establish a specification for the incoming mAb reagent, andperform testing before using the reagents in the manufacturing process. In addition to identitytesting for the incoming mAb reagent, drug substance manufacturers should carry out additional testing (e.g., binding activity, adventitious agents) to ensure that the reagent will perform asintended. Affinity and specificity studies are recommended to assess whether the characteristics of a mAb reagent are optimal for targeted binding to the appropriate substrate during themanufacture of the drug substance.Leaching of mAb or impurities from the solid support into the final product should beconsidered when specifications are established for the drug substance. The amount of column leachables is not uniform over the column lifespan and depends on several factors (e.g., length of storage, solutions used in the regeneration and/or sanitization steps, column operatingparameters). A variety of methods can be used to test for leachables such as sampling the4buffer flow-through prior to the load of the drug substance intermediate, in-process testing ofthe intermediate bulk, or testing the final drug substance. Alternatively, if documentation isavailable that the production steps that follow the use of the reagent mAb reduce the maximum amount of column leachables to appropriate levels, this documentation can be provided in lieu of routine testing for leachables.Data on the ability of the affinity column to achieve the intended purity under specified working conditions should be submitted. The stability of the mAb reagent during use, the columnperformance, and the microbial contaminants should be monitored during production of drugsubstance and documented by the drug substance manufacturer. Tests and acceptance criteria for residual mAb should be included in the specifications for drug substances processed withmAb reagents. Residual mAb should be monitored by sensitive and specific assay (e.g.,enzyme-linked immunosorbent assay (ELISA)).parabilityChanges in the mAb supplier or changes in the manufacturing process of mAb or solid support are considered to be drug substance manufacturing process changes that can have an effect on the biological safety and effectiveness of the drug substance and, consequently, the finalproduct. In cases where significant changes have been implemented in the mAb manufacturing process that may change the purity or the performance of the reagent (e.g., specificity, avidity,microbiological safety), appropriate product comparability testing should be performed. Theguidance document entitled FDA Guidance Concerning Demonstration of Comparability of Human Biological Products, Including Therapeutic Biotechnology-Derived Products(1996) contains a discussion of comparability testing for mAbs used parenterally.Comparability testing for mAb reagents should focus mainly on the performance characteristics of the reagent and its purity and stability. This is particularly important when changes in thereagent manufacture are likely to have an impact on the biological safety, purity, quality, orstability of the drug substance and/or drug product.V.SPECIFICATIONS FOR MONOCLONAL ANTIBODY REAGENTS Specifications for the mAb reagents should be provided. A certificate of analysis (COA) should be available for each individual reagent lot. For monoclonal antibodies linked to a solid support, COAs should be provided for both forms, unconjugated and linked. A copy of a representative COA should be provided.5The COA should provide the test results, including those for adventitious agents, expiration date, and a disclaimer statement in large bold lettering: REAGENT USE ONLY; NOT INTENDED FOR HUMAN USE.A.Testing of Unconjugated Monoclonal Antibody ReagentsTests to adequately characterize the unconjugated mAb reagent typically include:C Identity (e.g., reducing and nonreducing sodium dodecyl sulfate polyacrylamide gelelectrophoresis (SDS-PAGE) pattern, isoelectric focusing (IEF) profile)C Purity (e.g., high performance liquid chromatography (HPLC), SDS-PAGE, capillaryelectrophoresis)C Protein concentrationC Binding to the target moleculeC pHC Microbial and/or bacterial endotoxin limits, as appropriateC Preservatives, as appropriateB.Testing of Monoclonal Antibody Reagents Linked to Solid SupportTests for mAb reagents linked to solid support should include, at minimum, the following:C Physical characteristics (e.g., mean particle size, matrix structure)C Concentration of mAb (e.g., milligrams of mAb per gram of resin)C Specific binding capacity at recommended temperature and buffer rangesC Amount of leaching of mAbC Microbial and/or bacterial endotoxin limits, as appropriateC Preservatives, as appropriate6VI.STABILITY OF MONOCLONAL ANTIBODY REAGENTSThe mAb manufacturer should perform real-time stability studies of unconjugated and conjugated mAb. Based on these studies, the mAb manufacturer should determine and provide an expiry date for each lot of mAb reagent. Stability indicating tests should focus on performance and physical integrity of the mAb reagent. Either the drug substance manufacturer or the reagent manufacturer should provide data supporting the in-use chemical stability of the column and mAb reagent using the recommended storage buffer, regeneration and/or cleaning solutions under specific time and temperatures.7REFERENCESPoints to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use, FDA, 1997 (PTC 1997).Guideline for Submitting Supporting Documentation in Drug Applications for the Manufacture of Drug Substances, FDA, 1987.FDA guidance for industry on Demonstration of Comparability of Human Biological Products, Including Therapeutic Biotechnology-Derived Products, FDA, 1996.8。
System for production and screening of monoclonal
