Cambridge Healthtech Institute’s 10th Annual

Rapid Methods to Assess Stability and Impurities in Biologics

Technologies and Strategies for Improving Prediction, Screening, and Quality

August 15 - 16, 2022 ALL TIMES EDT

The popular 10th Annual Rapid Methods to Assess Stability and Impurities in Biologics conference will bring together experts in formulation, analytical sciences, and process scientists to discuss rapid and practical ways to accelerate prediction and screening for protein instabilities and impurities arising from products, excipients, processes, and packaging in early- and late-stage development. Top scientists will share new insights through new presentations, informative panel discussions, high-level poster presentations, and interactive discussions on new methods and tools employed in real-time and accelerated stability studies, high-throughput analytics, developability assessment, high-throughput analytics, multi-attribute methods, post-translational modifications, and stability studies for traditional and novel biologics.

Monday, August 15

9:00 am Main Conference Registration and Morning Coffee (Grand Ballroom Foyer)

ROOM LOCATION: Back Bay B

9:55 am

Chairperson's Opening Remarks

Alex Dow, PhD, Associate Principal Scientist, Merck & Co., Inc.
Twinkle Christian, MS, Senior Scientist, Amgen, Inc.

A biotherapeutic is exposed to a variety of stress factors throughout its production. The drug manufacturer is responsible to deliver a safe and efficacious product to the patient by maintaining the critical quality attributes of a drug product during manufacture, transport, and use.  The scope of this talk is to assess the impact of packaging, transportation, and handling of drug product quality and discuss appropriate mitigations.

IMPROVING PREDICTION AND SCREENING

11:00 am

Autonomous Pipeline for Characterization of Biotherapeutics: Integrating Rapid Analytics with Rapid Informatics

Harsha Gunawardena, PhD, Senior Scientist, Mass Spectrometry, Janssen Pharmaceutical Companies of Johnson & Johnson

High-throughput screening tools are needed to triage product quality attributes of molecules and clones. We demonstrate high-throughput RapidFire-mass spectrometry analysis (i.e., 96-samples in ~ 20 mins) integrated with cloud-based protein informatics (~ 30 min/96 samples), to report and aggregate product quality attributes. This to our knowledge is the first demonstration of a complete end-to-end and scalable mass analysis solution that is hands-off, automated, and rapid (96 samples < 1 hr.).

David Cetlin, Senior Director, MockV Products, Cygnus Technologies

To determine viral clearance efficacy of biomanufacturing steps, viruses are “spiked” into in-process solutions, processed and analyzed for reduction.  Due to the infectivity of these viruses, studies are conducted in BSL-2 facilities.  Costs and logistics limit analysis during process development.  Discussed in the presentation are results from several studies that utilized CHO-endogenous RVLP and  non-infectious MVM VLP’s as viral surrogates. The results demonstrate the feasibility and value of adding viral clearance predication to downstream process development and optimization. 

12:00 pm Enjoy Lunch on Your Own

IMPROVING PREDICTION AND SCREENING, CONT.

12:50 pm

Chairperson's Remarks

Alex Dow, PhD, Associate Principal Scientist, Merck & Co., Inc.
12:55 pm

Machine Learning-Based Prediction of Single and Multiple Point Protein Mutations Stability Changes

Andrzej Kloczkowski, PhD, Professor, Pediatrics, Nationwide Children's Hospital

Accurate prediction of protein stability changes resulting from amino acid substitutions is of utmost importance in medicine to better understand which mutations are deleterious, leading to diseases, and which are neutral. Since conducting wet-lab experiments to get a better understanding of protein mutations is costly and time-consuming, computational tools based on machine learning are becoming promising alternatives to tedious and highly costly mutagenic experiments. We will review them and will present a robust machine learning-based methodology to predict the energy changes of proteins upon mutations.

1:25 pm PANEL DISCUSSION:

Formulation Development during the Pandemic: What Did We Learn?

