IBI Volume 3 Issue 3

Page 1

Volume 3 Issue 3

Peer Reviewed

Transforming the Healthcare Landscape With Synthetic Biology and Living Medicines

Re-Modelling Drug Delivery Devices For High-Viscosity Formulations

Hydrogel Encapsulation

The New Paradigm for Ambient Temperature Cell Shipping

Key Challenges and Potential Solutions

For Optimizing Downstream Bioprocessing Production

Sponsor Company: www.biopharmaceuticalmedia.com


QUALITY

LIFE SCIENCES LIFE INSPIRED, QUALITY DRIVEN

RELIABILITY

SGS Life Sciences' integrated network of laboratories offers extensive experience in novel vaccines, providing biosafety, biologics characterization and bioanalytical testing solutions to support the fight against COVID-19.

© SGS Group Management SA – 2020 – All rights reserved – SGS is a registered trademark of SGS Group Management SA

Our biosafety testing laboratory provides testing support for the biosafety and characterization of raw materials, cell bank and virus seeds for vaccines, cell and gene therapies, monoclonal antibodies and other recombinant protein based biological medicines, including a vaccine testing solution for coronavirus.

EXPERTISE

We provide established expertise and resources for all aspects of biopharmaceuticals characterization – from physico-chemical properties, to primary, secondary, and tertiary structures, as well as aggregation. Our characterization services include antibody testing and batch release testing for large number of doses. Our GLP/GCP compliant laboratories offer the expertise to both develop assays from scratch (including LC-MS/MS, immunoassays and cell-based assays). We support large-scale routine sample analyses, from regulatory pre-clinical (toxicology) to early and late clinical studies (Phase I to IV).

CONTACT US lss.info@sgs.com

II INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

www.sgs.com/lifescience

www.sgs.com/LinkedIn-Life

Winter 2020 Volume 3 Issue 3


Contents 04 Foreword MARKET REPORT (REGULATORY & COMPLIANCE) 06 How Has the COVID-19 Pandemic Amplified the Importance of the IVD Industry? DIRECTORS: Martin Wright Mark A. Barker BUSINESS DEVELOPMENT: David Knight david@pharmapubs.com EDITORIAL: Beatriz Romao beatriz@pharmapubs.com DESIGN DIRECTOR: Jana Sukenikova www.fanahshapeless.com FINANCE DEPARTMENT: Martin Wright martin@ipimedia.com RESEARCH & CIRCULATION: Virginia Toteva virginia@pharmapubs.com COVER IMAGE: iStockphoto © PUBLISHED BY: Pharma Publications J101 Tower Bridge Business Complex London, SE16 4DG Tel: +44 (0)20 7237 2036 Fax: +44 (0)01 480 247 5316 Email: info@ibijournal.com www.biopharmaceuticalmedia.com All rights reserved. No part of this publication may be reproduced, duplicated, stored in any retrieval system or transmitted in any form by any means without prior written permission of the Publishers. The next issue of IBI will be published in Spring 2021. ISSN No.International Biopharmaceutical Industry ISSN 1755-4578. The opinions and views expressed by the authors in this magazine are not necessarily those of the Editor or the Publisher. Please note that although care is taken in preparation of this publication, the Editor and the Publisher are not responsible for opinions, views and inaccuracies in the articles. Great care is taken with regards to artwork supplied, the Publisher cannot be held responsible for any loss or damage incurred. This publication is protected by copyright. 2020 PHARMA PUBLICATIONS / Volume 3 Issue 3 – Winter 2020

www.biopharmaceuticalmedia.com

As the world is still coming to grips with the COVID-19 pandemic, the role of IVD in detecting, and thus helping in finding an appropriate immune response to, the novel coronavirus, has been praiseworthy. Anant Singh at Grand View Research explains how In vitro diagnostics have already witnessed widespread appeal in the medical and research fields, even before the advent of the coronavirus pandemic, and how they can affect the industry in the future. 10 The Benefits of Outsourcing for Early-stage Drug Development The investment drug companies are making into producing biosimilars has never been greater. This major push is driven by the faster time to market of a biosimilar product compared to a novel drug, achieved through the greater emphasis on analytical investigations into the product and a decreased clinical trial requirement. Richard L. Easton and Andrew J. Reason at BioPharmaSpec Ltd evaluate the benefits of outsourcing for early-stage drug development. 14 New Wave of R&D Points the Way to Tackling the Oldest Enemy The COVID-19 pandemic has forced businesses of every kind to adjust and pivot, demolishing all manner of received wisdoms along the way. The changed environment has also impacted the investment ecosystem in biotech and the perception of infectious disease. Dr. Peter Jackson at Infex Therapeutics explains the best way to tackle this issue. RESEARCH / INNOVATION / DEVELOPMENT 18 Base Editing, The Story So Far Gene editing technologies have been evolving since CRISPR was first commercialised in early 2012. The recent emergence of CRISPR base editing platforms have attracted attention in the cell and gene therapy fields. This is a gene editing technology that could drive the evolution of new therapeutic applications. Kevin Hemphill at Horizon Discovery touches upon the potential therapeutic applications of base editing and explains why there is so much hype around this latest incarnation of CRISPR-based gene editing. 22 Re-modelling Drug Delivery Devices for High-viscosity Formulations Biologic formulations for subcutaneous delivery allow patients to self-administer their treatment and reduce the need for them to consult hospital-based healthcare practitioners. Intravenous formulations are therefore being reformulated for subcutaneous injection (where possible) to capitalise on these benefits. George

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 1


Contents I’ons at Owen Mumford Pharmaceutical Services outlines the new challenges for the medical device industry and why drug delivery device design must be reconsidered to cater to these new biologics and their requirements. 24 A Global Approach to Nitrosamine Impurity Testing for Drug Products Manufacturers and regulatory authorities around the world are now aware of the potential threat posed by nitrosamine impurities in API/DP. By taking a global, collaborative approach to the problem, they have created a regulatory landscape which allows stakeholders to develop proactive risk assessment strategies that cover a wide range of markets. Romain Simon at SGS addresses the potential risk for contamination of biologic drugs with nitrosamines.

them into a final product. In pharmaceutical production, the constituent parts include the active pharmaceutical ingredient (API), excipients, and packaging and labelling materials. Tim Daniels at Autoscribe Informatics demonstrates how the pharmaceutical manufacturing process is essential to success. 44 Key Challenges and Potential Solutions for Optimising Downstream Bioprocessing Production The dramatic growth in the use of biologics across multiple therapeutic applications and categories will only continue to increase. As the demand for these drugs accelerates, there are growing concerns about their cost and availability. Nandu Deorkar and Claudia Berron at Avantor highlight the key challenges and potential solutions for optimising downstream bioprocessing production.

28 Transforming the Healthcare Landscape with Synthetic Biology and Living Medicines

48 Building a Gene Therapy: Challenges and Changes in Viral Sector Manufacture

There is a well-recognised global need for therapeutic options for diseases that are deemed difficult to treat or are currently untreatable. The emergence of synthetic biology tools and technologies has led to a growing awareness of the potential for engineered living organisms to tackle these challenging areas. Raquel Sanches-Kuiper at Evonetix evaluates the transformation of the healthcare landscape with synthetic biology and living medicines.

Retroviruses were an attractive first choice for viral vectors for gene therapy because the DNA they encode – either wildtype viral DNA or the therapeutic transgene – becomes integrated into the host cell genome. Dr. Sophie Lutter at OXGENE outlines the challenges and changes in viral sector manufacturing when building a gene therapy.

32 Helping Labs Operate at Peak Efficiency to Foster Innovation

52 Compliance Capacity: A Checklist to Assess Regulatory Readiness

Science, as Vannevar Bush wrote, is an endless frontier. Bush, the founder of Raytheon and Director of the Office of Science and Development at its inception in 1941, pushed the US government to invest heavily in research – leading to a renaissance of advancement in science and technology. Unfortunately, as more and more businesses have realised, the resources to explore that frontier are not endless. Experts at Avantor analyse how to help labs operate at peak efficiency to foster innovation.

As life science businesses focus on managing the impact of COVID-19 and working on solutions to fight this pandemic, regulatory responsibilities remain a pressing concern that must not fall in priority. In the immediate future, planning for compliance deadlines is a matter of urgency, whether to keep products on the market or meet emerging demands rapidly. Steve Cottrell, President, Maetrics focuses on the outsourcing solutions that will make more efficient use of limited internal resources.

PRE-CLINICAL & CLINICAL RESEARCH

SUPPLY CHAIN MANAGEMENT

36 The Solution to Improve Profitability in Pharmaceutical Development; How to Increase Pre- clinical Productivity and Success Rates in Clinical Trials

56 Hydrogel Encapsulation – The New Paradigm for Ambient Temperature Cell Shipping

The limited availability and usage of pre-clinical laboratory instruments which provide biologically relevant data has resulted in unnecessarily high failure rates of pharmaceutical drug candidates in clinical trials, the single most expensive component of the pharmaceutical development process. Teodor Aastrup et al. at Attana AB explain that the clinical results from Attana’s validated drug candidates have shown impressive progress in a short time and increased productivity in clinical trials. MANUFACTURING/TECHNOLOGY PLATFORMS 40 Tightly Coupling the Laboratory to the Pharmaceutical Manufacturing Process is Essential to Success All manufacturing flows – whether making cars, food products, medical devices, or pharmaceuticals – take raw materials, and through a series of manufacturing processes, transform 2 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

REGULATORY/QUALITY COMPLIANCE

Deep freezing has become the standard practice for long-term preservation of biological samples, especially for isolated and cultured cells. Its use has been extended to the shipping of cells from point of manufacture to the place of use. Ana Ribeiro at Atelerix outlines the different cryopreservation techniques available and evaluates the new paradigm for ambient temperature cell shipping. 60 Driving Demand for Parenteral Packaging: How Biologics are Changing the Face of Pharma Packaging? The biopharmaceutical sector has enjoyed robust and sustained growth in recent years. A recent BioPlan Associates report estimates worldwide sales of biopharma treatments reached more than $300 billion this year (2020), increasing at an annual rate of 12%. Marcelo Cruz at Tjoapack discusses more about the evolving biopharma landscape and the rising demand for packaging solutions for parenteral products to explore the future for the sector. Winter 2020 Volume 3 Issue 3


DEVELOPING PERSONALISED CANCER TESTS FOR DOCTORS AND PATIENTS

RGCC is a specialist medical genetics company, established in 2004 with headquarters in Switzerland. We are experts in developing and providing personalised cancer genetics tests for doctors and patients, and testing tumours for sensitivity and resistance to chemotherapy treatment and a range of natural substances. We are actively involved in pharmaceutical research and development. Our facilities are equipped with the most technologically advanced equipment and specialised software for data analysis. RGCC is a global organisation, and we work in collaboration with branch offices and distributors to provide a worldwide service. RGCC HEADQUARTERS RGCC International GmbH Baarerstrasse 95, Zug, 63OO, Switzerland email: office@rgcc-international.com, tel: +41 (O) 41 725 O5 6O LABORATORY FACILITIES RGCC SA Florina, Greece, email: office@rgcc-genlab.com, tel: +3O 2385 O 4195O RGCC India Gajularamarm, Hyderabad, India RGCC Central Europe Biozentrum, Martin-Luther-Universität Halle-Wittenberg, Weinbergweg 22 O612O Halle (Saale), Germany

rgcc-group.com


Foreword As the coronavirus pandemic continues to spread across the world and the likelihood of a second wave increases, the biotech sector has continued to see significant investment. The entire world faced personal and professional challenges and that was reflected in the way that the biopharmaceutical industry worked in the past few months. From gene therapies to intelligent drug discovery and development, the biopharma and medtech industry is working at lightning speed to commercialise gene therapies and aid the development and testing of new drugs. In this journal, you will find some articles that will evaluate the benefits of outsourcing for an early stage of drug development as well as the new challenges for the medical device industry. George I’ons at Owen Mumford Pharmaceutical Services outlines the new challenges for the medical device industry and why drug delivery device design must be reconsidered to cater to these new biologics and their requirements. Biologic formulations for subcutaneous delivery allow patients to self-administer their treatment and reduces the need for them to consult hospital-based healthcare practitioners. Improving the lives of patients requires a deep understanding of their medical conditions and needs and priorities. So how can the biopharmaceutical industry improve patient incomes? According to Raquel Sanches-Kuiper at Evonetix, the discovery of new living medicines has already resulted in major improvements in healthcare and patient outcomes, by harnessing the power of synthetic biology. By using ‘living medicines’ we are able to produce desirable therapeutic effects when treating patients. Alongside patient outcomes in drug development, another hot topic is the benefits of outsourcing for early stage drug development. Investment drug companies that are producing biosimilars have never been greater. Richard L. Easton and Andrew J. Reason at BioPharmaSpec Ltd explore how this major push is driven by the faster time-to-market of a biosimilar product compared to a

novel drug, achieved through the greater emphasis on analytical investigations into the product. Drug development is a costly and timely process, and includes the critical work of verification, optimisation and monitoring needed to take the most promising molecules through from clinic to market. In manufacturing, manufacturers are now aware of the potential threat posed by nitrosamine impurities in API/DP. Romain Simon at SGS addresses the potential risk for contamination of biologic drugs with nitrosamines and analyses the proactive risk assessment strategies that need to be taken by manufacturers to cover a range of markets. The biopharmaceutical sector has enjoyed robust and sustained growth in recent years. A recent BioPlan Associates report estimates worldwide sales of biopharma treatments reached more than $300 billion this year (2020), increasing at an annual rate of 12%. Marcelo Cruz at Tjoapack discusses more about the evolving biopharma landscape and the rising demand for packaging solutions for parenteral products to explore the future for the sector. Finally, providers of temperature-controlled packaging, storage, and shipping capabilities for the biopharmaceutical industry are about to face their potentially finest moment. In particular, deep-freezing has become the standard practice for long-term preservation of biological samples, especially for isolated and cultured cells. Ana Ribeiro at Atelerix outlines the different cryopreservation techniques available and evaluates the new paradigm for ambient temperature cell shipping. I would like to thank all our authors and contributors for making this issue an exciting one. We are working relentlessly to bring you the most exciting and relevant topics through our journals. As I write this, the holiday and the festive season is rapidly approaching. I hope everyone will take forthcoming opportunities to recharge their batteries and look forward to whatever may come our way in 2021. Beatriz Romao, Editorial Manager

IBI – Editorial Advisory Board •

Ashok K. Ghone, PhD, VP, Global Services MakroCare, USA

Jim James DeSantihas, Chief Executive Officer, PharmaVigilant

Bakhyt Sarymsakova – Head of Department of International Cooperation, National Research Center of MCH, Astana, Kazakhstan

Lorna. M. Graham, BSc Hons, MSc, Director, Project Management, Worldwide Clinical Trials

Catherine Lund, Vice Chairman, OnQ Consulting

Mark Goldberg, Chief Operating Officer, PAREXEL International Corporation

Cellia K. Habita, President & CEO, Arianne Corporation

Maha Al-Farhan, Chair of the GCC Chapter of the ACRP

Chris Tait, Life Science Account Manager, CHUBB Insurance Company of Europe

Deborah A. Komlos, Senior Medical & Regulatory Writer, Clarivate Analytics

Rick Turner, Senior Scientific Director, Quintiles Cardiac Safety Services & Affiliate Clinical Associate Professor, University of Florida College of Pharmacy

Robert Reekie, Snr. Executive Vice President Operations, Europe, Asia-Pacific at PharmaNet Development Group

Stanley Tam, General Manager, Eurofins MEDINET (Singapore, Shanghai)

Stefan Astrom, Founder and CEO of Astrom Research International HB

Steve Heath, Head of EMEA – Medidata Solutions, Inc

T S Jaishankar, Managing Director, QUEST Life Sciences

Elizabeth Moench, President and CEO of Bioclinica – Patient Recruitment & Retention

Francis Crawley, Executive Director of the Good Clinical Practice Alliance – Europe (GCPA) and a World Health Organization (WHO) Expert in ethics

Hermann Schulz, MD, Founder, PresseKontext

Jeffrey W. Sherman, Chief Medical Officer and Senior Vice President, IDM Pharma.

4 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


2.25

Spring-free

Simplicity for higher volume formulations

Now a reality. A springless, passive safety device for 2.25mL pre-filled syringes, designed for simple assembly and use.

Available now

Scan the QR code to find out more or visit ompharmaservices.com/ unisafe-2-25/

Also available in 1mL

® UniSafe is a registered trademark of Owen Mumford Ltd. © 2020 www.biopharmaceuticalmedia.com

OMPS/ibi/ad/ob/1120/7 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 5


Market Report

PEER REVIEWED

How Has the COVID-19 Pandemic Amplified the Importance of the IVD Industry? In Vitro Diagnostics – The general population may not be very familiar with this term, but it has been in and around the daily conversation in the present world. As the world is still coming to grips with the COVID-19 pandemic, the role of IVD in detecting, and thus helping in finding an appropriate immune response, to the novel coronavirus, has been praiseworthy. As the world, and more particularly the United States and European nations, is dealing with a second wave of the virus, the areas of vaccine development and IVD are firmly in the limelight. Let’s see more about the basics of IVD, and how effective they have proven to healthcare and government agencies in combating the pandemic on a war footing.

An Introduction… Simply put, in vitro diagnostics involves testing carried out on samples obtained from the human body, such as tissues or blood. The term ‘in vitro’ signifies the study of micro-organisms, cells, or biological molecules in laboratory settings such as petri dishes, test tubes, and flasks. The use of in vitro testing generally offers a more detailed analysis of an organism than in the in vivo studies, which involve studies conducted in living organisms. The utilisation of IVD rapid tests helps in offering quick results, which is especially useful during emergency situations; additionally, IVD also allows multiple varied markers to be detected at the same time. Other major advantages include ease of customisation for detecting specific chemicals or markers, convenience offered in recording data, and offering reliable results that can be easily repeated. In vitro diagnostics already witnessed widespread appeal in the medical and research fields, even before the advent of the coronavirus pandemic. As per a report by Grand View Research, in vitro diagnostics have been used in critical areas such as infectious diseases and oncology, with major companies launching products and services. The growing instances of diseases such as tuberculosis and HIV, among others, have led to the development of new IVD systems. Roche Diagnostics, in May 2019, introduced the Cobas MTB-RIF/INH test, which detects antibiotic resistance in tuberculosis DNA. The company had also launched VENTANA pan-TRK (EPR17341) Assay globally in November 2018, which is the first automated IVD immunohistochemistry assay used to detect tropomyosin receptor kinase in cancer-afflicted people. The Impact of COVID-19 However, the coronavirus pandemic has arguably become the biggest headache for healthcare institutes around the globe. As of the time of writing, the total number of cases globally has crossed 55 million, and has resulted in more than 1.3 million deaths. Powerhouse economies such as the United States, India, 6 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Italy, UK, and France, among others, have borne the brunt of the pandemic, which has resulted in high rates of testing becoming critical to control the pandemic in these countries. As per the latest CDC data on November 16, 2020, the number of tests reported across the US was over 166.3 million, with around 12.6 million turning out to be positive. For India, the number of tests conducted as of November 11, 2020 was over 121 million. Not just in these countries, the COVID-19 tests are expected to be necessary in countries that have attained control over the virus. These numbers are expected to continue increasing over the coming year, thus showcasing their significance. To cope with the high number of COVID-19 cases, the US FDA has provided EUA (Emergency Use Authorization) to a number of IVDs. An EUA is issued in the scenario of an extreme public health emergency, which allows the use of unapproved medical products, or the unapproved use/s of an approved medical product, for the diagnosis, treatment or prevention of severe or life-threatening conditions, wherein criteria such as lack of availability of any approved alternative are met. Some of the notable IVDs that the FDA considers for EUA include: •

Diagnostic tests – Include molecular tests and antigen tests; can detect parts of the SARS-CoV-2 virus, can diagnose infection with the virus.

Serology/antibody tests – Constitutes a blood test that looks for indications of any previous COVID-19 infection. It detects antibodies that fight off the infection. This test, though, does not guarantee complete individual immunity, as returning a positive antibody result does not guarantee that the concerned individual will not be affected by the virus again.

Tests for COVID-19 patient management – These tests are authorised to be used in the management of COVID-19 patients. Once the patient is diagnosed with the disease, such additional tests may be carried out for informing patient management decisions.

Molecular Tests and Antigen Tests – Some Key Differences The diagnostic tests are further classified into molecular tests and antigen tests. Molecular tests help in the identification of the genetic material of the virus. The majority of the molecular tests are polymerase chain reaction (PCR) tests, also known as nucleic acid amplification tests (NAAT). The field of molecular diagnostics had been showing promising growth in the pre-pandemic period, as per a report by Grand View Research, and the outbreak has only accelerated its growth. The antigen tests, on the other hand, detect one or more specific proteins from a virus particle. These tests are typically less sensitive than the molecular tests; however, their simpler design means that they can be scaled to test millions of individuals per day. Winter 2020 Volume 3 Issue 3


Market Report

Molecular test

Antigen test

Is also known as RT-PCR test, LAMP test, viral test, diagnostic test, nucleic acid amplification test (NAAT)

Also known as diagnostic test

The sample is collected mostly through nasopharyngeal, nasal or throat swab; a few tests include the utilization of saliva

Most of the tests involve nasal or nasopharyngeal swabs

Results may be obtained on the same day, or may also take up to a week

Most of the results can be obtained rapidly, in the range of 15-30 minutes

This is a highly accurate test and does not require a follow-up or repeat testing

Less accurate, with a tendency to show false positives; the negative results may require confirmation with a molecular test

Shows the diagnoses of active coronavirus infection

Shows diagnoses of active coronavirus infection

These tests cannot show if the concerned person ever had COVID-19, or was infected with the virus that causes COVID-19 in the past

More likely to miss an active COVID-19 infection when compared to molecular tests. Healthcare professionals may recommend a molecular test if the person shows COVID-19 symptoms, but the test shows a negative result

Following is an overall view of the differences in these two methods:

and specificity rates, which may sound confusing to general readers, but have clear and defined boundaries which doctors and health experts are, or should be, well aware of.

The use of rapid antigen testing has been ramped up in countries such as India, on account of its low cost and quick results. The test, though, carries the danger of showing a high number of ‘false negatives’, which has been pointed out by doctors and medical experts. However, this testing has been rapidly identified by manufacturers as a means to not only help the healthcare frontline fight against the pandemic, but also to increase their global footprint. In September 2020, Aytu BioScience announced that it would be distributing Pinnacle’s ‘CovID RAD Rapid Antigen Detection Test’ worldwide. This test delivers results in 15 minutes, by utilising a nasopharyngeal sample, and can be conducted without using any laboratory equipment. The manufacturer Pinnacle has planned to scale the manufacturing capacity of this test in the United States to 25 million tests per month. Sensitivity and Specificity – The Important Concepts An important aspect of COVID-19 testing is the sensitivity www.biopharmaceuticalmedia.com

Sensitivity rate – It is also known as the ‘true positive rate’, and concerns the proportion of people with the disease who will have a positive result. With regard to COVID-19, sensitivity pertains to the ability of the test to identify the population having the antibodies to SARS-CoV-2 in their system. So, for example, a test having 90% sensitivity would return a positive result correctly for 90% of the people that have the disease, but a false negative for the remaining 10% who have the disease and should have tested positive.

Specificity rate – It is the ability of a test to correctly show a negative result for people not suffering from the condition which they are being tested for. A test with high specificity would generate minimum false-positive results. Example: a test showing 90% specificity rate would generate a negative result correctly for 90% of INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 7


Market Report Company

Test

Launched

Features

Abbo�

SARS-CoV-2 IgG an�body blood test

April 2020

Was the company’s third COVID-19 test launch, and had a sensi�vity of 92·7% (95% CI 90·2–94·8), and specificity of 99·9% (99·4–100%)

BioMérieux

VIDAS an�-SARS-CoV-2 serology tests

Announced in May 2020

Includes two tests, the VIDAS® an�-SARSCoV-2 IgM and an�-SARS-CoV-2 IgG. Used for measuring the presence of an�bodies in the popula�on infected with SARSCoV-2. Provide results in less than 30 minutes and claim to have a specificity of 99.4% and 99.9% respec�vely. Received CE marking in May 2020.

DiaSorin

LIAISON® SARS-CoV-2 Ag

October 2020

Makes use of CLIA technology for detec�ng the presence of SARS-CoV-2 Nucleocapsid protein an�gen in nasopharyngeal swabs and nasal dry swabs. Clinical studies showed that within 10 days of the onset of symptoms, it delivered results with 97.1% sensi�vity and 100.0% specificity on nasal swabs and 94.6% sensi�vity and 99.5% specificity on nasopharyngeal swabs. The manufacturing capacity is poised to be around 10 million tests per month.

Roche

Elecsys An�-SARS-CoV-2 serology test

Announced in October 2020

An automated laboratory assay that is expected to aid in SARS-CoV-2 infec�on diagnosis. It is planned to be made available by the end of 2020 for European markets accep�ng the CE mark; addi�onally, the company plans to obtain EUA from the US FDA. Provides results in 18 minutes for a single test and has a throughput of up to 300 tests per hour from a single analyzer, on the basis of the analyzer.

Siemens Healthineers

CLINITEST Rapid COVID-19 An�gen Test

October 2020

Point-of-care test that u�lizes the nasopharyngeal swab method and delivers results in 15 minutes. As per Siemens, the test displayed 96.72 % sensi�vity and 99.22 % specificity based on a clinical study of 317 subjects across 6 sites. As of October, the company planned to submit the test for EUA from the US FDA.

the tested population that does not have the disease, but would also return a false-positive result for the remaining 10% population. Key Developments Major companies such as Abbott, Becton Dickinson and 8 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Company, Bio-Rad Laboratories, and Roche, among others, have developed a wealth of products, including antigen tests, molecular diagnostics tests, and serology tests, and why not? With most of these companies being established multinational organisations, they have had a clear picture of what the country has, or lacks, in terms of testing capabilities. Here, we will see Winter 2020 Volume 3 Issue 3


Market Report

some of the most notable organisations, and what they have brought to the table in the pandemic.

the different assays would be used to tackle very specific issues or questions.

Results from The Lancet Study Researchers recently conducted a comparative assessment of four widely available commercial antibody assays, in order to evaluate the performance of each assay. The four assays were:

In Conclusion With the high number of cases and the outbreak of the second wave of the pandemic, it is expected that there would be several more positive developments to the already high number of tests brought out by healthcare organisations over the past 6–7 months. With the availability of vaccines still expected to be in the early months of 2021, it is imperative that the field of IVD is strengthened further so that more and more testing capacity is achieved. Of course, this requires the presence of tremendous manpower, and a laser-focus approach. Another factor that can hugely benefit the population is bringing down the costs of this testing, especially in the economically-sensitive markets.

