“A long-awaited startâ€, today (December 22), the CFDA issued a notice saying that in order to standardize and guide the research and evaluation of cell therapy products based on drug development and registration, the General Administration of the People’s Republic of China has organized the research on cell therapy products. And evaluation technical guidelines (Trial) (hereinafter referred to as the guiding principles).
According to the Guiding Principles, in recent years, with the continuous development of basic theories, technical means and clinical medical research research such as stem cell therapy, immune cell therapy and gene editing, cell therapy products have provided new and serious refractory diseases. Treatment ideas and methods. These guidelines are developed to standardize and direct the research, development, and evaluation of such products in accordance with drug management practices. Due to the rapid development of cell therapy products and the large differences in products, this principle is mainly based on the current cognition, and proposes general technical requirements related to the safety, effectiveness and quality control of cell therapy products.
The Guiding Principles clarify that the cell therapy products described in the principles refer to human-derived living cell products for the treatment of human diseases, sources, operations and clinical trial processes that meet ethical requirements and are developed and registered in accordance with drug management regulations. It is not suitable for blood components for blood transfusion, hematopoietic stem cell transplantation, reproductive-related cells, and tissue and organ products composed of cells, which have been prescribed, have not been treated in vitro.
For the risk control of cellular products, the Guiding Principles believe that the risk of cell therapy products depends largely on the source, type, nature, function, production process, non-cellular components, non-target cell populations, and throughout the production process. Prevention and control of pollution and/or cross-contamination, as well as specific treatment routes and uses.
Therefore, the Guiding Principles require that the cell source, access and manipulation processes in cell therapy products should be ethical, and producers should establish an “informed and confidential†management system that allows donors to fully understand the use and use of cells, and On the one hand, the donor's personal information is fully protected. For cell treatment products or donor cells that are unqualified during the preparation process and remaining in clinical trials, treatments that are appropriate, legal, and ethical and biosafety-related requirements must be used.
At the same time, producers of cell therapy products should establish a traceable management system to ensure traceability of the product from donor to recipient. It is necessary to list the donor-product-recipient chain, or the self-product-recipient chain. It is necessary to standardize and monitor the production operation process, and strictly control the confusion of different donor samples (or different batch samples).
In addition, the Guiding Principles also provide detailed regulations and requirements for pharmaceutical research, non-clinical research, and clinical research of cell therapy products.
Subsequently, the CFDA also interpret the issues related to the Technical Guidelines for Research and Evaluation of Cellular Therapy Products (Trial).
1. What is the background for the development of the Technical Guidelines for Research and Evaluation of Cellular Therapy Products?
Cell therapy technology is currently the key development field of international medical frontiers, which provides new hopes for the treatment of some human difficult diseases. In recent years, new research results have been continuously obtained in the field of cell therapy, and the research and evaluation and evaluation of cell therapy products are increasingly receiving high attention from domestic and foreign pharmaceutical companies and government departments. In response to the rapid development and increasingly fierce research of current cell therapy products, in order to better create a cell product research and development environment and provide technical support to relevant scientific research institutions and enterprises, the Bureau organized the drug evaluation center to start drafting cell therapy products in 2015. Technical guidelines. After extensive research on foreign related guidelines and full consultation with the industry, the Drug Evaluation Center drafted the "Guidelines for the Research and Evaluation of Cellular Therapeutic Products" (Trial), clarified the scope and positioning of the guidelines, and proposed cell therapy products in pharmacy. General principles and basic requirements to be followed in research, non-clinical research, and clinical research. The publication of this guiding principle aims to further standardize the research and development of cell therapy products, improve their safety, effectiveness and quality controllability, and thus promote and promote the healthy development of cell therapy in China.
2. What is the scope of application of this guiding principle?
This guiding principle is mainly applicable to human-derived living cell products that are researched and registered according to drugs. For cell therapy technologies/products developed in accordance with medical technology or other management pathways, other relevant regulatory requirements and technical requirements shall be implemented. This guideline does not apply to cell preparations that have clear regulatory requirements and technical standards in other sectors, including blood components for blood transfusion, hematopoietic stem cell transplantation that has been prescribed, and has not been treated in vitro; based on current knowledge and management status This guideline also does not apply to reproductive-related cells and tissues and organ products composed of cells.
3. Can human serum be used during cell culture?
Sera from any source, including human serum, should be avoided as much as possible. Applicants should provide sufficient research data to demonstrate the need to use human serum during cell culture. At the same time, the value of the clinical application of the product will be assessed in terms of risk and benefit during the technical review process. In the case of a precursor that satisfies the necessity of human serum use, the applicant shall provide the selection basis of the human serum used, the safety research data, and the applicant's control strategy for human serum quality, etc., and the cell treatment product shall not be used without safety. Sexually verified serum. With the development and innovation of technology, applicants are encouraged to actively explore safer and more defined serum substitutes for subsequent research and production.
4. What are the quality requirements for genetically modified/modified material materials that require in vitro genetic modification/engineering?
Specific requirements can refer to the technical guidelines and documents related to gene therapy products. Applicants should conduct adequate research and evaluation on the quality, safety and stability of the genetic modification/modification materials, establish corresponding testing standards, and conduct release tests. Relevant research materials such as design, operation process, production process and quality control of genetically modified/modified materials shall be provided in the application materials. Since the genetic modification/modification material may accompany the production process as a material component of the cell treatment end product, it should comply with the production quality management regulations of the drug.
5. What should be paid attention to in the production process control of cell therapy products?
The production process of cell therapy products should follow the basic norms and related principles of the "Good Manufacturing Practices". Every production step of a cell therapy product should be studied and verified to ensure the rationality and stability of the process. The whole process should be monitored during the production process, and monitoring points of important process steps should be set according to the process characteristics, and monitoring should be strengthened. The cell therapy products are directly input into the human body and cannot tolerate the inactivation or removal process of viruses, bacteria, etc., and the control in the process is very important. Applicants should fully identify and control the risk of pollution in the process, especially the risk of cross-contamination should be combined with the specific situation and the process of research and development to achieve as close as possible, automated production. In addition, measures must be taken to avoid confusion between different samples or batches. The applicant is the responsible subject, and should self-assess and strictly control the various risks of the process, so that the quality of the cell products of each patient meets the standard control requirements. In addition, applicants are reminded to pay attention to the updates required by relevant laws and regulations or technical guidance documents for the production of cell therapy products.
6. How to set up the cell treatment product quality release test project? Can you refer to the release testing items and standards of products already listed in foreign countries?
The detection mechanism of cell therapy products is recommended to use a combination of quality testing of intermediate samples and final product release testing. The intermediate sample can be a suitable and critical control node in the production process. It is necessary to consider the docking time required for the completion of the reserve test, which is convenient for sampling and retention, and does not affect the limiting factors such as subsequent processing after the sample size is extracted. The verification project should be based on product quality research and a thorough understanding of the production process and production process, taking into account the characteristics of the product and the current scientific understanding and consensus. With the deepening of research, process-related information should be gradually accumulated, and the test methods should be gradually improved to meet the quality control requirements of each stage. It is recommended to maintain the quality control of the samples for confirmatory clinical trials and the quality control requirements during commercial production. Consistent.
The items and standards for product release testing can refer to the listed products, but considering the differences in raw materials, production processes, process control, and control methods and methods of operation, the applicants need to combine the actual products. The situation establishes control items and standards that are consistent with the characteristics of the product. For important items that cannot be effectively controlled by the production process, but important items necessary for clinical efficacy and safety control, quality control should be carried out during test release.
7. Can cell therapy product release detection methods use new, rapid detection methods instead of traditional detection methods?
The method of release inspection should be studied and verified, especially for the new method established, and the applicability should be verified for the methods included in the pharmacopoeia. For products with short expiration and small sample size, researchers should compare and evaluate new test methods with traditional test methods while establishing fast and trace new test methods, without completing sufficient methodological verification and comparative studies. Can't be replaced simply. Two test methods can be used for mutual verification during product testing during clinical trials. After accumulating enough methodological verification and comparative analysis data, consider replacing.
8. What is the overall strategy for non-clinical research evaluation of cell therapy products?
Due to the rapid development of cell therapy products and complex biological characteristics and clinical indications, this principle is mainly based on the current principles of cognition and non-clinical research evaluation of drugs, and proposes non-clinical research evaluation of cell therapy products. General technical specifications covering research projects that may be involved in non-clinical studies of cell therapy products; researchers are required to fully consider product characteristics, develop clinical indications, and have obtained safety effectiveness data, specific analysis of specific issues, and scientific rationality The evaluation method determines whether or not to conduct a certain test, refer to the general scientific principles of pharmacological toxicological evaluation, and provide research data to support the proposed clinical plan.
Throughout the development phase of cell therapy products, it is recommended to discuss the timing of non-clinical research and design to meet the seamless R&D needs of the product through communication and other forms.
9. How does non-clinical research in cell therapy products follow GLP normative?
According to the requirements of relevant regulations, drug non-clinical safety tests need to be carried out in a nationally accredited GLP institution. However, in non-clinical studies of cell therapy products, there may be animal ethical or technical issues, such as obtaining toxicity data from pharmacodynamic studies using disease models, or certain endpoints of integration in toxicological tests, such as cell distribution, survival, Special immunological endpoints, etc. cannot be completed in the GLP testing facility. Although GLP normative requirements are not applied to pharmacodynamic studies, if there is a planned safety endpoint assessment, it is recommended that this part of the research indicators comply with the GLP specification. For studies or tests conducted under non-GLP conditions, the impact of non-GLP conditions on the reliability, completeness, and overall safety evaluation of cell treatment products should be accounted for and evaluated.
