Science Pool

ICH M12 2024 vs Japanese PMDA 2018 DDI Guidelines. Navigating the Changes.

Posted by Evotec on Jul 5, 2024 12:00:29 PM

DOWNLOAD OUR HANDY COMPARISON SUMMARY

The new ICH M12 guideline harmonises drug interaction guidance from the major regulatory authorities. It is expected that the ICH M12 guideline will be implemented by the Japanese Pharmaceuticals and Medical Devices Agency (PMDA), the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) and presumably replace their existing guidelines/guidance. In this blog, we explore the differences in the in vitro studies between the new ICH M12 guideline adopted in May 2024 and the Japanese PMDA guideline from 2018. You may also be interested in our blogs comparing with the US FDA 2020 guidance (LEARN MORE), and the EMA 2013 guideline (LEARN MORE) to the ICH M12 guideline.

Reaction Phenotyping

The new ICH M12 and the PMDA guidelines are similar in terms of their recommendations. Both have the same cut-off of ≥25% of total elimination for determining if the enzyme needs further investigation in a clinical study. The ICH M12 suggest investigating a larger range of phase II enzymes if the investigational drug is not metabolised by the main CYP enzymes whereas the Japanese PMDA only specifically name UGT enzymes for phase II.

Enzyme Inhibition

For enzyme inhibition, both the ICH M12 and PMDA guidelines recommend evaluating the main seven CYP isoforms for enzyme inhibition as well as UGT inhibition if one of the major elimination pathways of the investigational drug is direct glucuronidation. However, the ICH M12 suggests evaluating a larger panel of UGT isoforms (UGT1A1, UGT1A4, UGT1A9, UGT2B7 and UGT2B15) compared to the PMDA guideline which suggests only UGT1A1 and UGT2B7 inhibition.

For reversible inhibition, the cut-off for determining if a clinical study is required is the same in both guidelines using the basic model. However, for time dependent inhibition, 5x Cmax,u is used in the calculation in the ICH M12 whereas 50x Cmax,u was used in the 2018 PMDA guideline, suggesting a less conservative approach is now being used in the new ICH M12 guideline. Interestingly, the PMDA guideline also recommended a different cut-off for CYP3A in the GI tract for time dependent inhibition – this is not covered in the new ICH M12 guideline.

Enzyme Induction

The ICH M12 and the PMDA guidelines are similar in terms of CYP induction. The equation for the relative induction score is the same in both guidelines. Similarly, the basic kinetic model cut-offs are the same for both the ICH M12 and PMDA guidelines.

Transporter Substrate Identification

Both guidelines recommend that the same transporters are assessed. The method for testing and cut-offs for clinical assessment are very similar between the PMDA and ICH M12 guidelines.

Transporter Inhibition

For transporter inhibition, the ICH M12 and PMDA guidelines are the same in terms of the cut-offs. The only omission from the PMDA is that it only considers the oral route for P-gp and BCRP inhibition whereas the ICH M12 also considers the parenteral route for these transporters.

DOWNLOAD OUR COMPARISON SUMMARY

Get in touch

Tags: Blog, ADME/DMPK, IND Enabling Studies/Preclinical Development

ICH M12 2024 vs EMA 2013 DDI Guidance. What’s New?

Posted by Evotec on Jul 4, 2024 2:13:31 PM

DOWNLOAD OUR HANDY COMPARISON SUMMARY

The new ICH M12 guideline on drug interaction studies provides a harmonised approach which is expected to be implemented by the major regulatory authorities including the European Medicines Agency (EMA), the US Food and Drug Administration (FDA), and the Japanese Pharmaceuticals and Medical Devices Agency (PMDA), and presumably replace their respective existing guidelines/guidance. In this blog, we explore the differences between the new ICH M12 guideline adopted in 2024 and the previous EMA guideline which came into effect in 2013. You may also be interested in our blog comparing the US FDA 2020 guidance to the ICH M12 2024 harmonised guideline (LEARN MORE). Our comparison of the Japanese PMDA 2018 guideline with the ICH M12 guideline will follow shortly.

Reaction Phenotyping

The new M12 ICH and the EMA guidelines are similar in terms of their recommendations. Both the ICH M12 and the EMA guideline have the same cut-off of ≥25% of total elimination for determining if the enzyme needs further investigation in a clinical study.

Enzyme Inhibition

For enzyme inhibition, both the ICH M12 and EMA 2013 guidelines recommend evaluating the main seven CYP isoforms for enzyme inhibition. For reversible inhibition, the cut-off for determining if a clinical study is required is the same in both guidelines using the basic model. However, for time dependent inhibition, 5x Cmax is used in the calculation in the ICH M12 whereas 1x Cmax was used in the EMA 2013 guideline, suggesting a more conservative approach is now being used in the new ICH M12 guidance. Furthermore, in the ICH M12, only a single cut-off is provided for time dependent inhibition whereas the EMA has cut-offs for intestinal enzymes for orally administered drugs as well as systemic enzymes. The EMA currently suggest drug interactions in the GI tract will be addressed with accompanying EMA documentation.

Both guidelines suggest evaluating UGT inhibition if one of the major elimination pathways of the investigational drug is direct glucuronidation, however, the ICH M12 references a larger panel of UGT isoforms including UGT1A1, UGT1A4, UGT1A9, UGT2B7 and UGT2B15 whereas the EMA 2013 references only UGT1A1 and UGT2B7.

Enzyme Induction

The ICH M12 and the EMA 2013 guidelines are similar in terms of CYP induction. The cut-offs for the basic fold-change method and the relative induction score (RIS) are the same. For the correlation method, the ICH M12 guideline gives better clarity on the cut-off value to be used compared to the EMA guideline. The ICH M12 provides clearer guidance on how to interpret the basic kinetic model whereas the EMA 2013 guideline incorporates the model into a mechanistic static equation.

Transporter Substrate Identification

Both guidelines recommend the same transporters are assessed. The method for testing and cut-offs for clinical assessment are very similar between the ICH M12 and EMA guidelines. However, once again the ICH M12 provides more clarity on interpretation of the results especially in the case of the uptake transporters.

Transporter Inhibition

For transporter inhibition, the EMA 2013 guideline recommends screening for OCT1 and BSEP inhibition in addition to the standard transporters recommended by the ICH M12. Although these transporters are not in the standard list for the ICH M12 guideline, it is suggested they may be assessed on a case-by-case basis with other transporters such as OATP2B1 and MRP2. The cut-off values also differ between the two guidelines with the EMA 2013 guideline tending to be more conservative for certain transporters. One final difference is that the ICH M12 recommend a pre-incubation with test article for transporters such as OATP1B1 and OATP1B3 whereas the EMA 2013 guideline does not refer to a pre-incubation as the scientific literature and consensus concerning this topic only started to appear later around 2017.

