Cambridge Healthtech Institute’s 10th Annual

Formulation and Delivery of High-Concentration Proteins and New Modalities

Overcoming Challenges in Viscosity, Aggregation, and Delivery with Formulation and Device Approaches

August 16 - 17, 2023 ALL TIMES EDT

The development of high-concentration protein formulations for traditional and novel biologic products can be challenging during manufacturing, storage, and delivery due to problems such as aggregation, precipitation, gelation, and high viscosity. The popular 10th Annual Formulation and Delivery of High-Concentration Proteins and New Modalities conference will feature informative, high-quality case studies and successful strategies to overcome the problems you are facing with the development and delivery of high-concentration protein formulations, new modalities, and cell and gene therapies and mRNA therapies.

Wednesday, August 16

Registration and Morning Coffee7:30 am

HIGH-CONCENTRATION PROTEIN FORMULATIONS

7:55 am

Chairperson's Opening Remarks

Namita Sawant, PhD, Principal Scientist, Amgen

8:00 am

Sequence Engineering to Improve the "Syringeability" of High-Concentration Monoclonal Antibody Formulations

Georgina Armstrong, PhD Student, Senior Scientist, BioPharm Process Research, Drug Substance Development, GSK

The trending preferential parenteral route of administration of monoclonal antibodies is via subcutaneous autoinjectors with increased patient autonomy, compliance and reduction in healthcare trips and costs. A notable source of device failure is from highly viscous protein solutions, requiring increased force for full injection. A multitude of studies have focused on alterations in formulation composition and there are increased emerging trends in device engineering solutions. However, an earlier and alternative cost and time-effective strategy is introducing single point mutations in the variable domains of mAbs. This project focuses on the latter, with unique aims to holistically characterize biophysical profiles of candidate mutants as well as their viscosity.

8:30 am

In silico-Based Design of High-Concentration Protein Formulation

Maral Adeli Koudehi, PhD, Scientist, Drug Product Development, Johnson & Johnson

Advanced AI/ML models can be applied to protein science, specifically in high-concentration antibody formulation. In silico methods improve the selection process and design steps in protein development. This presentation summarizes strategies being developed and implemented for structural assessment of biotherapeutic proteins in combination with designing high-throughput formulation using modeling and simulation.

9:00 am

Evaluation of a Silicone-Free Syringe and Stopper Presentation for Use in Biopharmaceutical Drug Product Development

Cait Sofa (Quaile), Principal Scientist, Biopharmaceutical Drug Product Development, GlaxoSmithKline

Subcutaneous biopharmaceutical drug products are often supplied in a siliconized syringe with stopper. Levels of component siliconization vary from lot to lot leading to different levels of drug product-silicone interactions during storage. This presentation includes an assessment of product quality for three drug products using two syringe systems, an unsiliconized glass syringe with polytetrafluoroethylene coated stopper, and a siliconized syringe and stopper. The study demonstrates that an unsiliconized presentation is a suitable alternative to traditional siliconized presentation.

9:30 am Pfanstiehl High Purity Carbohydrates and Amino Acids for Biopharma Upstream, Downstream and Formulation Applications

Sudhakar Voruganti, PhD, Director, Business Development, Pfanstiehl, Inc.

Pfanstiehl high purity Galactose & Mannose for upstream applications

  • Background 
  • Glycosylation and role in protein therapeutics
  • Importance of high purity Galactose & Mannose

Pfanstiehl Carbohydrates & amino acids for downstream and formulation applications

  • Advantages of Trehalose over Sucrose 
  • Amino Acid Buffers in commercial antibody formulations  
  • Importance of utilization of highly characterized AAs as Excipients  
  • Amino Acids as viscosity lowering Excipients
  • Methionine as Biopharmaceutical Stabilizer Antioxidant
9:45 am Novel Recombinant Hyaluronidase for Subcutaneous Delivery of Biologics

Hojun Choi, Executive Vice President, Chief Strategy Officer, BMI KOREA

BMI introduces BMI-2004, a novel recombinant ovine sequenced hyaluronidase for improved subcutaneous biologic delivery. The developmental goal of BMI 2004 is minimize the change of original Mab formulation. The main advantages of the enzyme is 1) the same pH range to Mab at pH 5, 2) highly concentrated form, 3) proprietary liquid formulation technology of hyaluronidase enzyme. Phase1 trial (n=250) and pre-clinical study results will be shared. 

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

HIGH-CONCENTRATION PROTEIN FORMULATIONS (CONT.)

