文档库 最新最全的文档下载
当前位置:文档库 › EMA-GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED therapeutic proteins

EMA-GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED therapeutic proteins

EMA-GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED therapeutic proteins
EMA-GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED therapeutic proteins

European Medicines Agency
London, 13 December 2007 Doc. Ref. EMEA/CHMP/BMWP/14327/2006
COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)
GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS
DRAFT AGREED BY BMWP ADOPTION BY CHMP FOR RELEASE FOR CONSULTATION END OF CONSULTATION (DEADLINE FOR COMMENTS) AGREED BY BMWP ADOPTION BY CHMP DATE FOR COMING INTO EFFECT
July 2006 January 2007 July 2007 October 2007 December 2007 April 2008
KEYWORDS
Immunogenicity, unwanted immune response, biotechnology derived proteins, immunogenicity risk factors, assays, clinical efficacy and safety, risk management
7 Westferry Circus, Canary Wharf, London, E14 4HB, UK Tel. (44-20) 74 18 84 00 Fax (44-20) 74 18 86 13 E-mail: mail@emea.europa.eu http://www.emea.europa.eu
?EMEA 2007 Reproduction and/or distribution of this document is authorised for non commercial purposes only provided the EMEA is acknowledged

GUIDELINE ON IMMUNOGENICITY ASSESSMENT OF BIOTECHNOLOGY-DERIVED THERAPEUTIC PROTEINS TABLE OF CONTENTS EXECUTIVE SUMMARY................................................................................................................... 3 1. 2. 3. 4. INTRODUCTION ......................................................................................................................... 3 SCOPE............................................................................................................................................ 4 LEGAL BASIS .............................................................................................................................. 4 MAIN GUIDELINE TEXT .......................................................................................................... 4 4.1 FACTORS THAT MAY INFLUENCE THE DEVELOPMENT OF AN IMMUNE RESPONSE AGAINST A THERAPEUTIC PROTEIN ............................................................................................................ 4 4.1.1 Patient- and disease-related factors................................................................................ 4 4.1.2. Product related risk factors of immunogenicity .............................................................. 6 4.2 NON-CLINICAL ASSESSMENT OF IMMUNOGENICITY AND ITS CONSEQUENCES ........................ 7 4.3 DEVELOPMENT OF ASSAYS FOR DETECTING AND MEASURING IMMUNE RESPONSES IN HUMANS................................................................................................................................... 7 4.3.1 Assay strategy.................................................................................................................. 7 4.3.2 Antibody assays ............................................................................................................... 7 4.3.3 Assay validation .............................................................................................................. 8 4.3.4 Characterisation of antibodies to a therapeutic protein ................................................. 9 4.4 POTENTIAL CLINICAL CONSEQUENCES OF IMMUNOGENICITY ............................................... 10 4.4.1 Consequences on Efficacy ............................................................................................. 10 4.4.2 Consequences on Safety ................................................................................................ 10 4.5 IMMUNOGENICITY AND CLINICAL DEVELOPMENT ............................................................... 11 4.5.1 Rationale for sampling schedule and kinetics of the antibody response ....................... 11 4.5.2 Consequences on pharmacokinetics of the product ...................................................... 12 4.5.3 Methodology aspects to assess comparability of immunogenicity potential as part of a comparability exercise .................................................................................................. 12 4.5.4 Immunogenicity in paediatric indications ..................................................................... 13 4.6 RISK MANAGEMENT PLAN ..................................................................................................... 13 REFERENCES .................................................................................................................................... 14 ANNEX 1 FURTHER DETAILS ON METHODS FOR ASSESSMENT AND CHARACTERISATION OF IMMUNOGENICITY....................................................................... 15 ANNEX 2 AN EXAMPLE OF A STRATEGY FOR ANTIBODY DETECTION AND CHARACTERISATION .................................................................................................................... 18
?EMEA 2007
Page 2/18

EXECUTIVE SUMMARY The number of biological/biotechnology-derived proteins used as therapeutic agents is steadily increasing. These products may induce an unwanted immune response in treated patients, which can be influenced by various factors, including patient-/disease-related factors and product-related factors. This document contains background information concerning the potential causes and impacts of immunogenicity and provides general recommendations for the performance of a systematic immunogenicity assessment from a marketing authorisation perspective. The predictive value of non-clinical studies for evaluation of immunogenicity of a biological medicinal product in humans is low due to inevitable immunogenicity of human proteins in animals. While non-clinical studies aimed at predicting immunogenicity in humans are normally not required, animal models may for example be of value in evaluating the consequences of an immune response. It is essential to adopt an appropriate strategy for the development of adequate screening and confirmatory assays to measure an immune response against a therapeutic protein. Assays may need to be capable of distinguishing neutralizing from non-neutralizing antibodies, and for use in pivotal clinical trials as well as in post-authorisation studies to be validated. In the clinical setting, careful planning of immunogenicity evaluation should include data systematically collected from a sufficient number of patients. For a given product, sampling should preferably be standardized across studies (e.g., sampling at baseline, under treatment and follow up samples). The sampling schedule for each product is determined on a case-by-case basis, taking into account also the risks associated with an unwanted immune response to patients. Data on the impact on efficacy and safety should be collected in order to fully understand the clinical consequences of the immune response. Immunogenicity issues should be further addressed in the Risk Management Plan. The scope of this guideline covers a wide applicability. Thus, the concepts might have to be adapted on a case-by-case basis to fit an individual development programme. Applicants should consider the possibility to seek Scientific Advice from EMEA or from National Competent Authorities. 1. INTRODUCTION
Most biological/biotechnology-derived proteins induce an unwanted immune response that is triggered by more than one single factor. This immunological response is complex and, in addition to antibody formation, other events such as T cell activation or innate immune response activation could contribute to potential adverse responses. The consequences of an immune reaction to a therapeutic protein range from transient appearance of antibodies without any clinical significance to severe life-threatening conditions. Potential clinical consequences of an unwanted immune response are a loss of efficacy of the therapeutic protein, serious general immune effects such as anaphylaxis, and, for therapeutic proteins used for substitution, a potential cross-reactivity with the endogenous counterpart in case it is still produced. Many factors may influence the immunogenicity of therapeutic proteins. They can be considered to be patient-, disease- or product-related. Patient-related factors that might predispose an individual to an immune response include: underlying disease, genetic background, immune status, including immunomodulating therapy, and dosing schedule. Product-related factors also influence the likelihood of an immune response, e.g. the manufacturing process, formulation, and stability characteristics. Although data on possible unwanted immune reactions to therapeutic proteins are required before marketing authorisation, problems may still be encountered in the post-authorisation period. In the marketing authorisation application, the applicant should include a summary of investigations of immunogenicity in the respective overview sections with full cross-reference to the data in the relevant modules. Depending on the immunogenic potential of the therapeutic protein and the rarity of the disease, the extent of immunogenicity data before approval might be limited. Further systematic immunogenicity testing might become necessary after marketing authorization, and may be included in the risk management plan.
?EMEA 2007
Page 3/18

2.
SCOPE
The general principles adopted and explained in this document mainly apply to the development of an unwanted immune response against a therapeutic protein in patients and how to systematically evaluate this. The guideline applies to proteins and polypeptides, their derivatives, and products of which they are components, e.g., conjugates. These proteins and polypeptides are mainly produced from recombinant or non-recombinant expression systems. Throughout this guideline, the term “therapeutic protein” is used. This guideline should be read in conjunction with other relevant guidelines, e.g.: ? ? Guidelines on similar biological (biosimilar) medicinal products; Guidelines on comparability of biotechnology-derived medicinal products after a change in the manufacturing process.
For coagulation factors, please, refer to the specific CHMP guidelines in this area (see references). 3. LEGAL BASIS
This guideline has to be read in conjunction with the introduction and general principles (4) and part III of the Annex I to Directive 2001/83 as amended. 4. MAIN GUIDELINE TEXT
The consequences of an immune reaction to a therapeutic protein range from transient appearance of antibodies without any clinical significance to severe life threatening conditions. As a rule, therapeutic proteins should be seen as individual products, and experience from related proteins can only be considered supportive. Also in this respect, concomitant medications and other patient-related factors like the underlying disease have to be taken into account, since these can also influence the clinical presentation of immunogenicity. Therefore, immunogenicity evaluation needs to be studied individually for each indication/patient population. Evaluation of immunogenicity should be a multidisciplinary task, encompassing joint efforts of quality, non-clinical and clinical experts. This document gives general recommendations and principles for developers and assessors of biotechnology-derived therapeutic proteins of how to approach immunogenicity evaluation from a marketing authorisation perspective. The scope of this guideline covers a wide applicability. Thus, the concepts might have to be adapted on a case-by-case basis to fit an individual development programme. For the justification of their approach to immunogenicity testing, Applicants should take into consideration both the risk for developing an unwanted immune response, and the potential clinical consequences as outlined below. The approach taken for the design of the immunogenicity development concept should be fully justified, e.g. when omitting assays or immune response measurements proposed in this guideline. Applicants should consider the possibility to seek Scientific Advice from EMEA or from National Competent Authorities. 4.1 Factors that may influence the development of an immune response against a therapeutic protein 4.1.1 Patient- and disease-related factors
Patient-related factors, which might influence the immune response to a therapeutic protein, may include genetic factors, age of the patient, disease-related factors including other treatments, and previous exposure to similar proteins. ? Genetic factors modulating the immune response
Genetic factors can alter the immune response to a therapeutic protein and lead to inter-patient variability. Allelic polymorphism in the major histocompatibility complex (MHC), impacting on affinity and stability of the interaction between MHC molecules and antigenic peptides, and genes
?EMEA 2007 Page 4/18

encoding the T cell receptor of helper T cells may influence immune responses and immunological tolerance induction. Immune responses may occur even if the amino acid sequence of the therapeutic protein is fully human. Other genetic factors influencing immunogenicity could be gene polymorphisms for cytokines that play a role in the fine-tuning of the immune response (e.g. interleukin-10, TGF-beta etc.). ? Genetic factors related to a gene defect
If the therapeutic protein is used for substitution of an endogenous protein, reduced levels or even the lack of this protein may influence immunological tolerance, since for these patients the physiological antigen may represent a neo-antigen. ? Age
The data from one age group cannot necessarily be projected to others since immune response against a therapeutic protein can be an age-related phenomenon. Children may possibly have a different immune response to these proteins. If the product is indicated in children, studies on immunogenicity should be carried out in this age group (see section 4.5.4). If indicated in elderly, consideration should be given to a potentially altered immune response. ? Disease-related factors
The patient’s underlying disease itself can be an important factor in the context of developing an unwanted immune response. Some patients with chronic infections may be more prone to an immune response, since their immune system is in an activated state. In other conditions (e.g. malnutrition, advanced metastatic disease, advanced HIV disease, organ failure), an immune response against a therapeutic protein might be less likely to occur due to an impaired immune system. For some products, it has been reported that the development of an antibody response can be different for different therapeutic indications or different stages of the disease. Therefore, immunogenicity normally needs to be studied separately for each disease or stage of the disease as part of the clinical studies. ? Concomitant treatment
Concomitant therapies may either decrease or increase the risk of an immune response to a therapeutic protein. Typically, the immune reaction against a therapeutic protein is reduced when immunosuppressive agents are used concomitantly. Consideration should also be given to previous treatments, that can modulate the immune reaction to a therapeutic protein and that have a long-term impact on the immune system. If clinical trials are performed in combination with immunosuppressants, a claim for use of the therapeutic protein in monotherapy must be accompanied by adequate clinical data on the immunogenicity profile in absence of immunosuppressants, i.e. immunogenicity data from the combination with immunosuppressants are not relevant for the monotherapy setting. ? Duration, route of administration, treatment modalities
Factors which may increase the immune response to a therapeutic protein may be the route of administration, dose, and the schedule of administration. Products given intravenously may be less immunogenic than those given subcutaneously or intramuscularly. Short-term treatment only is usually less likely to be associated with immune response than long-term treatment, and products given continuously are usually less immunogenic than those given intermittently.
?EMEA 2007 Page 5/18

