How the Netherlands became a key player for vaccine development

The Netherlands has a long history in the life sciences industry. Especially in the field of vaccine development and manufacturing, this small country has always played a significant role. Lately, the rich history proved a fertile ground for developing and manufacturing a wide variety of SARS-CoV-2 vaccines.

The website Invest in Holland has written an interesting article on how the Dutch became such a prominent player for vaccine development. Gerard Schouw, Director of the Association Innovative Medicines, states in the article:

“The Dutch life sciences & health sector has been at the innovative forefront of Europe for many years. The Netherlands has a lot to offer due to its central location in Europe and successful history of public-private partnerships.”

Role of Batavia in vaccine development

We, at Batavia, are proud to play our part in the development of new and innovative vaccines. In the recent years, we have announced partnerships for the development of vaccines against viruses like: SARS-CoV-2, polio, Marburg, Lassa, rota, measles, and rubella.

In each project, we strive to improve the affordability and availability of these life-saving medical countermeasures. For example, with our  HIP-Vax®  platform for low-cost, highly intensified manufacturing, we are able to bring the Cost of Goods of vaccine manufacturing well below <$1.00 per dose and reduce the time from bench to clinic to 9 months. This timeline is including the biosafety testing. These benefits are the result of significant reductions in the manufacturing footprint, increased cell density and less process steps.

With our extensive bioprocessing knowhow, we will continue to expand our footprint in the vaccine development sector until all those in need have access to vaccines.

Link  to full article

How to Work With a CDMO for Viral Vectors: 5 Steps to Success

How to Work With a CDMO for Viral Vectors: 5 Steps to Success

Batavia Biosciences - Favicon

Viral vector technologies are at the heart of many promising new therapies, such as vaccines, gene therapies, immune-oncology therapies and other advanced therapy medicinal products (ATMPs). In this rapidly evolving and competitive landscape, there is little scope for error or inefficiency in the development process.

A contract manufacturing and development organization (CDMO) specializing in viral vector products can be a lifeline for process development and CMC managers, who bear the lion’s share of responsibility for ensuring their company’s innovations make it from bench to bench clinic as quickly and efficiently as possible. Outsourcing to a CDMO is one of the best ways to de-risk and accelerate viral vector projects—provided it is carefully planned and managed. So how do you engage effectively with a CDMO and make the most of this vital relationship? Here are 5 steps to a successful collaboration.

1. Start Early

In viral vector product development, the seeds of success—or failure—are sown early. In particular, there are many challenges and pitfalls when taking a product from proof-of-concept to the first clinical trials. The decisions you make at this stage can have major consequences regarding the overall capability, reliability, quality and cost-efficiency of your process.

By reaching out to a CDMO in the early stages of development, you can benefit from the full range of their experience, capabilities and services. This usually starts with a comprehensive evaluation. For example, how scalable is your current process? Is your cell line suitable for GMP manufacturing? Do you have the appropriate analytical methods for in-process and release testing?

A CDMO with a strong track record in viral vector development and process engineering should be able to evaluate your project from every angle—scientific, technical, operational, cost and regulatory. An early assessment will maximize the number of design options available and ensure that strategic gaps, issues, and potential stumbling blocks are spotted early, before they become problematic.

2. Be Clear About What You Need and Want From a CDMO

The first step to getting what you want from a CDMO partnership is knowing what you want. This may sound trivial, but it is surprising how often companies reach out to a CDMO with a request for proposal (RFP) before having a clear idea about precisely what they need to get out of the partnership and how they prefer to collaborate.

Before engaging with a CDMO, it is a good idea to run a gap analysis to assess your in-house capabilities and identify any gaps you think need filling regarding skills, expertise and resources. Consider also the more intangible qualities that make for a good working relationship. Intangible qualities could be your preferred communication style, the importance of compatible values, and the degree of responsiveness, flexibility and support you expect from a potential partner.

3. Find the Right Fit

With your general needs and preferences in mind, you will be better positioned to research potential CDMOs, articulate your needs, and effectively interview candidates. At the same time, it is vital to remain open-minded in your initial discussions with CDMOs. In many cases, they will be able to spot gaps or needs that weren’t identified in your gap analysis.

Some of the key topics to explore with potential CDMOs include:

  • Range of services and competencies. Does the CDMO have the right capabilities to cover all of your needs, from virus and cell banking to regulatory sign-off and support for manufacturing handover?
  • Track record in viral vector process development and clinical manufacturing. How extensive is the range of viral vectors they have successfully manufactured? Will your vector system be new to them?
  • Flexibility to tailor their approach and processes to meet your vector-specific requirements.
  • Equipment and facilities. Are they appropriate for your current and future needs?
  • Regulatory history and experience with IND/IMPD dossier submissions. Can the CDMO prepare and provide all the necessary information for filing? Do they offer support in completing the CMC section?
  • Project and program management capabilities. How experienced are they at managing complex viral vector development projects? Would you be able to trust them to drive your project forward while you focus on the high-level strategy and manage your in-house team?
  • Capacity and willingness to support you at every stage of the project. Importantly, how will the CDMO prioritize your project against existing work or the needs of larger clients?

Before you make your final selection, arrange some face-to-face time with the leadership team. If possible, an on-site meeting will help you assess the culture. During such a meeting you can get a sense of what it would be like to work together. Do they listen? Are they open and easy to get along with? Do you feel comfortable working together? Is this a company you can trust to deliver?

4. Align and Organize

Once you have found a good match, proactive engagement is key to ensuring your project gets off to a good start. This includes putting the right mechanisms in place to keep the momentum going. Together with the CDMO, organize an on-site kick-off meeting to align on project vision and scope. Key milestones, priorities and ways of working can be established during the meeting. Ensure roles and responsibilities are clearly defined from the start so there is ownership when problems arise. Along the way, note how quickly the CDMO gets up to speed and takes action on your requests. If they are slow to respond in the signing of documents, for example, or to address your questions and concerns, this could be an early warning sign.

5. Maintain Strong Lines of Communication

Lastly, never underestimate the importance of transparency and regular communication throughout your project. Establishing a schedule of communication and sticking to it will help you build strong connections and establish trust. Your CDMO should also be willing and available to participate in more impromptu discussions to work out any issues as soon as they arise. The more proactive you are about ensuring the lines of communication are open, the better able you will be to build a productive and lasting partnership.

Partnering with a CDMO can make a world of difference for your project. To discover how, talk to one of our experts. They will be happy to discuss your needs and perform a free review of your current process.

Related

Guide To Viral Vector Production & Development

Guide To Viral Vector Production & Development

Batavia Biosciences - Favicon

Viral vector technologies are taking center stage in the development of novel vaccines and therapies to combat a wide spectrum of human diseases. Efficient development of safe, cost-effective, and robust methods for viral vector manufacture is therefore of vital importance for patients to benefit from these potentially life changing new medicines.

