Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 13th International Conference on Biopharma and Biotherapeutics Boston, Massachusetts, USA.

Day 1 :

Keynote Forum

Francois Xavier Frapaise

Biosimilars Consultant Paris, France

Keynote: The end of clinical trials in biosimilars development?

Time : 09:15-09:45

Conference Series Biopharma 2018 International Conference Keynote Speaker Francois Xavier Frapaise photo
Biography:

Francois Xavier Frapaise, M.D has over 30 years of international drug development experience at major pharmaceutical companies including Sanofi, Bayer, Boehringer, Merck and Abbott; he has held multiple C- level positions (CSO, CMO, CEO) in different pharmacies in the US and Europe. He has extensive experience of biosimilars development (Boehringer-Ingelheim, Pfenex, Merck KGaA) and is currently heading a biosimilar consulting company. He has been the CSO and SVP of Optimer, has served as the CEO of Asphelia Pharmaceuticals, Inc. VP R&D and corporate officer of TAP, CMO of Ocera Therapeutics, VP of Scientific Affairs at Abbott International, Head of Medical Affairs at Bayer Europe, Medical Director at Bayer France, VP of R&D at Delagrange, Head of Anti-thrombotics Strategic Marketing at Sanofi, Medical Director at Choay. He holds an M.D. degree from Faculté of Medecine and holds an academic position at the Thrombosis Research Center at the Loyola Medical Center in Maywood (IL).

Abstract:

In his book, “The End of History?”, published in the National Interest in 1992, Fukuyama argues that the advent of western liberal democracy may signal the endpoint of humanity's sociocultural evolution and the final form of human government. In the same vein, based on recent development of sophisticated analytical methods and evolution of regulatory guidance, one should discuss the relevance of the current clinical development model for biosimilars and raise the following long-termquestions: is it the end of clinical trials in biosimilar development,” or more realistically, “could we, in the foreseeable future, drastically reduce the financial and operational burden of biosimilars clinical trials?” It is well known that proteins have unique structural organisation patterns and even those that are chemically identical may have different biological effects due to structural folding differences, without mentioning the effect of post-translational modifications. However, with the current exponential development of multiple sophisticated analytical methods enabling comparability assessment between originators and candidate biosimilars both structurally (orthogonal methods) and functionally (compound-dependent), considering the lack of sensitivity of many clinical models to detect meaningful differences between follow-on biologics and reference compounds and the recent evolution of regulators perspective on this matter, the relevance of the current clinical development model should be legitimately questioned. Even if this is not ready for prime time, we observe, through the interaction with regulators, that current trends are compatible with this possible long-term perspective, namely: The reliance on healthy
volunteer PK/PD studies only to support some biosimilar compounds registration (without testing the candidate biosimilar in
oncology patients), wider acceptance by the FDA/EMA of extrapolation from one indication to all other approved indications
of the reference compound (e.g. infliximab) and agencies’ willingness, if not encouragement, to test clinical biosimilarity in
non- approved indications of the originator compound! When one considers the financial and operational burden of running pivotal trials based on clinical endpoints (skeletal-related events) in some indications (e.g. osteoporosis, metastatic prostate cancer), the likely acceptability of surrogate markers (BMD in osteoporosis), one can predict that clinical trial designs and endpoints for biosimilars are likely to significantly evolve in the coming years. However, one important question remains open, at least for immunogenic compounds (e.g. adalimumab), namely the opportunity to predict immunogenicity based on nonclinical models.

Conference Series Biopharma 2018 International Conference Keynote Speaker Peter H Kalinka photo
Biography:

Peter H Kalinka is chairman and principal consultant at Longmore 60 Biotech Inc. He possesses in-depth knowledge of drug development and directed numerous development projects including Therapeutic Proteins, Monoclonal Antibodies and Fusion Proteins. His experience in overall development spans cloning, process development, scale-up, (e-coli, CHO, Hybridoma etc.) analytical development, bioassays, pre-clinical, clinical Phases, to manufacturing and regulatory affairs. Working with more than 20 companies worldwide, he has directed all or spearheaded parts of development programs for biosimilars, biobetters and biologics.