专利名称:System for production and screening of monoclonal antibodies发明人:Robert Pytela,Dongxiao Zhang,Weimin Zhu 申请号:US10266387申请日:20021007公开号:US20040067496A1公开日:20040408专利内容由知识产权出版社提供摘要:The invention provides methods for producing a monoclonal antibody in a host cell. The methods involve introducing linear expression cassettes containing coding sequences for immunoglobulin heavy and light chains into a host cell and expressing a monoclonal antibody. In most embodiments, the immunoglobulin heavy and light chains are both derived from a single antibody-producing cell. The invention further provides methods for producing a plurality of monoclonal antibodies, and methods of screening a plurality of monoclonal antibodies to identify a monoclonal antibody of interest and its encoding nucleic acid. Also provided by the invention are host cells containing monoclonal antibody-encoding sequences, and libraries of monoclonal antibodies for use in screening methods. The invention further provides kits for carrying out the subject methods. The subjects systems, methods and kits find use in a variety of different industrial, medical and research applications.申请人:PYTELA ROBERT,ZHANG DONGXIAO,ZHU WEIMIN更多信息请下载全文后查看。
MONOCLONAL ANTIBODIES AND VACCINE DEVELOPMENT DIRE
专利名称:MONOCLONAL ANTIBODIES AND VACCINE DEVELOPMENT DIRECTED TO HUMANCANCER-ASSOCIATED ANTIGENS BYIMMUNIZATION WITH ANIMAL ANDHUMAN MUCIN AND WITH SYNTHETICCARBOHYDRATE-CARRIER CONJUGATES 发明人:KJELDSEN, Thomas, J. The BiomembraneInstitute,CLAUSEN, Henrik TheBiomembrane Institute,SINGHAI, Anil TheBiomembrane Institute,TOYOKUNI, TatsushiThe Biomembrane Institute,TAKAHASHI,Helio The BiomembraneInstitute,HAKOMORI, Sen-itiroh TheBiomembrane Institute申请号:EP89903892.1申请日:19890310公开号:EP0357767B1公开日:19950524专利内容由知识产权出版社提供摘要:we have developed a new process for the production of monoclonal antibodies which bind to mucin glycoprotein antigens of cancer.(1) immunization of a host with a structure of core of mucin type glycoprotein; and (2) fusing splenocytes from immunized with host of myeloma cells to produce hybridomas cells; (3) cultivating said hybridoma cells in a medium s\u00e9lecti (f); and (4) selecting the hybridoma cells that survived inthe step (3), and an antibody binding to themthe structure of the nucleus of a mucin glycoprotein of said cells; and (5) hydridomes clone selected from the stage (4) and (6) of the hydridomes cloned cells cultivation; and (7) to get the antibody.described here are the hybridomas and monoclonal antibodies produced according to the process mentioned above, as well as active and passive immunization procedures employing the monoclonal antibody and mucin glycoproteins in combination with the type of oligosaccharide synthesis.申请人:BIOMEMBRANE INST地址:US国籍:US代理机构:Woodcraft, David Charles更多信息请下载全文后查看。
免疫球蛋白生产工艺流程
免疫球蛋白生产工艺流程英文回答:The production process of immunoglobulin involves several key steps. Firstly, the source of antibodies needs to be identified. This can be done through various methods such as isolating antibodies from the blood of individuals who have recovered from a specific disease or by using hybridoma technology to produce monoclonal antibodies. Once the source is identified, the next step is to purify the antibodies. This is typically done using chromatography techniques to separate and remove impurities. The purified antibodies are then concentrated to increase their potency.After purification, the antibodies are formulated into a final product. This involves adding stabilizers, preservatives, and other excipients to ensure the stability and efficacy of the immunoglobulin. The formulation process may vary depending on the intended use of the product, such as whether it is for intravenous or subcutaneousadministration.Once the formulation is complete, the immunoglobulin is filled into vials or syringes and undergoes final packaging. This includes labeling, serialization, and quality control testing to ensure that the product meets the required specifications and is safe for use. The packaged immunoglobulin is then stored under appropriate conditions until it is ready to be distributed.中文回答:免疫球蛋白的生产过程涉及几个关键步骤。
单克隆抗体完整流程
单克隆抗体完整流程The complete process of monoclonal antibody production is a complex and intricate one that involves several steps and requires a high level of precision and expertise. From initial antigen selection to final antibody purification, each stage of the process plays a crucial role in ensuring the production of high-quality monoclonal antibodies that can be used for various therapeutic and diagnostic applications.The first step in the production of monoclonal antibodies is the selection of an appropriate antigen. This involves identifying a specific protein or molecule that is capable of eliciting an immune response in an animal model. The chosen antigen should be highly specific to the target molecule or protein that the antibody will ultimately bind to. This step requires careful consideration and extensive research to ensure that the selected antigen will result in the production of monoclonal antibodies with the desired specificity and affinity.Once the antigen has been selected, the next step in the process is immunization. This involves administering the antigen to an animal, typically a mouse or a rabbit, in order to stimulate an immune response. The animal's immune system will recognize the antigen as foreign and produce a diverse array of antibodies in response. The antibodies produced by the animal will include a mixture of different specificities and affinities, and the next step is to isolate and characterize the individual B cells that produce the desired antibodies.To isolate the individual B cells, the animal is euthanized and its spleen is removed. The spleen contains a large population of B cells, and these cells are isolated and fused with a myeloma cell line to create hybridoma cells. These hybridoma cells have the ability to produce monoclonal antibodies with the same specificity andaffinity as the original B cells. The hybridoma cells are then grown in culture to produce large quantities of monoclonal antibodies, which can be harvested and purified for use in various applications.The purification of monoclonal antibodies is a critical step in the production process, as it ensures that thefinal product is of high purity and quality. Thepurification process typically involves several steps, including protein A or protein G affinity chromatography, ion exchange chromatography, and size exclusion chromatography. These techniques allow for the isolation and purification of the monoclonal antibodies from the culture supernatant, removing any contaminants orimpurities that may be present.Once the monoclonal antibodies have been purified, they can be used for a wide range of applications, including therapeutic treatments for various diseases and conditions, as well as diagnostic tools for the detection of specific proteins or molecules. The specificity and affinity of monoclonal antibodies make them valuable tools in the field of medicine and research, and the production process must be carefully controlled and monitored to ensure the quality and consistency of the final product.In conclusion, the production of monoclonal antibodies is a complex and multi-step process that requires careful antigen selection, immunization, isolation of B cells, fusion with myeloma cells, cell culture, antibody purification, and quality control. Each step in the process is crucial for the successful production of high-quality monoclonal antibodies that can be used for a variety of therapeutic and diagnostic applications. The production of monoclonal antibodies requires a high level of expertise and precision, and the resulting antibodies play a vital role in advancing medical research and improving patient care.。