Can I use my cell line for clinical production? What is the quickest route to clinical trials? How can we effectively de-risk and accelerate viral vector development?

These are just a few of the many questions we address in this go-to resource for anyone navigating the complexities of viral vector process development, scale-up, and cGMP manufacturing.

Viral Vector Technologies

The use of viral vectors in vaccines and targeted therapies is revolutionizing the way we tackle some of nature’s most intractable human diseases and infectious pathogens. From rare diseases to cancer to COVID-19: viral vectors are becoming indispensible weapons in the armory of drug modalities.

What Are Viral Vectors?

Viral vectors are viruses that have been engineered in the lab to efficiently infect target cells and deliver a genetic payload. These properties make them highly attractive for a wide variety of clinical applications.

When genetically engineered with the code for an immunogenic antigen, viral vectors essentially hijack the cell’s own machinery to produce large amounts of the antigen, which is then effectively presented to the host immune system. This makes them an attractive platform for development of novel prophylactic and therapeutic  viral vector  vaccines.

Viral vectors also offer tremendous promise for the development of novel  cell and gene therapies. An estimated 70% of gene therapy trials worldwide involve the use of viral vectors.

The scope of viral vector technology further extends to the development of oncolytic viruses, a rapidly evolving class of anticancer agents that are tailored to specifically target cancer cells for destruction, working in combination with the patient’s immune system.

Viral Vector Vaccines 

The rising global threat of infectious diseases and cancers has been a major driver in the evolution of next-generation vaccines.

Viral vector technologies in particular have emerged as an enabling platform for vaccine development. Notably, the technology gave rise to the first FDA-approved vaccine for Ebola virus in late 2019, and in December 2020 the first viral vector vaccines for SARS-CoV-2 / COVID-19 were granted emergency use authorization (EUA) less than a year after the genetic sequence for SARS-CoV-2 was shared.

Milestones like these, underpinned by significant progress in addressing safety and scale-up challenges over the past few years, are paving the way to more widespread adoption of viral vectors in vaccine development.

Virus Vector Phase I Clinical Trial
Adenoviruses (Ad)
ChAd3 (Chimpanzee adenovirus) Ebola Zaire, Hepatitis C, Ebola Sudan, Ebola Marburg
ChAdOx (Chimpanzee adenovirus) Tuberculosis, Chikungunya, MERS-CoV
Ad5 (Adenovirus type 5) Cystic fibrosis, HIV, Ebola Zaire
VXA (Replication-deficient Ad5) Respiratory syncytial virus, Norovirus, Influenza
rAd26 (Recombinant Ad 26) HIV, Ebola Zaire
Ad35 Tuberculosis, HIV
Ad4 HIV, Anthrax
Alphaviruses
VEE Replicon (Venezuelan equine encephalitis) CMV
Measles Virus (MeV)
Measles virus COVID-19
Poxviruses
MVA (Modified vaccinia virus Ankara) Ebola, HIV, Hepatitis C, MERS-CoV
FPV (Fowlpox vector) HIV
ALVAC (canarypox vector) HIV
Vesicular Stomatitis Virus (VSV)

Replication-competent VSV

rVSV

HIV

Lassa virus

Virus Vector Phase II Clinical Trial
Adenoviruses
ChAdOx1 Malaria, SARS-CoV-2
Poxviruses
MVA CMV, Tuberculosis

 Table 1. Human clinical trials in progress with viral vectored vaccines1,2

Gene and Cell Therapies 

 According to the Alliance for Regenerative Medicine, at the end of 2020 there were 1,220 ongoing regenerative medicine and advanced gene and cell therapy trials worldwide.3 Of these, 152 were in phase 3 clinical trials, in line with EMA and FDA predictions that by 2025 the annual rate of cell and gene therapy approvals will reach 10-20 per year.

Valued at $4.4 billion in 2020, the global gene and cell therapy market is projected to reach $15.48 billion by 2025 at a growth rate of 28.7%.4 Viral vectors are playing a central role in this rapidly expanding market.

Although  in vivo  gene therapies have had their ups and downs recently, there are over 400 viral vector gene therapy assets in preclinical and clinical phases of development.5 The vast majority of these utilize adenovirus, adeno-associated virus (AAV), and lentivirus vectors.

While cancer is by far the predominant disease area, viral vector gene and cell therapies are under investigation for a wide variety of other indications including cardiovascular and monogenic disorders, as well as metabolic, inflammatory, neurological and ocular diseases.

In late 2020, two new viral vector-based therapies gained regulatory approval: Libmeldy (Orchard Therapeutics), approved by the European Medical Agency for treatment of metachromatic leukodystrophy (MLD), and Tecartus (Kite), the first CAR-T treatment approved by the FDA for relapsed or refractory mantle cell lymphoma. This was followed in February 2021 by FDA approval of Breyanzi, a lentiviral vector-based CAR-T therapy for relapsed or refractory Large B-cell Lymphoma.

Oncolytic Viruses 

Targeted eradication of cancer cells using oncolytic viruses is another exciting and constantly evolving application area for viral vector technologies.

Oncolytic viruses are able to selectively infect, replicate in and lyse cancer cells and/or deliver a genetic payload, leaving healthy cells untouched. In addition to directly mediating cell death, virus-induced lysis of the target cell can activate cytotoxic T cells. This wakes up the immune system to actively seek out and destroy tumor cells.

Many different virus families and subfamilies have been investigated for use in oncolytic therapies, including: adenovirus, coxsackievirus, herpes simplex, vaccinia, measles, reovirus and parvovirus. This diversity presents a potential manufacturing challenge in terms of the breadth of manufacturing strategies and capabilities required.

Roadmap for Viral Vector Process Development 

 While global markets for viral vector vaccines and therapies continue to expand, the journey from bench to clinic is rarely straightforward. Strategic end-to-end planning of the entire process development journey is an important first step to  avoid common development and scale-up pitfalls, mitigate risks across the development program, and identify the most cost-effective strategies for each stage of development. 

 

Common Roadblocks in Viral Vector Production

While the promise of viral vector-based vaccines and therapies is great, so too are the development risks. This is largely because of the complexities of working with viral particles and living cells, including the challenges of scaling up production for clinical manufacture. At the same time, rigorous safety standards must be met to ensure products are safe for patients and the environment.

The bottom line is that many potentially life-changing viral vector products never make it to market. It is not uncommon to invest considerable resources in viral vector development, only to discover at a late stage that the production process is not sufficiently scalable, robust or cost-effective to be commercially viable. As these statistics from the vaccine industry illustrate, the price of failure is high:

5 Frequently Reported Challenges in Viral Vector Manufacturing 

Recent business surveys10 have identified some of the main problems respondents mention when asked about the difficulties they face in viral vector manufacturing. These include:

  1. Complexity of working with viral particles and living cells
  2. Scaling up manufacturing processes
  3. Gaps in process engineering and regulatory expertise
  4. Temperature sensitivity and related storage and logistics challenges
  5. High capital investments required to establish and maintain production facilities

To address issues like these and de-risk development, many viral vector product developers are turning to contract manufacturing and development organizations  (CDMOs) that have the track record, specialized expertise, and facilities needed to bypass these issues.