Abstract:

When utilizing different cell lines, expression, productivity and upstream process design, in general, need to be adapted to the metabolism and behavior of these cells. Bacterial systems have the advantage of quickly reaching high cell densities but slow when it comes to protein expression. Apart from that, bacterial systems cannot glycosylated proteins which makes them unsuitable for the production of monoclonal antibodies and many therapeutic proteins. There are also big differences when looking at mammalian cells such as CHO and Hybridoma cells. This workshop will discuss the nature of these issues and why they necessitate very different ways of upstream processing.

Keynote Forum

Thomas J Webster

Northeastern University, USA

Keynote: The influence of nanomedicine on drug discovery

Time : 10:15-10:45

Conference Series Biopharma 2018 International Conference Keynote Speaker Thomas J Webster photo
Biography:

Thomas J Webster is the Chemical Engineering Department Char and Art Zafiropoulo Chair at Northeastern. Prof. Webster has graduated 144 students. His lab group published 9 textbooks, 48 book chapters, 403 articles and 32 provisional/full patents. He has received numerous honors: 2012, Fellow, American Institute for Medical and Biological Engineering; 2013, Fellow, Biomedical Engineering Society; 2015, Wenzhou 580 Award; 2015, Zheijang 1000 Talent Program; 2016, Fellow, Biomaterials Science and Engineering; and 2016, Acta Biomaterialia Silver Award. He also frequently appears on the BBC, NBC, ABC, Fox and other news outlets talking about science.

Abstract:

Nanotechnology has already revolutionized disease prevention, diagnosis and treatment, as evidenced by a significant number of FDA, approved materials. In fact, many studies have indicated that nanoparticles can act as drugs and even replace pharmaceutical agents. This talk will first cover advances in the use of nanoparticles as improved drug agents to treat cancer, infections and grow tissues. Moreover, this talk will highlight how the development of nanoparticles has influenced traditional drug discovery and development. There has been a noted increase in combined nanoparticle – drug approaches but some believe the development of nanoparticles has decreased efforts in traditional drug discovery and delivery. This talk will also highlight FDA approved nanoparticles and what is necessary for the field to continue to grow.

Keynote Forum

Candida Fratazzi

Bull Breed Coalition Registry Consulting, Massachusetts

Keynote: E-BABE: How to address immunogenicity in the development of a rare disease biosimilar

Time : 11:00-11:30

Conference Series Biopharma 2018 International Conference Keynote Speaker Candida Fratazzi photo
Biography:

Candida Fratazzi is a Founder and President of BBCR consulting. Leveraging extensive experience leading early clinical development and developing regulatory strategy, BBCR consulting is a valuable asset for a company requiring advisory related to life science product development; specifically, pipeline and clinical plan assessments, clinical trials, GTM strategy, safety, or biomarker. BBCR team areas of expertise include rare diseases, immune-oncology, immunology, inflammation, risk-benefit management, regulatory compliance and FDA issues. BBCR team is a specialized in orphan, autoimmune, oncology and CNS indications for an innovator, biosimilar and repurposing products. BBCR team has contributed innovative clinical regulatory strategies and trial design for products directed to regulatory compliance across international government entities for drug, device and combination products. BBCR team leads evidence-based trial strategies, cost-effective solutions reduces risks and facilitate patient centricity, recruitment and retention she is an accomplished Senior Executive MD with more than 25 years of success spanning biotechnology, pharmaceuticals and device. She led market approval for five major products, improved endpoint achievability and designed Natural History studies and registry protocols. An industry leader, she is a sought-after speaker presenting at international industry summits. Candida attained her MD and a higher degree in Immunology and trained at The Johns Hopkins University and The Harvard University 

Abstract:

Rare diseases hugely need moderate treatment choices. Vagrant medications have been ignored by the biosimilar business for quite a while. Patient’s advocates that people with rare diseases have access to safe and effective biologic and biosimilar medicines. Worldwide, 350M people are estimated to suffer from a rare disease, including 25-30M US and 30M EU residents. With >60% biologics represent most of the worldwide vagrant medication advertise. A noteworthy issue with protein-based therapeutics is their immunogenicity.Forms of an immune response are the activation of B cells and T-cells, which help to activate B cells. The T-cells react in a typical response to a fake protein remedial as though it were outside since it is unique in relation to the imperfect, common protein. A T-cell reaction confound like this occasionally happens on account of the protein FVIII. Virtually all therapeutic proteins (biologics) evoke an insusceptible reaction with the resulting creation of hostile to medicate antibodies (ADA). The ADA to therapeutic monoclonal antibodies (mAbs) that are coordinated against the antigenbinding
site of the therapeutic mAb is neutralizing. This nature of the ADA reaction explains why fully human antibodies can still be exceedingly immunogenic. Biosimilars have to be tested for their immunogenicity as it is impossible to predict if they will induce an immunogenicity similar to the one manifested by the corresponding innovator biologics. Infusion-Related Reactions (IRRs) include hypersensitivity reactions and cytokine release syndromes. Hypersensitivity reactions have classically been partitioned into type I, II, III and IV reactions; type I and III reactions are those regularly observed following administration of biologics. The infusion-related reaction is defined as a disorder characterized by an adverse reaction to the infusion of pharmacological or biological substances and as a disorder characterized by nausea, headache, tachycardia, hypotension, rash and shortness of breath and caused by the release of cytokines. Infusion-related reactions are common and timely related to drug administration and have been reported as anaphylaxis, anaphylactoid reactions and cytokine release syndrome, among other terms used. Animal toxicology studies are neither predictive of severe IRRs nor of hypersensitivity in human. With respect to intravenous (IV) administration, the SC route offers more convenience to patients, flexibility in dosing and potential to lessen medicinal services costs. There is a perception that SC organization can represent a higher immunogenicity hazard than IV administration for a given protein.However, a recent comparative clinical study of sc vs iv administration of abatacept showed that the efficacy and immunogenicity are comparable between the two routes of administration.

Conference Series Biopharma 2018 International Conference Keynote Speaker Leigh Ann M Durant photo
Biography:

Leigh Ann M Durant, Esq., is a senior legal and business executive with more than 24 years of experience in the global biopharmaceutical industry. She is currently the Head lawyer for all R&D activities in the US at EMD Serono, Inc., the US affiliate of Merck KGaA, Darmstadt, Germany. She also serves as an independent director on a privately-held biotech company. Earlier in her career, she was a trial lawyer and Partner at Nixon Peabody LLP and law clerk for Chief Justice Weisberger of the Rhode Island Supreme Court. Recipient of the national Top 30 Emerging Leaders in the Pharmaceutical Industry Award, Woman of the Year in Law Award and Top 10 Women Lawyers in Massachusetts Award, for over two decades she has served on a variety of boards and executive committees, has led various legal and non-profit organizations as President and speaks nationally on a variety of topics

Abstract:

Statement of the Problem: Biopharma’s 50-year old drug development model – which focuses on products and healthcare providers, rather than patients and health outcomes – is failing to meet modern-day stakeholders needs. Technology-driven patients and providers, evidence-focused regulators and cost-conscious payors are demanding more data, more innovation, improved health outcomes and answers to questions that matter most to patients, all of which are inspiring a radical shift in our drug development paradigm. A patient-centric drug development model offers solutions to these challenges. This paper provides an overview of some of the latest developments in this new model and paradigm shift, including conceptual definitions, widely adopted patient-centric activities, new regulatory guidance and a novel approach to estimating financial value of patient engagement, all of which provide opportunities to maximize innovation.
Methodology and Theoretical Orientation: This paper was developed from a systematic literature review of peer-reviewed and gray literature using PubMed and Google and a synthesis of peer-reviewed literature, reports, FDA guidance documents, recommendations, findings and observations from regulators, academic research centers, patient advocacy organizations, nonprofit
coalitions and commentators.
Findings: Despite wanting to meet stakeholder needs and an eagerness to embrace patient-centricity, adoption of patientcentered
drug development has been challenging for biopharma. Some of the latest developments in this area provide additional justification and validation for embracing this paradigm shift, however, new strategies, organizational structures and enterprisewide culture change are also needed to ensure wholesale organizational adoption and permanence.
Conclusion and Significance: Latest developments further support that patient-centered drug development reduces development timelines, improves data quality, increases probabilities of technical and regulatory success and increases return on investment. However, changes in strategies, structures and culture must occur to disrupt and supplant the existing productfocused drug development model and transition successfully and permanently to the new patient-centric model, enabling biopharma to maximize innovation.

