Thursday, September 26, 2024 - Day Three of Main Conference - ET (Eastern Time, GMT-05:00)
Thursday, September 26, 2024 - Day Three of Main Conference - ET (Eastern Time, GMT-05:00)
Take a break from the science and find your flow by joining us for yoga. See you on the mat!
More details and sign up form to follow.
- Don Jarvis, Ph.D. - President, GlycoBac, LLC
Production of biologics require adequate cellular capacity to maintain desired productivity and quality. Topics covered include how tuned energy supply improve secreted productivity in HEK293. How tuning of secretory helper proteins in CHO increase level of active sulfatase product 150-fold. We will also cover cell and protein engineering strategies for improved AAV production.
- Johan Rockberg - Professor, KTH Royal Institute of Technology
A product produced from cell-free protein synthesis (CFPS) has progressed to late-stage clinical trials in the REFRαME-O1 study for ovarian cancer. The XpressCF+® system from Sutro Biopharma is poised to launch the first commercial CFPS produced therapeutic pending successful completion of clinical trials. To enable a viable commercial process, challenges of scale, cost, and robustness have been addressed. Continued innovation in XpressCF+® promises to further improve efficiencies for all pipeline products and establish CFPS as a new therapeutic manufacturing platform.
- James Zawada - Associate Director Process Science, Sutro Biopharma
This presentation will outline the optimization conducted at each stage of the cell line development process. Through holistic enhancements in DNA vector design, transfection methods for pool generation, clone selection, and bioreactor process parameters, we achieved a 20-fold increase in titers across multiple transgenes. This case study showcases our platform's evolution from a partially adherent 293T system to a fully-suspension, T-antigen free cell line development process. This evolution eliminates the use of animal-derived components and significantly reduces the development timeline.
- Adam Carcella - Cell Line Development Scientist, Asimov
- Eden Turner - Portfolio Manager, Informa Connect
How it will work is we have 3 Round Tables each covering a different challenge of Upstream & Cell Culture in Bioprocessing. Each will be led by an expert. The expert will kick off and guide the discussion which will last for 30 minutes. At the end of the 30 minutes the leader will report their main conclusions to the room for a maximum of 10 minutes.
Roundtable Discussions
Cell Culture Optimization
- Cell Line Development: Selecting and developing an optimal cell line is crucial for achieving high product yields and quality. Identifying a cell line with desirable characteristics, such as growth rate, productivity, and stability, can be a time-consuming and complex process.
- Cultivation Conditions: Maintaining optimal conditions for cell growth and productivity is challenging. Factors like temperature, pH, nutrient availability, and gas exchange need to be carefully controlled to ensure optimal performance. Achieving consistent and reproducible results across large-scale bioreactors adds another layer of complexity
Bioreactor & Upstream Scale-Up:
- Transition from Lab Scale to Production Scale: Scaling up bioprocesses from small laboratory bioreactors to large-scale production facilities can lead to challenges in maintaining consistent conditions. Factors like mixing, mass transfer, and heat dissipation become more complex as the scale increases. Achieving uniform distribution of nutrients and gases becomes crucial for maintaining cell health and product quality. Take into account Perfusion versus Fed-Batch.
- Bioreactor Design: The design of bioreactors for large-scale production must consider factors such as hydrodynamics, shear stress, and heat transfer to ensure optimal cell growth and product formation. Choosing the right type of bioreactor and ensuring scalability without compromising performance is a significant challenge
Process Monitoring and Control:
- Real-time Monitoring: Continuous monitoring of various parameters, such as cell viability, metabolite concentrations, and product titer, is essential for process control and optimization. Implementing reliable real-time monitoring techniques can be challenging, particularly for complex bioprocesses.
- Process Control: Maintaining tight control over the bioprocess is crucial to achieve consistent product quality. Controlling variables such as pH, dissolved oxygen, and nutrient concentrations requires advanced control strategies. Deviations from optimal conditions can negatively impact cell growth, viability, and product quality
- Swetha Kumar - Scientist, Sanofi
- Jianfa Ou - Principal Scientist, Biologics Development, Global Product Development and Supply, Bristol Myers Squibb
- Nikki Nogal, PhD - Global Director, Technical and CMC, Lonza
The automation of monoclonal antibody (mAb) production necessitates real-time information on the concentrations of products and contaminants at various process stages, which current analytical methods cannot provide. This presentation introduces a fiber-optic nanoplasmonic biosensor system designed for automatic, real-time affinity-based detection and quantification of Immunoglobulin G (IgG) and IgG aggregates. These sensor systems can be integrated into both upstream and downstream unit operations, offering specific biodetection in complex sample matrices with high sensitivity and selectivity across a wide concentration range. The flexible flow cells enable continuous monitoring at different production scales and flow rates, thereby simplifying process development, enhancing production, and facilitating process intensification and automation in mAb manufacturing.