Foundations and Guiding Principles For Success 

Designing for quality, manufacturability, compliance and cost-efficiency are key foundations for success when developing any commercial product. This becomes all the more important when manufacturing viral vectors, which are especially complex, difficult to handle, and subject to stringent regulatory oversight.

Batavia’s guiding principles in viral vector process development

At Batavia Biosciences, we follow three guiding principles in viral vector process development:

Right First Time

 Getting process development right the first time around starts with a thorough understanding of the expected commercial manufacturing process.  This means ensuring that all raw materials, equipment and process architecture will allow seamless extrapolation to commercial scales. This is supported by Design of Experiment (DoE) approaches, scale-down models, assay platforms, and cost modeling tools that enable timely data-driven decision-making at every stage of the process.

Cost Efficiency

A cost-efficient manufacturing process is vital to ensuring you meet the Cost of Goods (CoG) requirements for your product. This includes fine-tuning upstream and downstream processes to maximize viral yield in bioreactors and obtain high recovery from purification steps.

Bullet-Proof Documentation

 Robust documentation is essential at every stage of development: from establishing a sound development roadmap, to submitting an IND or IMPD dossier that will hold up to regulatory scrutiny, to ensuring that QA release and technology transfers happen without a hitch. Batavia’s expert QA team provides vital support throughout the entire development cycle. 

    Step by Step: Troubleshooting Process Development

    Process Evaluation and Design 

    The starting point for clinical process development is usually a thorough evaluation of the current process for vector production. Raw materials, including plasmids, virus seeds, cell lines, media and supplements, are assessed for cGMP compliance and suitability for clinical manufacture, as well as ability to achieve the target product profile (TPP).

    Both the upstream and downstream processes are assessed for overall design, scalability and anticipated CoG. Time constraints and investment milestones are also factored into the mix to map out the most phase-appropriate and cost-efficient strategies. In addition, critical supply chains and any necessary licensing for raw materials or technology IP needs to be secured.

    Batavia Biosciences has a strong track record in cGMP process development for clinical manufacturing of viral vectors. Our experts can evaluate your project from every angle – scientific, technical, operational, financial and regulatory. An early assessment will maximize the number of design options available and ensure that strategic gaps, issues, and potential stumbling blocks are spotted early, before they become problematic.

    Cell Line Selection:   Can I Use My Cell Line for Clinical Production? 

    One of the most crucial steps in the evaluation phase is ensuring that a suitable cell line has been selected.

    Some of the questions asked at this stage include:

    • Does the cell line comply with cGMP requirements? For example, is the history of the cell line known? Has it been properly tested for purity and viral safety?
    • Is a cGMP-produced master cell bank (MCB) available?
    • How robust does the cell line need to be to tolerate upstream processing
    • Does the cell line support viral stability and yields?
    • Does it make sense to switch from an adherent to a suspension format?

    HEK293 cells and their derivatives are a common choice for clinical manufacture of AAV, adenovirus and lentivirus vectors. Vero cell lines are also widely used, particular for production of measles and VSV vectors.

    Cell and virus banking

    Once an appropriate cell line has been chosen, the MCB can be established. To do this, the cells are expanded under cGMP conditions and quality tested to demonstrate identity, purity and viral safety. From the MCB, working cell banks (WCB) can be generated. These are the banks that will be used for expansion to support production of the final product, without the chance of depleting the MCB.

    Similarly, a robust, traceable and regulatory-compliant master virus seed (MVS) must be established using the MCB or WCB. Once tested to confirm viral identity, purity and stability, the MVS gives rise to the working virus seed (WVS) used in the production process.

    Upstream process (USP) 

    Cell cultures are inherently heterogeneous, sensitive to mechanical and environmental disturbances, and susceptible to contamination. Factors such as these mean that the USP portion of viral vector manufacturing is particularly challenging to develop, optimize and control. 

    During USP development, many parameters need to be investigated to identify the critical process parameters. These must then be optimized to find the “sweet spot” for a robust, reproducible and cost-efficient design. 

    Upstream Processing Steps

    Given the large number of USP variables, the numbers of experimental conditions and replicates that need to be tested can quickly become unmanageable or require too much time and materials. Design of Experiments (DoE) provides a systematic way to maximize the information gained about the test system, while limiting the number of experiments. It can also uncover hidden relationships between variables that can have profound effects on process outcomes. In a nutshell, DoE approaches can enable you to more quickly hone in on the most optimal system and process settings. This significantly shortens the development cycle and reduces experimental costs.

    Batavia Biosciences combines DoE with its proprietary  SCOUT® platform to facilitate high-throughput cell culture in mini bioreactors that closely mimic what happens at clinical production scales. Not only does this reduce the amount of time and materials required for testing, it also allows parallel development of both upstream and downstream processes.

    Downstream process (DSP) 

    One of the many challenges of  DSP development is that processing needs are highly product-specific and dependent on the type of vector produced and USP choices. This means that the nature and order of processing steps can vary widely, there are no standardized solutions for vectors, and it can be difficult to develop both USP and DSP in parallel.

    Despite product-specific variations, there are certain commonalities in downstream workflows, usually starting with clarification of the harvest to remove large impurities like cell debris. This is typically followed by tangential flow filtration (TFF) to concentrate the virus and transfer it to the appropriate buffer. 

    If depth filtration is used, it is sometimes possible to streamline DSP by combining particle purification with removal of impurities such as host-cell or plasmid-derived nucleic acids. A number of chromatography steps may be needed to remove any remaining impurities. If the viral vector particle size is not prohibitive, a sterile filtration step is performed after formulating in the final buffer. Otherwise, aseptic process validation may be a necessity.

    Analytical Methods and Assay Development

    Success of viral vector process development and manufacturing strongly depends on having suitable analytical assays for a variety of different purposes:

    • Qualification of MCB and MVS
    • Materials testing and control
    • Process development 
    • Monitoring and QC during manufacturing 
    • Quality assurance and release testing 

    To develop the appropriate assays product release, the process must first be well characterized. This means being able to generate representative material. Assays intended for release testing must then be qualified for phase 1/2 clinical trials.  Regulatory guidelines such as those issued by the ICH and FDA for chemistry, manufacturing and control (CMC) require demonstration of suitable testing for key critical quality attributes relating to: identity, potency, purity, safety and stability.

     

    Batavia uses state-of-the-art assay technologies, and supports analytical assay development and implementation to support the entire development and manufacturing cycle. Examples of the types of analytical assays that may be implemented during viral vector development are shown in Table 2.