Conference Series Biopharma 2018 International Conference Keynote Speaker Aruna Dontabhaktuni, photo
Biography:

Aruna Dontabhaktuni is an internationally renowned pharmaceutical professional with 23 years of experience across all phases of drug development. She received her Pharmacy degree from Karnataka Pharmacy College, India and PhD from Long Island University, New York City. She is the founder and CEO of PharmaPro consulting, her company specializes in clinical pharmacology and regulatory submissions. She also serves on the scientific board for several pharmaceutical and biotech companies as a subject matter expert in clinical pharmacology. In her carrier, she has supported 30+ compounds from early, to late-stage of drug development, in oncology & immunology, other rare diseases including breakthrough therapy designations application for Olaratumab (LARTRUVO™). She has played the pivotal role in mentoring the next generation of scientists, thorough her managerial responsibilities. Her 50+ publications including posters, manuscripts and conference presentations as a keynote speaker at national and international conferences, are a clear testament to her achievements 

Abstract:

What does it take to qualify for the expedited FDA Drug approval? What is the breakthrough therapy designation application and how is it impacting the global drug development? In 2012, Congress created the breakthrough-therapy designation to expedite the testing and approval by the FDA, of medications with the potential to provide the substantial improvement over existing treatments. “Breakthrough Therapy Designation” is improved to hasten the growth and evaluation that are meant to treat the serious condition and introductory clinical proof indicates that the drug may reveal considerable increment over an available therapy on a clinically significant endpoint. To control whether the advancement over available therapy is considerable is a matter of discernment and depends on both the magnitude of the treatment effect, which could include the duration of effect and the elevation of an observed clinical outcome. In general, the primary clinical proof should show a clear advantage over available therapy. How the recent trends in globalization and the complexity of drug development have resulted in the possibility that expedited programs in one country may now influence drug development in another

 

  • Current Challenges in Developing Biosimilars | Clinical Development of Biosimilars | Biosimilars Analytical Strategies | Biosimilars Companies and Market Analysis | Current Trends in Pharmaceutical Industries | Challenges in Biosimilars Pharmacovigilance
Location: Boston, USA
Speaker

Chair

Peter H. Kalinka

Longmore 60 Biotech Inc., USA

Session Introduction

Neil Schauer

Schauer Biologics Consulting LLC, USA

Title: Risk based development and manufacturing of biosimilars
Speaker
Biography:

Neil Schauer is the Principal Consultant at Schauer Biologics Consulting where he provides strategic planning and tactical support for process development and manufacturing activities at startup and established biologics companies. At Avaxia Biologics, he was Senior VP of Technical Operations; where he was responsible for process development, manufacturing, quality and program management. In previous positions (Inspiration Biopharmaceuticals, Hospira, Millipore, Biogen Idec and Genetics InstituteWyeth), he held positions of increasing responsibility associated with biologics: technology, manufacturing, process development and projectprogramportfolio management.

Abstract:

The development of biosimilars presents a unique risk profile to biopharmaceutical companies when compared to proprietary biologics development. This risk profile will be examined and used to explain why biosimilars must be developed and manufactured differently than originator molecules, in order to insure commercial success. For biosimilars, commercial
endpoint oriented process development should start earlier for biosimilars than for originator products. To buy down risk, at-scale manufacturing may be desirable as early as nonclinical / phase I manufacturing.

