Heavy: The technical guidelines for the research and evaluation of cell therapy products (for trial implementation)

“A long-awaited start”, 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.

安全性相关的研究应根据细胞来源和制备工艺过程的特点考虑,可选择针对外源性因子、细胞恶性转化的可能性、成瘤性和致瘤性、相关杂质、病毒载体回复突变等方面开展研究。相关杂质研究中应包括工艺中引入的杂质(如蛋白酶、分化诱导试剂、病毒载体、微珠等)和产品相关的杂质(如细胞非预期表达的产物、死细胞残余和其他可能的生物降解产物等)。

2. QC

研究者需建立细胞治疗产品的质量控制策略。建议采用中间样品的质量检验和终产品放行检验相结合的机制。检定项目应当建立在产品质量研究以及对生产工艺和生产过程充分理解的基础之上,同时兼顾产品的特性和当下的科学认知与共识。随着研究的不断深入(如从临床前阶段进行至临床阶段),工艺相关信息应逐渐获得累积,检验方法应逐步完善,以适应各阶段的质量控制要求,建议确证性临床试验用样品的质量控制与商业化生产时的质控要求保持一致。质量控制一般应考虑鉴别、生物学效力、纯度、杂质、细胞数量(活细胞数、功能细胞数等)和一般检测(如无菌、支原体、内毒素、外观、除细胞之外的其他外源性异物等)等。验收标准的制订应以临床前研究批次、临床研究批次和验证批次中检测获得的数据,以及其他相关数据(如经验、文献报道和稳定性研究等)确定。

当放行检验受到时间限制时,可考虑加强工艺过程中的样品质量监控,将过程控制与放行检验相结合,通过过程控制简化放行检验。以上操作应经过研究与验证,并附有相应的操作规范。建议尽量在产品临床应用前完成全部放行检验,当有些放行检验结果可能后置时,应对可能出现的非预期检验结果制订处置方案。为对产品进行回顾性分析或进一步分析,建议研究者根据产品自身特点,并参照《药品生产管理规范》中的要求进行留样备查。

一些细胞治疗产品在给药前还需经过一系列操作步骤,研究开发时应开展模拟给药过程的相关研究。如果该操作在医疗机构开展,产品生产者应明确操作步骤和注意事项,以指导临床医护工作者正确操作使用。建议在给药前完成操作后对最终产品进行质量核准,如细胞形态、活细胞数及比率、颜色、除细胞之外的其他外源性异物等,以及操作步骤的复核和标签核对等。

放行检验用方法应经过研究与验证,特别是对于建立的新方法应进行全面的验证,对于药典中收录的方法应进行适用性的验证。对于有效期短和样本量小的产品,可采用快速、微量的新型检测方法。研究者应对新型检验方法与传统检测方法进行比较和评估,必要时,在产品放行检验时可以采用两种检验方法进行相互验证。

(五)稳定性研究

细胞治疗产品的生产建议采用连续的工艺,对于生产过程中需要临时保存的样品应进行稳定性研究,以支持其保存条件与存放期。细胞治疗产品稳定性研究的基本原则可参照一般生物制品稳定性研究的要求,并根据产品自身的特点、临床用药的需求,以及保存、包装和运输的情况设计合理的研究方案。应采用具有代表性的细胞样本和存储条件开展研究。其中,需要特别关注细胞治疗产品的运输稳定性研究和使用过程中的稳定性研究等,应开展研究证明在拟定的存储条件下,细胞治疗产品的质量不会受到运输、使用中或其他外界条件的影响。应根据产品自身的特点和存储条件等方面,合理地设计稳定性考察的项目和检测指标,例如,冷冻储存的样品或产品一般应模拟使用情形(如细胞复苏过程)开展必要的冻融研究。考察项目建议涵盖生物学效力、细胞纯度、细胞特性、活细胞数及比率、功能细胞数和安全性相关的内容等。