10. How do cell treatment products be selected for animal species?
When non-clinical studies of cell-based therapeutic products are not preferred, non-human primates are not preferred, and non-clinical studies of rodents and non-rodents are not required. The biological response of the selected animal to the product is similar or similar to the expected human response. Prior to formal non-clinical studies, it is recommended that in vitro studies (eg, functional analysis, immunophenotyping, morphological evaluation) and in vivo pre-tests be performed to determine the biological relevance of the selected animal species to the product.
Given the nature of cell therapy products, disease animal models can also be considered for non-clinical studies, while examining relevant safety endpoints to assess the potential toxicity of the product.
If there are restrictions on the choice of animals, consider using animal source substitutes for non-clinical studies, especially for new target cell therapy products. It is important to use alternative products for pharmacodynamic studies during pharmacodynamic proof-of-concept. Encourage the exploration of relevant model research.
11. What are the main considerations for the tumorigenic/carcinogenicity of cell therapy products?
In the non-clinical study, the specific cell types in the cell therapy products, the differentiation state of each cell group/subgroup typing, the influence of the production process on the cells, and the expression of transduction genes in the genetically modified cells (such as various growth) are considered. Factor), cell therapy products induce or enhance the potential of tumor formation in the host, target population and other factors, it is necessary to evaluate the risk of tumor treatment products causing tumors in the host cells or cell treatment products themselves. Traditional carcinogenicity tests may not be suitable for cell therapy products, but there is currently no scientific consensus on animal models for tumorigenicity evaluation and their predictive value. The study was described in this guideline using tumorigenicity/carcinogenicity to align with other non-cellular product safety test project names.
12. How to use existing human test data?
The purpose of non-clinical research evaluation is to assess and control the risk of developing a clinical plan to ensure the safety of the subject. If the existing human data is scientifically evaluated, it can indicate the effectiveness and safety of the cell treatment product. The safety of clinical subjects, non-clinical studies can be exempted from unnecessary animal testing based on the specific analysis of specific varieties.
For cell therapy products, risk management measures that require both non-clinical and clinical research and use the entire process of research are required. In clinical trials, it is recommended to develop risk control measures based on existing non-clinical data, human data, or related information related to similar products.
13. Can I accept non-registered clinical trial data?
In order to better meet the needs of public medicine in China, we will promote the urgent need for new drugs in our country in the morning, according to the "Opinions on Deepening the Review and Approval System Reform and Encourage Drug Medical Device Innovation" and other relevant regulations, combined with the actual situation of cell treatment product registration and supervision, based on scientific evaluation. The principle of reducing duplication of research and benefiting patients can accept non-registered clinical trial data to varying degrees to support the registration of drugs in China and the update of post-marketing safety and efficacy information. The acceptability of non-registered clinical trial data depends on the consistency of the clinical trial sample with the declared product and the process of clinical research data generation, the authenticity, completeness, accuracy and traceability of the data, as well as the National Food and Drug Administration. The General Administration of Management has comprehensively evaluated the results of the verification of clinical trials.
14. How do cell therapy products be clinically staged?
Due to the particularity of cell therapy products, the traditional Phase I, II, and III clinical study staging design cannot be fully applied to cell therapy products for clinical research. Applicants can formulate clinical research staging and research design according to the specific characteristics of the products to be applied. Generally, they can be divided into early clinical trial phase and confirmatory clinical trial phase according to the research progress. The content of the early clinical trial phase should include, in principle, preliminary safety assessments, pharmacokinetic studies, preliminary pharmacodynamic studies, and dose-exploration studies. It is recommended to obtain as much research evidence as possible in the early clinical trial phase to support subsequent confirmatory clinical trials. If necessary, encourage communication with drug review agencies to ensure the rationality of the design of confirmatory clinical trials and to facilitate research results. The research and the registration and listing of the products to be declared.
15. What are the special considerations for the selection of subjects in clinical trials of cell therapy products?
Due to the uncertainty of the risk of cell therapy products and the complexity of the mode of administration, early clinical trials should take into account the severity of the disease and the different stages of the disease as well as the existing treatments, and choose the ones that cannot benefit from the existing treatments. Test the subject and reduce the risk that the subject may bear.
16. What are the special considerations for the necessity of clinical pharmacodynamic evaluation and the selection of evaluation indicators?
The complexity and specificity of cell therapy products often lead to large population and individual differences between non-clinical studies and clinical studies. There may not even be a suitable in vitro and in vivo disease model for pharmacodynamic evaluation in non-clinical studies. Therefore, it is necessary to initially evaluate the effectiveness of the product during the early clinical trial phase. The selection of evaluation indicators should be based on the characteristics and mechanism of action of the cell treatment products, and select short-term effects or long-term outcomes that may prove potential efficacy, which is conducive to the development or even simplification of subsequent confirmatory studies.
17. Is pharmacokinetic research necessary? What should I pay attention to in research design?
It has been clarified in the guidelines that traditional pharmacokinetic studies are not suitable for pharmacokinetic studies of human cell therapy products and are therefore not necessary for cell therapy products that are not capable of performing pharmacokinetic studies at this stage. However, for cell therapy products whose mechanism of action is unknown, it is of great significance to understand the process in the human body to understand the effectiveness and safety of cell therapy products. Therefore, under the current technical conditions, cell therapy products should be carried out as much as possible. In vivo process studies, including cell viability, proliferation and differentiation, distribution/migration in vivo, and related biological functions.
18. What are the special considerations for dose discovery research design?
Unlike traditional small molecule drugs, the dose-exposure-effect relationship of cell therapy products can be complex. Therefore, the dose exploration research design for cell therapy products has its particularity. Exploring the optimal effective dose range within a safe dose range is the primary goal of a dose-exploration study, and the need to determine the maximum tolerated dose should be based on the specifics of the cell treatment product. The initial dose setting for early clinical trials can be based on previous clinical experience, and the first human trial should be performed in a single dose. Minimize exposure of the subject to ineffective doses based on the safety of the subject. The dose escalation setting should take into account the risks and activities associated with dose changes in preclinical data and any clinical data available, and the semi-log incremental method mentioned in the selection guidelines can also be chosen autonomously. Adequate dosing intervals and follow-up times should be set to observe acute and subacute adverse events.
19. What are the special considerations for the safety study of cell therapy products?
Since the mechanism of action of most cell therapy products at this stage is not completely clear, the safety monitoring of cell therapy products should be carried out throughout the product development process. Based on risk considerations, other subjects should be enrolled on a case-by-case basis after the first subject's safety is as fully exposed as possible. Safety monitoring indicators should be selected based on product characteristics, mechanism of action, study population, non-clinical findings, and any relevant clinical experience, with an emphasis on assessing and monitoring the specific expected safety risks of the product. For products that are expected to have long-term activity, patients should be followed up to determine the long-term effectiveness of the therapeutic product and adequate exposure to product-related safety issues. The duration of follow-up should provide preliminary evidence of efficacy and duration of activity of the product, and should consider whether the product causes delayed hair safety issues and other factors. Due to the high risk of some cell therapy products, in order to better protect the subjects, it is recommended to select researchers and clinical research institutions with corresponding risk prevention and control capabilities and experience, and systematic training of relevant staff involved in clinical trials. .
Technical guidelines for research and evaluation of cell-based therapeutic products
(Trial)
I. Introduction
In recent years, with the continuous development of basic theories, technical means and clinical medical exploration research such as stem cell therapy, immune cell therapy and gene editing, cell therapy products have provided new therapeutic ideas and methods for some serious and refractory diseases. These guidelines are developed to standardize and direct the research, development, and evaluation of such products in accordance with drug management practices. Due to the rapid development of cell therapy products and the large differences in products, this principle is mainly based on the current cognition, and proposes general technical requirements related to the safety, effectiveness and quality control of cell therapy products. With the development of technology, the improvement of cognition and the accumulation of experience, the specific technical requirements for products of different cell types will be gradually improved, refined and revised. Since this guideline covers products of multiple cell types, the applicability of technical requirements should also be based on the specific analysis of the specific issues.
Second, the scope
The cell therapy products described in the present guidelines refer to human-derived living cell products for the treatment of human diseases, source, operation and clinical test procedures in accordance with ethical requirements, research and development and registration in accordance with drug management regulations. This guideline does not apply to blood components for blood transfusion, hematopoietic stem cell transplantation, reproductive-related cells, and tissue and organ products composed of cells that have been prescribed, have not been treated in vitro.
Third, risk control
Due to the variety of cell treatment products, large differences, complex and variable nature, rapid research, rapid technology update, and different degrees of risk, for different types of products, based on risk characteristics and special control measures, the unique technology suitable for its products can be adopted. .
The risk of cell therapy products depends to a large extent on the source, type, nature, function, production process, non-cellular components, non-target cell populations, contamination and/or cross-contamination control throughout the production process, and specific Treatment routes and uses. The preparation and use of different cell therapy products may present different degrees of risk to patients. Cell therapy products should develop appropriate risk control programs based on different risks. From the initial development of cell therapy products, comprehensive analysis should be based on existing knowledge and intended use, and data should be continuously collected and updated throughout the product life cycle to clarify and prevent risks.
When assessing the overall risk of a product, consideration should be given to the effects of various factors on product risk, such as the source of the cell, the degree of cell manipulation, cell proliferation, differentiation, migration, cell exposure to specific culture materials, cell culture time. , cell survival and cell generation, toxic effects of non-cellular components, physical and chemical treatment or genetic modification/engineering changes in cell characteristics, combinations of cells and bioactive molecules or structural materials, activation of immune responses The ability to cross-react with immune recognition, the manner in which it is used, and the pre-treatment of the recipient, similar product experience or availability of relevant clinical data.
In the development of cell therapy products, various risk factors should be continuously analyzed and evaluated. In particular, comprehensive risk analysis results should be used to: identify risk factors related to product quality and safety; determine non-clinical and clinical applications. The scope and focus of the data required; the process of determining risk minimization measures, etc.