抗体偶联药物生产流程
抗体偶联药物生产流程英文回答:Antibody-drug conjugates (ADCs) are a class of biopharmaceutical drugs that combine the specificity of monoclonal antibodies (mAbs) with the potency of cytotoxic drugs. The production process of ADCs involves several key steps, including antibody selection, drug payload conjugation, and purification.Firstly, the production process starts with the selection of an appropriate monoclonal antibody. This involves screening a large library of antibodies to identify the one that specifically targets the desired antigen. Once the antibody is selected, it is produced using recombinant DNA technology, typically in mammalian cell culture systems.Next, the antibody is conjugated with a cytotoxic drug payload. This step involves attaching the drug to theantibody through a linker molecule. The linker must be stable in circulation but capable of releasing the drug once the ADC reaches the target cells. Various conjugation methods, such as chemical conjugation or enzymatic conjugation, can be used depending on the properties of the antibody and the drug.After conjugation, the ADC is purified to remove any unconjugated antibody, free drug, and other impurities. Purification methods typically include chromatography techniques, such as size exclusion chromatography oraffinity chromatography, to separate the ADC from the impurities. The purified ADC is then formulated into a suitable dosage form for administration.Finally, the formulated ADC undergoes rigorous quality control testing to ensure its safety, efficacy, and stability. This includes testing for potency, purity, identity, and stability under various conditions. Once the ADC passes all the quality control tests, it can be packaged and distributed for clinical use.中文回答:抗体偶联药物(ADCs)是一类将单克隆抗体(mAbs)的特异性与细胞毒性药物的效力结合在一起的生物制药药物。
抗体药生产检验流程
抗体药生产检验流程英文回答:Antibody Drug Production and Testing Process.Introduction.Antibody drugs, also known as monoclonal antibodies (mAbs), are a type of biologic medicine used to treat various diseases, including cancer, autoimmune disorders, and infectious diseases. The production and testing of antibody drugs involve a complex and multi-step processthat ensures their purity, potency, and safety.Upstream Processing.The first stage of antibody drug production is upstream processing, which involves the development of a cell line that can produce the desired antibody. This is typically done using hybridoma technology, where a myeloma cell isfused with a B cell that produces the desired antibody. The resulting hybridoma cell line is then cloned and expanded to create a large population of cells that can produce the antibody.Bioreactor Culture.Once a stable hybridoma cell line has been established, it is grown in a bioreactor, a large-scale culture vessel that provides optimal conditions for cell growth and antibody production. During bioreactor culture, the cells are fed with a nutrient-rich medium and monitored for growth, viability, and antibody production.Downstream Processing.Once the cells have produced sufficient antibody, the culture is harvested, and the antibody is purified from the cell debris and other impurities. Downstream processing typically involves a series of steps, including centrifugation, filtration, and chromatography.Purification.The antibody is purified using various chromatography techniques, such as protein A or protein G chromatography. These techniques capture the antibody based on its specific affinity for these proteins. The purified antibody is then subjected to further purification steps to remove potential contaminants.Formulation and Fill-Finish.Once purified, the antibody is formulated into a stable and injectable form. This typically involves adding excipients, such as buffers, stabilizers, and preservatives, to ensure the antibody's stability and efficacy during storage and administration. The formulated antibody is then filled into vials or syringes and prepared for packaging.Quality Control Testing.Throughout the production process, the antibody drug undergoes rigorous quality control testing to ensure itspurity, potency, and safety. This testing includes:Identity testing to confirm the antibody's structure and specificity.Potency testing to determine the antibody's ability to bind to its target antigen.Safety testing to assess the antibody's potential toxicity and immunogenicity.Regulatory Approval.Before an antibody drug can be marketed, it must undergo regulatory approval by agencies such as the FDA or EMA. The regulatory review process involves a thorough evaluation of the drug's safety, efficacy, and manufacturing process.Post-Market Surveillance.Once an antibody drug has been approved, it continuesto be monitored for safety and efficacy through post-market surveillance programs. These programs collect data on the drug's use, adverse events, and long-term outcomes.中文回答:抗体药物生产和测试流程。
单克隆抗体和多克隆抗体相关书籍
单克隆抗体和多克隆抗体相关书籍英文回答:Single-clone antibodies and multi-clone antibodies are two types of antibodies that are used in research and clinical applications. There are several books that discuss the production, characteristics, and applications of these antibodies.One notable book on single-clone antibodies is "Monoclonal Antibodies: A Practical Approach" by R. H. Kennett, T. J. McKearn, and K. D. Bechtol. This book provides a comprehensive overview of the methods for producing and characterizing monoclonal antibodies, as well as their applications in various fields such as immunology, cancer research, and infectious diseases.Another important book on multi-clone antibodies is "Polyclonal Antibodies: Production, Applications and Advantages" by Abdul R. Jan. This book covers theproduction of polyclonal antibodies in various animal species, their purification and characterization, and their use in research and diagnostics.These books provide in-depth knowledge of the production and applications of single-clone and multi-clone antibodies, making them essential resources for researchers and professionals in the field of immunology and antibody-based therapeutics.中文回答:单克隆抗体和多克隆抗体是研究和临床应用中使用的两种抗体类型。