- Daniel Aili - Professor of Molecular Physics, Linköping University/ArgusEye
- Barry Walsh - Conference Director & Project Leader, Informa
- Automation and multiscale modeling can be integrated to accelerate purification development and aid in process understanding.
- Two mAbs with approximately the same isoelectric point had notably different loading and elution behavior in cation-exchange chromatography.
- Protein surface properties should be considered during development of chromatographic polishing steps, since isoelectric point does not tell the full story.
- Scott Altern - Senior Scientist, AbbVie
- Tingting Cui, Ph.D. - Associate Principal Scientist, AstraZeneca
- Nandu Deorkar, PhD - Vice President, Research & Development, Avantor
- Qian Ruan, Ph.D - Senior Vice president of TechOps and Supply Chain, Arcturus Therapeutics
- What do we see as standards and regulatory science needs to increase adoption of advanced technologies?
- How can AI be incorporated into smart and advanced manufacturing?
- How can regulatory agencies and industry work together to make safe and smart manufacturing a reality?
- James Coburn - Senior Advisor for Emerging Technologies at FDA’s Office of Counterterrorism and Emerging Threats, FDA
Recent advancements in mRNA technology have enabled mRNA-based vaccine and therapeutics entering a new era in medicine, however, the availability of established production facility is still limited. While mRNA production is much faster and platform design is more desirable than conventional cell-based vaccine, the tech transfer could still be challenging, especially across the globe. Arcturus’ PMDA approved first self-amplifying mRNA COVID vaccine exemplifies this challenge. With numerous tech transfers, the endeavour has been both enlightening and enjoyable. We would like to share the journey of our global tech transfers, including the raw materials, process and assay tech transfers, and lost in translation situations.
- Qian Ruan, Ph.D - Senior Vice president of TechOps and Supply Chain, Arcturus Therapeutics
Enhancing productivity and efficiency is essential for converting discoveries into viable treatments. Although the objectives during clinical phases differ from those in commercial manufacturing, their significance remains high. During clinical phases, the primary aim is to expedite processes without compromising quality. In contrast, commercial manufacturing focuses on consistent performance, high quality, and maximizing throughput, productivity, and yield to reduce the cost of goods (COGs). To achieve these goals, a comprehensive approach is necessary, incorporating various process technologies, materials, equipment, and analytical methods. This presentation will highlight how the selection and integration of bioreactor technologies, harvest techniques, purification processes, and analytical tools can synergistically improve productivity. We will showcase examples illustrating how these choices, combined with process design and optimization, lead to high levels of productivity and sustainability. Moreover, we will demonstrate that these strategies are applicable to both small and large-scale operations, including new and legacy facilities, as well as clinical and commercial manufacturing environments. By aligning process development with product design, significant productivity gains can be realized, ensuring efficient and cost-effective biopharmaceutical production.