The source, acquisition and manipulation of cells in cell therapy products should be ethical. Producers should establish an “informed and confidential†management system that allows donors to fully understand the use and use of cells and, on the other hand, fully protect the donor's personal information. For cell treatment products or donor cells that are unqualified during the preparation process and remaining in clinical trials, treatments that are appropriate, legal, and ethical and biosafety-related requirements must be used.
Producers of cell therapy products should establish a traceable management system to ensure traceability of the product from donor to recipient. It is necessary to list the donor-product-recipient chain, or the self-product-recipient chain. It is necessary to standardize and monitor the production operation process, and strictly control the confusion of different donor samples (or different batch samples).
Fourth, pharmaceutical research
(1) General principles
Since the cells themselves have the ability to survive, proliferate or/and differentiate in vivo, their pharmaceutical research and quality control should fully consider the above basic characteristics, and the cell therapy products should meet the general requirements of drug quality management, and the whole process of clinical sample production should be It complies with the basic principles and related requirements of the "Good Manufacturing Practices". Special attention should be paid to personnel, environment, equipment and other requirements during the production process. The production of cell therapy products should establish a whole process control system. The production process should undergo strict process verification and establish clear key control points; the quality of production materials should be strictly controlled and the operation specifications of production lines should be established to avoid production raw materials and production. Exogenous or cross-contamination that may be introduced during operation; effective isolation measures should be developed to prevent confusion between different donor-derived products or different batches.
Researchers need to comprehensively assess the rationality of donor cell application based on the characteristics of the product itself. In general, donors should be screened prior to collection, including a comprehensive examination of health status (eg general information, past medical history and familial genetic disease), screening for infection by pathogenic microorganisms, and investigation of stays in hazardous areas. .
(2) Materials for production
Production materials refer to substances or materials used to prepare cell therapeutic products, including cells, culture media, cytokines, various added components, cryopreservation solutions, genetic modification/modification materials, and excipients. Production materials are directly related to the quality of the product, so researchers should establish a good and standardized quality management system for production materials, including assessment of the use of risks, auditing of suppliers of key production materials, and development of quality release testing mechanisms. Work program.
1. cell
1.1 donor cell
The donor cell source should meet the relevant national laws, regulations and ethical requirements. The operation steps of donor cell acquisition, transportation, sorting, inspection or preservation should be studied and verified, and clear specifications and requirements should be formulated on this basis. For example, characteristics of donor cells, culture conditions, generations, growth characteristics, preservation status, storage conditions, and inspection conditions. In principle, a cell bank should be established for preservation and production of donor cells suitable for establishing a cell bank. The level of the cell bank can be comprehensively considered according to the characteristics of the cell, the production situation and the clinical application; and the test standard of the cell bank should be established, and the test should meet the basic requirements of safety, quality controllability and/or effectiveness.
1.2 production process cells
Production process cells, such as cells for the production of viruses, should in principle be consistent with the clear culture of source and history, controllable safety risks, compliance with the needs of production technology and the establishment of basic principles for cell bank management.
2. Other production materials
2.1 Raw materials
The source, composition, use, dosage and quality control of raw materials for production should be clear and reasonable. The selection of raw materials should consider the necessity, rationality and safety of their use, such as unintended effects that may lead to cell mutation or the possibility of sensitization, and should carry out process removal verification and safety risk assessment, if necessary, The residual amount is subjected to release detection. Raw materials that have been approved for use in the human body or that meet Pharmacopoeia standards should be used as much as possible. Bio-sourced raw materials should be tested for comprehensive exogenous factors and should take into account the knowledge of new exogenous factors. Self-use products should be strictly protected against the risk of possible exogenous factors.
Screening kits, sorting reagents or materials used in the production of cell therapy products, antibodies or magnetic beads for cell separation or activation, culture media, additives to culture processes, and production equipment or materials in contact with products or intermediate samples Strict screening and applicability assessment should be followed, and safety risks such as infectious pathogenic microorganisms and immunogenicity should be considered. It is recommended to use products approved by regulatory authorities as much as possible. Otherwise, it is recommended to use high quality grade products suitable for the product. In the process of cell culture, animal or human-derived substances should be avoided as much as possible. For example, serum should be avoided as much as possible. If serum must be used, relevant research data should be provided to explain the necessity and rationality of use. Animal serum; serum that has not been tested for safety should not be used.
For products that require genetic modification/engineering, the source, composition, and quality control of the genetic material used in the genetic modification/reconstruction process should be clarified. For specific requirements, refer to relevant technical guidelines and/or documents. Since the genetic modification/modification substance can be used as the final product, it should comply with the production quality management regulations of the drug.
2.2 accessories
The use, dosage and quality of excipients in cell therapy products should be studied and verified to demonstrate the necessity, safety and rationality of their use. Excipients approved for use in the human body should be preferred, otherwise comprehensive research and evaluation is required. Appropriate non-clinical safety studies should be conducted for new types of excipients.
(III) Preparation process and process control
The preparation process of a cell therapy product refers to a series of in vitro procedures from the donor's acquisition of the donor cell to the entry of the cell product into the recipient. Researchers should conduct research and verification of the process to prove the feasibility and robustness of the process. The design of the production process should avoid unpredictable or abnormal changes in cells and meet the requirements for removal of related impurities; standard process steps, process control parameters, internal control indicators and waste standards should be established to monitor the entire production process. Researchers should continually optimize the preparation process to reduce the physical, chemical or biological effects that have unintended effects on cell characteristics and reduce the introduction of impurities such as proteases, nucleases, and selective inhibitors. It is recommended to use a continuous preparation process as much as possible. If there are discontinuous production in the production process, the storage conditions and duration of the cells should be studied and verified. It is recommended to use closed or semi-closed preparation processes as much as possible to reduce the risk of contamination and cross-contamination.
The monitoring of the whole process of the production process includes the monitoring of production process parameters and the achievement of process control indicators. Based on the understanding of the overall process and the accumulated experience of the production products, the researcher should clarify the key production steps in the process control and formulate the limited range of sensitive parameters to avoid process offset. If necessary, the quality of the cells in the preparation process can be monitored, and the quality monitoring and cell release detection in the process can be combined and complemented to achieve overall process and product quality control. For example, when a cell needs to be genetically modified/modified in vitro, it needs to pay attention to the transduction efficiency of the gene substance, the integration of the gene into the cell, the phenotype and genotype of the cell, the genetic stability of the target gene, and the genetic material for transduction. Residues, as well as virus replication ability back mutations; when cells are induced to differentiate in vitro, they need to pay attention to cell differentiation, cell growth characteristics (such as malignant transformation), cell phenotype and / or genotype, induced substances Residual conditions, etc.
The dosage form, formulation and prescription process of the product should be determined according to the clinical drug requirements and the stability of the product itself. Some cell therapy products require a change in the physical state of the product components, conversion of the container, filtration and cleaning, combination with other structural materials, and adjustment of the dosage of the drug before administration. The determination of these process steps should also be studied. With verification, and strictly enforced in practical applications.
(4) Quality research and quality control
1. Quality research
Quality studies of cell therapy products should be conducted by selecting representative production batches and appropriate production phase samples (eg, initially isolated cells, cells or finished products during preparation, etc.). Quality studies should cover aspects such as cell characterization, functional analysis, purity analysis, and safety analysis, and additional related research projects can be added based on the product's own characteristics.
Cell characteristics studies should be based on the characteristics of different types of cells, such as cell identification (genotype, phenotype, etc.), differentiation potential studies, expression of surface markers, biological activity, response to exogenous stimuli, and qualitative expression products. And quantitative aspects of research. For the expected product to be a mixture of many different types or different genotypes/phenotypes, it is recommended to conduct an identification study and quantitative control of the mixed characteristics of the cells.
In terms of functional analysis, functional research methods should be established for the nature, characteristics and intended use of the cells, and used for research and analysis. The study should consider the mechanism of action of the product (such as direct cell action, cell secretion factor or other), combined with clinical indications or other alternative indicators to establish a reasonable and effective method for the detection of biological efficacy.
In terms of cell purity, it is recommended to detect the percentage of viable cells, the percentage of subcellular fractions, etc.; if the cells are genetically modified/engineered or induced to differentiate, it is recommended to detect the ratio of functional cells. It is recommended that qualitative and quantitative research and/or quality control of other non-target cell populations be considered based on the risk profile of the clinical application.
