2021 BioMAN Summit

This year’s 2-day virtual Summit will bring together thought leaders from industry, academia, and government who are working to meet the novel scientific and technological challenges of expanding the reach of cell and gene therapies. In a series of invited presentations, we will:

Identify

what innovative cell and gene therapies are advancing through the development pipeline in the immuno-oncology space and across other disease indications.

Explore

what the cell and gene therapy ‘factory of the future’ looks like and how it will meet the technical and logistical challenges of scaled-up or scaled-out manufacturing.

Discover

what novel technologies can address current cell and gene therapy bottlenecks such as supply and variability of raw materials.

Examine

how analytics can improve process development and manufacturing and decrease the cost of goods for these modalities.

Investigate

strategies for introducing new technologies in the face of compressed development timelines and accelerated product approvals.

SPEAKERS

Peter Marks
plus

Peter Marks

United States Food and Drug Administration

Richard D. Braatz
plus

Richard D. Braatz

MIT

Krystyn Van Vliet
plus

Krystyn Van Vliet

MIT

Michael Birnbaum
plus

Michael Birnbaum

MIT

Stacy L. Springs
plus

Stacy L. Springs

MIT

Jacqueline M. Wolfrum
plus

Jacqueline M. Wolfrum

MIT

James C. Leung
plus

James C. Leung

MIT

Anthony J. Sinskey
plus

Anthony J. Sinskey

MIT

PROGRAM

Click HERE for a downloadable copy of the event agenda, and HERE for a copy of the Summit participant list!

Summit Welcome, Introduction & Framing of the Summit

30 minutes

Jacqueline M. Wolfrum

Dr. Wolfrum has been at the MIT Center for Biomedical Innovation (MIT CBI), since 2014. She is Director of the Biomanufacturing Program (BioMAN), a pre-competitive biopharmaceutical industry consortium focused on development of new knowledge, science, technologies, and strategies that advance the manufacture and global delivery of high-quality biologic medicines. She manages sponsored projects and activities […]

Stacy L. Springs

Dr. Stacy Springs is the Executive Director at the MIT Center for Biomedical Innovation (CBI). The Center integrates the Institute’s technical, scientific, and management expertise to solve complex biopharmaceutical challenges. CBI leads multi-stakeholder, multidisciplinary research and educational initiatives with real world impact, including MIT’s Biomanufacturing Consortium, (BioMAN), and it’s Consortium on Adventitious Agent Contamination in […]

Anthony J. Sinskey

Anthony J. Sinskey, Sc.D., is a Professor of Microbiology at the Massachusetts Institute of Technology. He also holds positions as Co-Director of the Malaysia-MIT Biotechnology Partnership Program and as Faculty Director of the MIT Center for Biomedical Innovation. He conducts interdisciplinary research in metabolic engineering focusing on the fundamental physiology, biochemistry and molecular genetics of […]

Keynote Presentation: The Importance of Manufacturing in Cell and Gene Therapy Development

The number of approved cell and gene therapy products in the United States continues to increase. However, further accelerating progress in the field will depend heavily on advancing manufacturing technologies for these products. In particular, the production of high-quality therapy gene therapy vectors used for very small populations remains quite challenging. The use of process automation, including in-process controls, in a similar manner to that which is now starting to be applied to chimeric antigen receptor T cell production may facilitate the production of gene therapy at notably reduced cost compared with current gene therapy manufacturing technologies. Application of such manufacturing technology could be transformational in facilitating the delivery of gene therapy to those in need.

30 minutes

Peter Marks

Peter Marks received his graduate degree in cell and molecular biology and his medical degree at New York University and completed Internal Medicine residency and Hematology/Medical Oncology training at Brigham and Women’s Hospital in Boston. He has worked in academic settings teaching and caring for patients and in industry on drug development.  He joined the […]

Keynote Presentation: Viral vectors – A manufacturer’s wish list

Gene delivery using Adeno- Associated Viruses has become the go to method for development for novel gene therapies. Numerous innovations have improved tissue targeting and gene expression, but innovations in the core AAV manufacturing process and in the characterization of these complex products are needed to improve productivity, consistency and enable processes to produce safer and more effective AAV products. I will outline a typical AAV manufacturing process and test methods and highlight areas where improvements and innovative solutions are needed.

