This page is archived

Links to external sources may no longer work as intended. The content may not represent the latest thinking in this area or the Society’s current position on the topic.

Delivering novel therapies in the 21st century

24 - 25 October 2018 07:30 - 16:30
673371430

Read the conference report (PDF).

Watch talks and presentations from the conference.

Delivering novel therapies in the 21st century is a two-day conference held by the Royal Society and Academy of Medical Sciences FORUM. Speakers will discuss recent scientific advances in therapeutic modalities and delivery. The meeting will also explore the infrastructure required to develop novel treatments and implications of these advances for policy and healthcare. Developments in areas including new immunotherapies, affinity reagents, bioelectronics, gene therapy and nanoparticle delivery will be considered within the broader context of personalised medicine, technology and data, regulation, value, access and care delivery.

Attending this event

This open event is free to attend and is intended for those with an interest in the life sciences sector, from a variety of backgrounds including academia, industry, government, the NHS, as well as regulatory, charitable and other scientific bodies.

Contact the Industry team for more information.

About the conference series

This meeting forms part of the Royal Society’s Transforming our Future series, and the Academy of Medical Sciences’ FORUM programme. The Transforming our Future meetings are unique, high-level events that address scientific and technical challenges of the next decade and bring together leading experts from wider scientific community, industry, government and charities.

The Academy’s FORUM programme brings together industry, academia and the NHS, and the charity, regulatory and wider healthcare sectors. It provides an independent platform to bring together leaders from across the life sciences sector to discuss scientific opportunities, technology trends, translational challenges and strategic choices in healthcare.

Organisers

  • Steve Rees, VP Discovery Biology, Discovery Sciences, AstraZeneca

    Steve currently leads the Discovery Biology department at AstraZeneca with global accountability for protein and cellular reagent generation and assay development, target identification and validation and chemical biology. Prior to this Steve led the Screening Sciences and Sample Management department and successfully implemented strategies for hit identification, compound profiling, sample management and open innovation. Steve has led multiple international collaborations and has authored >60 scientific papers. Steve is currently Chair of the European Laboratory Research and Innovation group (ELRIG), has served as Chair of the SLAS Europe Council, and is a member of the Scientific Advisory Board for Axol Biosciences, LifeArc and the Centre for Membrane Protein and Receptor Research at the Universities of Nottingham and Birmingham.

  • Professor Molly Stevens FREng, Professor of Biomedical Materials & Regenerative Medicine/Research Director for Biomedical Material Science, Imperial College London

    Molly M. Stevens is currently Professor of Biomedical Materials and Regenerative Medicine & Research Director for Biomedical Material Sciences in the Department of Materials, Department of Bioengineering and the Institute of Biomedical Engineering at Imperial College London. She received her PhD from The University of Nottingham in 2000, working within the School of Pharmaceutical Sciences. She conducted her postdoctoral research within the Department of Chemical Engineering at MIT in the labs of Prof Robert Langer, where she co-developed innovative techniques for the regenerative of bone and other tissues. She joined Imperial College in 2004 and was promoted as Professor in 2008. Research in the Stevens Programme focusses on designing and developing innovative bio-inspired materials for applications in regenerative medicine, tissue engineering and biosensing.  

    Molly Stevens’ research has been recognised by over 20 major awards, such as the 2016 Clemson Award for Basic Research from the Society for Biomaterials, the EU40 Prize for best material scientist under the age of 40, a listing in The Times as one of the top 10 scientists under 40 and the European Life Sciences 2014 Research Group of the Year Award, amongst many others. She was recently elected to the Fellowship of the Royal Academy for Engineering and delivered the Clifford Paterson Lecture for the Royal Society in 2012. She has previously served on the Board of Reviewing Editor for Science and is Associate Editor of ACS Nano. More information on the Stevens Group can be found at www.stevensgroup.org.

  • Sir John Skehel FMedSci FRS, Vice-President and Biological Secretary, The Royal Society

    John Skehel has provided major insights into the molecular basis of how viruses recognise and infect their host cells. John focusses on the virus that causes influenza, of which there are 3–5 million cases a year worldwide, resulting in up to 500,000 deaths. 