- Natraj Ram - Vice President, Innovation, BioProduction Group, Thermo Fisher Scientific
- Rajeev Boregowda - Associate Director, Bioassay & Molecular Analytical Development, Genomic Medicine CMC, Sanofi
- Co-mixtures add complexity to the assessment of product quality and potency
- Compromises may be required at the formulation stage to enable acceptable product quality/stability for each API
- Agency expectations with regards to stability profiles for the individual API’s within the co-mixture compared to as individual products
- Sorting through the impurity profiles in the co-mixture
- Christina Vessely - Senior Consultant, Biologics Consulting
- Christoph Herwig - Senior Scientific Advisor, Körber Pharma Austria GmbH, Austria
Integrated and continuous biomanufacturing (ICB) processes offer higher throughputs, tighter process control, and reduced material usage, all within a smaller footprint. While ICB offers unique challenges to various biologic modalities, there are several difficulties that are relevant across the industry. One such pain point is the ability to acquire accurate real-time data to better enable enhanced process control for faster and more efficient unit operations, especially in a development environment that may lack harmonization of digital and analytical platforms. The convergence of digital and process analytical technologies (PAT) offers unprecedented opportunities to expedite bioprocess innovation. One key aspect to accelerating this innovation is through data ontology, the homogenization and organization of data across varied systems and analytical units present in development labs. Without a defined structure and organized data system, this can be an untenable task for a development group to engage in. Secondly, the physical orientation of units needs to follow a rational approach to interface with other systems that will be leveraged for advanced process control strategies. This is especially important in lab environments that may be siloed based on functional areas, frequently separating upstream, downstream, and analytical development activities. In this presentation, we will address both the digital infrastructure and lab design aspects for a fully integrated “Digital Lab” used to further R&D applications work in ICB development. This lab ties together analytical systems with upstream and downstream processing equipment using automated on-line sampling and integrated in-line sensors. The data accumulated is historized and retrievable through digital platforms for real-time and near real-time monitoring and control via process orchestration and modular unit operation integration. Automation strategies and control schemes enable practical use-cases such as glycosylation profile control in perfusion cell culture and aggregate control for flow-through polishing purification. With this framework, development teams can have a more holistic view and oversight of complex processes, work toward solving implementation challenges, and gain valuable insights into leveraging cutting-edge technologies to drive efficiency, quality, and innovation in ICB. |
- James Angelo - R&D Manager, MilliporeSigma
- Brent Morse - Principal Consultant, Dark Horse Consulting Group
- Diane McCarthy - Senior Director, Science and Standards, U.S. Pharmacopeia
- Greg Adams - Senior Director, Global Analytical Strategy & Development, FUJIFILM Diosynth Biotechnologies
Detailed and accurate information about a molecule’s characteristics is critical for production. This data allows scientists to make informed decisions, pushing the strongest drug candidates forward, while maintaining a competitive pace. Recent advances in analytical techniques have enhanced the speed and reliability of data collection, making it easier to gather critical insights. Here, we explore case studies, demonstrating how SCIEX solutions support the development of biopharmaceuticals from routine analysis to deep characterization of PQAs.
- Matthew Stone - Sr. Advanced Workflow Specialist, SCIEX
- Jun Li - Associate Director, Upstream Process Development, Gene Therapy, Ultragenyx Pharmaceutical
- Downstream process development: challenges with respect to downstream, in-process analytics, scale up, regulatory landscape.
- Potential ways of mitigating these challenges.
- Benefits of platform process as a business driver.
- Parag Kumthekar - Lead Gene Therapy Downstream Process Sciences, UCB Pharma
- Approaches to reaching high yield downstream purification
- Removing aggregates and contaminates from your titer
- Removal of partially-full capsids from your full-capsid yield.
- Seema Bhatlekar - Scientist, Johnson & Johnson Innovative Medicine
Mass photometry is a revolutionary technology designed for quick and accurate assessment of biomolecule purity, stability, and heterogeneity. We present here the applications of two instruments designed for gene editing vectors: the SamuxMP for AAV, and the new KaritroMP for characterization of adenovirus (AdV), lentivirus (LVV), and lipid nanoparticles (LNPs). With experimental times of minutes per sample, these instruments remove bottlenecks for in-process analytics that are conventionally encountered with slower methods like analytical ultracentrifugation (AUC).
- Gael Nicolas - Key Account Manager, Refeyn
- Harry Starkey - Senior Conference Producer, Informa Connect
Moore's Law, a foundational principle in the realm of technology, posits that the number of transistors on a microchip doubles approximately every two years, leading to exponential growth in computing power and efficiency while reducing costs. While not directly mirroring technology, the concept finds resonance in the rapid pace of medical advancements and pursuit of new modalities driven by exponential growth in knowledge, capabilities, and understanding. This accelerated pace enables medical breakthroughs but also poses technical challenges, specifically in manufacturing and delivery of new modalities like mRNA and other nucleic acid medicines. We are on the precipice of a revolution in R&D that marries the rapid advancement in modalities with targeted delivery vehicles and streamlined manufacturing to produce the next generation of medicine.
Through leveraging technological advancements in machine learning, at Flagship we have built an open end-to-end platform that harnesses vast knowledge, data, and tools to design and manufacture near infinite medicines with the highest possibility of translational success against target indications across the body. This relentless drive for innovation across the industry has the promise to expand the boundaries of what is possible exponentially to realize future of nucleic acid medicine.