30 minutes

Andrew Topping

Dr. Topping has worked in contract development and manufacture of biologics for almost 30 years, both from microbial expression systems and from mammalian cells. He trained as a molecular biologist with a Ph.D. from the University of Wales in Cardiff and have led development and manufacturing projects bringing over 100 protein products into the clinic. […]

Refreshment Break

30 minutes

The potential of using crystallization for the separation of full and empty capsids

Purification of genome-loaded recombinant adeno-associated viral (full rAAV) capsids from empty capsids is challenging. The current purification technology has low yields (typically ~50%). This presentation discusses the potential of using crystallization for the separation of full and empty capsids, with the objective of achieving >90% yield. We propose an efficient method for purification of full rAAVs based on preferential crystallization. We have thoroughly mapped out the crystallization phase diagrams for full and empty rAAV capsid, which shows very large crystal nucleation and growth regions as a function of pH and precipitant concentrations. The phase diagrams and kinetic analyses for multiple serotypes indicate that preferential crystallization is feasible.

30 minutes

Richard D. Braatz

Dr. Richard D. Braatz is the Edwin R. Gilliland Professor of Chemical Engineering at MIT, where he conducts research into advanced biopharmaceutical manufacturing systems. In this role, he leads process data analytics, mechanistic modeling, and control systems for many projects, including on monoclonal antibody, viral vaccine, and gene therapy manufacturing within the Center of Biomedical […]

Vivekananda Bal

Vivekananda Bal received his combined undergraduate degree in Chemistry and Chemical Engineering, and masters degree in Chemical Engineering from the University of Calcutta, India and Indian Institute of Technology, Kanpur, India, respectively. He then completed Ph.D. in Chemical Engineering from Indian Institute of Technology Bombay and worked in the area of Computation Fluid Dynamics, reactor […]

Overcoming CRISPR delivery challenges using high capacity baculovirus vectors in human cells – Virtual

CRISPR-based precise gene-editing requires simultaneous delivery of multiple components into living cells, rapidly exceeding the cargo capacity of traditional viral vector systems. This challenge represents a major roadblock to genome editing applications. Delivery of complex gene editing approaches for knock-in, base editing or prime editing is nowadays typically achieved by deploying multiple viral vectors, severely limiting the rate of successful gene editing events and imposing and increasing burden on manufacturing processes. By contrast, baculovirus vectors (BVs) are large dsDNA insect-specific viruses which can efficiently transduce a range of mammalian and human cell lines, while remaining replication and integration deficient in the mammalian hosts. BVs can accommodate >30 kb synthetic DNA cargo and, backed by a novel hybrid DNA assembly system, we demonstrate successful delivery of multifunctional CRISPR toolkits in human cells. By encoding Cas9, sgRNA and Donor DNAs on a single, rapidly assembled baculoviral vector, we achieve with up to 30% efficacy whole-exon replacement in the intronic β-actin (ACTB) locus, including site-specific docking of very large DNA payloads. We use our approach to rescue wild-type podocin expression in steroid-resistant nephrotic syndrome (SRNS) patient derived podocytes. We demonstrate single baculovirus vectored delivery of single and multiplexed prime-editing toolkits, achieving up to 100% cleavage-free DNA search-and-replace interventions without detectable indels. Taken together, we provide a versatile delivery platform for single base to multi-gene level genome interventions, addressing the currently unmet need for a powerful delivery system accommodating current and future CRISPR technologies without the burden of limited cargo capacity.