趋化因子CXCL1在巨噬细胞-肝癌微环境中表达情况及对肝癌进展的影响
趋化因子CXCL1在巨噬细胞-肝癌微环境中表达状况及对肝癌进展的影响背景:趋化因子CXCL1在多种癌症中发挥着重要的作用,然而其在巨噬细胞-肝癌微环境中的表达及其对肝癌进展的影响尚不清晰。
目标:本文的目标是探究CXCL1在巨噬细胞-肝癌微环境中的表达状况,并探讨其对肝癌进展的影响。
方法:我们接受免疫组织化学和实时荧光定量PCR技术检测了CXCL1在肝癌组织和对应癌旁组织中的表达,并对肝癌病人的临床病理学参数进行了统计分析。
结果:CXCL1在肝癌组织中表达显著增加,并与肝癌的大小、浸润度以及肿瘤分期呈正相关。
此外,我们也观察到CXCL1高表达的肝癌患者预后较差。
结论:CXCL1在巨噬细胞-肝癌微环境中表达提高,并且与肝癌的进展和预后密切相关。
因此,CXCL1可能成为肝癌治疗的一个新的靶点。
关键词:肝癌;CXCL1;巨噬细胞;微环境;预后Abstract:Background: Chemokine CXCL1 plays an important role in various cancers. However, its expression in macrophage-liver cancer microenvironment and itsimpact on liver cancer progression are still unclear.Purpose: The purpose of this study was to investigate the expression of CXCL1 in the macrophage-liver cancer microenvironment and explore its impact on livercancer progression.Methods: We used immunohistochemistry and real-time quantitative PCR to detect the expression of CXCL1 in liver cancer tissues and corresponding adjacent tissues, and analyzed the clinical pathological parameters of liver cancer patients.Results: CXCL1 was significantly upregulated in liver cancer tissues and positively correlated with the size, invasion degree and tumor stage of liver cancer. In addition, we also observed that liver cancer patients with high CXCL1 expression had a poor prognosis.Conclusion: CXCL1 was upregulated in the macrophage-liver cancer microenvironment and closely related to the progression and prognosis of liver cancer.Therefore, CXCL1 may become a new target for liver cancer treatment.Keywords: liver cancer; CXCL1; macrophages; microenvironment; prognosi。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
UNIT2.5 Production of Monoclonal AntibodiesHighly specific antibodies can be obtained by fusing immune B cells from the spleen withtumor cells to produce hybridomas,each of which will then secrete a single antibody.The desired antibody-producing hybridoma can be identified by a screening process.If this hybridoma is subjected to a cloning process in which clones are selected,suchthat all progeny are derived from a single cloned parental cell,a monoclonal antibodyis obtained.Monoclonal antibodies have high specificity and can be produced in largequantities.Thus,these biological reagents have been used extensively as probes in awide range of systems including the characterization of novel cell-surface and solubleproteins and carbohydrates,as enzyme catalysts,and for targeting in immunotherapy(see Commentary).This unit describes the production of monoclonal antibodies beginning with protocolsfor immunization(Basic Protocol1)and cell fusion and selection(Basic Protocol2).AnAlternate Protocol describes cell fusion and one-step selection and cloning of hybridomasutilizing a semisolid methylcellulose-based medium(ClonaCell-HY,a registered trade-mark of StemCell Technologies,Inc.;).Methods are providedfor screening primary hybridoma supernatants for antibodies of desired specificity(Sup-port Protocol1),establishment of stable hybridoma lines(Support Protocol2),cloningof these B cell lines by limiting dilution to obtain monoclonal lines(Support Protocol3),recloning of hybridoma cells in semisolid medium(Support Protocol4),and preparationof cloning/expansion medium(thymocyte-conditioned medium;Support Protocol5).Figure2.5.1summarizes these stages and notes the protocols in this and subsequent unitsin which they are detailed.Selection and cloning of hybridomas using the techniquesdescribed in Basic Protocol2require a major commitment of time and labor.However,the Alternate Protocol reduces the overall time necessary to produce monoclonal anti-bodies by18to20days.When successful,the monoclonal antibody may be an extremelyvaluable reagent that will be available in large quantities.Submission of monoclonal antibodies to the American Type Culture Collection(ATCC)for distribution to the scientific community is encouraged.Moreover,the ATCC servesas a repository for cell lines should the line be lost in the investigator’s laboratory due tounforeseen circumstances.NOTE:All protocols using live animals mustfirst be reviewed and approved by an Insti-tutional Animal Care and Use Committee(IACUC)and must conform to governmentalregulations for the care and use of laboratory animals.NOTE:All solutions and equipment coming into contact with living cells must be sterile,and proper aseptic technique should be used accordingly.NOTE:All culture incubations are performed in a humidified37◦C,5%CO2incubatorunless otherwise specified.Some media(e.g.,DMEM)may require altered levels of CO2to maintain pH7.4.Contributed by Wayne M.Yokoyama,Michelle Christensen,Gary Dos Santos,and Diane Miller Current Protocols in Immunology(2006)2.5.1-2.5.25Copyright C 2006by John Wiley&Sons,Inc.Induction of Immune Response2.5.1 Supplement74Production of Monoclonal Antibodies2.5.2Supplement 74Current Protocols in ImmunologyFigure 2.5.1Stages of monoclonal antibody production,with references to the Basic,Alternate,and Support Protocols in this unit (as well as subsequent units)that describe the steps.Induction of Immune Response 2.5.3Current Protocols in Immunology Supplement 74BASIC PROTOCOL 1IMMUNIZATION TO PRODUCE MONOCLONAL ANTIBODIESA wide variety of antigen preparations have been used successfully to produce mon-oclonal antibodies (see Critical Parameters for discussion of antigen preparation).The following protocol provides an immunization schedule for the production of most an-tibodies,although several different schedules can be used.In this protocol,emulsified antigen is injected intraperitoneally into the species of choice.A booster injection is administered 10to 14days after the primary immunization.Three days after the booster injection,the animals’spleens are ready for cell fusion (Basic Protocol 2).MaterialsAntigenComplete