    What can be tested Example Assay
    IDENTITY  
    Genetic identity Genome sequencing (NGS) PCR
    Identity PCR
    Protein identity SDS-PAGE
    Western blot (immunoblot)
    STRENGTH/POTENCY  
    Physical viral titer ELISA
    qPCR
    Optical density (A260/280)
    HPLC
    Functional viral titer Plaque-forming assay
    Fluorescence foci assay
    TCID50 (end point dilution assay)
    PURITY  
    Residual testing ELISA (benzonase, BSA)
    Host cell-related impurities Host cell DNA/RNA: Picogreen, qPCR
    Host cell proteins: ELISA
    General impurities HPLC
    SAFETY*  
    Sterility Standard sterility tests (EP 2.6.1, USP71)
    Endotoxin LAL method (EP 2.6.14, USP85)
    Mycoplasma qPCR (EP2,6,7)
    Mycobacterium Culture medium method (EP 2.6.2)
    Environmental monitoring Environmental monitoring during production
    Adventitious viruses (human, bovine, porcine) qPCR 
    Adventitious agents In vivo and in vitro cellular assays
    STABILITY  
    pH Potentiometry
    Appearance Check for visible particles
    Osmolality Osmometry
    Aggregate formation HPLC
    DLS
    Stability indicating TCID50
    Western blot

    Table 2. Analytical Assays in viral vector development

    *Assays in these categories may be outsourced

    How to Accelerate The Journey to Clinical Trials

    In the high-stakes arena of viral vector vaccines and therapies, companies are under increasing pressure from changing market forces, competitors, investors and other stakeholders to get products to market as quickly as possible. At the same time, quality needs to be assured and program risks minimized.

    Finding the quickest route to clinical trials requires considerable experience and know-how across a wide range of disciplines. While there are no one-size-fits-all solutions, our experts at Batavia have found that there are 3 actions in particular that can help drive a swift and successful transition from bench to clinic.

    Use In Silico Cost Modeling

    When looking for opportunities to streamline processes and accelerate development it is crucial to be able to quickly understand the cost implications of any proposed changes. Without this insight, what seems like a timesaving measure could instead lead to costly delays. In silico cost modeling of production processes can help you make well-informed decisions at every stage of development, saving considerable time and resources.

    Combine DoE with Scale-Down Analysis

    To develop a robust and reliable manufacturing process, extensive experimentation is needed to identify and optimize all the critical quality attributes that can impact on process performance, yield and cost. Due to the complexity of viral vectors and cell culture systems, the number of parameter combinations that need to be tested in parallel can still run into the hundreds, even with a DoE approach.

    In many cases, running so many experiments would simply not be feasible or cost-efficient without miniaturization. To overcome this problem, it is possible to use scale-down platforms that faithfully mimic full-scale process steps—both upstream and downstream.

    Batavia Bioscience achieves this with its proprietary SCOUT® platform, which integrates mini bioreactors for high-throughput cell culture with high-throughput purification technology and matching analytical capabilities.

    DoE methodology paired with scale-down models is becoming an indispensible tool for viral vector process development. Using this approach it is possible to develop a representative process that can be used to generate enough material for phase 1 clinical trials without having to wait until the full scale process has been implemented. This saves considerable development time and cost.

    Partner to Mitigate The Risks

    Companies venturing into development of viral vector products for the first time often have a deep understanding of the biotechnology but more limited expertise and resources in other crucial aspects of development and manufacturing, such as process engineering, cleanroom operations, cGMP manufacture and regulatory affairs.

    This introduces significant risk into the program, and raises a red flag for venture capitalists and other potential investors. On top of that, in-house development and manufacture may require significant capital investment in specialized equipment and regulatory-compliant facilities. For less experienced players, there is also a high risk of failing to secure regulatory approval for phase 1 clinical trials, due to problems in completing the CMC section of the IND or IMPD dossier.

    What is the best way to mitigate these risks and speed the journey from bench to clinic? In many cases, a CDMO partnership is the answer. 

    If you are considering partnering with a CDMO to accelerate your development journey, our blog provides some top tips to ensure you make the most of this important relationship:  How to work with a CDMO for viral vectors: 5 steps to success.

    Common abbreviations and terms

    CDMO
    (Contract Development and Manufacturing Organization) – A company that provides development and manufacturing services on a contract basis.

    cGMP
    (Current Good Manufacturing Practice) – Refers to the Current Good Manufacturing Practice regulations issued by the United States Food and Drug Administration. cGMP regulations aim to assure the identity, strength, quality and purity of drug products by requiring that manufacturers to adequately control manufacturing operations. The “c” of cGMP stands for “current”, signifying the requirement for companies to use up-to-date technologies and systems that are fully compliant with the latest regulations.

    CMC
    (Chemistry, Manufacturing and Controls) – The body of information that defines product characteristics, manufacturing processes and product testing to ensure the safety, efficacy and batch consistency of pharmaceutical products.

    CoG
    (Cost of Goods) – In manufacturing, the abbreviation CoG is often used to refer to the cost of goods manufactured, comprising the total direct manufacturing cost of a product, including materials, labor and factory overhead.

    CPP
    (Critical Process Parameter)  A key variable in pharmaceutical manufacturing affecting the production process. CPPs impact critical quality attributes (CQA) and should therefore be controlled within a proven acceptable range to ensure the drug product meets its quality specifications.

    CQA
    (Critical Quality Attribute) – Any physical, chemical, biological or microbiological property or characteristic that must be maintained within a defined range, limit or distribution in order to ensure a product’s quality.

    DoE
    (Design of Experiments) – A systematic method of determining cause-and-effect relationships between factors affecting a process and the output of that process.

    DP
    (Drug Product) – A specific drug in dosage form.

    DS
    (Drug Substance) – An active ingredient that is intended to furnish pharmacological activity or other direct effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure or any function of the human body, but does not include intermediates used in the synthesis of such ingredient.

    DSP
    (Downstream Processing) – Describes the steps in biopharmaceutical manufacturing required to purify the desired ingredient, from the production harvest up to purified Drug Substance Bulk.

    IMP
    (Investigational Medicinal Product) – A pharmaceutical drug or active substance or placebo being tested or used as a reference in a clinical trial.

    IMPD
    (Investigational Medicinal Product Dossier) – A document that must be submitted to regulatory authorities in the European Union in order to gain approval for use of an IMP in clinical trials.

    IND
    (Investigational New Drug) – A drug that has not been approved by the US FDA, but is under investigation for use in human clinical trials. An IND application must be submitted to the FDA to obtain authorization to administer an IND or biological product to humans in a clinical trial.

    MCB
    (Master Cell Bank) – A bank of a cell substrate from which all subsequent cell banks will be derived. The MCB represents a collection of cells of uniform composition derived from a single source prepared under defined culture conditions.

    MVS
    (Master Virus Seed) – A viral seed of a selected vaccine virus, from which all future vaccine production will be derived—either directly or via Working Virus Seeds.