DOWNLOAD OUR COMPARISON SUMMARY

Get in touch

Tags: Blog, ADME/DMPK, IND Enabling Studies/Preclinical Development

Unlocking the Future of Drug Discovery & Development with Evotec’s Clinical Proteomics

Posted by Evotec on Jul 3, 2024 5:20:35 PM

In the ever-evolving landscape of biomedical research, proteomics has emerged as a crucial field for advancing drug discovery and development and biomarker discovery. Evotec stands at the forefront of this scientific frontier, offering cutting-edge mass spectrometry-based proteomics solutions. Here’s how Evotec’s innovative technologies and comprehensive services deliver exceptional value to customers, driving breakthroughs in clinical research, such as discovery of stratification biomarkers and diagnostic markers for the discovery of novel targets.

Revolutionizing Proteomics with Advanced Platforms

ScreenPep™ Platform: Evotec’s ScreenPep™ platform is a high-throughput, automated system designed for deep-coverage proteomics of cell lines, tissues or biofluids. When applied to plasma or serum it can identify up to 1,500 proteins , making it incredibly efficient and cost-effective. This platform is ideal for large-scale studies, in which sample quantity may be limited but extensive protein identification is required.

Proteograph™ Technology: As the only Center of Excellence in Europe offering Proteograph™ technology, Evotec provides an unparalleled capability to identify up to 5,000 proteins from human biofluids such as serum, plasma, and cerebrospinal fluid (CSF). This technology leverages nanoparticle-based methods to achieve deep proteome coverage, significantly enhancing the scope of biomarker discovery and translational research.

Integrated MultiOmics Solutions: Evotec excels in integrating proteomics data with genomics, transcriptomics, and metabolomics, offering a holistic approach to biomarker discovery. This integrated solution, facilitated by Evotec’s PanHunter platform, allows for comprehensive data analysis and interpretation, enabling researchers to uncover complex biological insights and accelerate drug development processes.

Customized and Scalable Services: Understanding that each research project has unique requirements, Evotec offers highly customized services tailored to meet specific client needs. From optimizing study designs to adapting sample preparation workflows for difficult and rare samples, Evotec ensures that every project receives the precise attention and expertise it demands.

Exceptional Expertise and Infrastructure: With over 20 years of experience and more than 50 mass spectrometers across its sites, Evotec is one of the largest providers of proteomics services worldwide. Their dedicated team of bioinformaticians and proteomics experts continuously develops and refines in-house pipelines, ensuring the highest standards in data quality and analysis.

High-End Mass Spectrometry: Utilizing cutting-edge mass spectrometry instruments, Evotec guarantees high-sensitivity and high-precision proteomics. These advanced technologies are crucial for detecting low-abundance proteins and analyzing post-translational modifications, providing deeper insights into protein functions and interactions.

Advantages of Mass Spectrometry-Based Proteomics

Unbiased Protein Measurement: Unlike antibody or aptamer-based techniques, mass spectrometry provides an unbiased measurement of all proteins containing tryptic peptides. This comprehensive approach allows for a more accurate and complete proteome analysis, essential for discovering new biomarkers and therapeutic targets. The quantification is also not impacted by any conformational changes.

Versatility Across Species: Evotec’s proteomics solutions are not limited to human samples; they are also applicable to various species, including animals used in health studies. This versatility broadens the research applications and facilitates translational research from preclinical models to clinical settings.

Detection of Protein Isoforms and Modifications: Mass spectrometry is uniquely capable of detecting protein isoforms and post-translational modifications, such as phosphorylation and ubiquitination. This capability is vital for understanding protein functions and regulatory mechanisms, paving the way for novel therapeutic strategies.

Cutting-Edge Technology and Continuous Innovation: Evotec’s commitment to innovation is exemplified by their continuous development of nanoparticle-based proteomics in collaboration with Seer Inc. This ongoing enhancement ensures that Evotec remains at the forefront of proteomics research, providing customers with the most advanced and reliable technologies available.

Conclusion

Evotec’s clinical proteomics capabilities offer a robust, scalable, and precise solution for drug development and biomarker discovery. By leveraging advanced technologies, integrated MultiOmics approaches, and customized services, Evotec empowers researchers to make significant scientific advancements. Partner with Evotec to unlock new possibilities in your research and drive the future of healthcare innovation.

For more information, please contact us at info@evotec.com or visit our website at PanOmics - Technology Platforms - Evotec Website (English)

LEARN MORE

Tags: Blog, Proteomics, Metabolomics & Biomarkers

Green Chemistry at Evotec: Pioneering Sustainable Drug Discovery

Posted by Evotec on Jul 2, 2024 4:34:16 PM

As the world increasingly turns its focus toward sustainability, the pharmaceutical industry is not exempt from this shift. One company leading the charge is Evotec, a global drug discovery and development biotech who offer CRO and CDMO services. Evotec’s commitment to green chemistry is setting new standards for sustainability in medicinal chemistry, particularly in how drug discovery processes are designed and executed. This blog post will delve into Evotec’s green chemistry initiatives and how they are revolutionizing the pharmaceutical industry.

Evotec’s Commitment to Green Chemistry

Evotec’s green chemistry philosophy centres around minimizing the environmental impact of their chemical processes while maintaining excellence in drug discovery. This commitment is evident in their comprehensive strategy that encompasses various innovative approaches and technologies designed to make their operations more sustainable.

Key Initiatives in Green Chemistry

  1. Safer Solvents and Reduced Plastic Waste
    • Evotec prioritizes the use of safer solvents, particularly those sourced from renewable bio-based origins, to reduce the reliance on chlorinated solvents known for their environmental and health hazards.
    • They actively work on reducing plastic waste by encouraging the reuse and recycling of plastic consumables in their laboratories.
  2. Energy Efficiency
    • Energy conservation is a significant aspect of Evotec’s sustainability efforts. Simple yet effective practices like shutting laboratory fume cupboards when not in use help in reducing unnecessary energy consumption.
  3. Raising Awareness and Continuous Improvement
    • Evotec fosters a culture of sustainability by raising awareness among their scientists about green chemistry alternatives through communication and poster sessions.
    • Continuous improvement of chemical processes with a sustainable vision is a cornerstone of their green chemistry initiatives.