• • • •

SARS-CoV-2 IgG assay (Abbott, Chicago, IL, USA) Elecsys Anti-SARS-CoV-2 assay (Roche, Basel, Switzerland) LIAISON SARS-CoV-2 S1/S2 IgG assay (DiaSorin, Saluggia, Italy) SARS-CoV-2 Total assay (Siemens, Munich, Germany)

Along with these assays, the study also involved a novel 384-well ELISA (the Oxford immunoassay). The study results were published in ‘The Lancet Infectious Diseases’. The sensitivity and specificity of these tests were derived from 976 pre-pandemic blood samples obtained from research studies in Oxford, UK, and 536 blood samples from patients with lab-confirmed SARS-CoV-2 infection. The results showed that each assay had high sensitivity (92.7%–99.1%) and specificity (98.7%–99.9%), with the Siemens assay and the Oxford immunoassay meeting the sensitivity and specificity target of at least 98% by utilising the pre-defined thresholds, and achieving this threshold at all the assessed timepoints, that is, at least 14 days, at least 20 days, and at least 30 days after the onset of symptoms. As per the study, the Roche assay met the sensitivity and specificity target on samples that were taken at least 30 days after the onset of symptoms. The study also noted that the differences observed in assay performance would potentially result in thousands of additional incorrect diagnoses between the worst and best assays, when considered for large population sets. This means that a 10% seroprevalence (number of persons who test positive for a disease on the basis of serology specimens) would translate to the Siemens assay generating 2800 total errors per million tests, while for the same number of tests, the DiaSorin assay would generate an estimate of 16,700 total errors. Additionally, the researchers stated that www.biopharmaceuticalmedia.com

REFERENCES 1.

2. 3.

https://www.grandviewresearch.com/industry-analysis/ in-vitro-diagnostics-ivd-market?utm_source=biopharmaceuticalmedia&utm_medium=referral&utm_campaign=rashmi-s_hc_ guest-post_19-nov-20_anant-s&utm_term=in-vitro-diagnosticsivd&utm_content=home_page https://covid.cdc.gov/covid-data-tracker/#cases_ casesper100klast7days https://www.infectiousdiseaseadvisor.com/home/topics/covid19/ comparing-performance-characteristics-of-five-immunoassaysfor-sars-cov-2/

Anant Singh Anant Singh is a content writer who takes a keen interest in the healthcare and consumer goods sectors, as well as all things technology. He currently works at Grand View Research Inc., a U.S. based market research and consulting company, headquartered in San Francisco, and has written a number of blogs and articles pertaining to various markets for the organization. Has also published reviews for TV shows on HBOWatch, where he is a guest writer.

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 9


Market Report

PEER REVIEWED

The Benefits of Outsourcing for Early-stage Drug Development Introduction The investment drug companies are making into producing biosimilars has never been greater. This major push is driven by the faster time to market of a biosimilar product compared to a novel drug, achieved through the greater emphasis on analytical investigations into the product and a decreased clinical trial requirement. The resulting decrease in development costs allows for a lower drug cost, resulting in a competitive position for market share. This approach to drug development will give benefit to patients and practitioners alike due to the greater choice of product at a decreased cost. As mentioned above, the biosimilars drug pathway places a very strong emphasis on the analytical component in order to demonstrate that the developed drug is “similar” to the innovator drug as defined by the regulatory authorities. Bearing this in mind, how is this similarity best achieved? Structural characterisation of biosimilars is detailed in regulatory agency guidance documents1,2,3, each of which clearly outlines the expectations for analytical investigations and how this work should be approached, as well as discussing the number of batches of biosimilar and innovator that should be considered for side-by-side comparative analysis of structural features. In terms of the focus for the analyses themselves, the EMA and FDA guidelines cite the document that has been the mainstay of structural characterisation of biological drug products for many years: The International Council for Harmonisation of Technical Requirements for Pharmaceutical Use (ICH) Q6B4. This document describes in detail the areas of structural analysis that are expected to be investigated for biopharmaceutical products, covering all aspects of primary, secondary and tertiary structure. For reference, these areas are listed below: Structural Characterisation • • • • • •

Amino acid sequence Amino acid composition Terminal amino acid sequence Peptide map Sulfydryl group(s) and disulfide bridges Carbohydrate structure

Physicochemical Analysis • • • • • •

Molecular weight or size Isoform pattern Extinction coefficient (or molar absorptivity) Electrophoretic patterns Liquid chromatographic patterns Spectroscopic profiles

10 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

The requirement to cover so many areas of structural assessment can prove challenging for analytical labs within the product development company, particularly as many of the techniques will require high-end, expensive analytical equipment that would see relatively little use outside of that specific need. It may not be economically feasible for drug manufacturers to set up internal labs for the full range of required structural characterisation techniques. Another point of consideration is the evolving nature of analytical techniques and instrumentation. Indeed, the biosimilar guidelines specify “state-of-the-art” instrumentation and techniques should be used for structural investigations1,3. Furthermore, the ICH Q6B documentation clearly requires structural features under investigation to be examined “to the extent possible”4, in order to take advantage of the development of new procedures and methods to give greater depth of understanding for the product. This approach allows the documentation to remain current as techniques evolve to give greater depth of analysis (e.g. resolution and sensitivity) across all areas that need to be considered for investigations of a biological product. The requirement not only covers the product itself but also residuals and impurities within the product (as requested in ICH Q6B). Host cell proteins (HCPs) are an important part of the residual/impurity analysis requirement. Analysis of HCPs has traditionally been performed using enzyme-linked immunosorbent assays (ELISA) but there is now a movement to employ mass spectrometric-based investigations, alongside the ELISA-based procedures, as an orthogonal technique to identify and extend the known HCPs in the product5. Analytical contract research organisations (CROs) are ideally placed to provide these latest methods of analysis and are therefore a valuable resource. Therefore, the best option for manufacturers is to consider outsourcing either all or a subset of their analytical needs to specialised analytical CROs. The use of a CRO for this type of work carries many advantages for a manufacturer and these will be considered below. It should also be stated that, whilst this article is focusing on the consideration of analytical support, these factors apply to all types of CROs including those involved in biological analyses (e.g. potency, cell-based and/or binding assays). Efficient Response to Evolving Need Structural analyses, as already stated, are the foundation upon which any claim of biosimilarity will stand or fall. For this reason, analysis of one form or another will take place right from the earliest stages of biosimilar product development and will grow and evolve with the evolution of the product. This will include, but not be limited to, cell clone selection, assessment of bioreactor conditions and scale-up investigations, and of course the drug product itself, as well as studies on any aspects of the drug product structure or observed properties of the drug Winter 2020 Volume 3 Issue 3


Market Report product that may be causing concern (e.g. increased aggregation). Outside of the analysis of the actual drug product itself, stability investigations and formulation studies also need to be considered. Changes of manufacturing practice or manufacturing site will also need to be investigated to assess any impact the changes may have had on the product structure itself. CROs provide a means of rapidly responding to these analytical requirements as and when needed, thus offering an efficient mechanism to generate data in a timely manner, allowing data-driven product decisions to be made efficiently and with confidence. Development of Analytical Requirements An extension of the point above is the consideration of analyses at the appropriate time in the development of a product. As an example, let us consider glycosylation. This is one of the most major, abundant and important Post Translational Modifications (PTMs) (Figure 1) on biological products and is directly and indirectly responsible for a wide range of biological functions6. Therefore, and as expected, it is a requirement of ICH Q6B to analyse the glycan structures of a glycosylated biopharmaceutical (e.g. monoclonal antibodies, Etanercept, Erythropoietin amongst many others). Glycosylation is complex and heterogeneous due to the nature of its biosynthesis and a variety of techniques need to be used to carry out the required in-depth investigations into their structure. However, there is no point in performing all of these detailed investigations at the cell clone selection stage since only one clone will be selected for development. At this stage it is often sufficient to investigate the population composition profile of any N-and O-glycans on the expressed product to find the optimal clone, with reference to any critical quality attributes that have been identified (e.g. a required level of sialylation) based on similarity to the innovator. Once the clone is selected, the more detailed structural analyses can be performed to investigate the nature of the linkages between the monosaccharides and any site specificity of glycosylation that may be present. It goes without saying that the precise glycan structures must be “similar” to the innovator product and if significant differences are found, then a new clone may need to be developed or the manufacturing process may need to be investigated. Since glycosylation is such a key area of analysis, with glycans carrying both the potential to be functionally significant and, if non-human epitopes are present, the potential to be immunogenic, having CROs on hand that have a high degree of understanding and ability to investigate and interpret glycan data is highly beneficial. The ability of different cell lines to produce what will be cell-specific and possibly relatively uncommon glycosylation means specialist knowledge in this area is invaluable when it comes to full structural glycan investigation. As an example, see BioPharmaSpec’s recent collaborative work on the glycan structure of recombinant SARS-CoV-2 spike protein RBD in HEK293 cells7. Orthogonality Orthogonality is a key term in investigative structural analyses www.biopharmaceuticalmedia.com

for all biopharmaceuticals and is vital for biosimilar analyses. The biosimilar guidelines from both the FDA and EMA emphasise orthogonality in the way analytical techniques are brought together to provide supportive data for various structural features, such as certain aspects of monoclonal antibody structure (e.g. C-terminal Lys), other PTMs such as deamidation, aggregation and secondary and tertiary structure1,3,8. As an example, the FDA biosimilars guideline document states “Methods that use different physicochemical or biological principles to assess the same attribute are especially valuable because they provide independent data to support the quality of that attribute (e.g. orthogonal methods to assess aggregation)”3 and the EMA biosimilars quality document states, with regard to the structural comparability exercise, “This should include comprehensive analyses of the proposed biosimilar and reference medicinal product using sensitive and orthogonal methods…”1. Examples of orthogonal techniques for selected analytical procedures are given in Table 1. Technique

Aspect

Orthogonal technique(s)

Orthogonal parameter

Peptide mapping mass spectrometry

Mass and fragment ion based identification of heavy chain C-terminal peptide with/without C-terminal Lysine

Imaging capillary isoelectric focussing

Charge based separation of monoclonal antibodies with/ without heavy chain(s) containing C-terminal Lysine

Peptide mapping mass spectrometry

Mass and fragment ion based identification of deamidated peptides

Imaging capillary isoelectric focussing

Charge based separation of monoclonal antibodies exhibiting different degrees of deamidation

Circular Dichroism

Spectroscopic assessment of higher order structure (secondary/tertiary structure)

Fourier Transform InfraRed spectroscopy, NMR

Alternative mechanisms for assessing higher order structure through different forms of spectroscopy

Size exclusion chromatography with Multi Angle Laserlight Scattering

Assessment of aggregation through chromatographic separation with light scattering assessment of aggregate size

Sedimentation Velocity Analytical Ultracentrifugation

Separation of aggregates through interaction with a centrifugal field and UV and interference optics measurement (also provides a column free assessment)

ELISA

Assessment of Host Cell Proteins through gel based separation and immunochemical detection

Mass Spectrometry

Assessment of protein fragments with database interrogation for protein identification. Quantitation of HCPs through proteomic type analyses

Table 1: Examples of selected analytical techniques and orthogonal procedures for the analysis of various structural parameters.

As mentioned above, orthogonality extends to the analysis of residuals and impurities. The increased use of mass spectrometry-based techniques for HCP analysis (covered above and in Table 1) is an example of this. CROs are ideally placed to generate orthogonal data since they will have both the equipment and the knowledge to produce a package of self-supporting data for the various aspects of molecular structure requiring investigation. Knowledge The most important benefit in choosing to outsource characterisation activities is that the product development company now has access to the analytical knowledge available at the CRO and can use this to effectively fulfil the above-described regulatory requirements. Access to a broad array of high-end analytical equipment and producing quality data is of course important, but partnering with an analytical CRO brings many other advantages. The CRO will likely have INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 11


Market Report

Asn

O Disulfide Bridges

and sound workflow where complementary methods are bought together to provide orthogonality as required and where the limitations of some procedures can be overcome by firm data from other techniques.

Deamidation

OH

Methionine Oxidation

Cys

S

Cys

Asn

The biosimilar guidelines from the FDA specifically state “A thorough understanding of each analytical method’s limitations will be critical to a sponsor’s successful identification of residual uncertainties and, in turn, to the design of subsequent testing”3. This is a very important consideration, particularly as current instrumentation is made simpler to use with regard to operation and data collection. However, collection of data is only the start: understanding and interpreting the data is all-important if meaningful and informed conclusions are to be drawn. An example of orthogonal data, showing how different techniques can generate different profiles is shown in Figure 2. Understanding techniques at a fundamental level allows for data of this nature to be assessed and rationalised.

O

S S Met

Ser Lys

O-linked glycosylation

Asn

Glycation

N-linked glycosylation

Experience within a CRO partner provides a huge bank of knowledge on which to draw and this only comes with years of working in this type of environment and having exposure to a wide variety of proteins/glycoproteins, each with their own analytical challenges.

Figure 1: Graphical representation of a selection of post-translational modifications commonly found on proteins and glycoproteins. Detailed methods are required for the assessment and structural characterisation of PTMs. Analytical CROs, with their range of instrumentation and experience in this type of analysis, are ideally placed to perform these types of detailed structural investigations.

considerable knowledge covering the regulatory expectations, the analytical techniques and the biosimilar products in development, and will also have the experience to interpret the data in a proper way and put the findings into context. All of this increases understanding of the product and ultimately accelerates the development of the product through the early phases and towards the clinic. Partnering with an experienced CRO will also allow for the development of a comprehensive

This knowledge is priceless to the drug developer, as CROs will not only generate the necessary data but also identify anything anomalous that may require further investigation. This type of rapid identification of anomalies can in turn lead to targeted structural investigations that feed information back to the manufacturing process, allowing changes/ improvements to be made. Rapid response of this nature

2749

TOF_060418_017

(2) ACQUITY FLR ChA Ex265,Em425 nm Range: 763268

B

700000.063 650000.063 600000.063 550000.063 500000.031

TOF_230218_004 215 (3.787) Cm (204:229)

A

100

450000.031

*

EU x 10e4

2749 R/A/Tryp digest, 10pmol/ul

1216.49 1216.50 1216.47

400000.031

300000.031

200000.016

*

150000.016 100000.008

1216.99

50000.004 0.000

1217.02

%

*

8.00

10.00

12.00

14.00

16.00

18.00

20.00

22.00

24.00

26.00

28.00

30.00

Time

1215.94 1297.50 1297.01

1217.48

1135.46 1134.95

1114.92 1115.43

0 1100

*

250000.016

1215.97

*

1: TOF MS ES+ 2.51e3

350000.031

1120

1298.01

1296.99

= N-Acetylglucosamine = Mannose = Fucose = Galactose

1135.97 1143.47

1217.97

1147.83

1140

1160

1296.96

1298.52

1242.94

1180

1200

1220

1240

1260

1280

1300

1399.03 1399.53

1318.01 1319.05

1320

1340

1360

1380

1400

1420

1440

1460

1480

m/z 1500

Figure 2: Orthogonal data obtained from analysis of monoclonal antibody N-glycans. Panel A represents the analysis of the glycopeptide observed in a peptide mapping analysis, following a proteolytic digest, with the N-glycan structures identified for the observed glycoforms of the peptide. Panel B represents the analysis of fluorescently labelled, released N-glycans. Truncated species are marked with an asterisk. The higher level of truncated species observed in the peptide mapping data is due to glycopeptide fragmentation occurring in the source of the mass spectrometer. 12 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


Market Report

can thus serve to save a manufacturer considerable time and money by identifying product issues at the earliest opportunity. Consideration should also be given to the fact that CROs, being a third party, are perceived as “neutral” and have no vested interest in the product being analysed. An independent assessment of a product is something that is valued highly by the various regulatory authorities. In summary, CROs are a vital resource for any drug development company, including those engaged in production of biosimilars, as they allow a drug developer to meet the analytical expectations of the guidelines along with the more generally applicable analytical guidance for structural investigations. They provide not only ready access to a wide range of state-of-the-art instrumentation whenever it is needed, but can serve as a long-term partner, providing an ever greater depth of structural information and product knowledge to the drug developer. The analytical support from a CRO can serve to assist in the development programme of the drug itself and design of the method of production and purification. Drug developers should therefore consider bringing CROs on-board early in the research and development stage of the product lifecycle in order to maximise their ability to support the product. REFERENCES 1.

2. 3.

4. 5.

6.

Guideline on similar biological medicinal products containing biotechnology-derived proteins as active substance: quality issues (revision 1). European Medicines Agency EMA/CHMP/ BWP/247713/2012 (2014) Guideline on similar biological medicinal products. European Medicines Agency CHMP/437/04 Rev 1 (2014) Development of therapeutic protein biosimilars: Comparative analytical assessment and other quality-related considerations. Guidance for Industry. Draft guidance. FDA (2019) ICH Topic Q6B Specifications: Test procedures and acceptance criteria for biotechnological/biological products. EMA CPMP/ICH/365/96 Roamer, J and Sharov V, Qualitative and quantitative host cell protein analysis using mass spectrometry. BioProcess International 17 (1-2), January – February 2019 Varki, A. Biological roles of glycans. Glycobiology 27, 3-49 (2017).

www.biopharmaceuticalmedia.com

7.

8.

Antonopoulos, A et al. Site-specific characterisation of SARS-CoV-2 spike glycoprotein receptor binding domain Glycobiology cwaa085 doi.org/10.1093/glycob/cwaa085 (2020). Easton, RL. Building orthogonality into biosimilar testing. BioProcess International 18(3), March 2020

Dr. Richard L. Easton Richard obtained his PhD in glycoprotein structural characterisation using mass spectrometry from Imperial College of Science, Technology and Medicine. He subsequently spent several years there as a postdoctoral research scientist working in the field of glycoprotein structural characterisation with emphasis on glycan elucidation. He moved to GlaxoSmithKline for a short time where he was head of mass spectrometry for the toxicoproteomics and safety assessment group. Richard joined M-Scan Limited (now part of SGS Life Sciences) as a biochemist and became the Team Leader for Carbohydrate Analysis before being appointed Principal Scientist. Richard joined BioPharmaSpec in 2016 as Technical Director for Structural Analysis and is responsible for management of all aspects of carbohydrate and glycoprotein characterisation at the primary structure level. Email: r.easton@biopharmaspec.com

Dr. Andrew J. Reason Andrew is the founder, CEO and MD of BioPharmaSpec. He has 25 years of experience in analysis of both novel and biosimilar biopharmaceuticals and has been involved in the commercialisation of a number of analytical methods for characterising proteins. In addition to his scientific and managerial duties, Andrew has contributed to many industry publications and is a regular presenter at conferences. Andrew is also currently a Visiting Professor at the University of Warwick. Email: a.reason@biopharmaspec.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 13


Market Report

New Wave of R&D Points the Way to Tackling the Oldest Enemy The COVID-19 pandemic has forced businesses of every kind to adjust and pivot, demolishing all manner of received wisdoms along the way. The changed environment has also impacted the investment ecosystem in biotech and the perception of infectious disease in particular. COVID-19 has given governments, public health authorities and insurance companies a harsh lesson in preparedness. The vast sums being committed now to mitigate the effects of the pandemic will have implications for years to come. Amid the crisis, it will not have escaped the attention of investors that the case for better bio-security has never been stronger.

COVID-19 is amplifying interest in funding therapeutics for infectious disease – and that includes antimicrobial resistance (AMR), where the business model has long been challenging, but where the failure to act is also having a disastrous impact on global health and economics. Every year, an estimated 700,000 people die as a result of antimicrobial resistance (AMR) after contracting drug-resistant infections that can no longer be treated with existing medicines. It is predicted that if no new antibiotics are found, the death toll from AMR could reach 50 million by 2050, wiping up to $100 trillion of economic growth from the global economy. Yet there is still a problem of perception for investors wary of the failed antibiotics market. What tends to be overlooked is that AMR research has itself fundamentally evolved. Today, it’s not so much about investing in the research and development of new antibiotics, but in new precision medicines that will save patients suffering life-threatening infections. There’s a new generation of technologies, across AMR therapeutics, preventatives and diagnostics. One focus is on stimulating the immune system to tackle the infection as opposed to attempting to directly destroy the bug. At the same time, we are also seeing the development of genetically-engineered phages, exclusively targeting the microbes that are going to kill you. I’m currently part of a team working on an anti-virulence programme, RESP-X, which we are developing through an agreement with the Japanese pharma company Shionogi. The therapy is designed to help tackle Pseudomonas aeruginosa infections, a hard-to-treat pathogen that features in non-cystic fibrosis bronchiectasis and other lung diseases. The RESP-X programme is not about directly killing the bugs but switching off the virulence mechanism, reducing inflammation in the lungs and allowing the body’s own immune system to clear the infection. The risks associated with COVID-19 include the grim reality that it has created a febrile environment for secondary bacterial infections to flourish. Thousands of patients may be affected, 14 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

and as more data is analysed, these could prove to be just as deadly as the virus. The SARS-CoV-2 virus weakens the body, making it easier for bacterial infections to take hold. According to a paper in The Lancet (Strathdee, Davies, & Marcelin, 2020), early research has found thatnearly 15 per cent of all COVID-19 patients acquiresecondary bacterial infections. These bugs can cause or worsen a patient’s pneumonia, potentially leading to septic shock, a life-threatening complication. Furthermore,public health authoritiesandscientistssuggest that of individuals who died from COVID-19, as many as half were afflicted with these secondary infections. Among these opportunistic bacteria are drug-resistant superbugs. In summary, we have exciting technologies in development, and an increasing burden of life-threatening infections, but there is still one vital element missing from the AMR ecosystem: effective market reform. Given the scale of global public health challenge posed by AMR, what the world really needs is leadership at the level of nation states, individually and collectively. The UK has punched above its weight in this respect by introducing a new reimbursement trial for innovative AMR drugs that can treat the most clinically-relevant drug-resistant infections. What we really need to see is a major step forward across the Atlantic, with the United States playing a vital and perhaps decisive role in rebuilding the broken market and facing down the infectious disease challenge. For AMR scientists, investors and patients, one of the best hopes is that politicians within the US Congress consider what needs to be achieved in AMR, and then deliver action through collaboration, transparency and bipartisanship. There is much to laud about what the US has already undertaken, and some very positive steps have been taken to start on the journey to reimbursement reform. A 2013 Centers for Disease Control (CDC) report identified AMR and the “potentially catastrophic consequences of inaction. This led President Obama to direct the National Security Council and the Office of Science and Technology Policy to develop an action plan detailing milestones to tackle the misuse of antibiotics and to propel the development of new antimicrobials and vaccines. However, the subsequent journey hasn’t been straightforward, with the Re-Valuing Antimicrobial Products (REVAMP) Act of 2018 failing to progress. At its core was an attractive offer: a transferable exclusivity voucher. If a company got an antibiotic approved and that new product addressed a priority medical need, the company would receive up to 12 months additional exclusivity on that product. It could have transferred or sold this to another drug as the prize. A step forward came in the form of the 2019 Developing an Innovative Strategy for Antimicrobial Resistant Microorganisms (DISARM) Act. It features a multifaceted strategy for developing new antibiotics and protecting the effectiveness of existing antibiotics. The legislation is designed to help reinvigorate the Winter 2020 Volume 3 Issue 3


Market Report

antibiotic development pipeline for effective new antibiotics, by increasing hospital reimbursement, and incentivise development of robust effective antimicrobial stewardship programmes. These are required to encourage judicious use of currently available antibiotics in order to conserve their effectiveness. The best hope for progress in 2021 is the Pioneering Antimicrobial Subscriptions to End Upsurging Resistance (PASTEUR) Act, which could be hugely impactful. A bipartisan bill introduced by Sen. Michael Bennet (Democrat) and Todd Young (Republican), PASTEUR seeks to create a new method of payment for new antibiotics, delinked from the sales or use of the drugs, ensuring a predictable return on investment for new antibiotics that while critical for patient care, will be used in a limited manner in order to maintain effectiveness. This approach is likened to a Netflix subscription and is similar to a model launched in the UK trial. The new payment system is proposed to incentivise the development of new drugs, with a subscription approach that will see companies paid upfront for access to new antibiotics based on their value to patients and the wider healthcare system, rather than simply on the volume of medicines sold. Passing the PASTEUR Act in 2021 would be a transformational moment in getting the R&D ecosystem moving, but the goal requires getting over some key obstacles. There may, www.biopharmaceuticalmedia.com

understandably, be a sense of ‘crisis fatigue’ as a result of the all-encompassing impact of COVID-19, and the public at large may be unwilling to pay attention to another health priority while we feel the impact of the pandemic. It’s therefore the responsibility of advocates to make the case loudly on behalf of patients, and lobby politicians across Congress to ensure PASTEUR becomes law. We certainly need the thousands of patients dying from AMR infections to be given a higher priority and for our politicians to approve radical action but as COVID shows us, having any issue centre stage still comes with the challenge of making and winning your case. The introduction of measures as dramatic as national lockdowns to control COVID has naturally meant that the models and data used to justify these decisions came under intense scrutiny – as have the scientists offering the advice. Deep divides over strategies to control the coronavirus epidemic have crystallised along political lines, with an accompanying media war. In the US, the opening salvos included Dr Rick Bright, director of the Biomedical Advanced Research and Development Authority (BARDA), who ran the office involved in developing a coronavirus vaccine, being fired by President Trump after they fell out over the latter’s enthusiasm for hydroxychloroquine. Dr. Bright then filed an INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 15


Market Report In this heaving ocean of subjectivity, what lessons are there for AMR, and what hope for any scientist or public health official demanding radical action, such as a ban on the use of certain antibiotics or radical reimbursement reform? As a scientist, I would put my faith in data. We need good data and reliable evidence to make the right choices about AMR, COVID or any other infectious disease. But we know that scientists, like anybody else, can have an agenda, and it’s regrettable that the intensely polarised political discourse around COVID will only have encouraged a general mistrust of official data and its interpretation. There is an obvious danger that by presenting selected data in support of a particular agenda, the public will not only lose trust in what the official position is, but in scientists in general. There is now a clear perception, on both sides of the Atlantic, that data may have been used selectively in order to reflect short term success or to ensure compliance with new measures. I believe that we need much greater transparency around the models and data that form the basis of policy-making. With openness and scrutiny, we can ensure that the public understand, and respect, the need for action. We have to believe that the discipline and rigour of good science and the weight of its evidence will win in the end. We must hope that lessons learned from the pandemic are properly understood and help inform future decisions. What is beyond debate is that AMR and infectious diseases do not respect borders, and that people, animals and other goods are able to transport infections to all geographies. It is only through global collaboration that we can overcome barriers, manage existing treatments adequately and get new drugs to market in order to better manage a further looming health crisis. It may be a little belatedly, but I do believe that big pharma companies have stepped up to the mark with COVID, launching a vast international effort to discover a vaccine and test new drugs Twenty major companies have also stepped forward with a new $1 billion AMR action fund. While we need governments to fix the reimbursement challenges around AMR, a billion dollars will keep new drugs moving through the pipeline whilst the problem is fixed. extensive whistle-blower complaint following his exit from office,alleging his early warnings about the coronavirus were ignored. He told the US media: "I was pressured to let politics and cronyism drive decisions over the opinions of the best scientists we have in government.” Dr. Anthony Fauci, theimmunologistwho heads theNational Institute of Allergy and Infectious Diseases(NIAID), and who led theWhite House Coronavirus Task Forceaddressing COVID-19, is one of the world's leading experts on infectious diseases and has been described as one of America’s most trusted medical figures. Yet Dr. Fauci was hounded by sections of the media following various disagreements with President Trump, and even received death threats that resulted in the need for a security detail. 16 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Infex Therapeutics – formerly the AMR Centre – is currently utilising an expanded focus to include viral as well as bacterial infections with pandemic potential. We are using our drug development capability and expertise to target a wider range of World Health Organisation criticalpriority viral and microbial pathogens, including coronaviruses. Over the past year, we have identified and acquired promising technology, as well as initiating our own in-house R&D programmes. We are also actively looking for other new opportunities in anti-viral therapeutics and AMR drugs over the next year. Professor Christopher Whitty, England’s chief medical officer, has described infectious diseases as our ‘oldest enemy’ – one that has dominated medicine and morbidity. History Winter 2020 Volume 3 Issue 3


Market Report

shows us what can be achieved when our drug discovery and healthcare systems function well globally. Smallpox is estimated to have killed around 300 million during the 20th century and has been controlled by a vaccine. Estimates of how many lives saved by the antibiotic penicillin, created in 1928, go as far as 200 million and counting. We know as scientists that we can have a major impact on infectious disease. Professor Whitty looks to these examples and, like us, is hopeful of new treatments, vaccines tests, pointing out during one press conference to the UK that we have to remember that 100 years ago infectious disease was the major killer on the planet. Thanks to the successes of academia and industry, that’s no longer the case. So we’ve done it before, and with a collective effort on the COVID and AMR challenges, we can – and must – do it again. www.biopharmaceuticalmedia.com

Dr. Peter Jackson Dr. Peter Jackson is an experienced UK-based serial entrepreneur in the life sciences sector. Over the past ten years, he has created six new companies, targeting novel therapeutics across infection, oncology and immunology, as well as in the agrochemicals and life sciences services. During 2015–16, Dr. Jackson was chairman of the steering committee created to establish the UK’s translational R&D centre focused on antimicrobial resistance, the AMR Centre, and now runs Infex Therapeutics as its executive director.

