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

Similar documents
Regulatory perspectives on CQAs, CPPs, and Risk Analyses for Combination Products.

Systematic Risk Management: An Overview of ICH Q-9

The Device Side of Combination Products

EU Annex 11 US FDA 211, 820, 11; other guidelines Orlando López 11-MAY-2011

INTRODUCTION TO MODERN RANDOMIZATION AND TRIAL SUPPLY MANAGEMENT SERVICES

Draft Guidance for Industry Development and Use of Risk Minimization Action Plans

FDA Medical Device HFE Guidance

TECTURA LIFE SCIENCES HIGHLIGHTS

Enabling faster insights to improve clinical trial efficiency and quality

EU Update on Regulatory Developments

Quality Agreements and Managing Contract Supplier Quality

Industry Experience: Early Collaboration with FDA on Combination Products. Kristi Kistner, Amgen Inc. CMC Strategy Form January 26, 2015

Office for Human Subject Protection. University of Rochester

Regulatory Challenges of Global Drug Development in Oncology. Jurij Petrin, M.D. Princeton, NJ

ICH Q3D Guideline Impact on the Users: Perspective of a Finished Product Manufacturer John Glennon

THE NEW QUALITY PARADIGM OPPORTUNITIES AND EXPECTATIONS IN ICH Q8 Q9 Q10 Q11 DR. FRITZ ERNI

Planning of Application

Industry Perspective on Manufacturing in Early Development

Mitigating Medical Device Risk through Human Factors. Christina Mendat, PhD Director, Research & Human Factors

CRO partner in Rx/CDx Co-Development

WHO GUIDELINE Stability testing of active pharmaceutical ingredients and finished pharmaceutical products

References Concept. Principle. EU Annex 11 US FDA , (g), (i), 11 Orlando Lopez 2/15/11. Old Annex 11.

PhUSE Single Day Event 2009, Frankfurt

Submission of comments on 'Reflection paper on the use of extrapolation in the development of medicines for paediatrics' (EMA/199678/2016)

Validation Requirements for Nutrition Products

Risk-Based Approach to SAS Program Validation

ICH Q11 Development & manufacture of drug substances

Session 7 Clinical Trial Assessment Bioequivalence Studies

Regulations governing the application of medical accelerators

APPLICATION OUTSOURCING. for life sciences

Pharma EXPO 6 9 November 2016 Chicago, IL

Challenges in Getting Global Approvals for Post-approval Changes

Current FDA Perspective for Continuous Manufacturing

Demonstrating a High Degree of Assurance in Stage 2 of the Process Validation Lifecycle

Type of Activity. Universal Activity Number L04-P

QuEST Forum. TL 9000 Quality Management System. Requirements Handbook

Food Beverage COMPLIANCE READINESS

Hot Topics in Drug Product Process Validation: A Reviewer s Perspective

Vendor Selection: Identifying Effective Procedures & Strategies

COMMERCIAL PRODUCT STABILITY

An Industry Perspective: Reducing the Complexity and Impact of Regulatory Changes in Latin America

PHARMACEUTICAL TESTING

A COMPARATIVE FRAMEWORK BETWEEN NEW PRODUCT & LEGACY PRODUCT PROCESS VALIDATION

Pre-Clinical Testing for Devices and Diagnostics

DESIGN AND DEVELOPMENT

Implementing Compliant Medical Device Best Practice Business Processes Using Oracle E-Business Suite

Challenges in Implementing Knowledge Management within ICH Q10

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

Joint WQHO/UNICEF/UNFPA suppliers meeting

Pharmacy. Medication. Checks

Medical Electronics: Risks and Opportunities for Electronics Manufacturers. by Stephen Las Marias

Spectroscopy Solutions for Pharmaceuticals:

The interface between Good Clinical Practice and Good Manufacturing Practice

Challenges of Analytical Method Transfer in the Pharmaceutical Industry

MEASURE FOR MEASURE: QUALITY METRICS

Extractables and leachables: An Introduction

Elemental Impurities: An Industry Perspective

Good Clinical Practice (GCP) & Clinical Trial Registries

Copyright GCI, LLC Remediation of Legacy Medical Devices

Application of PAT for Tablet Analysis. Case examples from Novartis Lorenz Liesum, Lead PAT Hamburg, 19 th of April 2013