Advanced Green Chemistry Techniques

Evotec employs several advanced green chemistry techniques that underscore their innovative approach to sustainable drug discovery:

  1. Micellar Chemistry
    • Utilizing micellar properties for chemical reactions offers a new paradigm in sustainable chemistry, reducing the need for harmful organic solvents.
  2. Mechanochemistry
    • This technique uses mechanical forces to drive chemical reactions, which can be more sustainable than traditional methods reliant on solvents and reagents.
  3. Flow Chemistry
    • Flow chemistry enhances productivity and safety, offering better control over reaction conditions and significantly reducing organic waste.
  4. Biocatalysis
    • By employing enzymatic processes, Evotec can replace traditional organic synthesis routes, reducing the use of toxic reagents and conditions, whilst often also increasing yields.

Practical Applications and Case Studies

Evotec has documented numerous case studies showcasing the practical application of green chemistry in their labs. These include:

  1. Solvent Replacement
    • Evotec has successfully replaced conventional hazardous solvents like DMF, DCM, and THF with greener alternatives such as Me-THF, CPME and dimethyl isosorbide (DMI), achieving comparable or superior yields in various reactions while reducing toxicity and environmental impact.
  2. Catalyst Optimization
    • By optimizing catalyst loading in reactions, such as pallado-catalyzed cross-couplings, the company has significantly reduced the amount of precious metals required, decreasing both environmental impact and costs.
  3. Sustainable Work-Up Methods
    • The adoption of FastWoRX™, a technique that significantly reduces the amount of solvent used in the work-up phase, demonstrates Evotec’s commitment to minimizing waste.
  4. Better purification processes
    • Rethinking our working habits in a smarter and sustainable way. Avoid systematic normal phase flash column chromatography (or reuse, if necessary). Favour reverse phase and crystallization/recrystallization.

The Future of Green Chemistry at Evotec

Evotec’s approach to green chemistry is not static; it is an evolving process that aims to continually integrate new technologies and methodologies. The company's dedication to sustainability is reflected in their global initiatives and partnerships, fostering a greener pharmaceutical industry.

In conclusion, Evotec is at the forefront of green chemistry in drug discovery. Their innovative approaches and commitment to sustainability set a benchmark for the industry, proving that excellence in drug discovery and environmental stewardship can go hand in hand. As Evotec continues to develop and implement green chemistry practices, they pave the way for a more sustainable future in medicinal chemistry.

LEARN MORE

Tags: Computational Chemistry, Medicinal Chemistry, Blog

Advancing Immunotherapy through Immunopeptidome Deciphering for Neoantigen Identification

Posted by Evotec on Jul 2, 2024 3:31:10 PM

In the quest to revolutionize immunotherapy, the identification of naturally presented peptides bound to HLA class I and II molecules from cancerous cells holds paramount importance. These peptides, crucial for developing immunotherapy-based treatments, also offer promising avenues for combating infectious diseases. Evotec's pioneering immunopeptidomics platform stands at the forefront, enabling the unbiased discovery of novel immunotherapeutic targets.

This comprehensive approach extends beyond cancer research, facilitating the identification of diagnostic and monitoring biomarker signatures across normal and altered cells in cohort studies. It sheds light on the intricate interplay between T cells and MHC-presenting cells, deepening our understanding of immunobiology.

Evotec's meticulously crafted experimental strategy, coupled with its state-of-the-art capabilities in high-end quantitative mass spectrometry, achieves unparalleled sensitivity. This precision is essential for distinguishing disease-specific neoantigens from their normally presented counterparts. By integrating whole exome sequencing and transcriptomics data, the platform empowers the discovery of neoepitopes, while advanced statistics and bioinformatics tools enable comprehensive data analysis and interpretation, facilitating peptide prioritization.

The platform's capabilities are exemplified by its ability to identify up to a thousand peptides per sample, providing direct detection of presented peptides, surpassing computation-intensive in silico predictions. Moreover, validation and accurate quantification of individual peptides are ensured through targeted mass spectrometry (PRM-MS).

In essence, Evotec's immunopeptidomics platform represents a transformative leap in the field, offering unparalleled insights into immunobiology and neoantigen identification, with profound implications for the development of immunotherapy-based treatments and the fight against various diseases.

For further inquiries, contact Evotec's experts at info@evotec.com or learn more here 

LEARN MORE

Tags: Blog, Proteomics, Metabolomics & Biomarkers

ICH M12 2024 vs FDA 2020 DDI Guidance - Exploring the Differences

Posted by Cyprotex on Jun 19, 2024 6:18:50 PM

ICH M12 2024 vs FDA 2020 DDI Guidance. Exploring the Differences.

Download our handy comparison summary

The new ICH M12 guideline on drug interaction studies is welcomed by industry as it provides a harmonized approach which is expected to be implemented by the major regulatory authorities such as the US Food and Drug Administration (FDA), European Medicines Agency (EMA) and the Japanese Pharmaceuticals and Medical Devices Agency (PMDA) and presumably replace their respective existing guidance. In this blog, we explore the differences between the now finalized ICH M12 guideline adopted in May 2024 (draft for comment released 2022) and the previous US FDA 2020 guidance released in January 2020 with a focus on the in vitro assays.

Before we launch into the individual assays, one general observation in the new ICH M12 guideline is the replacement of the term ‘victim’ to ‘object’ to denote an investigational drug (usually a substrate) which is affected by a concomitant drug, and the replacement of the term ‘perpetrator’ to ‘precipitant’ to denote an investigational drug (typically an inhibitor or inducer) which affects a concomitant drug. This amendment only appeared in the final version of the ICH M12 guideline as it was one of the comments from review of the draft version.

Reaction Phenotyping

If we consider reaction phenotyping initially, the differences between the new ICH M12 and the FDA 2020 guidance are relatively minimal. Although the ICH M12 refers to all the enzymes mentioned in the FDA guidance, it also refers to some additional Phase II enzymes which may need to be evaluated including glutathione S-transferases (GSTs) and N-acetyl transferases. Both the ICH M12 and the FDA guidance have the same cut-off of ≥25% of total elimination identified using in vitro studies (reaction phenotyping and metabolite identification) and human mass balance data for determining if the enzyme needs further investigation in a clinical study. One key difference is that the FDA guidance recommends two methods should be used for reaction phenotyping – the first method using selective inhibitors in the presence of human liver microsome or hepatocytes and the second method using human recombinant enzymes. In the ICH M12 guidelines, only one of these methods is required.

Both the ICH M12 guideline and the FDA guidance have recommendations for evaluating metabolites in reaction phenotyping studies if the exposure levels of the metabolite results in clinically relevant changes in efficacy or safety.

