PROACTIVE PHARMACOVIGILANCE

Similar documents
The Role of the Pharmacist in Pharmacovigilance A Regulatory Perspective

PHARMA CONGRESS. Pharmacovigilance and Drug Safety: Assessing Future Regulatory and Compliance Developments. October 28, 2008

Importance of Pharmacovigilance for Pharmaceutical Industry

DEMONSTRATING YOUR MEDICINE S VALUE TO ALL STAKEHOLDERS TRUSTED COMMERCIALIZATION AND MARKET ACCESS EXPERTISE

Advance Topics in Pharmacoepidemiology. Risk Management. Conflict of Interest Declaration. Benefit Harm Profile?

A practical guide to achieving and maintaining global oversight and ensuring end-to-end pharmacovigilance

Quality check of spontaneous adverse drug reaction reporting forms of different countries y

GENETIC TESTING A Resource from the American College of Preventive Medicine

Summary of Provisions in 21 st Century Cures Act (H.R. 6) as passed by full House of Representatives, July 10, 2015

Pharmacovigilance: Vigilantia initiative

CRO partner in Rx/CDx Co-Development

Pharmacovigilance Approach to Challenges with Use of Medicines. Oksana Lebega USAID SIAPS project Antalya, Turkey December 10-13, 2013

Medical Affairs Not a Sunshine Day Analyzing the Impact of the Sunshine Act on Medical Affairs: Challenges for 2014 and Beyond

Policy Position. Pharmacy-mediated interchangeability for Similar Biotherapeutic Products (SBPs)

Draft Guidance for Industry Development and Use of Risk Minimization Action Plans

The Challenge of Pricing Combination Therapies

RWE from pre-clinical to launch. RWE from pre-clinical to launch. Standard of care Unmet needs. Disease burden Budget impact.

A High-Touch Approach to Improving Patient Access. Using field support to navigate reimbursement challenges

Guideline on good pharmacovigilance practices (GVP)

Drug Development: Why Does it Cost so Much? Lewis J. Smith, MD Professor of Medicine Director, Center for Clinical Research Associate VP for Research

Data Analytics Pharmacovigilance

PAREXEL ACCESS MANAGED ACCESS PROGRAMS

Update on Real World Evidence Data Collection

Managing Risk and Uncertainty Through the Drug Life cycle. Recent FDA Initiatives. Theresa Mullin, PhD

Use of RWE in a regulatory context: issues and examples. Rob Hemmings, MHRA

WHITE PAPER. Engaging the Evolving Stakeholder Network: A New Approach for Medical Affairs

Report from the Paediatric Committee on its first anniversary

Pharmacovigilance. An agency of the European Union

The Role of the CRO in Effective Risk-Based Monitoring

Current Trends at FDA: Implications for Data Requirements

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) POSITION PAPER ON COMPLIANCE WITH PHARMACOVIGILANCE REGULATORY OBLIGATIONS

Contact Information: Rapture Biotech D-201, Sector - 10, Noida Uttar Pradesh. Phone , ,

RWE Market Impact on Medicines: A Lens for Pharma

Work programme Adopted by the Management Board on 18 December December 2014 EMA/773839/2014 Rev. 1 Management Board

EFPIA-PHRMA PRINCIPLES FOR RESPONSIBLE CLINICAL TRIAL DATA SHARING REPORT ON THE 2016 MEMBER COMPANY SURVEY

Nonproprietary Naming of Biological Products; Draft Guidance for Industry; Docket No. FDA 2013 D 1543 October 27, 2015

GxP Auditing, Remediation, and Staff Augmentation

EU support for Health Research from FP6 to FP7

Off-Label Use of FDA-Approved Drugs and Biologicals

3. Human Biomedical Research. Defining Human Biomedical Research

Int. J. Pharm. Sci. Rev. Res., 28(2), September October 2014; Article No. 18, Pages:

DIGITAL EDITION BONUS CONTENT

Università Cattolica del Sacro Cuore

Emerging Trends in Regulatory Outsourcing WHITE PAPER.