    Scale-down model
    – A scaled down or miniaturized model that mimics a larger scale unit operation in the manufacturing process. Scale-down models are developed and used to support process development studies. Models should account for scale effects and be representative of the proposed commercial process. A scientifically justified model can enable a prediction of quality, and can be used to support the extrapolation of operating conditions across multiple scales and equipment.

    SCOUT®
    – A scale-down model using a miniaturized production and purification platform to rapidly develop multivariate processes. The SCOUT® technology provides for an effective tool for“Design-of-Experiments” (DoE)  approaches.

    SIDUS®
    – The SIDUS® platform combines biological materials (cell lines and vector systems) with in-depth experience and complete protocol systems for manufacturing viral vector-based vaccine, oncolytic and gene therapy products.

    USP
    (Upstream Processing) – The entire process from early cell isolation and cultivation, to cell banking and culture expansion of the cells, and production of the desired biological substance until final harvest.

    WCB
    (Working Cell Bank) – A cell bank derived by propagation of cells from MCB under defined conditions and used to initiate host cell cultures required for virus production on a lot-by-lot basis.

    WVS
    (Working Virus Seed) – A viral seed derived by propagation of virus from the MVS under defined conditions and used to initiate production virus production lot-by-lot.

     


     

    Vrba SM et al. Development and applications of viral vectored vaccines to combat zoonotic and emerging public health threats. Vaccines (Basel) (2020) 8(4):680.

    2 ClinicalTrials.gov database. US National Library of Medicine, NIH, https://clinicaltrials.gov/ct2/home.

    3 Alliance for Regenerative Medicine. (2021). 2020: Growth & Resilience in Regenerative Medicine. Annual Report. https://alliancerm.org/sector-report/2020-annual-report

    4 Gene Therapy accounts for a major portion of the cell and gene therapy market and it is expected to have the most growth. The Business Research Company, Intrado GlobeNewswire, 23 February 2021, https://www.globenewswire.com/news-release/2021/02/23/2180767/0/en/Gene-Therapy-Accounts-For-A-Major-Portion-Of-The-Cell-And-Gene-Therapy-Market-And-It-Is-Expected-To-Have-The-Most-Growth.html.

    5 Capra E et al. (17 May 2021). Gene-therapy innovation: Unlocking the promise of viral vectors. McKinsey & Company web post. Retrieved from https://www.mckinsey.com/industries/pharmaceuticals-and-medical-products/our-insights/gene-therapy-innovation-unlocking-the-promise-of-viral-

    6 D’amore T and Yang Y-P. Advances and Challenges in Vaccine Development and Manufacture. BioProcess International (2019) Volume 17, September issue.

    7  This is how much it costs to develop a vaccine. The Cost of Things. MarketWatch, 1 October

    8  Pronker ES et al. Risk in vaccine research and development quantified. PLOS One (2013) 8(3): e57755.

    9  Gouglas D, et al. Estimating the cost of vaccine development against epidemic infectious diseases: a cost minimisation study. The Lancet (2018) 6(12): E1386-1396.

    10 Viral Vectors, Non-Viral Vectors and Gene Therapy Manufacturing Market (3rd and 4th Editions)Roots Analysis.

    Related

    Copyright © 2023. All rights reserved.
    Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16 2333 CL Leiden, The Netherlands

    Reg No: 27362965, Registered in The Netherlands

    Privacy Notice      |      Website Terms      |       Manage Preferences

    Copyright © 2023. All rights reserved.
    Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16 2333 CL Leiden, The Netherlands

    Reg No: 27362965, Registered in The Netherlands

    Privacy Notice      |      Website Terms      |       Manage Preferences

    How Important Is The Matrix Effect in Analyzing Bioprocess Samples?

    How Important Is The Matrix Effect in Analyzing Bioprocess Samples?

    Batavia Biosciences - Favicon

    The matrix effect is the effect on an analytical assay caused by all other sample components except the specific compound (analyte) to be analyzed.

    Matrix effects are observed either as a loss in response, resulting in an underestimation of the amount of analyte or an increase in response, producing an overestimated result. These effects have long been associated with bioanalytical techniques. However, their evaluation for each assay and each sample matrix can be very time-consuming. Also, these extra evaluation steps bring additional costs.

    So, how important is evaluating the matrix effects when analyzing bioprocess samples? What are the best approaches to evaluate the effect? And in what situations is the matrix effect acceptable?

    The Importance of Matrix Effect Evaluation

    To release final product for use in patients, the product undergoes different quantitative or quantitative analytical procedures. Based on the obtained data, a decision is made to approve or reject the product batch. Therefore, showing that generated analytical data is accurate and no matrix effect is involved is necessary. During the assay development and validation, the impact of the sample matrix is investigated. Each assay is developed to allow monitoring of the matrix effect and, if possible, eliminate it.

    Methods to Evaluate and Eliminate

    A few quick, qualitative options exist to determine whether a matrix effect is present, such as the dilution-based method. However, this article will focus on a few quantitative methods often used for bioanalytical techniques.

    Signal-Based Method

    This method allows for quantifying the matrix effect for one specific concentration.

    At this concentration, the analyte is measured in the matrix and subsequently measured in a solvent known not to induce any effect. The analyte signal in the matrix is then divided by the analyte signal in the solvent and multiplied by a hundred, resulting in the percentage of matrix effect. If the percentage is below a hundred, the matrix effect results in a suppression of the result. When it is above a hundred, it causes enhancement of the result.

    This method is useful when only this concentration is relevant. It doesn’t necessarily provide any indication of other analyte concentrations.

    Concentration-Based Method

    In this method, the matrix effect is measured with the signal-based method but for a range of analyte concentrations. The concentration-based method is used to show that the matrix effect is not analyte concentration dependent.

    Calibration-Based Method

    This method is particularly relevant when a blank matrix is not available. Different analyte concentrations are measured in solvent and the matrix, and obtained data are plotted in a graph, and linear regression model is used to generate a slope value.

    The slope of calibration analyzed in the matrix is then divided by the slope of the calibration curve prepared in the solvent. The ratio is then multiplied by a hundred to generate %ME. For %ME >100% matrix results in overestimation, and for %ME < 100% tested matrix leads to signal suppression.

    When a matrix effect is present, the effect-causing component must be removed from the sample before analysis. Unfortunately, this is not always an option.

    In those cases,  matrix minimization (dilution) will provide a way forward. Matrix minimization is especially useful when the analytical technique has sensitivity to spare. Eliminating matrix effect for the particular matrix is proved during the assay development or validation.

    When Can You Ignore Matrix Effect?

    Theoretically, samples consisting a pure compound could be ignored for matrix testing. However, even a supposedly pure compound may contain other elements in some cases. Elements such as reaction impurities or by-products may lead to matrix effects. Especially during process development, it is challenging to remove the matrix effect. This is due to the large number of matrices generated at each step of upstream or downstream process development.

    Analysis of PD samples usually serves to monitor processes and provide process developers with an indication of whether changes in certain process parameters have beneficial or undesirable consequences. As the absolute value of the analysis is less important during the process development compared to batch release analysis, matrix effects are often not completely removed. Instead, they are only monitored using a spike recovery approach. This approach enables sample analysis with sufficient information on a potential matrix effect while saving time and resources.