Enzyme Inhibition

Moving onto enzyme inhibition, both the ICH M12 and FDA guidance recommend evaluating the main seven CYP isoforms for reversible and time dependent inhibition. For reversible inhibition, the cut-off for determining if a clinical study is required is the same for both the ICH M12 and FDA. However, for time dependent inhibition, 5 x Cmax,u is used in the calculation in the ICH M12 whereas a higher safety factor of 50 x Cmax,u was used in the 2020 FDA guidance, suggesting a less conservative approach is used in the new ICH M12 guideline.

One addition to the ICH M12 guideline, which is not specifically included in the previous FDA 2020 guidance, is the inclusion of reversible UGT inhibition. The ICH M12 recommends that UGT1A1, UGT1A4, UGT1A9, UGT2B7 and UGT2B15 inhibition should be evaluated if direct glucuronidation is one of the major elimination pathways of the investigational drug. Although it is considered an area of ongoing research, it is currently recommended in the ICH M12 that the same cut-off values should be used for UGT inhibition that is applied to CYP enzymes (i.e., Cmax,u/Ki,u<0.02).

Enzyme Induction

For enzyme induction, the FDA guidance suggested either catalytic activity or mRNA could be used to assess induction. It also suggested that immortalized hepatic cell lines may be used to determine CYP induction potential. However, the new ICH M12 primarily recommends the analysis of mRNA (with the exception of CYP2C19 where catalytic activity should be measured) and that the CYP induction assessment should be performed in human hepatocytes. The ICH M12 guideline also addresses the issue of toxicity and recommends that cell viability assessment is performed before and at the end of the incubation.

For data analysis, both the ICH M12 and FDA cover the three basic methods (fold-change, relative induction score correlation method and basic kinetic model) and align on the cut-offs for indicating if an investigational drug has the potential to induce in vivo. However, for the basic mRNA fold method in the ICH M12 guideline describes assessing test drug concentration of 50 x Cmax,u whereas the FDA only suggests testing up to 30 x Cmax,u suggesting a more conservative approach now by the ICH M12. Furthermore, in the calculations for the correlation methods and the basic kinetic model, unbound EC50 (EC50,u) is specified in the ICH M12 whereas only EC50 is referred to in the FDA guidance. Finally, the ICH M12 describes in more detail an indicative positive control response for CYP1A2, CYP2B6 and CYP3A4 of typically at least 6-fold to ensure sufficient sensitivity of system, whereas this level of detail is absent from FDA.

Transporter Substrate Identification

Both the ICH M12 guideline and the FDA DDI guidance (2020) recommend the same transporters are assessed. The method for testing and thresholds for clinical assessment are very similar between the FDA and ICH M12.

Transporter Inhibition

For transporter inhibition, the same transporters are recommended in the ICH M12 as the previous FDA guidance, however, the cut-off value for determining if a clinical study should be performed are different in a couple of instances. Firstly, for P-gp and BCRP inhibition where the investigative drug is administered by the parenteral route or if a metabolite is formed post absorption, the cut-off value in ICH M12 is an IC50 of 50 x Cmax,u, whereas in the FDA guidance it is 10x Cmax total; important to note the additional difference for what is defined as [I]. This indicates a more conservative approach by the FDA when plasma protein binding is > 80%, or by ICH M12 when plasma protein binding is < 79%. The second instance applies to the cut-off value for MATE1 and MATE2-K, which has increased to an IC50 of 50 x Cmax,u rather than 10x Cmax,u, indicating a more conservative approach by the ICH M12 for these renal transporters. The ICH M12 also specifically refers to unbound IC50 (IC50,u) in the calculations whereas the FDA guidance only references IC50. In this regard, it is important to recognize that any correction of IC50 for potential non-specific binding that might occur within a transporter inhibition assay would only be required if the assay did not incorporate a pre‑incubation step with investigational drug as standard methodology for all transporters prior to the co‑incubation with fresh investigational drug solution and probe substrate. With such standard methodologies, the inclusion of the pre‑incubation step would be anticipated to mask any non-specific binding sites therefore the co‑incubated concentrations of investigational drug would be nominal for IC50 fitting purposes, i.e. IC50 = IC50,u.

DOWNLOAD OUR COMPARISON SUMMARY

Get in touch

Tags: Blog, ADME/DMPK, IND Enabling Studies/Preclinical Development

Commercial Active Pharmaceutical Ingredient manufacturing challenges

Posted by Evotec on Jun 19, 2024 5:15:46 PM

The top challenges in commercial API manufacturing

Once an investigational medicinal product has demonstrated its safety and efficacy during clinical trials, the next challenge for its Sponsor is to secure a reliable supply chain for its commercial production. Understandably, commercial pharmaceutical manufacturing, including commercial API manufacturing, is a highly regulated area to ensure patients’ ongoing access to a consistently high quality of medicines that guarantees their safety and efficacy. Transitioning from a clinical pilot scale to eventually the larger plant scale required for commercial API production can be a long and arduous process.

The main challenges faced in the journey to commercialization include scaling up production, establishing and maintaining process consistency and reproducibility, and ensuring compliance with regulatory guidelines. In this blog, we explore these areas in detail, discussing how a strategic approach, robust planning, and the incorporation of innovative technologies, can help ensure your drug smoothly and successfully reaches the market.

Developing robust and scalable processes

Increasing the production of active pharmaceutical ingredients (APIs) and drug excipients from milligram to multi-kilogram quantities is arguably the biggest challenge faced in paving the way to reliable commercial pharmaceutical manufacturing. Without the adoption of strategic planning during the process development stage, unexpected issues may arise and lead to significant increases in your program timelines and costs.

Here are our top tips for effectively planning and developing robust and scalable API manufacturing processes, and achieving faster and more seamless scale-up that supports marketing approval and supply security requirements:

  • Choice of starting materials – several considerations should be made when selecting the starting materials used in the manufacture of APIs and medicinal products. This includes quality, compatibility, long-term stability, affordability, safety, security of supply, and compliance with pharmaceutical regulatory guidelines. Ensuring your starting materials meet these criteria throughout the entire scale-up process, up to commercial scale, will pave the way for a safe and high-quality medicinal product.
  • Choice of equipment and processing technology – where possible, scalable equipment and processing technology should be chosen in API and drug manufacturing. This will help to streamline the technology transfer process, reducing the time and costs required for process optimization and validation, and minimizing the risk of failure. Cost, availability, material compatibility, cleaning processes, and the ability to implement automation and control systems at commercial scale should be considered.
  • Process development – the basis of every robust, scalable process is effective API process development. This requires the optimization of several components, including the selection of the synthetic route, and measures for process control and risk management. Scientific and risk-based approaches should be utilized to help gain an in-depth understanding of how any changes to the process during scale-up will affect the safety and quality of the product.