10:40 am

High Concentration Monoclonal Antibody Formulations: Critical Insights and Key Learnings

Namita Sawant, PhD, Principal Scientist, Amgen

Development of a high concentration monoclonal antibody formulation poses multiple challenges. Often, accelerated timelines and clinical targets to achieve a high dose require a fine balance between speed-to-market and development of a product for patients that meet the required viscosity and product quality specifications. This presentation will focus on high throughput formulation development strategies enabling faster product development with reduced material usage. Additional aspects, including device-related viscosity constraints, associated rheological characterization, and product quality assessments relevant to high concentration formulations will be covered.

11:10 am

Upper Concentration Limits of mAb Therapeutics—Strategies for High-Concentration (>200mg/ml) Drug Products

Ian Roy, Scientist, Formulation, Janssen R&D

  • Overconcentrating mAbs generates a web of problems. Most of the challenges arise because of increased viscosity or decreased stability.While viscosity does not directly present itself as an 
  • upper bound for DP concentration, the variability in viscosity and other formulation characteristics brought on by high concentrations does represent an effective limit to concentration.
  • The maximum concentration for a DP is a compromise between acceptable DP variability due to manufacturing ranges and a shrinking stable formulation space.


11:40 am

Strategies in the Development and Manufacturing of Low-Viscosity, Ultra-High-Concentration Formulation for IgG1 Antibody

Vaibhav Deokar, Principal Scientist, Formulation Development, Biotechnology Division, Lupin Ltd.

The present research provides a comparative evaluation of scalable manufacturing strategies to develop low viscosity (150mg/mL) formulation for lyophilized biosimilar IgG1; suitable for single, subcutaneous injection ~600mg/3.0mL, per dose.IgG1 was concentrated to ~200mg/mL and provides a comparative evaluation of manufacturing strategies and their impact on the chemical and structural stability of IgG1. Techniques used for the concentration of IgG1 are tangential flow filtration (TFF), spray drying (SPD), and spray freeze drying (SFD).

Enjoy Lunch on Your Own12:10 pm

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

IMPORTANT FORMULATION DEVELOPMENT CONSIDERATIONS FOR BIOLOGICS

1:25 pm

Chairperson's Remarks

Jennifer Litowski, PhD, Senior Principal Scientist, Drug Product Technologies, Amgen, Inc.

1:30 pm

In the Patient’s Hands: In-Use Compatibility Testing and Strategies

Jennifer Litowski, PhD, Senior Principal Scientist, Drug Product Technologies, Amgen, Inc.

Drug products undergo handling by patients and healthcare professionals, including reconstitution of lyophilized products, transfer to syringes, and dilution into IV bags for administration. In-use compatibility studies are performed to show that stability and concentration are maintained, ensuring patient safety and accurate dosing. However, regulatory expectations are not well-defined, leading to uncertainty in study parameters. Recent cross-product data and a multi-company collaboration offer guidance for streamlined experimental design.

Presentation to be Announced2:00 pm

Sponsored Presentation (Opportunity Available)2:30 pm

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

DRUG-DEVICE COMBINATIONS

7:55 am

Chairperson's Remarks

Christoph Brandenbusch, PhD, Assistant Professor, Bioprocess Separations & Biologics Formulation Development, TU Dortmund University

8:00 am

Variables Impacting Silicone Oil Migration and Biologic Drug Product in Prefilled Syringes 

Xi Zhao, PhD, Senior Scientist, Sterile and Specialty Products, Merck

Despite the benefits that Prefilled Syringes (PFS) can provide to patients, the silicone oil pre-coated on the glass barrel may migrate into the drug product and impact drug product quality. Due to current supply chain shortages and procurement preference for commercial product, the PFS source may have to change in the middle of development and a need of establishing source duality is also required by health authorities. The presentation focuses on risk assessment of silicone oil migration to enable a thorough and optimal selection of primary container closure and de-risk the impact of silicone oil on drug product quality.

8:30 am

Addressing Challenges in Combination Product Development for SC Delivery

Ajit M. D'Souza, PhD, Director Combination Product Development & Manufacturing, Combination Product Development & Manufacturing, Kiniksa Pharmaceuticals Corp.

As healthcare systems increasingly rely on medical care administered at home, the focus on ease of administration, safety, compliance and sustainability is driving the development of combination products for home use. This presentation will discuss approaches to addressing three key challenges: i) drug-device compatibility, ii) reliability and robustness of performance and iii) usability issues in the context of high concentration mAb formulations.