MEDICAL DEVICE CLINICAL INVESTIGATIONS AND ISO 14155

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

Draft agreed by Scientific Advice Working Party 5 September Adopted by CHMP for release for consultation 19 September

Medical Device Product Development for Startups

ISPE S GUIDES AND HOW THEY APPLY TO CLEANING AND CLEANING VALIDATION

Data Quality and Integrity: From Clinical Monitoring to Marketing Approval

The Risk Management + Design Controls Connection: What Device Makers Need to Know

CMC Challenges for cell therapy manufacturing

Launching drug product in Europe Q&A

ISO INTERNATIONAL STANDARD. Risk management Principles and guidelines. Management du risque Principes et lignes directrices

Building a Hosted Statistical Computing Environment: Is it Possible?

USP <797> Compliance Common Challenges and Potential Solutions

Intralinks Studyspace : Electronic Investigator-Controlled Site File (eisf)

QbD (Quality by Design) Has industry benefited from this? WHITE PAPER.

MARKETS & CUSTOMERS. Progressing globalization. Evolving markets

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

Revision. Quality Manual. Multilayer Prototypes. Compliant to ISO / AS9100 Rev C

Use in Process Characterization. Nathan McKnight

Five years as EMA Liaison at US FDA

Sensile Medical. Sensile Medical achieves solid growth with Siemens PLM Software solutions and Quadrix

Q10 PHARMACEUTICAL QUALITY SYSTEM

Structure and content of an IMPD. What is required for first into man trial?

Visions on the role of Statisticians in the Pharmaceutical Industry. Merete Jørgensen President EFSPI VP Biostatistics, Novo Nordisk A/S

Novartis E2E CM case study

Introduction to CMC Regulatory Affairs

Briefing Book Guidance for Company

Digital Solutions for the Life Sciences Industry The connected product and manufacturing concept

10 Things You Should Do Before You Validate Your Next Package

How to support compliance with GAMP 5

Oracle Buys ClearTrial Adds Leading Cloud-based Clinical Trial Operations and Analytics Applications to the Oracle Health Sciences Suite

Application of Quality Risk Management Tools for Cell Therapy Manufacturing

IS BIOSIMILARS COMPETITIVE INTELLIGENCE DIFFERENT?

Textvergleich. Verglichene Dokumente MEDIA3917.pdf. ICH_Q10_Step4.pdf

Questions And Answers To Support The

DEVICES ARE KEY TO DRUG EFFICACY

Emerging Trends in Regulatory Outsourcing WHITE PAPER.

GMPs: Distribution Centers

At the Heart of Connected Manufacturing

Transcription:

Challenges and Opportunities in the Development of integrated Drug Device Combination Products CMC Strategy Forum, Tokyo, 5 December 2017 F. Wildenhahn, Human Factors Engineering & U. Grauschopf, Head of Device Development Device Development, Hoffmann-La Roche, Basel, Switzerland

Introduction Device requirements in a Pharma environment Challenges Dynamic development programs Evolving regulatory expectations Complex supplier networks Opportunities

Pharma versus Device Extractables & Leachables Biocompatibility Clinical study patient Stability Testing Quality by Design ICH C9 vs. Human Factors study user Verification Testing Design Controls ISO14971

Pharma versus Device Extractables & Leachables Clinical study patient Stability Testing Quality by Design ICH C9 Biocompatibility Human Factors study user Verification Testing Design Controls ISO14971

Stability vs. Design Verification Legal Manufacturers are required to ensure device functionality by Design Verification, i.e. by demonstrating that Design Input Requirements are met. This may be done by testing, tolerance analysis, mathematical modeling or certificate review This may be, and often is, outsourced to suppliers, but it remains within the responsibility of the legal manufacturer, with respect to the adequacy of e.g.: Acceptance criteria Sample size Test methods Quality system

Stability vs. Design Verification typically small sample size statistically significant sample size Stability Testing Design Verification inter-batch variability extrapolated intra-batch variability rarely extrapolated

Clinical studies vs. Simulated use studies Use errors are a known source of risk for any medical device and therefore need to be understood and managed. Simulated-use Human Factors studies are an adequate (and safe!) way of studying user behaviour and provide insights on use errors a clinical study in many cases could not provide.