Read this blog for more detail on how to optimize API process development

 

Establishing and maintaining process consistency and reproducibility

Establishing and maintaining consistency and reproducibility during scale-up is a complex yet vital task in commercial pharmaceutical manufacturing. Increasing the vessel size and introducing any other necessary changes can drastically alter process performance and product quality. Failure to design and optimize the process to account for these differences could derail production, altering the safety, quality, and efficacy of the API.

The key to ensuring process consistency and reproducibility in commercial scale production lies in strategic planning during process development, with the use of systematic approaches that facilitate the implementation of a robust process control strategy

Implementation of systematic approaches

Several systematic approaches, such as design of experiments (DoE), quality by design (QbD), and process modeling and simulations performed with a software such as Dynochem® are used to improve knowledge of the product and the scale-up process. This includes the thorough impact assessment of any changes in processing parameters on process performance and final product quality.

DoE involves the use of factorial design to plan a series of experiments that test simultaneously variations in individual input factors. The aim of this is to gain an understanding of the individual and combined effects of parameters variability on the process performance and output. Following these experiments, mathematical models, such as response surface methodology (RSM), are used to identify the optimal set of process parameters as well as the acceptable operating envelope for the process.

DoE and process modeling and simulations can be integrated into a QbD approach, which incorporates statistical and analytical methodologies to enhance the process control strategy and build quality and risk management into the manufacturing process. This is achieved by identifying the critical process parameters (CPPs) and critical quality attributes (CQAs) that are associated with the quality, safety, and efficacy of the drug substance and excipients.

Using the QbD methodology, the determined parameters and quality attributes are used to establish a design space. For this, statistical tools are used to explore how combinations of CPPs might interact and impact on CQAs. This helps manufacturers optimize the production of APIs and drug excipients, ensuring the design of reproducible processes that minimize batch failure risks and significantly reduce batch-to-batch variability.

 

Click here to learn more on how to apply QbD principles to drug development and manufacturing

 

Adoption of advanced process control systems

The process knowledge gained from experimental modeling approaches, such as QbD and DoE, can be used to develop advanced process control systems. These commonly utilize process analytical technology (PAT), which monitors process parameters on-line in real-time.

When PAT is integrated into advanced process control systems, the analytical data may be used to make automatic system adjustments and maintain process parameters within their predefined limits. This reduces manual intervention and creates a closed-loop control system, allowing for the immediate detection of deviations in process conditions and subsequent feedback control.

By immediately correcting any deviations in the process, advanced process control systems enable real-time release testing, assuring manufacturers that the processes have remained consistent, and that the product meets quality and safety standards. By increasing efficiency and consistency, these systems also help to reduce waste, costs, and product cycle times.

Regulatory compliance

Compliance with current good manufacturing practice (cGMP) regulations is critical to ensuring the production of consistent, high-quality, pharmaceutical products. API manufacturing plants are subject to strict regulations, with complex and ever-evolving requirements, stringent quality standards, and severe consequences in case of non-compliance. To comply with cGMP guidelines, several robust management systems must be in place, including those for data integrity, process control, risk management, and supply chain management:

Data integrity

Consistent, accurate, timely, and complete records are required to provide regulators and stakeholders with the confidence that your medicinal product meets all safety and quality standards. Clarity, consistency, and conciseness of the documents must be maintained across the entire product lifecycle. The development of a robust documentation system can help manufacturers with this, establishing effective procedures for naming, authoring, reviewing, approving, updating, storing, and distributing documents.

Control strategy

There are increasing requirements for a clear, well-defined, and scientifically justified process control strategy in cGMP applications. This should include the selection and evaluation of starting materials, followed by approaches including QbD and DoE to link materials attributes and process parameters to product CQAs. These approaches are used to establish a design space and plan control measures to ensure that CQAs are met and a sound process validation methodology is implemented. Furthermore, the process control and validation strategy should be adapted to the increased production scale throughout all stages of the product’s lifecycle.

Risk management strategy

cGMP guidelines require a systematic approach to risk assessment in pharmaceutical manufacturing. This involves the process of identifying, assessing, controlling and reviewing risks based on their potential for impacting the performance of the process and the quality of the product. Risk management plays a central role in scaling up to commercial production in order to mitigate significant quality risks such as cross-contamination and minimize health and safety risks to operators, especially when handling highly potent APIs. Several tools are available for risk assessment, including failure mode and effects analysis (FMEA).

Supply chain management systems

In the pharmaceutical industry, securing patients’ access to drug supplies post-marketing authorization is a regulator’s number one priority. Regulatory guidelines are designed to cover the entire supply chain, from the supply of raw materials to be introduced in cGMP manufacturing operations through to the manufacturing, packaging, labeling, and distribution of the final product.

Digitalizing the management of manufacturing activities can enhance the visibility and efficiency of inventory management, product monitoring, and data exchange throughout the supply chain. Additionally, supply chain management systems should be designed to incorporate risk management strategies for the prediction, prioritization, and mitigation of risks of product stock-outs. This is not only essential for regulatory compliance and patient access, but it will also increase supply chain efficiency to help overcome issues such as inadequate forecasting, long lead times and build-up of working capital.

How to master commercial pharmaceutical manufacturing

The journey to commercialization can be challenging. Scaling up production while maintaining process consistency, product quality, and regulatory compliance, requires expert process development capabilities, and the adoption of innovative science and risk management methodologies. A common pitfall for the Sponsor of an innovative therapy is to under-estimate the complexity and intricacy of this enterprise, which involves the coordinated optimization of strategies for process control, risk management, data management, and supply chain management.

With ever-evolving regulatory requirements and the increasing urge to shorten drug development timelines, getting your drug to market can seem like a daunting undertaking. That’s why taking some of the pressure off your organization by outsourcing your drug development and manufacturing activities to an expert partner can be the smartest decision. This will ensure your drug is commercialized in the fastest and most cost-efficient way possible, utilizing expertise, facilities, equipment, and processes to anticipate and overcome any challenges thrown at your program with ease.

Evotec offers an integrated end-to-end solution for innovative drug R&D, with the capabilities to support all phases of your drug development program. Your projects are in safe hands with our team of expert scientists who are pioneers in QbD, process design, scale-up, and validation, operating to full cGMP within FDA, MHRA, AIFA and BfArM approved facilities.

 

Our chemistry, material science, analytical, and manufacturing operations are co-located at sites in Abingdon, UK, Verona, Italy, and Halle/Westfalen, Germany, with more than 25 years of experience in the development and manufacturing of small molecule APIs.

Alongside integrated process research and analytical development, Evotec supplies APIs for use in pre-clinical development, non-clinical use, clinical trials, and low volume commercial supply.