30 minutes

Francesco Aulicino

Dr Francesco Aulicino is a senior post-doctoral research associate at University of Bristol (UK), in the group of Prof. Imre Berger. Francesco is a molecular and cellular biologist by training (B.Sc., M.Sc., University of Pisa, Italy) and holds a Ph.D. in Biomedicine (UPF, Barcelona, Spain). During his Ph.D., he investigated the role of Wnt/β-catenin signalling […]

Engineering a pseudotyped lentiviral platform to enable lineage-specific transduction of immune cells

Cell-specific transduction remains one of the next frontiers for virally-delivered gene therapy. Efforts to achieve cell-specific transduction have largely been limited to borrowing of preexisting viral glycoproteins and pseudotyping viral surface envelopes to result in altered tropism. Our lab recently developed a “receptor-blinded” version of VSVG, which we call “VSVGmut”, enabling co-display of a new LV pseudotype ligand to drive specific lentiviral tropism. Initial experiments have shown modularity of this platform for achieving potent transduction of on-target cells via a range of co-expressed host proteins, with minimal infection of bystander cells, across a range of affinities (pM to uM) and at frequencies as low as 1 in 100,000.

30 minutes

Michael Birnbaum

Michael Birnbaum is an associate professor of biological engineering at MIT. He received his bachelor’s degree in chemical and physical biology from Harvard University and his PhD from Stanford University. At Stanford, he worked under K. Christopher Garcia, and studied the molecular mechanisms of T cell receptor recognition, cross-reactivity, and activation. After postdoctoral work in […]

Lunch

1 hour

Nanoparticle single-cell multiomic readouts reveal that cell heterogeneity influences lipid nanoparticle-mediated messenger RNA delivery

Cells that were previously described as homogeneous are composed of subsets with distinct transcriptional states. However, it remains unclear whether this cell heterogeneity influences the efficiency with which lipid nanoparticles (LNPs) deliver messenger RNA therapies in vivo. To test the hypothesis that cell heterogeneity influences LNP-mediated mRNA delivery, we report here a new multiomic nanoparticle delivery system called single-cell nanoparticle targeting-sequencing (SENT-seq). SENT-seq quantifies how dozens of LNPs deliver DNA barcodes and mRNA into cells, the subsequent protein production and the transcriptome, with single-cell resolution. Using SENT-seq, we have identified cell subtypes that exhibit particularly high or low LNP uptake as well as genes associated with those subtypes. The data suggest that cell subsets have distinct responses to LNPs that may affect mRNA therapies.

30 minutes

Curtis Dobrowolski

Dr. Dobrowolski graduated with a Bachelors of Science in Biology from Jacksonville University in 2008 and completed a Master’s in Infectious disease and Immunology at the University of Sydney in 2010. In 2018, he completed his Ph.D. in molecular virology at Case Western Reserve University focusing on the reversal of HIV latency. He joined the […]

Joint analysis of CAR T cell products before and after infusion reveals hidden biomarkers of effector potential

Cellular therapy products are often thought of as generally homogeneous, with standard quality control criteria during manufacturing focusing on broad genotypic or phenotypic characterizations (e.g., proportion of cells transduced). In many cases, however, cells utilized for manufacturing are often extremely heterogeneous, each with their own potentially distinct physiological experiences, developmental trajectories, and corresponding epigenetic differences that can influence therapeutic efficacy. For instance, in a pediatric clinical study utilizing CD19 chimeric antigen receptor-modified (CAR) T cells, we found that a subset of cells within products ultimately contributes to the majority of cellular expansion and effector response in patients. Although high-resolution analysis of the pre-infusion products alone was insufficient for identifying cellular subsets with particularly high effector potential, joint analysis of pre- and post-infusion CAR T cells linked by T cell receptor sequencing allowed us to trace successful lineages to a subset of cells within the pre-infusion product. By analyzing this pre-infusion subset in the context of patient samples, we then characterized unexpected gene expression signatures and phenotypic patterns corresponding to cells that gave rise to highly effective CAR T cells in patients. Functional validation confirmed that these cells were both more cytotoxic and less prone to exhaustion than other cells in the product upon antigen encounter. Using this study as an example, we provide an experimental and analytical framework for identifying biomarkers of success in cellular therapy products.