Intermittent treatment or re-exposure after a long treatment free interval may be associated with an enhanced immune response. ? Previous exposure to similar or related proteins
Previous exposure to similar or related proteins can lead to pre-sensitisation and cause an immune response. For certain proteins being used for replacement therapy, previous therapies may have induced cross-reacting antibodies or immunological memory that affects subsequent therapies. 4.1.2. Product related risk factors of immunogenicity
Product-related factors influencing the immunogenicity of biological/biotechnology-derived therapeutic proteins include the origin and nature of the active substance (structural homology, posttranslational modifications), modification of the native protein (e.g. pegylation), product- and processrelated impurities (e.g. breakdown products, aggregates and host cell proteins, lipids or DNA), and formulation. ? Protein structure
Biotechnology-derived analogs to human endogenous proteins may trigger an immune response due to variations in the amino acid sequence or changes to the protein structure as a result of posttranslational modifications, physical, chemical or enzymatic degradation and/or modification e.g. deamidation, oxidation and sulfatation during all steps of the manufacturing process and during storage. Fusion proteins composed of a foreign and self-protein are of particular concern because of the potential of the foreign moiety to provoke an immune response to the self-protein (epitopespreading). Identification of the antigenic moiety of the fusion protein is advisable. Glycosylation is a frequent posttranslational modification of biotechnology-derived therapeutic proteins. These modifications may differ in the number and position of glycosylation sites as well as sequence, chain length and branching of the attached oligosaccharide. Therefore, when the same protein is manufactured under different conditions (e.g. change in cell culture process) there might be changes in the pattern of post-translational modifications and the immunogenic potential of the protein. This means also that antibodies induced by one product may react differently with the analogous product manufactured under modified conditions. This might have to be considered for evaluation of immunogenicity. ? Formulation
The composition of a formulation is chosen in order to best maintain the native conformation of therapeutic proteins. A successful, robust formulation depends on the understanding of the physical and chemical nature of the active substance and the excipients alone and their interaction. The formulation and the source of excipients may alter immunogenicty of therapeutic proteins and should be considered as a possible cause of such events. This should be considered when variations to the formulation are made. Impact of the primary packaging material and the conditions of clinical use e.g. dilution in infusion solutions and infusion devices of different materials could also influence the immunogenic potential of a therapeutic protein. ? Aggregation and Adduct Formation
Aggregation or adduct formation of proteins may either reveal new epitopes or lead to the formation of multivalent epitopes, which may stimulate the immune system. Factors which could be considered to contribute to aggregate or adduct formation include formulation, purification processes, viral inactivation procedures, and storage conditions of intermediates and finished product. The use of proteins, e.g. albumin, as excipient may lead to the formation of more immunogenic aggregates. It is important to monitor the aggregate and adduct content of a product throughout its shelf life. ? Impurities
There are a number of impurities of therapeutic proteins, which potentially can serve as adjuvants. Host cell proteins (HCPs) from the cell substrate co-purified with the active substance could induce
?EMEA 2007 Page 6/18

immune responses against themselves. But it is also possible that these HCPs, host cell-derived lipids or DNA function as adjuvants for the protein of interest. 4.2 Non-clinical assessment of immunogenicity and its consequences
Therapeutic proteins show species differences in most cases. Thus, human proteins will be recognised as foreign proteins by animals. For this reason, the predictivity of non-clinical studies for evaluation of immunogenicity is considered low. Non-clinical studies aiming at predicting immunogenicity in humans are normally not required. However, ongoing consideration should be given to the use of emerging technologies (novel in vivo, in vitro and in silico models), which might be used as tools. Measurement of antibodies in non-clinical studies are however requested as part of repeated dose toxicity studies, in order to aid in the interpretation of these studies (as discussed in “Note for guidance on preclinical safety evaluation of biotechnology-derived pharmaceuticals.” ICH S 6). Also, the comparison of the antibody response to the reference product in an animal model may be part of the comparability exercise both for similar biological medicinal products (see Guideline on Similar biological medicinal products containing biotechnology-derived proteins as active substance: Non-clinical and clinical issues CHMP/42831/05 and product-specific annexes) and for changes in manufacturing processes (see Guideline on comparability of biotechnology-derived medicinal products after a change in the manufacturing process – Non-clinical and clinical issues CHMP/BMWP/202695/06). An immune response to a therapeutic protein representing a counterpart to an endogenous protein may result in cross-reactivity, directed to the endogenous protein in cases where endogenous protein is still produced. Any relevant experience on the consequences of induction of an immune response to the endogenous protein or its absence/dysfunction in animal models should be discussed. Both humoral and cellular immune responses (where relevant) should be considered. In absence of such data, and if theoretical considerations are suggestive of a safety risk, animal immunisation studies with the therapeutic protein or the animal homolog may be considered in order to gain information on the potential consequences of an unwanted immune response. 4.3 Development of assays for detecting and measuring immune responses in humans.
Unwanted immunogenicity induced by biologicals can comprise humoral and cellular immune responses. It is therefore very important to select and/or develop assays and assay strategies for assessment of such immune responses. Most effort is usually focused on antibody detection and characterisation, as this is technically feasible and often related to clinical safety and efficacy. However, cell-mediated responses could play an important role and their assessment may be considered by applicants on a case by case basis. 4.3.1 Assay strategy
Adopting an appropriate strategy for assessment of unwanted immunogenicity of biological products is essential. This should usually include a screening assay for identification of antibody positive samples/patients, analytical immunochemical procedures for confirming the presence of antibodies and determining antibody specificity and functional assays for the assessment of the neutralizing capacity of antibodies. In addition, non-antibody assays e.g., assays for relevant biomarkers or pharmacokinetic measurements will be required which assess and characterize the clinical impact of antibodies (and possibly other components of immune responses) if these are detected/induced. It is important to include baseline data from all patients where appropriate. Annex 2 shows an example of a possible strategy for antibody detection and characterisation. 4.3.2 ? Antibody assays Screening assays
A screening assay should be capable of detecting antibodies induced against the biological product in all antibody positive samples/patients. This implies that detection of some false positive results is inevitable as absolute screening-assay specificity is normally unattainable and false negative results
?EMEA 2007 Page 7/18

must be avoided. The desirable characteristics of screening assays are sensitivity, specificity, precision, reproducibility and robustness. ? Assays for confirming the presence of antibodies
These assays are necessary for elimination of false positive samples/patients following the initial screen. Various approaches can be adopted for this purpose but it is necessary to select assays taking account of the limitations and characteristics of the screening assay(s). To confirm specificity, it is not normally sufficient or appropriate to simply repeat the screening assay in its original form. ? Assays for dissecting the specificity of antibodies
Assays which provide information concerning the specificity of the antibodies detected may be useful in some cases. This data contributes to confirmation of the specificity of the immune response. ? Neutralization assays
Assessing the neutralizing capacity of antibodies usually requires the use of bioassays. An assay must be selected or developed which responds well to the biological product. Bioassays used for measuring the potency of biological products e.g. for lot release purposes can often be adapted to assess neutralising antibodies. However, they frequently require refining if they are to perform optimally for measuring the neutralizing capacity of antibodies. If neutralising cell-based assays are not feasible/available, competitive ligand binding assays or other alternatives may be suitable. However, when these are used it must be demonstrated that they reflect neutralizing capacity/potential in an appropriate manner. 4.3.3 Assay validation
Assay validation is an ongoing process throughout product development. Assays used for the pivotal clinical trials need to be validated for their intended purpose. Validation studies must be conducted to establish that the assays show appropriately linear, concentration dependent responses to relevant analytes as well as appropriate accuracy, precision, sensitivity, specificity and robustness. For pivotal clinical trials, the use of a central laboratory to perform the assays may be helpful to avoid interlaboratory variability. In the post-approval setting, it is also important to consider inter-laboratory variability. Assays must also be validated to show that matrix effects caused by reagents or substances present in samples do not adversely affect the results obtained. This is normally addressed by ‘recovery’ investigations conducted by observing the effects of such substances present in the matrix on the response obtained in their absence. This needs to be investigated for the full range of dilutions of samples, which are to be used in assays, and, at least in some cases, limits the dilutions, which can be validly assessed. Residual biological product present in patients’ blood can complex with induced antibody and hence reduce the amount of antibody detectable by assays. This may affect assays differently, depending on the assay, assay format or type and the antibody characteristics. If this occurs, it may be circumvented/resolved by using a number of approaches e.g. by dissociating the immune-complexes with acid, removing excess biological by solid-phase adsorption, use of long incubation times and/or using an assay which allows sufficient sample dilution to avoid this problem. Such approaches must themselves be validated for effectiveness and adopted on a case-by-case basis according to needs. In some cases this problem can be overcome by appropriate spacing of the timing between administration of product and sampling for antibody assessment i.e. allowing time for the product to be cleared from the circulation before sampling. However this latter approach must not significantly compromise the detection of antibodies or the treatment of the patient. ? Standardisation and controls
Assays must be standardised and this requires the identification and/or development of appropriate reference materials, i.e. the use of relevant biological standards and/or well characterized positive and negative controls. These reagents function as critical assay reagents and are essential for assay
?EMEA 2007 Page 8/18

calibration and validation. This is especially important for assays used in unwanted immunogenicity investigations/studies, as it is intimately associated with assay interpretation and with distinguishing antibody positive from antibody negative samples. 4.3.4 Characterisation of antibodies to a therapeutic protein
If antibodies are detected in patients undergoing therapy, these need to be characterized to establish their clinical significance. This normally involves an immunological and/or biological assessment of antibody characteristics and investigation of effects of the antibodies (or other induced immune responses) on the product. Some of this can be addressed by non antibody assays as part of in vitro studies but it may also require clinical assessment of the patients receiving therapy. ? Antibody Characteristics
If antibodies are induced in patients, serum or plasma samples need to be characterised in terms of antibody content (concentration/titre), neutralizing capacity and possibly other criteria determined on a case-by-case basis according to the biological product, the type of patients treated, the aim of the study, clinical symptoms and possibly other factors. These may include antibody class and subclass (isotype), affinity, specificity. The degree of characterization required may differ depending on the study purpose and stage of development of the product. The assays used should be qualified for their intended purpose. Antibodies present in confirmed positive samples need to be examined for specificity for the active protein and, where applicable, distinguished from antibodies which bind to product-related and process-related components. It has been shown that antibodies can be induced against all and or any of these. It is also useful to screen for cross reactivity with other products based on the particular protein as well as (if possible and relevant) its endogenous counterpart. The neutralising capacity of antibodies present in positive samples needs to be established as this often correlates with diminished clinical responses to biological product. In some cases, screening neutralizing samples for cross-neutralization of other products based on the same protein and the endogenous protein is important as it may have implications for clinical efficacy and safety. It should be noted that neutralizing activity does not necessarily correlate with binding antibody content i.e. samples containing significant or high amounts of binding antibodies may fail to neutralize biological activity whereas samples containing lower amounts of binding antibodies can neutralize variable (sample dependant) amounts. This may depend on product, but must be determined empirically. ? Immunogenicity Assessment strategy –design and interpretation
Immunogenicity studies need to be carefully and prospectively designed to ensure all essential procedures are in place before commencement of clinical assessment. This includes the selection, assessment, and characterisation of assays, identification of appropriate sampling points, sample volumes and sample processing/storage and selection of statistical methods for analysis of data. This applies to assays used to measure and characterise antibodies and to methods employed for assessing clinical responses to antibodies if they are induced. Much of this needs to be established on a case-bycase basis, taking account of product, patients, and expected clinical parameters. Such studies can provide valuable information concerning significant immunogenicity of biological products, its characteristics and potential clinical consequences. They can be valuable for comparative immunogenicity studies for biosimilar products or following production/process changes introduced for established products. However, unwanted immunogenity can occur at a level, which will not be detected by such studies when conducted at a pre-approval stage, due to the restricted number of patients normally available for study. In view of this, it is often necessary to continue assessment of unwanted immunogenicity and its clinical significance post-approval, usually as part of pharmacovigilance surveillance. In some cases, post-approval clinical studies may be needed to establish the risk associated with an unwanted immune response. For further details on methods for assessment and characterisation of immunogenicity see Annex 1.
?EMEA 2007
Page 9/18