ABPI response to European Commission consultation on advanced therapy medicinal products

GxP Auditing, Remediation, and Staff Augmentation

Testimony of Christopher Newton-Cheh, MD, MPH Volunteer for the American Heart Association

ZS Medical Affairs Outlook Report Analysis of Field Medical Growth and Industry Trends

Partnering with Rare Disease Patient Groups. Namrata Taak R&D External Communications, Rare Diseases

Q10 PHARMACEUTICAL QUALITY SYSTEM

Putting CDISC Standards to Work How Converting to CDISC Standards Early in the Clinical Trial Process Will Make Your CDISC Investment Pay

The interface between Good Clinical Practice and Good Manufacturing Practice

Reimbursement Strategy for Companion Diagnostics:

Use of Big Data to Support Regulatory Decision Making

PREVIEW. Benchmarking European Investigator Payments Industry Standard Research

Workshop on Access to and Uptake of Biosimilar Medicinal Products

Pharma Medical Affairs 2020 and beyond. Matthias Evers Edd Fleming Arnab Ghatak Jan Hartmann Arif Nathoo Ron Piervincenzi Lawrence Wai Ann Westra

VIVEK VIKRAM SINGH, SUBJECT MATTER EXPERT, RESEARCH & ANALYTICS CAN COMPETITIVE INTELLIGENCE STRATEGIES HELP IN ORPHAN DRUG DEVELOPMENT?

New Pharmacovigilance legislation. Post-authorisation safety studies. ENCePP Plenary meeting. 3 May 2012

Insights into the Evolving Pricing & Market Access Environment

STATEMENT SANDRA KWEDER, M.D DEPUTY DIRECTOR, OFFICE OF NEW DRUGS CENTER FOR DRUG EVALUATION AND RESEARCH U.S. FOOD AND DRUG ADMINISTRATION BEFORE THE

Pharmacovigilance. Training session for patients and consumers involved in EMA activities, 25 November Presented by: Priya Bahri

Adaptive Design for Medical Device Development

Mircea Ciuca, MD Global Head Medical & Clinical Drug Safety

Developing a European First-in-Human Study: Three Key Decisions

GxP Auditing, Remediation, and Quality System Resourcing

Office for Human Subject Protection. University of Rochester

Post-marketing Drug Safety Surveillance: Pharmacovigilance in FDA/CDER

Chin Koerner Executive Director US Regulatory and Development Policy

The Promise and Challenge of Adaptive Design in Oncology Trials

YASHAJIT SAHA & ABHISHEK SHARMA, SUBJECT MATTER EXPERTS, RESEARCH & ANALYTICS ADVANCED ANALYTICS: A REMEDY FOR COMMERCIAL SUCCESS IN PHARMA.

COMMITTEE FOR THE MEDICINAL PRODUCT FOR HUMAN USE (CHMP)

Optum Performance Analytics

Regulatory hurdles and opportunities

KINGSMANN CARE GROUP

Maximizing Market Access: THE 5 MOST CRITICAL QUESTIONS TO ASK WHEN LAUNCHING A SPECIALTY DRUGS

Reimagining Life Sciences With AI-Enabled Digital Transformation. Abstract

STATE OF THE ASIA-PACIFIC PHARMACEUTICAL INDUSTRY 2013: EXECUTIVE SUMMARY

Delivering on the promise: the clinical application of new diagnoses and treatments for RD K A T E B U S H B Y N E W C A S T L E U N I V E R S I T Y

Systematic Risk Management: An Overview of ICH Q-9

December 4, Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

HEDIS Measures NCQA s 20 Years of Experience with Measurement

Establishing Case Quality Metrics The Sciformix experience

Updated EU Medical Device Regula2ons: What s In, What s Out, and What s Hot?

Saudi FDA Drug Approval Process. Dr. Mohammed A. Alquwaizani Consultant- Chief of the scientific office Saudi Food and Drug Authority

Decoding Phase II Clinical Trial Terminations

MEDICAL DEVICE CLINICAL INVESTIGATIONS AND ISO 14155

Trinity ConTRact: Cornerstone Projects in Contracting Strategy

Guideline on good pharmacovigilance practices (GVP)

ADVANCING SCIENTIFIC INNOVATION Together, the possibilities are endless.