    To infect a cell, the influenza virus must bind to a sialic acid-containing receptor on the cell surface, which it achieves through its own haemagglutinin glycoprotein. John was able to isolate, crystallise and subsequently determine the three-dimensional structure of this molecule. He also observed that under conditions of low pH, haemagglutinin changes shape — allowing the virus to fuse with and enter the cell.

    John has been a leader in virology research for over thirty years. He headed the WHO Collaborating Centre for Reference and Research on Influenza between 1975 and 1993 and was the Director of the National Institute for Medical Research from 1987–2006. His pioneering research was recognised in 1996 when he received a knighthood.

Schedule

Sir John Skehel FMedSci FRS, Vice-President and Biological Secretary, The Royal Society

Professor Sir Robert Lechler PMedSci, President, Academy of Medical Sciences

The majority of today’s medicines are based on small molecule, protein or monoclonal antibody platforms. With an ever increasing need to better understand complex disease biology we need our scientists to be able to prosecute the best validated targets even if they are viewed as traditionally intractable from a traditional drug discovery perspective. Exploring novel drug modalities which complement existing strengths in small molecule and biologic platforms has the potential to open new therapeutic options.

Over the past few years we have made significant progress across seven additional drug discovery platforms, complementing existing capabilities in chemistry and protein engineering. These include modified mRNA therapies, antisense oligonucleotides (ASOs), oligonucleotide conjugates, bicyclic peptides, proteolysis targeting chimeras (PROTACs), anticalins and CRISPR. By combining our distinctive skills and technologies with those of our partners we are accelerating our goal to bring new medicines to patients in areas of high unmet medical need.

Sir Menelas (Mene) Pangalos PhD FRSB FMedSci, Executive Vice President & President BioPharmaceuticals R&D, AstraZeneca

Chair

Steve Rees, VP Discovery Biology, Discovery Sciences, AstraZeneca

09:40 - 10:00 Future opportunities for small molecule therapeutics

The vast majority of small molecule drug discovery projects explored by medicinal chemists to date have targeted protein function, predominantly although not exclusively by blocking activity.  Typically leads are defined from high throughput screens and structure or substrate based design all of which are biased towards active site binders.  The past decade has seen the emergence of a number of technologies that are beginning to challenge this practise.  I will review progress in 3 areas during the presentation.  Namely, encoded library synthesis and screening that massively increases the number of compounds that can be evaluated in an unbiased binding mode, the emergence of small molecules that program protein targets for destruction and finally the evolution of chemical biology tools and technologies for assigning mode of action to cell based screening hits.  These technologies are poised to fundamentally change the landscape of application of small molecule drug discovery adding new mode of action options.

Dr Tony Wood, Senior Vice President, Platform Technology and Science, GSK

10:05 - 10:25 Bicyclic peptides as novel therapeutics

Bicyclic peptides or Bicycles® are a new therapeutic modality combining the attributes of antibodies, small molecules and peptides within one molecule. Bicycles® can be used as standalone therapeutics or coupled to deliver various therapeutic payloads, including cytotoxins, immune modulators, radionuclides or nucleic acids in a tissue selective manner. Bicycle Therapeutics’ has unique proprietary technology for the discovery of Bicycles® and is focused in oncology where Bicycle Toxin Conjugates® (BTCs) selectively deliver toxins to tumours, killing them with a high degree of precision and minimal effects on healthy cells. BTCs take advantage of Bicycles’ inherent properties of high target affinity and specificity, fast penetration into the tumour and low systemic exposure, combined with rapid renal elimination. BT1718, Bicycle’s lead BTC is currently under clinical exploration in collaboration with CRUK. Bicycles®also offer a range of unique opportunities in immune oncology and other therapeutic areas outside oncology.

Dr Kevin Lee, Chief Executive Officer, Bicycle Therapeutics

Chair

Steve Rees, VP Discovery Biology, Discovery Sciences, AstraZeneca

11:00 - 11:20 The landscape for new biologics formats

Antibody engineering has come of age and there are now over 500 antibody therapeutics in clinical development and nearly 70 approved for a wide range of different disease indications. Amongst these, there are many examples of antibodies with engineered properties such as increased potency, half-life extension and improved stability. The landscape for biologics therapies is evolving to deliver additional functionality with the introduction of novel formats, such as bispecific antibodies, peptide-antibody fusions, scaffolds and antibody -drug conjugates. Case studies will be presented illustrating the benefits of different formats and how they can be applied in different disease settings.