4.4 4.4.1
Potential clinical consequences of immunogenicity Consequences on Efficacy
Factors which influence whether antibodies to a therapeutic protein will induce clinical consequences include the epitope recognised, affinity, class of the antibody, the amount of antibodies generated, and the pharmacological properties of the biotechnological medicinal product. In addition, the ability of immune complexes to activate complement or be cleared may be a factor that impacts clinical outcome. Usually, antibodies recognising epitopes on the therapeutic protein not linked to activity are expected to be associated with less clinical consequences. However, as discussed below, such antibodies can influence pharmacokinetics and, as such, influence efficacy indirectly. “Neutralising” antibodies, interfering with biological activity by binding to or near the active site, or by induction of conformational changes, can induce loss of efficacy. Determination of neutralizing antibodies from confirmed positives, and the assays used, should be appropriate (see section 4.3). Most importantly, neutralizing antibody assays should be capable of detecting clinically relevant neutralizing antibodies. Correlation of antibody characteristics with clinical responses requires a comparison of data generated in assays assessing antibody responses (see above) with results generated using patients’ samples and assays designed to assess clinical responses. Most of the latter are product-specific, e.g. assessing expansion of leukocyte populations by colony-stimulating factors, or increased reticulocyte numbers by erythropoietin. Such assays need to be selected according to product and need. In many cases, it might be difficult to identify a clinical endpoint which is sensitive enough to establish the impact on clinical outcome directly, and adoption of a surrogate measure of response may be an option, e.g. biomarkers/pharmacodynamic markers. The choice of such markers should be justified. In vivo comparison of patient’s clinical responses to product before and following antibody induction can provide information on the correlation between antibody development (and antibody characteristics) and clinical responses. This can be done either by intra-group analysis (response in patients before and after occurrence of antibodies), or by comparison with patients within the study who do not show an immune response.
4.4.2
Consequences on Safety
Loss of efficacy and alteration of the safety profile are not necessarily linked. Safety issues, like infusion-related reactions, can occur even when there is no loss of efficacy. ? Acute consequences
Usually, patients who develop antibodies are more likely to show infusion-related reactions. Acute infusion reactions including anaphylactic reactions may develop during (within seconds) or within a few hours following infusion. Applicants should differentiate between the terms “infusion reaction” and “anaphylaxis” and carefully define which symptoms to label as “infusion-related reaction”. “Infusion reactions” usually represent symptoms occurring in a close timely relationship to an infusion and are not necessarily linked to anaphylaxis or even hypersensitivity. However, acute reactions can be true allergic, namely IgE-mediated type I reactions (anaphylactic reactions), including hypotension, bronchospasm, laryngeal or pharyngeal oedema, wheezing and/or urticaria. The term “anaphylaxis” should be restricted to such situations and represents a strict contraindication to further exposure to the drug. However, the majority of infusion reactions are characterized by more non-specific symptoms, for some products more frequently occurring on initial exposure and sometimes less frequent/severe reactions are observed on re-exposure. An infusion might not represent a contraindication to further exposure. A range of symptoms including headache, nausea, fever or chills, dizziness, flush, pruritus, and chest or back pain have been described in relation to infusions. It is acknowledged that the distinction between an infusion reaction and anaphylaxis can be challenging, but nevertheless such distinction in necessary due to the different clinical consequence. Applicants should not only focus on infusion reactions and anaphylactic symptoms since the consequence of immunogenicity is product-specific and can elicit unexpected clinical symptoms.
?EMEA 2007
Page 10/18

?
Non-acute consequences
Delayed hypersensitivity and immune complexes In addition to acute reactions, delayed type (T-cell mediated) hypersensitivity and immune complex mediated reactions have to be considered. The risk of these reactions may be higher with an increasing drug free interval. Such delayed hypersensitivity reactions should be clearly delineated from infusion reactions. Applicants should ensure the systematic collection of non-acute clinical sequelae following application of the therapeutic protein. Clinical signs can include myalgia, arthralgia with fever, skin rash, pruritus etc., but also other, less obvious clinical symptoms should be systematically collected. Besides consequences on pharmacological characteristics, immune-complexes can potentially be deposited in tissues. The underlying disease and the potential consequences of immune complexes on the further clinical course should be considered and critically evaluated, e.g. potential worsening of renal involvement in patients with underlying autoimmune disease. Cross-reactivity with an endogenous counterpart Antibodies developing against therapeutic proteins with endogenous counterparts can cross-react with this endogenous counterpart in cases where it is still produced (e.g., erythropoietins). In-depth characterization of the antibody response including cross-binding and close surveillance of the clinical consequences should be part of the pre-approval development programme. Experiences with similar products can be supportive, but are not sufficient per se. Applicants developing novel constructs like hybrid molecules fused to physiological functional molecules should carefully consider the potential consequences of cross-reactivity of antibodies against all endogenous (or self) components. 4.5 Immunogenicity and Clinical Development
4.5.1
Rationale for sampling schedule and kinetics of the antibody response
Immunogenicity assessment should be part of the clinical trials, since the correlation to clinical efficacy and safety is important. For a clinical trial, Applicants are encouraged to evaluate immunogenicity in all patients and not only in a symptom-driven manner (i.e. only for patients when a change in safety or efficacy profile is suspected). However, further to scheduled routine repetitive sampling, patients should also be evaluated in a symptom-driven manner with additional samples, when the occurrence of an unwanted immune response is suspected. Several factors such as dose, schedule and treatment modalities influence the development of an immune response against a therapeutic protein (see 4.1). Therefore, the sampling schedule for detection of an immune response should be adapted and selected individually for each product, taking into account also its pharmacokinetics. Baseline samples should always be collected. The overall incidence of immunogenicity should be evaluated for a given product in all indications, thus sampling schedules should preferably be comparable between different trials in order to enable for direct comparison of the incidence of anti-drug antibodies. Deviations from this concept should be justified. Applicants should endeavour to standardise sampling schedules, assays, definitions etc. taking into account also experiences with comparable products. During treatment samples should always be taken before administration of the product, since residual levels of the active substance in plasma can interfere with the assay (see section 4.3). Adequate conditions for storage and shipment of samples need to be established to ensure appropriate quality of the test material. The frequency of sampling and the timing and extent of analyses will also depend on the risk identified for a particular drug and the clinical consequences, and has to be justified. Sampling schedules should include repetitive sampling and be designed to clearly distinguish patients being transiently positive from patients developing a persistent antibody response. Both transient and persistent antibody responses should be combined to determine the overall immunogenicity of a product in a given condition. In particular, persistent antibodies are of high importance, since patients with persistent antibodies are more likely to experience clinical sequelae in terms of safety and efficacy, while a transient antibody response can resolve without further consequence.
?EMEA 2007 Page 11/18

For products intended for chronic use, it may be necessary to study the evolution and persistence of an observed immune response. Efforts should be engaged to collect data on potential changes in the character of the antibody response over time, e.g. change from non-neutralizing to neutralizing in a given patient, where applicable. On a case-by-case basis, e.g. when required according to a risk assessment, potential long-term consequences of an unwanted immune response should be considered when planning the clinical programme of immunogenicity evaluation. More frequent sampling will usually be employed in the earlier phase of treatment, where patients are normally most at risk of antibody development. Since longer-term treatment is more likely to result in an immune response, routine sampling later in the treatment course should be implemented in clinical trials. In case of continuous chronic treatment, usually immunogenicity data for one year of treatment should be available pre-authorisation. Deviations should be fully justified, e.g. shorter exposures or differences as regards the extent of data for different routes of administration. If used for different routes of administration, Applicants should justify their approach as regards immunogenicity assessment for each route at the time of Marketing Authorisation Application. Depending on the medicinal product and the potential risks associated with the occurrence of an unwanted immune response, it might become important to cover a sufficient number of exposures. If feasible, sampling should also be done after completion of the treatment regimen to determine persistence of response. While a decrease of anti-drug antibodies might occur over time in patients initially positive for such antibodies, also a rise in such antibodies might occur, e.g. if the therapeutic protein has immunosuppressive properties and by its mechanism of action suppresses an immune response against itself. Where feasible and possible, Applicants should provide guidance for the prescriber as part of the marketing authorisation application on how a patient with loss of efficacy should be handled over time, e.g. by an increase of dose or a reduced dosing interval or cessation of treatment. The results of the immunological studies should be included in the relevant sections of the SPC.
4.5.2
Consequences on pharmacokinetics of the product
Antibodies recognising epitopes on the therapeutic protein not linked to activity are associated with fewer clinical consequences. However, such antibodies can influence pharmacokinetics and, as such, influence efficacy. “Clearing” antibodies may be neutralizing or non-neutralizing, and reduce efficacy by removing the therapeutic protein from circulation. Non-neutralizing, “binding”, antibodies, may sometimes also increase, rather than decrease, the efficacy of a product by prolonging the half-life, or stimulating a pathway or mechanism.” Neutralizing antibodies may inactivate the drug with or without clearance. The loss of efficacy may be characterized through the Assay Strategy described in Section 4.3 as needed. A change in pharmacokinetics may be an early indication of antibody formation. If antibodies are detected during the clinical programme, their possible interference with the pharmacokinetics should be studied (see also Guideline on the Clinical Investigation of the Pharmacokinetics of Therapeutic Proteins).
4.5.3 Methodology aspects to assess comparability of immunogenicity potential as part of a comparability exercise
It has been reported that variations to the production process may induce alterations of the immunogenic properties of the product. When variations to the manufacturing process of a licensed product are made, the comparability exercise is a stepwise approach (see Guideline on comparability of biotechnology-derived medicinal products after a change in the manufacturing process). If the initial physicochemical and biological testing indicates a difference between the pre- and post-change product, the potential consequences to safety and efficacy need to be considered including altered immunogenicity. Even when initial physicochemical and biological testing do not indicate a difference, the potential for altered immunogenicity undetected by such tests needs to be considered. The extent of immunogenicity studies, if required, should be based on risk analysis, taking into consideration the nature of the observed difference, the potential clinical impact, and knowledge gained with this product and product class before. The determination of the appropriate target population will be selected to where best detect differences, not restricted to immunogenicity only. Applicants should make an effort to select a homogeneous and clinically relevant patient population
?EMEA 2007 Page 12/18

