Cambridge Healthtech Institute’s 9th Annual

Cell Therapy Manufacturing

Scaling and Industrializing Cell-Based Therapies

August 16 - 17, 2023 ALL TIMES EDT

CHI’s Cell Therapy Manufacturing conference examines the practical challenges in manufacturing autologous and allogenic cell therapies at scale, with dedicated sessions on cell processing, scalability, next-generation production technologies, automation, closed systems, artificial intelligence, supply chain, and facility design. The meeting takes examples from autologous and allogeneic therapies and the field of CAR Ts, NK cells, Gamma Deltas, TCRs, TILs and IPSCs.

Wednesday, August 16

Registration and Morning Coffee7:30 am

MANUFACTURING CELL THERAPIES BEYOND CAR Ts

7:55 am

Chairperson's Opening Remarks

Dominic Clarke, CSO, Orange County Bio; Committee Chair, ISCT

8:00 am KEYNOTE PRESENTATION:

Manufacturing Gamma Deltas

William Ho, President, CEO, Co-Founder, IN8bio

IN8bio is a clinical-stage gamma-delta T cell therapy company with two clinical programs in Phase I and one in Phase II, in both solid and hematological tumors. Gamma-delta T cells are part of the innate immune system with the ability to recognize and kill malignant cells and our clinical manufacturing experience has shown that evaluation of the CQA and cellular effector memory profile may help predict expansion and potency.

8:30 am FEATURED PRESENTATION:

The (Re)emerging Field of Xenotransplantation

Knut Niss, PhD, CTO, eGenesis, Inc.

Through our transformative research, we are developing HuCo organs and cells to meet the increasing need. Our eGenesis Genome Engineering and Production (EGEN) platform leverages advances in gene editing technologies to address the historical challenges of xenotransplantation.

9:00 am

Tackling Starting Material Heterogeneity to Overcome the Challenges of Autologous TCR T Cell Manufacturing

Gagan Bajwa, PhD, Senior Scientist, Process Development, Immatics

This presentation will discuss: Heterogeneity of the starting leukapheresis poses challenges for successful manufacturing of autologous TCR T cells; cellular composition of the starting material impacts product characteristics; and optimization of starting material is critical to achieve adequate quantity and quality of the TCR T cell product.

9:30 am

POSTER PRESENTATION: A Mathematical Approach to Bridge Metabolic Parameters to CAR T Cell Growth in Limited Data Conditions

Keshav Patil, PhD, Scientist, Advanced Therapies, Janssen Pharmaceuticals Inc.

Cell expansion is critical to manufacturing of cell therapies and is quantified using cell count and the underlying cell growth profile. Mathematical models provide a non-invasive technique to estimate cell count, however, face constraints during limited data conditions. We perform experiments measuring growth in T cells samples from healthy donors and propose a framework that maps metabolic parameters to cell growth based on a hybrid modeling strategy to deal with the limited data.

Coffee Break in the Exhibit Hall with Poster Viewing10:00 am

SPECIAL ISCT SESSION – PAT, PROCESS CONTROL, AND SCALE-UP

10:40 am

Cell Therapy Process Development and Manufacturing

Jerry Eriksson, MSc, Senior Research Scientist, AstraZeneca R&D

11:10 am

Process Analytical Technologies for Process Control Strategy Development of Cell & Gene Therapy Products

John Churchwell, PhD, Associate Lead Scientist, Cell & Gene Therapy Catapult

Advanced control strategies using PAT technologies have the potential to increase product consistency, reduce process variability and increase potential for process automation during the production of cell and gene therapy products. This presentation will discuss examples of real-time monitoring strategies and how an automated PAT lab set up can be utilized for improved process characterization and future manufacturing automation.

11:40 am

Decentralized Manufacturing of Cell Therapy Products

Scott R. Burger, Principal, Advanced Cell & Gene Therapy LLC

This presentation provides an overview of centralized and decentralized manufacturing models, pros/cons and suitable applications, approaches to overcoming challenges of decentralized manufacturing, and regulatory considerations.

Sponsored Presentation (Opportunity Available)12:10 pm

Refreshment Break in the Exhibit Hall with Poster Viewing12:40 pm

SPECIAL ISCT SESSION – PAT, PROCESS CONTROL, AND SCALE-UP (CONT.)