å®‰å…¨æ€§ç›¸å…³çš„ç ”ç©¶åº”æ ¹æ®ç»†èƒžæ¥æºå’Œåˆ¶å¤‡å·¥è‰ºè¿‡ç¨‹çš„特点考虑,å¯é€‰æ‹©é’ˆå¯¹å¤–æºæ€§å› åã€ç»†èƒžæ¶æ€§è½¬åŒ–çš„å¯èƒ½æ€§ã€æˆç˜¤æ€§å’Œè‡´ç˜¤æ€§ã€ç›¸å…³æ‚è´¨ã€ç—…毒载体回å¤çªå˜ç‰æ–¹é¢å¼€å±•ç ”究。相关æ‚è´¨ç ”ç©¶ä¸åº”包括工艺ä¸å¼•å…¥çš„æ‚质(如蛋白酶ã€åˆ†åŒ–诱导试剂ã€ç—…毒载体ã€å¾®ç ç‰ï¼‰å’Œäº§å“相关的æ‚质(如细胞éžé¢„期表达的产物ã€æ»ç»†èƒžæ®‹ä½™å’Œå…¶ä»–å¯èƒ½çš„生物é™è§£äº§ç‰©ç‰ï¼‰ã€‚
2. QC
ç ”ç©¶è€…éœ€å»ºç«‹ç»†èƒžæ²»ç–—äº§å“çš„è´¨é‡æŽ§åˆ¶ç–略。建议采用ä¸é—´æ ·å“çš„è´¨é‡æ£€éªŒå’Œç»ˆäº§å“放行检验相结åˆçš„机制。检定项目应当建立在产å“è´¨é‡ç ”究以åŠå¯¹ç”Ÿäº§å·¥è‰ºå’Œç”Ÿäº§è¿‡ç¨‹å……分ç†è§£çš„基础之上,åŒæ—¶å…¼é¡¾äº§å“的特性和当下的科å¦è®¤çŸ¥ä¸Žå…±è¯†ã€‚éšç€ç ”究的ä¸æ–深入(如从临床å‰é˜¶æ®µè¿›è¡Œè‡³ä¸´åºŠé˜¶æ®µï¼‰ï¼Œå·¥è‰ºç›¸å…³ä¿¡æ¯åº”é€æ¸èŽ·å¾—累积,检验方法应é€æ¥å®Œå–„,以适应å„阶段的质é‡æŽ§åˆ¶è¦æ±‚,建议确è¯æ€§ä¸´åºŠè¯•éªŒç”¨æ ·å“çš„è´¨é‡æŽ§åˆ¶ä¸Žå•†ä¸šåŒ–生产时的质控è¦æ±‚ä¿æŒä¸€è‡´ã€‚è´¨é‡æŽ§åˆ¶ä¸€èˆ¬åº”考虑鉴别ã€ç”Ÿç‰©å¦æ•ˆåŠ›ã€çº¯åº¦ã€æ‚è´¨ã€ç»†èƒžæ•°é‡ï¼ˆæ´»ç»†èƒžæ•°ã€åŠŸèƒ½ç»†èƒžæ•°ç‰ï¼‰å’Œä¸€èˆ¬æ£€æµ‹ï¼ˆå¦‚æ— èŒã€æ”¯åŽŸä½“ã€å†…æ¯’ç´ ã€å¤–观ã€é™¤ç»†èƒžä¹‹å¤–的其他外æºæ€§å¼‚物ç‰ï¼‰ç‰ã€‚éªŒæ”¶æ ‡å‡†çš„åˆ¶è®¢åº”ä»¥ä¸´åºŠå‰ç ”究批次ã€ä¸´åºŠç ”究批次和验è¯æ‰¹æ¬¡ä¸æ£€æµ‹èŽ·å¾—çš„æ•°æ®ï¼Œä»¥åŠå…¶ä»–相关数æ®ï¼ˆå¦‚ç»éªŒã€æ–‡çŒ®æŠ¥é“å’Œç¨³å®šæ€§ç ”ç©¶ç‰ï¼‰ç¡®å®šã€‚
当放行检验å—到时间é™åˆ¶æ—¶ï¼Œå¯è€ƒè™‘åŠ å¼ºå·¥è‰ºè¿‡ç¨‹ä¸çš„æ ·å“è´¨é‡ç›‘控,将过程控制与放行检验相结åˆï¼Œé€šè¿‡è¿‡ç¨‹æŽ§åˆ¶ç®€åŒ–放行检验。以上æ“作应ç»è¿‡ç ”究与验è¯ï¼Œå¹¶é™„有相应的æ“作规范。建议尽é‡åœ¨äº§å“临床应用å‰å®Œæˆå…¨éƒ¨æ”¾è¡Œæ£€éªŒï¼Œå½“有些放行检验结果å¯èƒ½åŽç½®æ—¶ï¼Œåº”对å¯èƒ½å‡ºçŽ°çš„éžé¢„期检验结果制订处置方案。为对产å“进行回顾性分æžæˆ–进一æ¥åˆ†æžï¼Œå»ºè®®ç ”ç©¶è€…æ ¹æ®äº§å“自身特点,并å‚照《è¯å“生产管ç†è§„范》ä¸çš„è¦æ±‚è¿›è¡Œç•™æ ·å¤‡æŸ¥ã€‚
一些细胞治疗产å“在给è¯å‰è¿˜éœ€ç»è¿‡ä¸€ç³»åˆ—æ“作æ¥éª¤ï¼Œç ”究开å‘时应开展模拟给è¯è¿‡ç¨‹çš„ç›¸å…³ç ”ç©¶ã€‚å¦‚æžœè¯¥æ“作在医疗机构开展,产å“生产者应明确æ“作æ¥éª¤å’Œæ³¨æ„事项,以指导临床医护工作者æ£ç¡®æ“作使用。建议在给è¯å‰å®Œæˆæ“作åŽå¯¹æœ€ç»ˆäº§å“进行质é‡æ ¸å‡†ï¼Œå¦‚细胞形æ€ã€æ´»ç»†èƒžæ•°åŠæ¯”率ã€é¢œè‰²ã€é™¤ç»†èƒžä¹‹å¤–的其他外æºæ€§å¼‚物ç‰ï¼Œä»¥åŠæ“作æ¥éª¤çš„å¤æ ¸å’Œæ ‡ç¾æ ¸å¯¹ç‰ã€‚
放行检验用方法应ç»è¿‡ç ”究与验è¯ï¼Œç‰¹åˆ«æ˜¯å¯¹äºŽå»ºç«‹çš„新方法应进行全é¢çš„验è¯ï¼Œå¯¹äºŽè¯å…¸ä¸æ”¶å½•çš„方法应进行适用性的验è¯ã€‚对于有效期çŸå’Œæ ·æœ¬é‡å°çš„产å“,å¯é‡‡ç”¨å¿«é€Ÿã€å¾®é‡çš„æ–°åž‹æ£€æµ‹æ–¹æ³•ã€‚ç ”ç©¶è€…åº”å¯¹æ–°åž‹æ£€éªŒæ–¹æ³•ä¸Žä¼ ç»Ÿæ£€æµ‹æ–¹æ³•è¿›è¡Œæ¯”è¾ƒå’Œè¯„ä¼°ï¼Œå¿…è¦æ—¶ï¼Œåœ¨äº§å“放行检验时å¯ä»¥é‡‡ç”¨ä¸¤ç§æ£€éªŒæ–¹æ³•è¿›è¡Œç›¸äº’验è¯ã€‚
ï¼ˆäº”ï¼‰ç¨³å®šæ€§ç ”ç©¶
细胞治疗产å“的生产建议采用连ç»çš„工艺,对于生产过程ä¸éœ€è¦ä¸´æ—¶ä¿å˜çš„æ ·å“åº”è¿›è¡Œç¨³å®šæ€§ç ”ç©¶ï¼Œä»¥æ”¯æŒå…¶ä¿å˜æ¡ä»¶ä¸Žå˜æ”¾æœŸã€‚细胞治疗产å“ç¨³å®šæ€§ç ”ç©¶çš„åŸºæœ¬åŽŸåˆ™å¯å‚照一般生物制å“ç¨³å®šæ€§ç ”ç©¶çš„è¦æ±‚ï¼Œå¹¶æ ¹æ®äº§å“自身的特点ã€ä¸´åºŠç”¨è¯çš„需求,以åŠä¿å˜ã€åŒ…装和è¿è¾“的情况设计åˆç†çš„ç ”ç©¶æ–¹æ¡ˆã€‚åº”é‡‡ç”¨å…·æœ‰ä»£è¡¨æ€§çš„ç»†èƒžæ ·æœ¬å’Œå˜å‚¨æ¡ä»¶å¼€å±•ç ”究。其ä¸ï¼Œéœ€è¦ç‰¹åˆ«å…³æ³¨ç»†èƒžæ²»ç–—产å“çš„è¿è¾“ç¨³å®šæ€§ç ”ç©¶å’Œä½¿ç”¨è¿‡ç¨‹ä¸çš„ç¨³å®šæ€§ç ”ç©¶ç‰ï¼Œåº”å¼€å±•ç ”ç©¶è¯æ˜Žåœ¨æ‹Ÿå®šçš„å˜å‚¨æ¡ä»¶ä¸‹ï¼Œç»†èƒžæ²»ç–—产å“çš„è´¨é‡ä¸ä¼šå—到è¿è¾“ã€ä½¿ç”¨ä¸æˆ–其他外界æ¡ä»¶çš„å½±å“ã€‚åº”æ ¹æ®äº§å“自身的特点和å˜å‚¨æ¡ä»¶ç‰æ–¹é¢ï¼Œåˆç†åœ°è®¾è®¡ç¨³å®šæ€§è€ƒå¯Ÿçš„é¡¹ç›®å’Œæ£€æµ‹æŒ‡æ ‡ï¼Œä¾‹å¦‚ï¼Œå†·å†»å‚¨å˜çš„æ ·å“或产å“一般应模拟使用情形(如细胞å¤è‹è¿‡ç¨‹ï¼‰å¼€å±•å¿…è¦çš„冻èžç ”究。考察项目建议涵盖生物å¦æ•ˆåŠ›ã€ç»†èƒžçº¯åº¦ã€ç»†èƒžç‰¹æ€§ã€æ´»ç»†èƒžæ•°åŠæ¯”率ã€åŠŸèƒ½ç»†èƒžæ•°å’Œå®‰å…¨æ€§ç›¸å…³çš„内容ç‰ã€‚
(å…)容器和密é—系统