Safety Measures in the new Pharmacovigilance System

Copyright. Jeremiah J. Kelly (2015). All rights reserved. Further dissemination without express written consent strictly prohibited.

Challenges and Opportunities in the Development of integrated Drug Device Combination Products CMC Strategy Forum, Tokyo, 5 December 2017

The Drug Safety and Effectiveness Network (DSEN) Dr. Diane Forbes, Associate Director, DSEN CAPT 2012 Annual Conference, May 8, 2012

Sonja Brajovic Medical Officer, Office of Surveillance and Epidemiology Center for Drug Evaluation and Research Food and Drug Administration

Appropriate Control Strategies Eliminate the Need for Redundant Testing of Pharmaceutical Products

Explanatory Note to GVP Module VII

Transcription:

PROACTIVE PHARMACOVIGILANCE RISK BENEFIT A New Model for the 21st Century 21st century technologies will create significant opportunities and challenges for all health care stakeholders. Pharmacovigilance too is in transition, with new sources of medical information and methods for its analysis that will transform today s largely reactive system into proactive benefit-risk management for all medication users. The discipline of pharmacovigilance (PV) largely originated in reaction to public health disasters caused by medications, e.g. tetanus toxoid (1912), sulfanilamide (1938), thalidomide (1962), etc. The detection of significant, previously unrecognized hazards of medications following their introduction into clinical use was traditionally based on collation and evaluation of essentially anecdotal reports of suspected adverse drug reactions (ADRs). Subsets of these reports were communicated to regulators, individually and/or in the aggregate, within specific timeframes; however, these regulations were established at a time when modern communication tools and databases did not exist and all communication was on paper, greatly limiting the analysis and evaluation of potential safety signals. Furthermore, regulatory oversight of company safety activities for approved medical products was, until very recently, largely limited to assessing the adequacy of and compliance with the procedures license holders established to meet their legal obligations, rather than on any identifiable public health benefit.

In recent years, the combination of greater public awareness of drug safety issues and emerging informatics-based methods for acquiring and analyzing medical information has sparked global research initiatives for transforming the present largely reactive system into one that is proactive, robust, and more useful clinically. While technologies such as data mining may permit reader identification of rare, medically important ADRs, the detection of clinically important drug-associated events and drug interactions cannot be significantly improved simply by applying increasingly sophisticated analytical tools to ever-increasing volumes of anecdotal data that remains inadequate and flawed. Pharmacovigilance is therefore undergoing a major transition, in which improving both the quality of data sources and the tools used for data analysis pivotal. Post-approval PV will continue to be the mainstay for detecting previously unidentified harms due to drugs after their licensing based on the provisional benefit-risk assessment identified during development. However, one especially challenging scenario is detection of a drug-related increase in the incidence of an event that is relatively common in the unexposed population a classical signal to noise problem. For example, the increased risk of cardiovascular events associated with NSAIDs (nonsteroidal anti-inflammatory drugs) was discovered only in a large comparative clinical trial of a newer drug, having gone undetected by traditional PV methods during decades of widespread NSAID use. Similarly, provisional evidence sufficiently convincing for regulatory action, e.g. the supposed increase in cardiovascular risk attributed to rosiglitazone, may prove unfounded by subsequent studies. Since it is not feasible to conduct large prospective trials for more than very small number of drugs, a principal focus of new PV initiatives is the identification and/or creation of complete, validated data sets reflecting real-world clinical use which can be analyzed to assess the true risks of a drug in very large numbers of patients. Why do we need a new PV model? PV needs to be refocused as a function closely aligned with actual public health benefit rather than its historical primary focus on regulatory compliance concerns conducted by an often isolated safety function within life science companies. For medicines to be properly appreciated as important public health interventions, all their users, especially patients, require a clear understanding of their attributable benefits and harms. Without that understanding, individual prescribers and patients cannot make informed, decisions on what benefits and risks are acceptable. PV must therefore

