Biomarkers.

Slides:



Advertisements
Similar presentations
Post Research Benefits Mandika Wijeyaratne MS, MD, FRCS Dept. of Surgery, Colombo.
Advertisements

Regulatory Framework Leigh Shaw, Director.
4 pictures 1 word! Learning Outcomes Examine how future medicine may take into account an individuals genome for personalised medicine Distinguish between.
Breakout Session 4: Personalized Medicine and Subgroup Selection Christopher Jennison, University of Bath Robert A. Beckman, Daiichi Sankyo Pharmaceutical.
Clinical Trial Designs for the Evaluation of Prognostic & Predictive Classifiers Richard Simon, D.Sc. Chief, Biometric Research Branch National Cancer.
Approaches to incorporating pharmacoeconomic data into early drug discovery Kevin Sheehy Acting CEO Medicines New Zealand.
Personalized Medicine Presentation by: Alex Wiki by: John and Lydia.
Personalized Medicine
Statistics for Health Care
Cancer Clinical Trials:
Preventing HIV/AIDS There is no way to tell just by looking whether a person is infected with HIV. Because people are unaware that they are HIV-positive,
University of Utah Department of Human Genetics Pharmacogenomics Louisa A. Stark, Ph.D. Director.
Improving Phase II Designs: Increasing phase III success Methods in Clinical Cancer Research Feb 6, 2015.
Clinical Trials. What is a clinical trial? Clinical trials are research studies involving people Used to find better ways to prevent, detect, and treat.
Testing People Scientifically.  Clinical trials are research studies in which people help doctors and researchers find ways to improve health care. Each.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication A substance used in the diagnosis, treatment,
Ethics of Biotechnology. CLONING What is CLONING? Creating new and identical organisms using biotechnology.
Statistics for Health Care Biostatistics. Phases of a Full Clinical Trial Phase I – the trial takes place after the development of a therapy and is designed.
Biomedical Research Objective 2 Biomedical Research Methods.
Biomedical Research.
Pharmacogenetics & Pharmacogenomics Personalized Medicine.
Biomedical Research. What is Biomedical Research Biomedical research is the area of science devoted to the study of the processes of life; prevention.
 A test of a new intervention or treatment on people.
Developing medicines for the future and why it is challenging Angela Milne.
Cancer 101: A Cancer Education and Training Program for [Target Population] Date Location Presented by: Presenter 1 Presenter 2.
European Patients’ Academy on Therapeutic Innovation Ethical and practical challenges of organising clinical trials in small populations.
Patients access to investigational drugs Steinar Aamdal Dept. of Clinical Cancer Research Oslo University Hospital.
European Patients’ Academy on Therapeutic Innovation Aspects of pharmacovigilance: Post-Authorisation Efficacy Studies (PAES)
Gene Therapy. What is Gene Therapy? Gene Therapy is the insertion of genes into an individual’s cells and tissues to treat a disease. Gene Therapy is.
INTERPRETING GENETIC MUTATIONAL DATA FOR CLINICAL ONCOLOGY Ben Ho Park, M.D., Ph.D. Associate Professor of Oncology Johns Hopkins University May 2014.
Learning objectives Know the stages of drug development Explain why animals are used in research Analyse why new drugs may fail Starter: 1.List 5-10 medications.
WHAT IS THE IMPACT OF THE HUMAN GENOME PROJECT FOR DRUG DEVELOPMENT? Arman & Fin.
I farmaci innovativi in una prospettiva europea Giuseppe M.C. Rosano, MD, PhD Cardiovascular and Cell Sciences Research Institute, St George's University.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication recognized or defined by the U.S. Food, Drug,
European Patients’ Academy on Therapeutic Innovation Challenges in Personalised Medicine.
Tumor markers 1111.
CLINICAL TRIALS.
Moiz Bakhiet, MD, PhD, Professor and Chairman
New research areas in personalised medicines
Myotonic Dystrophy Research: What’s Next
Off-label Use.
KEY CONCEPT Technology continually changes the way biologists work.
When things don’t go as planned.
How Can You Study Human Heredity?
Evidence-based Medicine
Genetic Engineering and Animal Research
CLINICAL PROTOCOL DEVELOPMENT
The brain.
The Many Careers of Pharmacy
Principles of Epidemiology E
Within Trial Decisions: Unblinding and Termination
Jeopardy Testing 1, 2, 3 She Has The Cancer Radiation or Chemo?
Clinical Study Results Publication
Critical Reading of Clinical Study Results
Regulatory perspective
KEY CONCEPT Technology continually changes the way biologists work.
Biomedical Research.
Speeding access to therapies
So …What’s the future of medicine?
Comments on design and sequence of biomarker studies
Complete Station Race Assignment…
Personalised Medicine ‘into the future’
GENERAL PATHOLOGY Diagnostic Services in Health Care that apply to the various diseases The role of diagnostics in Healthcare Quality procedures.
Ethical Considerations for Pediatric Clinical Investigations
Thank you to the Congressional Public Health Caucus, which has brought us today to talk about an important issue that affects patient care and treatment,
A New Approach to Clinical Trials
Objective 2 Biomedical Research Methods
An introduction to EMA’s support for medicines development
BioCapital Europe 2019, Amsterdam
Using clinical trial data as real-world evidence
Presentation transcript:

Biomarkers

Biological Markers A biological marker is a measurable indicator that can tell us something about a person’s health or disease state, for example, disease (pathological) processes in the body, for example, disease stage, biological processes in the body (heart rate, blood pressure, temperature), a person’s response to a treatment or medicine, or a psychological condition. Biomarkers are used in many scientific fields, in different ways at different stages of medicines development. The accuracy of biomarkers can vary, therefore, not all biomarkers are suitable for medicines development.

Examples of Biomarkers Glucose levels that are used as a biomarker in managing diabetes. Brain images such as Magnetic Resonance Imaging (MRI) that can provide information about the progression of Multiple Sclerosis. biological substances such as enzymes, which may be found in the blood or in tissue samples and are often used in cancer research, genetic (DNA) changes, medical images, or X-rays.

Aims of Biomarker Use 1. Improving the processes of medicines development Clinical trials seek to measure patients’ responses to a treatment. If it is not possible to measure the response directly, biomarkers may provide an alternative way of measuring an outcome - they serve as surrogate endpoints.

There are advantages of using validated biomarkers as surrogate endpoints, they might be able to be measured earlier, more easily, or frequently, with high precision; they may be less affected by other treatments, reduce the sample size required, and allow researchers to make faster decisions; they offer important ethical advantages as surrogate endpoints in diseases with poor prognoses.

Example of the use of a biomarker as a surrogate endpoint An example of the use of a biomarker as a surrogate endpoint comes from the development of antiretroviral medicines for HIV and AIDS. Previously, studies would have been based on hard clinical endpoints such as the progression of the HIV infection to AIDS and/or patient survival. Now, cell changes such as levels of ‘CD4 lymphocytes’ and changes in the levels of HI-virus RNA in plasma can be used as surrogate endpoints.

Aims of Biomarker Use 2. Tailoring treatment to individuals Biomarker research is helping to improve how well we can predict a person’s risk of disease, how a disease might progress once it is diagnosed, how an individual will respond to a medicine. This will enable safer and more effective treatment decisions. Examples: Blood sugar levels in a patient’s blood can be used to monitor if an individual is responding to diabetes treatment. Magnetic Resonance Imaging (MRI) scans of a patient’s brain can be used to monitor the progress of the disease in Multiple Sclerosis.

Many new biomarkers are being discovered and used during the development of new medicines. Many of these use: genomics (analyses of changes occurring at the gene level), proteomics (analyses of changes on the protein level), and/or metabolomics (analyses of differences in chemical molecules that play an important role in body and cell function).

Biomarkers in Medicines Development Cancer (oncology) research was one of the first areas where the use of biomarkers was adopted. Biomarkers are used to make exploratory trials and Phase II trials (‘Proof of Concept’ trials) of medicines more efficient. Only a limited number of biomarkers can be used for clinical endpoints in Phase III trials (confirmatory trials). Biomarkers may be used in late-stage trials in combination with clinical outcomes (clinical endpoints).

For some medicines, only a minority of patients might respond. It is helpful to identify these patients for clinical trials using biomarkers.

Companion Diagnostics Companion diagnostics are tests that are validated and approved for marketing alongside a new medicine. These tests may help to: select patients likely to respond to a medicine, exclude those patients likely to have an adverse reaction, and determine the best dose for a patient.