Dr Jane Osbourn, VP Research and Development and Site Leader, MedImmune

11:25 - 11:45 Antigen-targeted TCRs in cancer immunotherapy

Cancer immunotherapies function to harness the body’s own immune system to eradicate cancer cells. Critical to this process is the T cell, capable of directing potent and antigen-specific immune responses. T cell receptor (TCR) antigens are short intracellularly processed peptides presented on the cell surface, offering distinct advantages over antibody-based therapies that recognise secreted or cell surface proteins. Here, we describe two novel platforms designed to overcome the limitations of the natural T cell response to deliver high-affinity engineered TCRs to an identified and validated target antigen. The platforms can be distinguished based on a cellular versus soluble approach. SPEAR (Specific Peptide Enhanced Affinity Receptor) T cell therapies targeting MAGE-A10, MAGE-A4, AFP and NY-ESO are progressing through clinically studies in multiple solid and hematologic cancers. The lead soluble ImmTAC® (Immune mobilising monoclonal TCR against cancer) molecule, IMCgp100, has entered pivotal monotherapy trials for the treatment of patients with metastatic uveal melanoma.

Dr Bent Jakobsen FMedSci, Chief Scientific Officer, Immunocore and Scientific Founder, Adaptimmune

11:50 - 12:10 Antisense oligonucleotide therapy for Huntington's disease - results from the first HTT lowering clinical trial

Background: HD is an autosomal dominant neurodegenerative disease caused by CAG repeat expansion in the HTT gene resulting in polyglutamine expansion in the mutant huntingtin protein (mHTT) with a toxic gain-of-function disease mechanism. No disease-modifying treatments are currently available. In transgenic rodent models of HD, suppressing HTT production delays disease progression and reverses disease phenotype. A comprehensive drug discovery effort, including extensive preclinical testing, was undertaken to design a well-tolerated ASO with high specificity to human HTT mRNA that potently suppresses HTT production.

Design/Methods: In this first-in-human, multi-center, double-blind clinical trial (NCT02519036), 46 patients were randomized (3:1) to receive four doses of IONIS-HTTRx (RG6042) or placebo by monthly bolus intrathecal (IT) injection followed by a 4-month untreated period. Five ascending-dose cohorts were enrolled with independent Data Safety Monitoring Board review of safety, PK and target engagement prior to dose escalation.

Results: IONIS-HTTRx was well-tolerated at all doses tested.  Adverse events were mostly mild and unrelated to study drug. There were no adverse trends in laboratory parameters. No patients prematurely discontinued from treatment. ASO was measurable in CSF and plasma. Significant, dose-dependent reductions in CSF mHTT were observed.

Conclusions: ASO technology has the potential to provide disease-modifying benefits to patients with neurodegenerative diseases. In this Phase 1/2a trial in early stage HD patients, IONIS-HTTRx delivered via IT injection was well tolerated with no study drug-related adverse safety signals during the treatment or follow-up periods.  Significant dose-dependent reductions in CSF mHTT were observed, suggesting that IONIS-HTTRx is a promising therapeutic for the treatment of HD.

Study Supported By: Ionis Pharmaceuticals

Authors: Sarah J Tabrizi, Blair Leavitt, Bernhard Landwehrmeyer, Edward Wild, Carsten Saft, Roger Barker, David Craufurd, Josef Priller, Hugh Rickards, Anne Rosser, Holly Kordasiewicz, Christian Czech, Eric Swayze, Daniel A. Norris, Tiffany Baumann, Irene Gerlach, Scott Schobel, Anne Smith, Roger Lane, C. Frank Bennett - on behalf of the study teams

Professor Sarah J Tabrizi FMedSci, Director of UCL Huntington's disease centre, UCL Institute of Neurology, and UK DRI UCL

12:15 - 12:35 Bioelectronics - is the next novel therapy class an electronic implant?

Electrical and molecular signalling constitute main axes of control in biology. Peripheral nerves conduct electrical impulses to and from organs, regulating a wide range of physiological processes, from smooth muscle tone to cellular programming and molecular secretion. Modulating such signals is proving to have therapeutic potential across chronic diseases such as rheumatoid arthritis, diabetes, and asthma. Bioelectronic medicine is a new therapeutic modality that seeks to treat such diseases with miniaturised, implantable devices delivering precision neuromodulation. This talk will outline the therapeutic potential, describe how mechanistic learnings from animal models, surgical approaches, and new technology solutions are coming together into the device development. The clinical development steps ahead will also be discussedss. A glimpse will be provided into how smart device-based therapies may change medical practice in the coming decades and highlight opportunities for collaborative research towards that goal.