become a means of evaluating, understanding, and effectively communicating both the real-world benefits and risks for each patient-drug combination, and not merely a mechanism for identifying and approximately quantifying overall population risks. In the near future advances in genomics, epigenetics, metabolomics, microbiomics, etc., will transform our understanding of disease mechanisms and individual patient response factors, both positive and negative, to therapeutic agents. This will provide new qualitative and quantitative tools for clinical decision making and therapeutic choices, especially the comparative benefits and risks of different interventions. PV should be in the forefront of making this explosion of knowledge accessible and comprehensible to every stakeholder in the therapeutic chain. What might effective PV look like in 20 or 25 years? Scientific advances in biotherapeutics will create a shift towards individualization of therapy and away from our historical model of one size fits all, with relatively minor modifications for relatively broad patient sub-populations, e.g. the elderly, renally-impaired, etc. Personalized medicine will use patients specific biological and disease characteristics to tailor therapies for an individually optimized benefit-risk balance. Current PV relies largely on the analysis of relatively large numbers of poorly defined individual patients taking palliative drugs for prolonged durations, and does not support an individualized model of therapeutic decision making. New models will be needed for effective safety evaluation of novel therapies (stem cell transplants, gene therapy, live biotherapeutics, etc.), many of which may involve single or short-duration, narrowly targeted treatments of small numbers of highly selected patients in whom adverse effects may be delayed for very long periods. In this setting, effective therapeutic interventions will require accurate, reliable, and comprehensible information on benefits and risks. When you can measure what you are speaking about, and express it in numbers, you know something about it; but when you cannot express it in numbers, your knowledge is of a meager and unsatisfactory kind (Lord Kelvin). One major challenge for PV will be to develop tools for accurate qualitative and quantitative assessment of both benefits and risks using data that will differ greatly from that used historically. Safety & Risk & Risk Management Clinical Research & Post & Post Approval Support Regulatory Affairs Affairs

New data sources Regulatory agencies have been at the forefront of using electronic health record data and developing signalling and analytical tools for active surveillance of real-world clinical information to supplement or supplant today s largely anecdotal safety data. The European Network of Centres for Pharmacoepidemiology and Pharmacovigilance (ENCePP) comprises over 170 research centers and health care data providers, with a comprehensive, searchable database and a registry of data sources for post-authorization studies. In the US, FDA and the National Institutes of Health established the Observational Medical Outcomes Partnership to research analytical methods for systematic, proactive, and cost-effective use of electronic medical databases. The FDA also created the Mini-Sentinel system for active surveillance, which links multiple healthcare data sources; this system contained >130 million individual patient records as of December 2012. New safety signals may be identified or communicated via the web, social media, "cloud" storage etc. Early examples include analysis of web searches to identify influenza epidemiological trends and data-mining of free-text clinical notes for hypothesis generation and analysis of suspected ADRs. There are several commercial developers and providers of various types of software for analysis of clinical "big data". The multiplicity of disparate data sources requires new methods, analytical tools, and standardization of attributes including benefit and risk outcomes, terminologies, data structures, signal triage, privacy protection, etc., which are presently at a very preliminary, exploratory stage. Once validated, such standards and tools should allow more accurate determination of drug benefits and risks in actual clinical practice than the limited information derived from controlled trials. Multivariate analysis will identify patient sub-populations in whom the benefit-risk balance is optimal or unfavorable, permitting assessment of true clinical practice effectiveness, as opposed to today s "two adequate well-controlled trials" required to prove statistical efficacy in a limited patient population. Additionally, these analyses should permit actual clinical usage comparison of the benefit-risk balance for multiple treatment options which is now almost completely lacking.

Better clinical decision making Marketers, regulators, and dispensers of therapeutic products are obligated to ensure that reliable, actionable clinical information is effectively communicated to prescribers and users to allow informed therapeutic choices based on balancing benefits and risks. The ever-increasing flood of medical information far exceeds individual human capacity to assimilate and apply it. Passive dissemination of minimally structured information, such as text-based prescribing information, is insufficient to provide robust patient protection; rather, sophisticated decision support systems, which are presently at a very early developmental stage, are needed to help prescribers choose appropriate medications based on patient and drug-specific characteristics. It is also insufficient for such information to be available and comprehensible only to health care providers; patients and care givers need ready access to relevant information in an intelligible and actionable format. Greater understanding of the drivers of prescriber and patient behavior and factors that influence them is necessary for successful interventions to improve the understanding of benefits and risks. This will require research into the sociological and psychological bases of such decision making, the development of incentives and methods for data-driven risk prevention, and reorientation towards prophylaxis of today s reactive, illness-focused, intervention-driven medical systems. Patients and care givers will need to be active, health-literate participants in both regulatory and individual decisions regarding acceptable medication use.