Speaker
Biography:

Louis Boon received his PhD in Biochemistry at the University of Amsterdam. In 2003 he was one of the founders of Bioceros BV were he currently hold a position of CSO. In addition, he held positions as CSO for 4AZA Bioscience NV and for FF Pharma: he was VP Preclinical for PanGenetics BV and Tanox. He is an author of over 280 papers in international scientific journals in the field of medical biotechnology. Bioceros generates production cell lines and production processes for the industry and uses its proprietary CHOBC® platform and its SPOT™ technology to generate a portfolio of cell lines producing biosimilar monoclonal antibodies. He developed a complete process modulation toolbox to fit biosimilar CQAs. Currently, he is also involved in the development of innovative new molecular antibody entities, which now within the Polpharma Biologics group can be progressed to GMP production and clinical testing.

Abstract:

Innovative cell line generation and early process development are the cornerstones of the success of a biosimilar antibody since the costs of goods (COGs) needs to be very low. To achieve this, a high producing cell line in combination with a modulatory upstream processing (USP) strategy to meet similarity to the originator that do not limit productivity are obligatory. The major strategy generally used in USP to optimize productivity, is an elevation of viable cell density (VCD) in the fermenter. This USP solution however creates difficulties in downstream processing (DSP), since clarification will be difficult and in addition host cell related impurities will be high. Since Bioceros recognizes the biosimilar process as USP and DSP together, we have a different USP strategy for productivity optimization. Ultimately COGs are determined by USP and DSP together. Therefore, we create using our SPOT™ technology to increase Qp values, already during the cell line generation. The high Qp values facilitate high volumetric productivity at low VCD of these cell lines which enables a simple and efficient DSP process. Alongside we observed that irrespective of the VCDs, cell lines with a high productivity had a very high demand for oxygen, agitation, gasflow and nutrients. These high demands result in process issues, like oxidation of the product, shear stress, high sparging rates and high costs. Furthermore, these high demands will limit the ultimate innovations to further increase productivity. To avoid these issues we applied metabolic engineering and developed the SLIM™ technology on our CHOBC® platform. The SLIM™ technology decreases oxygen and feed consumption and also decreases sparging and agitation rates in the bioreactors showing the high efficiency of these process on which the SLIM™ technology was applied. Together, SPOT™ and SLIM™ technology in our CHOBC® platform reduce the cost of goods of biosimilars.

Anupam Chanda

Alembic Pharmaceuticals Ltd, India

Title: Packaging challenge for biosimilar products
Speaker
Biography:

Anupam Chanda has his expertise in creating innovative design in packaging material and device technical book “ Packaging Technology An advance Practical approach “published from germany and amazon is distributing to 77 countries. Book is available in www.amazon for Biologics, biosimilar, anti Cancer and HIV products. He is having two decades ( approx. 25 years) of experience in India and abroad. He is a renowned speaker, widely travel throughout the world. Gain and share his thoughts with scientist from various institution and industry professionals. Author’s.de, www.amazon.com and www.morebooks.de. He is attached with various charitable institutions and research institutes.

Abstract:

Presently packaging plays a significant role for biosimilar product. The process of selecting materials and the type of packaging offers an opportunity for the packaging scientist to look for new biological delivery choices. Most injectable
protein products were supplied in some sort of glass vial, prefilled syringe and cartridge. Those products having high pH content there is a chance of “delamination” from inner surface of glass vial. With protein-based drugs, the biggest issue is the effect of packaging derivatives on the protein’s three-dimensional and surface structure. These are any effects that relate to denaturation or aggregation of the protein due to oxidation or interactions from contaminants or impurities in the preparation. The potential for these effects needs to be carefully considered in choosing the container and the container closure system to avoid putting patients in jeopardy.
Cause of Delamination:
• Formulations with a high pH include phosphate and citrate buffers increase the risk of glass delamination.
• High alkali content in glass could accelerate erosion.
• High temperature during the vial-forming process increases the risk of glass delamination.
• Terminal sterilization (irradiated at 20-40 kGy for 150 min) also is a risk factor for specific products (veterinary parenteral
administration), could cause delamination.
• High product-storage temperatures and long exposure times can increase the rate and severity of glass delamination.
• How to prevent Delamination
• Treating the surface of the glass vials with materials, such as ammonium sulfate or siliconization can reduce the rate of
glass erosion.
• Consider alternative sterilization methods only in rare cases.
• The correct specification for the glass to ensure its suitability for the pH of the product.
• Use COC/COP vial
Present market demand is syringe barrel and/or plunger coated with silicone. Non-uniform silicone coatings can affect protein stability. The proteins can absorb into the walls of the container. While silicone can reduce absorbtion, excess silicone can form suspended oil-like droplets. Proteins can form around those droplets and change their natural state. New lubricant coatings, such as inert fluoropolymers, are being introduced to replace silicone. As a very inert material, fluoropolymer provides smoothness for the syringe plunger without the irregularities or the issues that have come with silicone. Other new coating
materials are being introduced with new types of packaging related to self-injectors, especially injector pens, patches and transdermal and wearable devices for self-infusion. Extractable and leachable are most important for inhalers and catheters.

Sung Nyeo Shim

Chong Kun Dang Pharmaceuticals, South Korea

Title: Biosimilars analytical strategies
Speaker
Biography:

Sung Nyeo Shim has approximately 18 years of experience in the field of QA, QC, Compliance Of Biologics; QMS; Gmp Training & Biologics Analytical Test Methods Tech Transfer; Filing Support with Regulatory Affairs. She is enthusiastic, energetic, organized, the multifunctional individual with integrity, highly self-motivated. She has delivered milestones in a timely manner. She has QC lab qualification, analytical method development, qualification and validation per International Council for Harmonization (ICH) guidelines. She has also Monitored biologics manufacturing, data integrity/analysis/troubleshooting.

Abstract:

Biosimilar products (follow-on versions of pioneering biopharmaceutical products which are already licensed) are complex structurally and functionally. Regulatory authorities require biosimilars to have appropriate and comparable safety, quality and efficacy with a corresponding reference biologic product. In order to investigate the critical quality attributes of a biosimilar and a reference product for interchangeability and comparability, various analytical approaches are employed, such as LC/MS, Peptide mapping, Thermal analysis, SDS-PAGE, Isoelectric focusing and Capillary isoelectric focusing, Particulate matter analysis, In-vivo and In-vitro bioassay, etc. An analytical similarity of a biosimilar to a reference biological product is assessed with the state-of-the-art of analytical strategies. One of the key analytical techniques is Mass Spectrometry (MS) for assessment of similarity, detection and identification of primary sequence differences and evaluation of batch variability. Analytical differences observed need to be characterized and understood for a cross-confirmation of the data using orthogonal methods to prove the differences are not clinically relevant. Post-translational modifications, three-dimensional structures and protein aggregation have been identified as three important characteristics in developing strategies for the biosimilar. For quality control of the biosimilar production process and product, in-process samples, drug substance and drug product are evaluated in terms of protein content, purity, potency, identity and impurity. All analytical data includes the stability study data impact on clinical trials, the product expiration date and the product approval.

Speaker
Biography:

Anita Burrell is Vice President, Global Market Access at Health Strategies Group where she is responsible for leading and growing the global market access practice including syndicated and custom market research services. Prior to joining Health Strategies Group, she spent over 18 years with legacy Sanofi companies (23 years in the pharmaceutical industry) where she led worldwide strategy for launch pricing and reimbursement including evidence generation strategies. She was also the product champion for Aubagio (oral MS therapy) and coordinated commercial effectiveness for diabetes franchise. Her work is regularly published and presented at conferences. She is frequently invited to speak on real-world evidence, managed entry agreements, value proposition development and value frameworks.