Dr Kristoffer Famm, President, Galvani Bioelectronics

Chair

Professor Molly Stevens FREng, Professor of Biomedical Materials & Regenerative Medicine/Research Director for Biomedical Material Science, Imperial College London

13:30 - 13:50 Chair's introduction

The Chair's introduction will set the scene for this session by highlighting latest developments in the field as well as the challenges and opportunities that lie ahead in the design of new approaches for drug delivery.

Professor Molly Stevens FREng, Professor of Biomedical Materials & Regenerative Medicine/Research Director for Biomedical Material Science, Imperial College London

13:50 - 14:10 Early days of gene therapy – are we there yet?

Gene therapy is emerging as a new medical modality for treating genetic disorders. Recombinant viral, and non-viral vectors represent multiple approaches for delivery that is showing significant promise for the treatment of several types of diseases in humans including heart disease, musculoskeletal diseases, hemophilia, rheumatoid arthritis, and others. This presentation will first provide a historical review of developing a viral vector: namely Adeno-associated virus (AAV), as an efficient delivery system and more importantly, highlight the limitations that are still prevalent with respect to everyday success. Highlighted examples of successful gene transfer will focus on the recent data accumulating in the clinic for genetic disorders such as Hemophilia, Blindness, and muscular dystrophy. As with most new areas of drug development, new issues arise after entering the clinic. I will outline the observations that are still appearing problematic (e.g immune response, high costs of manufacturing, treating ultra rare orphan diseases) for further discussion. Furthermore, for any biologic drug to become commercially viable - and to be able to attract the significant drug development resources of industry - a cost effective drug manufacturing process must be available. A portion of this presentation will focus on current methods of vector production.

As the field continues to mature, we see more and more novel therapeutics entering the clinic. The ability to generate high titer, high quality, GMP-grade vector products will be paramount to advance such efforts. With this backdrop, we anticipate accelerated establishment of “proof of concept” studies in humans. This will be achieved by multiple groups of researchers, using viral and non-viral vectors as well as specific academic and commercial vector cores, that will assist translational and clinical research communities as the journey continues.

Professor Richard Jude Samulski, Professor of Pharmacology, Gene Therapy Center, University of North Carolina School of Medicine

14:15 - 14:35 Nucleic acid delivery systems for RNA therapy and gene editing

High throughput combinatorial approaches have revolutionized small molecule drug discovery. Here we describe our work on high throughput methods for developing and characterizing RNA delivery and gene editing systems. Libraries of degradable polymers and lipid-like materials have been synthesized, formulated and screened for their ability to delivery RNA, both in vitro and in vivo. A number of delivery formulations have been developed with in vivo efficacy, and show potential therapeutic application for the treatment of genetic disease, viral infection, and cancer.

Professor Daniel G Anderson, Professor of Applied Biology, Associate Professor of Chemical Engineering, and member of the Koch Institute for Integrative Cancer Research, MIT

14:40 - 15:00 Generation and testing of clinical-grade engineered exosomes to facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer

Exosomes are extracellular vesicles naturally generated by cells in the blood. Exosomes transfer genetic material, including exogenously loaded siRNA, to cancer cells. The mutant form of the GTPase KRAS is a key driver of pancreatic cancer but remains a challenging therapeutic target. Exosomes derived from normal fibroblast-like mesenchymal cells were engineered to carry short interfering RNA, or short hairpin RNA specific to oncogenic KrasG12D, a common mutation in pancreatic cancer. Engineered exosomes (iExosomes) target oncogenic Kras with an enhanced efficacy dependent on CD47 and facilitated by macropinocytosis. Treatment with iExosomes suppress cancer in pancreatic cancer mouse models and significantly increase survival by direct and specific targeting of oncogenic Kras in tumours using iExosomes. To translate to the clinic, we developed a bioreactor-based, large-scale production of clinical-grade exosomes to generate engineered exosomes to target oncogenic Kras. Clinical-grade GMP iExosomes were tested in vitro and in vivo to confirm suppression of oncogenic Kras and an increase in the survival of several mouse models with pancreatic cancer. Our studies determine the shelf life, biodistribution, toxicology profile, and efficacy in combination with chemotherapy for future clinical testing of GMP iExosomes. This illustrates the process and feasibility of generating clinical-grade exosomes for various therapies of human diseases.