30 minutes

Jeremy Chase Crawford

Dr. Jeremy Chase Crawford is a Principal Bioinformatics Research Scientist and the Director of Immunoinformatics in the Department of Immunology and the Center for Translational Immunology and Immunotherapy (CETI2) at St. Jude Children’s Research Hospital. He received a B.S. from Duke University in 2008, and in 2016 he received a Ph.D. from the University of California, […]

Implantable Biomaterial Cell Factories to Streamline CAR T Cell Manufacturing

Despite their clinical success, chimeric antigen receptor (CAR)-T cell therapies for B cell malignancies are limited by lengthy, costly and labor-intensive ex vivo manufacturing procedures that lead to cell products with heterogeneous composition. In this talk, I will describe several biomaterials technologies that streamline CAR T cell manufacturing and reduce processing times to a single day. Biomaterials provide the appropriate interface and signaling for T cell activation, viral vector-mediated gene transfer, expansion and release of functional CAR-T cells. Further, in vivo-generated CAR-T cells enter the bloodstream and control distal tumor growth in mouse models of lymphoma, lung, ovarian and orthotopic pancreatic cancer. Biomaterial cell factories could transform CAR-T cell therapy by fast-tracking manufacture and reduce the complexity and resources needed for provision of this type of therapy.

30 minutes

Yevgeny Brudno

Yevgeny is a Joint Assistant Professor in the Department of Biomedical Engineering at UNC – Chapel Hill and NC State – Raleigh. He earned dual B.A. degrees in Chemistry and Biophysics with minors in Math and Philosophy from the University of Pennsylvania. He obtained his Ph.D. in Chemical Biology at Harvard University, developing directed evolution […]

Refreshment Break

15 minutes

Expansion scale-up and automation of an iPSC derived NK cell therapy

Allogeneic cell therapies, and even more so iPSC derived therapies, have the potential to reach hundreds, if not thousands of patients. However, to be able meet demand, the manufacturing process has to be reliably scaled up with no significant impact to product quality. Fate is exploring two strategies for scale up: volumetric scaling and culture intensification. Here we present preliminary results from feasibility studies demonstrating the impact of both strategies on our drug substance expansion process. Both strategies appear to be feasible per the growth kinetics and phenotype at harvest.

30 minutes

Heather Leu

Heather Leu is a process development engineer and a member of the Process Engineering group within MSAT at Fate Therapeutics. She currently works on scaling up expansion in the manufacturing process. Prior to Fate, she worked at Kite Pharma doing autologous process development.

Pinpointing correlations for quality by integrating process and data analytics

Cell-based therapies and diagnostics are promising medicines to treat disease and injury, ranging from a child’s blood cancer to an otherwise healthy athlete’s damaged knee joints. But cell therapies are ”living medicines” for which the positive impact on patients is limited by critical bottlenecks in the manufacturing of even approved biopharmaceutical therapies. All cell therapies begin with a human donating the cells that will be manufactured into medicine, and that is just the start of the manufacturing complexities when biology (rather than just chemistry) is used to design and engineer medicine. Here, we will discuss recent advances in the research, development, and workforce training developed by engineers at MIT and our scientific, clinical, and regulatory science partners across the world, including the CAMP (Critical Analytics for Manufacturing Personalized-Medicine) at MIT and in Singapore. These new approaches lay the groundwork for a world that does not just invent exciting new cell therapies, but also produces them in a way that makes the resulting medicine safe, effective, and accessible for each patient.

30 minutes

Krystyn Van Vliet

Krystyn J. Van Vliet serves currently as Director of Manufacturing Innovation for the MIT Innovation Initiative. She is the Michael and Sonja Koerner Professor of Materials Science and Engineering, and holds a dual appointment in the Department of Biological Engineering.  Dr. Van Vliet earned her Sc.B. in Materials Science & Engineering from Brown University and […]

Day 1 Closing Remarks

15 minutes

Jacqueline M. Wolfrum

Dr. Wolfrum has been at the MIT Center for Biomedical Innovation (MIT CBI), since 2014. She is Director of the Biomanufacturing Program (BioMAN), a pre-competitive biopharmaceutical industry consortium focused on development of new knowledge, science, technologies, and strategies that advance the manufacture and global delivery of high-quality biologic medicines. She manages sponsored projects and activities […]