that allows for such comparisons. Due to expected differential susceptibility, immunogenicity data from healthy volunteers are not suitable substitutes. For most products, immunogenicity is studied in previously unexposed patients, and integrated in the clinical study to establish that the change in the manufacturing process has had no adverse impact on efficacy and safety. Immunogenicity evaluation as part of a clinical trial for a comparability exercise should preferably involve head-to-head studies of pre- and post-change product. The same assays should be used. Changes in immunogenicity as a result of a change in the manufacturing process might require a specific risk management strategy and an update of the risk management plan (see section 4.6). If there is a risk of rare immune-mediated adverse effects, this may be addressed after the implementation of the change in a post-marketing setting.
4.5.4
Immunogenicity in paediatric indications
Therapeutic proteins are increasingly used in children. It has to be considered that children may differ from adults in their immune response. When studying the product in a paediatric indication, posology and treatment schedules should be selected and justified accordingly. If applicable and feasible, results should be analysed by age groups, and immunogenicity data should be evaluated and presented separately for each age group to potentially identify vulnerable age groups. As regards substitution therapy, recombinant technology has allowed the development of proteins for use in genetic disorders where previous substitution treatment has not been available. Children are the most likely subjects exposed to these products and may be at high risk for antibody development. 4.6 Risk management Plan
Within the marketing authorisation application, the applicant should present a risk management plan in accordance with current EU legislation and pharmacovigilance guidelines including the CHMP Guideline on Risk Management Systems for Medicinal Products for Human Use (EMEA/CHMP/96268/2005). Immunogenicity should always be addressed in the Risk Management Plan (RMP), taking into account risks identified during product development, and potential risks and consequences of an unwanted immune response to patients. The risk specifications and minimization should follow the principles outlined in this guideline. Again, it should be emphasized that evaluation of immunogenicity is a multidisciplinary approach, at best providing input of quality, non-clinical and clinical experts. The extent of data on immunogenicity that can be obtained during the clinical development programme of a biotechnology-derived product before approval depends on the event rate, driven both by the immunogenic potential of the protein and the rarity of the disease. Therefore, further systematic immunogenicity testing might become necessary after marketing authorization, and may be included in the Risk Management Plan. The extent of immunogenicity data to be collected in the post-marketing setting will depend on various factors including: ? ? ? ? Disease-related factors like its prevalence, the vulnerability of the patients, availability of alternative therapies, duration of treatment, etc. Pre-authorization immunogenicity findings including impact on efficacy and safety Experience on immunogenicity with similar proteins or related members from that class of proteins, including proteins manufactured with similar production processes. Seriousness of the immune reaction.
However, biotechnology-derived proteins should be considered individually, and therefore the possibility for extrapolation from other related proteins is limited and needs to be fully justified. Immunogenicity can be further studied in a post-marketing setting e.g. by enhanced reporting of possibly immune-related adverse events (including loss of efficacy), or by pharmacoepidemiological studies.
?EMEA 2007
Page 13/18

Since systematic sampling might not be feasible in a post-marketing setting, it is important to conclude on potential unwanted immune responses based on suspicious safety and (loss of) efficacy signals. This requires that the evaluation of such events is defined prospectively in the RMP. The MAH should establish a standardized algorithm on how to further investigate those cases of suspected immune responses, including how to confirm the development of antibodies in a given case. The RMP should include: ? ? ? ? Risk Identification & Characterisation (e.g. case definitions, antibody assays); Risk Monitoring (e.g. specific framework to associate risk with product); Risk Minimization & Mitigation strategies (e.g. plans to restrict to intravenous use where necessary, actions proposed in response to detected risk etc.); Risk communication (e.g. minimization and mitigation messages for patients and physicians, communication to physicians of how to access specific investigation tools like antibody testing assays); Monitoring activities to ensure effectiveness of risk minimization.
?
Applicants should respond to evolving data on immunogenicity by taking adequate measures, e.g. changes in the Product Information, update of the RMP, and other risk minimization activities (e.g. educational programmes etc.). For planning immunogenicity assessment in the post marketing setting, the same recommendations apply as discussed in previous sections of this guideline. For changes in the manufacturing process, implications of this change on the immunogenic potential might have to be addressed in the RMP. REFERENCES ? ? ? ? ? ? ? Note for guidance on pharmaceutical development (ICH Q8 Step 4) Note for guidance on preclinical safety evaluation of biotechnology-derived pharmaceuticals (ICH S6) Similar biological medicinal products containing biotechnology-derived proteins as active substance: Non-clinical and clinical issues. (CHMP/42831/05) Guideline on comparability of biotechnology-derived medicinal products after a change in the manufacturing process. Non-clinical and clinical issues. (CHMP/BMWP/101695/2006) Guideline on the Clinical Investigation of the Pharmacokinetics of Therapeutic Proteins (CHMP/EWP/89249/2004). Clinical Investigation (CPMP/BPWG/198/95) of Human Plasma Derived Factor VIII and IX Products
Clinical Investigation of Recombinant Factor VIII and IX Products (CPMP/BPWG/1561/99).
?EMEA 2007
Page 14/18

ANNEX 1 Further details on methods for assessment and characterisation of immunogenicity Types of antibody assays ? Screening assays
The need to accommodate screening of relatively large numbers of samples necessitates use of an assay with high throughput and appropriate automation. Screening methods include immunoassays, radioimmunoprecipitation assays and surface plasmon resonance assays. All procedures detect antigen-antibody interaction (binding) but may differ in their underlying scientific/ technical principles. Immunoassays constitute a large group of assays and are based on a variety of formats and detection systems. These include direct binding assays, bridging assays, capture (sandwich) assays and competitive immunoassays using radioligand, enzymatic, fluorescent, chemiluminescent or electrochemical luminescence detection systems. ? Assays for confirming antibody positivity
Different assays can be used for this purpose and high sample throughput may be less important than for screening assays due to the smaller number of samples requiring analysis. To achieve confirmation of specificity, it is necessary to include an assay which evaluates specificity. For example, addition of an excess of antigen to the sample prior to evaluation in binding assays, which should result in a adsorption of antibody and therefore reduction of positive signal for true positive samples. Identification of immunoglobulin as the analyte in some assays e.g., by using specific antiimmunoglobulin reagents can also aid in identifying genuine antibody positive samples. In certain problematical cases, it may be useful to include an assay based on a different scientific/technical rationale than that used for the screening assay but the characteristics of the assays e.g., different sensitivities need to be considered. ? Assays for dissecting the specificity Analytical immunoassays such as immunoblotting and radioimmunoprecipitation analysis can be used to dissect the specificity of the detected antibodies. ? Neutralization assays Bioassays or other functional assays need to be selected using a product-based approach. Usually a single concentration of biological is chosen for the assay and dilutions of each sample assessed for their inhibitory effect on the assay response. This allows a neutralizing dose response to be determined and calculation of neutralizing capacity (‘titre’) for each sample. ? Assays for assessing cell-mediated immune responses
The strategy for assessing cell-mediated immune responses induced by biologicals is generally less clear than for humoral responses. Assays need to be developed or selected on a case-by-case basis if these are required. In most cases, development of a mature IgG response implies underlying antigen specific helper T-cell involvement. Examples of assays of use for detecting/assessing cell-mediated responses are T-cell stimulation/proliferation assays and cytokine (e.g. IL2, IL4, IFN-gamma) production/release methods. These involve the use of T-cell preparations sometimes co-cultured with preparations of other cell types, e.g. dendritic cells. Elispot and flow cytometry procedures are commonly used for these assays. Cell-mediated cytotoxicity assays may be useful in some cases. In some cases more detailed studies involving assessment of cell-mediated immune responses may be useful. Memory B-cell (and sometimes memory T-cell) assays can provide useful information regarding the nature of the immune response and may contribute to prediction of development of immunogenicity problems. Studies using peptides or full-length protein (depending on the assays and purpose of the assays) and Elispot methodologies can be used for these. In some cases more complex
?EMEA 2007 Page 15/18

investigations of cell-mediated immunity e.g. involving study of regulatory T-cells may be valuable. The need for such investigations must be decided on a case-by-case basis depending on the aims and purpose of the studies. Assay characteristics Assays need to be selected, optimized and analysed according to and taking account of their intended use. The importance and requirements of assay characteristics (see above under screening assays for a list of some of these) depends on the use of the assay. For example great sensitivity may not be required for an assay if this is not needed for detection of the amounts of antibodies, which are induced by a particular biological product in patients receiving therapy. Developing unnecessarily sensitive assays for such antibodies would be inappropriate especially if this sensitivity can only be achieved by sacrificing other desirable characteristics e.g. specificity, robustness. Adoption of the simplest assay suitable for all requirements is normally a valid approach to assay selection (particularly when high throughput is important e.g. for screening assays). However care with this is necessary to ensure that it does not compromise other stages of immunogenicity assessment. For example direct binding ELISAs, with antigen directly immobilized on plate well surfaces are often the simplest assay approach, but may be associated with a very high incidence of false positivity. They may also be associated with a high incidence of false negatives for samples containing low affinity antibodies and certain isotypes or subclasses. In such cases, it is often necessary to adopt a more suitable assay, e.g. ‘bridging’ assays, electrochemiluminescent or SPR methods to avoid this. False negative results in screening assays due to epitope masking can be encountered and a strategy to avoid these may be necessary e.g. by using assays that avoid specific masking of particular epitope(s). Standardisation, reference materials, well characterized controls and assay validation An antibody positive standard/reference material/control is clearly needed for all assays. This is used to demonstrate assay response and can be used for calibration purposes. If possible this should be a human preparation with a significant antibody content which is available in sufficient quantity for continued use. It should be stored appropriately (lyophilized or frozen at a suitable temperature) and well characterized. Reference preparations for neutralization bioassays should have significant neutralizing activity, but it is also useful to include a non-neutralizing antibody preparation in assays, at least in validation studies. However, in several cases, sufficient human serum may not be available to allow preparation of an appropriate reference preparation. In such cases, pooling of samples is usually the best approach and this may also avoid problems due to the specific characteristics of a single donor sample. In some cases human serum is unavailable in the quantities required either as a pool or even at all e.g. early in product development/trials and in such cases use of an animal serum as a reference is the only realistic option. However, this needs to be selected carefully and its use is more limited than for human reference preparations e.g. immunochemical procedures, which involve the use of an anti-human immunoglobulin reagent, will not reliably respond to non-human antibodies and the response in all assays may differ in characteristics from responses to human antibodies in human samples. Calibration of immunoassays is problematical as the immunoglobulin present in standards and samples is heterogeneous in structure, specificity and avidity. This makes direct valid comparison between samples and reference materials, especially on a mass basis difficult, if not impossible. This implies that calibration of such assays should be carried out using an acceptable, valid approach, which is clearly described. An option is to report immunoassay data as a titre based on a standard procedure for calculating this value. An alternative to this is to calculate the relative antibody concentration of samples and positive controls. Biological assays used to assess the neutralizing capacity of antibodies should be calibrated using International Standards/Reference Preparations where these are available. This allows expression of neutralizing activity in terms of meaningful units of biological activity of product/preparation and also provides information relevant to assay validation. If such standards are not available, appropriate inhouse preparations need to be established. In many cases it is useful to express the neutralizing capacity of samples in terms of the volume of sample required to neutralize a constant biological
?EMEA 2007 Page 16/18

activity of product e.g. ml of serum/defined unit of bioactivity of biological. In other cases, using the sample dilution or titre required to neutralize the biological activity of the product is also an option. It is also very useful to prepare a panel of reference materials containing different amounts of antibodies and antibodies with different characteristics, which can be used to characterize/validate assays and act as assay performance indicators. If possible this should include one or more preparations with low antibody content (close to the minimum detection limit) and containing low avidity antibodies. Negative standards/controls are needed to establish assay baselines and characterize/validate the assays. Assay baseline for normal (healthy) individuals is clearly fairly easily determined by measuring the assay response using samples derived from an appropriate number of such individuals and analysing this to provide a statistically valid background value. However, this may not represent the baseline response of the assay to samples derived from the patient population, which would therefore need to be established separately, using pre-treatment samples from patients, or from some other valid, relevant population. In any case, some individual’s/patient’s samples may contain preexisting (pre-treatment) antibodies or possibly other substances which produce significant positive responses in assays, and so screening patients for this is necessary to ensure that post-treatment data can be interpreted correctly. Reagents used in assays need to qualified and acceptance specifications set, at least for those, which are most important. Interpretation of Results It is essential to establish clear criteria for deciding how samples will be considered positive or negative, and also how positive results will be confirmed. Approaches to these can differ according to assay etc. and need to be decided accordingly. A common procedure for establishing positive cut-off for immunoassays is to establish assay background (see above). A statistical approach should preferably be used to establish the assay cut-off value. Alternatively, real data (e.g. double background value) can be used to determine what will be considered the lowest positive result.
?EMEA 2007
Page 17/18