Dr Raghu Kalluri, Professor and Chairman, Department of Cancer Biology and Director, Metastasis Research Center, University of Texas MD Anderson Cancer Center

Chair

Professor Molly Stevens FREng, Professor of Biomedical Materials & Regenerative Medicine/Research Director for Biomedical Material Science, Imperial College London

15:35 - 15:55 Shaken and stirred: Ultrasound-enhanced drug delivery

The thermal and mechanical effects produced by ultrasound have a major role to play in enabling therapeutics to cross biological barriers, such as those presented by the highly irregular vasculature and elevated interstitial pressure in tumours, or the cellular membrane. We first present results from a first-in-man clinical trial (TarDox) of targeted drug delivery to liver tumours using thermosensitive liposomes in combination with mild hyperthermia delivered non-invasively using focused ultrasound. In a further development, a mechanical effect of ultrasound, known as acoustic cavitation, is exploited for convective transport of therapeutics. This approach necessitates lower-intensity ultrasound delivered from a portable device and does not require drug modification or encapsulation. In oncology, sub-micron cavitation-inducing particles co-administered with small-molecule drugs, therapeutic antibodies or viruses are shown to increase both the penetration and total dose delivered to tumours following systemic administration, significantly enhancing their therapeutic efficacy even at a reduced systemic dose.

Professor Constantin Coussios, Professor of Biomedical Engineering (Drug Delivery), Head of the Biomedical Ultrasonics, Biotherapy & Biopharmaceuticals Laboratory, Oxford University

16:00 - 16:20 Cancer evolution and immune escape: TRACERx

Increasing evidence supports complex subclonal relationships in solid tumours, manifested as intratumour heterogeneity. Parallel evolution of subclones, with distinct somatic events occurring in the same gene, signal transduction pathway or protein complex, suggests constraints to tumour evolution that might be therapeutically exploitable. Emerging data from TRACERx, a longitudinal lung cancer evolution study will be presented. Drivers of tumour heterogeneity change during the disease course and contribute to the temporally distinct origins of lung cancer driver events. APOBEC driven mutagenesis appears to be enriched in subclones in multiple tumour types. Oncogene, tumour suppressor gene and drug induced DNA replication stress are found to drive APOBEC mutagenesis. Evidence that intratumour heterogeneity and chromosomal instability is finely tuned will be presented, to create sufficient diversity for adaptation mitigating the risks of excessive genome instability resulting in cell autonomous lethality. On-going chromosomal instability, manifested as Mirrored Subclonal Allelic Imbalance (MSAI) is found to be a major driver of intratumour heterogeneity in non-small cell lung cancer, contributing to parallel evolution and selection. The finding of subclonal driver events, evidence of ongoing selection within subclones, combined with genome instability driving cell-to-cell variation is likely to limit the efficacy of targeted monotherapies, suggesting the need for new approaches to drug development and clinical trial design and integration of cancer immunotherapeutic approaches. The clonal neo-antigenic architecture may act as a tumour vulnerability, targeting multiple clonal neo-antigens present in each tumour to mitigate resistance and treatment failure. The role of cancer genome instability driving immune evasion and HLA/MHC loss and immune escape will be presented.

UCl-CRUK-2014

Professor Charles Swanton FRS FMedSci, Translational Cancer Therapeutics Laboratory Group Leader, The Francis Crick Institute

Chair

Dr Nicholas Medcalf, Senior Innovation Lead in Advanced Therapies, UK Research and Innovation, Innovate UK

In this session the panellists consider the ways in which manufacturing is changing to address the challenges of customisation, lot size, shelf life and inventory management for the new categories of medicinal products. The impact of the nature of the product, the integration of the supply chain and local finishing with the traditional manufacturing steps will be considered and the roles and responsibilities of those responsible for the chain of custody will be re-examined. The potential reduction of the R&D timeline by providing manufacturing technology that can be applied during the discovery phase will be discussed.