Freunds adjuvant (CFA;Sigma)Animal:pathogen-free mouse,hamster,or rat (Armenian hamsters from Cytogen Research are recommended;see Critical Parameters for discussion of animal choice and UNIT 1.1)Incomplete Freunds adjuvant (IFA;Sigma),optional1-to 2-ml glass syringes with Luer-Lok tips,sterile3-way stopcock20-and 22-G needles,sterileAdditional reagents and equipment for handling and restraint of animals (UNIT 1.3)and intraperitoneal injection (UNIT 1.6)CAUTION:CFA is an extremely potent inflammatory agent,particularly if introduced intradermally or into the eyes.Profound sloughing of skin or loss of sight may occur.Self-injection can cause a positive TB skin test and lead to a granulomatous e gloves and protective eyewear when handling CFA.1.Prepare antigen using 2×106to 5×107cells or 1to 50µg protein or peptide per animal to be immunized in normal saline.The antigen may be in several different forms depending on the desired property of the MAb and the method of screening (see Critical Parameters for discussion of antigen preparation and screening assays).If cells are the immunogen,wash three times in serum-free medium before immunization.Plan the immunization of several animals (enough for several fusions)so that primed and boosted animals will be ready 3days before fusion (see Basic Protocol 2).To minimize the risk of introducing a pathogen into the rodent colony,screen cells for pathogens by antibody-production assay (UNIT 1.1).2.Draw up antigen into a sterile 1-to 2-ml glass syringe with a Luer-Lok tip.Connect syringe to a 3-way stopcock.pletely resuspend CFA to disperse the Mycobacterium tuberculosis bacilli,which settle to the bottom of the container with time.Draw up a volume of CFA equal to the antigen volume in a syringe and connect to the antigen-containing syringe.4.Emulsify antigen and CFA by discharging antigen into CFA,then discharging back and forth until a thickened mixture results.Test whether the emulsion is stable—a stable emulsion will not disperse when a drop of it is placed in water.See UNIT 2.4for further discussion of immunization.Figure 2.4.1illustrates the double-syringe device.5.Transfer all of the CFA/antigen emulsion to one syringe and remove the other syringe and stopcock.Attach a sterile 20-G needle to the syringe containing the emulsion.Production ofMonoclonal Antibodies2.5.4Supplement 74Current Protocols in Immunology6.Inject emulsion intraperitoneally into the animal using <0.2ml/mouse,0.5to 1ml/rat,or 0.2to 0.4ml/hamster.Be careful not to force the syringe plunger,since excessive pressure may dislodge the needle and spray the emulsion.Introduce the needle through the skin and tunnel the needle between the skin and peritoneal wall before entering the peritoneal cavity at a site distant from the dermal puncture site.Twirl needle before withdrawal to minimize leakage.Rats are generally anesthetized (UNIT 1.4)whereas mice and hamsters can be manipulated with one hand and do not require anesthetic.7.Boost animal after 10to 14days with approximately the same dose of antigen as in step 5.If cell fusion is planned for 3days after boosting,immunize with antigen alone in aqueous solution,or intact cells in suspension.If a fusion is not immediately planned,boost the animal with antigen emulsified in IFA (which does not contain Mycobacterium tuberculosis bacilli).Do not use CFA for the booster immunizations as this will cause intense inflammation and increased anti-TB antibody response.Blood can be collected from a tail bleed (UNIT 1.7)after the first intraperitoneal boost to obtain antibodies from the blood as a positive control in subsequent detection assays.If desired,antibody titers can be assayed by ELISA (UNIT 2.1)or immunoprecipitation (UNIT8.3),7to 10days after the primary and booster immunizations.BASICPROTOCOL 2CELL FUSION AND SELECTION OF HYBRIDOMAS While animals should be immunized as soon as the decision has been made to produce a monoclonal antibody and the antigen prepared,do not perform cell fusion until the screening assay (Support Protocol 1)has been perfected.Artifactual results that may arise from conditioned media must be identified before cell fusion,because after a fusion there is only a finite amount of time available to assay for the desired monoclonal antibody.Prior to cell fusion,the partner (myeloma)cell line is expanded and a booster injection of antigen is administered to the primed animals.On the day of fusion,the spleens are harvested.Spleen cells and partner cells are washed,harvested,and mixed.Cell fusion is performed at 37◦C in the presence of polyethylene glycol (PEG).The resulting pellet is harvested and plated into tissue culture plates.After incubation with hypoxanthine,aminopterin,and thymidine (HAT)medium and feeding over ∼2weeks,the hybridomas are ready for screening (Support Protocol 1).Materials SP2/0-Ag14myeloma cell line (drug-marked,nonsecretory;ATCC #CRL 1581)Complete DMEM-10and -20media (APPENDIX 2A )with 10mM HEPES and 1mM sodium pyruvate Primed animal;mouse,hamster,or rat (10to 14days after primary immunization;(see Basic Protocol 1)Complete DMEM medium (APPENDIX 2A ),serum-free 50%polyethylene glycol (PEG),sterile Ammonium chloride solution (see recipe)Complete DMEM-20/HEPES/pyruvate/HAT (or HT)medium (see recipe)175-cm 2flasks Fine-mesh metal screen 50-ml conical polypropylene centrifuge tubes Beckman TH-4rotor or equivalent 96-well flat-bottom microtiter platesInduction of Immune Response 2.5.5Current Protocols in Immunology Supplement 74Additional reagents and equipment for animal euthanasia (UNIT 1.8),spleen removal (UNIT 1.10),and counting cells and assessing cell viability by trypan blueexclusion (APPENDIX 3B )Prepare myeloma cells (1week before fusion)1.One week before fusion,begin expansion of SP2/0-Ag14myeloma cell line (the fusion partner cell line)in complete MEM-10/HEPES/pyruvate (see Critical Pa-rameters).By the day cell fusion is to be performed,the following total number of myeloma cells must be available (in multiple 175-cm 2flasks containing 100ml each),depending upon the source of the primed animal:mouse spleen,1×108cells in two or three flasks;hamster spleen,2×108cells in three or four flasks;and rat spleen,5-10×108cells in ten flasks.Two mouse or hamster spleens,or one rat spleen,will provide enough cells for the fusion (see step 7).Boost primed animal (3days before fusion)2.Three