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 17


Research / Innovation / Development

PEER REVIEWED

Base Editing, the Story So Far

Gene editing technologies have been evolving exponentially since CRISPR was first commercialised in early 2012. The recent emergence of CRISPR-based base editing platforms has attracted attention in cell and gene therapy fields as a gene editing technology that could drive the evolution of new therapeutic applications. This article will touch upon the potential therapeutic applications of base editing and explain why there is so much hype around this latest incarnation of CRISPR-based gene editing.

With the rapid emergence of CRISPR-Cas gene editing over the past decade, the field of genome engineering has taken a major step forward in its ability to support both cell and gene therapy. The transition from initial gene editing applications in mammalian cells1,2 to the US Food and Drug Administration’s (FDA) investigational new drug (IND) for CRISPR-engineered cell-based therapies has occurred at an unprecedented rate: the use of CRISPR-based approaches has revolutionised cell-based therapeutics. Expanding on the use of nucleases such as zinc fingers and transcription activator-like effectors, CRISPR-Cas gene editing systems personify the benefit of making efficient and site-specific genomic changes. The advent of base editing has taken this desire to make more precise genomic changes a step further3,4. Like all CRISPR-Cas systems, base editing uses a short guide RNA in partnership with a Cas enzyme. For base editing, a nickase

version of Cas is used to nick a single strand of DNA along with a deaminase enzyme to enable alteration of a single nucleotide (Figure 1). The use of a nickase as opposed to a nuclease substantially reduces the occurrence of DNA double-strand breaks (DSBs). The power of editing the genome with a high degree of specificity while not causing a DNA DSB is pushing base editing into the therapeutic spotlight. Researchers, clinicians and governing bodies are now looking to harness the potential of base editing as an efficient means for introducing stop codons to disrupt gene function5, an area that is gaining a lot of traction in the engineering of chimeric antigen receptor (CAR) T cells to target and kill cancer cells. The Limitations of CRISPR-Cas Gene Editing RNA-guided Cas enzymes are directed to a DNA target site by a guide RNA leading to the introduction of a DSB into the DNA at that precise location. This break is repaired by the cell’s intrinsic DNA repair pathways. Often these repair processes are imprecise, leading to new nucleotide sequences being generated through random insertions and deletions (indels). These indels often produce combinations of missense mutations, nonsense mutations and premature stop codons, which effectively disrupt or knockout the transcription of the gene of interest. However, for clinical applications, the introduction of one or more DSB into the DNA might cause additional safety concerns due to the random nature of indel formation and the possibility of additional DNA breaks occurring elsewhere in the genome with the potential to lead to chromosomal translocations. Additionally, this imprecise nature of indel formation can lead to other undesirable

Figure 1. Overview and potential outcomes of CRISPR-Cas9 mediated genome editing and base editing 18 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


Research / Innovation / Development genomic changes where the downstream impacts are not yet fully characterised. Some of these issues with nucleasebased gene editing have been overcome with carefully selected guide RNAs that do not lead to Cas9-mediated DNA DSBs at sites other than at the gene of interest. However, for some cell therapy applications, multiple genes need to be edited at the same time in the same cell. If standard CRISPR editing platforms are used, then multiple DNA DBSs will be introduced into the genome at the same time, with the potential for chromosomal rearrangements increased. As base editors use a nickase to mediate gene alterations, they could have greater utility in cell therapies where multiple edits are needed to produce the cell-based therapeutic product. An Emerging Technology The initial description of a base editing platform was with cytidine base editors (CBEs)3,4 that enabled recruitment of cytidine deaminase enzymes to a DNA target site via the CRISPR-Cas system, converting C-G base pairs to T-A base pairs, a transition mutation where one pyrimidine base is converted to a different pyrimidine base. These base editors were rapidly followed by adenine base editors (ABEs)6, which convert A-T base pairs to G-C. More recent systems can convert C-G base pairs to G-C7,8 and C-G base pairs to A-T8, demonstrating the capability of base editing to also make transversion mutations, where a pyrimidine base is changed to a purine base (Figure 2). These systems, as well as other novel base editing systems9, have undergone and continue to undergo phases of evolution and refinement with the goal of providing an efficient, precise, and safe method of changing nucleotide base pairs for therapeutic applications. This ability to make a plethora of both transition and transversion mutations presents an opportunity for highly specific editing of the genome, adding to the therapeutic potential that base editors have. Therapeutic Potential Cell-based therapeutics are not a new inception, but their development has been greatly facilitated in recent years by the ability to tailor the genetic make up of a cell using

gene engineering. For example, the engineering of T cells to recognise and destroy cancer cells through the introduction of a CAR and deletion of the T cell receptor, has led to new treatments for B cell leukaemias. These CAR-T cell therapies can be classified as either autologous, where a patient's own T cells are removed, engineered, and then infused back into the patient, or allogeneic where a single source of donor cells can be used to create a steady supply of “universal” CAR-T cells to treat multiple patients. While both methods have advantages and disadvantages, they have both had clinical success. In 2017, the FDA approved the use of autologous CAR-T cells for treating B cell lymphoblastic leukaemia, while in 2019 the first allogeneic CAR-T cell approach was approved for investigational use in patients with multiple myeloma. Despite these successes, CAR-T cells and other CAR-like immune cell-based therapeutics will need to be further modified to treat solid cancers and potentially other human diseases. Some have proposed that CAR-T cells might require upwards of 10 modifications or more to increase their efficacy and longevity, meaning that precision technologies such as base editing could provide a very attractive means to make these multiplex changes. In 2019, clinical trials were initiated using T cells that had been edited with CRISPR-Cas9 to disrupt the function of three genes. Subsequent analysis of the cells nine months later showed healthy-looking T cells with the CRISPR-Cas9 edited cells persisting and showing few off-target changes10. This study offers an important launching point as the field progresses forward with safely editing T cells at multiple sites. It also demonstrates how base editing could offer a compelling route forward. What if T cells and CAR-T cells could be edited precisely without the introduction of a DNA DSB and with much lower levels of indel formation? Base editing has been successfully used for the multiplex knockout of T cell receptor targets11 and it is only a matter of time before some of these base-editing multiplex approaches advance into the clinic. On the Horizon Researchers around the world are working to further characterise and improve base editing technologies as they advance through therapeutic pipelines. Aside from the use of a nickase, base editing can also profit from being similar to but different from standard CRISPR-Cas systems by learning from the methods used to drive the rapid and safe adaption of CRISPR-Cas gene editing in IND-approved therapeutics. Base editing also has a crucial role in treating diseases that arise owing to single point mutations12,13 emphasising the vast number of therapeutic applications and treatments that could be addressed and expediated by base editing. Point mutations associated with Alzheimer’s disease3, sickle-cell anaemia6, β-thalassemia14, and progeria15 are just a few of the thousands of clinically relevant disease variants in the human genome that have been either corrected or modelled by base editing.

Figure 2. Base editing conversion of transition and transversion nucleotide changes www.biopharmaceuticalmedia.com

While several questions about base editing remain unanswered, including the efficient DNA-free delivery of this system into primary cells and the major hurdle of in vivo delivery for gene therapies as opposed to cell therapies, the INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 19


Research / Innovation / Development

impact that base editing will have on therapeutic development is becoming clear. The exciting precision and efficacy in which base editing can edit the genome is just the start of the base editing story. REFERENCES 1.

Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems.Science339, 819–823 (2013). 2. Mali, P. et al. RNA-guided human genome engineering via Cas9.Science339, 823–826 (2013). 3. Komor, A. C. et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016). 4. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science (80-). 353, aaf8729–aaf8729 (2016). 5. Billon, P. et al. CRISPR-mediated base editing enables efficient disruption of eukaryotic genes through induction of STOP codons. Mol. Cell. 2017;67:1068–1079.e4 (2017). 6. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551 (7681): 464-471 (2017). 7. Kurt, I. C. et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 10.1038/ s41587-020-0609 (2020). 8. Zhao, D. et al. New base editors change C to A in bacteria and C to G in mammalian cells. Nat. Biotechnol. s41587-020-0592-2 (2020). 9. https://horizondiscovery.com/en/news/2020/Horizon-Discoveryto-provide-access-to-novel-base-editing-technology, visited on 10 May 2020. 10. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with 20 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

refractory cancer. Science 367, eaba7365 (2020). 11. Webber, B. R. et al. Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors. Nature 10(1): p5,222 (2019). 12. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–868, 10.1093/nar/gkv1222 (2016). 13. Landrum, M. J. et al. ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 42, D980–985, 10.1093/nar/gkt1113 (2014). 14. Liang, P. et al. Correction of β-thalassemia mutant by base editor in human embryos. Protein Cell. 8(11):811-822. (2017). 15. Liu, Z. et al. Efficient generation of mouse models of human diseases via ABE- and BE-mediated base editing. Nat Commun. Jun 14;9(1):2338 (2018).

Kevin Hemphill Kevin Hemphill is an R&D Scientist at Horizon Discovery. Since joining the Company in 2017, he has supported the development of new products for gene editing and modulation and has contributed towards the commercialisation of Horizon’s base editing platform. Previously, Kevin worked as a scientist at GE Healthcare Dharmacon after obtaining degrees in biochemistry from New Mexico State University and molecular biology from the University of Colorado Boulder. Email: kevin.hemphill@horizondiscovery.com

Winter 2020 Volume 3 Issue 3


Corporate Profile

Diamond – 15-year Anniversary

As it celebrates its 15 birthday, Maureen Graham, Managing Director and founder of East of England based technical services and regulatory affairs consulting group, Diamond Pharma Services (Diamond), reflects on how far the company has progressed, including the recent realisation of her biggest ambition to date – to work on a gene therapy within the ophthalmic space. th

Maureen Graham founded Diamond in 2005, initially employing just two people. The organisation has now grown to over eighty and is a key player in the field of pharmaceutical regulatory affairs and product development. Diamond provides support from early-stage development through to life-cycle management of approved products, across all therapeutic areas, medicinal products, and devices, including small-molecule and next-generation biologics, to cell and gene therapy products. Diamond Then vs Now Maureen established Diamond after 20 years gaining broad experience in leadership roles in the regulatory affairs teams of some of the world’s leading pharma and biotech companies. Her background meant there was little within the regulatory field that she had not touched throughout her career, enabling the company, even in those early days, to offer services of every type across the spectrum. Diamond very quickly became a specialist in the field of advanced therapies, with its first project in this niche area in early 2006 leading to the approval of the first gene therapy product in Europe. Other focus areas in the early days were patient information leaflet (PIL) user testing, to help companies deal with retrospective testing of information in the wake of new legislation that came into force in October 2005, and from 2007 Diamond also took on GMP compliance consulting. Diamond PV Services, offering full pharmacovigilance support, was formed in August 2007 to join all services under one umbrella, or as Maureen says: “to complete the diamond necklace”. The organisation now offers a complete service through regulatory, compliance and pharmacovigilance. Key Achievements From inception, Diamond grew into an organisation with truly talented people, enabling it to become a key player in the field of pharmaceutical regulatory affairs and product development. Reflecting on the proudest achievements since founding the company, Maureen said that, “It is hard to pick out ‘proudest’ achievements, as everything we do is a great achievement no matter how small the task. If I had to, I would say my staff are the proudest achievement”. A quick reflection on some of the achievements of Diamond sheds light on just how far the company has come; Diamond has assisted with market authorisation for multiple ‘firsts’ including the first gene therapy approval in Europe and one of the first CAR T cell therapy approvals in Europe. In addition, www.biopharmaceuticalmedia.com

the team provides valued UK/Irish support for clinical trial activities for a major multinational, with a relationship spanning years. Challenges There has been a period of uncertainty around the United Kingdom’s exit from the European Union since the vote to leave in 2016. With the end of the transition period fast approaching in December, the full impact on the pharmaceutical and biotechnology industries is still not known. What is known, however, is that Diamond is well placed to help its clients adjust in response to the impact of Brexit on the industry. Diamond is a global organisation, with UK-based teams, fully-trained staff in mainland Europe, and a tight network of affiliates across Europe which will allow it to continue to operate all EU activities across the regulatory and pharmacovigilance sectors, ensuring active engagement with European national competent authorities and the European Medicines Agency (EMA), plus the added bonus of being able to support for Brexit and the UK. Like every organisation this year, Diamond has had to adapt to the challenges that the COVID-19 pandemic has forced upon us. “At Diamond we were slightly ahead of the game, having made sure all staff were fully set up for remote working weeks prior to the March lockdown here in the UK,” said Maureen. “We found that we were able to operate business as usual, and thank all of our staff for adapting so well to the changes.” On the easing of the lockdown, Diamond ensured that the office was compliant with government guidelines and the team will be able to return to the office when the time is right, with another smooth transition. A Diamond Future Looking ahead, Diamond is on course to consolidate its already sparkling reputation. Continued growth into other territories including the USA is a key focus, as is continuing to offer quality service and advice to clients both new and old. Maureen is very clear that her vision for Diamond is to surpass expectations, and to continue to work on exciting products and projects. “I really enjoy the relationship we have with our clients; nurturing these relationships helps us understand their needs and deliver our excellent service. Working with and surpassing the expectations of such exciting, cutting-edge companies is at the heart of everything. We look forward to working on more ‘firsts’, and to continuing to strive to be the best!”

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 21


Research / Innovation / Development

Re-modelling Drug Delivery Services for High-viscosity Formulations Biologic formulations for subcutaneous delivery allow patients to self-administer their own treatment and reduce the need for them to consult hospital-based healthcare practitioners. Intravenous formulations are therefore being reformulated for subcutaneous injection (where possible) to capitalise on these benefits. Adding to this, the continued entry of biosimilars into the market in recent years has made these therapies more readily accessible and affordable, further accelerating the growth of the biologics market. As the subcutaneous route of administration continues to gain in popularity, however, there is an ever-growing need for innovation in drug delivery devices. In this article, we look at the challenges faced by manufacturers and device engineers when adapting drug delivery devices to new biologic formulations and key strategies and developments within the industry to overcome these.

Reformulating Intravenous Drugs for Subcutaneous Delivery Therapies formulated for intravenous delivery are usually administered in a clinical setting, which is cost- and resourceintensive. In a context of healthcare worker shortages and limited budgets, the subcutaneous route for drug delivery offers many benefits, notably allowing self-medication by patients away from the hospital. While subcutaneous injections may vastly improve treatment for a range of diseases, these also come with a new set of challenges for the industry. The new wave of biological drugs is typically made up of higher-viscosity and higher-volume formulations which are more difficult to administer. There is therefore an imperative to overcome the challenges associated with these biological drugs through adapted drug delivery device design, and to ensure continued safety and efficiency of injection procedures. This is particularly crucial at a time where self-administration of therapies by patients in their home environments is more common – placing ease-of-use, patient comfort and safety at the top of the agenda for device designers and manufacturers. Molecular Structure and Viscosity Biologics for subcutaneous delivery present a particular challenge for device development due to their molecular structure. Biotherapeutics tend to require injection due to their large, complex protein molecules, but they are highly viscous compared with chemically-based medicines. Monoclonal antibodies (MAbs), for instance, usually require a high dose and are therefore formulated at high concentrations, causing the viscosity to increase significantly. Standard prefilled syringes and safety devices are typically designed with 1mL fill volumes in mind, and for formulations with a viscosity below 10cP. New high-viscosity, high-volume 22 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

biological formulations are quickly surpassing these design parameters for drug delivery devices and creating an impetus for innovation within this field. Furthermore, patient preferences and the need for increased compliance have fuelled a desire for less frequent injections. While there is a clear benefit in enabling less frequent injections from a patient perspective, this poses additional challenges in requiring larger volumes and potentially higher-viscosity formulations. Viscosity: Its Relationship with Temperature While some subcutaneous formulations are already high in viscosity, other elements may come into play to further increase viscosity and make injection more difficult. For instance, viscosity increases with decreasing temperatures. Yet biologics are typically refrigerated at 2–8 °C. Manufacturers will therefore need to map the time it takes for a drug solution to return to a temperature that is comfortable for patient administration. Patients must then factor in that time when removing the product from storage to allow for it to reach room temperature before use. Elements such as this should be factored into the instructions for use for the final combination product. How does Viscosity Impact Injection Force and Time? The impact of viscosity on ease of injection is significant. For instance, high-viscosity formulations require a higher injection force. Human factors studies have shown that in practice, a patient’s typical injection force is 10 N or under, but this may be hard to achieve with higher-viscosity drugs. Furthermore, higher-viscosity (and higher-volume) therapies will also take longer to administer. Given that patients typically only hold the device for 10-15 seconds during drug delivery, this creates new obstacles for device designers. If the drug delivery exceeds the time limitations for patients self-administering, this can interfere with patients’ ability to receive the full dosage and complete their treatment. This clearly has implications for achieving compliance to their given therapies and achieving good outcomes. Larger-gauge Needles and Pain Another challenge to subcutaneous injection is the size of the needles required for some biologic formulation. While needle gauges with larger diameters can facilitate the delivery of high-viscosity formulations, they may be more painful during the injection procedure for the patient. For more viscous drugs, thin-wall needles have been developed and are commonly used to help improve the flow of the drug in needles of a small diameter. Getting Design Engineers Involved When designing drug delivery devices, design engineers and human factors specialists will therefore need to consider all of these important factors which could otherwise negatively impact the ability to inject the biologic as well as patient Winter 2020 Volume 3 Issue 3


Research / Innovation / Development formulations; however, the formulations are small enough in volume that any drug leakage during the delivery process would have consequences on the efficacy of the treatment. Ensuring the full dosage is delivered is essential to the effectiveness of the treatment, making manufacturability and a robust device design key to the successful injection of high-viscosity new-generation biologics. For instance, while most safety syringes are designed to include an internal spring to activate the safety mechanism, this may cause complications during transit. More specifically, this may lead the device to activate and could cause leakage in the process. Furthermore, manufacturing processes using springs must be carried out when they are under a high tension, which makes it a complex procedure with a potential level of product rejections. A springless solution is therefore ideal for manufacturers and patients alike. An added benefit of this design is that the absence of a spring allows patients to more clearly see and check the contents of the syringe before injection, as well as afterwards, to ensure that the full dosage has been delivered.

adherence. There are a number of strategies which can be considered to lower the viscosity of these therapies, but though they may be advantageous, each comes with its own challenges. For instance, lowering the concentration of the drug may be effective in reducing the viscosity but will in turn increase the required volume of the dose. Given that the recommended maximum volume for a single subcutaneous injection is 2–3mL, this may not be a feasible route of action. Beyond this point, wearable devices that enable selfadministration are available, but these have seen a low uptake to date. This may be because wearables must be worn in place on the injection site for a period of time to allow for drug delivery. Patients may be hesitant to use wearables if they are visible and can impact daily activities such as showering or swimming. For this reason, single periodic injections, even if they are required more frequently, may be preferred by patients. The Emergence of 2.25mL Syringes As a response to the challenges discussed above regarding biologic injection, we are starting to see a growth in prefilled syringes suitable for volumes of up to 2.25mL, as well as safety devices to administer them. Designed for higher-volume and higher-viscosity drugs, 2.25mL syringes are ideal for biologics and biosimilars. Since the level of pain and the difficulty of injection can hinder patient compliance, the ergonomics and ease-of-use of the drug delivery device is critical. Prefilled safety syringes adapted for higher volumes enable patients to control injection speed and force, and potentially reduce the level of pain on injection. Safety Features: Springless Designs and Non-removable Plungers New biologics may require comparatively larger-volume www.biopharmaceuticalmedia.com

Ensuring the syringe plunger at the rear of the device cannot be removed is another important design consideration to prevent leakage. Non-removable plungers help prevent wasting costly biologics and creating issues, especially where dose accuracy is critical. Plungers designed to stay fixed also serve to impede tampering or multiple use of devices that are appropriate for single-use only. Conclusions The new generation of biologics has the potential to provide better treatment for a range of diseases. However, the highervolume, higher-viscosity nature of their formulations has also brought about new challenges for the medical device industry. In particular, drug delivery device design must be reconsidered to cater for these new biologics and their requirements. With home-administration on the rise, optimising drug delivery devices design will be important to patient adherence, quality of life and treatment effectiveness as well as commercial success. Manufacturers and device engineers will need to give full consideration to the impact that biologics have on injection time, force and pain – and strive to optimise the comfort and ease-of-use of their devices for healthcare professionals and patients alike.

George I’ons George is currently Head of Product Strategy and Insights at Owen Mumford, having worked for the former OEM and now Pharmaceutical Services division of the organisation since 2006. His current focus is on deciphering the rapidly changing pharmaceutical and biotech sectors in relation to their needs for combination products. In his previous roles in business development, he worked closely alongside R&D to develop devices for a variety of global pharmaceutical and diagnostic clients. Prior to Owen Mumford, George worked for Abbott in EMEA marketing roles in Germany, focusing on their diabetes business.

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 23


Research / Innovation / Development

A Global Approach to Nitrosamine Impurity Testing for Drug Products At first glance it may seem strange for an article about nitrosamine impurities to be in a journal targeted at a biologic audience, since nitrosamines are not formed during the biosynthesis of living organisms. Nevertheless, there is potential for such impurities to occur in biopharmaceutical products via a number of different routes.