Dr Annette Bak, Head of Advanced Drug Delivery, AstraZeneca (Sweden)

Keith Thompson, CEO, Cell and Gene Therapy Catapult

Dr Derek Adams, Chief Technology and Manufacturing Officer, Bluebird Bio

Professor Richard Jude Samulski, Professor of Pharmacology, Gene Therapy Center, University of North Carolina School of Medicine

Steve Rees, VP Discovery Biology, Discovery Sciences, AstraZeneca

Chair

Professor Dame Angela McLean DBE FRS, Professor of Mathematical Biology, University of Oxford and Co-Chair, Data Theme Community Group of Interest, Royal Society

09:40 - 10:05 The potential for genomics to transform drug discovery

Currently 90% of drug candidates taken into clinical trials fail to be approved.  Drug targets are often motivated by studies in model systems, but the modulation of these targets often fails to be effective in humans, underlining out relative lack of understanding of human biology.  Analysis of human genetic variation and its phenotypic consequences offers the potential to study perturbations of human biology directly.  Combining large-scale genomic data sets now allows simultaneous analysis of thousands of different phenotypes so that for the first time it is possible to exploit what has been called Nature’s clinical trial so as to begin to reveal the human wiring diagram.  The resulting biological insights offer a powerful new approach to empower target discovery and other aspects of drug development.

Professor Peter Donnelly FMedSci FRS, CEO, Genomics PLC and Professor, Statistical Science, University of Oxford

10:10 - 10:35 Is Digital Health the snake oil of the 21st century?

At the June 2016 Annual Meeting of the American Medical Association, CEO James L Madara, M.D speaking of “Digital Health” observed “From ineffective electronic health records, to an explosion of direct-to-consumer digital health products, to apps of mixed quality – it's the digital snake oil of the early 21st century.” Without doubt “Digital Health” has been greatly hyped over the last few years and has come to mean different things to different people. However, by considering alternative definitions of digital health and understanding the origins of “digital snake oil” a new realisation of digital health is presented. Based on the concept of the “digitised patient” represented in the form of high resolution dynamic physiological images, this view points to new clinical pathways and more efficient ways of delivering care both in and out of hospital.

Keith Errey, CEO, Isansys Lifecare Ltd

10:40 - 11:05 Companion AI: seeking dynamic precision and better drug-AI-outcomes

As most health systems face escalating demands from more people living longer with multiple long-term conditions the returns on therapeutic investments, especially in multimorbidity, are highly uncertain. Precision medicine investments have focused on biological stratification and current clinical delivery models, which concentrate resources on late-stage, single-condition care. Concurrently, patient data are being integrated across healthcare, research and civic settings to pursue digital futures. AI has been marketed beyond current evidence of successful harnessing of such data for actionable information, apart from mature niches such as medical image analysis. Nevertheless, many health behaviours, from physical activity to mental resilience, are now affected by AI and ubiquitous technologies, intended or not. The digital health industries are pursuing interoperability standards as key to a universal app store to assemble different elements of clinical and consumer healthcare. One app per drug/clinic, however, does not provide an organising framework for the 'companion AI' to understand and improve multi-morbid drug-outcomes. Concepts such as 'health avatar' or 'digital twin' have been put forward as ways to study unions of models for better clinical prediction and self-care interaction. This talk will explore the potential for multi-sector partnerships to move beyond the current app blizzard to a more scientific and governable approach to companion AI for better therapies.

Professor Iain Buchan, Professor of Public Health and Clinical Informatics, University of Liverpool

Chair

Professor Dame Angela McLean DBE FRS, Professor of Mathematical Biology, University of Oxford and Co-Chair, Data Theme Community Group of Interest, Royal Society

11:40 - 12:05 Novel approaches to assessing the safety and efficacy of new medicines

Traditional approaches to investigating the safety and efficacy of new medicines are primarily based on randomised controlled trials. There is, however, increasing interest in newer techniques that include adaptive designs, basket trials, umbrella trials, step wedge trials, ring trials and - in some circumstances - Baskerville trials. The advantages and disadvantages of these approaches will be discussed.

Sir Michael Rawlins GBE Kt FMedSci, Chair, Medicines and Healthcare products Regulatory Agency

12:10 - 13:00 Panel discussion

This panel session, chaired by Professor Angela McLean FRS, will focus on the future for data, technology and regulation. The panel will discuss the following questions:

How can the UK realise the value of patient data to discover the next generation of medicines?