 

 

API platform

 

Learn more about our API and drug product manufacturing capabilities

Tags: Blog, Formulation & CMC, IND Enabling Studies/Preclinical Development

Key considerations for API process development and optimization

Posted by Evotec on Jun 7, 2024 1:15:16 PM

Active pharmaceutical ingredient (API) manufacturing for new chemical entities (NCEs) is a lengthy undertaking involving many complex chemical processes, including the production of intermediates and purification and isolation steps. Optimizing the manufacturing process, through rational API process design and development, is the key to enabling fast, safe, reproducible, and cost-effective API production. And with the increasing pressure on accelerating drug development while staying within budget and meeting increasingly complex and stringent regulatory requirements, optimizing the API process development workflow has never been more crucial.

However, designing and executing an effective API process development strategy is no small or easy feat. From the early phases of drug development through to commercial manufacturing, the fine-tuning of many individual steps is required to optimize and de-risk API production. This blog outlines some of the key considerations involved in successfully designing and developing an optimal commercial API manufacturing process, including the selection of the synthetic route and regulatory starting materials, the development of a highly effective process control strategy, as well as risk management methodologies, and process safety considerations.

Synthetic route selection

When designing or selecting a synthetic route, it’s important that you get it right, as the use of suboptimal, inefficient routes can drain resources and substantially increase your time to market. Indeed, changing the synthetic route at the late development stage can prove very costly and time-consuming, not least due to the regulatory constraints being faced; in the worst-case scenario, such a change can even trigger the requirement for additional in vivo studies of the drug.

A key consideration for selecting the synthetic route for any type of chemical API is efficiency. Ideally, the chosen synthetic route requires the least number of steps and results in the shortest time to market.

In this regard, scalability is a critical aspect. All chosen materials, unit operations, processing equipment, and conditions should be suitable for scale-up to enable the manufacture of both clinical and commercial scale API batches. Building robustness into the synthetic route by selecting chemical transformations for ease of scalability will help maintain high levels of reproducibility, quality, and cost-effectiveness at increasing scales. This will ultimately ensure continuity in API supply by minimizing the time and costs involved in technology transfer activities.

Additional essential considerations in API synthetic route selection and optimization include those outlined by the SELECT principle: safety, environmental impact, legal requirements, economics, control, and throughput. These principles were first proposed in a 2006 consortium of pharmaceutical manufacturers, including AstraZeneca, GSK, and Pfizer, providing a good basis for selecting or designing a robust, commercially viable synthetic route.

Starting material selection

The selection and introduction of all processing materials, including raw materials, intermediates, solvents, and reagents, will depend on the chosen synthetic route and the API. However, all materials should have well-defined chemical properties, structures, and impurity profiles.

Additionally, a thorough understanding of the selection criteria for starting materials is essential, as outlined in global regulatory guidelines, including ICH Q7 ‘Good manufacturing practice for active pharmaceutical ingredients’. This requires a risk-based approach, which entails gaining a detailed understanding of how changes in the proposed API starting materials can influence the critical quality attributes (CQAs) of the drug substance, including its impurity profile, and the consequences this may have on the medicinal product’s quality.

All starting materials should also be evaluated for their security of supply, in addition to their quality, safety, and environmental impact. It is of particular importance to develop a robust supply chain strategy for custom-made starting materials, including the evaluation and qualification of reliable suppliers.

Developing a process control strategy (PCS)

Each process involved in API manufacturing requires the definition of a unique set of process parameters, such as mixing, temperature, pressure, and time. These parameters must be adequately monitored and tightly controlled to avoid the formation of impurities and prevent inconsistencies in output quality and yield. To develop an effective PCS, several considerations should be made, including the control of input materials, and the use of quality-by-design (QbD) principles to characterize the process and develop appropriate analytical methods to be implemented for quality control.

Quality-by-design (QbD)

QbD is a systematic, rigorous, data-driven approach that should be adopted to improve process and quality control in API manufacturing. QbD relies on predefined objectives to gain a thorough understanding of process control. This starts with determining the drug’s quality target product profile (QTPP), which consists of several design specifications that ensure the product is safe to use and has the desired therapeutic effect.

Based on the definition of the QTPP for the medicinal product, the API CQAs, i.e. the measurable properties of the compound that characterize its quality, including purity, potency, particle size, and stability, are identified. Once API CQAs are defined, the manufacturing process must be thoroughly studied and characterized to evaluate the critical process parameters (CPPs), which are the variables, such as temperature, pH, agitation, and processing time, that impact process performance and consequently product quality.

Essentially, QbD involves the consideration of all materials and processing parameters that could influence product quality. By gaining a sound scientific understanding of the processes through the execution of a rational experimental design, e.g. using a statistical design of experiments (DoE) methodology, this approach enables the elaboration of a more effective control strategy. As documented in the international pharmaceutical guidelines, ICH Q8, Q9, and Q10, the QbD methodology enhances drug development by helping to reduce the risk of batch failure and improving final product quality, safety, and consistency.

 

Learn more about the role of QbD in drug development

 

Analytical method development

QbD principles can also be extended to analytical method development. The API’s CQAs provide a good basis for selecting the most appropriate analytical methods and determining their parameters. Well-designed test methods are paramount to supporting the accurate and reliable monitoring of intermediates and product quality, in line with international quality standards and regulatory guidelines.

For in-process analysis, chromatographic methods, like high-performance liquid chromatography (HPLC) and gas chromatography (GC), are commonly preferred to monitor process execution due to their accuracy and reliability. More recently, the development of process analytical technology (PAT) based on the use of in-line spectroscopic methods, such as Fourier transform infrared (FTIR) spectroscopy, Raman spectroscopy, and focused beam reflectance measurement (FBRM), has helped to improve process control by enabling real-time process monitoring.

When choosing the best analytical method for each process, molecule type and stability arekey considerations. These attributes drive requirements for achieving the appropriate sensitivity and robustness of each assay, including the choice of detection method, range of detection, sample preparation procedure, and analysis time, in addition to cost and ease of routine implementation.

Implementing quality risk management

Risk of failures associated with the facilities, equipment, materials, personnel, processing, product testing, and storage are inherent to API manufacturing. Effective risk management strategies are required to prevent these risks from impacting the quality, efficacy and safety of the API, and from causing costly delays. Recognizing this, ICH guideline Q9 outlines a framework for quality risk management (QRM) applied to pharmaceutical products. This is a strategic approach that involves assessing, controlling, and continuously reviewing potential risks that could occur in API manufacturing, using the following steps:

  • Risk assessment – this step considers the identification of hazards, the probability of them occurring, and the evaluation of the severity of their consequences.
  • Risk control – this next step consists of the use of a decision-making tool to identify and implement control strategies designed to either eliminate or reduce the probability of occurrence, and lessen the severity of the impact down to a level that assures the quality, efficacy, and safety of the product.
  • Risk review – this step is essential to maintaining the effectiveness of the quality risk management framework since it is designed to incorporate the risk assessment and control activities described above while reviewing the new data and knowledge generated throughout the product lifecycle activities.