为é¿å…由于å˜å‚¨è€Œå¯¼è‡´çš„产å“è´¨é‡å‘生éžé¢„期å˜åŒ–ï¼Œç ”ç©¶è€…åº”å¯¹ç»†èƒžæ²»ç–—äº§å“生产过程ä¸çš„æ ·å“å’Œï¼æˆ–æˆå“ä¿å˜ç”¨çš„包装容器和密é—ç³»ç»Ÿè¿›è¡Œå®‰å…¨æ€§è¯„ä¼°å’Œç›¸å®¹æ€§ç ”ç©¶ï¼Œä»¥è¯´æ˜Žå…¶ä½¿ç”¨çš„åˆç†æ€§ï¼Œä¾‹å¦‚密å°æ€§ç ”究ã€å†·å†»å‚¨å˜é€‚åº”æ€§ç ”ç©¶ç‰ã€‚
细胞治疗产å“生产过程ä¸å¯èƒ½æ¶‰åŠæ ·å“与容器çŸæš‚直接接触的æ¥éª¤ï¼Œå¦‚采集的组织或细胞ã€åˆ¶å¤‡è¿‡ç¨‹ä¸çš„细胞ã€æˆå“回输ç‰æ¥éª¤ï¼Œç ”ç©¶è€…åº”å¯¹ä½¿ç”¨çš„å®¹å™¨è¿›è¡Œå®‰å…¨æ€§è¯„ä¼°æˆ–æŽ¥è§¦çš„ç›¸å®¹æ€§ç ”ç©¶ç‰ã€‚
对于è¿è¾“用的次级包装容器(éžç›´æŽ¥æŽ¥è§¦ç»†èƒžï¼‰æˆ–æ料也应ç»è¿‡éªŒè¯ï¼Œå¦‚对其é®å…‰æ€§ã€å¯†å°æ€§å’ŒæŠ—击机械压力ç‰æ–¹é¢è¿›è¡Œç ”究和验è¯ã€‚
五ã€éžä¸´åºŠç ”究
(一)一般原则
1.ç ”ç©¶è¯„ä»·å†…å®¹
由于细胞治疗产å“的物质组æˆåŠä½œç”¨æœºåˆ¶ä¸Žå°åˆ†åè¯ç‰©ã€å¤§åˆ†å生物è¯ç‰©ä¸åŒï¼Œæ‰€ä»¥ä¼ 统ã€æ ‡å‡†çš„éžä¸´åºŠç ”究评价方法å¯èƒ½ä¸å®Œå…¨é€‚用于细胞治疗产å“。细胞治疗产å“çš„éžä¸´åºŠç ”究评价内容å–决于细胞类型åŠä¸´åºŠç”¨é€”,与细胞æ¥æºã€ç§ç±»ã€ç”Ÿäº§è¿‡ç¨‹ã€åŸºå› 修饰ï¼æ”¹é€ ã€å¤„æ–¹ä¸éžç»†èƒžæˆåˆ†ç‰å› ç´ å¯†åˆ‡ç›¸å…³ï¼Œè¿˜ä¸Žç ”å‘计划åŠç›¸åº”的临床试验方案有关。由于细胞治疗产å“ç§ç±»ç¹å¤šï¼Œä¸åŒäº§å“其治疗原ç†ã€ä½“内生物å¦è¡Œä¸ºã€ä¸´åºŠåº”用å˜åœ¨å·®åˆ«å’Œä¸ç¡®å®šæ€§ï¼Œå› æ¤ï¼Œå¯¹ä¸åŒäº§å“çš„ç ”ç©¶è¯„ä»·åº”éµå¾ªâ€œå…·ä½“情况具体分æžâ€çš„原则;åŒæ—¶ï¼Œäººç”¨è¯å“注册技术è¦æ±‚国际å调会(ICH)é¢å¸ƒçš„《生物技术è¯å“çš„éžä¸´åºŠå®‰å…¨æ€§è¯„价指å—》(S6)å¯ä¸ºç»†èƒžæ²»ç–—产å“çš„éžä¸´åºŠç ”究评价æä¾›å‚考。
2.å—试物è¦æ±‚
éžä¸´åºŠç ”究评价试验应尽å¯èƒ½ä½¿ç”¨æ‹Ÿç”¨äºŽä¸´åºŠè¯•éªŒçš„细胞治疗产å“;用于进行éžä¸´åºŠè¯•éªŒçš„å—试物,其生产工艺åŠè´¨é‡æŽ§åˆ¶åº”与拟用于临床试验的å—试物一致(如果ä¸ä¸€è‡´åº”给予说明,并评估其对预测人体å应的影å“)。
(1)人æºçš„细胞治疗å—试物å‚考本指导原则的è¯å¦è¦æ±‚ï¼›
(2)如果由于相关动物选择的é™åˆ¶ï¼Œå¯è€ƒè™‘使用动物æº
替代å“进行éžä¸´åºŠç ”究评价;动物æºæ›¿ä»£å“应与人æºçš„细胞治疗å—试物的生产工艺åŠè´¨é‡æ ‡å‡†å°½å¯èƒ½ç›¸ä¼¼ï¼Œå¹¶æ供必è¦çš„比较数æ®ä»¥ç¡®è®¤æ›¿ä»£å“çš„è´¨é‡å±žæ€§ã€‚å¯è€ƒè™‘如下对比:
â‘ ç»„ç»‡æˆ–æ ·æœ¬èŽ·å–的程åºï¼›
②细胞识别ã€åˆ†ç¦»ã€æ‰©å¢žä»¥åŠä½“外培养程åºï¼›
③细胞生长动力å¦å‚数(例如细胞å€å¢žæ—¶é—´ã€ç»†èƒžç”Ÿé•¿æ›²çº¿ã€ç»†èƒžå¢žæ®–高峰时间);
â‘£è¡¨åž‹å’ŒåŠŸèƒ½ç‰¹æ€§ï¼ˆæ¯”å¦‚ç”Ÿé•¿å› åå’Œç»†èƒžå› å的分泌,细胞群体特异性表型ï¼åŸºå› åž‹æ ‡å¿—ï¼‰ï¼›
⑤终产å“é…æ–¹ï¼ç»†èƒžæ”¯æž¶ç§æ¤æ–¹å¼ï¼ˆå¦‚果有);
⑥终产å“的储å˜æ¡ä»¶åŠç»†èƒžæ´»åŠ›ï¼›
⑦动物替代细胞作用方å¼ä¸Žç»ˆäº§å“细胞作用方å¼çš„异åŒã€‚
éžä¸´åºŠè¯•éªŒå—è¯•ç‰©å’Œä¸´åºŠç”¨æ ·å“的异åŒå‡åº”在新è¯ç”³æŠ¥æ—¶äºˆä»¥è¯´æ˜Žã€‚
3.动物ç§å±žé€‰æ‹©
éžä¸´åºŠç ”究评价应选择åˆé€‚ç§å±žçš„动物进行试验,所选动物对细胞治疗产å“的生物å应与预期人体ååº”æŽ¥è¿‘æˆ–ç›¸ä¼¼ã€‚åŠ¨ç‰©æ¨¡åž‹é€‰æ‹©éœ€è€ƒè™‘ä»¥ä¸‹å‡ ä¸ªæ–¹é¢ï¼š
(1)生ç†å¦å’Œè§£å‰–å¦ä¸Žäººç±»å…·æœ‰å¯æ¯”性;
(2)对人体细胞产å“或æºå¸¦äººç±»è½¬åŸºå› 的细胞产å“çš„å…ç–«è€å—性;
(3)临床给è¯ç³»ç»Ÿï¼æµç¨‹çš„å¯è¡Œæ€§ï¼Œè½¬è¿ç‰¹å®šå‰‚é‡çš„细胞到治疗é¶ç‚¹çš„å¯è¡Œæ€§ï¼›
(4)å…疫缺陷动物的适用性,对产å“进行长期安全性评估的å¯è¡Œæ€§ã€‚
å…疫功能æ£å¸¸çš„动物给予人æºç»†èƒžæ—¶å¯èƒ½å‡ºçŽ°å…疫应ç”å应,æ¤ç§æƒ…况下,å¯è€ƒè™‘采用以下模型开展éžä¸´åºŠç ”究:
①给予å…疫抑制剂于具有å…疫能力的动物;
â‘¡é—ä¼ æ€§å…疫缺陷动物;
③人æºåŒ–动物;
④在å…ç–«è±å…部ä½ç»™è¯ï¼›
⑤或者以上形å¼çš„组åˆã€‚
æŸäº›æƒ…况下,也å¯é‡‡ç”¨åŠ¨ç‰©æºæ›¿ä»£å“进行评价。
鉴于细胞治疗产å“的特性,如产å“效应æŒç»æ—¶é—´é•¿ã€åœ¨ä½“内æŒç»å˜åœ¨ã€åœ¨ç»†èƒžæ²»ç–—产å“与疾病环境间å¤æ‚的作用机制ã€ä¾µå…¥æ€§çš„ç»™è¯é€”径ç‰ï¼Œä¹Ÿå¯è€ƒè™‘采用疾病动物模型进行éžä¸´åºŠç ”究。
4.ç»™è¯æ–¹å¼ï¼ˆé€”径)
éžä¸´åºŠç ”究评价ä¸ï¼Œç»†èƒžæ²»ç–—产å“çš„ç»™è¯æ–¹å¼åº”能最大程度模拟临床拟用给è¯æ–¹å¼ã€‚如果在动物试验ä¸æ— 法模拟临床给è¯æ–¹å¼ï¼Œä¸´åºŠå‰ç ”究ä¸éœ€æ˜Žç¡®æ›¿ä»£çš„ç»™è¯æ–¹å¼ï¼æ–¹æ³•ï¼Œå¹¶é˜æ˜Žå…¶ç§‘å¦æ€§å’Œåˆç†æ€§ã€‚
当使用特殊的给è¯è£…置给è¯æ—¶ï¼Œéžä¸´åºŠè¯•éªŒé‡‡ç”¨çš„ç»™è¯è£…置系统应与临床一致。
5.å—试物分æž
应æä¾›å—试物分æžæ•°æ®ã€‚
细胞治疗产å“在给è¯å‰å¯èƒ½è¿˜éœ€ç»è¿‡ä¸€ç³»åˆ—æ“作æ¥éª¤ï¼Œåœ¨å®Œæˆæ“作åŽéœ€å¯¹å—试物进行质é‡æ£€æµ‹ï¼Œæ£€æµ‹æŒ‡æ ‡åŒ…括细胞形æ€ã€æ€»æ´»ç»†èƒžæ•°ã€ç»†èƒžå˜æ´»çŽ‡ã€é¢œè‰²ã€é™¤ç»†èƒžä¹‹å¤–的其他外æºæ€§å¼‚物ç‰ã€‚