- Hari Pujar, PhD - Operating Partner, Flagship Pioneering
Cell-based manufacturing of gene therapy molecules presents multiple challenges in maintaining high product yield and quality. Traditionally, isolating the desired DNA molecules is challenging. Additionally, cell-based approaches require long manufacturing timelines. To address these speed and quality challenges, we developed an enzymatic method capable of producing DNA drug substance at g/L scale and with >99% purity using chromatographic methods. This cell-free process has enabled highly accelerated production timelines compared to cell-based methods and the enzymatic platform facilitates diverse production application with constructs up to at least 7000bp in size.
- Anthony Dawson - Senior Scientist, Generation Bio
Explore our other tracks to curate your ideal conference experience, and discover exciting new content that sparks your curiosity!
- Introduction to Biopharmaceutical Life Cycle.
- Explain what upstream bioprocessing involves: the early stages of production, including cell culture and fermentation.
- Outline the key objectives: generating the desired biological product through cell growth and expression.
- Discuss the selection of cell lines (e.g., CHO cells, microbial cells).
- Introduce bioreactors and their role in providing a controlled environment for cell growth.
- Discuss different types of bioreactors (e.g., stirred-tank, wave, single-use) and their applications.
- Explain the fermentation process and its parameters (e.g., pH, temperature, oxygen levels).
- Explain the importance of culture media in supporting cell growth and productivity.
- Describe the process of scaling up from lab-scale to commercial-scale production.
- Highlight current trends in upstream bioprocessing (e.g., single-use technologies, continuous processing).
- Discuss future directions and innovations in the field.
- Michael Butler - Principal Investigator, NIBRT
- Explain what downstream bioprocessing involves: the purification and formulation of the biological product after cell culture and fermentation.
- Outline the key objectives: ensuring product purity, quality, and stability.
- Describe the process of harvesting cells or extracellular products from the bioreactor.
- Explain the methods used for cell separation (e.g., centrifugation, filtration).
- Introduce the main purification methods: chromatography, filtration, and precipitation.
- Describe different types of chromatography (e.g., affinity, ion-exchange, size-exclusion) and their applications.
- Explain the principles and applications of ultrafiltration and diafiltration.
Welcome To Group Therapy. Let's share the pain. Please join us and come prepared with your bioprocessing related problems. How it will work is we have 4 Round Tables each covering a different area of Bioprocessing. Each will be led by an expert. The expert will guide the discussion allowing participants to share their challenges and time to discuss within the group. These will last for 40 minutes. Included are some examples below:
Round Table Discussions
Cell Line Development & Upstream
- Expanding use of various cells: from Bacteria, yeast, and animal cell platforms to therapeutic cells
- The expanding nature of products: from therapeutic cells, to peptides, proteins, nucleic acids, and viruses
- Upstream sustainability: defining clear goals, establishing unambiguous metrics. and evaluating available options
- Upstream implications of AI: from process control, to supporting personalization, to heightened PAT, to continuous process verification
- Process intensification: Tools for increasing final product yield per cost, time, volume, footprint, personnel, and environmental burden generation
- Process, equipment, and suite flexibility: considering specific goals, performance and financial costs, and the various tools and solutions available
Recovery & Purification
- Standards and Validation: Meeting regulatory requirements and documenting processes for quality assurance
- Product Stability: Maintaining product integrity is essential for safety and efficacy. Degradation or aggregation can compromise the biologic's effectiveness and lead to safety concerns
- Yield & Efficiency: High yield and efficient recovery are crucial for the economic viability of the production process. Low yield or inefficient processes can significantly impact the cost and scalability
- Impurity Removal: Effective removal of impurities is vital to ensure product purity and safety. This includes removing contaminants like host cell proteins, endotoxins, and residual nucleic acids
- Cost and Time: Balancing cost and time with quality is essential for commercial success. High costs or prolonged production times can affect the economic feasibility of the biologic
- Solubility and Precipitation: Maintaining protein solubility and managing precipitation issues
Innovation & Adaptation:
- Compliance: Ensuring that new processes comply with existing regulations and standards can be challenging
- Compatibility: New technologies must integrate seamlessly with existing systems and workflows, which may require significant adjustments
- Data Integration: Managing and integrating data from new systems with legacy systems can be complex
- Economic Justification: Demonstrating the return on investment (ROI) and economic benefits of adopting new technologies
- Operational Disruption: Implementing new technologies can disrupt existing processes and workflows
- Cultural Resistance: Overcoming resistance to change within the organization
Cell & Gene Therapies
- Approval Processes: Lengthy and complex approval processes for new therapies
- Production Scale-Up: Difficulty in scaling up manufacturing processes from lab to commercial scale while maintaining consistency and quality
- Batch-to-Batch Variability: Managing variability between production batches
- Economic Viability: Demonstrating cost-effectiveness and economic viability for widespread use
- Material Sourcing: Securing reliable sources of high-quality raw materials and reagents
- Cold Chain Logistics: Maintaining cold chain logistics for sensitive products
- Customization: Customizing therapies for individual patients, which complicates standardization and scalability
- Long-Term Safety: Monitoring and ensuring the long-term safety of patients receiving cell and gene therapies
- Off-Target Effects: Minimizing off-target effects and unintended consequences of gene editing
- Advanced Analytics: Employing advanced analytics to develop and validate robust assays for measuring potency, purity, and safety.