days before fusion,boost primed animal(s)according to step 7of Basic Protocol 1.Prepare reagents and split myeloma cells (1day before fusion)3.One day before fusion,prepare all reagents and media,particularly 50%PEG.4.One day before fusion,split SP2/0-Ag14myeloma cells (from step 1)into fresh complete DMEM-10/HEPES/pyruvate medium.Vigorous growth of the SP2/0-Ag14cells is generally required for good fusion.Check myeloma cells and prewarm reagents (day of fusion)e an inverted microscope to check the SP2/0-Ag14myeloma cells to make sure they are growing vigorously (refractile and not pyknotic),they are not contaminated (no obvious bacteria or fungi),and there are enough cells for the fusion.It is better to postpone the fusion than to perform an ill-advised fusion,since the entire selection and screening effort will take ∼3weeks.6.Prewarm the following in a 37◦C water bath:Three 400-and three 600-ml beakers,each containing ∼100ml H 2O20ml sterile complete serum-free DMEM5ml sterile 50%PEG solution.Harvest spleen and prepare cells7.Sacrifice boosted animal(s)(UNIT 1.8)and aseptically harvest spleen(s)(UNIT 1.10).Do not use anesthetics for sacrifice.Instead,use cervical dislocation for mouse,or CO 2asphyxiation for mouse,hamster,or rat to avoid introducing an anesthetic into the bloodstream and therefore into the cultures.8.Transfer spleen to a sterile 100-mm-diameter petri dish filled with 10ml sterile complete serum-free DMEM.Perform all subsequent steps in a laminar flow hood.9.Tease spleen into a single-cell suspension by squeezing with angled forceps or by chopping with fine-tipped dissecting scissors.Remove debris and disperse cells further by passage through a fine-mesh metal screen.10.Transfer spleen cell suspension to a sterile 50-ml conical centrifuge tube and fillwith sterile complete serum-free DMEM.Do not use protein-or HEPES-containing medium because the PEG will precipitate proteins and HEPES can be toxic to cells during fusion.Production ofMonoclonal Antibodies2.5.6Supplement 74Current Protocols in Immunology11.Centrifuge 5min in TH-4rotor at 1500rpm (500×g ),room temperature,and discard supernatant.12.Lyse red blood cells (RBC)by resuspending pellet in 5ml ammonium chloride solution.Let stand 5min at room temperature.13.Add 45ml sterile complete serum-free DMEM,and centrifuge as in step 11.14.Resuspend pellet in 50ml sterile complete serum-free DMEM.Centrifuge as in step 11.Repeat DMEM addition and centrifuging once (each repeat is a wash).15.While spleen cells are being washed,separately harvest the SP2/0-Ag14myeloma cells (from step 5)by transferring the cells to 50-ml conical centrifuge tubes.Centrifuge as in step 11.Resuspend myeloma cells in DMEM and pool all cells into one 50-ml conical centrifuge tube.Wash myeloma cells three times as in step 14.16.Separately resuspend the spleen and myeloma cells in 10ml complete serum-free DMEM.Count cells and assess viability in each cell suspension using a hemacytometer and trypan blue exclusion (APPENDIX 3B );there should be nearly 100%viability of both suspensions.17.On basis of cell counts (from step 16),calculate the amount of complete DMEM-20/HEPES/pyruvate needed to plate cells at ∼2.5×106total cells/ml.Prewarm this amount of complete DMEM-20/HEPES/pyruvate in 37◦C water bath.Prepare 96-well flat-bottom plates by labeling them sequentially:one plate is required for each 10ml of final cell suspension.Perform cell fusion 18.Mix SP2/0-Ag14myeloma and spleen cells at a 1:1ratio in a 50-ml conical cen-trifuge tube.Fill the tube with complete serum-free DMEM.Other cell ratios work.Successful fusions have been performed with a ratio of myeloma/spleen cells as low as 1:20.19.Centrifuge cell mixture 5min at 500×g ,room temperature.20.While cells are in the centrifuge,prepare three 37◦C double-beaker water baths in the laminar flow hood by placing a 400-ml beaker (from step 6)containing 100ml of 37◦C water into 600-ml beaker containing 75to 100ml of 37◦C water.Place the tubes of prewarmed 50%PEG solution and complete serum-free DMEM (from step 6)into two of the 37◦C water baths in the hood.21.Aspirate and discard supernatant from the mixed-cell pellet (from step 19).22.Perform the cell fusion at 37◦C by placing the tube containing the mixed-cell pellet in one of the double-beaker water baths in the laminar flow ing a 1-ml pipet,add 1ml prewarmed 50%PEG to the mixed-cell pellet drop-by-drop over 1min,stirring the cells with the pipet tip after each drop.Stir for an additional ing a clean pipet,add 1ml prewarmed complete serum-free DMEM to the cell mixture drop-by-drop over 1min,stirring after each drop.Repeat once with an additional 1ml of prewarmed complete serum-free DMEM.25.With a 10-ml pipet,add 7ml prewarmed complete serum-free DMEM drop-by-drop over 2to 3min.Macroscopic clumps of cells should be obvious at this point.26.Centrifuge 5min at 500×g ,room temperature.Induction of Immune Response 2.5.7Current Protocols in Immunology Supplement 7427.While the cells are in the centrifuge,rewarm the beaker water baths to 37◦C andplace in the hood.Place prewarmed complete DMEM-20/HEPES/pyruvate (from step 17)in the beaker water bath.28.Discard the supernatant (from step 26).Place tube in the beaker water bath.29.With a clean 10-ml pipet,forcefully discharge 10ml prewarmed complete DMEM-20/HEPES/pyruvate to the cell pellet.30.Repeat step 29until the total volume of prewarmed complete DMEM-20/HEPES(calculated in step 17)is added.If necessary,allow clumps to settle and disrupt with the pipet tip.Further warming of cell suspension is no longer required.If the total volume exceeds 50ml,gently aspirate and transfer to another sterile container such as a tissue culture flask.31.Gently aspirate 10ml of cell suspension with a 10-ml pipet.Add 2drops (100to 125µl)of suspension to each well of a 96-well flat-bottom plate (continue until entire suspension is plated).Incubate overnight in a humidified 37◦C,5%CO 2incubator.Vigorous pipetting of the cell suspension should be avoided at this point,as the newly formed hybrids are unstable.Moreover,the vigorous addition of cells to the wells with repeating micropipettor is not e a pipet aid and hold the 10-ml pipet at a 45◦angle with the tip 1to 2cm above the well,bracing the pipet with a finger from the opposite hand.To avoid introducing contaminants,do not hold hands above the plate.A steady,even