    What are examples of matrix effects?

    Matrix effect is predominantly observed in mass spectrometry when there’s a suppression or enhancement of the ionization efficiency of the analyte due to the presence of other compounds in the sample. 

    In bioprocessing, several factors can influence and potentially interfere with the accurate detection and quantification of specific proteins or molecules. The presence of salts, lipids, or other organic compounds can mask or distort the detection of certain proteins, and the addition of detergents or buffer components present in a sample can introduce interference during protein quantification, further complicating the process. During the chromatography process, compounds that co-elute may affect the precise quantification of the analyte in focus.

    What causes the matrix effect?

    The matrix effect arises from a myriad of factors. Primarily, other components in the sample can vie for ionization, potentially overshadowing or altering the analyte’s presence. This competition is further intensified by co-eluting compounds, which can either suppress or amplify the ionization of the analyte. Notably, even subtle changes in the sample’s pH can influence the ionization potential of the analyte, affecting detection and quantification. 

    Additionally, chemical interplays between the analyte and the matrix components can introduce deviations. Beyond these chemical influences, disparities in physical properties, like volatility or polarity, between the matrix and the analyte can also contribute to the matrix effect.

    What is a matrix effect and how might it impact your results?

    A matrix effect describes the changes observed in the detection or quantification of an analyte when other substances are present in the sample. The implications of this phenomenon are manifold. For one, the actual concentration of the analyte may not be accurately represented, leading to reduced precision in results. This is further complicated by decreased sensitivity, where the presence of matrix components can lower the detection limits for the analyte.

    Moreover, even if the analyte concentration remains steady, variations in the matrix composition can cause inconsistency between samples, adding to the variability. Additionally, the interference from these matrix components can sometimes result in false positives or negatives, further skewing the detection of the analyte.

    How do you determine the matrix effect?

    To ascertain the matrix effect, several methods can be employed. One common approach is the Spike and Recovery Method, where a predetermined quantity of the analyte is added to the sample matrix. The concentration is then measured, and the observed value is compared with what was anticipated.

    Another technique is post-extraction spiking, which involves comparing the response of an analyte that’s introduced into a sample post-extraction to the response observed when the analyte is added to a pure solvent. Additionally, matrix-matched calibration can be utilized. In this method, the calibration curve derived from an analyte in a matrix-free solvent is juxtaposed against a curve where the standards are calibrated within a sample matrix.

    Lastly, a blank matrix analysis can be conducted to scrutinize a sample matrix devoid of the analyte, helping identify potential interference or background signals.

    How do you avoid the matrix effect?

    To mitigate or altogether sidestep the matrix effect, several strategies can be employed.

    One fundamental approach is sample purification, where methods like solid-phase extraction (SPE) or liquid-liquid extraction (LLE) are deployed to refine samples. Additionally, simply diluting samples can help diminish the concentration of interfering matrix components. Further precision can be attained by optimizing chromatographic conditions; tweaking parameters ensures that the analyte doesn’t co-elute with any interfering substances.

    Incorporating internal standards can also prove invaluable, as they assist in rectifying variations brought on by the matrix effect. Equally crucial is instrument optimization, where fine-tuning instrument settings can substantially reduce matrix interference. It’s important to note that while it might be challenging to entirely eliminate the matrix effect, a comprehensive understanding and proactive management can significantly bolster the accuracy and reliability of results.

    Related

    Adenovirus Vectors in Gene Therapy – Vaccine Development & Delivery Tools

    Adenovirus Vectors in Gene Therapy – Vaccine Development & Delivery Tools

    Adenovirus Vectors in Gene Therapy – Vaccine Development & Delivery Tools

    Batavia Biosciences - Favicon

    Adenovirus  vectors are a form of viral vectors which are used to develop gene therapy products and vaccines. Decades of research stand behind these vectors which have gone through several iterations and improvements to make them what they are now.

    A History of Adenovirus Vectors

    As mentioned in a previous article, the E1 proteins which control the adenoviral replication cycle were removed in very early iterations of the adenovirus vectors. This prevented the virus from replicating in humans, thus only allowing the virus to deliver its genetic payload. In addition, the E3 region, which is not essential for viral propagation, was removed. The space freed up in the wildtype virus genome by the removal of E1 and E3 allowed for the insertion of up to 6.5 kb of foreign DNA.

    This was the first generation of adenovirus vectors.

    In the second generation more improvements were made when E2a, E2b and E4 were removed. The removal freed up yet more space in the genome for up to 10.5 kb of transgene DNA to be inserted into the chromosome. This was particularly relevant for researchers desiring to put multiple antigens into one vector. For instance, in pursuit of AIDS vaccines it was shown that a combination of HIV virus-derived envelope together with Gag and Pol provided broad immune responses. The deletion of more genes encoding for E proteins from the viral backbone also improved the safety of Ad vectors by making it less likely that spontaneous recombination events during the vector propagations would lead to replication-competent viral particles. The deletion of multiple E-encoding genes from the viral genome also led to a significant reduction in viral gene expression in host cells, lowering a cytotoxic T-lymphocyte response against the vector itself. This made second generation Ad vectors much less likely to be cleared by the immune system.

    Third generation Ad vectors are void of all viral sequences except for the inverted terminal repeats, which are the signals for the DNA to be effectively packaged into the adenoviral capsid proteins. These high-capacity adenovirus vectors can package up to 36 kb of foreign genetic material. To produce these vectors, adenoviral helper viruses are required to produce the proteins needed for replication and packaging of the Ad vector genome.

    Recombinant Ad vectors have been around for a long time and the active research around them has led to great leaps forward in their usage and in the fields of gene therapy and viral vaccine production. Currently, it is possible to make conditionally replicating Ad vectors, which only replicate inside tumor cells. When this is combined with the effective cell targeting provided by transgenic knob proteins on the capsid surface, Ad vectors become a truly powerful tool for cancer therapies as well.

    Main Advantages for Adenovirus Vectors

    Ad vectors have advanced further than any other vector system owing to continued active research from academia and industry together. This research has made Ad vectors more effective while also making them safe. Modern Ad vectors have four key advantages in gene therapy and vaccine development.

    1. High Transduction Efficiency in Dividing and Quiescent Cells

    This is one of the reasons that adenoviruses were first considered as vectors for gene therapy and vaccines. Ad vectors can deliver genetic cargo to cells very efficiently, so that therapeutic levels can be achieved with fewer viral particles. This is extremely important for in vivo systemic applications where high concentrations of Ad vector are typically more challenging.