1:25 pm

Chairperson's Remarks

Dominic Clarke, CSO, Orange County Bio; Committee Chair, ISCT

1:30 pm

Be More Closed-Minded

Ian D. Gaudet, PhD, ISCT Process & Product Committee Member, and Senior Director and Site Head, Process Sciences, Miltenyi Biotec, Inc.

Autologous cell therapy manufacturing costs still limit bringing these impactful medicines to more patients. Open process manipulations requiring expensive engineering controls are a significant driver of the overall manufacturing costs, including significant expenses in labor, equipment, materials, cleaning, validation, and facility footprint. Progress towards fully-closed system design suitable for commercial scale manufacturing in both centralized and point-of-care settings are discussed.

2:00 pm PANEL DISCUSSION:

ISCT SESSION – Process Analytics, Automation, and Digitalization

PANEL MODERATOR:

Dominic Clarke, CSO, Orange County Bio; Committee Chair, ISCT

  • ​The role of process analytics to inform decisions
  • Emerging technologies and modalities
  • Automation and AI in cell therapy manufacturing 
PANELISTS:

Jerry Eriksson, MSc, Senior Research Scientist, AstraZeneca R&D

Ian D. Gaudet, PhD, ISCT Process & Product Committee Member, and Senior Director and Site Head, Process Sciences, Miltenyi Biotec, Inc.

John Churchwell, PhD, Associate Lead Scientist, Cell & Gene Therapy Catapult

Refreshment Break in the Exhibit Hall with Poster Viewing3:00 pm

PLENARY KEYNOTE: LEADING TO TOMORROW'S ADVANCES

3:50 pm

Chairperson's Remarks

Ran Zheng, CEO, Landmark Bio

4:00 pm

Implementing Advanced Manufacturing Technologies to New Biotech Modalities

Konstantin B. Konstantinov, PhD, CTO, Ring Therapeutics

Using exosomes as an example, this presentation examines the current and future trends in biomanufacturing, and the technologies needed to manufacture emerging modalities at scale. Traditional biomanufacturing methods do not provide the industrialized, commercially scalable, highly efficient and reproducible manufacturing process essential for this new class of biotherapeutics—so we built it from the ground up.

4:30 pm

The Digitalization of Biomanufacturing

Richard D. Braatz, PhD, Edwin R. Gilliland Professor, Chemical Engineering, Massachusetts Institute of Technology

A fully instrumented testbed is described for the end-to-end integrated and continuous manufacturing of monoclonal antibodies. The testbed consists of parallel bioreactors, simulated moving bed chromatography systems for capture and polishing, bespoke viral inactivation, and a MAST auto-sampling system. Experimental results are compared with a digital twin for continuous runs lasting 30 to 60 days each, which include variations in metabolites and glycosylation profiles in designed experiments. The increased consistency in the glycosylation profile of the monoclonal antibodies being produced is quantified when going from batch to semi-batch to perfusion mode, and when moving from start-up to quasi-steady conditions.

Networking Reception in the Exhibit Hall with Poster Viewing5:00 pm

Close of Day6:00 pm

Thursday, August 17

Registration and Morning Coffee7:30 am

LENTIVIRUS, AAV PROCESS DEVELOPMENT AND QUALITY

7:55 am

Chairperson's Remarks

Nathalie Clément, PhD, Vice President, Vector Development, Translational Gene Therapies, Siren Biotechnology

8:00 am

Overcoming the Challenges of Biomanufacturing Lentiviral Vector

Martin Loignon, PhD, Team Leader, Cell Engineering, National Research Council Canada

The demand for lentiviral vectors (LVs) for R&D and engineering cell therapies stems from their efficacy to deliver genes into targeted cells. Current LVs' production bioprocesses vary widely, significantly impacting quantities, quality, and costs. We have used a holistic approach to address challenges of upstream and downstream bioprocesses to increase titers and recovery.

8:30 am

Generation of Rhabdoviral-free Sf9 Cells

Stanley Chung, PhD, Engineer II, Voyager Therapeutics

Coffee Break in the Exhibit Hall with Poster Viewing9:00 am

9:30 amBreakout Discussion Groups

Breakout discussions provide an opportunity to discuss a focused topic with peers from around the world in an open, collegial setting. Select from the list of topics available and join the moderated discussion to share ideas, gain insights, establish collaborations or commiserate about persistent challenges. Please visit the breakout discussions page on the conference website for a complete listing of topics and descriptions.