(六)容器和密闭系统

为避免由于存储而导致的产品质量发生非预期变化,研究者应对细胞治疗产品生产过程中的样品和/或成品保存用的包装容器和密闭系统进行安全性评估和相容性研究,以说明其使用的合理性,例如密封性研究、冷冻储存适应性研究等。

细胞治疗产品生产过程中可能涉及样品与容器短暂直接接触的步骤,如采集的组织或细胞、制备过程中的细胞、成品回输等步骤,研究者应对使用的容器进行安全性评估或接触的相容性研究等。

对于运输用的次级包装容器(非直接接触细胞)或材料也应经过验证,如对其遮光性、密封性和抗击机械压力等方面进行研究和验证。

五、非临床研究

(一)一般原则

1.研究评价内容

由于细胞治疗产品的物质组成及作用机制与小分子药物、大分子生物药物不同,所以传统、标准的非临床研究评价方法可能不完全适用于细胞治疗产品。细胞治疗产品的非临床研究评价内容取决于细胞类型及临床用途,与细胞来源、种类、生产过程、基因修饰/改造、处方中非细胞成分等因素密切相关,还与研发计划及相应的临床试验方案有关。由于细胞治疗产品种类繁多,不同产品其治疗原理、体内生物学行为、临床应用存在差别和不确定性,因此,对不同产品的研究评价应遵循“具体情况具体分析”的原则;同时,人用药品注册技术要求国际协调会(ICH)颁布的《生物技术药品的非临床安全性评价指南》(S6)可为细胞治疗产品的非临床研究评价提供参考。

2.受试物要求

非临床研究评价试验应尽可能使用拟用于临床试验的细胞治疗产品;用于进行非临床试验的受试物,其生产工艺及质量控制应与拟用于临床试验的受试物一致(如果不一致应给予说明,并评估其对预测人体反应的影响)。

(1)人源的细胞治疗受试物参考本指导原则的药学要求;

(2)如果由于相关动物选择的限制,可考虑使用动物源

替代品进行非临床研究评价;动物源替代品应与人源的细胞治疗受试物的生产工艺及质量标准尽可能相似,并提供必要的比较数据以确认替代品的质量属性。可考虑如下对比:

①组织或样本获取的程序;

②细胞识别、分离、扩增以及体外培养程序;

③细胞生长动力学参数(例如细胞倍增时间、细胞生长曲线、细胞增殖高峰时间);

④表型和功能特性(比如生长因子和细胞因子的分泌,细胞群体特异性表型/基因型标志);

⑤终产品配方/细胞支架种植方式(如果有);

⑥终产品的储存条件及细胞活力;

⑦动物替代细胞作用方式与终产品细胞作用方式的异同。

非临床试验受试物和临床用样品的异同均应在新药申报时予以说明。

3.动物种属选择

非临床研究评价应选择合适种属的动物进行试验,所选动物对细胞治疗产品的生物反应与预期人体反应接近或相似。动物模型选择需考虑以下几个方面:

(1)生理学和解剖学与人类具有可比性;

(2)对人体细胞产品或携带人类转基因的细胞产品的免疫耐受性;

(3)临床给药系统/流程的可行性,转运特定剂量的细胞到治疗靶点的可行性;

(4)免疫缺陷动物的适用性,对产品进行长期安全性评估的可行性。

免疫功能正常的动物给予人源细胞时可能出现免疫应答反应,此种情况下,可考虑采用以下模型开展非临床研究:

①给予免疫抑制剂于具有免疫能力的动物;

②遗传性免疫缺陷动物;

③人源化动物;

④在免疫豁免部位给药;

⑤或者以上形式的组合。

某些情况下,也可采用动物源替代品进行评价。

鉴于细胞治疗产品的特性,如产品效应持续时间长、在体内持续存在、在细胞治疗产品与疾病环境间复杂的作用机制、侵入性的给药途径等,也可考虑采用疾病动物模型进行非临床研究。

4.给药方式(途径)

非临床研究评价中,细胞治疗产品的给药方式应能最大程度模拟临床拟用给药方式。如果在动物试验中无法模拟临床给药方式,临床前研究中需明确替代的给药方式/方法,并阐明其科学性和合理性。