Abstract:

Biosimilars were touted as a payer’s tool to gain savings in specialty markets, but a recent survey by Avalere of the top 25 payers in the U.S. (about 189 million covered lives) using publicly available coverage policies found that biosimilars are commonly subject to step through policies, including those that require the patient to “fail” first on a branded product and only then will the payer cover a biosimilar of that same branded product (i.e., the biosimilar’s reference product). Policies like this may be why although the Congressional Budget Office (CBO) originally estimated a 10-year decrease in federal spending of $5.9 billion attributable to “follow-on biologics”/biosimilars in 2009, it is estimated the actual savings have only been 8 percent of that amount (approximately $241 million). Some of the biggest challenges to biosimilar uptake are patent and intellectual property (IP) issues since we have 12 approved agents but only three which have been launched commercially. While the USA is currently the largest single market in the world overall by dollar value, the healthcare system comprises of thousands of payers and a lot of the incentives on how their choices are made are less than transparent. Contracting incentives from originator companies may play a part in the lackluster performance of those biosimilars which are available on the market but the bigger issue for a multisource market is that the promise of increased access for patients is not currently being realized. This presentation will review the barriers and possible future opportunities for biosimilar uptake in the USA.

Karlheinz Landauer

Quality Biotech Development & Cells GmbH, Switzerland

Title: How to start a biosimilar development: A risk based approach
Speaker
Biography:

Karlheinz Landauer holds a PhD in Applied Microbiology, focus on animal cell culture technologies, with 20 years of work experience. He has specialized in cell line engineering, upstream process development, scale-up and tech transfer strategies, GMP operations and was a program manager of an NBE (New biological entity). He had several positions in the above-mentioned fields with increasing responsibilities. His career started at igeneon GmbH in Vienna, later he moved to Celltrion R&D Center when in 2011 he eventually was Head Manufacturing and COO at Celonic AG in Basel. In 2016, he has founded QBDC, with its core focus on consultancy for biotech companies in CMC matters, process development, tech transfer & scale-up, facility layout & design, project management and QM & QA. At QBDC, he is driving the company towards high-quality standards and problem-solving communication strategies. For QBDC’s customers, he is an easily accessible, goal-oriented technical solution provider.

Abstract:

With the big success of biologicals and the experience in generics, the biosimilar development was born. In Europe, the first biosimilars were already approved in the 2006 and since then many more followed. Regulators, especially in Europe,developed new guidelines promising fast access and fewer investment costs for biosimilar development programs. With the approval of Celltrion’s Remsima the first monoclonal antibody reached the market and today many companies around the world are increasing the efforts to develop and market their own biosimilars. Often the companies and here especially small molecule pharma companies, do not intend to market their products worldwide but are addressing their own regional markets where they are already well established. Entering the biosimilar development and finally, markets is a risky venture. There are many candidate originators which could be copied and thus need to be analyzed form business opportunity perspective but also from a manufacturing/technology perspective. As already mentioned, the companies often quite well understand their markets, thus business analysis is performed in detail and thoroughly. However, the manufacturability of biosimilars, like the type of originator cell line, mode of fermentation, the stability of formulation, etc. are not analyzed and understood in detail and not appropriately taken into account. After the decision for a specific biosimilar, newcomers underestimate the needs for thorough originator characterization and process development needs and the effort for process characterizations prior entering phase I clinical trial. Not only one company failed based on amino acid aberrations of their product, could not copy post-translational modification or even fail in needed formulation stability and finally, but the program was also stopped. Most often companies, who are intending to enter the biosimilar world, do not have R&D nor production capacities, thus they need to start with contract development and manufacturing organization for their development programs and at the same time invest resources in the planning and construction of production facilities. Another trend is to in-license already established and approved products and starts with fill/finish works in-house and working itself backward till finally own R&D and APIproduction capabilities are established, based on the revenues from the in-licensed products. We are presenting an overall investment analysis tool for biosimilars, a quick method to see how much resources need to be invested when and effects on the program if not done in a timely manner. After the main road was defined as a second tool, to select the best candidate molecule will be shown in detail based on 30 analyzed molecules and finally, our standard CDMO selection guide will be briefly discussed. Having done these first three steps, biosimilar development is still not an automatic success, but it will help to overcome many obstacles by being aware that biosimilar development programs are time-consuming, resource intensive and for sure no generics.