Firstly, they may be present in the ingredients used for formulation of the product and in various chemicals used for downstream processing of the active biopharmaceutical ingredient, particularly during certain chromatographic steps. Secondly, nitrosamines can be generated during the production of antibody drug conjugates (ADC), where there is a risk that free amino groups on the antibody could be transformed into nitrosamines. This modification can be very challenging to identify due to their low incidence and, moreover, their impact on the safety of the product is difficult to evaluate at the current time. Finally, exposure to certain packaging, such as nitrocellulose blister packs, has also been suggested as a potential source of nitrosamine contamination. In summary, there is some potential risk for contamination of biologic drugs with nitrosamines and this is likely linked to the quality of the ingredients used for formulation and/or the chemicals used during production or packaging. Discovery and Concern In June 2018, a manufacturer of active pharmaceutical ingredients (APIs) in China was made aware of an impurity in its Valsartan product. By July, this impurity had been identified as N-nitrosodimethylamine (NDMA), one of several nitrosamines. This was the first time a nitrosamine impurity had been detected in a pharmaceutical product and it resulted in multiple regulatory warnings and product recalls. This discovery, and the identification of further nitrosamine impurities in APIs, has been a wake-up call to pharmaceutical manufacturers that they need to adopt a more proactive approach to risk assessment and mitigation in relation to genotoxic contaminants. Nitrosamines are organic compounds with the chemical structure R2N−N=O, where R is usually an alkyl group. They have long been recognised as environmental contaminants and have been found in both water and food, but their presence in an API was unexpected. The investigations conducted by the European Medicines Agency (EMA), U.S. Federal Drug Administration (FDA) and Health Canada all identified the Valsartan API contaminant as NDMA. Valsartan and ARBS Valsartan is part of a group of angiotensin II receptor blockers (ARBs) used to treat hypertension. Other ARBs include Olmesartan, Candesartan, Irbesartan and Losartan. The presence 24 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

of nitrosamine impurities in these drug products (DPs) led to the worldwide recall of batches of ARB, with various regulatory bodies listing the recalled products and their manufacturers on their websites. The identification of NDMA in July 2018 inevitably led to the question of how the impurity contaminated the API. The manufacturer’s analysis concluded that the impurity originated in the use of sodium nitrite to remove excess sodium azide during the manufacturing process. This may have allowed the formation of nitrous acid, which then reacted with trace amounts of dimethylamine to produce NDMA. This change in manufacturing process from the originator (which used tributylin azide) was approved by the U.S. FDA in 2012. Looking at the wider picture of NDMA contamination, it was conjectured that the impurity had developed during the formation of the tetrazole group in the molecular structure, common to this group of APIs. This meant that ARBs without a tetrazole group, such as Azilsartan, Eprosartan and Telmisartan, were exempted from the recalls by the EMA. There are several possible causes for the presence of nitrosamine impurities in the finished DP, including formation in the presence of two or three amines or nitrites, inadvertent use of contaminated equipment or reagents, or the reuse of materials, e.g. solvents. Recent U.S. FDA guidance, published in 2020, lists at least seven potential root causes for nitrosamine contamination, while the most recent guidance by EMA lists eleven. The variety in possible root causes can make accurate risk assessment a challenging proposition. Amines and nitrite salts formed under acidic reaction conditions have emerged as potential generators of nitrosamines from tertiary and quaternary amines used as reagents. Amide solvents have also been considered as a source for secondary amines. Such impurities may also arise from vendor-sourced materials, which has highlighted the importance of full transparency in manufacturers raw material supply chains. Stakeholders should be aware that nitrosamine impurities within a DP can also vary widely between batches, increase during storage, and may even be present in potable water. The threat posed by these genotoxic substances and their unexpected presence in API/DP has made it clear a new, proactive approach to risk mitigation needs to be adopted by manufacturers. Testing Regulatory agencies around the world have set acceptable limits for the level of nitrosamine impurity in relation to ARB. The U.S. FDA has determined that nitrosamines should not be present in either the ARB API or DP. They have also published interim acceptable intake limits based on the maximum daily dose for NDMA, NDEA and NMBA. These are: Winter 2020 Volume 3 Issue 3


Research / Innovation / Development

• • •

NDMA – 96 ng/day NDEA – 26.5 ng/day NMBA – 96 ng/day

The same limits for NDMA and NDEA have also been published by the EMA. If laboratory testing finds that nitrosamine impurity levels exceed these limits, manufacturers can be required to conduct voluntary recalls. The regulatory agencies have also published several test methods for the detection and quantification of nitrosamine impurities in ARB. Health Canada has published a GC/MS/MS method that uses direct injection. This was developed in relation to the presence of NDMA and NDEA in Sartan API and DP. In addition, Health Canada has also published both a limit of detection (LOD) and limit of quantitation (LOQ) for these substances. They are: • •

NDMA: 0.002 ppm LOD and 0.0054 ppm LOQ NDEA: 0.002 ppm LOD and 0.0073 ppm LOQ

The FDA has published a GC/MS method for NDMA in Valsartan API and DP, with an LOD of 0.05 ppm and LOQ of 0.3 ppm. More recently, they have also published an LC-HRMS method that covers six different nitrosamine impurities in the Sartan ARB group of molecules. The six nitrosamine impurities are: • • • • • •

N-nitrosodimethylamine (NDMA) N-nitrosodiethylamine (NDEA) N-nitrosoethylisopropylamine (NEIPA) N-nitrosodiisopropylamine (NDIPA) N-nitrosodibutylamine (NDBA) N-nitroso-N-methyl-4-aminobutyric acid (NMBA)

www.biopharmaceuticalmedia.com

The LC-HRMS method was needed because GC/MS methods were unable to detect and quantify NMBA. In the European Union (EU), the Official Medicines Control Laboratories (OMCLs) network has undertaken similar investigations and has developed methodologies for the specific testing of nitrosamines in Sartans. They have developed both LC-MS/MS and GC-MS methods. Ranitidine In September 2019, following a collaborative effort by Health Canada, the FDA and EMA, it was announced that NDMA had now been found in Ranitidine, which is an H2 (Histamine-2) blocker that helps to reduce the production of stomach acid. At the same time as the agencies made this announcement, they also published test methods for detecting and quantifying NDMA and the acceptance criteria. The FDA published an LC-HRMS method, for both drug substances (DS) and DP, after it was observed that GC-based methods elevated NDMA levels in tested materials. This validated method had an LOD of 0.011 ppm and an LOQ of 0.033 ppm. The FDA also published an LC-MS/MS method for detecting NDMA in Ranitidine. The EMA has published two methods – the first is a GC-MS screening method and the second is the CVUA Karlsruhe method. Metformin In December 2019, it was announced, also in a joint statement between the EMA, U.S. FDA and Health Canada, that NDMA had been found in Metformin. Metformin is a prescription drug used to control high blood sugar in patients with type 2 diabetes. As with the Ranitidine announcement, the agencies also published methods for detecting and quantifying NDMA and the acceptance criteria. The U.S. FDA published two methods – an LC-HRMS and an LC-ESI-HRMS method – and the EMA has issued both GC-MS and GC-MS/MS methods. INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 25


Research / Innovation / Development Validation Stakeholders should be aware that, for the data resulting from these test methods to be used to support quality assessments or regulatory submissions, the analytical methodology must be validated by the user. The U.S. FDA also states that the detectable amount of nitrosamine impurity should be based on one of three criteria: • • • •

Limit of detection established in one of the U.S. FDA’s published methods A method equivalent to the U.S. FDA method but published by another regulatory agency Any appropriately developed and validated method capable of an LOD and LOQ equivalent to the FDA method

Risk Assessment – Regulatory Perspective In September 2019, the EMA released guidelines relating to risk assessment in relation to the presence of nitrosamine impurities in API and DP. This was followed by the publishing of similar guidance by Health Canada in December 2019. In July 2020, the EMA’s Committee for Medicinal Products for Human Use (CHMP) concluded that some biological medicines might also be at risk for containing nitrosamine impurities, so the call for assessment has now been extended to include biological medicinal products. These guidelines ask the market authorisation holder (MAH) and/or manufacturer of the API/DP to consider three possible sources of nitrosamine impurity: • • •

API synthetic route – formed during the API synthetic process Manufacturing process – formation during the manufacture of the DP or during storage/shelf-life Contamination – for example, recovered materials such as solvents, reagents, catalysts; equipment, degradation, starting materials and intermediates

mutagenic impurities, which will help them establish an interim acceptable intake. Risk Assessment Process Step 1 – Risk Evaluation The MAH should conduct a risk evaluation that identifies if products are at risk of N-nitrosamine formation or contamination/ cross-contamination. If a risk is identified, the MAH should proceed to step 2. If they do not identify a risk to the active substance, the MAH should report these findings and conduct a risk evaluation of the finished product. Step 2 – Confirmatory Testing Products identified as being at risk of N-nitrosamine formation or contamination/cross-contamination are required to report the confirmed presence of the impurity as soon as possible. The various testing and analytical methodologies which have been published by the regulatory authorities in relation to nitrosamine impurities in API/DP act as a sensible starting point for developing and validating an analytical methodology. However, there is no regulatory requirement to use them. What is important is that the MAH ensure their analytical methods are validated and appropriately sensitive to the API. It is also important for all testing to be conducted in a GMP compliant facility. Whichever analytical methodology is chosen by the MAH, it should be quantitative in nature and fully validated prior to the commencement of the confirmatory testing. If limit-based tests are used, they should be accompanied by appropriate scientific justification in the risk assessment document. Finally, method validation should be performed using the drug product at a strength appropriate to the market. When developing analytical methods, it is important to remember: •

Looking at the question of nitrosamine impurities in this way represents a comprehensive approach to risk assessment – from synthesis to finished product. Stakeholders should be aware this current approach only relates to chemically synthesised APIs. They should also be aware that, as the regulators have noted, the regular references to NDMA, NDEA, NMBA, NEIPA and NDIPA in the literature should not be taken as an exhaustive list of possible nitrosamine impurities. At the same time, the assessment report published by the EMA, which contains multiple nitrosamines, probably contains many substances that are not relevant to the manufacturing of API/ DP. A better approach for manufacturers is to ensure their risk assessment processes can consider and identify all possible nitrosamine impurities. Employing a risk assessment strategy that achieves this goal would ensure that all nitrosamine impurities that could potentially form or be introduced into the API/DP during the manufacturing process are covered in the confirmatory testing programme. If a nitrosamine is not included in the EMA’s assessment report, the MAH should follow ICH M7(R1) principles on 26 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

• •

Possible interference from trace amounts of nitrosamines in testing materials In situ formation of nitrosamines The need to use accurate mass techniques to overcome interferences in the identification of the specific peak of a nitrosamine (e.g. DMF coeluting with NDMA)

If no nitrosamines are detected, this should be reported to the regulatory authority. If a nitrosamine is detected, the MAH or manufacturer of the API/DP should continue to Step 3. Step 3 – Corrective Actions The detection of nitrosamine(s) means the MAH or manufacturer must apply the necessary changes to the manufacturing process to remove the potential for contamination. If one or more nitrosamine impurities are detected below the interim acceptable limit(s), the MAH should: • • • •

Determine the origin of the detected impurity Determine if product recalls are required Perform quality investigations in accordance with written procedures Assess possible root causes and corrective and preventative actions (CAPAs) Winter 2020 Volume 3 Issue 3


Research / Innovation / Development

• •

Evaluate whether potential changes to the facility, materials, equipment and/ or process will reduce the levels of nitrosamine impurities Establish a risk mitigation plan to ensure levels remain below the interim acceptable limit(s) Initiate measures to further reduce the levels of nitrosamine impurities

Collaboration Manufacturers and regulatory authorities around the world are now aware of the potential threat posed by nitrosamine impurities in API/DP. By taking a global, collaborative approach to the problem, they have created a regulatory landscape which allows stakeholders to develop proactive risk assessment strategies that cover a wide range of markets, including Australia, Canada, the EU, Japan, Singapore, Switzerland, and the United States. www.biopharmaceuticalmedia.com

Romain Simon Romain Simon is currently Operation Manager at SGS. Following a PhD from the University of Lyon (France), Romain took up a postdoc position at the University of Victoria (BC, Canada) where he worked on absolute quantitation of proteins in biological fluid by mass spectrometry. More recently, and back at the University of Lyon, he led the core lab dedicated to small molecule analysis by mass spectrometry. Romain joined SGS in Geneva (Switzerland) in 2015 and became Team Leader of quantitative studies, then Operation Manager.

Email: romain.simon@sgs.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 27


Research / Innovation / Development

Transforming the Healthcare Landscape with Synthetic Biology and Living Medicines An exciting emerging field of technology, living medicine uses living cells to treat disease. Using synthetic biology tools, the field is tackling difficult-to-treat diseases, including those previously thought to be untreatable. The development of increasingly complex and diverse engineering tools will facilitate the progress of living programmable therapeutics, with a range of applications including neurodegenerative disorders, autoimmune diseases, metabolic diseases and gut health, infections and some cancers.

There is a well-recognised global need for therapeutic options for diseases that are deemed difficult to treat, or even currently untreatable. The emergence of synthetic biology tools and technologies has led to a growing awareness of the potential for engineered living organisms to tackle these challenging areas. The field of ‘living medicines’ is harnessing the power of synthetic biology to design cells (non-pathogenic bacterial, human or fungal cells) or viruses that are able to produce desirable therapeutic effects. For example, cells can elicit an immune response to target cancer cells and infections, regenerate diseased tissue or deliver therapeutic effectors. Living cells can be engineered to sense and respond to environmental signals, to detect harmful compounds and perform a specific function, thereby targeting specific mechanisms of these unmet diseases. The potential applications for living medicines could also open new avenues into personalised medicine. The discovery of new living medicines has already resulted in major improvements in healthcare and patient outcomes. For example, the field of immuno-oncology has yielded chimeric antigen receptor technology (CAR-T), a cancer cell therapy which engineers a patient’s own immune cells to fight cancer and has already shown significant benefits for certain patients1. CAR-T cells are not obtainable naturally and have to be engineered. There are currently two CAR-T therapies on the market, which have been available since 2017 (Novartis’ Kymriah for the treatment of B-cell precursor acute lymphoblastic leukaemia (ALL) and Gilead’s Yescarta for patients with relapsed or refractory large B-cell lymphoma), and there are many more advancing through the clinic as scientists work to overcome the remaining technical and regulatory challenges2,3. Stem cell therapies offer the potential to treat a range of diseases, injuries and health conditions. Research efforts to establish cell therapies using stem cells are focused on the sources of precisely reprogrammed stem cells as it is currently limited by the availability of human cells. Today, the only stem cell based treatment that is established and approved is hematopoietic stem cell transplantation for the treatment of patients with cancers and disorders that affect the blood and immune system. Although this approach does not involve 28 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

reprogramming, the hope is that this will become possible in the future. Engineered bacterial cells have been shown to respond to the body’s cues, to deliver optimal treatment, with the correct doses of enzymes, biologics and small molecules released at the right time, directly where they are needed. In addition, these engineered bacteria can absorb and break down potentially toxic molecules. For example, strains of E. coli are currently being developed that can live in the gut and consume molecules such as phenylalanine, for treatment of diseases where patients’ bodies are unable to process these waste products. Within the bacterial therapeutics field, the focus of significant active research is on two main areas: the identification of microbes that naturally produce therapeutic effects and the engineering of bacteria to produce desirable therapeutic effects. The latter involves the design of genes or complex gene circuits to synthesise molecules inside the cell that in turn will perform a specific function, which in a therapeutic context could mean producing a drug or protein. Researchers are able to hijack the complex system of communication within a live cell by encoding RNA or proteins, controlling the way these cells navigate their environment, respond and interact with one another, and build intricate patterns by responding to specific signals that trigger gene expression. Focus on Applications of Microbial Living Medicines As increasingly complex synthetic biology tools become available, we are seeing therapies being developed to treat a wider range of health conditions, including infections, metabolic disorders, immunotherapy for cancers and other health conditions, as well as gut-related disorders. One of the most active application areas for living bacterial medicines currently is gut health. Probiotics have long been used to treat gastrointestinal conditions, including irritable bowel syndrome, due to their ability to restore balance to the intestinal microflora. Disorders of the gut can be particularly difficult to treat due to the harsh environment of the digestive tract, meaning that therapies delivered orally must withstand exposure to stomach acid, bile salts and digestive enzymes. However, bacteria can be engineered to survive in this challenging environment and deliver therapeutics that can be absorbed by the body in situ or deliver their therapeutic effect in the gut. Several strains of bacteria have been genetically modified for therapeutic use in the gut, with popular chassis organisms including E. coli , Bacteriodes, and lactic acid bacteria (LAB), with E. coli being the most studied. Research has demonstrated the success of E. coli in reducing bacterial infection of Pseudomonas aeruginosa and Salmonella typhimurium by delivering the anti-biofilm enzyme, dispersin B, to the gut4,5. Winter 2020 Volume 3 Issue 3


Research / Innovation / Development While naturally occurring microbiota-directed therapeutic approaches for digestive disorders, such as faecal transplants, already exist, engineered bacterial approaches possess advantages over these due to their ability to confer functions that are not naturally present in the microbiota. Engineered bacteria can be designed to improve the capabilities of regular microbes in several ways. This includes carrying out natural biological processes at enhanced rates, producing effectors that are not native to bacteria, such as human proteins, or degrading harmful amino acids that are produced by genetic mutations in patients with phenylketonuria, for example6. Another exciting research area and potential treatment application for bacterial therapeutics is for solid tumours, which display abnormal blood vessel structure resulting in hypoxic areas, where anaerobic bacteria can survive. The danger of solid tumours is that they can grow freely if undetected by the body’s immune system. Treatment can pose a challenge as the dosage of the drug must be balanced to deliver sufficient toxicity to the tumour to kill the target cells, whilst also being a low enough dose so as not to damage healthy cells surrounding the tumour site. Engineered bacteria could therefore provide a new wave of cancer immunotherapies by triggering a controlled immune response, localised at the tumour site, to target and kill tumour cells without damaging healthy tissue. A study has shown that non-pathogenic E. coli can act as a signal to the immune system when engineered to activate STimulator of INterferon Genes (STING) in phagocytic antigen presenting cells (APCs) and trigger immune pathways in tumours. The resulting effect was shown to remain local to the tumour site, significantly delaying tumour growth, and in some cases even stimulated complete tumour rejection. Not only are the results extremely hopeful in terms of immediate action, but there is also evidence of long-term anti-tumour immunity, with animal models showing 40% long-term survival rates and resistance to secondary tumour challenge7.

and modulation of the immune response, competition for nutrition and specific adhesion sites, and inhibition of toxic protein expression9. For example, many bacterial pathogens’ mode of infection involves formation of a biofilm, which can lead to antibiotic and immune system resistance. As mentioned above, engineered bacteria have proved extremely successful for blocking this, with one study showing an E. coli variant inhibiting P. aeruginosa biofilm formation by 90%10. Another approach to combatting infection through microbial therapy is to orally ingest engineered bacteria, which then enter the lymphatic nodules in the small intestine where they are phagocytosed by APCs and express antigen genes. The antigens are processed by the APCs, stimulating mucosal immune response – the body’s first defence against infection, for example in the gut or nose. This approach is very similar to a vaccine, offering protection against transmission with little to no side-effects, as it is made by the body’s own immune system. With applications ranging from Salmonella to Zika, this platform of oral, living vaccines could play a key role in addressing the challenge of distributing vaccines rapidly across the globe11. The Importance of Synthetic Biology in the Development of Living Medicines Synthetic biology plays a crucial role in facilitating the development of living medicines, providing researchers the ability to fine-tune organisms, using precision DNA synthesis or DNA editing to re-engineer desired characteristics onto the framework, to control cellular or viral behaviour and functions. For example, the first step in developing a novel bacteria-based therapeutic requires identification and design of therapeutic genes or pathways, followed by a series of processes taking the idea from initial concept through to prototype generation, strain optimisation, lead and candidate selection, as shown in Figure 1.

Another mechanism for microbial cancer treatment is by engineering bacteria to deliver anti-tumour cargo to tumour sites, which disrupts the tumour microenvironment. The delivery of these payloads can be controlled through the regulation of bacterial gene expression, making it possible to control the timing of delivery as well as limiting further accumulation of payload. The microbes can secrete bacterial toxins to facilitate tumour regression. Toxins from S. typhimurium, Listeria, Salmonella and Clostridium have been shown to directly kill tumour cells through inducing apoptosis or autophagy. This offers an exciting advancement over traditional methods of dosing patients, as the precision reduces any damage to healthy tissues whilst maintaining optimal dosage at the tumour site. These treatments can also be used in conjunction with immune activation therapy, for a multi-targeted approach for cancer therapy8. Antibiotic resistance is recognised as one of the world’s greatest public health threats, with bacterial infections being a major cause of morbidity globally. There is growing evidence that engineered probiotics could provide an innovative, alternative treatment for infections through various mechanisms. They have been shown to demonstrate great specificity and efficacy, including the secretion of antibacterial chemicals, stimulation www.biopharmaceuticalmedia.com

Figure 1: Development of Bacterial Therapeutics: Schematic representation of a workflow for developing clinical candidate-quality engineered strains. The development workflow should incorporate technologies for optimising strain potency, as well as predictive in vitro and in vivo assays, and quantitative pharmacology models, to maximise translational potential for patient populations6. INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 29


Research / Innovation / Development Synthetic biology gives researchers a ‘genetic toolbox’ to enable engineering of more complex functions of organisms and cells. Platforms in development, such as DNA writers, will give researchers access to DNA synthesis, allowing them to identify and test pathways and gene circuits in real time. CRISPR has played a key part in the development of living medicines, and advancements in this field have been developing rapidly over recent years. There now exists a wide set of genetic tools which give living medicines their desired qualities by controlling input sensing, gene expression control devices, memory, and the production of molecules. There is even the ability to develop biocontainment-enabling tools to address safety concerns, including ‘kill-switches’, which prevent uncontrolled replication and spread, and protection of therapeutic organisms from antibiotics. Safety measures such as these are vital for the advancement of bacteria-based therapeutics into human trials. Some therapies have shown great promise in animal models but safety regulations mean they are not Generally Recognised As Safe for human use. The development of synthetic biology tools will be key to driving this forward11. The ability to control the timing and locality of therapeutics delivery is a huge potential advantage of synthetic biology in the living medicines field. For example, therapies involving bacteriaproducing tumour-lethal treatment must only confer toxicity at the target site (i.e. at the tumour mass) to avoid damaging healthy surrounding tissues. In addition, gene expression may be induced through the application of small molecules that ‘turn on’ molecule production as demonstrated for example by salicylateinducible gene expression in Salmonella typhimurium, which confers the ability to synthesise more salicylate12. Future Outlook – Challenges and Opportunities It remains a challenge to understand the complexity of human diseases and disorders, as well as the relationship with the engineered organisms being used to target them, and this continues to present a major barrier. However, with increasing knowledge of these mechanisms, both the exploitation of living biotherapeutics and development of improved tools will be enabled. A particular challenge is the development of orthogonal regulators (such as promoters, transcription regulators, and regulatory RNA) to facilitate more complex genetic circuits. This is key to ensuring the safety of therapies and will help them progress through clinical trials. Biocontainment poses a particular challenge in therapies that require repeat dosing, but with more regulators available, it will be easier to incorporate safety features such as biocontainment12. Currently, there is no single, broad solution for the manufacture of engineered living biotherapeutics. The scale-up production of engineered cells also presents challenges from a cost and quality perspective. For bacterial cells, cell viability must be confirmed during and after fermentation, downstream processing, formulation and storage. There is also the challenge of maintaining genetic stability during cell production. In addition, the inclusion of engineered genetic circuits encoding novel effector functions may come at a cost to bacterial fitness and/or growth rates, and 30 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

this can lead to selective pressure for strain variants that have lost the engineered function. There is therefore a need for tight regulatory control of gene expression and robust assays to assess this. The expansion of the toolbox for engineering living medicines is expected to address these challenges, facilitated by the fast growth and progression of synthetic biology. Recent years have seen significant progress, and with gene editing technology constantly improving, this is expected to continue apace. A huge advancement will come when we have reached the milestone of proof of concept and progression of a living therapeutic to clinical trial. Once achieved, living medicines will create a new category of drugs to address significant unmet therapeutic needs and improve outcomes for patients. REFERENCES 1.

CAR T Cells: Engineering Patients’ Immune Cells to Treat Their Cancers (Originally published by National Cancer Institute 2019) 2. Press Release: Novartis receives first ever FDA approval CAR T therapy, Novartis (2017) 3. Press Release: Kite’s YescartaTM (Axicabtagene Ciloleucel) becomes first CAR T therapy approved by the FDA for the treatment of adult Patients with relapsed or refractory large B-cell lymphoma after two or more lines of systemic therapy, Gilead (2017) 4. Hwang, I.Y. et al. Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat. Commun. 8, 15028 (2017) 5. Palmer, J.D. et al. Engineered probiotic for the inhibition of Salmonella via tetrathionate-induced production of Microcin H47. ACS Infect. Dis. 4, 39–45 (2018) 6. Charbonneau, M.R., Isabella, V.M., Li, N. et al. Developing a new class of engineered live bacterial therapeutics to treat human diseases. Nat Commun 11, 1738 (2020) 7. Leventhal, D.S., Sokolovska, A., Li, N. et al. Immunotherapy with engineered bacteria by targeting the STING pathway for anti-tumor immunity. Nat Commun 11, 2739 (2020) 8. Duong, M.T., Qin, Y., You, S. et al. Bacteria-cancer interactions: bacteriabased cancer therapy. Exp Mol Med 51, 1–15 (2019) 9. Zhou, Z., Chen, X., Sheng, H. et al. Engineering probiotics as living diagnostics and therapeutics for improving human health. Microb Cell Fact 19, 56 (2020) 10. Saeidi, N., Wong, C.K., Lo, T.M., Nguyen, H.X., Ling, H., Leong, S.S.J., Poh, C.L. & Chang, M.W. Engineering microbes to sense and eradicate Pseudomonas aeruginosa, a human pathogen. Mol Syst Biol. 2011;7:521 11. Flehinger, R. Vaxonella vs. Zika. Drug Discovery News. 16:10 (2020) 12. Pedrolli, D.B. et al. Engineering Microbial Living Therapeutics: The Synthetic Biology Toolbox. TIBTech 37, 1:100-115 (2019)

Raquel Sanches-Kuiper Raquel Sanches-Kuiper was appointed Director of Biology of Evonetix in February 2017. Raquel is an experienced R&D leader with a track-record of taking new ideas from concept phase to commercialised products in the next generation sequencing (NGS) space. Raquel was a post-doctoral researcher in the Department of Chemical Engineering and Biotechnology, University of Cambridge, in the area of cancer gene therapy. Raquel holds a PhD in molecular and cell biology from the Faculty of Medicine, University of Auvergne and an MSc in biochemistry from the Faculty of Sciences, University of Lisbon. Email: raquel.sanches-kuiper@evonetix.com

Winter 2020 Volume 3 Issue 3


Producing breakthrough treatments requires continual advances in bioprocessing productivity. That’s why Avantor® is your trusted partner in producing life-saving therapies. Our J.T.Baker® BAKERBOND® PROchievA™ affinity chromatography resin offers best-in-class purification performance in the critical protein A chromatography step of monoclonal antibody production, using a proprietary ligand to improve efficiency. We focus our solutions on your needs so that you can focus on science. See how we help advance monoclonal antibody manufacturing at www.biopharmaceuticalmedia.com avantorsciences.com/moves-science-forward/ibi/performance

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 31


Research / Innovation / Development

Helping Labs Operate at Peak Efficiency to Foster Innovation Science, as Vannevar Bush wrote1, is an endless frontier. Bush, the founder of Raytheon and Director of the Office of Science and Development at its inception in 1941, pushed the US government to invest heavily in research – leading to a renaissance of advancement in science and technology. Unfortunately, as more and more businesses have realised, the resources to explore that frontier are not endless. Although the pursuit of knowledge and scientific advancement knows no limits, the price tag for carrying out research is too often strictly defined and increasingly prohibitive. Simply put, science costs a lot of money.

The Tufts Center for the Study of Drug Development estimated the cost for developing and marketing a new drug at over $2.5 billion2. One study3, tracking 12 leading companies in the pharmaceutical industry, revealed that in a span of six years, costs to develop a scientific asset rose 33% while the projected sales, or return on investment (ROI) of those assets, declined by 50% [Fig. 1].

Figure 3: Global pharma sales of conventional products are forecasted to drop almost 10% by 20204

the burgeoning success in research. Changes in the market are causing companies to transform their approach and adapt to stay viable. Many innovative drugs have already been discovered and pushed to market while others are running up against the patent expiration clock. In 2017 alone, the patents of at least 20 drugs representing over $9 billion in estimated annual revenue expired5. Losing patent protection is costly; it’s estimated that 90% of sales are lost to generic drugs6. Available time and resources have only added to the difficulty faced by labs. Scientists spend, on average, 42% of their time handling non-core, administrative tasks7 – time that is taken away from discovery and innovation [Fig. 4].

Figure 1

Figure 4

Figure 2: These industry leaders have seen their R&D returns drop from 10.1% to 4.2% in the same time span

The era of the “blockbuster” drug – medicines that brought in billions of dollars a year – led to pharmaceutical companies pouring vast amounts of money into infrastructure to support 32 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Despite these obstacles, demand is growing. With the rise of speciality drugs and personalised medicine, the biotechnology sector is forecasted to realise a near 50% increase in sales8. Ageing populations living longer and an expanding global market push growth further, as the amount of clinical trials tracked by the NIH have nearly quintupled in the past decade9 [Fig. 5]. Winter 2020 Volume 3 Issue 3


Research / Innovation / Development In order to meet growing market demand and offset decreasing ROI, companies have turned the trend of outsourcing lab work into an important business truth: Science is now a service needed to propel companies forward. Benefits of Outsourcing There are six main factors driving pharmaceutical and biotech companies to outsource their laboratory experiments, clinical trials and manufacturing production work. These areas cause additional complexity that is often difficult to address by a scientific organisation alone.