ANNEX 2 An example of a strategy for antibody detection and characterisation
-ve denotes negative; +ve denotes positive
?EMEA 2007
Page 18/18

物流自动分拣输送系统

随着我国经济和社会的不断发展,电商、快递行业的发展突飞猛进,这对于输送和分拣作业提出了更高的要求,要高效地处理大批的物流量,离不开自动分拣输送系统。用自动化的快速分拣技术,取代大量的人工分拣,不仅降低了人力成本,同时还大幅提高了分拣作业的效率与准确率。下面六维智能物流就为大家详细的介绍下物流自动分拣输送系统的相关信息,希望对你有所帮助。 商品或货物的到达、卸货、分类、存储,再到出货、分类、发出等作业,势必要通过不同复杂程度的输送分拣设备,以实现在最短的时间内将这些商品卸下并按商品品种、货主、储位或发送地点进行快速准确的分类,将这些商品运送到指定地点(如指定的货架、加工区域、出货站台等)。 当供应商或货主通知物流中心按配送指示发货时,自动分拣系统须在最短的时间内从庞大的高层货架存储系统中准确找到要出库的商品所在位置,并按所需数量出库,并将从不同储位上取出的不同数量的商品按配送地点的不同运送到不同的理货区域或配送站台集中,以便转运或装车配送,充分实现快速输送分拣功能。 除了电商、快递市场的需求大增,输送分拣设备应用的主要领域还包括烟草、医药、流通、食品、汽车等各个行业。这些领域的输送分拣设备市场需求量仍然占据总需求的大部分比例。 自动分拣系统(Automatic sorting system)是先进配送中心所必要的设施条件之一。具有很高的分拣效率,通常每小时可分拣商品6000-12000箱,可以说,自动分拣机是提高物流配送效率的一项关健因素。

输送设备主要有: 皮带输送机、网带输送机、滚筒输送机、钢带输送机、链条输送机、链板输送机、伸缩式输送机、倍速链输送机。 分拣设备分类: 摆臂式、弹出轮式、滑靴推块式、翻盘式、交叉皮带式、推杆式、直角移载式、斜导轮式。

Sayatoo卡拉字幕精灵制作VEGAS和会声会影的卡拉OK字幕的方法

Sayatoo卡拉字幕精灵制作VEGAS和会声会影的卡拉OK字幕的方法 1.先用“Sayatoo卡拉字幕精灵”制作好卡拉OK字幕并保存为kaj文件。(制作方法见“Sayatoo卡拉字幕精灵”应用) 2.然后点击“Sayatoo卡拉字幕精灵”中的“工具”菜单→“生成虚拟字幕AVI视频”打开对话框,输入kaj文件路径,点击“开始生成”生成虚拟字幕AVI文件。 3.在“Sony Vegas”中打开前面生成的AVI文件,并将AVI文件拖入视频轨道并叠加于其他视频背景之上。 4.该AVI视频本身带有Alpha透明通道,需要在Sony Vegas打开该通道: (1)首先对在“项目媒体(Project Media)”中打开的AVI视频文件(或者对视频轨道上的AVI视频),右键菜单选择“属性(Properties)”项打开媒体属性对话框。 (2)然后在“媒体”属性对话框中将“透明通道(Alpha 通道)”设置为“直接(非蒙版的)”。 (3)然后再将相应的音乐文件拖入轨道并与虚拟AVI视频对齐。这样就可以直接输出高质量的音乐字幕视频。 用会声会影X2直接就可以用32位的AVI视频,不需要设置,背景是透明的。VEGAS导入32位的AVI视频则要进行如上设置,背景才是透明的。 在会声会影中应用必须注意以下二点,才能保证Kai文件能在会声会影中打开,否则会提示错误。 1.将“Sayatoo卡拉字幕精灵”安装程序中的“host”文件夹里面的“uvKAJ.vio”这个文件复制到会声 会影应用程序“vio文件夹”中。 2.在“会声会影”→“文件”→“参数设置”→“智能代理”里面的“启用智能代理”勾去掉。

自动分拣系统

自动分拣系统 自动分拣系统一般由自动控制和计算机管理系统,自动识别装置,分类机构,主输送装置,前处理设备及分拣道口组成。 1)自动控制和计算机管理系统是整个自动分拣的控制和指挥中心,分拣系统的各部个的一切动作均由控制系统决定。其作用是识别,接收和处理分拣信号,根据分拣信号指示分类机构按一定的规则(如品种,地点等)对产品进行自动分类,从而决定产品的流向。分拣信号的来源可通过条形码扫描,色码扫描,键盘输入,质量检测,语音识别,高度检测及形状识别等方式获取,经信息处理后,转换成相应的拣货单,入库单或电子拣货信号,自动分拣作业。 2)自动识别装置是物料能够实现自动分拣的基础系统。在物流配送中心,广泛采用的自动识别系统是条形码系统和无线射频系统。条码自动识别系统的光电扫描安装在分拣机的不同位置,当物料在扫描器可见范围时,自动读取物料上的条码信息,经过对码软件即可翻译成条码所表示的物料信息,同时感知物料在分拣机上位置信息,这些信息自动传输到后台计算机管理系统。 3)分类机构是指将自动识别后的物料引入到分拣机主输送线,然后通过分类机构把物料分流到指定的位置。分类机构是分拣系统的核心设备。 4)主输送装置的作用是将物料输送到相应的分拣道口,以便进行后续作业,主要由各类输送机械成,又称主输送线。 5)前处理设备是分拣系统向主输送装置输送分拣物料的进给台及其他辅助性的运输机和作业台等。进给台的功能有两个,一个是操作人员利用输入装置将各个分拣物料的目的地址送入分拣系统,作为物料的分拣作业指令;二是控制分拣物料进入主输送装置的时间和速度,保证分类机构能准确进行分拣。 6)分拣道口也称分流输送线,是将物料脱离主输送线使之进入相应集货区的通道。一般由钢带、传送带、滚筒等组成滑道,使物料从输送装置滑入缓冲工作台,然后进行入库上架作业或配货工作。

快递自动分拣运输系统

快递自动分拣运输系统 发表时间:2018-12-24T17:14:30.133Z 来源:《基层建设》2018年第32期作者:杨春旭姜峰姚华[导读] 摘要:析目前物流的分拣现状,特别是走在前沿科技的京东物流分拣系统中心、天狼SHUTTLE货架穿梭车、AGV及其现代化物流设备的发展及其研究,找出自动分拣系统的不足与其发展趋势,为我们下一步的研究工作打下基础。 哈尔滨远东理工学院黑龙江哈尔滨 150025 摘要:析目前物流的分拣现状,特别是走在前沿科技的京东物流分拣系统中心、天狼SHUTTLE货架穿梭车、AGV及其现代化物流设备的发展及其研究,找出自动分拣系统的不足与其发展趋势,为我们下一步的研究工作打下基础。 关键词:系统;研究现状;发展趋势 随着经济的快速发展与经济形势的快速转型,在互联网的快速发展下,从原有的实体经济运营模式转变成了“互联网+”的时代。在互联网催动下互联网购物模式兴起,同时也促进了物流业的发展。在互联网时代,加快了商品在全国甚至整个世界范围内的流动,大量的商品流动给物流业带来了巨大的财富,但与此同时也带来巨大的挑战。在原有的物流行业里,所有的货物分拣都靠人力去完成,不仅给工人带来巨大压力,同时速度与效率都很低下。因此,我们将建立一个自动分拣系统,采用中央控制器,全程无人化操作,避免了繁重的人工劳动,可全天候工作,且大大减短分拣时间,可以使快递物流的速度更加迅速。目前发展前景广阔,其研究现状如下: 1 国内物流分拣现状 物流的自动分拣系统最早可追述到二战之后,二战之后在美国、日本的物流中心就已经采用了自动分拣系统。近年来,在“互联网+”商业模式下,促进了物流业的快速兴起,在国内具有代表性的物流企业有中通、申通、圆通顺丰、韵达,还有我们众所周知的邮政,但在分拣最具有代表性的并不是他们,而是京东快递。 目前我国的自动分拣系统呈现出运用不广泛、设备不全面,大多数还处于半自动化模式,极少部分实现自动化及智能化控制自动分拣。在我国的物流企业中,自动分拣系统只在比较发达且设备及技术比较完善的发达地区,而其他技术及设备落后地区,还处于半自动化分拣模式。半自动化及自动化模式,分拣过程都将需要大量的人力来完成,分拣过程还需要靠人为的装卸和搬运,由于我国的市场规模大,很难在短时间内完成分拣模式的转变,主原因在于完成智能化需要投入大量的资金,关键在于设备和技术还处于试运行阶段,并没有达到可以广泛普遍流传阶段。因此,我国物流行业的分拣大多数还处于以往的人工分拣模式,半自动化及智能化控制还处于试运行,尚未普遍使用。 2 自动分拣系统阐述 2.1 物流的分拣系统发展历程简述 最开始物流分拣是通过人力来完成的,通过人工将其货物分类、搬运货物来实现货物的提取。在传统的人工分拣货物模式中,对于文件的制作、查找、人工搬运等等,都是一个非常耗时间、耗人力的过程、,而且速度低、作业效率低、准确性差。在当今“互联网+”的商业模式下,是不能瞒住人们的需求。 随着科技的发展,分拣系统开始运用各种机械化设备,加快了物流运输速度,同时作业效率也有所提高。近年来,随着软件的开发及互联网的发展,在互联网模式下的经济发展更是突飞猛进,进一步的推动了物流行业的发展,同时也带来巨大的挑战。在随着时代发展脚步,物流的分拣模式同样在革新,从人工分拣模式到半自动化模式,如今在最前沿的科技下,我们已经开始向智能化方向行进。我国具有代表性的就是京东物流分拣系统中心及其所研发的天狼SHUTTLE货架穿梭车,实现了智能控制。计算机空控制,、信息技术及其自动化机械设备是自动分拣系统不可缺少缺少的核心组成部分。 2.2 自动分拣中心系统 物流的自动分拣中心将包括物流机械化系统、信息系统及智能控制系统。物流机械化系统是物流设备之间的有效组合和配合,实现货物的运输到所指定的位置。信息系统将所分拣的货物信息进行采集、储存及处理等。智能控制系统是根据所设定的指令,将有效的控制机械化系统每台设备的运作及其监测。 3 自动分拣展趋势 物流的自动分拣系统最显著的优点,大批量、连续的分拣货物,大大减少了人力的参与,整个过程能实现无人化的分拣。全程的实现了智能控制、自动化作业,自动分拣系统并且不受时间、气候及其人力的限制,与人工分拣相比,自动分拣系统有了新的飞越性突破,提高的分拣效率,大幅度减少的人为劳动力。而且自动分拣系统的分拣误差为零,其原因在于分拣系统是时采用条形码扫描输入方式,除非条形码的印刷出错,否者是不会出现错误的。如果遇到损坏的条形码,是扫描不出来的,同时扫描器会自动发出警报,将信息报告给部门主管,将其交予人工处理,由此一来,分拣的错误率将降至零。 经济市场竞争日益加剧,货物的流通数量和频率都在逐渐增长,物流企业也在新起,竞争也在不断增加,要想在物流业占据一席之地,那么必须要提高自身的运输速度、缩短运输时间、降低运输成本。然而现在大多数的物流分拣还是传统的人工分拣模式,不仅作业效率低下,而且还存在分拣错误。然而,走在科技前沿,同时也是物流分拣革新的先行者,他们已经开始研发自动分拣系统,例如京东的物流分拣系统中心及天狼SHUTTLE货架穿梭车、太原刚玉仓储设备公司和贵阳普天通信机械厂也已经设计出设计生产货架电子标签拣选系统,小车式数字显示拣选系统等,还有珠海普天慧科信息技术有限公司研发了PTL电子标签拣选系统等。 自动分拣系统,可以实现分拣过程全程无人化操作,实现智能化控制,减少员工的劳动力,改变了传统人工分拣模式,同时大幅度的提高了分拣效率,同时也提高了经济效益。在未来的物流产业中,自动分拣将趋于普遍化、全球化,否则将会被从物流业中被淘汰出局。物流自动分拣系统是现在物流分拣革新的目标,也是“互联网+”商业模式下的需求,在未来将会被广泛的使用,有非常好的发展前景。 参考文献 [1]刘磊. 配送中心设施布局规划与分拣系统仿真研究[c]. 中南大学,2008. [2]杨玮,曹巨江. EIQ分析法在物流配送中心拣货系统设计中的应用[J]. 机械设计与制造,2006,9:160-161. [3]李暄,洪怡恬,郑慧,陈小静. Flexsim系统仿真软件在配送中心分拣系统设计中的应用[J]. 物流工程与管理,2009,31(1):37-39. [4]文静. 配送中心设施布局规划与分拣系统仿真研究[J]. 企业导报,2012,24:276-277. [5]《分拣系统应用现状及趋势https://www.wendangku.net/doc/fd18652623.html,/view/fa5e91da5022aaea998f0ff1.html