- William Whitford - Founder, Oamaru BioSystems
- Christopher Brown - Technical Operations, Tourmaline Bio, Inc
- John Quarles - Director, Tourmaline Bio
- Mark Duerkop - Chief Executive Officer, Novasign
- Bob Castellucci - Founder and President, Partnership To Prosperity
- How do you avoid greenwashing and ensure your efforts are viewed as credible? What role does third-party verification play in that?
- What are the key needs and goals of pharma and biopharma laboratories when it comes to sustainability?
- Biotechnology and pharma are among the world’s largest carbon-emitting industries. What steps need to be taken in the future to lower their position in this list of carbon emitters?
- What are the most significant challenges or barriers that the pharma industry faces in implementing more sustainable practices?
- Renewable energy purchasing and PPA's
- The health sector generates four to five percent of total global emissions
- Mainly to do with the supply chain and the impact from globally shipping
- Challenges in contract manufacturing and M&A
- Focus on Scope 3 and how to address it: What is one cool industry collective action initiative
- Focus on product development (80% for design for sustainability), going upstream
- Complex global value chain- need to work together to solve sustainability and create circularity.
- James Connelly - Chief Executive Officer, My Green Lab
- Manu Juneja - Director – Global Sustainability, Takeda
- Christine Riley Miller - Executive Director of Global Responsible Business and Sustainability, BeiGene
- Byron Austin - Global Head of ESG, Sustainability, and Corporate Responsibility, Organon
To make a landing pad cell line that generates high titer TI clones requires screening many cell lines to find a landing pad in a hot spot in a hot cell. Here we describe a novel robust strategy of data mining to find a hot cell with a verified hot spot and convert it directly into a commercially viable landing pad cell line.
- Duncan McVey, Ph.D. - Senior Scientific Director, Bristol Myers Squibb
Transposase-mediated semi-targeted transgene integration systems deliver highly productive and genetically stable clones; however, it is sometimes challenging to characterize target gene integration sites for production cell lines with relatively higher gene copy numbers. This presentation will discuss the use of NGS and in silico predictive tools to support the generation of cell line stability data package to meet regulatory expectations.
- Hyo-Young Jeong,PhD - Senior Scientist, Process Cell Sciences, Merck & Co Inc.
Many of today’s expression platforms for recombinant protein expression have been optimised for the expression of monoclonal antibodies, often with the platform fundamentals originating from the 1990s. However, as new modalities increase in prevalence, particularly multichain asymmetric antibodies and bispecifics, there is a demand for improvements to expression vector design to enable optimal expression of these diverse formats. In this talk, we present on how Lonza is developing enhanced expression vector technology, and combining improved genetic instructions with high-throughput approaches to accelerate the discovery of new tools to improve titres.