Figure 5

Pharmaceutical sales have increased by 22.6% in the fast-industrialising market of BRICS countries (Brazil, Russia, India, China and South Africa) as many barriers to free trade have recently been removed through economic and policy changes in global trade10. Thus, constricting profit margins, matched with a growing demand and time constraints, are leading to a greater focus on the bottom line. Companies want to propel innovation and discovery while offsetting costs through business tactics and strategies11 – seeking ways to tighten the belt. Doing so has become a game of analysing every “loop” in the “belt” of research. In any major research and development programme, there's a long chain of diversely talented professionals – technologists, scientists, researchers, directors, managers, medical doctors and others – who lead to the ultimate success or failure of the research outcome. For each one of these resources, another resource must be situated to manage the asset and the process. It is through critical evaluation of this R&D value chain that organisations across the research spectrum are able to realise gains. Costs are lowered and efficiency improved as more companies outsource routine lab and science work to third parties and CROs. Experts believe that companies will soon outsource all aspects of drug research and development, in addition to clinical trials and manufacturing, thereby creating virtual pharma organisations12. To date, roughly 67% of pharma companies now outsource Figure 6 their manufacturing process13 [Fig. 6]. Outsourcing also allows for greater agility in meeting heavily fluctuating project pipeline demands while maintaining regulatory compliance with the increasingly stringent protocols required to ensure patient safety. Issues with regulatory compliance can result in costly project delays and adversely affect a company's ability to bring its product to market efficiently. www.biopharmaceuticalmedia.com

Bottom line pressure: As the data in the graphs above show, the business environment is growing more complex with patents expiring, innovation and discovery becoming more targeted to smaller populations leading to fewer blockbuster drugs and ROI diminishing related to asset R&D. This has led more companies to offload research, development, quality and production costs to reliable third parties and CROs. Physical transformation: With the cost of a new lab now reaching over $1000 per square foot14, it is clear that space is at a premium in today’s laboratory [Fig. 7]. Organisations are moving away Figure 7 from traditional labs that once sequestered small teams who focused on a single project. Now the emphasis is on open and flexible space or even virtual labs. The open-space lab facilitates interdisciplinary interaction and collaboration – connecting areas of scientific specialisation under one umbrella to truly master innovative technology for the advancement of science. Doing so also centralises complex, high-cost equipment (like automation used in high-throughput screening and high-content biology platforms), reducing overhead and maintenance costs. Virtual labs focus on modelling and simulation 15, requiring communication between many resources spread out globally. Furthermore, organisations are seeking to be closer to target patient groups16 for therapeutic research and clinical trials, requiring labs and scientists to be available to emerging global communities. Technology-driven evolution: As a new generation enters the world of research, the expectation for technologybased process and communication increases, giving rise to a digitally driven workforce17. Labs must evolve to keep up with the latest technologies that allow for more immediate collaborative communication between remote/regional/ international associates, cloud-based data for large dataset storage and analysis, and an easily searchable repository of shared intellectual discovery. The costs to keep up with the ever-expanding technological advancement can become overbearing. Compliance: The overhead costs related to staying compliant with all regulations and regulatory bodies in an increasing global market are complex. Managing the oft-moving rules, INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 33


Research / Innovation / Development staying aligned with organisational values and operating efficiently in cGMP and GxP environments are constant challenges that can make it difficult to stay ahead.

productivity and increasing efficiency in processes and protocolbased work returns valuable time to researchers in achieving scientific discovery, ramping up a lab’s speed to market.

Managing resources: Recruiting and hiring skilled employees and getting them trained in time to be effective becomes a management task outside of the core science. Many labs can no longer keep temporary workers for more than two years due to stringent laws preventing long temporary work; thus, turnover can be high, restarting the time-consuming cycle of hiring and training. Training can become expensive, particularly for high-content biology and gene array profiling technologies. Companies are finding it fruitless to invest in temporary employees who offer no long-term value.

Globalisation: Organisations are realising cost savings by relocating R&D throughout the world. Harnessing the economic advantages of emerging markets demands clear and immediate communication between multiple internationally situated sites, resources and personnel, as well as a reliable global supply chain and logistics network.

Additionally, new laws in some countries require increased pay for postdocs – whom many labs rely on – due to improved overtime rules18, causing a further strain on tight salary allotments. As temporary staffing can be time-prohibitive, CROs are becoming more cost-prohibitive and inflexible to the evolving needs of labs. One recent survey of over 300 global clinical operations leaders revealed that delays in CRO data analysis and reporting lead to an average delay of three months and over $100,000 per CRO change order19. Driving efficiency: Streamlining data management, workflows, global operations and logistics, along with managing employee productivity and quality output in compliance with myriad regulations can swiftly become arduous. Balancing all of these essential elements while driving down costs turns an organisation's focus away from innovation and discovery. With the benefits of outsourcing clear, the question organisations must ask, then, is not if they should outsource, but to whom should they outsource? Finding a trusted, reliable third party with a proven track record is critical to success with the outsourcing model. Delivering Science as a Service to the Lab of the Future As business in innovation and discovery progresses forward through the limitless frontier, so must the environment and technology in which science is conducted. Organisations must be prepared to bring a modern lab into the advanced age, allowing their scientists and researchers to excel in the lab of the future. Doing so means adjusting to the major factors driving the need for the lab of the future. Marketisation: Quickly turning innovative research into a marketable product has never been more important. Improving

34 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Digitisation: More sophisticated tools are enhancing the role of the researcher while digital solutions are revolutionising the entire supply procurement and management process. Furthermore, younger scientists entering the field are well acquainted with technology, which instils them with a more flexible approach to work. This new generation has come to view aspects of work, like remote access, flexible work environments, and digital tools and solutions, as the norm instead of the product of future advancements. Solution: Outsource Science as a Service These significant and ongoing challenges make it clear that, for labs to accomplish their work, there are significant and long-term benefit from outsourcing the key tasks of managing the essential, routine lab operations tasks to expert, professional service organisations. Outsourcing science as a service gives scientists the time and resources to do what they do best: science. Scientists, spared from routine procedural work, can then spend more time on innovation and discovery. Take, for example, the emerging need for cold-chain biosample management. Discovery depends on the integrity of test samples and analysis, but managing the process is time-consuming, tedious, and costly. Trained specialists in these processes remove scientists from the logistical process, while also optimising in-lab space for researchers. Specialists can provide immediate archival and delivery of any test sample or analysis via an advanced database inventory system on demand. Stability: Outsourcing delivers stable and compliant provision of non-core tasks through recruiting, employing and managing highly talented personnel, from lab technicians to career scientists, and developing a career ladder for associates to expand their skillset. This provides stable, trusted services from a competent team devoid of the high

Winter 2020 Volume 3 Issue 3


Research / Innovation / Development 5.

6.

7.

8.

turnover and lowered engagement endemic to temporary staffing agencies.

9.

Quality, safety & compliance: Science as a Service providers have experts dedicated to documenting processes, including scope of work, working instructions and SOPs; managing training matrices and job safety profiles; integrating onsite specific EH&S programmes; and supporting audit programmes.

10.

Reduced operating costs: Various hard and soft cost savings – from inventory and procurement management to employee training and HR management – can be achieved through improved processes and collaboration. Having logistics, supply chain and lab operations HR experts create the processes and deploy the technology tools, driven by mutually agreed-upon KPIs that can identify areas where savings can be achieved and implement programmes that deliver measurable results. Technological advancements: Many lab operations outsourcing organisations invest continually in advanced digital database and process management tools. These systems replace outmoded, time-consuming and error-prone paper processes, allowing for more organised collaborative reporting, swift accounting of product inventory and timely equipment calibration/validation.

11.

12.

13.

14.

15.

16.

REFERENCES 1.

2.

3.

4.

Bush, Vannevar. United States. National Science Foundation. Science the Endless Frontier: A Report to the President by Vannevar Bush, Director of the Office of Scientific Research and Development. Washington, D.C.: GPO, 1945. https://www.nsf.gov/od/lpa/nsf50/ vbush1945.htm. Accessed 8 May 2017. Tufts Center for the Study of Drug Development. Tufts CSDD Assessment of Cost to Develop and Win Marketing Approval for a New Drug Now Published. Boston, MA: Tufts Center for the Study of Drug Development, 10 Mar. 2016. http://csdd.tufts.edu/news/ complete_story/tufts_csdd_rd_cost_study_now_published. Accessed 8 May 2017. Terry, Colin, et al. Deloitte Centre for Health Solutions. Measuring the Return from Pharmaceutical Innovation 2015: Transforming R&D Returns in Uncertain Times. United Kingdom: Deloitte LLP, 2015. https://www2.deloitte.com/content/dam/Deloitte/global/ Documents/Life-Sciences-Health-Care/gx-lshc-pharma-innovation. pdf. Accessed 8 May 2017. Deloitte Centre for Health Solutions. 2016 Global Life Sciences Outlook: Moving Forward with Cautious Optimism. United Kingdom: Deloitte LLP, 2015. https://www2.deloitte.com/be/en/pages/ life-sciences-and-healthcare/articles/global-life-sciences-sectoroutlook.html. Accessed 8 May 2017.

www.biopharmaceuticalmedia.com

17.

18.

19.

20.

Renoe, Jeff. Dickson, Inc.. “[Infographic] The Major Pharmaceuticals Losing Patent Protection in 2017.” DicksonData.com, 5 Jan. 2017. https://www.dicksondata.com/blog/2017/01/05/drugs-losingpatent-protection-in-2017/. Accessed 8 May 2017. Renoe, Jeff. Dickson, Inc.. “[Infographic] The Major Pharmaceuticals Losing Patent Protection in 2017.” DicksonData.com, 5 Jan. 2017. https://www.dicksondata.com/blog/2017/01/05/drugs-losingpatent-protection-in-2017/. Accessed 8 May 2017. National Science Board. United States. Reducing Investigators’ Administrative Workload for Federally Funded Research. Washington, D.C.: National Science Foundation, 10 Mar. 2016. https://www.nsf.gov/ pubs/2014/nsb1418/nsb1418.pdf. Accessed 8 May 2017. Deloitte Centre for Health Solutions. 2016 Global Life Sciences Outlook: Moving Forward with Cautious Optimism. United Kingdom: Deloitte LLP, 2015. https://www2.deloitte.com/be/en/pages/ life-sciences-and-healthcare/articles/global-life-sciences-sectoroutlook.html. Accessed 8 May 2017. United States. ClinicalTrials.gov. “Number of Registered Studies Over Time.” Trends, Charts, and Maps. U.S. National Institutes of Health, 2017. https://clinicaltrials.gov/ct2/resources/ trends#RegisteredStudiesOverTime. Accessed 8 May 2017. 10. Arlington, Steve, et al. From Vision to Decision: Pharma 2020. PricewaterhouseCoopers, 2012. http://www.pwc.com/pharma2020. Accessed 8 May 2017. Mullin, Rick. “Costly Drugs: Cornered on Pricing, Drugmakers Fire Back with Their Standard Message About World-Class Science.” Chemical & Engineering News, vol. 95, no. 9, 27 Feb. 2017. http://cen.acs.org/ articles/95/i9/Pushback.html. Accessed 8 May 2017. Denny-Gouldson, Paul. “The Future Trends in Bioanalytical Outsourcing.” Bioanalysis Zone, 16 Feb. 2017. https://www. bioanalysis-zone.com/2017/02/16/spotlfuture-outsourcing-thefuture-trends-in-bioaanalytical-outsourcing/. Accessed 8 May 2017. Industry Standard Research. “Two-Thirds of Pharmaceutical Manufacturing is Outsourced.” Contract Development and Manufacturing Outsourcing Models. ISR Reports, 18 Nov. 2016. https:// www.isrreports.com/two-thirds-pharmaceutical-manufacturing/. Accessed 8 May 2017. Gering, John. “2015 Lab Construction Outlook.” Laboratory Design, 11 Aug. 2015. https://www.labdesignnews.com/article/2015/08/2015lab-construction-outlook. Accessed 8 May 2017. PwC. Pharma 2020: Virtual R&D – Which Path Will You Take? PricewaterhouseCoopers, June 2007. https://www.pwc.com/gx/ en/pharma-life-sciences/pdf/pharma2020_virtualrd_final2.pdf. Accessed 8 May 2017. Walker, Nigel. “CRO Outsourcing Trends for 2015: The Move to the Cloud, the Web and Mobile Technology Continues.” Pharmaceutical Outsourcing, 28 May 2015. http://www.pharmoutsourcing.com/ Featured-Articles/174600-CRO-Outsourcing-Trends-For-2015-TheMove-to-the-Cloud-the-Web-and-Mobile-Technology-Continues/. Accessed 8 May 2017. Tulsi, Bernard B.. “A New Generation.” Lab Manager, 4 Feb. 2016. http://www.labmanager.com/business-management/2016/02/anew-generation. Accessed 8 May 2017. Benderly, Beryl Lieff. “Postdoc Pay to Increase Due to New Overtime Rule.” Science, 19 May 2016. http://www.sciencemag.org/ careers/2016/05/postdoc-pay-increase-due-new-overtime-rule. Accessed 8 May 2017. Hublou, Rani and Bruno Gagnon. Comprehend Systems, Inc. Clinical Operations Benchmark Report: Survey of Leading Life Sciences Companies. Redwood City, CA: Comprehend Systems, Inc., 2016. http://info.comprehend.com/hubfs/Downloads/2016_ClinOps_ Benchmark_Report.pdf. Accessed 8 May 2017. Paul, Steven M., et al. “How to Improve R&D Productivity: The Pharmaceutical Industry’s Grand Challenge.” Nature Reviews Drug Discovery, vol. 9, Mar. 2010, pp. 203-214. doi: 10.1038/nrd3078. Accessed 8 May 2017.

by Avantor Sciences

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 35


Clinical Research

The Solution to Improve Profitability in Pharmaceutical Development; How to Increase Pre-clinical Productivity and Success Rates in Clinical Trials

PINPOINTING THE OPPORTUNITY FOR INCREASED PROFITABILITY Costs Related to Pharmaceutical Development Pharmaceutical development is associated with substantial costs and long lead times. The average cost to develop a pharmaceutical drug was $2 billion in 2019, an increase of 76 per cent over the last decade1. Clinical trials are the single most expensive component of the pharmaceutical development process, constituting roughly half of the total development cost of a drug2. The increased costs of clinical development are a consequence of performing more extensive, and thus more expensive, quality assurance (QA) assessments in clinical trials. The costs successively increase in each clinical phase as they become more comprehensive, emphasising the importance of identifying potential flaws in early stages and avoiding late-stage failures. Average R&D Cost of a Pharmaceutical Drug from Discovery to Commercial Market 2 500

2 327

Average cost (million USD)

2 250

2 091 1 882

2 000

Average Failure Rates between Pharmaceutical Development Stages 100%

90,4%

80% 69,3% 60%

40%

41,9%

36,8%

20%

0%

14,7%

Phase I to Phase II

Phase II to Phase IIII

Phase III to NDA/BLA

NDA/BLA to Approval

Phase I to Approval

Figure 2. Average failure rates between pharmaceutical drug development stages (Phase I to approval)3.

The limited availability and usage of pre-clinical laboratory instruments which provide biologically relevant data, has resulted in unnecessarily high failure rates of drug candidates in clinical trials. More thorough and insightful evaluations during pre-clinical testing will contribute to improved characterisation and optimisation of candidates before they enter billion-dollar clinical studies. This article will investigate a solution to the challenge that the pharmaceutical industry is facing: How can productivity and profitability be increased in pharmaceutical development? THE QCM TECHNOLOGY – A PROVEN SOLUTION TO IMPROVE THE DEVELOPMENT PROCESS

1 585

1 750

reach Phase III2. One explanation is that pharmaceutical development has traditionally relied on pre-clinical research in artificial in vitro laboratory systems. The artificial experimental conditions, in which the drug candidates are initially assessed, result in artificially positive test results. Accordingly, this often leads to an overestimation of the drug’s efficacy and an underestimation of the side-effects that are only revealed later in subsequent human clinical trials.

Failure rate

The limited availability and usage of pre-clinical laboratory instruments which provide biologically relevant data has resulted in unnecessarily high failure rates of pharmaceutical drug candidates in clinical trials, the single most expensive component of the pharmaceutical development process. Attana’s QCM technology and pre-clinical validation of drug candidates has been shown to improve the efficiency and reduce the costs associated with bringing new pharmaceuticals to market, by increasing success rates in clinical studies and detecting unqualified candidates in the pre-clinical stage. The clinical results from Attana’s validated drug candidates have shown impressive progress in a short time and increased productivity in clinical trials by an estimated 80 per cent, demonstrating an unparalleled offering to clients seeking to maximise the returns of their R&D efforts.

1 497 1 500 1 250 1 000

1 310 1 188

2010

1 175

2011

2012

1 270 1 290

2013

2014

2015

2016

2017

2018

2019

Figure 1. Average R&D cost of a pharmaceutical drug from discovery to commercial market (2010–2019)1.

The Challenge Lies in Clinical Trial Preparation Only one in ten pharmaceutical drug candidates that enter clinical trials successfully reach the commercial market. Although a majority of all candidates entering clinical trials move from Phase I to Phase II, only 22 per cent of candidates 36 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Valuable Pre-clinical Insights from QCM-based Biosensors Historically, the QCM (quartz crystal microbalance) technology has been utilised to measure picogram-level changes in mass per unit area. It was discovered in 1959 by Prof. Günter Sauerbrey and has since then contributed to multiple industries. In the last two decades, Attana has leveraged the technology and pioneered the development of QCM systems for applications in pharmaceutical research & development. Attana’s proprietary instruments have been proven as a reliable solution to several of the technical challenges facing the pharmaceutical industry in the drug development process. Winter 2020 Volume 3 Issue 3


Clinical Research specialists engage with R&D personnel at the institution or pharmaceutical company developing the drug candidate. Upon completing the Attana-enabled studies, data is generated and evaluated, allowing the drug candidates to be categorised: a.

Promising candidates are identified and optimised for clinical trials. An initial assessment determines the candidate’s potential for success. Some candidates show sufficient performance and are recommended for clinical studies without modification. Others are modified based on Attana’s recommendations and if follow-up analyses prove successful, may be recommended for clinical trials.

b.

Unqualified candidates are identified prior to clinical trials. The high clinical trial failure rates are partly a consequence of too many de facto unqualified candidates (poor efficacy, poor safety, or both) being recommended for clinical trials, and partly due to factors beyond the laboratory’s control (such as a competitor reaching the market with a similar drug). Attana has a strong track record in identifying unqualified drug candidates with high accuracy.

Figure 3. Attana’s most recent instrument, the Cell™ 250, which was launched H1 2020.

The Attana Cell™ 250, launched earlier this year, has the ability to analyse how a given pharmaceutical drug candidate interacts with the human body by mirroring in vivo conditions (for more details on Attana’s analyses, see section Attana’s validation process explained below). By enabling the study of molecular interactions between, e.g., proteins, DNA, nanoparticles and viruses, as well as interactions with living cells cultured on the Attana sensor surface, the biosensors can, with exceptional accuracy, analyse a drug candidate’s efficacy and potential side-effects – prior to the commencement of clinical trials. Validating and Optimising Drug Candidates Prior to Clinical Trials To improve the success rates of clinical trials, Attana can validate and optimise drug candidates in the pre-clinical phase. Typically, these analyses are initially performed through contract research projects where Attana’s team of application Attana’s categorization

Financing for clinical studies

Pre-clinical candidates

Pharmaceutical Drug Candidates Assessed by Attana (2013-2020)

39%

30 candidates

61%

47 candidates

Unqualified candidates Promising candidates (validated)

Figure 4. Pharmaceutical drug candidates assessed by Attana (2013–2020) categorised as either promising or unqualified. Phase I

Phase II

Phase III

1 2 3

77

Analyzed Candidates

30

Yes

4 5

Validated

6 7 8

47

Unqualified

No

22

No

47 Total: 77

Figure 5. The clinical results from 77 pharmaceutical drug candidates analysed by Attana. Eight of the 30 validated candidates have received financing for clinical trials to date, and several of the other 22 validated candidates have financing pending. None of the unqualified candidates have entered clinical trials to Attana’s knowledge. www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 37


Clinical Research

Clinical Results from Drug Candidates Validated by Attana A pharmaceutical drug candidate’s chances of both entering clinical trials and successfully reaching the market can be vastly improved with Attana’s solution to characterise, validate and directly recommend modifications in pre-clinical studies. The R&D pipeline of the 77 candidates which have been analysed by Attana in pre-clinical studies is shown below. Of the validated candidates, eight have secured financing for clinical studies to date. Of these, six candidates have successfully completed Phase I, of which two have commenced Phase II studies and one has reached Phase III. In 2016, Attana conducted an analysis of Index Pharmaceuticals’ drug candidate, Cobitolimod4, which has now successfully concluded clinical Phase IIb and is entering Phase III5 (other candidates are undisclosed due to confidentiality). Track Record Illustrating Significant Impact in Short Time To date, Attana-validated drug candidates have had a 100 per cent success rate in clinical trials. Six of six candidates have Phase I success, and one of one has Phase II success with an additional two candidates showing good promise. Conversely, of the 47 candidates which Attana determined as unqualified, none have entered clinical trials (0 of 47).

Attana’s Validated Pharmaceutical Drug Candidates Expected to Enter Each Development Stage 10

Number of candidates

Attana’s Track Record in Pharmaceutical Drug Development Attana has analysed 77 pre-clinical drug candidates between 2013 and 2020 in 26 contract research projects with over 20 international clients. Of the 77 candidates, Attana studies have rejected 47 (61%) for either showing poor safety or poor efficacy. The other 30 candidates (39%) have been validated and determined to have good chances for clinical trial success. Several of these validated candidates have also been modified based on Attana’s recommendations (for details on modification recommendations, see subsection Optimising the characteristics of validated candidates).

8

8,0 7,3

6

4

8,0

2,9 1,7

4,1

2

1,6 0

Phase I

Phase II

Phase III

1,5 0,9

NDA/BLA

Phase II c ompleted

Phase I completed

Phase I ongoing

Indu stry Average

0,8 Approval

Figure 6. Attana’s validated pharmaceutical drug candidates expected to reach the commercial market. The data is based on Attana’s eight validated candidates in clinical trials and their current clinical phase. The industry average failure rates from Figure 2 are applied in all transitions between Phase I to approval.

Improving Clinical Trial Success Rates by 80 Per Cent The Attana technology’s ability to predict in vivo results helps to increase productivity in pre-clinical studies and improves success rates in clinical trials. By applying conservative marketbased averages on failure rates in clinical trials from Figure 2, it can be illustrated how productivity can be increased in the drug development process. Attana’s eight validated candidates have already improved clinical trial success rates by over 80 per cent by having an estimated 1.5 candidates reach the market instead of the industrial average of 0.8 candidates. By extrapolating the results to the pre-clinical stage, candidates that are validated by Attana will generate 4.8x more marketed pharmaceuticals than the industry average6. ATTANA’S VALIDATION PROCESS EXPLAINED

Pre-clinical validation with Attana can save, per average client, 50 per cent of historic clinical trial costs, by eliminating candidates which are highly unlikely to achieve clinical trial success and optimising those which show promise. The early identification of flawed candidates also ensures resources can be allocated to more promising candidates, which ultimately may lead to more pharmaceuticals reaching the market in less time. 1. Prepare analyte for testing

3. Select the appropriate assays for in vivo-like conditions

2. Calibrate the Attana Cell™ 250

Vaccines Antibodies

Predicting in vivo Results through in vitro Assays Enabled by Attana’s third-generation biosensor instruments and the expertise of Attana’s experienced research professionals, the validation process begins with one or more pre-clinical drug candidates not yet evaluated in vivo (clinical trials). The Cell™ 250-instrument is easily calibrated to suit the type of analyte(s) being tested. In order to best mirror in vivo conditions, the

Attana’s QCM biosensor

5. Gather in vivo insights and select top candidate(s)

4. Monitor candidates’ interactions in real time

Cell-based Tissue-based

Proteins

Biochemical

Fabs

Crude-based

Peptides

Sera-based

Other

Off-target Competition Whole blood

Affinity Kinetics Avidity Enthalpy Entropy Buffer variation Epitope binning Active concentration

a. Validate and optimize candidates for clinical trials b. Identify unqualified candidates prior to clinical trials

Degree of aggregation Active receptors per cell

Figure 7. Step-by-step illustration of Attana’s pre-clinical validation process, utilising the Cell™ 250 to mirror in vivo results through in vitro assays. 38 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


Clinical Research appropriate assays, including competition assays if needed, are selected. The assays are then performed and can be monitored in real time, creating a comprehensive interaction profile. From the insights generated by the interaction profile, Attana can (1) expose and understand any off-target interactions and (2) evaluate if the quality of the target interactions is sustained in the in vivo-like conditions.

3.

Selecting the Best Candidates to Enter Clinical Trials The evaluation of target and off-target interactions constitute the foundation of validating a candidate. Attana’s off-target evaluations resemble GLP toxicity studies and is an essential precursor to clinical trials. The candidate’s interaction profile exposes any off-target properties and Attana can attribute these unwanted interactions to a specific part of the antigen. For target interactions, it can be determined from the interaction profile whether their quality meets the requirement for the candidate’s purpose and the desired degree of efficacy is obtained. Based on the degree of target and off-target interactions, Attana provides recommendations for selecting the appropriate candidate(s) to proceed with into clinical trials, along with further optimisation recommendations.

6.

Optimising the Characteristics of Validated Candidates The objective of increasing efficacy and minimising the off-target properties can be achieved in various ways. By attributing the off-target property to a specific part of the antigen, Attana exposes its modifiable characteristics and provides guidance on how to improve the molecular composition of the drug candidate (for case study on improving off-target properties, see Forssén et al.7). To improve the quality of target interactions, Attana can validate the suitable target, analyse the off-and association-rate characteristics and adjust the candidate’s composition accordingly (for case study on target validation, see Bode et al.8). Summary of Results Pre-clinical validation of drug candidates by Attana has been shown to improve the efficiency and reduce the costs associated with bringing new pharmaceuticals to market. Clients have experienced considerable success both in contract research projects with Attana specialists, as well as by employing the proprietary Attana biosensor instruments in their own R&D labs. The versatility of Attana instruments combined with breadth of knowledge and experience possessed by the team of specialists, enables Attana to provide an unparalleled offering to clients seeking to maximise the returns of their R&D efforts. REFERENCES 1.