Several risk management tools are available to aid quality risk management, including failure mode effects analysis (FMEA) and its extension, failure mode, effects, and criticality analysis (FMECA). FMECA is a critical risk analysis tool derived from FMEA and used to determine the potential sources of failure during the execution of the manufacturing process, evaluating the severity and likelihood of such failure, in addition to how it can be detected before it occurs.

Process safety

The development of API manufacturing processes requires particular attention to health and safety considerations to avoid serious and potentially fatal hazards. In addition to ensuring that operators are effectively protected from direct exposure to chemicals, particularly those with high potency, the process chemist also needs to ensure that the safety of the API manufacturing process is carefully assessed and established.

The first step to developing an effective process safety strategy consists of performing hazard assessments for each processing step. These require a detailed understanding of the chemical processes involved, in addition to several other components, including the use of material safety data sheets (MSDS) to take into account specific chemical risks associated with the starting materials and API, and the consideration of any requirement for specific operator training, worker exposure control measures, and compliance with regulatory standards. Although conducting thorough hazard assessments and using these to develop safe processes is time-consuming, it is essential to manage the operational risks associated with small molecule drug development.

Key considerations and future directions for API process development

In summary, a clear API process development strategy is essential to streamline and de-risk drug development, helping your drug reach the market in the most cost and time-effective way possible. Additionally, utilizing science- and risk-based methodologies in your strategy is essential for successful API production and for demonstrating a thorough understanding of the product and the process, including how any changes in the manufacturing process will affect the safety, efficacy, and quality of the final product.

Looking to the future, the adoption of innovative technologies, such as high-throughput experimentation (HTE) and predictive process modeling, will undoubtedly change the landscape of API process development. These methods can effortlessly generate large amounts of data, aiding the prediction of how processes will respond to varied reaction conditions and scale-up configurations. These data-driven techniques will further help to improve decision-making and design more efficient, robust, and scalable API manufacturing processes.

 

Outsourcing your API process development

With these many complex considerations, outsourcing your API process development can maximize your chances of success while taking the pressure off your drug development teams. Leveraging the scientific expertise and tried-and-tested tools and methodologies offered by a specialist partner to develop your processes will pave the way for streamlined and de-risked API manufacturing.

When looking to outsource your API process development, you’re in safe hands with Evotec. The combination of our 25 years of experience in the development and manufacturing of small molecule APIs and our end-to-end shared R&D platform offers a fully integrated approach to process research and analytical development, supplying APIs for use in pre-clinical development, non-clinical use, and clinical trials, all the way through to commercial supplies.

 

API platform

Head to our API capabilities webpage to find out more about how we can optimize your API process development strategy, and de-risk and accelerate your drug development program.

CONTACT OUR EXPERTS

Tags: Blog, Formulation & CMC

Tackling Autoimmune Disease with CAR iNK Cells

Posted by Evotec on Jun 6, 2024 9:29:29 AM

Autoimmune diseases (ADs) represent a significant challenge in healthcare, affecting millions worldwide. These conditions arise when the body's immune system mistakenly attacks healthy tissues, leading to chronic inflammation and tissue damage. While current treatments for ADs focus on immunosuppression, they often come with significant side effects and provide only symptomatic relief. In recent years, there has been growing interest in cell-based therapies, particularly CAR (chimeric antigen receptor) cell therapy, as a potential solution to address the underlying causes of ADs.  

Autoimmune diseases encompass a wide range of conditions, including rheumatoid arthritis, lupus, multiple sclerosis and inflammatory bowel disease, among others. Despite their diverse manifestations, these diseases share a common underlying pathology of immune dysregulation. In healthy individuals, the immune system is finely tuned to distinguish between self and non-self antigens, but in autoimmune disorders, this distinction becomes blurred, leading to an attack on the body's own tissues. 

CAR cell therapy offers a promising approach to treating ADs by depleting disease-driving immune cells and rebalancing immune homeostasis. CAR T cells, which are engineered to express synthetic receptors targeting antigens expressed on the surface of pathogenic cells, have shown remarkable success in treating certain cancers. Now, researchers are exploring the potential of CAR T cells and their counterparts, CAR iNK (natural killer) cells, in targeting the aberrant autoreactive cells implicated in autoimmune diseases to reset the immune sytsem1. We believe that cell therapy approaches could provide long-lasting drug-free remission and potentially a curative treatment for AD patients.

Understanding CAR Therapeutics

CAR T cell therapy first emerged as a groundbreaking treatment modality for hematological malignancies, such as leukemia and lymphoma. Building on this clinical success, researchers have recently turned their attention to applying CAR cell therapy to treat autoimmune diseases. 

In the context of ADs, CAR T cells have been investigated for their ability to target autoreactive T cells or B cells. B cells, in particular, play a central role in many autoimmune disorders by producing autoantibodies and driving inflammation. One promising target for CAR cell therapy in ADs is the cell surface protein CD19. High-level expression of CD19 is maintained throughout the majority of B-cell differentiation stages. By targeting CD19-positive B cells, CAR T cells or CAR NK cells can selectively eliminate the autoreactive B cell populations responsible for driving autoimmune responses2

A recent early clinical study showed promising results for using CD19 CAR T cell therapy to treat ADs. Patients with systemic sclerosis, severe Systemic Lupus Erythematosus (SLE), and idiopathic inflammatory myositis showed remission following therapy3. However, CAR NK cell therapy may be beneficial over CAR T cell therapy, particularly in ADs that have dysfunctional T cells. CAR T cell therapy may also result in side-effects such as GvHD (Graft-versus-host disease, worsening the AD symptoms), neurotoxicity, and cytokine release syndrome; CAR NK cell therapy may offer a safer alternative4

Despite the potential of CAR cell therapy in ADs, several challenges remain, including scalability, persistence, and off-target effects. Evotec's innovative approach to addressing these challenges involves the use of induced pluripotent stem cells (iPSCs) to generate allogeneic off-the-shelf CAR iNK cells with enhanced scalability and precision. iPSCs, reprogrammed from adult somatic cells, offer a potentially inexhaustible source of immune cells that can be genetically engineered and differentiated into various cell types to tackle a range of diseases.

iPSC-Derived CD19 CAR iNK Cells for Targeted B Cell Depletion

A study by researchers at Evotec investigated the performance of iPSC-derived CD19 CAR iNK cells as a novel therapeutic approach for ADs. Using Evotec’s validated GMP iPSC line, researchers produced genetically modified cells with a knock-in of CD19-CAR. The modified cells successfully differentiated into CD19 CAR iNK cells using a feeder-free 3D differentiation process, which could be confirmed by flow cytometry. The established protocol can ensure high purity and functionality of the resulting cells. Results showed that iNKs generated from the GMP iPSC line were homogenous and phenotypically comparable to blood-derived (BD) NK cells form healthy donors.  