快递自动化分拣系统的开发

【最新资料,Word版,可自由编辑!】

目录 1研究背景 (3) 1.1研究背景 (3) 1.2研究意义 (3) 2文献综述 (4) 2.1快递自动化分拣系统国内外的发展状况 (4) 2.2快递自动化分拣系统的发展方向 (5) 3研究目标 (5) 4研究内容 (5) 5技术路线 (6) 5.1系统总体解决方案 (6) 5.2硬件系统设计 (7) 6预期的研究成果及课题的创新点 (7) 6.1预期的研究成果 (7) 6.2课题的创新点 (7) 7进度安排 (7) 8参考文献 ............................... 错误!未定义书签。

快递自动化分拣系统的开发 摘要:随着多品种少批量社会需求模式的推进,商品的流通速度迅猛提高,流通方式却是越来越复杂。因此,在人工成本不断高涨的环境下,需要引入适当的机械化分拣系统,以减少作业错误的发生,提高商品流通作业率,最终实现JIT(JUST IN TIME)配送。 本文将快速图像识别技术和快递自动化分拣装置相结合,研究基于自动化分拣系统的开发,介绍了一种应用DSP 技术、视频捕获技术,结合FPGA 逻辑控制、USB 通讯等多项技术,能够准确、快速识别物流系统商品大小、位置等信息的检测装置,可以有效解决自动分拣系统对复杂商品识别的难题。同时设计出相应的分拣装置,从而快速准确完成物流行业自动化分拣。 关键词:机械化分拣,图像识别,分拣装置 1研究背景 1.1研究背景 物流就是将物品从供应地往接收地传送的流动过程。根据现实需要,将储存、运输、搬运、装卸、流通加工、包装、信息处理、配送等基本功能的实施有机结合起来。它根据物品的种类、质量和数量,在最适当的时刻,以最低成本将其送到正确地点,并且准时完成物品信息的修改和传输及回收输送工具的全过程。 随着消费与生产在空间和时间上的分离不断扩大,为了保障企业生产和社会经济活动顺利进行,并且能取得好的经济效益;物流作为“需”、“供”之间桥梁,已发展成为了一门综合性的学科。物流学理论基础是多学科综合,并融合了社会科学与自然科学,它既是经济科学又是技术科学;同时,它还与其它学科有密切的联系,如自动控制、计算机科学等。物流学研究对象一般是复杂的、多目标决策的动态系统,在系统分析时,既要考虑技术的科学性、先进性,又要考虑它的 经济指标。 1.2研究意义 目前,现代物流作为一种比较先进的管理理念和组织方式,被认为是提高劳动生产率,企业降低物耗以外的第三利润的源泉。由于超大规模集中配送的效率难题和成本优势,提高配送中心的运营效率这已成了大中型企业改善现代物流的重要举措。作为配送中心的主要作业环节和核心设备,分拣系统的技术与分拣作业的效率也越来越受到工程界和的理论界关注。 我国现代物流发展比较晚,对自动分拣系统的研究也刚起步,分拣作业大部分都是采用的人工分拣,少数采用了机械分拣和人工分拣相结合的半自动分拣方式,而自动分拣所占比例非常低,其中一个非常重要原因是目前我国自动分拣系统技术还不够成熟,跟国外相比差距甚远。但自动分拣系统技术作为分拣领域中

premiere,cs6,字幕模板

竭诚为您提供优质文档/双击可除premiere,cs6,字幕模板 篇一:premierecs6调用卡拉字幕精灵虚拟avi文件 premierecs5.5调用sayatoo1.53卡拉ok字幕虚拟avi 的测试[ 复制链接] wf108 16 212 18 好0 主题友 积分 Vip会员 注册时间 20xx- 11-30 积分

6280 帖子电梯直达1#发表于20xx-11-511:32|只看该作者| 倒序浏览本帖最后由wf108于20xx-11-511:42编辑premierecs5.5调用sayatoo1.53卡拉ok字幕虚拟avi的测试wf10820xx/11/5sayatoo1.53卡拉ok字幕文件在64位windows7系统中不能使用,它生成的32位虚拟aVi文件也 无法导入64位的premierecs5.5使用。根据网上经验,我 用premierecs5.5调用sayatoo1.53卡拉ok字幕虚拟avi,测试成功。一、安装顽固不化版的sayatoo1.53傻丫头卡拉ok字幕精灵sayatooinstall1、右键选择“以管理员模式”运行sayatooinstall,自动注册,安装成功。2、右键点击 桌面上的sayatoo卡拉字幕精灵的属性,勾选“按 xpsp3模式兼容” 2739 发消息 图1 3、启动sayatoo卡拉字幕精灵,导入项目文件,一切 正常,制作虚拟aVi文件以备用。 图2 二、安装proxycodec64编码器代理 proxycodec64就是使64位程序可以调用32位的编码器,sayatoo的虚拟aVi是32位的,需要调用32位的编码

自动分拣系统仿真

实验2 自动分拣系统仿真 1.实验目的 通过建立一个传送带系统,学习Flexsim提供的运动系统的定义;学习Flexism提供的conveyor系统的建模,进一步学习模型调整和系统优化。 2.实验内容 (1)系统描述和系统参数 分拣系统的流程描述和系统参数如下。 ①四种货物A,B,C,D各自独立到达高层的传送带入口端: A的到达频率服从正太分布函数normal(400,50)s。 B的到达频率服从正态分布函数normal(200,40)s。 C的到达频率服从正态分布函数normal(500,100)s。 D的到达频率服从正太分布函数normal(500,100)s。 D的到达频率服从均匀分布函数uniform(150,30)s。 ②四种不同的货物烟一条传送带,根据品种的不同由分拣装置将其推入到四个不同的分拣道口,经各自的分拣道到达操作台。 ③每个检验包装操作台需操作工一名,货物经检验合格后打包,被取走。 ④没检验一件货物占用时间为uniform(60,20)s。 ⑤每种货物都可能有不合格产品。检验合格的产品放入箱笼;不合格的通过地面传送带送往检修处修复;A的合格率为95%,B为96%,C的合格率为97%,D的合格率为98%。 传送带的传送速度采用默认速度。 (2)实验要求 对上述传送分拣系统进行建模,仿真系统一天8h的运行状况,并完成思考。 3.实验步骤 (1)构建模型布局。 打开Flexism3.0,新建一个模型。从对象库中拖放所需的对象到建模视图中并根据实验内容的描述修改各实体的名字,如图1。 图1

(2)定义工件流程。 按住A键,同时用鼠标左键点击SourceA对象并且按住鼠标左键不放,然后拖动鼠标至Queue1对象。此时会出现一条黄线连接SourceA 和Queue1对象。然后松开鼠标左键,黄线将变成一条黑线,表示SourceA对象和Queue1对象的端口已经连接上。如上所述将SourceB,SourceC,SourceD和Queue1相连接;Queue1和Conveyor1相连;Conveyor1和ConveyorA 相连;Conveyor1和ConveyorB相连;Conveyor1和ConveyorC相连;Conveyor1和ConveyorD相连;ConveyorA和ProcessorA相连;ConveyorB和ProcessorB相连;ConveyorC 和ProcessorC相连;ConveyorD和ProcessorD相连;ProcessorA和Conveyor2、QueueA 相连;ProcessorB和Conveyor2、QueueB相连;ProcessorC和Conveyor2、QueueC相连,ProcessorD和Conveyor2、QueueD相连;Conveyor2和Sink相连。 按住S键将ProcessorA和OperatorA相连;同理将ProcessorB和OperatorB相连;ProcessorC和OperatorC相连;ProcessorD和OperatorD相连。 连接后的模型如图2所示。 图2 (3)定义对象参数 双击SourceA对象,打开其参数对话框,在“发生器的界面”将物品类型选取默认设置“1”;修改产品流出间隔时间,从“到达时间间隔”下拉框中选择使用正态分布,如图3;并修改选项的默认参数点击Template按钮修改其中的棕褐色的参数值:将“10”改为“400”,“2”改为“50”如图示4。点击“发生触发器”,在“离开触发”下拉菜单中选择颜色设置使用默认设置将物品设置为红色,如图5;点击Source参数框中的“使用”,“确定”。

自动分拣系统开题报告解析

自动分捡系统设计开题报告 一、课题介绍 1. 课题名称:自动分捡系统设计 2. 课题背景: 自动分拣系统(Automated Sorting System)是二次大战后率先在美国、日本的物流中心中广泛采用的一种自动化作业系统,该系统目前已经成为发达国家大中型物流中心不可缺少的一部分。该系统的作业过程可以简单描述如下:流动中心每天接收成百上千家供应商或货主通过各种运输工具送来的成千上万种商品,在最短的时间内将这些商品卸下并按商品品种、货主、储位或发送地点进行快速准确的分类,将这些商品运送到指定地点(如指定的货架、加工区域、出货站台等),同时,当供应商或货主通知物流中心按配送指示发货时,自动分拣系统在最短时间内从庞大的高层货架存储系统中准确找到要出库的商品所在位置,并按所需数量出库,将从不同储位上取出的不同数量的商品按配送进点的不同运送到不同的理货区域或配送站台集中,以便装车配送。 3.国内外发展现状: 纵观国内外物料自动分拣系统的应用情况可以发现,国外发达国家的物料自动分拣系统倾向于采用自动化程度很高的物料自动分拣系统。而在我国,由于起步晚,物料自动分拣系统中人工作业的比例也较高。国外物料自动分拣系统的广泛使用,以美国、日本及欧洲为代表的发达国家,在物料自动分拣系统的应用上呈现出自动化程度越来越高的特点。物料自动分拣系统已成为发达国家工业自动化不可缺少的一部分。主要应用在大中型物流中心、配送中心、流通中心、邮局分拣信件等等随着交流自动控制技术的发展,特别是电子技术的迅速发展,计算机的广泛应用。自动分拣技术在20世纪70年代被引入国内,我国的邮政系统最早并已多年使用自动分拣设备,并在长期的实践中不断创新、不断进步。例如,邮电部相关部门相继开发和研制出具国际水平的CORE-NT物料自动分拣系统,性价比很高的扁平邮件高速物料自动分拣系统;上海邮政通用技术设备公司研制成功了速递邮件网络化物料自