Poster Session and Evening Networking Reception

2 hours

Day 2 Opening Remarks

5 minutes

Stacy L. Springs

Dr. Stacy Springs is the Executive Director at the MIT Center for Biomedical Innovation (CBI). The Center integrates the Institute’s technical, scientific, and management expertise to solve complex biopharmaceutical challenges. CBI leads multi-stakeholder, multidisciplinary research and educational initiatives with real world impact, including MIT’s Biomanufacturing Consortium, (BioMAN), and it’s Consortium on Adventitious Agent Contamination in […]

Keynote Presentation: Partnering in Early Development and Biomanufacturing to Accelerate Life Science Innovation

An expanding toolbox is driving unprecedented acceleration in life science innovation.  Many new and emerging advanced therapies are entering the clinic driven by academia and a vibrant start-up community.  A significant challenge in this space is access to drug development expertise to help in the translational journey from the lab bench to the clinic.  Landmark Bio was created to bring together the best of academia, industry and research hospitals to accelerate breakthrough innovations in advanced therapies.  This talk will explore how Landmark is bringing drug development expertise and flexible biomanufacturing capacity to enable therapeutic translation of advanced therapies.

30 minutes

Gregg Nyberg

Gregg Nyberg is a biopharmaceutical executive with twenty-five years of experience in process development, clinical manufacturing, technology transfer and commercialization. He is currently Chief Technology Officer at Landmark Bio, where he is responsible for establishing process development and clinical manufacturing capabilities for emerging technologies such as cell and gene therapy.  Prior to joining Landmark, Gregg […]

The development of a novel technology platform for preservation of cells

Since the discovery of dimethlysulfoxide (DMSO) as a cryoprotective agent in the 1970’s, there have been many efforts to replace this molecule for the preservation of cells.  Nature uses combinations of molecules called osmolytes to preserve biological systems against environmental stresses.  We hypothesized that combinations of molecules could be used to preserve cells without DMSO. Testing this hypothesis requires the development of a technology platform.  One element of the technology platform was low temperature Raman spectroscopy.  This label-free technology enables us to understand where protective molecules are and how they interact with water or the cell to protect it.  The other technology is an optimization strategy to reduce the experimentation required to optimize a preservation protocol.  An algorithm that enables efficient optimization of composition, freezing rate and enables preservation of heterogeneous cell populations will be described.  This platform has been used to develop DMSO-free methods of preserving a variety of cell types.

45 minutes

Allison Hubel

Dr. Hubel is a Professor in Mechanical Engineering at the University of Minnesota and Director of the Technology Leadership Institute. She is President-elect of the Society for Cryobiology. Dr. Hubel has studied both basic science and translational issues behind preservation.  Her work spans from the study of molecular mechanisms of damage during preservation to the […]

In-use physicochemical stability studies: Strategies to support the clinical administration of cell therapies

In-use physicochemical stability studies are aimed at ensuring patient safety during the clinical administration of drug products. These studies are intended to demonstrate drug product stability and compatibility with the administration process by mimicking recommended in-use hold-times and clinical administration methods. There is limited health authority guidance on specific in-use testing practices. As such, current strategies should be guided by factors that can impact potential critical quality attributes (pCQAs) including 1) country or region-specific expectations for clinical administration, 2) anticipated drug product hold-times and administration practices during clinical use, and 3) required administration materials in the fluid path. For cell therapies in particular, in-use stability studies are complicated by the fact that the administered cells are living drugs that require diverse and potentially complex methods to relate product CQAs to clinical outcomes. Here, we discuss specific challenges with the administration of cell therapies and strategies to guide in-use testing practices for this new therapy.