当使用特殊的给药装置给药时,非临床试验采用的给药装置系统应与临床一致。

5.受试物分析

应提供受试物分析数据。

细胞治疗产品在给药前可能还需经过一系列操作步骤,在完成操作后需对受试物进行质量检测,检测指标包括细胞形态、总活细胞数、细胞存活率、颜色、除细胞之外的其他外源性异物等。

(二)药效学研究

药效学研究应采用可靠的方法验证细胞治疗产品的基本治疗机理,确定生物学效应标志物。试验设计应考虑细胞治疗产品的作用机制、疾病周期长短以及给药方式等因素,结合细胞治疗产品的特性和存活时间。建议采用相关的体外和体内模型完成细胞治疗产品的药效学研究。

(三)药代动力学研究

药代动力学研究应能阐明细胞的体内过程以及伴随的生物学行为,应根据细胞治疗产品类型和特点选择合适的动物模型,一般考虑雌雄各半。根据研究目的及检测指标的临床价值,建立合适的生物分析方法并对方法进行必要的验证。药代动力学研究要关注目标细胞在体内的增殖、生物分子的表达和/或分泌,以及与宿主组织的相互作用;相互作用还包括细胞治疗产品的非细胞成分(辅料成分)及分泌的生物活性分子引起的相关组织反应。药代动力学研究内容包括但不仅限于以下方面:

1.细胞的分布、迁移、归巢

应采用一种或多种合适的细胞追踪方法评价细胞产品的分布、迁移、归巢及其存续和消亡特性,并阐述方法的科学性。细胞治疗产品的分布及存续时间是影响细胞治疗产品有效性和安全性的最重要因素,应进行动态观察,必要时观察直至这些细胞的消失或功能丧失。可选择的技术方法有影像技术、PCR技术、免疫组化技术等,试验设计需要考虑技术方法的适用性和优缺点。

2.细胞分化

细胞在分布、迁移和归巢后进一步分化为功能细胞发挥其治疗作用或功能衰退;对于细胞产品分化的程度及其后果(功能化或去功能化、安全参数),可应用体外方法和动物体内方法进行定量或定性评价研究。

3.对于经基因修饰/改造操作的人源细胞的特殊考虑

对于基因修饰/改造的细胞,除上述要求外,还需对目的基因的存在、表达、以及表达产物的生物学作用进行必要的研究,以体现基因修饰/改造的体内生物学效应。

(四)安全性研究评价

1. GLP遵从性要求

细胞治疗产品的安全性研究评价应遵从《药物非临床试验质量管理规范》(GLP)。对于某些在非GLP状况下开展的研究或检测,应予说明并评估非GLP对试验结果可靠性、完整性及对细胞治疗产品总体安全性评价的影响。

2.安全药理学试验

细胞在体内分泌的活性物质可能会对中枢神经系统、心血管系统、呼吸系统的功能等产生影响;细胞本身分布或植入于重要器官,细胞治疗产品的处方成分等也可能影响器官功能。因此,对于细胞治疗产品应考虑进行安全药理试验。如果在毒性试验中发现有潜在风险,应补充开展有关安全药理试验。

3.单次给药毒性试验

单次给药毒性试验可获得剂量与全身和/或局部毒性之间的剂量-反应关系,有助于了解其毒性靶器官,也可为重复给药毒性试验的剂量设计提供一定的参考。由于细胞治疗产品能够长时间地发挥功能或诱导长期效应,因此单次给药的观察时间应考虑细胞或者细胞效应的存续时间,一般应长于单次给药毒性试验常规的观察时间。

4.重复给药毒性试验

试验设计应包含常规毒理学试验研究的基本要素,并结合细胞治疗产品的特殊性来设计,以期获得尽可能多的安全性信息。

动物种属选择:根据产品的不同特性,采用能够对细胞治疗产品产生生物学活性的动物种属进行重复给药毒性研究。一般情况下应采用雌雄动物进行试验。如无相关种属可开展非临床研究时,非相关种属的动物试验对评价生产工艺过程、全处方的安全性及非靶效应也可能具有价值。