flow of drops from the pipet will allow the most efficient delivery of cell sus-pension or medium to the e a fresh pipet to withdraw additional cell suspension.As an optional step to minimize fibroblast overgrowth,permit the fibroblasts in bulk-fused cell suspension to adhere overnight to tissue culture flasks before seeding the 96-well plates.Many investigators select their hybridomas under bulk conditions—i.e.,they incubate large numbers of cells per well in larger plates or flasks.This makes feeding easier,but allows fast-growing hybridomas to overgrow the others.Since nonproducing hybridomas tend to grow faster,especially in the hamster-mouse fusions,hybridomas are isolated initially in multiple small wells in this protocol.The primary hybridomas tend to be monoclonal.This is especially important when screening procedures are used that require differential reactivities,e.g.,to different cell lines by flow cytometry analysis or to different antigen preparations.In those cases,multiple hybridomas per well will obscure the reactivity of the MAb of interest.Monitor and feed cells32.After one day of incubation,check wells under an inverted microscope.If seededwith the appropriate number of cells,there should be a nearly confluent monolayer of highly viable cells on the bottom and obvious clumps of cells.33.Add 2drops complete DMEM-20/HEPES/pyruvate/HAT to each well with a 10-ml pipet (see step 31).Place in humidified 37◦C,5%CO 2incubator.Use a separate pipet for each microtiter plate and keep the same covers with each plate to ensure that each plate remains a separate unit and to avoid spreading contamination.It cannot be overemphasized that it takes practice and meticulous attention to possible sources of contaminants to keep these plates sterile during the subsequent 2-to 3-week feeding and monitoring schedule.If plates become contaminated,discarding them is advised.Alternatively,contamination in one or two wells may be treated by aspirating the contents of the contaminated well with a sterile Pasteur pipet attached to a vacuum flask,rinsing the well with 70%ethanol,and wiping with a sterile cotton swab.Wash twice with ethanol.Finally,blot the well dry with the sterile cotton swab and blot the appropriate area of the cover with a sterile cotton swab soaked in 70%ethanol.Do not open contaminated plates while other plates are in the hood.Production ofMonoclonal Antibodies2.5.8Supplement 74Current Protocols in Immunology34.On days 2,3,4,5,7,9,and 11,aspirate half the volume of each well using a sterile,short Pasteur pipet attached to a vacuum flask,holding pipet at a 45◦angle and touching tip to surface of supernatant at the point where the liquid meets the opposite wall of the well.Feed the cells by adding 2drops complete DMEM-20/HEPES/pyruvate/HAT from a 10-ml pipet (see steps 31and 33)to each well,and return to humidified 37◦C,5%CO e a separate Pasteur pipet for each plate to minimize spreading contamination.Since the frequency of successful viable hybridoma formation is ≤10−5,when HAT is added,profound cell death should be apparent at days 2and 3and the remaining viable cells should not be readily apparent until they have expanded.By day 7to 9for mouse-mouse fusions,day 11for rat-mouse fusions,and day 14for hamster-mouse fusions,clusters of hybridoma cells should become visible under the inverted microscope.If profound cell death is not apparent on days 2and 3,check the medium containing HAT and the parental cell line by incubating an aliquot of the parental myeloma line with the medium containing HAT.The feeding schedule is not rigid except for the first 4days,when it is necessary to remove the toxic products of cell death.Thereafter,feedings will depend on the actual number of cells deposited in the wells,efficiency of fusion,and appearance and growth of hybridomas.Do not allow wells to become yellow (acidic)for more than a day.Examine plates daily,even if cells are not scheduled to be fed,and feed plates if acidic wells are noted.35.On day 14,repeat feeding as outlined in step 34except use complete DMEM-20/HEPES/pyruvate/HT to feed cells.Return to 37◦C,5%CO 2incubator.Cells do not require more than one change of complete DMEM-20/HEPES/pyruvate/HT.After this change,the aminopterin (from prior addition of HAT medium)is apparently diluted out enough so that the cells can survive without additional HT.36.On day 15and subsequently,feed wells as noted using complete DMEM-20/HEPES/pyruvate without HAT or HT.The hybridomas are ready for screening when most of the wells containing growing cells demonstrate 10%to 25%con-fluence and when those with denser populations turn yellow within 2days after feeding (see Support Protocol 1).If the screening assay requires a [3H]thymidine incorporation assay (APPENDIX 3D ),be aware that the large amount of thymidine in complete DMEM-20/HEPES/pyruvate with HAT and HT will serve as a cold-label inhibitor of [3H]thymidine incorporation.At least 3to 4changes of complete DMEM-20/HEPES/pyruvate without HT are required to dilute out excess thymidine.ALTERNATEPROTOCOLCELL FUSION,SELECTION,AND CLONING OF HYBRIDOMAS USING A SEMISOLID MEDIUM (CLONACELL-HY)Traditionally,monoclonal antibody development has involved selecting hybridomas in suspension cultures,identifying cultures that produce antibodies specific for the targeted antigen,and cloning the specific antibody-producing hybridoma(s)by at least one round of culture under limiting-dilution conditions (as described in Basic Protocol 2and Support Protocols 1to 3).This approach is laborious and time consuming and may result in the selection of identical,duplicate clones.This section describes protocols for the use of a methylcellulose-based medium system,ClonaCell-HY ,for cloning and selection of mouse hybridomas.Performing hybridoma selection and cloning simultaneously in ClonaCell-HY reduces the time and reagents necessary to obtain a monoclonal hybridoma producing antibody against the antigen of interest.Cultures do not need feeding or maintenance during the selection process.This approach also allows all daughter cells to remain together during the selection process,decreasing the number of clones that need to be tested for antibody production.Selection of duplicate hybridomas,a common occurrence with hybridoma cloning in suspension cultures,is avoided.An additionalInduction of Immune Response 