    2. Epichromosomal Persistence in Cells

    This feature of Ad vector has led to some very specific and highly sought-after applications. Persistence of the vector is important to allow the genetic payload to be delivered, transcribed, and expressed as therapeutic proteins. Without the ability to persist, these therapies would not be present long enough to be effective. However, a common mode of persistence for viruses is chromosomal integration. This is often undesirable or unnecessary in gene therapy and carries many safety concerns specifically for DNA vectors. Because Ad vectors do not integrate into the chromosome of the host cells, there is no risk that they will permanently alter the host genetic make-up.

    3. Broad Tissue Tropism

    A wide variety of wild-type adenoviruses have been modified to create Ad vectors, providing a range of different tissue tropisms. More specific tropism has been developed in many therapeutic cases thanks to the genetic manipulation of the knob protein, which is the protruding end of the fiber capsid that is primarily responsible for host cell attachment. This protein is the key to unlock the entry into specific cells through receptor-specific binding. By modifying the knob protein, it is possible to make Ad vectors that can be precisely targeted to specific tissues.

    4. Scalable Production

    Gene therapy products that cannot be scaled to commercial manufacture cannot be used to treat real-world diseases. As we saw during the rollout of the COVID-19 vaccines, Ad vector-based products can be rapidly scaled to meet market demand. To meet demand for the COVID-19 vaccine, each 5-day production cycle readily delivered over 15 million doses of vaccine to battle SARS-CoV2 virus.

    Together, the described benefits of Ad vectors provide a broad and versatile platform to achieve sought-after clinical breakthroughs in the treatment and prevention of debilitating and life-threatening diseases

    Clinical applications

    Clinical applications of Ad vectors can be broadly split into two categories: vaccines and gene therapies.

    Vaccines

    Basic research in pursuit of improved Ad vectors revealed that the high prevalence of wild-type adenoviruses in the human population has led to widespread pre-existing immunity, particularly to some human serotypes like Adenovirus serotype 5. Developments in the field have turned this potential drawback into an important feature of Ad vector-based vaccines.

    With respect to vaccine development, the main purpose of a DNA-based vector is to deliver epitopes from other viruses to host cells and to ensure production of such epitopes in order to raise an broad and effective immune response against the virus. Over the iterations of development, Ad vectors have become better at delivering increasingly large nucleic acid payloads. In addition, the immunogenicity of some Ad vectors has been tweaked and harnessed to boost the production of pro-inflammatory cytokines that can enhance the humoral and cellular immune responses.

    A successful application of this approach is the Ad vector-based Ebola vaccine developed by Janssen. This vaccine, which takes advantage of Janssen’s rare Ad26 serotype, induces specific antibody and T-cell responses against Ebola virus. Clinical results demonstrated that the vaccine elicits a powerful humoral immune response in humans that persists for more than a year. Rapid and durable T-cell responses using Ad26 were also seen in adenovector-based COVID-19 vaccines, which induced strong humoral and cellular immune responses in 100% of clinical trial participants after two doses.

    Ad vectors have also been widely used for the production of cancer vaccines. Current vaccine research is focused on prostate cancer, human papilloma virus, colorectal, and pancreatic cancers. In addition to cancer prevention through vaccines, Ad vectors are also used for anticancer therapies.

    Oncolytic Virotherapy

    Anticancer therapies using adenovirus vectors fall broadly into three main categories:

    1. Delivery of Suicide Genes

    Many tumor cell types proliferate rapidly and have a dysfunctional p53 tumor suppressor pathway. Ad vectors can be engineered to induce p53 expression inside tumors, triggering cell death. Another successful application has been using Ad vectors to deliver genes that convert a pro-drug into an active cytotoxic agent. For example, the enzyme purine nucleoside phosphorylase converts the pro-drug fludarabine monophosphate into fluoroadenine, which kills proliferating cells. Trials using this enzyme have already been conducted using Ad vectors. The ability of Ad vectors to remain localized is extremely valuable when delivering cytotoxic therapies that can harm heathy cells if the treatment is not constrained to the target cell populations.

    2. Delivery of Immune-Regulatory Genes

    Ad vectors can also be loaded with genes that stimulate an antitumor immune response. Antitumor interferon-β and interferon-α-2b have both been safely delivered to the lungs of patients via intrapleural injection.

    3. Chimeric and Tropism-Modified Oncolytic Adenovirus Vectors

    A problem that often arises during cancer therapies is poor recognition of tumors by immune cells and Ad vectors. In this case, Ad vector knob proteins can be modified to bind more strongly to receptors on the surface of the tumor. One example of this is in an Ad vector used to treat ovarian cancer where the entire fiber knob domain of Ad5 was replaced with that of Ad3. The result was targeted ablation of ovarian cancer cells displaying elevated levels of Ad3 receptors.

    Adenoviral vectors have come a long way since their initial use several decades ago. Research has shown that they can be used extremely effectively for a number of gene therapy and vaccine applications.

    What are the advantages of adenovirus vector in gene therapy?

    Adenovirus vectors offer several advantages in gene therapy.

    Their large genome allows them to carry significant genetic material, enabling the delivery of larger therapeutic genes, unlike retroviruses, they don’t integrate into the host genome, reducing the risk of insertional mutagenesis. Adenoviruses can transduce both dividing and non-dividing cells, broadening their applicability and elicit a robust immune response, which can be beneficial in vaccine applications.

    Is adenovirus gene therapy approved?

    Adenovirus-based gene therapies have received approval in specific contexts. One of the most notable approvals is Glybera, which was the first gene therapy approved in the Western world for a rare condition called lipoprotein lipase deficiency (LPLD), approved by the European Medicines Agency (EMA) in 2012, although its high cost and limited demand led to its withdrawal from the market.

    Additionally, adenovirus vectors have been explored in various other therapeutic and vaccine applications, with some progressing through clinical trials and gaining approval. It’s essential to check current regulatory databases for the most up-to-date information on approvals.

    What is the difference between adenovirus and AAV?

    Adenoviruses and adeno-associated viruses (AAVs) are distinct yet central vectors in gene therapy.

    Adenoviruses, from the Adenoviridae family, have double-stranded DNA and can cause mild human infections, whereas AAVs – from the Parvoviridae family – are smaller, with single-stranded DNA, and are non-pathogenic. While adenoviruses don’t integrate into the host genome, reducing mutagenesis risks, AAVs occasionally do, often targeting a specific chromosome site. Adenoviruses carry larger genetic payloads and elicit strong immune responses, making them suitable for cancer therapies and vaccines.

    Related

    Taking your adenoviral vector product from R&D to the clinic

    Taking your adenoviral vector product from R&D to the clinic

    Taking your adenoviral vector product from R&D to the clinic

    Batavia Biosciences - Favicon
    Nicky Veringmeier

    Nicky Veringmeier

    Marketing

    Your content goes here. Edit or remove this text inline or in the module Content settings.

    You can also style every aspect of this content in the module Design settings and even apply custom CSS to this text in the module Advanced settings.

    Adenoviruses have been shown to be very effective vehicles for delivering genetic material to cells and are therefore in demand to develop gene therapies and vaccine products. The road from early-stage R&D to final production of an adenoviral vector product is well trodden, and there is a lot of detailed information out there to help you on your journey.