Panel Moderator:
Björn Boll, Fellow Drug Product Design – Senior Expert for Particle Characterization and Analytics, ten23 health
Panelists:
Andrzej Kloczkowski, PhD, Professor, Pediatrics, Nationwide Children's Hospital
Sanket Patke, PhD, Associate Director, Sanofi
Harsha Gunawardena, PhD, Senior Scientist, Mass Spectrometry, Janssen Pharmaceutical Companies of Johnson & Johnson
1:55 pm Sponsored Presentation (Opportunity Available)
2:25 pm Networking Refreshment Break (Grand Ballroom Foyer)

ROOM LOCATION: Back Bay D

LC-MS IN HCP DETECTION AND CONTROL

2:40 pm

Detection and Quantitation of Host Cell Proteins in Monoclonal Antibody Drug Products Using Automated Sample Preparation and Data-Independent Acquisition LC-MS/MS

Jonathan Bones, PhD, Principal Investigator, Characterisation and Comparability Laboratory, National Institute for Bioprocessing Research and Training (NIBRT), Ireland

Data from LC-MS analyses of host cell proteins focusing on quantitation aspects and application to recombinant proteins and AAV-based gene therapy will be presented. Additionally, combination with ribosomal footprint profiling (Ribo-seq) to improve the depth and quality of the database used for MS data searching will also be discussed. Using this approach, we identified microprotein-based HCPs present in commercial drug products and investigated their behaviour during cell culture.

3:10 pm

Advanced LC-MS/MS Workflows for HCP Quantification, Including Optimized Standards and Data Independent Acquisition (DIA) MS Combined with Ion Mobility Separation

Christine Carapito, PhD, Co-Head of the BioOrganic Mass Spectrometry Laboratory, CNRS Research Director, University of Strasbourg, France

The implementation of new MS-compatible standards for robust and accurate quantification of Host Cell Proteins (HCP) will be presented. The potentialities of Data Independent Acquisition approaches for HCP profiling will be highlighted and benchmarked against commonly used targeted and Data Dependent Acquisition methods. Finally, benefits of the addition of an ion mobility separation in the LC-MS/MS workflow will be assessed on various instrumental platforms including High-Field Asymmetric-Waveform Ion-Mobility Spectrometry (FAIMS) and Trapped Ion Mobility Spectrometry (TIMS). Results will illustrate how the methods can support bioprocess understanding and development and ultimately be applied for final drug product purity assessment.

3:40 pm Session Break and Transition to Plenary Keynote

ROOM LOCATION: Constitution A&B

PLENARY KEYNOTE: SOLVING TODAY'S CHALLENGES

4:20 pm

Plenary Introduction

James Warren, PhD, Vice President, Pharmaceutical Development, Ultragenyx Pharmaceutical
4:30 pm

Lessons Learned from the Pandemic: mRNA-LNP Vaccine Development

Nicholas Warne, PhD, Vice President, Pharmaceutical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer Inc.

The speed and scale of industry response to the COVID pandemic was unprecedented, ultimately leading to the availability of several vaccines in under a year. This presentation will discuss the approach taken by Pfizer, with their partner BioNTech, in the development, manufacture, and distribution of the vaccine drug product while reflecting on lessons that may, or may not, be applicable to future product development.

5:00 pm

Advances in Vaccine Formulation and Stability

David B. Volkin, PhD, Distinguished Professor, Pharmaceutical Chemistry, University of Kansas, Lawrence

This presentation will provide an overview of analytical characterization and formulation development considerations for new vaccine candidates targeted for use in low- and middle-income countries (LMICs). Illustrative case studies with vaccine candidates (e.g., live-virus, adjuvanted recombinant protein) will highlight implementing state-of-the-art stability-indicating assays to enable development of stable formulations. Challenges with developing lower-cost formulations (e.g., multi-dose, combination, non-parenteral) to expand vaccine coverage in LMICs will also be discussed.