Broad accessibility of appropriate information Open access to benefit-risk information for both investigational and marketed drugs is steadily increasing. Spontaneously reported adverse event data from the FDA safety database has been publicly available for many years, although not in a readily accessible or usable format, while comparable data from the EU is now directly accessible via the web. FDA and EU public websites list ongoing clinical trials, FDA posts the most frequently occurring clinical trial adverse events, and there are initiatives to make summary and even raw study data available appropriately. EU Pharmacovigilance Risk Assessment Committee deliberations are posted on the web, and FDA publishes information about ongoing activities such as unconfirmed safety signals under going evaluation. These actions are intended to improve clinical decision making and enhance public confidence in the system by making source data and benefit-risk assessments available and transparent. However, this is predicated on the users' ability to evaluate both benefits and risks accurately, and the impact, if any, of this level of transparency on actual health outcomes remains to be determined. Cost-benefit considerations Future PV activities will require demonstrable cost-effective public health benefits, and regulatory trends towards bureaucratization and global disharmonization must be reversed. Drug development costs have been increasing steadily for many years, while the pipeline of new products continues to shrink despite increasing clinical need, e.g. for infectious diseases and degenerative disorders in an increasingly elderly population. The business model in which giant global companies develop small numbers of highly profitable blockbuster products taken long-term by millions of patients for disease palliation, rather than cure, will become less common. It is reasonable to assume that multiple smaller companies with specifically focused research expertise will emerge to develop specialized therapies for much smaller numbers of highly selected patients. Such companies will generally not have the in-house expertise to navigate the global regulatory maze, and will

almost certainly need support from external providers or partners with that expertise. The trends will create greater difficulty, and fewer opportunities, to recoup expenses and generate profits, which will in turn require new models of drug development and reimbursement. All stakeholders - patients, health care providers, payers, regulators, and industry - will need to be fully engaged to minimize resource limitations and eliminate unnecessary costs. Demographic changes, including global population growth, increased prevalence of disorders associated with western lifestyles, emerging infectious diseases, and the increasing proportion of elderly people who are both more susceptible to ADRs and use more medications, will also alter perspectives on cost-benefit-risk. Future PV activities will need to demonstrate cost-effective public health benefits, and, recent regulatory trends towards increased bureaucratization and reduced global disharmonization must be reversed as quickly as possible. Conclusion The impact of 21st century technologies on health care delivery will create significant challenges and opportunities for all stakeholders in life sciences. PV is currently in transition, with new sources of medical information and methods for its analysis being explored that will transform the current reactive system into proactive benefit-risk management using emerging medical information technology. Regulations are gradually evolving towards a more proactive approach, but remain too procedurally focused, often inhibiting innovation, while the technical resources required for effective benefit-risk assessment are still at a nearly development stage. In summary, PV is entering uncharted territory requiring industry, regulators, health care providers, and patients to address multiple unknowns and should eventually lead to greatly enhanced benefit-risk assessment, communication and implementation. References 1. Kahn, S. (2013). Proactive pharmacovigilance A new model for the 21st century. International Pharmaceutical Industry, 5(3), pp. 62-66 2. Lele, C. (2013). A successful path to fulfilling pharmacovigilance obligations. Manufacturing Chemist, 84(7/8),pp. 24-26

Sciformix Sciformix Corporation is a global scientific process organization (SPO) that partners with life science companies to develop, launch and sustain medical products that aim to improve the quality of healthcare worldwide. We collaborate with our clients through the entire product development lifecycle to provide a full range of services from study design to post marketing surveillance and commercialization support. Sciformix consistently delivers scientific insight, improved productivity, and high quality results, in every engagement, through a deep understanding of the regulations governing the global life science industry. Sciformix Corporation 1500 West Park Drive, Suite 210 Westborough, MA 01581 USA Phone : 1 (877) 576-5005 Fax : 1 (508) 302-6520 Email : bizteam@sciformix.com