2.

Deloitte LLP, 2019. Weighted average based on data from 12 Large Cap and 4 smaller specialized pharmaceutical development companies. Pharmaceutical Research and Manufacturers of America, PhRMA Annual Membership Survey, 2019. R&D from corporate funding included.

www.biopharmaceuticalmedia.com

4.

5.

7. 8.

BIO, Biomedtracker and Amplion (2016) Clinical Development Success Rates 2006-2015. Schmitt, Heike, et al. “TLR9 Agonist Cobitolimod Induces IL10-Producing Wound Healing Macrophages and Regulatory T Cells in Ulcerative Colitis.” Oxford University Press, 20 Oct. 2019. Atreya, Raja, et al. “Cobitolimod for moderate-to-severe, left-sided ulcerative colitis (CONDUCT): a phase 2b randomised, doubleblind, placebo-controlled, dose-ranging induction trial.” The Lancet Gastroenterology & Hepatology, 05 Oct. 2020. Deloitte Centre for Health Solutions, Measuring the return from pharmaceutical innovation 2019. Forssén, Patrik, et al. “Reliable Strategy for Analysis of Complex Biosensor Data.” ACS Publications, 28 Mar. 2018. Bode, Kevin et al. “Dectin-1 Binding to Annexins on Apoptotic Cells Induces Peripheral Immune Tolerance via NADPH Oxidase-2.”Cell reports vol. 29,13 (2019). National Library of Medicine, 24 Dec. 2019.

Teodor Aastrup Teodor Aastrup is founder and CEO of Attana AB. He holds an MSc degree in material physics from Uppsala University, Sweden and a PhD in corrosion science from KTH Royal Institute of Technology, Stockholm, Sweden.

Diluka Peiris Diluka holds a PhD in biochemistry from University of Westminster, UK. Following her post-doctoral training in cancer glycobiology she joined Attana AB as a senior application specialist and currently holds a position as a scientific advisor.

Ahmed Ibrahim Ahmed Ibrahim is an application scientist at Attana. Ahmed holds a PhD (Dr. rer. nat.) in molecular biology and cancer research from the Philipps University of Marburg in Germany. Ahmed continued his postdoctoral studies at Yale University, the Ohio State University in USA and at KTH and Karolinska Institute in Sweden.

Amica Johansson Amica Johansson is as application specialist and product manager at Attana. She holds a BSc in biotechnology and a MSc.eng. in medical biotechnology from KTH Royal Institute of Technology, Stockholm, Sweden.

Cecilia Furugård Cecilia Furugård is an application specialist and product manager at Attana. She holds a BSc in molecular biology from Uppsala University and holds a MSc in molecular techniques in life science from KTH Royal Institute of Technology, Stockholm, Sweden.

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 39


Manufacturing/Technology Platforms

Tightly Coupling the Laboratory to the Pharmaceutical Manufacturing Process is Essential to Success Since the time of Henry Ford, the manufacturing process has relied on driving uniform production techniques with close tolerances to reduce costs and drive up volumes. All manufacturing flows, whether making cars, food products, medical devices, or pharmaceuticals, take raw materials, and through a series of manufacturing processes, transform them into a final product. In pharmaceutical production, the constituent parts include the active pharmaceutical ingredient (API), excipients, and packaging and labelling materials. The laboratory, through the function of quality control, plays a central role in ensuring these constituent parts, and the overall product, meet the required quality standards. Since batch release of ingredients and the final product is dependent upon these tests, the speed of batch testing and release through the laboratory is critical to the overall efficiency of the manufacturing plant.

Pharmaceuticals in tablet form may have coating agents added to make them easier to swallow and colouring to differentiate them. Tablets are often packaged in blister packs of 20 or so with labelling on which key consumer information is printed. As with cars or ice-cream, each constituent part from the API to the foil of the blister pack must be traceable back to the original suppliers and batches and be part of the production record. The key is traceability; traceability of everything that went into the final product. A Typical Pharmaceutical Manufacturing Flow

An overview of a typical pharmaceutical tablet production line is shown in Figure 1. The exact proportions of the various raw ingredients are controlled by a recipe which defines the amount of API, and excipient(s) required. The various ingredients may be milled and mixed to ensure an even distribution of API and the resulting bulk powder may be mixed with further excipients before being formed into tablets in a tableting machine. The tablets may be coated and finally packaged as required. Testing will be performed at each critical stage of the flow from raw materials (RM) to the final packaged product. For example, the incoming raw materials may be tested to ensure they conform to the supplier’s specification before being released for use. The intermediate product (IP) from the granulator may be tested to ensure the API is evenly mixed and the IP from the milling checked to ensure the mixture still contains the correct proportions of API and is ground evenly. Samples of tablets from the final product (FP) are retained for dissolution, stability, and other quality assurance purposes. Each batch of final product is linked with the relevant batches of raw materials and intermediate product, ensuring complete traceability through the production process, as shown in Figure 2. The Role of a LIMS Testing of the RMs, IPs and FPs, and ensuring they meet the required quality control and safety standards, is the responsibility of the laboratory, which therefore has a key role in product release. This entails the generation and management of large

Figure 1: A simplified pharmaceutical tablet manufacturing flow 40 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


Manufacturing/Technology Platforms stages and final product); to link test results from the originating raw materials/suppliers to the final product/customers; statistical tools to monitor process control limits; and the ability to quickly retrieve and report on all this data.

Figure 2: Laboratory view of a simplified pharmaceutical tablet manufacturing flow

amounts of critical data. Many organisations have implemented laboratory information management systems (LIMS) to help with this, as well as to control the laboratory workflow. A fully integrated LIMS will track samples and associated test results, use barcoding to speed up data input, associate samples with key information such as batch and supplier data, and create certificates of analysis (CoA) as formal proof of compliance. LIMS can also manage the calibration and maintenance of laboratory instruments, and record which instruments were used for specific tests. By managing and tracking training and competency of staff, LIMS can also ensure both the recording of who carried out the testing and their competency to do the testing. LIMS can also manage the inventory of reagents and consumables used. Additional LIMS Requirements for a Pharmaceutical Laboratory While a typical QC sampling flow monitors and reports on discreet samples, a pharmaceutical manufacturing flow requires a broader range of functionality. A pharmaceutical manufacturing workflow requires the addition of recipe management to associate the raw materials and intermediate products to final product batches; the ability to record test results for each part of the flow (raw materials, intermediate

Batch genealogy, the ability to maintain these relationships between raw materials and final product, and to trace which batches are used in which products, is fundamental to a LIMS for pharmaceutical manufacturing. By way of comparison, a contract laboratory, for example, will receive discrete samples for testing, and provide the individual sample results to the external client. However, maintaining complete traceability of related samples adds significant extra complexity that differentiates LIMS used in manufacturing flows and those used in more general QA laboratories. A stability module is the other key requirement for the pharmaceutical industry. Allowing stability protocols to be defined and managed, including the storage conditions and time periods needed, an integrated stability module will provide the functionality required to meet regulatory requirements for stability and shelf-life studies. Stability studies can be created for each batch of product with the pull schedule, tests, conditions, cycles, and test specifications specific to the study protocol automatically assigned. Operators can be notified when samples need to be taken, and the inventory of product available for testing maintained. The system can produce projected shelf-life analysis, with an associated graphical representation, using the test results obtained from testing each batch within that study. The Cost of Substandard Quality Control Where quality control is not placed at the heart of a business, things can go badly wrong. There are many documented instances of businesses being hurt financially, as well as denting the reputation of their brand, when product has not met expected quality thresholds. Yet businesses seem slow to learn.

Figure 3: Example Stability Shelf-life Report www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 41


Manufacturing/Technology Platforms In the 2019 fiscal year (Oct 18–Sep 19), 81 FDA warning letters1,3 were issued to drugs companies, continuing the upward trend of the previous years; emphasising the increased scrutiny being placed on this aspect of drug safety. FDA warning letters are the official mechanism used to tell manufacturers that rules have been violated. Stability Testing (21CFR211.166)2,3 was mentioned in approximately one in five of these warning letters, mimicking the overall rising trend.

Taking a closer look at recent warning letters in 2020 provides typical quotes that refer to 211.166 Stability Testing violations: •

• •

Your firm failed to establish and follow an adequate written testing programme designed to assess the stability characteristics of drug products and to use results of stability testing to determine appropriate storage conditions and expiration dates (21 CFR 211.166(a)). You lacked an adequate stability programme for your OTC drug products. Your stability programme is inadequate because your procedure does not require a long-term stability study for your OTC drug product. Your firm lacked appropriate stability data to support your firm’s expiration date for your drug products.

problems. However, if issues should arise, the information required to take decisive action and establish the root cause of the problem is readily available. Putting the laboratory at the heart of the pharmaceutical manufacturing process and giving them the authority to drive the quality process gives the business a solid structure to ensure its long-term success and profitability. REFERENCES

Defendable Data Framework A LIMS specifically designed to meet the needs of regulated manufacturing organisations, such as the Matrix Gemini Pharmaceutical LIMS, provides the framework for manufacturers to model their production processes and manage their data and information. This is done in such a way that it is much easier to demonstrate that the stringent requirements of regulators, including the US FDA, UK MHRA and EMA, are met compared to the use of paper-based or spreadsheet-based quality systems. The analytical test data recorded throughout the manufacturing process can be quickly referenced to the delivered final product, as well as back to the initial raw ingredients, ensuring complete traceability. Reagents and consumables used, equipment employed, and the analysts involved in the QA/QC process can be automatically recorded and automatic data capture from laboratory instruments eliminates manual transcription errors. Certificates of analysis can be produced and managed for each batch. The stability module provides the framework for manufacturers to demonstrate they meet the stringent requirements of FDA 21CFR211.166 (Stability Testing). Study protocols are properly defined, and the studies performed according to those protocols. Controls (as defined above) for reagents, consumables, equipment, and staff etc. are in place. The result of stability testing is readily available and accessible to authorised personnel and the integration of stability with the QA/QC process means that stability results can be easily referenced to production data. A LIMS enables the laboratory to manage the quality control data required and ensure the business has robust data management procedures to help prevent product quality 42 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

1.

2.

3.

List of FDA Warning Letters (FDA) https://www.fda.gov/inspectionscompliance-enforcement-and-criminal-investigations/ compliance-actions-and-activities/warning-letters FDA 21CFR211.166 Stability Testing (FDA) https://www.govinfo.gov/ content/pkg/CFR-2019-title21-vol4/pdf/CFR-2019-title21-vol4sec211-166.pdf FDA Warning Letter Analysis (Oct18-Sep19) (ECA) https://www.gmpcompliance.org/gmp-news/batch-release-without-determinationof-identity-and-strength-and-other-gmp-violations-a-look-at-fdaswarning-letters-over-the-la

Tim Daniels Tim has over 30+ years of experience working in national and international markets across a range of software/high-technology products. As worldwide Marketing Manager at Autoscribe Informatics, Tim works with product development, technical services, sales, and management teams across the company to drive marketing goals and achieve growth plans. Autoscribe Informatics provides database management solutions such as laboratory information management systems (LIMS) that, uniquely, are graphically configurable, requiring no scripting or custom coding to configure solutions. Tim’s broad background in marketing and technical roles provides a unique blend of practical knowledge and insight to drive all aspects of product and corporate marketing at Autoscribe Informatics. Email: tim.daniels@autoscribe.co.uk

Winter 2020 Volume 3 Issue 3


Accelerated timelines. Increased regulatory scrutiny. Expanding global needs. To develop life-changing treatments, you need scalable, customizable solutions, like inventory management, chemical and equipment management, scientific services, digital solutions and specialty procurement and sourcing for a full range of consumables. You need a solutions partner with the expertise to work with you through every step of the workflow. Avantor® brings a collaborative approach to solving your challenges in lab services, clinical services and production services so you can focus on science. Get the freedom to do what you do best — make scientific breakthroughs. avantorsciences.com/moves-labservices-forward

www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 43


Manufacturing/Technology Platforms

Key Challenges and Potential Solutions for Optimising Downstream Bioprocessing Production The term “optimise” is often applied to complex manufacturing, automation and business processes and implies that the most efficient function of all the elements of a process – technologies, sequences and procedures – has not been achieved. While no method can be perfect, every process can benefit from careful consideration where new thinking, new techniques and new technology can yield significant improvements. The dramatic growth in the use of biologics across multiple therapeutic applications and categories will only continue to increase. As the demand for these drugs accelerates, there are growing concerns about their cost and availability. Biologics manufacturers are investigating ways to address these concerns – and downstream production in bioprocessing operations is one such area.

Key Challenges in Downstream Production Downstream production currently encompasses about 60 per cent of the total cost of producing a biologic drug. Finding ways to remove bottlenecks and improve yields in downstream could lead to more cost-effective production results, but there are several challenges associated with this goal:

Downstream production, on the other hand, involves multiple steps where the biological material is moved from harvest, centrifugation and polishing to multiple chromatography steps before reaching final fill and finish. Each step requires a unique set of resins and buffers among other materials; storage and production systems at multiple steps; and analytical and quality control sampling activities in parallel. Finding efficiencies and economies of scale across downstream processing steps involves more complex analysis and optimisation. Improvements may be reached after investigating key aspects of current purification steps and technologies, including: •

Expanding the use of mixed-mode and multimode chromatography resins, including resins with targeted ligands with increased selectivity to more efficiently process targeted molecules Exploring ways to make chromatographic buffers more effective by using new kinds of additives, as well as utilising prepackaged single-use buffer materials to streamline buffer exchange steps Making wider use of data analysis tools to develop deeper insight into complex material interactions in downstream process steps, particularly as they relate to raw material characterisation

Increased upstream yields: Significant investments have been made in the technologies and processes used in upstream processes with the goal of improving yields. Efforts to improve raw material characterisation, and to add single-use systems, perfusion systems and more precisely controlled bioreactors, are leading to measurable increases in upstream yields. However, improvements in downstream throughput have not kept a similar pace to those for upstream, leading to potential bottlenecks in the end-to-end process.

Improving Process Chromatography Technology The ultimate goal of downstream optimisation is to improve recovery and therefore normalise, and potentially reduce, the cost per gram of protein produced. That means producing more drug product, using less time, with the same amount of resin and buffer material. One of the most effective ways to do this is by making better use of the newest generation of mixed-mode and multimode resins.

A significant capital investment could be made to create larger chromatography systems to handle the increased production, but it would do little to accomplish the goal of cost-effectively aligning the productivity of downstream production with upstream yields.

Biologic drugs are becoming more diverse, with more complex molecular structures. However, producing these precisely targeted drugs can also yield by-products that are very closely related chemically or biologically to the target molecule, with no therapeutic value.

Loss from upstream to downstream: One of the fundamental structural challenges in biologic production the approximately 30 per cent yields loss as harvest material goes through downstream purification. Any percentage of improvement in downstream recovery can contribute to improving the ultimate process yield for drug product of the target biologic.

Separating undesired glycosylated molecules and aggregates presents major challenges, as they are likely to have limited differential binding to traditional ion exchangers and can coelute. Thus, the combination of increased upstream yields and more complex molecules calls for new approaches to chromatography resins. In response, chemistry suppliers have been focusing their efforts on process chromatography selectivity and efficiency.

Complexity of downstream production: Upstream productivity may be improving because it involves a more straightforward process. Once the target molecule and raw materials are loaded into the bioreactor, the process runs to completion with the appropriate testing and quality control.

The traditional solution to this type of challenge is to utilise multiple downstream ion exchanges. While it yields the targeted drug, the cost per gram of this approach can become prohibitive. More advanced methods of achieving effective selectivity are based on new ligand chemistries engineered to achieve very

44 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Winter 2020 Volume 3 Issue 3


Manufacturing/Technology Platforms precise, selective interaction with the targeted protein. This approach increases selectivity while reducing the required number of steps, helping control process complexity and costs.

to boost chromatography yields while merging two process steps – and the ancillary time and costs associated with each step – ultimately impacting cost per gram.

Targeted affinity chromatographic media are based on ligands tailored to interact with specific proteins, offering high selectivity for a target drug molecule. This can be a time-consuming approach if implemented for every new molecule; instead, mixedmode and multimode approaches offer advantages to be considered.

For example, typical chromatography processes may first use a separate cation exchange step, then an ion exchange step. The yield is about 80 per cent pure after the first step, reaching upwards of 95 per cent after the second step.

Mixed-mode ligand structure: Mixed-mode media offer more interaction possibilities with the targeted drug molecule.

With a multimode resin, it is possible to reach 95 per cent purity in one step. Even with using a multimode resin in the column, it is more efficient to process 70 grams in one batch versus running 100 grams through separate cation and ion exchange steps. This is due to the overall higher throughput and the lower cost of materials, since this approach reduces the buffer consumption, types of filtration systems used, and ancillary costs associated with each chromatographic step. And since each step typically takes up to two hours, costly production time and labour can be cut in half. Another method for optimising process chromatography is using the continuous chromatography method. In continuous multicolumn chromatography, the large column is effectively split into a number of much smaller columns that operate in series over a larger number of cycles. While product is loaded onto some columns, other columns in the set are going through the wash, elution and regeneration phases. Combined with resin optimisation and merging two chromatography steps into one, there is the potential for a three-fold improvement over traditional processes. New Approaches to Buffers Optimising the resin chemistry presents significant opportunities to improve downstream production. Improving the ways buffers are formulated and delivered to the end user can also positively impact productivity.

Multimode ligand structure: Multimode resins have the capacity to simultaneously interact with different sites or regions of the protein molecule.

Mixed-mode chromatography media are based on ligands that offer two or more interaction possibilities with the targeted drug molecule. The mixed-mode approach has proven to be effective and more productive in applications, such as intermediate and polishing steps, for purifying proteins based on differential salt-induced hydrophobicity. The advantage with the mixed-mode approach is that the same media can be used for different purification steps, modulated by solution conditions, such as using multiple buffers or multiple elution steps. However, newer mixed-mode resins have ligand chemistry that enable use of multiple, sequential interactions during the normal chromatographic process. Multimode resins offer greater potential for efficiencies and improved yields. Rather than requiring multiple chromatographic purification steps, the simultaneous interaction makes it possible to separate very closely related proteins in a single step. This means it is possible to have the primary, secondary and tertiary interactions all happening at the same time. This simultaneous purification can occur without requiring additional intermediate steps, such as buffer exchange, buffer titration or dilution. There have been studies showing that using a multimode or mixed-mode ligand with multiple interactions has the potential www.biopharmaceuticalmedia.com

Traditionally, buffers have been very targeted: One type of buffer targets one type of pH in a column, then a different buffer is used to target a different pH. There is a move to more universal buffers that can be used in multiple process steps; while it’s not possible to reuse the buffer, having to acquire, store and manage fewer buffers can help control costs. There is also a greater focus on the use of additives to improve buffer performance. For example, in hydrophobic interaction chromatography (HIC), bioprocessors are working to fine-tune the selectivity of HIC functional groups. One method being explored is to use a select range of additives in the chromatographic media to improve the retention and selectivity of proteins as they move through the media, modulating their hydrophobic interaction and improving separation efficiency with decreased retention time, thereby improving throughput. Taking a more advanced approach to additives is one way buffers can be improved to optimise production. Another innovative advancement in buffer technology is the standardisation of buffer packages, predesigned for specific applications and delivered ready-to-use in single-use packaging. Leading buffer suppliers are now implementing “buffer-ondemand” programmes designed to eliminate buffer-related costs INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 45


Manufacturing/Technology Platforms • • • •

CofA data for all raw material lots manufactured Manufacturing in-process data In-test actuals for conforming specs Stability testing interval data

This approach could enable a biologics manufacturer to more accurately assess and predict the process performance of any given raw material ahead of its use. This is a more holistic and data-driven way to assess the total impact of other downstream optimisation steps, such as adopting more targeted resins or implementing the use of novel additives in buffers.

Avantor-biopharma

in terms of labour, time and capital expenses from downstream production. Buffers are supplied in single-use packages – either pre-weighed and ready-to-use in solution, or as concentrates that can then be diluted and used in the columns. This can eliminate capital expenses associated with buffer preparation tanks and equipment as well as storage space. It also can eliminate multiple buffer preparation, testing and validation production steps, directly impacting labour costs, time and management and documentation activities. Chemistry suppliers who are providing these buffers on demand can work with the manufacturer to recommend and formulate very specific buffer materials, with stringent and well-documented materials characterisation, so that the biologics manufacturer can be assured that the buffer’s performance in the chromatographic step is always on-target. Enhancing Use of Data Analytics Tools One area where the biopharmaceutical industry lags behind other manufacturers is the aggressive use of data and predictive analytics to mine for and uncover ways to improve productivity, process yield and costs. The industry is making significant investments to improve its use of data. Currently, much of the focus is on using process data to optimise a process, then reliably repeat that batch by reaching the perfect balance of process parameters. These efforts are being conducted for both upstream and downstream production; however, much of the focus is on the process data itself, somewhat in isolation. There is an opportunity to expand the application of data analytics beyond the process to more precisely and completely characterise the raw materials used in both upstream and downstream production, then integrate the data into overall optimisation efforts. In many cases, the biologics manufacturing sites are simply using standard certificate of analysis (CofA) data received with the delivery of chromatography resins, buffers and other production materials. Raw material variability presents serious issues impacting downstream efficiency, resulting in long investigations and potential delays in making drugs available for patients. Leading chemistry suppliers are implementing more advanced characterisation efforts that provide manufacturers with further insights into the variability of materials within the integrated supply chain. These include supplying e-datasets, such as: 46 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

“Buffer-on-demand” programmes can eliminate buffer-related costs in terms of labour, time and capital expenses from downstream production. Buffers are supplied in single-use packages – either pre-weighed and ready-to-use in solution, or as concentrates that can be diluted and used in columns.

New Potential for Downstream Optimisation The ultimate goal for downstream optimisation is clear: controlling or even reducing the cost per gram of value biologic drugs. Many of the more expensive elements of current downstream process steps, such as chromatography media, can be utilised more efficiently by investigating how newer, state-of-the-art materials offer ways to condense and streamline process steps. Each downstream process has requirements unique to each target drug. Working with suppliers of downstream materials who have deep insights into how process chemistries and raw materials perform can be a highly productive way to advance downstream biologic optimisation.

Nandu Deorkar Nandu Deorkar, PhD, MBA, is Vice President, Research & Development for Avantor. His expertise in materials technology research & development includes chemical/polymer R&D, drug development, formulation, drug delivery technologies, process development, and technology transfer. Dr Deorkar earned his PhD in chemistry from the Indian Institute of Technology, Bombay, and his MBA from Fairleigh Dickinson University, New Jersey (USA).

Claudia Berron Claudia Berron is the Senior Vice President, Clinical Services for Avantor. Berron’s expertise includes B2B strategic marketing ideation, value proposition strategies, market segmentation, marketing and sales plan, through product launch. Berron holds an MBA from the University of North Carolina, Kenan-Flagler Business School, Chapel Hill, and a BA from the Monterrey Institute of Technology.

Winter 2020 Volume 3 Issue 3


NON-INVASIVE FLOW SENSORS SONOFLOW CO.55 V2.0 NEW

l tro c on

C3S oftw are |c

NON-PRODUCT CONTACT

SONOTEC GmbH sonotec@sonotec.de www.biopharmaceuticalmedia.com www.sonotec.eu

HIGH ACCURACY

on

ct figure | colle

SUSTAINABLE & REUSABLE

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 47


Manufacturing/Technology Platforms

PEER REVIEWED

Building a Gene Therapy: Challenges and Changes in Viral Vector Manufacture In 1970, Freidman and Roblin published a seminal paper in Science proposing that genetic diseases could be cured by using “good” DNA to replace the “defective” DNA causing genetic disease1. This idea – the central tenet of gene therapy – remains sound but has proved unexpectedly difficult to turn into a mainstream therapeutic reality. As of today, the FDA has approved only two gene therapies (as well as one oncolytic virus and 15 cell therapies; eight of which are cord blood suppliers, and four are CAR-T or other cancer immunotherapies). Here, we look back over the challenges the gene therapy industry has faced from viral vector selection through vector engineering to scaling up manufacture and obtaining regulatory approval.

Carrying New DNA into the Host Cell At around the same time that scientists proposed the idea of gene therapies to treat genetic disease, other teams explored the idea of harnessing viral infection as a mechanism to carry new DNA into a mammalian cell2. These first experiments didn’t have gene therapies in mind. The first retroviral vector was designed to facilitate the study of protein-protein interactions, and for overexpression of proteins as commercial or research reagents3. Yet there were only six years between the construction of the first retroviral vector and initiation of a clinical trial using viral vectors to treat two children with severe combined immunodeficiency (ADA- SCID; 4). From Retroviral… Retroviruses were an attractive first choice for viral vectors for gene therapy, because the DNA they encode – either wildtype viral DNA or the therapeutic transgene – becomes integrated into the host cell genome, raising the possibility of long-term expression and persistence of the genetic information, thereby providing stable management or even a cure for genetic disease. However, there are obvious risks to retroviral-based gene therapies – the first being recombination events leading to the reformation of replication competent wildtype viruses, and the second that integration into the host cell genome could cause oncogenesis5. The latter risk is not trivial, as demonstrated by the onset of T-cell leukaemia in two of ten patients treated with a retroviral-based gene therapy for X-linked severe combined immunodeficiency disease6. …to Lentiviral Vectors In recent years, lentiviral vectors – a subtype of retrovirus – have become a more popular 48 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

vector choice. Like all retroviruses, lentiviruses can copy their single-stranded RNA genome into double-stranded DNA to integrate into the host genome. But unlike other retroviruses, lentiviral vectors can transduce non-dividing cells, making them an excellent choice for targeting quiescent cell populations. To reduce the risk of insertional mutagenesis in off-target cell populations and to improve targeting of the virus to the appropriate cell population, lentiviral vectors are frequently used for ex vivo cell therapies. Additional lentivirus is then ‘washed off’ the cells before reinjecting into the patient. A significant amount of development work has also been done to improve vector safety. First-generation plasmid systems included a large part of the HIV genome. The next-generation systems removed additional virulence factors without compromising the transfer of genetic material7. Third-generation plasmids are safer still, because gag-pol (genes encoding the viral core structure and reverse transcriptase) and rev (which encodes a transactivating protein essential for HIV-1 protein expression) are kept in different plasmids. This means an additional recombination event would have to occur to recreate a replication competent lentivirus. Third-generation packaging systems also eliminate tat, a gene that activates viral transcription. This means that when the viral DNA integrates into the host genome, it cannot activate transcription, so even if a recombination event were to occur, the provirus would still not be transcribed. The enhanced safety features of a third-generation lentiviral packaging plasmid system do come at a cost; viral titre is often lower in these systems. To combat this, commercial groups have made additional modifications to improve packaging efficiency, transgene expression and overall viral yield.