Premiere cs6调用卡拉字幕精灵虚拟avi文件-推荐下载

Premiere cs5.5调用Sayatoo1.53卡拉OK 字幕虚拟avi 的测试 2739 发消息 2011-11-5 11:32 |只看该作者 |倒序浏览 wf108 于 2011-11-5 11:42 编辑 调用Sayatoo1.53卡拉OK 字幕虚拟avi 的测试卡拉OK 字幕文件在64位 Windows 7系统中不能使32位虚拟AVI 文件也无法导入64位的Premiere Premiere cs5.5调用卡拉OK 字幕虚拟avi ,测试成功。 Sayatoo 1.53傻丫头卡拉OK 字幕精灵“以管理员模式”运行sayatooINSTALL ,自动注册,Sayatoo 卡拉字幕精灵的属性,勾选“按”

图1 3、启动Sayatoo 卡拉字幕精灵,导入项目文件,一切正常,制作虚拟AVI 文件以备用。

图2 二、安装Proxy Codec 64编码器代理 Proxy Codec 64就是使64位程序可以调用32位的编码器,Sayatoo 的虚拟AVI 是32位的,需要调用32位的编码器。Proxy Codec 64安装简单,我把它装在D 盘。三、Proxy Codec 64编码器代理选项中设置 1、安装后,会弹出下图,或在程序列表中找到 Proxy Codec 的 Config 选项,点击会弹出下图。

图3 2、如图4,点击下拉框,选择Sayatoo 的虚拟AVI 解码器“KAVC-Kara Title Avi” 。

图4 3、在图5中选择“KAVC-Kara Title Avi”后,在Proxy Codec0的选项上打勾。点击 “Install32Codec” 菜单,弾出图6 。

自动分拣系统毕业设计教材

重庆工业职业技术学院 毕业设计(论文) 课题名称:自动生产线分拣系统设计 专业班级:10 电气301 学生姓名:廖国强 指导教师:王俊洲 二O一三年月 摘要 PLC控制是目前工业上最常用的自动化控制方法,由于其控制方便,能够承受

恶劣的环境,因此,在工业上优于单片机的控制。PLC将传统的继电器控制技术、计算机技术和通信技术融为一体,专门为工业控制而设计,具有功能强、通用灵活、可靠性高、环境适应性强、编程简单、使用方便以及体积小、重量轻、功耗低等一系列优点,因此在工业上的应用越来越广泛。 本文主要讲述PLC在材料分拣系统中的应用,利用可编程控制器( PLC) ,设计成本低、效率高的材料自动分拣装置。以PLC 为主控制器,结合气动装置、传感技术、位置控制等技术,现场控制产品的自动分拣。系统具有自动化程度高、运行稳定、精度高、易控制的特点,可根据不同对象,稍加修改本系统即可实现要求。 关键词:可编程控制器,分拣装置,控制系统,传感器 ABSTRACT PLC control is the most commonly used industrial automation control method, because of its convenient control to withstand an adverse environment, it is better than MCU control in the industrial. PLC traditional relay control technology, computer and communication technologies are integrated specifically for industrial control and design, have strong function, common flexible, high reliability and environmental adaptability, and programming simple, easy to use and small size, light weight, a series of low-power advantages in industrial applications become more extensive. This paper focuses on the PLC in the canned beverage production, The design of an automatic sorting device with low cost and high efficiency is presented in the paper, which regards programmable logic controller ( PLC) as the master controller and combines pneumatic device, sensing technology, position control and other technology to implement automatic selecting of the products live. The device is characteristic of high automation, steady running, high precision and easy control, which can fulfill the requirement according to different situations with little modifications. Key words:programmable logic controller,sorting device,control system,sensors

分拣系统及分拣机的设计和应用综述

分拣系统及分拣机的设计和应用综述摘要:随着《2016-2020年全国电子商务物流规划》的颁布,到2020年基本形成布局完善、结构优化、功能强大、运作高效和服务优质的电商物流体系,信息化、标准化、集约化发展 取得重大进展,先进物流装备、技术在行业得到广泛应用。其中,提升物流设施设备智能化 水平是重要任务,随着近年来人工成本的快速上升,对智能分拣系统等信息化产品需求增 加,本论文对分拣系统和分拣机的研究进行简单的总结和归类,以便未来更好的研究分拣技 术。 1 分拣系统的设计 随着人们对网络购物越来越青睐,电子商务已经进入了一个高速发展的时期,为了配合市场发展的趁势,国很多企业都在积极建设自有的电商物流系统,比如知名的电商企业阿里巴巴和京东。对于物流系统而言,最具核心价值的环节就是它的配送中心。配送中心承担着衔接上游供应商与下游客户,实现高效地物品流通服务的重要责任,配送中心布局如果直接应用于电商的配送中心,可能会不太适用。配送中心的仓储和分拣都要占据大量的空间,包括商品存储的货架,分拣缓存区和补货区等,所以,合理设计和布局分拣系统是至关重要。订单量巨大时,为保证客户满意度,还要尽快完成订单的分拣配送,所以对配送中心的效率要求也会很高。 Gray等[1]综合了成本与拣选效率方面的需求,提出一种多级分层模型,求解拣选系统中涉及到的拣货区数量、品项分配及订单分批等配置优化问题。 Le Du等[2]建立了一种混合整数规划模型,对分区拣选策略下的人工拣选系统涉及到的品项分配及拣选顺序问题进行求解。该模型以订单处理总时

间最短为优化目标。与此同时,文章还分析了拣货区数量对订单处理总时间的影响,若减少拣货区的数量,会增加系统中的拣货人员行走距离,但同时也能够减少子订单的合流时间,考虑到这种互相影响互相制约的情形,指出了寻求拣货区数量平衡点的方式。 Guenov等[3]在分区拣选的策略下,研究了AS/RS系统的拣货区形状。以AS/RS系统可以划分为三个拣货区为前提,按照拣选量将所有品项分为A,B,C三类。文章分别给出了三种拣货区的外形配置,并设计了分段启发式算法求解每种外形配置下的订单处理总时间。最后通过仿真分析了每种配置下拣货区的特点。 Bartholdi等[4-7]在分区拣选策略下,针对人工拣选系统提出了救火队模型,克服拣货区固定划分后存在的问题。该模型中各拣货人员可以根据与相邻拣货员的相遇情况动态确定拣货区的边界。救火队模型具有极强的自组织性,各拣货人员能依据自身的工作效率,动态确定工作围,使工作时间快速达到平衡,有效消除整个作业过程中可能出现的效率瓶颈。 诗珍等[8]提出一种订单分批的拣选模型,该模型以最小化行走距离为目标,与此同时,文章基于启发式的聚类算法建立了订单分批问题的聚类模型,并设计了基于包络解码的混合遗传算法对模型进行求解。 Parikh等[9]研究了如何选择拣选策略,其中主要针对订单分批拣选策略与分区拣选策略进行了讨论。选择的过程综合地考虑了拣选频率、拣选人员可能出现的交叉、拣选人员工作量均衡及订单分流需求等因素,基于此建立了以最小化拣选成本为目标的选择模型,同时基于模型分析了拣选效率、订单数量、订单结构等因素会对拣选策略的选择带来何种影响。 王艳艳等[10]研究了人工拣选系统的订单分批拣选策略方面的问题,建立了以最小化拣选成本为目标的模型,该模型的建立综合考虑了包括拣选路

物流自动分拣系统设计

物流自动分拣系统设计 摘要:物流自动分拣系统是先进配送中心所必需的设施条件之一,自动分拣装置是提高物流配送效率的一项关健因素。只有在自动分拣系统中合理地选用分拣装置才能保证整个系统的安全高效运行。本文首先对自动分拣系统做了简单地介绍,然后提出了系统设计的一般方法,最后通过实例对自动分拣系统进行了分析和探讨。 关键字:自动分拣系统;分拣装置;设计方法;自动分拣系统设计 1.自动分拣系统概述 自动分拣系统是二次大战后在美国、日本等发达国家的物流中心、配送中心或流通中心所必需的设施条件之一。该系统的作业过程可以简单描述如下:物流中心每天接收成百上千家供应商或货主通过各种运输工具送来的成千上万种商品,在最短的时间内将这些商品卸下并按商品品种、货主、储位或发送地点等参数进行快速准确的分类,并将这些商品运送到指定地点(如指定的货架、加工区域、出货站台等);同时,当供应商或货主通知物流中心按配送指示发货时,自动分拣系统在最短的时间内从宠大的高层货架存储系统或其他指定地点中准确找到要出库的不同数量的商品按配送地点的不同运送到不同的理货区域或配送站台集中,以便装车配送。 2.自动分拣系统组成及特点 2.1自动分拣系统结构组成 如图1所示,自动分拣系统一般由上件装置、输送装置、分拣格口、控制系统组成。 1-输送装置2-上件装置3-控制系统4-分拣道口5-分类装置 图1 自动分拣系统结构组成 上件装置的作用是识别、接收和处理分拣信号,根据分拣信号的要求去指示分类装置按商品品种、商品送达地点或货主的类别等方式对商品进行自动分类。这些分拣需求可以通过不同方式,如可以通过人工输入、条形码扫描、色码扫描、键盘输入、重量检测、语音识别、高度检测及形状识别等方式,输入到分拣控制系统中去,根据这些分拣信号判断,来决定某一种商品该进入哪一个分拣格口。 分类装置的作用是根据上件装置发出的分拣指令,当具有相同分拣信号的商品经过装置时,该装车动作,使改变在输送装置上的运行方向进入其它输送机或进入分拣格口或其他接口设备。分类装置的种类很多,一般有推出式、浮出式、倾斜式、输送式和分支式几种,不同的装置对分拣货物的包装材料、包装重量、包装物底面的平滑程度等有不完全相同的要求。

edius教程

大家都知道用Flash可以制作出放大镜的效果,那么在Edius中是否也可以制作放大镜的效果呢?当然了可以了,在Edius中我们甚至运用和Flash中相同的原理来制作放大镜,即使用两个同样的素材,将其中一个放大比例后作为遮罩,放在另一层素材的上面。下面就让我们看看制作的过程,先看一下效果(本示例中素材图像来源于网络,仅供学习之用,若您认为侵犯您的权利请联系我们)。 1、首先导入两个完全相同的素材,分别放置在视频1轨和视频2轨道上。如图1 2、放大视频2轨道上的素材:在视频2轨道上,鼠标右键单击选择“布局”,打开布局设置窗口,剪裁源素

材中不需要放大的部分,将图像拉伸设置为全屏扩展,将填充颜色设置为黑色100%。3 3、为轨道2素材添加区域滤镜。如图3 4、设置区域内部滤镜参数:构选椭圆,柔边,(注意这里构选椭圆后在区域窗口中并不显示椭圆形状,但

是在监视窗口中会实时显示形状)。添加内部滤镜为组合滤镜,单击设置打开组合滤镜设置对话框。如图4 分别添加锐化和焦点柔化滤镜,锐化可以采用默认值,焦点柔化将其模糊值降低其他参数可取默认值。如图5 图片:6.jpg