Cytotoxicity Fig 1

Figure 1: Cytotoxicity against SLE patient B cells. NK killing assays of effector cells - iNK without CAR (WT), CD19 CAR iNK or healthy donor BD NK cells (BD-NK) co-cultured 1:1 (E:T) with SLE patient B cells, + 10μg/ml anti-CD20 antibody (Obinutuzumab (Obi)) or isotype control (Iso) (n=2). 

In vitro experiments demonstrated the cytotoxic effector function of CD19 CAR iNK cells in selectively targeting and eliminating CD19-positive B cells. Co-culture assays using patient-derived primary B cells from patients with SLE autoimmune disease, showed robust cytotoxicity of CD19 CAR iNK cells. The CD19 CAR iNK cells were more efficient than iNK cells without a CAR or BD NK cells in depleting the patient-derived primary B cells. These findings highlight the therapeutic potential of CD19 CAR iNK cells in treating ADs, offering a targeted and scalable alternative to conventional immunosuppressive therapies.

Evotec's Scalable Therapeutics Approach

Evotec's commitment to allogeneic cell therapy innovation is exemplified by its scalable therapeutics approach, which leverages cutting-edge technologies and infrastructure to develop next-generation therapies for ADs. Central to this approach is the use of iPSCs as a platform for generating CAR iNK cells with enhanced scalability and precision. By introducing CARs targeting CD19 into iPSCs and differentiating them into iNK cells, Evotec aims to create scalable and precise off-the-shelf therapies for ADs. 

Pipeline co-creation Fig 2

Figure 2: Evotec’s pipeline to co-create iPSC-based cell therapeutics with partners in Inflammation & Immunology.

Evotec's end-to-end process for iPSC-based therapeutics encompasses differentiation, gene editing, preclinical and clinical development, ensuring the efficient generation and characterization of CAR iPSC-derived cells. By utilizing validated GMP iPSC lines and GMP-compatible differentiation protocols, Evotec ensures the safety and quality of its allogeneic cell therapy products, paving the way for clinical translation.

iPSC Fig 3

Figure 3: Evotec’s end-to-end process for iPSC-based therapeutics. 

The scalability of Evotec's approach enables the production of large quantities of CAR iNK cells, making them suitable for widespread use in treating ADs. Additionally, the precision of iPSC-derived CAR iNK cells allows for targeted and personalized therapies tailored to individual patients' needs, reducing the risk of off-target effects and enhancing treatment efficacy.

Future Potential with Evotec

Evotec's iPSC-derived CD19 CAR iNK cells represent a promising new approach to treating autoimmune diseases. By harnessing the power of iPSC and CAR technology, allogeneic cell therapy can help revolutionize the treatment landscape for ADs, offering patients a targeted, scalable and potentially curative alternative to conventional therapies. 

As research in this field continues to advance, Evotec remains at the forefront of allogeneic cell therapy innovation, driving the development of next-generation treatments for ADs (Figure 2). Evotec’s GMP-compliant production pipelines provides an efficient, reproducible, and scalable way to produce CAR iNK cells derived from iPSC for clinical development. With its commitment to precision medicine and scalable therapeutics, Evotec is well-positioned to meet the growing demand for effective and accessible off-the-shelf therapies for autoimmune diseases.

 

See more iPSC-based Cell Therapies for I&I Diseases

To discover more about this research, download our scientific poster

 

References: 

(1) Blache, U.; Tretbar, S.; Koehl, U.; Mougiakakos, D.; Fricke, S. CAR T Cells for Treating Autoimmune Diseases. RMD Open 2023, 9 (4), e002907. https://doi.org/10.1136/rmdopen-2022-002907.

(2) Jin, X.; Xu, Q.; Pu, C.; Zhu, K.; Lu, C.; Jiang, Y.; Xiao, L.; Han, Y.; Lu, L. Therapeutic Efficacy of Anti-CD19 CAR-T Cells in a Mouse Model of Systemic Lupus Erythematosus. Cell. Mol. Immunol. 2021, 18 (8), 1896–1903. https://doi.org/10.1038/s41423-020-0472-1.

(3) Müller Fabian; Taubmann Jule; Bucci Laura; Wilhelm Artur; Bergmann Christina; Völkl Simon; Aigner Michael; Rothe Tobias; Minopoulou Ioanna; Tur Carlo; Knitza Johannes; Kharboutli Soraya; Kretschmann Sascha; Vasova Ingrid; Spoerl Silvia; Reimann Hannah; Munoz Luis; Gerlach Roman G.; Schäfer Simon; Grieshaber-Bouyer Ricardo; Korganow Anne-Sophie; Farge-Bancel Dominique; Mougiakakos Dimitrios; Bozec Aline; Winkler Thomas; Krönke Gerhard; Mackensen Andreas; Schett Georg. CD19 CAR T-Cell Therapy in Autoimmune Disease — A Case Series with Follow-Up. N. Engl. J. Med. 2024, 390 (8), 687–700. https://doi.org/10.1056/NEJMoa2308917.

(4) Műzes, G.; Sipos, F. CAR-Based Therapy for Autoimmune Diseases: A Novel Powerful Option. Cells 2023, 12 (11), 1534. https://doi.org/10.3390/cells12111534.

 

Tags: Induced pluripotent stem cells, Blog, Biologics, In vitro Biology, In vivo Pharmacology, Immunology & Inflammation

Achieving Continuous Manufacturing through Equipment Design

Posted by Evotec on Jun 5, 2024 4:42:25 PM

Continuous biomanufacturing is reducing the cost of goods of biopharmaceuticals. Achieving continuous manufacturing requires expertise in equipment design.

Download the highlights of Andrea Isby's presentation at Repligen's DSP Workshop in Estonia from May 23rd, 2024 to learn more. 

Download the presentation

Tags: Neuroscience, Respiratory, Oncology, Kidney diseases, Women's health, Presentations, Blog, Formulation & CMC, Biologics, Age-Related Diseases, IND Enabling Studies/Preclinical Development, Anti-Infectives, Immunology & Inflammation, Metabolic Disease & Complications, Rare Diseases, Clinical Development