5:30 pm Welcome Reception in the Exhibit Hall with Poster Viewing (Grand Ballroom)
6:30 pm Close of Day

Tuesday, August 16

7:30 am Registration and Morning Coffee (Grand Ballroom Foyer)

ROOM LOCATION: Back Bay B

STABILITY & DEVELOPABILITY

7:55 am

Chairperson's Remarks

Dhanashri Bagal, Principal Scientist, Discovery Attribute Sciences, Amgen, Inc.
8:00 am

High-Throughput Transformation of Biologics Formulation Development Workflows

Hanlin Ouyang, Senior Scientist, Merck & Co., Inc.

The HTS group collaborated with formulation and analytical departments to build from scratch platform HT formulation screening workflows that covers study design, sample analytical plan, and data visualization. This ongoing workflow development synergizes cutting-edge robotic liquid handling system, HT study setups, HT analytical instruments, DOE JMP design, and data analysis to deliver richer information for faster and robust decision-making with less material requirement.

8:30 am

Assessing New and Sensitive Mass Spectrometry-Based Techniques to Rapidly Characterize Protein Therapeutics

Dhanashri Bagal, Principal Scientist, Discovery Attribute Sciences, Amgen, Inc.

Low material consumptive and rapid analytics performed on early-stage engineering panels of protein therapeutic candidates can inform on protein stability and developability. Herein we examine sensitive and robust mass spectrometry-based methods using online capillary SEC and HIC – LC/MS to inform on protein attributes such as aggregation and mispairing. We will also discuss a sensitive N-terminal chemical tagging and rpLC-MS/MS-based approach to accurately identify low-level proteolytic clips.

9:00 am

Challenges and Opportunities in Cell Therapy Drug Product Development 

Bharathi Vellalore, PhD, Senior Scientist, Biotherapeutics Drug Product Development, Janssen
  • Overview of drug product development from formulation, fill-finish, storage, to delivery 
  •  Formulation and process considerations to improve end-to-end drug product stability 
  •  Integrated drug product design to suit clinical and commercial supply chain needs
9:30 am Sponsored Presentation (Opportunity Available)
10:00 am Coffee Break in the Exhibit Hall with Poster Viewing (Grand Ballroom)
10:45 am Breakout Discussions

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: Stability and Developability of Novel Modalities

Bharathi Vellalore, PhD, Senior Scientist, Biotherapeutics Drug Product Development, Janssen
  • ​Analytical methods for high concentration biologics and cell and gene therapies
  • Phase-appropriate methods for process monitoring and control
  • Analytical methods for drug products characterization
  • Stability and impurities in novel modalities

SURFACTANT AND AGGREGATION

11:30 am

Identification and Rapid Characterization of Impurities Resulting in PS-80 Degradation by Proteomics and Charge-Reduced Mass Spectrometry

Shannon Rivera, PhD, Senior Scientist, Merck & Co., Inc.

Polysorbates are commonly used excipients in biotherapeutic formulations. These molecules may be degraded by process-related impurities, ultimately resulting in negative impacts on quality, efficacy, safety, and stability of the biopharmaceutical. Here we present work using two mass spectrometry techniques – 1) LCMS proteomics for identification of impurities and 2) RP-HPLC coupled with charge-reduced HRMS for rapid characterization of PS-80 degradation – used to identify sources of and track PS-80 degradation.

12:00 pm

Surfactants – Cause or Cure for Aggregates

Björn Boll, Fellow Drug Product Design – Senior Expert for Particle Characterization and Analytics, ten23 health

This presentation will highlight surfactants, e.g. polysorbate or poloxamer, as an important part of drug formulations responsible for stabilizing the active ingredient against interfacial stresses caused by shaking, stirring, or freezing. The stability of the surfactants within the shelf-life of the drug product is critical to ensure the quality of the final drug product since their degradation could lead to aggregation and even compromise the stability of the active ingredient.