- Bernadette Sweeney - Senior Director, R&D, Lonza Biologics, Lonza
This presentation describes a process characterization (PC) study that evaluated the effect of three vial thaw parameters using a fractional factorial design. This study aimed to identify an optimal thawing process that maximizes cell viability and density. Results from this study have contributed to the standardization and refinement of the vial thaw process, particularly through a DOE approach focused on two molecules, thereby establishing best practices for the vial thaw procedure platform
- Yuan Yuan - Sr Scientist, Cell Culture Development, Teva Pharmaceuticals
Glycosylation profiles are essential to the quality and potency of therapeutic biologics produced from CHO cells. Culture medium formulations significantly impact the N-glycosylation of biologics, which is critical for protein structure and bioactivities. In this case study, improvements in the culture medium by supplementing specific metal ion / small molecule components altered the N-glycan profile and enhanced its biological activity in-vivo.
- Yingxue Zhang - Scientist, Upstream Process Development, Jecho Labs
- Jason Souvaliotis - Associate Scientist, Biopharmaceutical Drug Substance Process Development, GSK
- Simona Serban - Director, Global Life Sciences Applications, Sunresin New Materials
- Aaron Moulin - Field Applications Specialist, Ecolab Life Sciences
In the slowly evolving landscape of bioprocess development and manufacturing, digital bioprocess-twins have emerged as potential accelerators. While advanced algorithms are at the heart of this endeavor, they are just one piece of the puzzle. The talk delves into key discussion points that are integral to this paradigm shift. The foundation of accelerated process development and automated process control starts with a clever experimental design, in-time data accessibility combined with powerful modeling algorithms. The talk will highlight the advantages of using hybrid modeling, while emphasizing the other critical aspects of his journey. Several industrial relevant upstream showcases for microbial and mammalian cell lines will be highlighted. Thereby, concepts to save experimental effort by up to 70% will be elaborated, and the modeling structure created in the late-stage development will be reused for real-time monitoring and control in the later stages. Additionally, a downstream optimization showcase for UF/DF/SPTFF will be highlighted.
- Mark Duerkop - Chief Executive Officer, Novasign
Continuous improvement methodology has been employed for many years to improve manufacturing where effectiveness depends on team experience. Advanced modeling technology for biomanufacturing promises to make continuous improvement methodology more widespread and allow teams to achieve expert results with far less experience. This presentation will illustrate model based continuous improvement across a drug substance biomanufacturing process using an industrial case study.
- Joseph Pekny - Professor, Chemical Engineering, Purdue University
- Steve Harding - President, Advanced Process Combinatorics, Inc.
Explore our other tracks to curate your ideal conference experience, and discover exciting new content that sparks your curiosity!
- Yiwei Zhao - Director, Analytical Development, Mural Oncology
- Jie Chen - Director, API Pilot Plant, AbbVie
- Thaddaeus Webster - Lead Scientist, Lonza Biologics
- Vamsikrishna Kandhi - Principal Scientist, Pfizer
- Ramani Raghavan - Vice President, Mersana Therapeutics
- Monica Sadek - Technical Development Scientist 3, Protein Analytical Chemistry, Genentech
This presentation delves into the evolving landscape of supply chain management for CGTs, offering insights into the latest developments that are reshaping the industry. The session begins with an overview of traditional supply chain practices, highlighting their limitations and the pressing need for adaptation in an ever-changing global market. From there, it transitions into an exploration of cutting-edge innovations such as artificial intelligence (AI), machine learning, blockchain technology, and Internet of Things (IoT). These technologies are revolutionizing supply chain processes, enhancing efficiency, visibility, and responsiveness across the entire network. We will also consider predictive analytics to digital twins and autonomous systems, which can empower organizations to anticipate disruptions, optimize inventory management, and streamline operations for maximum effectiveness. In addition, we will consider sustainability initiatives, circular economy practices, agile supply chains, and the integration of advanced robotics and automation. By offering a comprehensive overview of these innovations, tools, and trends, attendees will be equipped with the knowledge and insights necessary to navigate the complexities of modern supply chain management.
- Jared Auclair - Vice Provost Research Economic Development, Director of Bioinnovation, College of Science at Northeastern University
Many pharma and biotech companies onboarded thousands of materials and increased inventory during the pandemic. With the endemic in full swing, organizations have a surplus of material that is about to or has already expired. This presentation seeks to explain how the presenter worked to extend the shelf life of various materials for continuity improvement and financial savings.
- Michelle Parziale - Senior Engineer - MS&T, Moderna
- Jerry Keybl - Vice President, Avantor
- Leveraging AI to design personalized gene therapies based on individual patients' needs and target new, personalized delivery sites.