How can the regulatory environment keep up with (and enable) innovation in therapy development?

How can we use data to change the paradigm from disease treatment to disease prevention?

How can technological advances enhance the discovery and development of novel therapies?

Professor Peter Donnelly FMedSci FRS, CEO, Genomics PLC and Professor, Statistical Science, University of Oxford

Keith Errey, CEO, Isansys Lifecare Ltd

Professor Iain Buchan, Professor of Public Health and Clinical Informatics, University of Liverpool

Sir Michael Rawlins GBE Kt FMedSci, Chair, Medicines and Healthcare products Regulatory Agency

Professor Jonathan K.C. Knowles, Visiting Professor, Medical Sciences Division, University of Oxford

Chair

Sir John Skehel FMedSci FRS, Vice-President and Biological Secretary, The Royal Society

14:00 - 14:25 How can we value novel medicines? A health economic perspective

In February of this year, a Special Task Force (STF) of the International Society for Pharmacoeconomics and Outcomes Research (ISPOR) published its report and recommendations on “A Health Economics Approach to US Value Assessment Frameworks.”  In assessing the potential value of novel medicines, the STF recommended that US public and private payers begin with a metric that compares incremental costs with incremental health benefits—with the latter measured in terms of quality-adjusted life years (QALYs) gained:  this is used in numerous country health systems and particularly by NICE in the UK.  Taking the cost-per-QALY metric as a starting point, the STF recommended to expand the concept of value and augment its measurement to include novel elements of value, such as insurance value, the value of hope, real option value, and scientific spillovers.  As a co-chair of the STF, Professor Garrison will provide an overview of the report, focusing on the implications for defining, measuring, and assessing the value of novel medicines globally.

Professor Lou Garrison, Professor Emeritus in the Comparative Health Outcomes, Policy, and Economics Institute, University of Washington

14:30 - 14:55 Health technology assessment of histology independent cancer drugs

A histology-independent cancer drug targets tumours with a certain genomic alteration, regardless of where in the body the tumour might be located. This means that a single indication for the drug can cover many different types of cancer, and that the clinical evidence in support of the drug's efficacy might look quite different to what decision makers such as regulatory agencies and health technology assessment (HTA) bodies are familiar with. As a consequence, assessing whether these drugs might provide value for money to a healthcare system - the task of HTA bodies such as NICE - is not straightforward.

As the pipeline of histology-independent cancer drugs is growing, exploring the challenges these new drugs will pose to HTA methods, as well as possible solutions that will enable their assessment, is becoming increasingly important. This talk will cover the key challenges that HTA bodies will face in assessing a histology-independent drug and discusses possible solutions to these challenges.

Dr Jacoline Bouvy, Senior Scientific Adviser, National Institute for Health and Care Excellence

15:00 - 15:25 The boundary between cognitive enhancement and treatment

While many people monitor their physical health using mobile devices and wearable technology to preserve their physical health, they rarely consider improving and monitoring their brain health. If we are going to have good mental capital and wellbeing throughout our lives, it is imperative that we consider mental health as being every bit as important as physical health and move to game-changing initiatives which include early detection and early effective treatment of neuropsychiatric disorders. Cognitive enhancing drugs, such as methylphenidate and modafinil, which were developed as treatments are increasingly being used by healthy people. Cognitive abilities are becoming progressively more important for work performance and successful competition in a global environment. However, work-stress, long hours, lack of sleep, shift work, and jet lag affect cognitive functions. Therefore, increasingly, healthy people are reported to use cognitive-enhancing drugs, brain-training apps and non-invasive brain stimulation to maintain or improve work performance.

Professor Barbara Sahakian FMedSci

Despite numerous attempts to encourage rapid adoption of new drugs and therapies, the NHS has remained remarkably slow at this. This has negative implications for more than just patient care. To get the conditions right for rapid adoption, the following conditions need to be considered: money, staff, and priorities within the NHS. Furthermore, the cost of new therapies needs to be considered across society, not just for the NHS, and this should include the importance of the life sciences industry to the UK.

Professor Dame Julie Moore, Professor of Health Systems, Warwick University

Sir John Skehel FMedSci FRS, Vice-President and Biological Secretary, The Royal Society