2.5.9Current Protocols in Immunology Supplement 74advantage is that smaller,slow-growing clones,which can easily be lost due to overgrowth by larger,faster-growing hybridomas in traditional liquid suspension culture,remain physically separated in semisolid medium from the larger,faster-growing clones,and can thus be isolated and screened separately.NOTE:All solutions and media should be prewarmed to 37◦C unless otherwise indicated.MaterialsMyeloma cell line (e.g.,SP2/0,X63Ag8.653;available from ATCC)ClonaCell-HY Monoclonal Antibody Production Kit (StemCell Technologies,Inc.)containing:Medium A—ClonaCell-HY Pre-Fusion Medium and Hybridoma Expansion Medium,500mlMedium B—ClonaCell-HY Fusion Medium,500mlMedium C—ClonaCell-HY Hybridoma Recovery Medium,100mlMedium D—ClonaCell-HY Hybridoma Selection Medium containing HAT,90ml Medium E—ClonaCell-HY Hybridoma Growth Medium containing HT,500ml Polyethylene glycol—ClonaCell-HYPEGSolution,pretestedforcellfusion,1.5ml Immunized mouse,1to 4days after final antigen boost (Basic Protocol 1)3%(v/v)acetic acidLiquid nitrogen (optional)Fetal bovine serum (FBS)containing 20%(v/v)DMSO15-and 50-ml conical polypropylene centrifuge tubes100-mm petri dishesFine-mesh metal screenLow-speed tabletop centrifuge3-ml and 12-ml syringes25-and 75-cm 2tissue culture flasks16-G blunt-ended hypodermic needles96-and 24-well tissue culture platesCryotubes (e.g.,Nunc)Liquid nitrogen freezer (Dewar flask and canes to accommodate cryotubes;optional)Additional reagents and equipment for determining cell viability by trypan blue exclusion (APPENDIX 3B ),animal euthanasia (UNIT 1.8),spleen removal (UNIT 1.10),preparing a single-cell suspension of splenocytes (UNIT 3.1),counting cells using a hemacytometer (APPENDIX 3),assaying for antigen production from hybridoma clones by ELISA (UNIT 2.1),flow cytometry (Chapter 5),or immunoblotting (UNIT 8.10),and cryopreservation of cells (APPENDIX 3G )Prepare myeloma cells1.Culture the parental myeloma cells in 25-cm 2tissue culture flasks with Medium A (Pre-Fusion Medium from ClonaCell-HY kit)for at least 1week prior to fusion to ensure they are well adapted to this medium.Seed cells at a density of ∼5×104cells/ml and passage every 2days.Suggested maximum cell density is 4×105cells/ml.The parental myeloma cells must not secrete any of their own immunoglobulin chains.They should be mycoplasma-free and efficiently fuse to form stable hybridomas that continuously secrete specific monoclonal antibodies.Parental myeloma cells that meet these criteria (such as SP2/0and X63Ag8.653)are widely available.2.Calculate the cell growth rate at every passage (APPENDIX 3A ).The day before the fusion,count the viable cells and split cells so that there will be at least 2×107parental myeloma cells available the next day.The recommended cell density for fusion is 2×105cells/ml.Only 100ml of these cells will be needed,but 200ml should be cultured to ensure an adequate supply.Production of Monoclonal Antibodies2.5.10Supplement 74Current Protocols in Immunology 3.Harvest the required number of parental myeloma cells in a 50-ml conical polypropylene centrifuge tube.Centrifuge 10min at 300×g ,room temperature or 37◦C,and remove the supernatant.Wash three times,each time by adding 30ml of Medium B (Fusion Medium),centrifuging again as before,and removing the supernatant.Resuspend final pellet in 25ml Medium B.This step may be performed simultaneously with or subsequent to spleen cell preparation (steps 6to 10)to ensure that the myeloma cells do not sit for an extended period of time.It is important to remove all the serum adhering to the cells by washing with serum-free Medium B.If the serum is not removed,the PEG will not fuse the cell membranes and the fusion frequency will drop drastically.4.Count live cells using a viability stain (APPENDIX 3B ).Viability of parental myeloma cells should be >95%.5.Calculate the volume of cell suspension that contains 2×107cells,to be used in step 11.Place cells at room temperature or 37◦C.Harvest spleen and prepare spleen cells 6.Sacrifice immunized animal(s)(UNIT 1.8)and aseptically remove spleen(s)(UNITS 1.8&1.10).Place spleen in a sterile 100-mm petri dish containing 5ml Medium A (Pre-Fusion Medium).IMPORTANT NOTE:The ClonaCell-HY kit has been optimized for use with mouse hybridomas.Hybridomas from other species have not been tested.Do not use anesthetics for sacrifice.Instead,use cervical dislocation or CO 2asphyxia-tion to avoid introducing anesthetic into the bloodstream and therefore into the cultures.It is important to collect blood from the animal to obtain serum as a source of antibodies for a positive control in subsequent screening assays.Blood can be collected from a tail bleed (UNIT 1.7)after the first intraperitoneal boost (see Basic Protocol 1,step 7)or from the heart at the time of spleen harvest.To collect blood from heart use a sterile Pasteur pipet and place blood into a 1.5-ml microcentrifuge tube.Let blood stand at room temperature with the cap off to allow blood to clot.After 30min,remove the blood clot with a sterile Pasteur pipet and place tube at 4◦C.The next day,centrifuge tube for 15min at 400×g,4◦C or room temperature.Collect serum and add sodium azide to a final concentration of 0.1%.Store at −20◦C 7.Disaggregate the spleen into a single-cell suspension (UNIT 3.1).Transfer the spleen to a fine-mesh metal screen placed on top of a 50-ml conical centrifuge tube,and use the plunger of a 3-m1syringe to gently grind the cells out of the spleen.Rinse the screen with Medium B to help cells pass through the screen.Pipet the cells up and down in the tube with a 10-ml pipet to break up lumps.Try not to cause the solution to foam.Only the spleen membrane should be left in the screen.See UNIT 3.1for additional dis-cussion of the above procedure.Other spleen disaggregation methods may also be used.8.Centrifuge cell suspension 10min at 400×g ,room temperature or 37◦C,and remove supernatant.Wash the splenocytes three times,each time by adding 30ml Medium B,centrifuging 10min at 400×g ,room temperature or 37◦C,and discarding the supernatant.Resuspend the final cell pellet in 25ml Medium B.It is important to remove all the serum adhering to the cells by washing with serum-free Medium B.If the serum is not removed,PEG will not fuse the cell membranes and the fusion frequency will drop drastically.。