- Engineering multifunctional non-viral vehicles that combine gene delivery with imaging, additional therapeutic payloads, and controlled release capabilities.
- Utilizing AI to predict and optimize not just physicochemical properties, but also cellular interactions, biosensing, and response to specific triggers.
- Shashi Murthy, PhD - CTO and Co-Founder, Nanite Inc.
- Scalable methods to optimize EV Isolation & Purification
- Efficient techniques for manipulating EV properties (size, surface ligands, cargo loading etc.) for enhanced targeting and in vivo delivery
- Challenges and potential strategies for adhering to strict GMP regulations for clinical-grade EV production.
- Kumkum Saxena - Vice President, Preclinical R&D, Versatope Therapeutics
- Patrick Paez, PhD - Technical Marketing Manager, Aldevron
- How do you avoid greenwashing and ensure your efforts are viewed as credible? What role does third-party verification play in that?
- What are the key needs and goals of pharma and biopharma laboratories when it comes to sustainability?
- Biotechnology and pharma are among the world’s largest carbon-emitting industries. What steps need to be taken in the future to lower their position in this list of carbon emitters?
- What are the most significant challenges or barriers that the pharma industry faces in implementing more sustainable practices?
- Renewable energy purchasing and PPA's
- The health sector generates four to five percent of total global emissions
- Mainly to do with the supply chain and the impact from globally shipping
- Challenges in contract manufacturing and M&A
- Focus on Scope 3 and how to address it: What is one cool industry collective action initiative
- Focus on product development (80% for design for sustainability), going upstream
- Complex global value chain- need to work together to solve sustainability and create circularity.
- James Connelly - Chief Executive Officer, My Green Lab
- Manu Juneja - Director – Global Sustainability, Takeda
- Christine Riley Miller - Executive Director of Global Responsible Business and Sustainability, BeiGene
- Byron Austin - Global Head of ESG, Sustainability, and Corporate Responsibility, Organon
Acknowledged as an emerging cluster for biotechnology across the world, India’s biotech sector is valued at approximately $150 billion and is on track to reach $300 billion. This session will spotlight India’s cutting-edge research capacities, advanced biomanufacturing facilities, and expertise in biomanufacturing, bio-services, and drug development capabilities. The session will explore how tapping into India’s rapid growth and innovation propels advancements in global biomanufacturing.
- Raveesh Malik - Bachelor of Technology, Biotechnology, Senior Investment Specialist, Invest India
- Cheryl Scott - Editor in Chief, BioProcess International
- Sebastien Walpen - CEO, Ceidos SA
- Logan Nichols - Senior Research Associate II, Downstream Process Development, Ultragenyx
- Hunter Reese - Scientist II, Asklepios Biopharmaceutical
- Highlight the importance of ensuring viral safety in biopharmaceutical products
- Describe methods for viral clearance, such as viral filtration and inactivation
- Present a few real-world examples of successful downstream bioprocessing projects
- Discuss lessons learned and best practices from these case studies
- Highlight current trends in downstream bioprocessing (e.g., continuous processing, single-use technologies)
- Discuss future directions and innovations in the field.
- Andrew Zydney, Ph.D. - Bayard D. Kunkle Chair, Chemical Engineering, Penn State University
- Briefly explain the growing importance of digitalization, AI, and machine learning in the biopharma industry
- Key Areas of Digitalization
- Data Management and Integration
- Automation and Robotics
- Real-time Monitoring and Analytics
- Applications in Manufacturing and Supply Chain
- Use of digital twins and AI to optimize manufacturing processes and improve yield.
- AI-driven real-time monitoring and anomaly detection in production lines
- Use of AI for demand forecasting, inventory management, and logistics optimization
- Challenges and Considerations
- Case Studies
- Future Trends and Directions.
- Christoph Herwig - Senior Scientific Advisor, Körber Pharma Austria GmbH, Austria
- Hunter Reese - Scientist II, Asklepios Biopharmaceutical
- Amana Mohiuddin - Process Development Associate, Regeneron Pharmaceuticals
Cultivated meat offers a more sustainable solution to the growing population’s increasing hunger for meat. Here we examine sustainability aspects of cultivated meat, life cycle analysis (LCA), and novel nutrient streams. We also examine parallels with biotherapeutic bioprocessing and how cultivated meat looks to upend traditional biopharma paradigms. This is most evident in the sources and characteristics of raw materials, design specifications, cost, safety assessment, and mission of cultivated meat.