30 minutes

Abbygail Foster

Abbygail Foster is a Principal Scientist and Cell Therapy Team Lead, Pharmaceutical Development at Genentech. As team lead, she oversees several cell therapy programs in various stages of clinical development. Prior to her current role, Abby was the formulation lead for one of Genentech’s T-cell bispecific antibody programs and was responsible for supporting Phase 1 […]

Refreshment Break

30 minutes

Perfusion-based microbioreactors for production of CAR T cell therapies

With six CAR T cell therapy products currently approved by the FDA for the treatment of hematological cancers, there are increasing efforts to develop novel and better manufacturing technologies and processes to improve efficacy, reduce variability, and reduce cost. However, due to limited or costly materials, such as human PBMCs and viral vectors, research- and preclinical-scale experiments in cell therapy are often done in milliliter-sized static culture with minimal environmental control, which can result in variabilities that may prove difficult to translate to larger culture systems. We leveraged an automated perfusion microbioreactor, which can replicate bench-scale perfusion processes, to generate functional CAR T cells at high cell densities in a 2-mL culture volume. We showed that with improved media exchange from continuous perfusion, cell expansion is improved. These proof-of-concept data demonstrate the utility of the system as both a process development tool as well as a potential future manufacturing platform.

30 minutes

Michael Birnbaum

Michael Birnbaum is an associate professor of biological engineering at MIT. He received his bachelor’s degree in chemical and physical biology from Harvard University and his PhD from Stanford University. At Stanford, he worked under K. Christopher Garcia, and studied the molecular mechanisms of T cell receptor recognition, cross-reactivity, and activation. After postdoctoral work in […]

Automation solutions in cell therapy development

Over the last decade, T cell therapy has proven to be a revolutionary clinical solution to several devastating, and often fatal diseases. Yet, the T cell therapy manufacturing process can be a complex, manual and labor-intensive process. This has an impact on regulatory compliance and can prolong timelines to get biopharmaceuticals to the patients who need it the most. To address these process development challenges, at Thermo Fisher, we are focused on developing automation solutions that can mitigate these manufacturing roadblocks, with the goal of getting therapies to patients faster. Through digital automation, our series of closed, modular instruments can be both physically and digitally integrated to increase standardization, and help customers successfully reach their cell therapy manufacturing milestones. This physical and digital connectivity using automation could potentially help reduce the overall cost of production of cell therapies. During this talk, we will discuss specific challenges biotech and pharma customers may face within the T cell therapy process workflow, and how those challenges can be overcome by leveraging automation across a broad range of production scales.

30 minutes

Yongchang Ji

Yongchang Ji, Ph.D. is a process development manager in the Cell and Gene Therapy Group at Thermo Fisher Scientific in Carlsbad, CA. Yongchang is currently working on development of closed, modular and semi-automatic workflow solutions for viral and non-viral cell therapy. Dr. Ji received his Ph.D. in Cell Biology and Anatomy from State University of […]

Removal of empty AAV capsids to undetectable levels using orthogonal purification steps – Product quality and process robustness considerations

Typical AAV Gene Therapy production systems generate cell culture harvests with a greater abundance of empty capsids (lacking DNA genomes) than full capsids (containing the desired DNA genome).  Removing the empty capsids during purification is a central production challenge since empty and full capsids possess similar physio-chemical properties. Design of a robust purification process capable of completely separating empty capsids from full capsids is presented in this work. This process employs orthogonal purification steps (Ion Exchange Chromatography and Zonal Ultracentrifugation) for removing empty capsids and controlling other process- and product-related impurities to assure the safety of AAV products. Scalability of this process and features that allow the combination of these orthogonal steps to provide robust product quality are also presented.

30 minutes

Yanhong Zhang

Yanhong Zhang is a Principal Scientist in the Drug Substance Technology group within the Technical Development Organization at BioMarin. She obtained her PhD in Biophysics from Medical College of Wisconsin before joining BioMarin. Yanhong has over 15 years of experience in downstream process development and scale-up/technology transfer for recombinant protein therapeutics and gene therapy vectors.