IN-PERSON ONLY BREAKOUT:

TABLE 14 - Manufacturing Cell and Gene Therapies

Michael D. Jacobson, PhD, Managing Partner, Cambridge Biostrategy Associates LLC

  • ?Trends in commercializing cell and gene therapies
  • De-centralized versus centralized manufacturing
  • Pricing trends, reducing costs
  • New trends
  • In vivo CAR T engineering and delivery?
  • Gene editing

MANUFACTURING IPSCs

10:25 am

Chairperson's Remarks

Mo Heidaran, PhD, Head, Translational and Regulatory Strategy, GC Therapeutics, Former FDA Reviewer

10:30 am

Scalable Production of Pluripotent Stem Cell-Derived Hematopoietic Progenitor Cells and Functional T Cells in Stirred Tank Bioreactors

Liz Csaszar, PhD, Senior Director, Manufacturing Sciences, Tech Operations, Notch Therapeutics

Stirred suspension-based cell manufacturing can be used for scalable and controllable production of cell therapy products. We have developed custom reagents to modulate Notch signaling, which are compatible with suspension culture, and have implemented the production of pluripotent stem cell-derived CD8aß+ T cells in stirred tank bioreactors (STRs). STR-based culture enables process optimization and characterization using bioprocess solutions including automated feeding and in-process monitoring.

11:00 am

Engineering & Manufacturing iPSC-Derived Innate Cells to Provide Globally Scalable, Allogeneic Innate Therapies

Allen Qiang Feng, PhD, Founder and CSO, HebeCell Corp.

Human pluripotent stem cell (PSC)-derived natural killer (NK) cells combine the advantages of PSC and the safety profile of NK cells. At HebeCell we have developed our proprietary technology platform that is bioprocessing friendly and adaptable to GMP standards. Our feeder-free platform utilizes 3D spheroids to mimic the in vivo hematopoiesis to generate cytotoxic protoNK cells in bioreactors. Our platform offers a highly scalable approach for off-the-shelf cell therapies.

11:30 am

POSTER PRESENTATION: Scalable Production of Induced Pluripotent Stem Cell-Derived CD8+ CAR-T Cells in Stirred Tank Reactors Using DLL4/VCAM-1 Coated Beads

Vaisakh Rajan, Scientist II Cell & Molecular Biology, Mfg Sciences, Notch Therapeutics

Adoptive T-cell therapies have shown promise for certain blood cancers but face challenges in production and logistics when using genetically modified patient-derived T cells. Induced pluripotent stem cells (iPSCs) offer a potential solution, yet scalable T-lineage commitment remains an obstacle. We developed an Engineered Thymic Niche (ETN) platform that enables scalable production of iPSC-derived CD8+ CAR-T cells with equivalent function to primary CD8+ CAR-T cells in serial killing assays.

11:45 am

POSTER PRESENTATION: Exploring the Production Potential and Mechanoresponsiveness of Induced Pluripotent Stem Cell-Derived Mesenchymal Stem/Stromal Cells for Extracellular Vesicle Biomanufacturing

Emily Powsner, Graduate Research Asst, Bioengineering, Univ of Maryland College Park

Mesenchymal stem cell extracellular vesicles (MSC EVs) are an emerging biotechnology that possess regenerative properties. However, donor MSCs exhibit substantial variability that impacts their ability to consistently produce bioactive EVs, limiting large-scale biomanufacturing potential. Thus, we evaluated induced pluripotent stem cell-derived MSCs for their responsiveness to mechanical cues, subsequent EV angiogenic bioactivity, and production potential compared to bone marrow-derived MSCs.