- Kalle Johnson - Senior Director, UPSIDE Foods
Gather around the 'fire' for a captivating conversation with our Keynote Speakers. Gain candid insights, get inspired, and dive deeper in a relaxed, conversational setting hosted by BioProcess International/BioProcess Insider Editors & Thought Leaders.
- Kalle Johnson - Senior Director, UPSIDE Foods
- Josh Abbott - Editor, BPI Insider, Bioprocess International
This presentation will cover the points to consider for enabling multi-use systems in a Next Generation Biotechnology (NGB) facility. Multi-use systems improve operability while reducing cost and facility environmental impact. A framework for enabling multi use systems will be proposed including a review of batch definition, genealogies, hold times, product quality assessments, microbial control, compliance, validation, and regulatory considerations.
- Brian Gregory Youchak - Sr. Advisor, Eli Lilly and Company
- Andrew Zydney, Ph.D. - Bayard D. Kunkle Chair, Chemical Engineering, Penn State University
- Stefano Menegatti - Associate Professor, North Carolina State University
- Andrew Falconbridge - Senior Independent Biotech Professional, Independent
- Can AI-ML benefit all stages of biopharmaceutical development (discovery, development, clinical, and manufacturing)?
- How can data quality and integrity concerns be addressed in AI-ML applications?
- How can companies ensure adherence to regulatory standards when employing AI-ML technologies?
- What cultural factors affect the acceptance and adoption of AI-ML within an organization?
- How crucial are collaborations and partnerships in driving AI-ML progress in biopharmaceutical manufacturing?
- How would you recommend an AI/ML novice gets started?
- Garima Thakur - Process Development Engineer III, Regeneron Pharmaceuticals
- William Whitford - Founder, Oamaru BioSystems
- Christoph Herwig - Senior Scientific Advisor, Körber Pharma Austria GmbH, Austria
- Marinna Madrid - Co-Founder & Chief Product Officer, Cellino
- Mark Duerkop - Chief Executive Officer, Novasign
- Lili Wang - Fellow, National Institute of Standards and Technology
- Lili Wang - Fellow, National Institute of Standards and Technology
- Ben Doak - Head of Innovative Treatments, NHS England
- Understanding emerging therapies: distinctions between cell therapy, gene therapy, etc.
- Therapeutic potential and current clinical landscape of different emerging therapies, unique challenges and opportunities presented.
- Differences and similarities from ‘traditional’ biologics:
- What learnings can we take from traditional modalities to approach novel modalities?
- Understanding the Cell Therapy and Gene Therapy manufacturing processes.
- Best practices when entering/transitioning into the advanced therapy industry.
- Leveraging experiences from your background into industry.
- Strategies and approaches to best utilise available technologies in the development & production of emerging therapies.
- Moving and translating research from academia, to start up, industry, and beyond.
- Understanding the difference between these, how to transition, pros and cons.
- Lessons and experiences from our panellists.
- Seema Bhatlekar - Scientist, Johnson & Johnson Innovative Medicine
- Tushar Patel - Director, Analytical CMC Strategy, Vertex Pharmaceuticals
- Kathryn Golden - SVP, Technical Operations and Cell Manufacturing, bit.bio
- Kevin Zen, PhD - Senior Director, IGM Biosciences
The finals competition winner announcement will run from 3:00pm-4:00pm
The event will showcase the BTI ’24 cohort of best-in-class life science tools companies and a pitch competition featuring the BioTools Innovator 2024 Finalists. In addition to startup presentations, the event includes panel discussions with industry experts, and a Final Competition where one winner will take home the $250,000 Grand Prize, selected by a live audience vote.
Attendees will have the opportunity to network with BioTools Innovator partners, including investors, and senior executives from Life Science tools companies and others.
The finals competition winner announcement will run from 3:00pm-4:00pm
The event will showcase the BTI ’24 cohort of best-in-class life science tools companies and a pitch competition featuring the BioTools Innovator 2024 Finalists. In addition to startup presentations, the event includes panel discussions with industry experts, and a Final Competition where one winner will take home the $250,000 Grand Prize, selected by a live audience vote.
Attendees will have the opportunity to network with BioTools Innovator partners, including investors, and senior executives from Life Science tools companies and others.