剂量组设计:参考“《药物重复给药毒性研究技术指导原则》”,需设计多个剂量水平、包含临床拟用剂量范围和最大可行剂量,并结合处方组成及生产工艺,设置合适的对照组。

观察指标:除常规观察指标外,需结合产品特点,选择合适的观察指标,尽可能包括形态学与功能学的评价指标,如行为学检测、神经功能测试、心功能评价、眼科检查、异常/异位增生性病变(如增生、肿瘤)、生物标志物、生物活性分子的分泌、免疫反应以及与宿主组织的相互作用等。

5.免疫原性和免疫毒性试验

细胞治疗产品或细胞分泌产物需要研究其潜在的免疫原性,免疫原性研究可参考最新版技术研究指导原则;此外,还需关注细胞治疗产品诱导产生的免疫毒性。

6.致瘤性/致癌性试验

细胞治疗产品的致瘤性/致癌性风险取决于产品中不同细胞的分化状态、生产过程中采用的细胞培养方式引起的生长动力学改变、基因修饰/改造细胞的转基因表达(例如多种生长因子)、诱导或增强宿主体内形成肿瘤的可能性以及目标患者人群等,需要根据以上特点进行综合考虑。

目前,如何选择致瘤性/致癌性研究的动物模型尚未达成科学共识。致瘤性/致癌性试验应采用临床拟用产品进行试验。致瘤性/致癌性试验需确保细胞可在体内长期存活以考察是否有潜在肿瘤形成。试验设计需注意以下方面:(1)合适的对照组(例如阳性对照、空白对照);(2)每组需有足够的动物数量,使肿瘤发生率的分析满足统计学要求;(3)需包含最大可行剂量;(4)受试物应到达拟定的临床治疗部位;(5)足够长的试验周期。

由于免疫排斥反应,人源细胞治疗产品的致瘤性/致癌性试验可考虑使用免疫缺陷的啮齿类动物模型进行。

7.生殖毒性试验

细胞治疗产品的生殖和发育毒性评价主要是取决于产品的特性、临床适应症以及临床拟用人群,应根据具体情况具体分析。

8.遗传毒性试验

对于人源的细胞治疗产品,如果该产品与DNA或其他遗传物质存在直接的相互作用,需进行遗传毒性试验。

9.特殊安全性试验

根据细胞治疗产品的特点与临床应用情况,应考虑对局部耐受性、组织兼容性及对所分泌物质的耐受性进行评估。

10.其他毒性试验

对于采用基因修饰/改造的细胞治疗产品,需关注有复制能力的病毒的产生和插入突变,特别是致癌基因的活化等特性带来的安全性风险。具体要求可参见相关的技术指导文件。

六、临床研究

当细胞治疗产品进入临床试验阶段时,应遵循《药物临床试验质量管理规范》(GCP)的要求。临床试验的研究内容原则上应包括临床安全性评价、药代动力学研究、药效学研究、剂量探索研究和确证性临床试验等。根据不同细胞治疗产品的产品性质,可酌情调整具体的试验设计。

鉴于细胞治疗产品特殊的生物学特性,在临床试验研究中,需要采取不同于其他药物的临床试验整体策略。为了获得预期治疗效果,细胞治疗产品可能需要通过特定的手术措施、给药方法或联合治疗策略来进行给药。因此,在临床试验方案设计中应一并考虑。

细胞治疗产品很多特有的性质也会影响其临床试验的设计,包括产品特性、生产特点以及临床前研究的结果等。

(一)受试者的保护

受试者的选择需要考虑多方面的因素,选择宗旨是能使研究受试者的预期风险与潜在获益经过慎重评估,同时能实现研究的科学目的。

在早期临床试验阶段,所预期的获益或风险存在很大的不确定性。对于预期作用持久或永久以及侵入性给药等高风险特点的细胞治疗产品,在试验中应选择预期治疗可能获益的患者。

选择患者作为受试者时,应充分考虑患者疾病的严重程度和疾病的不同阶段以及现有治疗手段,如果不能获得有效的治疗,特别是不可治愈性疾病重度致残和危及生命时,患者接受细胞治疗临床研究是合理的。同时应确定并减小受试者可能承担的风险。