(二)è¯æ•ˆå¦ç ”究
è¯æ•ˆå¦ç ”究应采用å¯é 的方法验è¯ç»†èƒžæ²»ç–—产å“的基本治疗机ç†ï¼Œç¡®å®šç”Ÿç‰©å¦æ•ˆåº”æ ‡å¿—ç‰©ã€‚è¯•éªŒè®¾è®¡åº”è€ƒè™‘ç»†èƒžæ²»ç–—äº§å“的作用机制ã€ç–¾ç—…周期长çŸä»¥åŠç»™è¯æ–¹å¼ç‰å› ç´ ï¼Œç»“åˆç»†èƒžæ²»ç–—产å“的特性和å˜æ´»æ—¶é—´ã€‚建议采用相关的体外和体内模型完æˆç»†èƒžæ²»ç–—产å“çš„è¯æ•ˆå¦ç ”究。
(三)è¯ä»£åŠ¨åŠ›å¦ç ”究
è¯ä»£åŠ¨åŠ›å¦ç ”究应能é˜æ˜Žç»†èƒžçš„体内过程以åŠä¼´éšçš„生物å¦è¡Œä¸ºï¼Œåº”æ ¹æ®ç»†èƒžæ²»ç–—产å“类型和特点选择åˆé€‚的动物模型,一般考虑雌雄å„åŠã€‚æ ¹æ®ç ”究目的åŠæ£€æµ‹æŒ‡æ ‡çš„临床价值,建立åˆé€‚的生物分æžæ–¹æ³•å¹¶å¯¹æ–¹æ³•è¿›è¡Œå¿…è¦çš„验è¯ã€‚è¯ä»£åŠ¨åŠ›å¦ç ”究è¦å…³æ³¨ç›®æ ‡ç»†èƒžåœ¨ä½“内的增殖ã€ç”Ÿç‰©åˆ†å的表达和ï¼æˆ–分泌,以åŠä¸Žå®¿ä¸»ç»„织的相互作用;相互作用还包括细胞治疗产å“çš„éžç»†èƒžæˆåˆ†ï¼ˆè¾…æ–™æˆåˆ†ï¼‰åŠåˆ†æ³Œçš„生物活性分å引起的相关组织å应。è¯ä»£åŠ¨åŠ›å¦ç ”究内容包括但ä¸ä»…é™äºŽä»¥ä¸‹æ–¹é¢ï¼š
1.细胞的分布ã€è¿ç§»ã€å½’å·¢
应采用一ç§æˆ–多ç§åˆé€‚的细胞追踪方法评价细胞产å“的分布ã€è¿ç§»ã€å½’å·¢åŠå…¶å˜ç»å’Œæ¶ˆäº¡ç‰¹æ€§ï¼Œå¹¶é˜è¿°æ–¹æ³•çš„科å¦æ€§ã€‚细胞治疗产å“的分布åŠå˜ç»æ—¶é—´æ˜¯å½±å“细胞治疗产å“有效性和安全性的最é‡è¦å› ç´ ï¼Œåº”è¿›è¡ŒåŠ¨æ€è§‚察,必è¦æ—¶è§‚察直至这些细胞的消失或功能丧失。å¯é€‰æ‹©çš„技术方法有影åƒæŠ€æœ¯ã€PCR技术ã€å…疫组化技术ç‰ï¼Œè¯•éªŒè®¾è®¡éœ€è¦è€ƒè™‘技术方法的适用性和优缺点。
2.细胞分化
细胞在分布ã€è¿ç§»å’Œå½’å·¢åŽè¿›ä¸€æ¥åˆ†åŒ–为功能细胞å‘挥其治疗作用或功能衰退;对于细胞产å“分化的程度åŠå…¶åŽæžœï¼ˆåŠŸèƒ½åŒ–或去功能化ã€å®‰å…¨å‚数),å¯åº”用体外方法和动物体内方法进行定é‡æˆ–å®šæ€§è¯„ä»·ç ”ç©¶ã€‚
3.对于ç»åŸºå› 修饰ï¼æ”¹é€ æ“作的人æºç»†èƒžçš„特殊考虑
å¯¹äºŽåŸºå› ä¿®é¥°ï¼æ”¹é€ 的细胞,除上述è¦æ±‚å¤–ï¼Œè¿˜éœ€å¯¹ç›®çš„åŸºå› çš„å˜åœ¨ã€è¡¨è¾¾ã€ä»¥åŠè¡¨è¾¾äº§ç‰©çš„生物å¦ä½œç”¨è¿›è¡Œå¿…è¦çš„ç ”ç©¶ï¼Œä»¥ä½“çŽ°åŸºå› ä¿®é¥°ï¼æ”¹é€ 的体内生物å¦æ•ˆåº”。
ï¼ˆå››ï¼‰å®‰å…¨æ€§ç ”ç©¶è¯„ä»·
1. GLPéµä»Žæ€§è¦æ±‚
细胞治疗产å“çš„å®‰å…¨æ€§ç ”ç©¶è¯„ä»·åº”éµä»Žã€Šè¯ç‰©éžä¸´åºŠè¯•éªŒè´¨é‡ç®¡ç†è§„范》(GLP)。对于æŸäº›åœ¨éžGLPçŠ¶å†µä¸‹å¼€å±•çš„ç ”ç©¶æˆ–æ£€æµ‹ï¼Œåº”äºˆè¯´æ˜Žå¹¶è¯„ä¼°éžGLP对试验结果å¯é 性ã€å®Œæ•´æ€§åŠå¯¹ç»†èƒžæ²»ç–—产å“总体安全性评价的影å“。
2.安全è¯ç†å¦è¯•éªŒ
细胞在体内分泌的活性物质å¯èƒ½ä¼šå¯¹ä¸æž¢ç¥žç»ç³»ç»Ÿã€å¿ƒè¡€ç®¡ç³»ç»Ÿã€å‘¼å¸ç³»ç»Ÿçš„功能ç‰äº§ç”Ÿå½±å“;细胞本身分布或æ¤å…¥äºŽé‡è¦å™¨å®˜ï¼Œç»†èƒžæ²»ç–—产å“的处方æˆåˆ†ç‰ä¹Ÿå¯èƒ½å½±å“å™¨å®˜åŠŸèƒ½ã€‚å› æ¤ï¼Œå¯¹äºŽç»†èƒžæ²»ç–—产å“应考虑进行安全è¯ç†è¯•éªŒã€‚如果在毒性试验ä¸å‘现有潜在风险,应补充开展有关安全è¯ç†è¯•éªŒã€‚
3.å•æ¬¡ç»™è¯æ¯’性试验
å•æ¬¡ç»™è¯æ¯’性试验å¯èŽ·å¾—剂é‡ä¸Žå…¨èº«å’Œï¼æˆ–局部毒性之间的剂é‡ï¼å应关系,有助于了解其毒性é¶å™¨å®˜ï¼Œä¹Ÿå¯ä¸ºé‡å¤ç»™è¯æ¯’性试验的剂é‡è®¾è®¡æ供一定的å‚考。由于细胞治疗产å“能够长时间地å‘æŒ¥åŠŸèƒ½æˆ–è¯±å¯¼é•¿æœŸæ•ˆåº”ï¼Œå› æ¤å•æ¬¡ç»™è¯çš„观察时间应考虑细胞或者细胞效应的å˜ç»æ—¶é—´ï¼Œä¸€èˆ¬åº”长于å•æ¬¡ç»™è¯æ¯’性试验常规的观察时间。
4.é‡å¤ç»™è¯æ¯’性试验
试验设计应包å«å¸¸è§„毒ç†å¦è¯•éªŒç ”究的基本è¦ç´ ,并结åˆç»†èƒžæ²»ç–—产å“的特殊性æ¥è®¾è®¡ï¼Œä»¥æœŸèŽ·å¾—å°½å¯èƒ½å¤šçš„安全性信æ¯ã€‚
动物ç§å±žé€‰æ‹©ï¼šæ ¹æ®äº§å“çš„ä¸åŒç‰¹æ€§ï¼Œé‡‡ç”¨èƒ½å¤Ÿå¯¹ç»†èƒžæ²»ç–—产å“产生生物å¦æ´»æ€§çš„动物ç§å±žè¿›è¡Œé‡å¤ç»™è¯æ¯’æ€§ç ”ç©¶ã€‚ä¸€èˆ¬æƒ…å†µä¸‹åº”é‡‡ç”¨é›Œé›„åŠ¨ç‰©è¿›è¡Œè¯•éªŒã€‚å¦‚æ— ç›¸å…³ç§å±žå¯å¼€å±•éžä¸´åºŠç ”究时,éžç›¸å…³ç§å±žçš„动物试验对评价生产工艺过程ã€å…¨å¤„方的安全性åŠéžé¶æ•ˆåº”也å¯èƒ½å…·æœ‰ä»·å€¼ã€‚
剂é‡ç»„设计:å‚考“《è¯ç‰©é‡å¤ç»™è¯æ¯’æ€§ç ”ç©¶æŠ€æœ¯æŒ‡å¯¼åŽŸåˆ™ã€‹â€ï¼Œéœ€è®¾è®¡å¤šä¸ªå‰‚é‡æ°´å¹³ã€åŒ…å«ä¸´åºŠæ‹Ÿç”¨å‰‚é‡èŒƒå›´å’Œæœ€å¤§å¯è¡Œå‰‚é‡ï¼Œå¹¶ç»“åˆå¤„方组æˆåŠç”Ÿäº§å·¥è‰ºï¼Œè®¾ç½®åˆé€‚的对照组。
è§‚å¯ŸæŒ‡æ ‡ï¼šé™¤å¸¸è§„è§‚å¯ŸæŒ‡æ ‡å¤–ï¼Œéœ€ç»“åˆäº§å“特点,选择åˆé€‚çš„è§‚å¯ŸæŒ‡æ ‡ï¼Œå°½å¯èƒ½åŒ…括形æ€å¦ä¸ŽåŠŸèƒ½å¦çš„è¯„ä»·æŒ‡æ ‡ï¼Œå¦‚è¡Œä¸ºå¦æ£€æµ‹ã€ç¥žç»åŠŸèƒ½æµ‹è¯•ã€å¿ƒåŠŸèƒ½è¯„ä»·ã€çœ¼ç§‘检查ã€å¼‚常ï¼å¼‚ä½å¢žç”Ÿæ€§ç—…å˜ï¼ˆå¦‚增生ã€è‚¿ç˜¤ï¼‰ã€ç”Ÿç‰©æ ‡å¿—物ã€ç”Ÿç‰©æ´»æ€§åˆ†å的分泌ã€å…ç–«å应以åŠä¸Žå®¿ä¸»ç»„织的相互作用ç‰ã€‚