    That said, taking an adenoviral product to the clinic always requires some process development prior to bringing the vector into GMP. This is based on the many examples in practice where the transgene had detrimental effects on vector yield or purity once inserted into the vector. For instance, the foreign gene may encode for a protein that interferes with HEK293 cell propagation, resulting in low yield of vector particles per cell. Likewise, the protein produced may bind to the Adenoviral vector capsid proteins, which are generally carry a net negative charge that can substantially affect the purification strategy.  A good way of mitigating risk is to seek expertise at the relevant stages of development to fill informational gaps and reduce the possibility of unexpected problems while moving from bench to clinic.

    Working with a center of excellence contract development and manufacturing organization (CDMO) is an attractive option for many who embark on the adenoviral production journey. Other than a general vendor type CDMO, A center of excellence CDMO has in-depth experience and can help guide the process by providing practical solutions to challenges that may pop up.   If you are developing a therapy from the very beginning, a “center of excellence CDMO” can often complement your expertise and help you navigate the complexities of the scale-up process.

    In this article, we highlight how a “center of excellence CDMO” can help you to smoothly navigate the transitions from R&D to scale-up and clinical manufacturing. The information here will provide you with a stable foundation when you start a conversation with a CDMO, allowing you to identify important traits that will give you a strong relationship going forward.

    Common challenges that a CDMO can overcome

    Concentration challenges

    One of the most important considerations that need to be made early in the product development trajectory is the concentration of the vector needed to be an effective therapy in the clinic (part of Target Product Profile setting).

    For adenovector based vaccines, a dose of 1010-1011 virus particles per milliliter typically suffices for systemic administration i.e. intramuscular injection. For oncolytic therapies, either higher or lower doses could be required, depending on the route of administration and the target tissue. For instance, treatment of a wound-bed post cancer surgery may require doses of up to 1013 virus particles per milliliter to effectively deliver a high dose in a confined location with minimal spill-over to surrounding tissue.

    At such high vector concentrations, aggregation of virus particles may occur, which reduces therapeutic efficacy. The high concentrations needed for many oncolytic and gene therapies exacerbate this problem. Aggregation can manifest itself in quite spectacular fashion, as concentrated adenovirus preparations become cloudy before “snow” appears on the bottom of the vial. Understanding how adenovirus particles behave and good forward planning are essential to reduce the chances of aggregation.

    On the other end of the spectrum, there is also a risk that during the scale-up process the concentration step will not be sufficiently effective, leading to viral titers that are lower than expected. This is a critical issue that can arise due to the significant alterations that are often required during scale-up of concentration steps, such as switching from ultracentrifugation to chromatography.

    Since the stakes and capital investment are always higher the further down the scale-up path you are, it pays to address any issues relating to concentration as early as possible.

    Release of intracellular DNA

    Eukaryotic cells have to be lysed in order to release the adenoviruses, and in the process the release of intracellular DNA is inevitable. The more viral particles needed, the greater the number of cells that must be lysed, and therefore the more host cell DNA is released. Unfortunately, DNA often forms an intricate and sticky web that easily traps adenovirus particles, causing them to aggregate. As discussed in the previous paragraph, adenoviruses are already prone to aggregation, so adding DNA into the mix only increases the risk. The DNA webs also are notorious for clogging filters, which further hampers the purification process. These problems can be circumvented in the process development stages by adding a DNA reduction step such as the introduction of benzonase. With careful quality control, interventions like this can mitigate the negative effects of extracellular DNA.

    Process yield & scalability

    Perhaps the biggest consideration when manufacturing Ad vectors is the process yield at large scales. In the R&D phase, simply ensuring there is “enough” virus to perform preclinical experiments is acceptable. However, production at larger scales inherently leads to larger losses. During the R&D phase it is therefore essential to prioritize yield. This can be done by reducing the number of empty capsids that are present in your viral preparation and ensuring that your process is ready for the switch to large scale production. This often requires changing from ultracentrifugation, which is a common way to concentrate adenovirus in the research lab but not appropriate for scaling. With technologies used for scale up such as chromatography, it is often difficult to achieve the same yield as ultracentrifugation which is why process development is so important.

    How does a CDMO help?

    Understanding the different applications for adenoviral vectors is a good way of highlighting how a good relationship with a center of excellence CDMO should work. The center of excellence will have industry experts who can save you a lot of time and expense in developing robust and scalable manufacturing processes for your vector. For example, at Batavia, our in-house experts have extensive experience in developing adenoviral vector-based therapies be it vaccines or oncolytic products. These experts are deployed to relevant projects to help guide the strategy and ensure the highest likelihood of manufacturing success.

    Clear communication is a must for a good center of excellence CDMO. Clearly articulating the challenges and opportunities of a scale-up project is the only way to make effective progress. While good communication may seem less relevant than the technical capabilities of a prospective CDMO, it is nevertheless important and very easy to assess early on your interactions. Pay close attention to how your CDMO communicates. Make sure that misunderstandings are quickly resolved, and that communication is clear and timely. The way a CDMO communicates and structures their meeting and interactions is a reflection of how they operate, so if you are not satisfied, it’s best to move on.

    A good CDMO should bring balance and clarity to the scale-up project. When you partner with a center of excellence CDMO, you should feel confident that you are in good hands. You should also feel that you acutely understand the process going forward, including the potential risks.

    At some point when moving from R&D to scale-up and production, you will meet a whole range of different regulations that have to be thoroughly studied and strictly adhered to, if your adenovirus vector is to make it successfully into the clinic. A good center of excellence CDMO has experience with the relevant and most up-to-date regulations and requirements, and will help you navigate the complex landscape of rules and paperwork. Always ask about track record.

    Choosing a CDMO

    If you decide that working with a center of excellence CDMO is the right path for you, the next question is who should you choose? Fortunately, the CDMO field is competitive, and you do have a choice. It’s important that you exercise this choice and do your due diligence by thoroughly researching and interviewing your CDMO candidates. Pay close attention to the way they communicate with you and how they discuss your project and bring up challenges. Most importantly pay close attention to the quotation and make sure to read the small footnotes to understand what is included in price and what is excluded and needs to be paid additionally.

    It’s never too early to begin discussions with a partner to help you scale your adenoviral vector product. Even informal conversations early on can give you insights and help you make decisions that will have a positive impact on the future of your project. To start with, why not book a call with one of our experts. We’re looking forward to hearing about your project and you can be sure that we’ll give you clear advice and an all-included costing overview for your project.

    Related

    Your content goes here. Edit or remove this text inline or in the module Content settings. You can also style every aspect of this content in the module Design settings and even apply custom CSS to this text in the module Advanced settings.

    Low-cost viral vector manufacturing

    High-throughput screening for viral vectors

    Viral vector manufacturing

    Thermostable viral envelope protein formulation

    Maximizing protein expression