在受试者的选择中,还应关注,如果患者将来需要通过细胞、组织或器官移植治疗该或其他疾病,异体细胞治疗产品诱导产生的抗体可能会影响到移植的成功率。

受试者选择可能会影响临床试验的风险和获益,应尽可能减少风险、提高分析结果的能力,并增加对个体受试者和社会的获益。

对受试者可能带来的风险和获益应在知情同意书中给予充分表述。

(二)药效学

即使受试的细胞治疗产品的作用机制尚不清晰,但对其主要的作用应有所了解。

在早期临床试验中,通常其主要目的是评价产品的安全性,常见的次要目的则是初步评估产品有效性,即药效学评价。评估指标为可能提示潜在有效性的短期效应或长期结局。这些概念验证数据可以对后续的临床开发提供支持。在细胞治疗产品的活性评估中,可以包括基因表达、细胞植入、形态学变化和其他生物标志物等特殊指标,也可以包括免疫功能变化、肿瘤体积改变或各种类型的生理应答等更常见的指标以及因技术发展可以检测的指标。

如果使用细胞治疗产品的目的是纠正功能缺陷或受损的细胞或组织的生物学功能,则应进行细胞治疗产品功能学检测。如果细胞治疗产品的预期用途是修复/免疫调节/替换细胞/组织,并有望能够终生发挥功能,则相关的结构/组织学检测指标可作为潜在的药效学标志物而进行检测,包括镜检、组织学检测、成像技术或酶活性指标检测等。

当细胞治疗产品包含非细胞成分时,应对该产品进行生物相容性、体内降解速率和生物学功能等进行综合评估。

(三)药代动力学

传统的药代动力学研究方法并不适合人的细胞治疗产品的药代动力学研究。应尽可能开展细胞治疗产品的体内过程研究。临床试验中应对研究要求、可能采取的方法及可行性进行讨论,并注意在细胞治疗产品预期的活性过程中,重点检测细胞的活力、增殖与分化能力、体内的分布/迁移和相关的生物学功能。

如果需对细胞治疗产品进行多次(重复)给药,临床方案设计时应考虑细胞治疗产品在体内的预期存活时间及相应的功能。

(四)剂量探索

早期临床试验的目的之一是探索细胞治疗产品的有效剂量范围。如可能,还应确定最大耐受剂量。

应基于在产品的质量控制研究和临床前研究中所获得的结果来确定细胞治疗产品给药剂量,并充分考虑产品的生物学效力。

与小分子药物不同,细胞治疗产品的首次人体试验起始剂量一般难以从传统的非临床药代动力学和药效学中评估确定,但其既往临床使用经验(如有)可能有助于合理地确定临床起始剂量。很多细胞治疗产品会长期存在于受试者体内或作用时间持久,所以首次人体试验应采用单次给药方案,只有在初步了解产品的毒性和作用持续时间之后,才可考虑重复给药。

细胞治疗产品通常采用半对数递增(100.5倍)的方法来制定剂量递增方案。给药剂量增幅的设定应该考虑到临床前数据中与剂量变化有关的风险和活性以及现有的任何临床数据。同时应充分考虑细胞治疗产品特有的安全性风险,设定足够的给药间隔和随访时间,以观察急性和亚急性不良事件。

尽管细胞治疗产品的给药剂量可能取决于患者的个体情况,然而早期临床试验所提供的剂量探索研究的证据仍然是确证性临床试验中给药剂量确定的重要依据。

(五)临床有效性

通常,临床有效性的确证性试验应在目标适应症人群中开展,应有足够样本量、合理的对照并选择具有临床意义的终点指标。同时,该临床试验应能够提供可产生预期治疗效果的临床给药方案、治疗效果的

PT141 Powder

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

Posted on