12:30 pm Sponsored Presentation (Opportunity Available)
1:00 pm Luncheon Presentation (Sponsorship Opportunity Available) or Enjoy Lunch on Your Own
1:30 pm Refreshment Break in the Exhibit Hall with Poster Viewing (Grand Ballroom)

CHARACTERIZATION & CONTROL OF IMPURITIES IN BIOLOGICS: HCPs, SURFACTANTS, AND MORE

2:10 pm

Chairperson's Remarks

Kevin Zen, PhD, Executive Director, Chemistry Manufacturing and Controls, AnaptysBio, Inc.
Kevin Zen, PhD, Executive Director, Chemistry Manufacturing and Controls, AnaptysBio, Inc.

The impurities in therapeutic protein drug products can originate from raw material, bioprocess, product itself, and during patient dosing. This presentation will highlight potential impurities of therapeutic biotech products during upstream process and downstream purification and share the best strategy to mitigate and control such impurities. Some common queries from health authorities will be exemplified as case studies. 

2:45 pm

Impact of Four Inorganic Impurities on the Quality Attributes of a Fc-Fusion Protein

Alessandra Pistacchio, PhD, Biotech Pharmaceutical Development, Drug Product Development, Merck KgaA

Regulatory guidelines such as ICH Q3D provide Permitted Daily Exposure (PDE) limits for those impurities considered having a higher potential safety risk. However, one of the limits of such PDE values is that they account for the safety risk, while alterations of certain Quality Attributes of a biologic may also take place. To this aim, an Fc-fusion protein was treated with increasing concentrations of Ni2+, Cu2+, Zn2+ andFe3+ and analyzed under normal storage conditions, after 6 and 44 weeks of incubation at different temperatures (+5°C, +25°C, +40°C). The potential changes in conformation, oxidation, aggregation and fragmentation were monitored.

3:15 pm

Informing Downstream Control of Polysorbate Degradation Using a High-Throughput, Lipolytic Activity Assay

Alex Dow, PhD, Associate Principal Scientist, Merck & Co., Inc.

Polysorbate, a stabilizer within biologics formulations, is known to degrade via active, low-concentration lipases and esterases. The need for a rapid assay probing this specific HCP mechanism is required to improve the use of proper control strategies. A high-throughput, rapid activity assay using a fluorescent substrate allows for quantitation of lipolytic activity. This enabled an improved approach for the measurement of impurity impact and control.

3:45 pm Refreshment Break in the Exhibit Hall with Poster Viewing (Grand Ballroom)

ROOM LOCATION: Back Bay D

4:30 pm

Identification and Control of Active Enzymes for Polysorbate Degradation in Biotherapeutics by Activity-Based Protein Profiling

Shawn Li, PhD, Principal Scientist, Analytical Research and Development (AR&D) Mass Spectrometry, Merck & Co., Inc.

Enzymatic activity from residual host cell enzymes such as lipases and esterases plays a major role in polysorbate degradation. Their high activity, often at very low concentration, constitutes a major analytical challenge in the biopharmaceutical industry. In this study, we evaluated and optimized the activity-based protein profiling (ABPP) approach to identify active enzymes responsible for polysorbate degradation, which enables more meaningful polysorbate degradation investigations for biotherapeutic development.

5:00 pm

Best Practices for Internal Development of HCP Monitoring Kits

Olivier Ducoudret, Senior Development Specialist, Quality Control, Macrogenics

Host cell proteins (HCPs) are a class of process-related impurities during the manufacturing of biologics. HCPs must be removed in biologics manufacturing to ensure final product purity, manufacturing robustness, and safety. HCP analysis is often performed using an enzyme-linked-immunosorbent-assay (ELISA) due to method sensitivity and ease of use. When using an ELISA kit for HCP monitoring best practices involve determining kit sensitivity (quantification and coverage), product dilutional linearity, and robustness.

5:30 pm Close of Rapid Methods to Assess Stability and Impurities in Biologics Conference