Figure 1: An at-a-glance comparison of three frequently used viral vectors and their applications. Winter 2020 Volume 3 Issue 3


Manufacturing/Technology Platforms And from Adenoviral… Adenoviral (AdV) vectors were another popular choice for early gene therapies and are still widely used for certain applications. There have been over 400 clinical trials using adenoviral vectors. These include gene therapy trials, where the adenoviral vector carries a therapeutic transgene and vaccine trials, although most are for cancer treatments8. In this case, the risk of damage to the patient genome is minimal, because these viruses do not integrate their DNA into host chromatin. In fact, these trials demonstrated that adenoviral vectors are – for the most part – safe and well tolerated at mid to low doses9–10; however their strong immunogenicity – an advantage for vaccine development and the delivery of oncolytic viruses – is a major drawback for long-term expression of therapeutic transgenes.

particle packaging, infectivity and how efficiently the vector expresses the gene of interest. These efficiency metrics matter, because they will impact the effective dose of the eventual therapeutic.

...to AAV Vectors Instead, adeno-associated viral (AAV) vectors are often used for the delivery of gene therapies. They have a favourable safety profile – AAV is not known to cause any human pathology – it readily transduces numerous cell and tissue types, and delivers persistent transgene expression without genome integration11. For AAV, there are multiple serotypes with widely divergent tissue tropisms, differing immunologic profiles, and diverse phenotypes with some serotypes (serotype 9) able to cross the blood-brain barrier.

Engineered viral and target genome DNA is introduced into mammalian cells for the purposes of viral vector production by transfection; an inherently variable process. It gives no way to ensure that all cells are successfully transfected with all the plasmids, or that by repeating the same steps twice, you will get the same results. This is a familiar problem for biologists, but problematic as part of a carefully regulated therapeutic manufacturing process.

AAV requires adenoviral help to replicate; consequently much of the work to improve AAV vector systems has concentrated around maximising adenoviral help, while minimising subsequent AdV contamination. The best quality adenoviral help (i.e. that which results in the most infectious particles containing a properly packaged AAV genome) is provided by wild type adenovirus12. However, as well as replicating AAV, the adenovirus itself replicates. In the industrial context, the costs and complexity of downstream purification to remove unwanted adenovirus are considerable. So, although it comes at the cost of packaging efficiency and particle infectivity, three-plasmid transfection has evolved as the preferred method of AAV production. Manufacturing at Scale Transient viral vector production systems have three key ingredients; plasmid, process (transfection) and cell line, all of which can be optimised to improve yields. Plasmids and Process Clever plasmid engineering can increase the number of viral particles recovered from each cell, as well as optimising

Arguably the biggest limiting factors in scalable gene therapy manufacture centre around the plasmid production and transfection steps. Producing GMP plasmids at scale is extremely expensive; and the majority of upstream production costs come down to the cost of producing DNA at such a high standard. In addition, limitations on contract development and manufacturing organisations (CDMO) capacity mean that there may well be a long lead time involved in guaranteeing plasmid supply before a manufacturer can even reach the stage of perfecting their production process.

Choosing a Cell Line and Scaling Up The choice of cell line can also make a difference to production yields. Human embryonic kidney (HEK)293 cells are a common choice for AAV and lentiviral production, because they are easy to transfect and have a relatively short doubling time. However, HEK293 cells are naturally heterogeneous, which means that there will be variation in the efficiency of viral vector production between cells within a parental pool. Choosing a clonal cell line that has been carefully selected for optimal viral production can overcome this issue and increase the overall yield per cell. The most obvious solution to producing more viral vector is to grow more cells. This isn’t a major issue for gene therapies targeting localised areas, or even for systemic treatments with very small patient populations. In either case, the quantities of viral vector required to deliver the treatment are limited. However, results from a successful haemophilia gene therapy trial in 2011 highlighted an urgent need for scalable technologies that would support the manufacture of gene therapies for systemic diseases that require high treatment doses13. The development of systemic gene therapies that required huge numbers of cells to produce sufficient quantities of

Figure 2: Lentiviral producer cell lines have all the genetic elements needed to produce a gene-of-interest expressing lentivirus stably integrated into their genome. www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 49


Manufacturing/Technology Platforms viral vector had serious implications in terms of the physical space required for manufacture. Gene therapy manufacturers are highly dependent on contract development and manufacturing organisations (CDMOs), who have custombuilt GMP (Good Manufacturing Practice) facilities for large-scale cell culture, and often plasmid manufacture as well. This brings its own problems. There are currently over 1000 regenerative medicine companies worldwide, and even more regenerative medicine and advanced therapy clinical trials underway14, so manufacturing demand far exceeds supply. There are a limited number of CDMOs, and these often operate at full capacity, with waiting lists months or even years long15. The move from adherent to suspension cell culture, where cells can be grown in ever larger bioreactors, is going some way towards widening this bottleneck. Not only can CDMOs increase their manufacturing capacity as the square footage required for cell culture decreases, but it becomes increasingly cost-effective and practicable for gene therapy manufacturers to build their own GMP facilities so they can better control their supply chains. Regulatory Compliance When faced with so many challenges in manufacturing a new gene therapy, it’s not surprising that regulatory compliance is another hurdle. Quality standards for gene therapies are understandably and necessarily high, but this can also be problematic given the current manufacturing processes. Early optimisation of production protocols and reagents, as well as using the same genetic system throughout clinical development, can go some way towards addressing this challenge, by promoting data consistency. The choice of cell line used to produce the viral vector for a gene therapy is also an important factor in influencing the regulatory decision. For example, regulators will consider everything from the cell line’s traceability and its previous genetic modifications to the presence or absence of animal components within its cell culture medium. However, in such a rapidly evolving field where patient safety is always the primary consideration, the biggest challenge of all is uncertainty around how to gain regulatory approval. What are the standards a new gene therapy must meet? With a precedent of only two FDA-approved therapies, how does a manufacturer using a new AAV serotype or a new production process go about ensuring that their clinical products meet standards that are still being set? The race to set – and influence – the development of new regulatory standards and guidelines is well underway, as the industry recognises that this challenge will only be overcome through collaboration and cooperation. Technologies of the Future Despite the difficulties, the future for gene therapy is bright. The industry now has a clear collective understanding of the issues it is facing, and there is a concerted effort underway to address these from across the spectrum of academia, biotech and big pharma. New technologies are coming to market that are set to change the way cell and gene therapies are manufactured, with a focus on scalable, cost-effective – and transfection-free – viral vector production. 50 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Producer cell lines are being developed for lentiviral manufacture, where all the genetic elements required for lentiviral production are stably integrated into the host cell line, removing the need for any GMP plasmid in the process. (Figure 2) This is a promising solution for manufacturers ready to embark on commercial manufacture of a cell or gene therapy product. The costs of scaling up production are minimal compared to GMP plasmid manufacture and removing the transfection step eliminates both process variability and complexity. It has proved harder to get to grips with the ins and outs of AAV biology. There have been several attempts to create an AAV producer cell line16–17, but so far it seems likely that to really transform AAV manufacture a novel approach will be required. Such technologies are now entering the market, and we expect to see an impact on AAV manufacturing over the next couple of years. Ultimately, these innovations will help deliver the promise of gene therapies to change the lives of patients with genetic disease. And as cost of goods reduces, process complexity decreases and supply chains shorten, there is good reason to hope that in the near future more of these treatments will reach the market, at a price point that both patients and health services are willing to pay. REFERENCES 1.

Friedmann T & Roblin R (1972).Gene Therapy for Human Genetic Disease? Science, 175(4025), 949–955. 2. Goff SP & Berg P (December 1976). Construction of hybrid viruses containing SV40 and lambda phage DNA segments and their propagation in cultured monkey cells. Cell. 9 (4 PT 2): 695–705. 3. Cepko CL, Roberts BE & Mulligan RC (July 1984). Construction and applications of a highly transmissible murine retrovirus shuttle vector. This Vector is used for entering a cell in the humans cell body. Cell. 37 (3): 1053–1062. 4. Blaese RM, Culver KW, Miller AD, Carter CS, Fleisher T, Clerici M et al. (October 1995).T lymphocyte-directed gene therapy for ADASCID: initial trial results after 4 years.Science.270(5235): 475–480. 5. Anson DS. The use of retroviral vectors for gene therapy – what are the risks? A review of retroviral pathogenesis and its relevance to retroviral vector-mediated gene delivery.Genet Vaccines Ther. 2004;2(1):9. Published 2004 Aug 13. 6. Hacein-Bey-Abina S, von Kalle C, Schmidt M, Le Deist F, Wulffraat N, McIntyre E & Fischer A. (2003). A Serious Adverse Event after Successful Gene Therapy for X-Linked Severe Combined Immunodeficiency. New England Journal of Medicine, 348(3), 255–256. 7. Milone MC & O’Doherty U (2018). Clinical use of lentiviral vectors. Leukemia 32, 1529–1541 8. Wold WS & Toth K. Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Curr Gene Ther. 2013;13(6):421-433. 9. Crystal RG, Harvey BG, Wisnivesky JP et al. Analysis of risk factors for local delivery of low- and intermediate-dose adenovirus gene transfer vectors to individuals with a spectrum of comorbid conditions.Hum Gene Ther.2002;13:65–100. 10. Harvey BG, Maroni J, O’Donoghue KA et al. Safety of local delivery of low- and intermediate-dose adenovirus gene transfer vectors to individuals with a spectrum of morbid conditions.Hum Gene Ther.2002;13:15–63. 11. Daya S & Berns KI (2008). Gene therapy using adeno-associated virus vectors.Clin Microbiol Rev. 2008;21(4):583-593. 12. Zeltner N, Kohlbrenner E, Clément N et al. (2010). Near-perfect infectivity of wild-type AAV as benchmark for infectivity of recombinant AAV vectors. Gene Ther 17, 872–879. https://doi. Winter 2020 Volume 3 Issue 3


Manufacturing/Technology Platforms

org/10.1038/gt.2010.27 13. Nathwani AC, Tuddenham EGD, Rangarajan S, Rosales C, McIntosh J, Linch DC & Davidoff AM (2011). Adenovirus-Associated Virus Vector–Mediated Gene Transfer in Hemophilia B. New England Journal of Medicine, 365(25), 2357–2365. doi:10.1056/ nejmoa1108046 14. Alliance for Regenerative Medicine Sector Report, H1 2020: https:// alliancerm.org/sector-report/h1-2020-report/ 15. Ambler N. Building manufacturing infrastructure for cell and gene therapies. https://www.phacilitate.co.uk/article/buildingmanufacturing-infrastructure-cell-and-gene-therapies 16. Emmerling VV, Holzmann K, Lanz K, Kochanek S & Hörer M (2013). Novel approaches to render stable producer cell lines viable for the commercial manufacturing of rAAV-based gene therapy vectors.BMC Proc. 2013;7(Suppl 6):P12. Published 2013 Dec 4. doi:10.1186/1753-6561-7-S6-P12 17. Zhou J & High KA, Qu G (2010). 673. A Novel Approach to Developing Recombinant AAV Producing Cell Lines. Molecular Therapy, 18, S263. www.biopharmaceuticalmedia.com

Dr. Sophie Lutter Dr. Sophie Lutter is Scientific Communications and Marketing Manager at OXGENE, a biotechnology company that provides technologies and services to accelerate the development of gene therapies from discovery to bio-manufacture. OXGENE’s biomanufacturing solutions include their novel transfection-free TESSA™ technology for AAV production, and lentiviral packaging and producer cell lines. For more information about these, or any other of OXGENE’s technologies, please email business@oxgene.com. Email: sophie.lutter@oxgene.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 51


Regulatory/Quality Compliance

Compliance Capacity: A Checklist to Assess Regulatory Readiness As life science businesses focus on managing the impact of COVID-19 and working on solutions to fight this pandemic, regulatory responsibilities remain a pressing concern that must not fall in priority. In the immediate future, planning for compliance deadlines is a matter of urgency, whether to keep products on the market or meet emerging demands rapidly. In the long term, regulatory and quality compliance raises the bar beyond risk management, by playing a vital role in setting up businesses for their strategic objectives. RA and QA professionals must take the time to thoroughly review their compliance processes, resources and responsibilities, in order to understand how well-positioned they are to meet current and future regulatory obligations.

In addition to these existing regulatory obligations, the temporary measures introduced by regulatory bodies as a result of COVID-19 may be extended if they prove to strengthen efficiency. For instance, it is likely that remote inspections will be used more widely in future, in combination with on-site audits. The time saved by doing desk audits allows auditors to give greater attention to documentation, so businesses can expect closer scrutiny. The need to limit site visits – in the near future, at least – may also give rise to more information-sharing between regulatory organisations, further improving efficiency. Regulatory functions will need to adapt to these changes, and assess how they impact existing operations. Maintaining Product Supply •

An important part of this review is assessing the balance between in-house capabilities and strategic outsourcing relationships. The healthcare crisis has put a spotlight on the challenges of mobilising resources quickly to address urgent requirements. By intelligently using outsourced solution providers, businesses can access the specialist support they need, as and where they need it, providing much-needed agility during this volatile period. Even before the crisis, the life science industry was catching on to the value of leveraging regulatory managed services providers for third-party support. The whole medical device outsourcing market is expected to be worth over €230 billion by 2027.1 As regulatory professionals review their resource needs, they will need to assess preparedness for a number of regulatory obligations, while taking into account new and evolving developments. In this article, we will take a look at key areas of assessment, and provide a checklist of compliance questions that businesses should be asking. Preparing for Increased Regulatory Scrutiny • •

Are you ready for enhanced capability and compliance reporting? Are you ready for enhanced scrutiny of audits?

The medical device and diagnostic industries were already undergoing significant regulatory change before the pandemic. The EU Medical Device Regulation (EU MDR) comes into force in 2021, while the EU In Vitro Diagnostic Regulation (EU IVDR) will be fully implemented in 2022. Both regulations have considerably overhauled existing requirements, and most affected businesses have been dedicating appropriate time and resources to achieve compliance. For their part, the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) have introduced measures to strengthen risk management and supply chain oversight, such as the Drug Supply Chain Security Act (DSCSA) which is now in its second phase of implementation. 52 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

Are you able to assess readiness for production capacity or facility expansion, i.e. rapid, agile scaling? Can you conduct rapid assessment of product qualification Emergency Use Authorization to expedite product release?

An immediate concern for regulators, buyers and suppliers alike is to be able to manage peaks or fluctuations in demand during the course of the pandemic and its aftermath. Regulators have introduced a number of measures so far to prevent shortages. In the European Union, Member States were asked to lift export bans impeding the trade of medicines within the internal market. They were also called to ensure that companies increase production where needed and work at full capacity.2 The European Medicines Agency (EMA) set up the i-SPOC (industry single point of contact) system, to fast-track industry reporting on medicine shortages.3 As in the US,4 extending expiry dates where possible was raised as one solution to ensure optimal use of medicines in hospitals. Short expiration dates force the disposal of drugs even when there may be scientific justification to extend shelf-life. Assessing expiry is therefore a useful tool during public emergencies where shortages are more likely. As well as preserving supply of existing medical products, it has also been important to quickly develop and test new therapies. In the US, Emergency Use Authorizations (EUAs) have seen a spike during the pandemic; as of October 30, 2020, 287 tests were authorised under EUAs,5 compared to just a handful prior to 2020.6 Businesses that wish to take the EUA pathway for a critical medical product must ensure that they have properly understood the submission criteria and process. Producing an EUA submission rapidly is likely to put internal resources under additional pressure, so it will be beneficial to assess whether external support is needed to put together a plan, collate information or write the final submission. A robust submission will position manufacturers well after their EUA expires, contributing to smoother regulatory approval to keep the product on the market. Winter 2020 Volume 3 Issue 3


Regulatory/Quality Compliance De-risking Supply Chains • • •

Supply chain monitoring and reporting: do you have the resources and do they have the capacity? How will you address/resource your supplier development and management programmes? Do you have the capabilities to assess CMO regulatory compliance?

An essential part of preventing medical product shortages is securing the supply chain. The FDA put forward specific proposals in this regard as early as February,7 and the ensuing Coronavirus Aid, Relief, and Economic Security (CARES) Act introduced provisions to address supply chain vulnerabilities. The Act introduces additional reporting requirements in response to drug shortages, and manufacturers of lifesaving drugs must now maintain contingency and redundancy plans to identify risks to drug supply.8 With regard to medical devices, the Act provides the FDA with new capabilities to help prevent or mitigate medical device shortages "during, or in advance of, a public health emergency".9 As a result, manufacturers of certain devices must notify the FDA if manufacturing of a device ceases or is significantly interrupted during a declared public health emergency. In future, the lessons learned from the health crisis are likely to lead to overall tighter scrutiny of the supplier chain, and expectations of strengthened transparency and reporting. Businesses must carry out supplier audits consistently to stay on top of current and emerging requirements, but also to pre-emptively identify where problems could occur. Experienced audit specialists are skilled at identifying issues and suggesting practical solutions, and they should also be able to advise on how to integrate reporting and processes into overall quality programmes. Assessing Supply Networks • •

Supply shortage and trade restrictions: can you effectively assess the implications? Are you prepared for any design or manufacturing transfer activities?

One of the primary concerns in early 2020 was that many US, UK, and EU production and supply capabilities are in China, India and South East Asia. For instance, in 2019, China was the largest global exporter of personal protective products (PPE),10 with a 50% share in US PPE imports11 and 45% in the EU.12 Many businesses will be looking to reduce this dependency by expanding their manufacturing networks and broadening their geographic footprint. This will increase complexity for regulatory professionals, who will need to address the compliance aspects of manufacturing products in alternative or even additional geographies. Specifically, there may be an immediate impact on critical design and validation activities, so compliance in these areas must be reviewed before manufacturing can take place in other locations. Medical device manufacturers working towards EU MDR compliance will already have made some headway in assessing their supply networks. For the first time, the regulation makes manufacturers, authorised representatives and importers jointly and severally liable for device compliance. Along with distributors, these entities are collectively termed economic www.biopharmaceuticalmedia.com

operators, and are subject to new regulatory requirements. To ensure that their devices can remain on the market following the EU MDR date of application on May 26, 2021, manufacturers must verify that each of their economic operators is prepared for implementation. This is no small task, especially for larger businesses with a footprint in multiple countries. An external partner with a sound understanding of the requirements will be able to quickly identify the information that is needed from each entity, and help to implement clear processes for continual monitoring of economic operators. Complying with EU MDR & EU IVDR •

EU IVDR transition programme: what is your capability, taking into consideration post-pandemic regulatory demands? IVD and CDx platform expansion: how will you manage it?

The extension of the EU MDR deadline to May 2021 has afforded manufacturers additional time to bring their processes and documentation up to speed for compliance. Pharmaceutical and biopharmaceutical manufacturers should note that they may also be impacted by the EU MDR if: they manufacture medical devices, such as devices with an ancillary medicinal substance; partner with or supply to companies that manufacture medical devices; or manufacture a drug that is sold pre-packaged in a delivery device. Since pharmaceutical businesses may not have prior experience with medical device regulation, they must take advantage of the deadline delay to assess whether their products fall under the remit of the EU MDR, and plan how to achieve compliance. As well as economic operator relationships and agreements, manufacturers will need to thoroughly review their clinical evaluations, technical file documentation, and post market surveillance documentation. Since the EU MDR is still relatively new to the industry, and introduces wide-reaching changes, there is a need for greater guidance on how to meet the requirements. Suppliers that have a proven record in assisting organisations with EU MDR compliance should be able to provide an effective industry benchmark based on their experience, and informed advice based on feedback from notified bodies for submissions they have worked on. This adds immediate value to compliance activities, since businesses may otherwise have to rely on their own rationale for what a good quality submission looks like. At the time of writing, there are no plans to extend the transition period for the EU In Vitro Diagnostic Regulation, ending in May 2022. Affected businesses must therefore continue their EU IVDR compliance activities, though they may need additional support to simultaneously manage the impact of the pandemic. External partners working in this regulatory area can help to make sense of the requirements and efficiently identify gaps in existing product data and documentation, where additional work is needed to achieve compliance. As the EU IVDR includes revised definitions for in vitro diagnostic devices, as well as new risk-based classifications, many IVDs will be under regulatory review for the first time. Pharmaceutical manufacturers should also note that the EU IVDR introduces a definition for companion INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 53


Regulatory/Quality Compliance

diagnostics, referring to IVD devices used to identify patients most likely to benefit from a corresponding medicinal product, and patients likely to be at increased risk of serious adverse reactions. Making Strategic Use of Outsourced Support Listed above is a selection of critical questions that regulatory and quality professionals are evaluating in order to prepare for the months and years ahead, building a clearer picture of how they can manage regulatory requirements on the horizon. Although the use of resources on an individual or group basis has been utilised extensively to date, the time has come to look at utilising alternative solutions for addressing the short- and long-term regulatory and quality needs in the medical device and diagnostic industries. Re-thinking outsourcing solutions will make more efficient use of limited internal resources and leverage specialist support in a manner that will realise strategic assistance in achieving goals and improving efficiency and effectiveness in internal processes. As circumstances continue to evolve, and as healthcare systems enhance their response capabilities, access to third-party support will be invaluable to remain flexible and adapt to new demands. Slow responses could be detrimental, prolonging time to market or necessitating corrective action later down the line. Looking further ahead, the ability to leverage strategic outsourced solutions will help businesses to stay one step ahead of their competitors. REFERENCES 1. 2.

3. 4.

Medium, Covid-19 impact: What is the Medical Device Outsourcing Market size? 13 May 2020 European Commission, COMMUNICATION FROM THE COMMISSION: Guidelines on the optimal and rational supply of medicines to avoid shortages during the COVID-19 outbreak, 08 April 2020 https://ec.europa.eu/info/sites/ info/files/communication-commission-guidelines-optimalrational-supply-medicines-avoid.pdf EMA, EU authorities agree new measures to support availability of medicines used in the COVID-19 pandemic, 6 Apr 2020 FDA, Coronavirus (COVID-19) Supply Chain Update, 27 Feb 2020

54 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

5.

FDA, Coronavirus (COVID-19) Update: October 30, 2020 https:// www.fda.gov/news-events/press-announcements/coronaviruscovid-19-update-october-30-2020#:~:text=As%20of%20 today%2C%20287%20tests,tests%2C%20and%207%20 antigen%20tests. 6. FDA, Emergency Use Authorization: Emergency Use Authorization (EUA) information, and list of all current EUAs https://www.fda.gov/emergency-preparedness-and-response/ mcm-legal-regulatory-and-policy-framework/emergency-useauthorization#othercurrenteuas 7. FDA, Coronavirus (COVID-19) Supply Chain Update, 27 Feb 2020 8. White & Case, Impact of the CARES Act on the Pharmaceutical and Medical Devices Industries, March 2020 https://www.whitecase. com/publications/alert/impact-cares-act-pharmaceutical-andmedical-devices-industries 9. FDA, Medical Device Shortages During the COVID-19 Public Health Emergency, 20 August 2020 10. Statista, Top personal protective products exporting nations worldwide in 2019, 02 June 2020 11. Statista, Chinese share among selected U.S. imports of medical supplies and equipment in 2019, 20 July 2020 12. European Parliament, EU imports and exports of medical equipment, October 2020

Steve Cottrell Stephen Cottrell is the President of Maetrics and is responsible for client service delivery, growth, and overall performance of the company. He leads an experienced team of industry professionals who formulate and deliver consulting services dedicated to the life sciences industry. Steve’s experience encompasses leading a wide array of business services within the life sciences sector, including business process outsourcing, strategic sourcing, and clinical trial offerings. Steve is a business executive with more than 25 years of experience in the life sciences industry. His career has been dedicated to supporting services in the medical device, pharmaceutical, and biotech market segments. He has a proven ability to evaluate client challenges and bring together unique solutions to meet the current and future needs of clients.

Winter 2020 Volume 3 Issue 3


YOUR BRIDGE BETWEEN LIFE-CHANGING THERAPIES AND PATIENTS

Introducing

Accelerate Into Early Phase Trials OUR SPEED TO STUDYTM SERVICES HELP YOU TO:

• Shorten timelines to proof of concept • Maximize dosage flexibility from cohort to cohort • Reduce API and drug waste significantly

Accelerate your Phase I / II studies with PCI services www.pciservices.com/speed-to-study

www.pci.com www.biopharmaceuticalmedia.com

talkfuture@pciservices.com INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 55


Supply Chain Management

Hydrogel Encapsulation – The New Paradigm for Ambient Temperature Cell Shipping Deep-freezing has become the standard practice for long-term preservation of biological samples, especially for isolated and cultured cells, and its use has been extended to the shipping of cells from point of manufacture to the place of use. Currently, there are different cryopreservation techniques available depending on the cell type, cryopreservants used or culturing conditions practised, but they all aim to reach the same outcome: similar levels of post-thaw cell viability and metabolic activity, when the thawed cells are compared to the original freshly produced material. Cryopreservation has become an art, whereby the scientist tries to achieve a balance between concentration of the cryopreservant used and the temperature cooling rate, so that there is minimal ice crystal formation within the cells, reducing overall cell loss, yet maintaining cell viability. This is a fine balance as high concentration of cryoprotectants becomes cytotoxic, while fast-rate freezing protocols can easily dehydrate cells. Neither scenario is, therefore, ideal for post-thaw cell recovery. Additionally, when shipping cells, the temperature needs to be maintained at a deep-freeze level throughout the cells’ journey, which involves the use of a cooling agent (like dry ice or liquid nitrogen), polystyrene package or cryogenic storage dewars, materials that add to the overall shipping cost, environmental impact and the complexity of the logistical journey.

developments are being limited by conventional freezing protocols, which is hindering the adoption and use of emerging cell-based applications, like those derived from stem cells. As the stem cell therapy market is growing rapidly, stem cells can be used for different cellular applications, like fresh stem cell treatments, clinical trials or long-term cryopreservation of umbilical cord and blood samples, the successful shipment of these therapies becomes a rate-limiting step in the manufacturing process. It should be noted that stem cell banking centres are thriving and estimated to become a billiondollar value market by 20235. Logistics Challenges of Cell-based Therapies The provision of cells as a research or therapeutic tool is a rapidly growing business, which forms a cornerstone of the life science industry. Cells are not just the fundamental building blocks of an organism, but also the fundamental component when used clinically as a therapeutic, as a disease model for drug discovery, as the product sold by a biosupplier and as a tool in blue-sky research. This diversity in use means that there isn’t such a thing as “one cryopreservation approach fits all”, and each application requires its own approach for cost-effective and reliable cold-chain supply of temperaturesensitive cell-based products. This means development of cryopreservation solutions for a wide applications portfolio: • •

The Ugly Reality of Freeze-thawing Cells More than just cell viability, it is important to understand if deep-freezing can impact cell functionality. Will thawed immune cells retain their activation phenotype? Will thawed mesenchymal stem cells lose their immunomodulatory profile? More and more research papers have been published highlighting how cryopreservation is impacting the cell’s phenotype and its functionality. Among different cell types, results showed: • • •

Thawed regulatory T cells can show reduced immunosuppressive potential1 Decreased cytokine expression from thawed mononuclear blood cells2 Thawed mesenchymal stem cells are less able to successfully graft on the injury site3 and to promote cell growth/ regeneration4

The fact that thawed cells can have a decreased therapeutic profile is alarming for cell-based clinical therapeutic applications. Pharmaceutical companies invest heavily in development of new clinical applications, so cryopreservation of cell-based therapeutic applications needs to provide a homeostatic microenvironment for cells to survive freezethaw cycles. Consequently, new cell culture technological 56 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

• •

patient-tailored medicine – cell therapies or blood-derived treatments cell-based products – plated cells, organoids or 3D tissueengineered products clinical sample processing (like tissues biopsies and bodily fluids) specialised applications – ready-to-use cell-based assays for toxicology testing, genomic studies.