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 17 - Formulation and Stability Considerations Delivery for Proteins and New Modalities

Christopher P. Locher, PhD, Co-Founder & CEO, Biology, Versatope Therapeutics

  • Polypeptide composition and glycosylation profiles
  • Lyophilization,gelatin, simple sugars and amino acids as excipients and Ionic and zwitterionic buffers and pH to support solubility and reconstitution
  • Silanized glass vials and drug/product silicone interactions​

PLATFORM TECHNOLOGIES, FORMULATION, AND DELIVERY TECHNOLOGIES

10:30 am

Next-Generation PEG: Application towards Safer mRNA Vaccine

Soumen Saha, PhD, Senior Research Scientist, Duke University

In 2022, FAERS showed over 39,657 adverse event reports related to the COVID vaccine, an incredible ~440% increase compared to 2020. There are at least ~350,000 individuals in the USA with known PEG allergy. These numbers highlight why a PEG-free vaccine option is needed, and why all COVID vaccines carry an FDA contraindication warning to patients with known allergies to PEG. For the realization of the full potential of this technology, a safe and functional replacement for PEG is urgently needed that exhibits several desirable features of PEG while maintaining equivalent capabilities for mRNA delivery, comparable to current products.

11:00 am

Formulation Development Considerations for AAVs

Paria Moxley, PhD, Scientist, Biologics Drug Product Development & Manufacturing, Sanofi

Recombinant adeno-associated virus (AAV) has emerged as a promising gene delivery vector for the treatment of various diseases. There are marked differences in buffer selection for formulation development with AAVs and protein therapeutics, which must be considered in the context of product manufacturing, long-term storage, in-use administration, and shipping/handling. This entails screening for buffer pH, ionic strength, and the impact of added surfactants on stability/degradation trends of drug product.

Enjoy Lunch on Your Own11:30 am

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

LNPs & NOVEL DELIVERY SYSTEMS: FORMULATION, ANALYSIS, PROCESS DEVELOPMENT, AND DELIVERY

1:05 pm

Chairperson's Remarks

Amey Bandekar, PhD, Associate Director, Drug Product Development, Sanofi

1:10 pm FEATURED PRESENTATION:

Advances in Chemistry Made RNAi Therapeutics Possible

Mano Manoharan, PhD, Distinguished Scientist & Senior Vice President, Innovation Chemistry, Alnylam Pharmaceuticals

For siRNAs, chemical modifications are necessary to regulate metabolic stability, potency (through effects on the interaction with the Ago2 enzyme and the targeted mRNA strand), and safety (impacted by metabolites and on-target specificity). We have evaluated numerous chemical modifications beyond the standard 2’-O-methyl, 2’-fluoro, and phosphorothioate linkages. These include backbone chiral phosphorothioates, glycol nucleic acids, altriol nucleic acids, gem 2′-deoxy-2′-α-F-2′-β-C-methyl, 5’-morpholino, and amino-oxy click chemistry (AOCC) mediated conjugates. Furthermore, novel spatial architectures like circular siRNAs have also been evaluated. This presentation will summarize how chemistry has made possible the currently exciting world of RNAi therapeutics.

1:40 pm

Equilibrium and Stability Considerations in the Development and Manufacturing of Liposome and LNP Formulations

Christoph Brandenbusch, PhD, Assistant Professor, Bioprocess Separations & Biologics Formulation Development, TU Dortmund University

Liposome and LNP formulations have evolved as highly potent drug delivery systems. Recent literature gives valuable insights in developing and manufacturing these formulations, with focus mainly set on delivering different liposome and LNP compositions/sizes. This presentation will give some insight into thermodynamic equilibrium considerations, such as long-term (size) stability of liposomes and LNPs at various temperature, as well as equilibrium radius considerations in manufacturing liposomes and LNPs.

2:10 pm

Considerations and Challenges in Early LNP Development for Non-Viral Gene Therapy

Yuefei Shen, PhD, Principal Scientist, CMC Drug Product Development, Sanofi

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). Compared to intramuscular vaccine delivery, intravenous (i.v.) delivery of a LNP formulation for gene therapy shows unique challenges. An LNP formulation for gene therapy via i.v. may require different lipid and formulation design. Here, we will discuss the considerations and challenges in early LNP development and impact on tissue targeting for NVGT.

Networking Refreshment Break2:40 pm

LNPs & NOVEL DELIVERY SYSTEMS (CONT.)

2:55 pm

Process Development Considerations for LNP Manufacturing

Amey Bandekar, PhD, Associate Director, Drug Product Development, Sanofi

In the development of LNP-based drug product, the choice of manufacturing technology is one of the key factors for success. The choice of technology can have a significant impact on the biophysical properties, structural characteristics, colloidal stability, and efficacy of the LNP. This study describes the impact of different manufacturing parameters and the scale-up consideration to enable successful LNP drug product manufacturing.

3:25 pm

De-Risk mRNA Adduct Formation in Lipid Nanoparticle Formulation Intended for Glycogen Storage Disease Type-1a Using Analytical Tools

Siddharth Bhoraskar, PhD, Scientist II, Beam Therapeutics

In this talk, we discuss de-risking mRNA adduct formation in lipid nanoparticle formulation intended for glycogen storage disease type-1a using analytical tools.

Close of Summit3:55 pm