Skip to main content

Article Filters

Part two: Has lentiviral purification been left behind?

Published date: 12 December 2023

Back to Article Listing

In our last blog we looked at why lab-scale lentiviral production is crucial to gene and cell therapy development and how do lentivirus characteristics impact purification strategies. Today we add to this, with commentary from Sujeong Yang and Ian Scanlon on the limitations of existing lab-scale purification approaches.

What are the limitations of existing lab-scale purification approaches?

Sujeong Yang: Ultracentrifugation and density gradient can both purify and concentrate LVVs. However, these have limitations including a lack of scalability, the need for high-powered centrifuges and special operational expertise. Additionally, the process is often time-consuming which limits throughput.

Ian Scanlon: There is no standard downstream purification process for these smaller volumes. Molecular weight cut off filters can be used with benchtop centrifuges to concentrate clarified feedstocks but have limited capability to remove impurities. The same considerations apply for tangential flow filtration, but additionally the LVV is concentrated along with salts, proteins, and DNA, generally for extended periods due to the length of time required for concentration.

Precipitation is sometimes used to selectively remove impurities. PEGylation of the vector can enable selective product concentration, for instance. The problem here is that, once again, adding any additional unit operations with LVVs typically reduces the final yield of product considerably.

Alternatively, ion exchange membrane adsorbers allow the purification process to be more aligned with chromatography steps commonly used at clinical scale LVV manufacturing. However, functional product recovery is compromised using high salt elution steps.

What would be the impact of improved lab-scale lentiviral vector purification on therapeutic development?

Ian Scanlon: When comparing LVV discovery workflows to those applied in protein and antibody drug discovery, it is clear there is much room for improvement. The latter leverages high-throughput technologies with multi-well plates for rapid separation and analysis. It would be highly beneficial to truly transfer that approach to the LVV space.

Sujeong Yang: Although there are upstream technologies that can work in parallel to produce vector targets, the capabilities of existing purification solutions restrict the throughput of LVV purification for use in preclinical studies, and therefore extends the time needed to obtain material representative of that required for clinical and commercial applications. A more efficient purification process would shorten the vector development time, so reducing costs, and allow therapies to progress faster to clinical testing.

How could a lab-scale chromatography solution facilitate lentiviral therapeutic development?

Sujeong Yang: The process must be easy to implement with equipment generally available in the lab. Many LVV purification methods that can provide pure, highly functional material are not very practical. The process should also be adapted to reduce LVV degradation. For example, using mild elution conditions to avoid the degradation seen at high salt concentrations, and a short process time should boost LVV recovery. Increased product recovery would increase production efficiency and reduce costs, so reducing the pressure on the upstream process to increase volumes of viral feedstocks.

Ian Scanlon: It would also need to increase throughput so researchers could advance more targets and assets that are likely to succeed through the drug development process faster, ultimately providing a greater number of therapies in the hands of patients.

A chromatography-based purification technology that could be deployed simply and easily, be readily scalable, and afford robust LVV purifications in a rapid manner would be the best solution.

Astrea Bioseparations is addressing this need through our development of an unprecedented and proprietary fiber-based technology, empowering therapeutic innovators with the tools they need to purify the quantities they desire.

Your basket

Your basket is empty. Continue shopping to add products to your basket.

Search our catalogue

Call Centre

Request Quote

Please enter the quantity of products you want a quote for

Please enter the date you need these products byPlease enter a date in the format DD/MM/YYYY

View Content

Please complete the form below to receive the requested content from Astrea Bioseparations. We will use the data you provide to send you relevant updates, special offers, and product-related information. See our Privacy Notice for details on how we use personal data.

If you have an account please log in to gain access to content. You can apply for an account here

Please enter a First Name

Please enter a valid First Name, the maximum length is 50 characters.

Please enter a Last Name

Please enter a valid Last Name, the maximum length is 50 characters.

Please enter a Company Name

Please enter a valid Company, the maximum length is 100 characters

Please enter a valid Email AddressPlease enter a valid Email AddressThe Email Address entered is already registered, please sign in with the Email Address or enter a different one

Please select a valid Telephone Number, the maximum length is 30 characters

Please enter a valid Telephone Number consisting only of the following characters and spaces ( ) + 0 1 2 3 4 5 6 7 8 9

Please select a State / Province / Territory

Please select a valid State / Province / Territory, the maximum length is 150 characters

Please select a Country

Data entered into form will be stored in your session (not in a cookie) to avoid having to re-enter in future form submissions.

Part two: Has lentiviral purification been left behind?

Published date: 12 December 2023

Back to Article Listing

In our last blog we looked at why lab-scale lentiviral production is crucial to gene and cell therapy development and how do lentivirus characteristics impact purification strategies. Today we add to this, with commentary from Sujeong Yang and Ian Scanlon on the limitations of existing lab-scale purification approaches.

What are the limitations of existing lab-scale purification approaches?

Sujeong Yang: Ultracentrifugation and density gradient can both purify and concentrate LVVs. However, these have limitations including a lack of scalability, the need for high-powered centrifuges and special operational expertise. Additionally, the process is often time-consuming which limits throughput.

Ian Scanlon: There is no standard downstream purification process for these smaller volumes. Molecular weight cut off filters can be used with benchtop centrifuges to concentrate clarified feedstocks but have limited capability to remove impurities. The same considerations apply for tangential flow filtration, but additionally the LVV is concentrated along with salts, proteins, and DNA, generally for extended periods due to the length of time required for concentration.

Precipitation is sometimes used to selectively remove impurities. PEGylation of the vector can enable selective product concentration, for instance. The problem here is that, once again, adding any additional unit operations with LVVs typically reduces the final yield of product considerably.

Alternatively, ion exchange membrane adsorbers allow the purification process to be more aligned with chromatography steps commonly used at clinical scale LVV manufacturing. However, functional product recovery is compromised using high salt elution steps.

What would be the impact of improved lab-scale lentiviral vector purification on therapeutic development?

Ian Scanlon: When comparing LVV discovery workflows to those applied in protein and antibody drug discovery, it is clear there is much room for improvement. The latter leverages high-throughput technologies with multi-well plates for rapid separation and analysis. It would be highly beneficial to truly transfer that approach to the LVV space.

Sujeong Yang: Although there are upstream technologies that can work in parallel to produce vector targets, the capabilities of existing purification solutions restrict the throughput of LVV purification for use in preclinical studies, and therefore extends the time needed to obtain material representative of that required for clinical and commercial applications. A more efficient purification process would shorten the vector development time, so reducing costs, and allow therapies to progress faster to clinical testing.

How could a lab-scale chromatography solution facilitate lentiviral therapeutic development?

Sujeong Yang: The process must be easy to implement with equipment generally available in the lab. Many LVV purification methods that can provide pure, highly functional material are not very practical. The process should also be adapted to reduce LVV degradation. For example, using mild elution conditions to avoid the degradation seen at high salt concentrations, and a short process time should boost LVV recovery. Increased product recovery would increase production efficiency and reduce costs, so reducing the pressure on the upstream process to increase volumes of viral feedstocks.

Ian Scanlon: It would also need to increase throughput so researchers could advance more targets and assets that are likely to succeed through the drug development process faster, ultimately providing a greater number of therapies in the hands of patients.

A chromatography-based purification technology that could be deployed simply and easily, be readily scalable, and afford robust LVV purifications in a rapid manner would be the best solution.

Astrea Bioseparations is addressing this need through our development of an unprecedented and proprietary fiber-based technology, empowering therapeutic innovators with the tools they need to purify the quantities they desire.

Call Centre Product Compare