问:Edius中的波纹模式和同步模式有什么区别? 答:波纹模式和同步模式的区别如下。 波纹模式:删除或剪辑片段后,后续片段前移,时间线上不留下空隙。编辑范围仅限于正在编辑的轨道之内。即使在非波纹模式下,编辑也会影响某些操作,例如波纹删除、波纹剪切或用快捷键进行剪辑。 同步模式:在此模式中,当插入片段或剪辑片段时,编辑结果会影响其它轨道。当要求插入片段时不影响轨道素材的对齐时使用。 波纹模式适用于以下条件: 1、修整素材时 2、删除素材时 3、插入或编辑时(波纹模式中,保留要插入的空白长度) 4、更改“速度”时 非波纹模式时,波纹删除、波纹剪切或用快捷键进行波纹剪辑都会影响相同的编辑内容 如果你目前为止还没接到这种类型的制作项目,我猜它可能快要来了。一个老朋友或者亲戚会打电话过来询问你是否能够将他们过去一年中用数码相机拍摄的照片制作成DVD。你可能立即会想:根据以前的经验,这可能是一个5-10小时的工作量。但是现在使用EDIUS的话,那也许只是5-10分钟而已。 告诉你的朋友:他们只需要将喜爱的照片挑出来,放到一个文件夹中并且按想要的显示顺序为照片编一下号即可。因为如果不手动重命名的话,数码相机会按照拍摄顺序自动命名它们。你可以按照日期排列一下,或者使用Picasa(一种免费的图像管理软件)或其他图像软件来批量重命名。 接下来按照这些步骤做: 1. 将图像文件从CD或者其他可移动媒介上复制到你的本地硬盘中。 2.启动EDIUS并将图片导入素材库。使用素材库窗口的文件夹图标打开“添加文件”对话框,你可以使用“选择全部”来一次性导入所有需要的图像文件。

会声会影制作卡拉OK字幕的方法

会声会影制作卡拉OK字幕的方法 1 既然Sayatoo制作的字幕会10能导入,那只装Sayatoo制作字幕不就行了吗?何必这么麻烦装这么多软件? 问: 在网上看了好几天,理不清个头绪来,有人说:1.首先下载并安装Sayatoo卡拉字幕精灵。 2.使用KAJConvert3将小灰熊字幕的ksc文件简单地转换为Sayatoo卡拉字幕精灵的kaj文件,不受歌词行数限制。(为何这样就不受歌词限制了,我搞不懂。) 3.在会声会影里直接把kaj文件拖到覆盖轨道上即可。又看到一句是:并不是用Sayatoo卡拉字幕精灵制作字幕,而是用它搭桥,就像FS搭桥一样。本人是个菜鸟,不明怎样搭桥,FS搭桥又是什么?厚着脸皮问了:)我就不明白,究竟装Sayatoo卡拉ok字幕精灵有何用?如果用小灰熊制作的KSC文件,直接用KAJConvert3转换成kaj文件不就行了吗?还有,既然Sayatoo制作的字幕会10能导入,那只装Sayatoo制作字幕不就行了吗?何必这么麻烦装这么多软件?[/size] 答: (1)如果你有正版的Sayatoo的话,那就只要装Sayatoo直接制作字幕就行了。不注册的Sayatoo 输出字幕文件是受歌词行数的限制的,装了它绘声绘影就能识别kaj文件,并不用它来输出字幕,即所谓的搭桥。 (2) 装这么多软件的原因是为了自己方便。看上去虽然不方便,但事实是为了自己的方便。因为 ①网上的KAJ文件少,网上的LRC文件多。下载方便②如果每个要自己制作歌词(KAJ)就比较麻烦,费力。③没有直接LRC转KAJ的软件。 2 会系中导不进KAJ文件 https://www.wendangku.net/doc/fd18652623.html,/read-htm-tid-11294.html 问:我安装了Ulead VideoStudio 9 手动安装: * *:\Program Files\Sayatoo Soft\Sayatoo KaraTitleMaker\host 的插件uvKAJ.vio 复制到会9中vio插件文件夹中会系中有“KAJ”这一项,就是插不进。 答虽然你手动安装:* *:\Program Files\Sayatoo Soft\Sayatoo KaraTitleMaker\host 的插件uvKAJ.vio 复制到会9中vio插件文件夹中,但你没有完全安装好Sayatoo制作字幕,建议重装Sayatoo制作字幕。 3 我用小灰熊做了一个卡拉OK歌词文件是KSC文件,如何能加在会9或10里 问: 我用小灰熊做了一个卡拉OK歌词文件是KSC文件,如何能在会9或10中使用? 答打开ksc转kaj软件,就是“友源”推荐的KAJConvert3软件,将ksc文件转换成kaj,

(完整版)(整理)自动分拣系统的设计

绪论 分拣是把很多货物按品种从不同的地点和单位分配到所设置的场地的作业。按分拣的手段不同,可分为人工分拣、机械分拣和自动分拣。 目前自动分拣已逐渐成为主流,因为自动分拣是从货物进入分拣系统送到指定的分配位置为止,都是按照人们的指令靠自动分拣装置来完成的。这种装置是由接受分拣指示情报的控制装置、计算机网络,把到达分拣位置的货物送到别处的的搬送装置。由于全部采用机械自动作业,因此,分拣处理能力较大,分拣分类数量也较多。 随着社会的不断发展,市场的竞争也越来越激烈,因此各个生产企业都迫切地需要改进生产技术,提高生产效率,尤其在需要进行材料分拣的企业,以往一直采用人工分拣的方法,致使生产效率低,生产成本高,企业的竞争能力差,材料的自动分拣已成为企业的唯一选择。针对上述问题,利用PLC 技术设计了一种成本低,效率高的材料自动分拣装置,在材料分拣过程中取得了较好的控制效果。 物料分拣采用可编程控制器PLC 进行控制,能连续、大批量地分拣货物,分拣误差率低且劳动强度大大降低,可显著提高劳动生产率。而且,分拣系统能灵活地与其他物流设备无缝连接,实现对物料实物流、物料信息流的分配和管理。其设计采用标准化、模块化的组装,具有系统布局灵活,维护、检修方便等特点,受场地原因影响不大。同时,只要根据不同的分拣对象,对本系统稍加修改即可实现要求。 PLC控制分拣装置涵盖了PLC技术、气动技术、传感器技术、位置控制技术等内容,是实际工业现场生产设备的微缩模型。 应用PLC技术结合气动、传感器和位置控制等技术,设计不同类型材料的自动分拣控制系统。该系统的灵活性较强,程序开发简单,可适应进行材料分拣的弹性生产线的需求。本文主要介绍了PLC控制系统的硬件和软件设计,以及一些调试方法。 精品文档

毕业设计---机械手自动分拣监控系统仿真

毕业设计说明书 题目基于组态软件MCGS的机械手自动分拣监控系统仿真专业机电一体化技术 班级 姓名 学号 指导教师 二○一○年十二月

2011届毕业设计(论文)任务书 设计题目: 基于组态软件MCGS的机械手自动分拣监控系统仿真 设计条件:要求利用组态软件MCGS仿真满足控制要求的机械手自动分拣监控系统的运行过程。 设计任务: 机械手分拣系统主要由三个机械手和一条传送带组成,三个机械手的功能分别是上料,正品捡拾和次品捡拾,在每个机械手旁边都有料盒,上料机械手按照一定要求将待分拣产品放在传送带上,分拣机械手是按照检测结果将产品分类,分别放入各自身旁的料盒中,传送带按一定速度运转,其上安装三个间隔相同的位置传感器,第一个位置传感器旁装有产品质量传感器,用来判断到来的产品是否合格,第二个和第三个位置传感器分别放置在两个分拣机械手附近,当传感器感应到产品到时可发出信号驱动相应的机械手动作。控制要求如下: 1.传送带按间歇方式工作,除在上料和产品捡拾时处于停滞状态,其他时间连续运转。 2.初始时,传送带停止,上料机械手实现上料操作,完成后启动传送带;当产品运行到位置传感器1时,传送带停止,进行产品质量检测,判断是否合格,同时上料机械手再上料,完成后启动传送带。 3.两个产品同时分别到达位置传感器1和位置传感器2,传送带停止,系统判断位置传感器2处的产品是否合格,如合格驱动正品机械手动作,如不合格,正品机械手不动作,等该产品到达位置传感器3时次品机械手动作,位置传感器1处的产品接受质量检测,记录该产品的质量信息,同时上料机械手再进行上料,完成后启动传送带。毕业设计(论文)内容包括: 1)组态监控画面的设计及实时数据库的构建。 2)脚本程序的设计思路及流程图。 3)脚本软件的编程及设计要求的实现。 起止日期:2010年月日- 2010年月日(共周) 指导教师:赵建伟 审核(教研室主任):批准(系主任):

物流配送中心的自动分拣系统由哪些部分组成

中山新永一测控设备有限公司 物流配送中心的自动分拣系统由哪些部分组成 1、输入装置:被拣商品由输送机送入分拣系统。 2、货架信号设定装置:被拣商品在进入分拣机前,先由信号设定装置(键盘输入、激光扫描条码等)把分拣信息(如配送目的地、客户户名等)输入计算机中央控制器。 3、进货装置:或称喂料器,它把被拣商品依次均衡地进入分拣传送带,与此同时,还使商品逐步加速到分拣传送带的速度。 4、分拣装置:它是自动分拣机的主体,包括传送装置和分拣装置两部分。前者的作用是把被拣商品送到设定的分拣道口位置上;后者的作用是把被拣商品送入分拣道口。 5、分拣道口:是从分拣传送带上接纳被拣商品的设施。可暂时存放未被取走的商品,当分拣道口满载时,由光电管控制阻止分拣商品不再进入分拣道口。 6、计算机控制器:是传递处理和控制整个分拣系统的指挥中心。自动分拣的实施主要靠它把分拣信号传送到相应的分拣道口,并指示启动分拣装置,把被拣商品送入道口。物流配送中心的分拣机控制方式主要是脉冲信号跟踪法。 分拣系统由哪些设备组成? 自动分拣系统一般由控制装置、分类装置、输送装置及分拣道口组成。控制装置的作用是识别、接收和处理分拣信号,根据分拣信号的要求指示分类装置、按商品品种、按商品送达地点或按货主的类别对商品进行自动分类。这些分拣需求可以通过不同方式,如可通过条形码扫描、色码扫描、键盘输入、重量检测、语音识别、高度检测及形状识别等方式,输入到分拣控制系统中去,根据对这些分拣信号判断,来决定某一种商品该进入哪一个分拣道口。 分类装置的作用是根据控制装置发出的分拣指示,当具有相同分拣信号的商品经过该装置时,该装置动作,使改变在输送装置上的运行方向进入其它输送机或进入分拣道口。分类装置的种类很多,一般有推出式、浮出式、倾斜式和分支式几种,不同的装置对分拣货物的包装材料、包装重量、包装物底面的平滑程度等有不完全相同的要求。 输送装置的主要组成部分是传送带或输送机,其主要作用是使待分拣商品贯通过控制装置、分类装置,并输送装置的两侧,一般要连接若干分拣道口,使分好类的商品滑下主输送机(或主传送带)以便进行后续作业。 分拣道口是已分拣商品脱离主输送机(或主传送带)进入集货区域的通道,一般由钢带、皮带、滚筒等组成滑道,使商品从主输送装置滑向集货站台,在那里由工作人员将该道口的所有商品集中后或是入库储存,或是组配装车并进行配送作业。 以上四部分装置通过计算机网络联结在一起,配合人工控制及相应的人工处理环节构成一个完整的自动分拣系统。

相关文档