Therefore, specialised cryopreservation logistics need to be established for each market segment and/or cell application, always trying to guarantee cell viability after freeze-thaw. The cell therapy clinical trials market is, undoubtedly, the major driving force for improvement of cold-chain supply logistics for temperature-sensitive cell products as clinical trials-specific cold-chain logistics is estimated to reach more than $3 billion by 20226. This is because the current logistics around fresh autologous immunotherapies (patient-matched engineered immune cells) is challenging. Patient-tailored cell therapies are produced in small batches, with transfer of valuable biological material from the patient into the manufacturer’s facility for engineering and back into the patient from a matter of hours to days. With this, two problems arise: a) cryopreservation can be too harmful for preservation of vulnerable engineered immune cells like regulatory T cells or Chimeric Antigen Receptor T-cells (CAR T-cells) as it can alter the immunophenotypic profile of the cells (discussed above); b) cell therapy treatments are geographically planned so that the Winter 2020 Volume 3 Issue 3


Supply Chain Management cell therapy manufacturer is in in close proximity to the clinical setting. This enables the cell therapy to be administered back to the patient as fast and fresh as possible. However, this is not always the case as globalisation of clinical trials will mean that multiple clinical sites often receive cryopreserved cell therapy products from a single remote manufacturing site. With globalisation comes wider access to clinical treatments, meaning that the supply chain will need to reach remote clinical sites, cross different climatic zones or enter different chain of custody regulations. This puts pressure on the cold-chain logistics services to deliver these high-value cell therapy products in a time-constrained, reliable, and cost-effective manner. Shipping of cryopreserved biological samples is very seldom cost-effective, as couriers will charge heavily for cryopreserved shipping services - package handling, storing, weight, chain-ofcustody documentation, coolant top-up for possible customs clearance delay and expedited delivery times. On top of that, and to compensate for the high risk of post-thaw cell death, manufacturers are choosing to overload the final cryopreserved cell therapy product with higher cell concentrations. Altogether, this imposes a high financial burden as it requires a higher production of therapy-targeted cells and expensive cold-chain logistics, which will impact the manufacturer’s revenue for that therapy. The latest data from IQVIA Institute for Human Data Science estimates that the biopharma industry loses approximately $35 billion every year on failed temperature-controlled logistics. Sadly, this estimate does not consider the impact that failed cell therapy logistics have on the patient’s quality of life and the added value for a successful treatment. New cost-effective cold-chain shipping logistics with minimal impact on cell viability are therefore required. How to Maintain Cell Homeostasis During Shipping Cells are always shipped in a temperature-controlled manner, either frozen (cryopreserved) or at a low temperature (4–8°C). This depends either on the cell type or if the cell culturing conditions allow for deep-freeze transport without compromising cell viability and functionality. Many cells are, therefore, shipped at a cool temperature with liquid medium culture on a race against time, in the hope that they will arrive at their destination still within the required temperature range. The most obvious issues one can think of from shipping cells in liquid medium culture is the likelihood of leaks or contaminations from the container, the nutrient deficiency from the continuous cell metabolic activity and build-up of metabolites, or the constant mechanical stress cells are under from the liquid shaking. As a mitigative solution, stabilising agents are often added to the cell culture before shipping as these additives have anti-bacterial and anti-fungal properties to avoid contaminations, while also protecting the biological sample’s DNA and general cell integrity during transport. Depending on the application, scientists may need to go a step further and fixate cells with alcohol or methanol protocols before shipping to ensure cells maintain their phenotype and functionality during shipping. This is often the case for single cell sequencing applications. Addition of fixation preservants will alter the transcriptomics profile of the fixed cells, leading www.biopharmaceuticalmedia.com

to overall skewed results. This is a current problem for transcriptomic users, where they often must compromise on sample quality by resourcing to fixation protocol when shipping samples for analysis. How is it then possible to maintain cell homeostasis during shipping without resourcing to additives or worse, fixation preservants that ultimately halt cell viability? Hydrogel-based Encapsulation – A Safe Solution for Cells Hydrogel-based cell encapsulation provides a safe extracellular microenvironment and protects the cell from mechanical stress during shipping. The applications of hydrogel-based encapsulation are not just limited to cellular therapies, as many other cell-based applications or products require complex shipping logistics – induced pluripotent stem cell (iPSC)-derived cells (like cortical neurons) or multicellular bio-engineered models. Many of these cell-based applications are currently developed in a “ready-to-use” as they are targeted for drug discovery, research tools, toxicology screening or diagnostics use. The Advantage of Hydrogel Encapsulation for Drug Discovery Cell-based assay developments for drug discovery is the major current revenue stream. Drug discovery-specific cell assays are commonly marketed as fully functional human-based plated cells for ready-to-use testing, like iPSC-derived cardiomyocytes or cortical neurons. Drug developers can take advantage of these cell-based assays for high-throughput drug screening at early development stages, identifying the most promising leads for further testing. This ultimately accelerates the drug development pipeline as drugs are developed, screened, safety tested and commercialised much faster. Any biosupplier that distributes iPSC-derived cells for drug screening service providers knows how challenging iPSC-derived cells are to ship, as they need to retain their embryonic-like pluripotent state. During shipping, cells can easily be exposed to uncontrolled mechanical (shaking) and temperature stress which is enough to initiate their differentiation cascade. This means that iPSC-derived cells will lose their pluripotency profile at arrival, no longer being suitable for the end user. Biosuppliers often ship plated partially differentiated progenitor cells instead and the end user is required to follow complicated and expensive cell maturation protocols until cells are fully differentiated. Leaving this responsibility to the end user is risky because it eliminates any manufacturer’s quality control, assumes that the end user has adequate cell culturing knowledge and in-house facilities, and can ultimately compromise the testing experimental outcome if cells are not correctly differentiated. Therefore, manufacturers are constantly searching for improved cold-chain solutions for shipment of finalised cells for testing. Would hydrogel encapsulation work for commercial distribution of iPSC-derived cells for drug discovery? Alginate gel would, in fact, prove to be a great solution as cells would be encapsulated as adherent cells, with finalised quality control tests. This way, the manufacturer would have absolute guarantee that the cells’ phenotypic profile remains unchanged before and after shipping. At the site of arrival, cells would be ready to use after release from the hydrogel (Figure 1). INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 57


Supply Chain Management

Figure 1. Hydrogel encapsulation – overview of its applications and how it could substitute cryopreservation for short-term storage and shipping of cell products. Illustration by Dean Hallam: dean.hallam@atelerix.co.uk

How Can Hydrogel Improve Stability of Ready-to-use Cell Products? Advancement of human-relevant cell culture research from in vitro 2D to 3D combined with increased regulatory and governmental efforts to replace in vivo animal studies with in vitro cell-based alternatives for safety testing, boosted the need for ready-to-use cell-based products. The range of cell-based products can go from cell-based assays (monolayer culture) to organoids, spheroids, reconstructed 3D tissue models (i.e. skin, liver, heart, brain). Regardless of the product, resemblance to the original in vivo microenvironment is vital. Therefore, development of these tissue-engineered products is often an expensive and time-consuming process for companies. The finalised cellular construct requires a well-defined combination of cells (single or multiple cell types), biocompatible scaffolds and molecules (proteins, cytokines, growth factors…) to promote formation of the tissue with in vivo cellular resemblance. While this is a challenge on its own, commercialised products also need to withstand shipping and long-term stability. Combine this with high-throughput screening (assay ready) and large-scale batch production, and you end up with stringent requirements for cell stability and preservation. As the cell products get more complex (combination of different cell types, cellular arrangement and size), it can be harder to guarantee its stabilisation throughout shipping. Cryopreservation would not be the most suitable option for different reasons, with the most obvious one being that different cells have different cryopreservation requirements 58 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

and freezing rates, so it would be difficult to establish one cryopreservation protocol suitable for all cell types included in the cell model. Furthermore, this could lead to ice crystal formation, which would not only damage the cells itself but also affect the cellular arrangement of any tissue-engineered 3D model. Ultimately, cryopreservation cannot compete with hydrogel preservation for shipment of ready-to-use cell products as it presents a higher risk of cell damage and higher shipping costs (temperature-controlled shipping versus ambient shipping). Altogether, any provider of ready-to-use cell products should consider hydrogel encapsulation as the most cost-effective solution. Thinking on a smaller scale, academic collaborations often require transport of biological materials back and forth between labs at either a local or global distance. Shipping of cryopreserved cell-based and tissue-engineered products between collaborators can be quite expensive to manage on low-funded project grants. Cost-effective solutions like hydrogel encapsulation not only provide a more affordable option, but on a higher level, facilitate scientific collaboration, inter-laboratory validation of results and protocols, share of knowledge and ultimately, help move science forward. Scientific progress is the driving force behind technological development, fuelled by the need for better, faster, more precise human-tailored treatments. Therefore, any scientific progress needs to be followed and matched by companion technologies to fully utilise breakthrough discoveries. Recent developments Winter 2020 Volume 3 Issue 3


Supply Chain Management across different markets – drug discovery, cell therapy or tissue engineering – suggest that cryopreservation has failed to keep up with the latest cell-based developments. Cryopreservation is an outdated solution that can jeopardise the technological development invested in these applications and should soon be replaced with safer, more cost-effective and standardised solutions like hydrogel encapsulation for short-term storage and ambient shipping of biological material. REFERENCES 1.

2.

3.

4.

5.

Florek M, Schneidawind D, Pierini A, Baker J, Armstrong R, Pan Y, et al. Freeze and Thaw of CD4+CD25+Foxp3+ Regulatory T Cells Results in Loss of CD62L Expression and a Reduced Capacity to Protect against Graft-versus-Host Disease. PLoS ONE. 10, 1-8 (2015) Kvarnström M, Jenmalm MC, Ekerfelt C. Effect of cryopreservation on expression of Th1 and Th2 cytokines in blood mononuclear cells from patients with different cytokine profiles, analysed with three common assays: an overall decrease of interleukin-4. Cryobiology, 157-68 (2004) Chinnadurai R, Garcia MA, Sakurai Y, Lam WA, Kirk AD, Galipeau J, Copland IB. Actin cytoskeletal disruption following cryopreservation alters the biodistribution of human mesenchymal stromal cells in vivo. Stem Cell Reports. 3, 60-72 (2014) Otsuru S, Hofmann TJ, Raman P, Olson TS, Guess AJ, Dominici M, Horwitz EM. Genomic and functional comparison of mesenchymal stromal cells prepared using two isolation methods. Cytotherapy. 17, 262-70. (2015) Stem Cell Banking Market by Cell Type (Umbilical Cord Stem Cell [Cord Blood, Cord Tissue, and Placenta], Adult Stem Cell, and Embryonic Stem Cell), Bank Type (Public and Private), Service Type (Collection

6.

& Transportation, Processing, Analysis, and Storage), and Utilization (Used and Unused) - Global Opportunity Analysis and Industry Forecast (2017-2023) 2019 Biopharma Cold Chain Logistics Survey: What Matters Most – and What it Means for the Future, Pelican BioThermal

Ana Ribeiro Ana Ribeiro is the Scientific Sales Specialist at Atelerix, a biotech company from Newcastle specialising in room-temperature storage of cells and tissues. Ana’s expertise includes cell and tissue culture, immunology, and cell-based applications. Ana’s role within the company is to provide scientific sales expertise, brand awareness and lead generation. Email: ana.ribeiro@atelerix.co.uk

ADVERT

PATIENTVIEW HAS LAUNCHED #PAGC19 ON TWITTER AND IS ENCOURAGING PATIENT GROUPS TO ADOPT THE HASHTAG IN ANY OF THEIR TWEETS ABOUT COVID-19 HOW DOES IT WORK? All tweets containing #PAGC19 are automatically picked up and pooled on the website: https://patientviewblog.com. The #PAGC19 tweets are categorised on the website according to disease area – so that patients can quickly find tweets relevant to their own medical condition. Importantly, the source of every tweets included on the website is easily identifiable (via the Twitter username that begins with @), so that patients can be sure who is actually providing the information and support. Patients are having difficulty finding healthcare information on Covid-19 that is both relevant to their needs, and trustworthy. Fortunately, patient groups comprise a powerful and responsible source of tailored support and information for people living with a disease condition.

The website also files patient-group-recommended videos of experts pointing out solutions to the problem of living with a medical condition during the pandemic. The aim, during this difficult time, is that #PAGC19 and https://patientviewblog.com become a hub that allows patients with a medical condition to access information from patient groups on how Covid-19 affects their specific medical needs. For further information email: report@patient-view.com.


Supply Chain Management

Driving Demand for Parenteral Packaging: How Biologics are Changing the Face of Pharma Packaging? The biopharmaceutical sector has enjoyed robust and sustained growth in recent years. A recent BioPlan Associates report estimates worldwide sales of biopharma treatments reached more than $300 billion this year (2020), increasing at an annual rate of 12%1. But what is behind this growth, and what impact is this booming sector having on the field of pharmaceutical packaging?

We spoke to Marcelo Cruz, Director Business Development and Marketing at Tjoapack, to understand more about the evolving biopharma landscape and the rising demand for packaging solutions for parenteral products to explore the future for the sector. The Boom in Biologics Biologics are fast establishing themselves as a key engine of growth in an already booming pharmaceutical sector. This category of medicine is playing a significant role in the growth of the global industry as a whole, which is forecast to reach a value of $1.5 trillion by 2023. A major reason for their success in recent years is the growing use of monoclonal antibodies for use in treatments to fight chronic diseases, such as diabetes, cancer and auto-immune conditions. Diagnoses of these conditions are on the rise worldwide, driven by a combination of ageing populations, poor diet, and environmental pollution. This surge in biopharma demand is having an impact on other parts of the pharmaceutical supply chain, particularly on drug delivery and packaging requirements. One key effect has been the resulting growth in the use of parenteral drug delivery – specifically self-administered injectable formulations. Effective Parenteral Delivery For many of these biologic treatments, the only effective delivery mechanism is parenteral administration, such as injection. Given the boom in biopharma treatments and their parenteral delivery requirements, it is no surprise that the injectables market is expected to grow considerably in turn – in fact MarketsandMarkets forecasts it to reach $624.5 billion by 2021, having nearly doubled in size in five years2. At the same time as ensuring new biologic treatments use the best administration method for their needs, manufacturers need to be conscious of making sure their biologic treatments are as patient-centric as possible to maximise patient access and to optimise dose compliance. Ease-of-use for patients is a pressing issue for drug developers, particularly when it comes to parenteral drugs 60 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

which are traditionally unpleasant to administer. Patients need to be able to administer the drugs themselves easily. Not only does this free up clinicians’ precious time and resources, but can go a long way towards ensuring that all patients take their treatment, at the right frequency and dose. This is vital to maximise the effectiveness of the treatment and, in doing so, can relieve pressure on healthcare systems by minimising the need for chronic patients to be seen by doctors. With this in mind, it is no surprise that there has been considerable innovation in the parenteral space in recent years to achieve this useability goal. A number of pharma companies have been investing time and funding in developing new technologies to support easier self-administered injections, particular in a non-clinical setting, such as a patient’s home, school or workspace. The most significant innovation in this area has been the rise of prefilled parenteral drugs, such as prefilled syringes, which in turn is driving demand for packaging partners that can meet the increasingly complex requirements of these products. Overcoming Prefilled Packaging Obstacles The new generation of prefilled injectable biologics is already transforming the pharmaceutical market. This diversity of injectable design is positive news for the market. However, the growth and diversification of prefilled injectable technologies to support biologic drugs highlights the vital role played by packaging and delivery solutions in creating effective and successful treatments and changing the lives of patients. Manufacturers now face a number of challenges when producing prefilled injectable solutions. In the case of prefilled syringes, for instance, it is vital to maintain a clean, sterile environment while packaging them to safeguard the health of users. With this in mind, manufacturers need production line solutions capable of maintaining the highest sanitary standards throughout the manufacturing process. In addition, the variety of potential components associated with prefilled injectables brings an extra layer of complexity to both the primary and secondary packaging process. The variety of requirements are far more varied than those of other dosage forms. For example, depending on the treatment, the syringe may be accompanied by a staked needle, or a loose needle, a safety device which may or may not include a finger flange or just a backstop. The only components that are always present regardless of the syringe are the plunger rod and label. Moreover, it is common to include additional inserts, such as Winter 2020 Volume 3 Issue 3


Supply Chain Management

alcohol pads or multiple leaflets and booklets in the secondary packaging, meaning expertise and flexibility is essential to ensure product safety and integrity. In addition, extra safety procedures must be observed when it comes to the handling of prefilled biologic treatments throughout the production process. The products involved are often more toxic than those in other dosage media, so it is vital that there are good spill responses in place in the event of a breakage, to protect line operatives and minimise the impact of the spillage on neighbouring products.

prefilled syringes is leading to a change in the way pharma companies outsource their drug manufacturing. In the past, packaging was seen as just one relatively minor part of the wider manufacturing process. This meant that it was often outsourced to contract development and manufacturing organisations (CDMOs) as part of their wider end-to-end development offering. Now, though, these CDMOs lack the capacity and specialised processing capabilities needed to cater for the complicated packaging required by prefilled injectable drugs.

Moreover, the product must be kept at the appropriate temperature to maintain its efficacy. If these steps aren’t taken, the batches could be unsafe and inappropriate for use by patients.

Increasingly, expert contract packaging organisations (CPOs) are stepping in to fill the gap, playing a greater role in the pharmaceutical supply chain as a means of providing cost- and time-efficient packaging services for injectable drugs of all shapes and sizes.

Achieving Packaging Efficiency The challenge presented by the complex packaging needs of

The primary reason for this is that CPOs offer in-depth specialist knowledge of both the needs of the market, and

www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 61


Supply Chain Management the regulatory landscape, which means they are best placed to develop a tailored, more effective packaging service to customers. This knowledge and expertise add significant value to their contract partner that benefits their business. In the case of prefilled injectable biologic treatments, whatever the product’s specific packaging needs, CPOs are well placed to provide packaging services, offering a solutionfocused approach to maximise efficiency and output while maintaining product quality. In doing so, they are offering more than a transactional contract service, they are establishing themselves as an integral strategic supply chain partner. Ensuring Supply Chain Integrity In addition to supporting pharmaceutical companies in overcoming the complexities of scaling up injectables packaging, CPOs are able to provide other benefits that add further value both for their customer and for patients, as well. Increasingly, parenteral therapeutics – and biologics in particular – require special handling and strict chain-of-custody integrity during transit from the manufacturing facility all the way to the point where the patient takes possession. Such a step is vital to protect the global drug supply from counterfeit medication – a growing problem in a large and competitive international marketplace. The World Health Organisation (WHO) estimated that, last year, the prevalence of falsified drugs ranged from less than 1% of sales in developed countries to more than 10% in developing countries, with some regions having more than 30% of counterfeit medicines on sale. These false products not only cause the loss of legitimate sales and revenues for genuine pharmaceutical companies, they pose a serious risk to public health as well. According to PWC, counterfeit medicines cause around 450,000 preventable malaria deaths per annum, with up to half of treatments for sale on the internet being counterfeit. The EU’s Falsified Medicines Directive, which came into force in 2019, and the US FDA’s Drug Supply Chain Safety Act 2013 contain requirements to tackle this issue, and demand companies print unique identifiers and serial codes on drug packaging. This is designed to verify the product within is genuine and to track each unit through the supply chain. To comply with this, prefilled drug products and similar require dedicated and complex logistical solutions, as well as specialist labelling to support traceability throughout the product’s journey. CPOs are able to tailor their offering to support drug companies in managing these necessarily complex traceability needs for injectable products to comply with global and local legislation. From vial labelling, re-labelling and over-labelling, to serialisation for single and multipacks, to inventory control, CPOs can help customers in ensuring each and every one of their injectable products can be tracked throughout the transport process. They can also support in providing cold-chain packaging and storage, helping to maintain the product’s shelf-life. Adding Patient-centric Value In addition to all of this, CPOs can support pharmaceutical 62 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

companies in delivering injectable products containing the additional components often required by this kind of solution. Those that provide a “kitting service” can supply packaged injectable drugs that feature not just the information leaflets, but additional items, such as swabs, disposable needles and bandages – all the accoutrements a patient might need to administer their dose safely and comfortably. Advanced CPOs are able to provide this service at scale, and for multiple markets as well – supplying leaflets and other items that comply with local language and regulatory requirements. By offering this additional support, CPOs are able not only to add value to biopharmaceutical companies – enhancing operational efficiency and maximising product quality – but to patients as well. They are able to help deliver more patientcentric solutions that make it easy for users to administer their doses on schedule, boosting compliance. A Bright Future for Biologics Biopharma treatments offer considerable potential in the fight against cancer and other chronic diseases, so it is no surprise that the sector is on track to grow for the foreseeable future. However, ensuring the right packaging solutions is essential to ensuring safe, patient-friendly medication. Access to the right regulatory expertise is also important to ensuring a smooth process to market for relatively complex products. It is imperative that companies work closely with experts. Collaborating with specialist CPOs as strategic outsourcing partners can help companies meet this goal. Not only will they benefit from their expertise in managing the complexities of parenteral biologic drugs, they can also enjoy access to innovative supply chain management approaches and years of experience. With all of this, biopharma companies can ensure they are able to deliver for their patients, time and again, when and where their drugs are needed. REFERENCES 1.

2.

https://www.manufacturingchemist.com/news/article_page/ Trends_and_opportunities_in_biopharmaceutical_product_ development_and_manufacturing/170557 https://www.marketsandmarkets.com/Market-Reports/injectabledrug-delivery-market-150.html

Marcelo Cruz Marcelo Cruz is Director Business Development and Marketing at Tjoapack. With over a decade of experience in the pharmaceutical industry, and over 15 years of driving global strategic marketing and sales, Marcelo is responsible forthe overall Business Development strategy and organic growth activitiesat Tjoapack. In his role he also leads the development and implementation of inbound and outbound marketing strategies to accelerate lead generation and drive the wider commercial strategyfor the business.

Winter 2020 Volume 3 Issue 3


INSIGHT / KNOWLEDGE / FORESIGHT

SUPER PUBLICATIONS FOR SUPER PHARMACEUTICALS IPI

Peer Reviewed, IPI looks into the best practice in outsourcing management for the Pharmaceutical and Bio Pharmaceutical industry.

www.ipimediaworld.com

JCS

Peer Reviewed, JCS provides you with the best practice guidelines for conducting global Clinical Trials. JCS is the specialist journal providing you with relevant articles which will help you to navigate emerging markets.

PHARMA’S DNA

www.jforcs.com

Listen to industry experts on the latest in drug discovery, development, research, industry regulations and much more at Pharma,s DNA, the podcast channel by Pharma Publications, available on Sound Cloud, Spotify, iTunes and YouTube.

IAHJ

IBI

Peer Reviewed, IAHJ looks into the entire outsourcing management of the Veterinary Drug, Veterinary Devices & Animal Food Development Industry.

www.animalhealthmedia.com

Peer reviewed, IBI provides the biopharmaceutical industry with practical advice on managing bioprocessing and technology, upstream and downstream processing, manufacturing, regulations, formulation, scale-up/technology transfer, drug delivery, analytical testing and more.

www.biopharmaceuticalmedia.com www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 63


Ad Index

Page 31 & 43

Avantor Inc

Page 21

Diamond Pharma Services

Page 5 Owen Mumford Page 55

Packaging Coordinators Inc

Page 59 PatientView Page 63

Pharma Publications Ltd.

BC

Plasmid Factory

Page 3

R.G.C.C. Group

IFC SGS SA Page 47 Sonotec

IBC RICHTER-HELM BIOLOGICS GMBH & CO. KG

I hope this journal guides you progressively, through the maze of activities and changes taking place in the biopharmaceutical industry

IBI is also now active on social media. Follow us on: www.facebook.com/Biopharmaceuticalmedia Subscribe today at www.biopharmaceuticalmedia.com or email info@pharmapubs.com

64 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

www.plus.google.com/biopharmaceuticalmedia www.twitter.com/biopharmace www.biopharmaceuticalmedia.tumblr.com/

Winter 2020 Volume 3 Issue 3


ARE YOU LOOKING F OR EXPE RTS IN

MICROBIAL PRODUCTION?

CONTRACT DEVELOPMENT AND MANUFACTURING OF BIOPHARMACEUTICALS Richter-Helm is a Germany-based GMP manufacturer specialized in products derived from bacteria and yeasts, with a proven 30-year track record. Count on us to flexibly provide a comprehensive range of services and customized solutions. Clients worldwide have already benefited from our commitment to good manufacturing practice and total transparency. Our work focuses on recombinant proteins, plasmid DNA, antibody fragments, and vaccines. Richter-Helm consistently works to the highest standards of pharmaceutical quality.

Contact us +49 40 55290-801 www.richter-helm.eu www.biopharmaceuticalmedia.com

INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY 65


20 YEARS

cines c a v A N l for mR ! a i r e t a m tities n a u Starting q e in larg e: We provid

Customized minicircle & plasmid production g Extensive expertise in manufacturing of minicircles for CAR-T cell production High Quality Grade DNA for GMP production of viral vectors and RNA Helper & Packaging plasmids for AAV production In Stock QC including CGE service for topology analysis

66 INTERNATIONAL BIOPHARMACEUTICAL INDUSTRY

PlasmidFactory.com

Winter 2020 Volume 3 Issue 3

PlasmidFactory GmbH & Co. KG | Meisenstraße 96 | D-33607 Bielefeld Germany | Fon +49 521 2997350

grafeting gmbh | Bildmaterial: stock.adobe.com

The better way to DNA


Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.