12:00 pmSession Break

Refreshment Break in the Exhibit Hall & Last Chance for Poster Viewing12:30 pm

OPTIMIZING CELL THERAPY MANUFACTURING

1:05 pm

Chairperson's Remarks

Scott R. Burger, Principal, Advanced Cell & Gene Therapy LLC

1:10 pm

Streamlined Expansion of PBNK and CD147-CAR-NK Cells in the Grex-100M Closed System: Achieving Scalability and Efficiency

Xuening Wang, PhD, Senior Research Associate, Rutgers-New Jersey Medical School

Natural killer (NK) cells efficiently eliminate tumors. Engineering them with a chimeric antigen receptor (CAR) enables adoptive cancer treatment. Reliable large-scale production methods are crucial for clinical application. The G-Rex 100M bioreactor expands NK cells, with ample medium volume and enhanced oxygen delivery. It supports both PBNK and CAR-NK cell growth, displaying high cytotoxicity against hepatocellular carcinoma cells and resembling G-Rex 6M plates. Cryopreservation minimally affects cytotoxicity. Our method offers a robust and scalable platform to produce NK and CAR-NK cells, facilitating the clinical use of "off-the-shelf" NK immunotherapy.

1:40 pm

NK and CAR-NK Processing Development

Dongfang Liu, PhD, Associate Professor, Director Immunoassay Development, Pathology & Immunology & Lab Medicine, Rutgers University

Currently available technologies for ex-vivo expansion of NK and CAR-NK cells using feeder cell expansion systems (e.g., K562 cells ) and cytokines (e.g., IL-2 in combination with IL-15) show several limitations telomere shortage, exhaustion, fratricidal killing, and regulatory concerns. Previous studies show that a human 721.221-mIL21 as a feeder cell can rapidly expand primary NK and CAR-NK cells. Based on this technology, we developed a non-feeder cell system to expand NK and CAR-NK cells. The results provided in this study show strong promise with the non-feeder cell expansion approach for functional NK and CAR-NK cells. 

2:10 pm

Encapsulated Cell Therapy: An Off-the-Shelf, Scalable Approach to Treating Solid Tumors

Lauren E. Jansen, PhD, Director, Process Development, Avenge Bio

Avenge Bio’s LOCOcyte platform consists of polymer encapsulated, allogeneic cells genetically engineered to produce immunomodulatory molecules for the treatment of previously intractable cancers. This presentation will highlight our approach to a successful technology transfer of our lead IL-2 program, AVB-001, into Phase I GMP manufacturing. In addition, we will discuss key considerations for future development and scale-up of this innovative cell therapy.

Networking Refreshment Break2:40 pm

DIGITAL INTEGRATION, DECENTRALIZED MANUFACTURING

2:55 pm

The Application of Digital Informatics Methods to Manage Development and Production Data for Cell and Gene Therapies

William E. Janssen, PhD, Principal, WEJ Cell & Gene Therapy Consulting Services LLC

Explosive growth in the fields of informatics and cell and gene therapy (CGT) has occurred over the last four decades. Application of Informatics tools in CGT manufacturing can facilitate record keeping, quality management reviews and communications. An informatics backbone will be essential for deployment of disseminated manufacturing. While electronic records can completely replace paper and ink records, the potential benefits are much greater, including prospective quality management, error mitigation in manufacturing, release and administration, and reduction in redundancies. Implementation of informatics in CGT requires substantial planning, resources, and collaboration between CGT and informatics teams.

3:25 pm

Could Your Cell Therapy Manufacturing Facility Be Working Harder for You?

Peter Walters, Fellow of Advanced Therapies, CRB

Optimizing cell therapy manufacturing facilities is challenging due to small-scale cell therapy batch sizes. With batch sizes as small as one patient, manufacturers must scale out rather than up, duplicating every inefficiency in the process. This presentation explores four manufacturing approaches for small-scale cell therapies: dedicated rooms, chasing workstations, segregated unit operations, and process-in-a-box systems. Each approach has its challenges, and manufacturers must balance risk tolerance, capital spending, and growth expectations to achieve manufacturing goals. Case studies comparing facility designs that enable throughput will be presented and compared.

3:55 pm

Considerations in Development of Lentiviral-Based in vivo Gene Therapy

Mukesh Mayani, PhD, Head of Process Development, Gene Therapy, National Resilience, Inc.

Lentiviral vectors (LVV) have demonstrated noteworthy clinical success in patients during ex vivo CAR T as well as stem cell therapy gene therapy (GT) applications. The third-gen SIN LVVs have shown improved safety profiles for a conceivable durable treatment of rare disease and cancer indications. However, it’s application as in vivo therapy option is severely limited due to manufacturing, safety, and quality challenges. In this presentation, we will highlight several development and manufacturing considerations for generation of LV vector suitable for in vivo GT use.

Close of Summit4:25 pm