5.å…疫原性和å…疫毒性试验
细胞治疗产å“或细胞分泌产物需è¦ç ”究其潜在的å…疫原性,å…ç–«åŽŸæ€§ç ”ç©¶å¯å‚è€ƒæœ€æ–°ç‰ˆæŠ€æœ¯ç ”ç©¶æŒ‡å¯¼åŽŸåˆ™ï¼›æ¤å¤–,还需关注细胞治疗产å“诱导产生的å…疫毒性。
6.致瘤性ï¼è‡´ç™Œæ€§è¯•éªŒ
细胞治疗产å“的致瘤性ï¼è‡´ç™Œæ€§é£Žé™©å–决于产å“ä¸ä¸åŒç»†èƒžçš„分化状æ€ã€ç”Ÿäº§è¿‡ç¨‹ä¸é‡‡ç”¨çš„细胞培养方å¼å¼•èµ·çš„生长动力å¦æ”¹å˜ã€åŸºå› 修饰ï¼æ”¹é€ ç»†èƒžçš„è½¬åŸºå› è¡¨è¾¾ï¼ˆä¾‹å¦‚å¤šç§ç”Ÿé•¿å› å)ã€è¯±å¯¼æˆ–增强宿主体内形æˆè‚¿ç˜¤çš„å¯èƒ½æ€§ä»¥åŠç›®æ ‡æ‚£è€…人群ç‰ï¼Œéœ€è¦æ ¹æ®ä»¥ä¸Šç‰¹ç‚¹è¿›è¡Œç»¼åˆè€ƒè™‘。
ç›®å‰ï¼Œå¦‚何选择致瘤性ï¼è‡´ç™Œæ€§ç ”究的动物模型尚未达æˆç§‘å¦å…±è¯†ã€‚致瘤性ï¼è‡´ç™Œæ€§è¯•éªŒåº”采用临床拟用产å“进行试验。致瘤性ï¼è‡´ç™Œæ€§è¯•éªŒéœ€ç¡®ä¿ç»†èƒžå¯åœ¨ä½“内长期å˜æ´»ä»¥è€ƒå¯Ÿæ˜¯å¦æœ‰æ½œåœ¨è‚¿ç˜¤å½¢æˆã€‚试验设计需注æ„以下方é¢ï¼šï¼ˆ1)åˆé€‚的对照组(例如阳性对照ã€ç©ºç™½å¯¹ç…§ï¼‰ï¼›ï¼ˆ2)æ¯ç»„需有足够的动物数é‡ï¼Œä½¿è‚¿ç˜¤å‘生率的分æžæ»¡è¶³ç»Ÿè®¡å¦è¦æ±‚;(3)需包å«æœ€å¤§å¯è¡Œå‰‚é‡ï¼›ï¼ˆ4)å—试物应到达拟定的临床治疗部ä½ï¼›ï¼ˆ5)足够长的试验周期。
由于å…疫排斥å应,人æºç»†èƒžæ²»ç–—产å“的致瘤性ï¼è‡´ç™Œæ€§è¯•éªŒå¯è€ƒè™‘使用å…疫缺陷的啮齿类动物模型进行。
7.生殖毒性试验
细胞治疗产å“的生殖和å‘育毒性评价主è¦æ˜¯å–决于产å“的特性ã€ä¸´åºŠé€‚应症以åŠä¸´åºŠæ‹Ÿç”¨äººç¾¤ï¼Œåº”æ ¹æ®å…·ä½“情况具体分æžã€‚
8.é—ä¼ æ¯’æ€§è¯•éªŒ
对于人æºçš„细胞治疗产å“,如果该产å“与DNA或其他é—ä¼ ç‰©è´¨å˜åœ¨ç›´æŽ¥çš„相互作用,需进行é—ä¼ æ¯’æ€§è¯•éªŒã€‚
9.特殊安全性试验
æ ¹æ®ç»†èƒžæ²»ç–—产å“的特点与临床应用情况,应考虑对局部è€å—性ã€ç»„织兼容性åŠå¯¹æ‰€åˆ†æ³Œç‰©è´¨çš„è€å—性进行评估。
10.其他毒性试验
å¯¹äºŽé‡‡ç”¨åŸºå› ä¿®é¥°ï¼æ”¹é€ 的细胞治疗产å“,需关注有å¤åˆ¶èƒ½åŠ›çš„病毒的产生和æ’å…¥çªå˜ï¼Œç‰¹åˆ«æ˜¯è‡´ç™ŒåŸºå› 的活化ç‰ç‰¹æ€§å¸¦æ¥çš„安全性风险。具体è¦æ±‚å¯å‚è§ç›¸å…³çš„技术指导文件。
å…ã€ä¸´åºŠç ”究
当细胞治疗产å“进入临床试验阶段时,应éµå¾ªã€Šè¯ç‰©ä¸´åºŠè¯•éªŒè´¨é‡ç®¡ç†è§„范》(GCP)的è¦æ±‚ã€‚ä¸´åºŠè¯•éªŒçš„ç ”ç©¶å†…å®¹åŽŸåˆ™ä¸Šåº”åŒ…æ‹¬ä¸´åºŠå®‰å…¨æ€§è¯„ä»·ã€è¯ä»£åŠ¨åŠ›å¦ç ”究ã€è¯æ•ˆå¦ç ”究ã€å‰‚é‡æŽ¢ç´¢ç ”究和确è¯æ€§ä¸´åºŠè¯•éªŒç‰ã€‚æ ¹æ®ä¸åŒç»†èƒžæ²»ç–—产å“的产å“性质,å¯é…Œæƒ…调整具体的试验设计。
鉴于细胞治疗产å“特殊的生物å¦ç‰¹æ€§ï¼Œåœ¨ä¸´åºŠè¯•éªŒç ”究ä¸ï¼Œéœ€è¦é‡‡å–ä¸åŒäºŽå…¶ä»–è¯ç‰©çš„临床试验整体ç–略。为了获得预期治疗效果,细胞治疗产å“å¯èƒ½éœ€è¦é€šè¿‡ç‰¹å®šçš„手术措施ã€ç»™è¯æ–¹æ³•æˆ–è”åˆæ²»ç–—ç–ç•¥æ¥è¿›è¡Œç»™è¯ã€‚å› æ¤ï¼Œåœ¨ä¸´åºŠè¯•éªŒæ–¹æ¡ˆè®¾è®¡ä¸åº”一并考虑。
细胞治疗产å“很多特有的性质也会影å“其临床试验的设计,包括产å“特性ã€ç”Ÿäº§ç‰¹ç‚¹ä»¥åŠä¸´åºŠå‰ç ”究的结果ç‰ã€‚
(一)å—试者的ä¿æŠ¤
å—试者的选择需è¦è€ƒè™‘多方é¢çš„å› ç´ ï¼Œé€‰æ‹©å®—æ—¨æ˜¯èƒ½ä½¿ç ”ç©¶å—试者的预期风险与潜在获益ç»è¿‡æ…Žé‡è¯„估,åŒæ—¶èƒ½å®žçŽ°ç ”究的科å¦ç›®çš„。
在早期临床试验阶段,所预期的获益或风险å˜åœ¨å¾ˆå¤§çš„ä¸ç¡®å®šæ€§ã€‚对于预期作用æŒä¹…或永久以åŠä¾µå…¥æ€§ç»™è¯ç‰é«˜é£Žé™©ç‰¹ç‚¹çš„细胞治疗产å“,在试验ä¸åº”选择预期治疗å¯èƒ½èŽ·ç›Šçš„患者。
选择患者作为å—试者时,应充分考虑患者疾病的严é‡ç¨‹åº¦å’Œç–¾ç—…çš„ä¸åŒé˜¶æ®µä»¥åŠçŽ°æœ‰æ²»ç–—手段,如果ä¸èƒ½èŽ·å¾—有效的治疗,特别是ä¸å¯æ²»æ„ˆæ€§ç–¾ç—…é‡åº¦è‡´æ®‹å’Œå±åŠç”Ÿå‘½æ—¶ï¼Œæ‚£è€…接å—ç»†èƒžæ²»ç–—ä¸´åºŠç ”ç©¶æ˜¯åˆç†çš„。åŒæ—¶åº”确定并å‡å°å—试者å¯èƒ½æ‰¿æ‹…的风险。
在å—试者的选择ä¸ï¼Œè¿˜åº”关注,如果患者将æ¥éœ€è¦é€šè¿‡ç»†èƒžã€ç»„织或器官移æ¤æ²»ç–—该或其他疾病,异体细胞治疗产å“诱导产生的抗体å¯èƒ½ä¼šå½±å“到移æ¤çš„æˆåŠŸçŽ‡ã€‚
å—试者选择å¯èƒ½ä¼šå½±å“临床试验的风险和获益,应尽å¯èƒ½å‡å°‘风险ã€æ高分æžç»“æžœçš„èƒ½åŠ›ï¼Œå¹¶å¢žåŠ å¯¹ä¸ªä½“å—试者和社会的获益。
对å—试者å¯èƒ½å¸¦æ¥çš„风险和获益应在知情åŒæ„书ä¸ç»™äºˆå……分表述。
(二)è¯æ•ˆå¦
å³ä½¿å—试的细胞治疗产å“的作用机制尚ä¸æ¸…晰,但对其主è¦çš„作用应有所了解。
在早期临床试验ä¸ï¼Œé€šå¸¸å…¶ä¸»è¦ç›®çš„是评价产å“的安全性,常è§çš„次è¦ç›®çš„则是åˆæ¥è¯„估产å“有效性,å³è¯æ•ˆå¦è¯„ä»·ã€‚è¯„ä¼°æŒ‡æ ‡ä¸ºå¯èƒ½æ示潜在有效性的çŸæœŸæ•ˆåº”或长期结局。这些概念验è¯æ•°æ®å¯ä»¥å¯¹åŽç»çš„临床开å‘æ供支æŒã€‚在细胞治疗产å“的活性评估ä¸ï¼Œå¯ä»¥åŒ…æ‹¬åŸºå› è¡¨è¾¾ã€ç»†èƒžæ¤å…¥ã€å½¢æ€å¦å˜åŒ–å’Œå…¶ä»–ç”Ÿç‰©æ ‡å¿—ç‰©ç‰ç‰¹æ®ŠæŒ‡æ ‡ï¼Œä¹Ÿå¯ä»¥åŒ…括å…疫功能å˜åŒ–ã€è‚¿ç˜¤ä½“积改å˜æˆ–å„ç§ç±»åž‹çš„生ç†åº”ç”ç‰æ›´å¸¸è§çš„æŒ‡æ ‡ä»¥åŠå› 技术å‘展å¯ä»¥æ£€æµ‹çš„æŒ‡æ ‡ã€‚