Companion Diagnostics Many companies developing targeted therapies for cancer have also begun to consider the potential benefits of developing a diagnostic to pair with that treatment. The trend is to develop medicines and companion diagnostics together, rather than have both developments happen separately.

Biomarkers potential to increase the efficiency of medicines development Speeding up clinical trials Biomarkers can be used to detect an effect (or lack of effect) earlier and more frequently than if only a clinical outcome (endpoint) is used. Example: a panel of biomarkers has been used in the early phases of a clinical trial for a psoriasis treatment. The biomarkers included ‘epidermal thickness’ (thickness of the outer layer of skin) and the activity levels of several genes. These were both measured in tissue samples.

Biomarkers potential to increase the efficiency of medicines development Streamlining clinical trials Biomarkers are used to identify those patients who are most suitable for a treatment, specifically, genomic biomarkers can be used to: identify patients with a particular disease sub-type or severity, exclude patients at increased risk of serious side effects (adverse reactions), for example, melanoma patients are at risk of their condition getting worse if their tumours do not have a certain mutation in the ‘BRAF’ gene and they are treated with kinase inhibitors, identify patients with a high chance of benefiting from a particular medicine.

Biomarkers potential to increase the efficiency of medicines development Improve our understanding Biomarkers can improve understanding of how new medicines work and may lead to novel approaches to medicines development in both non-clinical and clinical phases Improving the ethics of trial recruitment Biomarkers can help exclude people who won’t benefit from starting a non-helpful treatment, thus providing an ethical benefit. Improving trial monitoring and stopping unhelpful trials early Biomarkers may help decide whether to stop a trial early if there is no benefit to be gained by the patients in the trial or an obvious benefit where withholding treatment would be unethical.

Biomarkers potential to increase the efficiency of medicines development Speeding up authorisation A medicine that is having a positive effect may be authorised sooner based on information provided using biomarkers and therefore may be prescribed earlier to patients who will benefit.

Challenges of using biomarkers in medicines development As the use of biomarkers in pharmaceutical research grows, companies face new technical challenges, regulatory challenges, ethical challenges.

Technical Challenges Biomarkers used in clinical trials must be validated through scientific evidence to ensure that the biomarker test is sufficiently accurate, reliable, sensitive, and specific. The biomarker needs show a reasonable relationship to the disease being studied. If a biomarker is used to predict how severe a disease may become, is there enough evidence of the ‘predictive ability’ with this biomarker?

Technical Challenges IT systems for data management and data analysis must be reliable and fast to cope with the amount of data generated. All biomarker measurements must be correctly linked with individual patients. Where the use of a companion diagnostic is required for a new medicine to be prescribed, a new platform or kit for testing patients in the clinic may need to be developed.

Regulatory Challenges The regulation of the use of novel methods such as biomarkers in medicines development is evolving. ‘Biomarker’ and ‘surrogate endpoint’ are not interchangeable terms. For a biomarker to be used as a surrogate endpoint, studies will be done to assess the direct relationship of the biomarker with: the development of the disease, a treatment intervention with an important clinical endpoint.

Regulatory Challenges Developers of novel biomarkers are being encouraged to engage with regulators at an early stage. They can submit their plans to use biomarkers to the EMA. Validating biomarkers to meet regulatory standards can be complex and expensive. This is especially challenging if a biomarker is intended to be used as a surrogate endpoint. In this case, a dedicated clinical trial is required, designed to test the link between the biomarker and the clinical endpoint.

Regulatory Challenges In the EU, medicines and diagnostics are regulated differently. Licensing a medicine and its companion diagnostic together adds an extra layer of complexity to the approval process.

Ethical Challenges Many of the ethical issues that arise in biomarker research are linked to the storage and use of tissue samples and the associated handling of personal medical data. Wider concerns have been raised about the impact of targeted medicine (which is largely based on biomarker research). As targeted treatments only bring benefits to the sub- population of patients that respond to them, the challenge is to ensure that medicines are developed for those who fall outside of this sub-population.

References Industry Pharmacogenomics Working Group (2012). Understanding the intent, scope, and public health benefits of exploratory biomarker research. A guide for IRBs/IECs and investigational site staff. Retrieved 1 September, 2015 from http://i- pwg.org/index.php?option=com_docman&task=doc_dow nload&gid=187&Itemid=118