PatheonTM Quick to ClinicTM: A Serotype Agnostic AAV Vector Manufacturing Platform

Recombinant adeno-associated viral (rAAV) vectors are currently one of the most widely used gene therapy products in development due to their lack of pathogenicity, gene expression persistence and presence of various serotypes that enable a diverse cell tropism. The rapidly growing demand combined with challenges arising from long process development timelines, lack of cGMP-suitable fit-for-purpose components and uncertainty in regulatory approval has led to a critical need for scalable and cost- effective standardized manufacturing platforms. Thermo Fisher Scientific has created a high quality scalable robust end-to-end manufacturing rAAV platform that uses fit-for-purpose materials with matched analytics. Despite the subtle biochemical and biophysical properties of various AAV therapeutics/ serotypes, we were able to develop a purification platform that would be tolerant of these differences. Utilizing this characterized platform will enable clients to bring their products to the clinic on a de-risked and accelerated pathway.

30 minutes

Hetal Brahmbhatt

Dr. Hetal Brahmbhatt is a Senior Manager, Science and Technology Team, Pharma Services, and Viral Vector Services at Thermo Fisher Scientific. She has a Ph.D. in Biochemistry and Biomedical Sciences from McMaster University where she characterized small molecule modulators of apoptotic proteins. In her previous role at Thermo Fisher Scientific (former Brammer Bio), she developed […]

Lunch

1 hour

Continuous harvesting of lytic viruses using a VHU perfusion system

Lytic viruses such as Influenza A Virus are normally produced using adherent cell processes since suspension cell processes display a “cell density” effect where the cell-specific virus yield (virions/cell) decreases significantly as the cell density increases beyond 1-5 million cells/mL.  Prior investigations have shown that the cell-specific virus yield can be sustained with increasing cell density using a perfusion system consisting of a virus harvest unit (VHU®, Cambridge, MA) which allows for continuous nutrient supply and removal of toxic compounds. Virus titers more than 10 times higher in perfusion mode compared to previously reported batch cultivations were observed as a result of achieving similar or higher cell-specific virus yield (~50-90 PFU/cell) at cell densities increasing from 2x106 cells/mL in batch to 2x107 cells/mL in perfusion. During the virus replication phase, the VHU®, in contrast to the hollow fiber membrane, allowed 100% of the produced virus particles to pass through into the permeate including cell debris and host cell proteins as well as host cell DNA released with cell lysis. The VHU® appeared not to retain any protein.  In strong contrast, a hollow fiber membrane filter was shown to withhold most proteins which may result in greater fouling of the membrane over time. Overall, the VHU® perfusion process offers interesting possibilities for advanced process integration strategies for next-generation virus manufacturing.

20 minutes

Maurizio Cattaneo

Maurizio Cattaneo did a Ph.D. at McGill in the area of enzyme replacement therapy for ureamia. He then proceeded to National Research Council as a Research Officer in the area of biosensor development. He founded several companies in the area of drug delivery and cell culture and finally but not least, he developed and patented […]

Viral vector processing: Scalable ultra/diafiltration for improved process efficiency and viral recovery

Adeno-associated virus (AAV) & lentivirus (LV) are the most common vectors used for delivering genetic material in viral gene therapy and CAR-T cell therapy applications. There is currently no scalable, high yield and high throughput, platform approach for viral vector purification. Self-contained, pre-sterilized tangential flow filtration (TFF) filters with scalable and reliable performance offer opportunities for efficient viral vector UF/DF processing operations. This study evaluated the performance of 100 and 300 kDa regenerated cellulose membranes for concentrating AAV and LV feed streams. TMP control (typical antibody applications) and permeate control (microfiltration applications) are two operating modes for batch TFF processes. The 100 and 300 kDa membrane cut-offs lie between these two applications. This presentation compares two operating control strategies for the UF/DF with the 100 and 300 kDa membranes. We also show how a 50 cm2 lab-scale device can predict performance of large-scale devices, providing a starting point for process development with viral vectors. Additional comparison between flat sheet filters and hollow fiber modules with LV feed streams will also be presented.

20 minutes

Anand Alembath

Anand Alembath is a Development Engineer III working in bioprocessing filtration R&D team at MilliporeSigma. He is the applications technical lead for tangential flow filtration (TFF) product development projects that are focused on downstream purification of viral vectors. Anand received his M.S. and Ph.D. in Chemical Engineering from Missouri University of Science and Technology, Rolla.