如果使用细胞治疗产å“çš„ç›®çš„æ˜¯çº æ£åŠŸèƒ½ç¼ºé™·æˆ–å—æŸçš„细胞或组织的生物å¦åŠŸèƒ½ï¼Œåˆ™åº”进行细胞治疗产å“功能å¦æ£€æµ‹ã€‚如果细胞治疗产å“的预期用途是修å¤ï¼å…疫调节ï¼æ›¿æ¢ç»†èƒžï¼ç»„织,并有望能够终生å‘挥功能,则相关的结构ï¼ç»„织å¦æ£€æµ‹æŒ‡æ ‡å¯ä½œä¸ºæ½œåœ¨çš„è¯æ•ˆå¦æ ‡å¿—物而进行检测,包括镜检ã€ç»„织å¦æ£€æµ‹ã€æˆåƒæŠ€æœ¯æˆ–é…¶æ´»æ€§æŒ‡æ ‡æ£€æµ‹ç‰ã€‚
当细胞治疗产å“包å«éžç»†èƒžæˆåˆ†æ—¶ï¼Œåº”对该产å“进行生物相容性ã€ä½“内é™è§£é€ŸçŽ‡å’Œç”Ÿç‰©å¦åŠŸèƒ½ç‰è¿›è¡Œç»¼åˆè¯„估。
(三)è¯ä»£åŠ¨åŠ›å¦
ä¼ ç»Ÿçš„è¯ä»£åŠ¨åŠ›å¦ç ”究方法并ä¸é€‚åˆäººçš„细胞治疗产å“çš„è¯ä»£åŠ¨åŠ›å¦ç ”究。应尽å¯èƒ½å¼€å±•ç»†èƒžæ²»ç–—产å“çš„ä½“å†…è¿‡ç¨‹ç ”ç©¶ã€‚ä¸´åºŠè¯•éªŒä¸åº”å¯¹ç ”ç©¶è¦æ±‚ã€å¯èƒ½é‡‡å–的方法åŠå¯è¡Œæ€§è¿›è¡Œè®¨è®ºï¼Œå¹¶æ³¨æ„在细胞治疗产å“预期的活性过程ä¸ï¼Œé‡ç‚¹æ£€æµ‹ç»†èƒžçš„活力ã€å¢žæ®–与分化能力ã€ä½“内的分布ï¼è¿ç§»å’Œç›¸å…³çš„生物å¦åŠŸèƒ½ã€‚
如果需对细胞治疗产å“进行多次(é‡å¤ï¼‰ç»™è¯ï¼Œä¸´åºŠæ–¹æ¡ˆè®¾è®¡æ—¶åº”考虑细胞治疗产å“在体内的预期å˜æ´»æ—¶é—´åŠç›¸åº”的功能。
(四)剂é‡æŽ¢ç´¢
早期临床试验的目的之一是探索细胞治疗产å“的有效剂é‡èŒƒå›´ã€‚如å¯èƒ½ï¼Œè¿˜åº”确定最大è€å—剂é‡ã€‚
应基于在产å“çš„è´¨é‡æŽ§åˆ¶ç ”究和临床å‰ç ”究ä¸æ‰€èŽ·å¾—的结果æ¥ç¡®å®šç»†èƒžæ²»ç–—产å“ç»™è¯å‰‚é‡ï¼Œå¹¶å……分考虑产å“的生物å¦æ•ˆåŠ›ã€‚
与å°åˆ†åè¯ç‰©ä¸åŒï¼Œç»†èƒžæ²»ç–—产å“的首次人体试验起始剂é‡ä¸€èˆ¬éš¾ä»¥ä»Žä¼ 统的éžä¸´åºŠè¯ä»£åŠ¨åŠ›å¦å’Œè¯æ•ˆå¦ä¸è¯„估确定,但其既往临床使用ç»éªŒï¼ˆå¦‚有)å¯èƒ½æœ‰åŠ©äºŽåˆç†åœ°ç¡®å®šä¸´åºŠèµ·å§‹å‰‚é‡ã€‚很多细胞治疗产å“会长期å˜åœ¨äºŽå—试者体内或作用时间æŒä¹…,所以首次人体试验应采用å•æ¬¡ç»™è¯æ–¹æ¡ˆï¼Œåªæœ‰åœ¨åˆæ¥äº†è§£äº§å“的毒性和作用æŒç»æ—¶é—´ä¹‹åŽï¼Œæ‰å¯è€ƒè™‘é‡å¤ç»™è¯ã€‚
细胞治疗产å“通常采用åŠå¯¹æ•°é€’增(100.5å€ï¼‰çš„方法æ¥åˆ¶å®šå‰‚é‡é€’增方案。给è¯å‰‚é‡å¢žå¹…的设定应该考虑到临床å‰æ•°æ®ä¸ä¸Žå‰‚é‡å˜åŒ–有关的风险和活性以åŠçŽ°æœ‰çš„任何临床数æ®ã€‚åŒæ—¶åº”充分考虑细胞治疗产å“特有的安全性风险,设定足够的给è¯é—´éš”å’Œéšè®¿æ—¶é—´ï¼Œä»¥è§‚察急性和亚急性ä¸è‰¯äº‹ä»¶ã€‚
尽管细胞治疗产å“çš„ç»™è¯å‰‚é‡å¯èƒ½å–决于患者的个体情况,然而早期临床试验所æ供的剂é‡æŽ¢ç´¢ç ”究的è¯æ®ä»ç„¶æ˜¯ç¡®è¯æ€§ä¸´åºŠè¯•éªŒä¸ç»™è¯å‰‚é‡ç¡®å®šçš„é‡è¦ä¾æ®ã€‚
(五)临床有效性
通常,临床有效性的确è¯æ€§è¯•éªŒåº”åœ¨ç›®æ ‡é€‚åº”ç—‡äººç¾¤ä¸å¼€å±•ï¼Œåº”æœ‰è¶³å¤Ÿæ ·æœ¬é‡ã€åˆç†çš„对照并选择具有临床æ„ä¹‰çš„ç»ˆç‚¹æŒ‡æ ‡ã€‚åŒæ—¶ï¼Œè¯¥ä¸´åºŠè¯•éªŒåº”能够æä¾›å¯äº§ç”Ÿé¢„期治疗效果的临床给è¯æ–¹æ¡ˆã€æ²»ç–—效果的
Palatin Technologies Inc. (NYSE: PT-141) announced today that it has shown positive results in a Phase I clinical trial of pT-141 for the treatment of sexual dysfunction in normal premenopausal women.
The trial included 32 healthy female volunteers and evaluated their response to pT-141 and placebo in response to visual stimuli. Pt-141 was found to be safe and resulted in a significantly greater improvement in vaginal blood flow than placebo users (P< 0.05).
role
Such antibiotics first affect the outer membrane of sensitive bacteria. Drug ring part of polypeptide amino and lipopolysaccharide in the outer membrane 2 valence cations combining site to produce electrostatic interaction, destroy the integrity of the outer membrane damage, fatty acid part of drugs to penetrate the outer membrane, thus increase the permeability of cell size, lead to intracytoplasmic phosphate, nucleosides and other small molecules to escape, straight die cause cell dysfunction. Gram-positive bacteria do not respond to such antibiotics because they have a thick cell wall that prevents drugs from entering the bacteria.
Pt141 Powder,Bodybuilding Ipamorelin,Sermorelin Powder,Weight Loss Peptide
Shaanxi YXchuang Biotechnology Co., Ltd , https://www.peptidenootropics.com