Development of high throughput purification platform to accelerate AAV process development and product understanding

Early-stage development is a fast-paced phase in the life cycle of gene therapy product development. For adeno-associated viral vectors, this phase comes with modality specific challenges including limited available material and serotype-specific biophysical properties that impact the process development strategies. High throughput technology makes a significant impact in AAV process development because it helps circumvent modality specific challenges. This presentation highlights utilization of high throughout purification platforms for AAVs which enables accelerated process development and product quality understanding.

20 minutes

Mahsa Hadidi

As Senior Scientist of CMC Process Development, Mahsa Hadidi leads process development, characterization, optimization and scale up for viral vectors at Sanofi Genomic Medicine Unit. Prior to joining the CMC team, Mahsa was part of the Global Process Science team in Sanofi MSAT. Mahsa received her Ph.D. in Chemical Engineering from the Pennsylvania State University.

Refreshment Break

15 minutes

Placing filtration into gene therapy platforms – potential and remaining challenges

Early gene therapy manufacturing has largely borrowed technologies developed for recombinant protein processing. This has generally translated well, but there is still significant work to be done to improve process outputs (e.g. yield, cost, product quality) and assess their platform potential. In this work we will focus on filtration technologies and assess which have translated well, and how process development has changed for these new modalities. We will also highlight where new technologies need to be developed specifically for gene therapy processing. This talk will put particular focus on adventitious agent clearance (virus, bioburden) with an eye towards converging on platforms for some of the major viral vector classes. Data is available from adeno-associated virus (AAV) and lentivirus processes.

20 minutes

Nicholas Marchand

Nick Marchand is a manager within Pall’s Research & Development group focused on bioprocess testing of new equipment and consumables. He has previous experience in process development groups at Genzyme and Amgen. He holds a B.S. and Ph.D. from Rensselaer Polytechnic Institute.

Lipid nanoparticle development and impact on tissue targeting for non-viral gene therapy

Lipid nanoparticle (LNP) technology, as currently one of the most promising and emerging technologies, shows the ability to deliver nucleic acid therapeutics for non-viral gene therapy (NVGT). The recent approvals of Onpattro and COVID-19 vaccines have demonstrated the feasibility, manufacturability and clinical safety of lipid based nano carriers. Compared to intramuscular vaccine delivery, intravenous (i.v.) delivery of a LNP formulation for gene therapy shows unique challenges. A LNP formulation for gene therapy via i.v. may require different lipid and formulation design. LNP tropism can be modulated via changing LNP physical and chemical properties (passive delivery) and adding targeting ligands (active targeting). In this talk, we will discuss the considerations and challenges in LNP development and impact on tissue targeting for NVGT.

20 minutes

Yuefei Shen

Yuefei Shen is a Principal Scientist within the CMC Drug Product Development Organization at Sanofi wherein she leads a team responsible for early drug product development for gene therapy programs. Prior to Sanofi, she worked at Novartis leading research projects. She obtained her Ph.D. in Chemistry from Washington University in St. Louis and Postdoc from […]

Summit Closing Remarks

20 minutes

Stacy L. Springs

Dr. Stacy Springs is the Executive Director at the MIT Center for Biomedical Innovation (CBI). The Center integrates the Institute’s technical, scientific, and management expertise to solve complex biopharmaceutical challenges. CBI leads multi-stakeholder, multidisciplinary research and educational initiatives with real world impact, including MIT’s Biomanufacturing Consortium, (BioMAN), and it’s Consortium on Adventitious Agent Contamination in […]

VENUE

An email with a link to join the virtual Summit has been sent to all attendees.  Please contact Marisia Ketchum (mketchum@mit.edu) or Jessica McGrath (mcgrath2@mit.edu) if you have yet to receive the event credentials.

REGISTER

Registration for the 2021 Summit has closed.
Please reach out to Marisia Ketchum if you are interested in joining the meeting.

Thanks to Our Members

SUMMIT SPONSORS

Copyright MIT CBI. All rights reserved