Clinical studies vs. Simulated use studies Clinical data Higher risk Clinical trial Behavioral data Lower risk Human Factors study Quantitative results Statistically powered Semi-quantitative results Not statistically powered

Dynamic programs Why is it a challenge to be agile and respond swiftly to changing requirements of dynamic development programs?

Dynamic programs Potential drivers, a few examples The program is accelerated, due to breakthrough therapy designation Clinical trial results prohibit home-use Indications are added or dropped from the development program Competitive intelligence mandates a change in strategic direction altogether

Dynamic programs Example 1 - Late Design Input Drug and device characteristics have the potential to impact each other but with the primary mode of action being the drug constituent part, it is typically the device constituent part that will be adjusted. For delivery devices of parenteral drugs, critical Design Input becomes available rather late. Viscosity Dose accuracy Fill Volume Shear stress Materials Light exposure

Quick detour: Design Controls For Device Development activities a specific methodology is mandated by ISO13485 (EU) and 21CFR820 (US). Design Input is supposed to be established at the beginning of that process. Any change to that initial element will need to be traced through the entire development documentation. USER NEEDS DESIGN REVIEW DESIGN INPUT DESIGN PROCESS VERIFICATION DESIGN OUTPUT VALIDATION MEDICAL DEVICE

Dynamic programs Platforms and their limitations One way to address this challenge is by establishing technology platforms. With these standardized technologies generic development work can be frontloaded, e.g. by non-molecule specific Design Verification testing. This also may offer the opportunity to leverage data across programs. But: The device that works for everything will likely not be ideal for anything. Generally a nimble quality system, well embedded processes and (flexible) templates will help to minimize the development timelines.

Dynamic programs Example 2 Changing indications Often the mode of action of a particular drug allows the treatment of different diseases. Patient & User Pharma companies therefore regularly expand the label of approved medicines to new indications. The process to justify these additional indications is well established from a clinical perspective. There is little involvement from technical development, as long as the primary packaging does not change. For a device (and a combination product) the indication is relevant Design Input. A change may impact the entire Design History File and requires diligent assessment. Dosing Regime Injection Site Experience User capability Education Environment Lifestyle Other diseases

Dynamic programs Example 2 Changing indications What could go wrong? And how often? Any difference between the established and the new indication will be evaluated to understand, whether the risk profile changes. SEVERITY (S) PROBABILITY (P) RISK Probability hazardous situation leads to harm Probability failure leads to hazardous situation Probability a failure or use error occurs

Dynamic programs Strategies to deal with it Build device awareness throughout the organization. Ensure that relevant information (i.e. Design Input) is made available as early as possible. Standardize to an extent feasible, i.e develop platforms on different levels as well as templates.

EVOLVING REGULATIONS Why can it be a challenge to keep up with regulations that are constantly evolving?

Evolving regulations Example 1 Risk management Roche had used descriptive categories for probability of harm in device FMEA s. Score Category Criteria 10 Very high Certain to occur routinely 8 High Occurs frequently 6 Moderate Occurs occasionally 4 Low Has not occurred often 2 Remote Not expected to occur Probability of harm (ppm) > 6210 6210 > 1350 1350 > 233 233 > 3.4 3.4 This, while previously accepted, was criticized in a pre-approval inspection as inadequate. Roche then changed procedures to quantitative categories for probability of harm.

Evolving regulations Example 1 Risk management Ultimately this resulted in a more coherent and robust risk management file, that facilitates communication and is easier to defend, for example in respect to: Direct traceability of Human Factors study results to the usability FMEA. Assessment of Design Verification issues to determine whether these result in unacceptable risk However, establishing this approach required significant effort and adds considerable complexity to the risk management process.

Evolving regulations Example 2 Untrained user data For home-use medication administered by self-injecting patients, Roche s intended use statement typically prescribes that users will have to be trained. This training is reflected in HF validation study protocols, an approach consistent with previous health authority guidance. In a recent HF validation study, the protocol was submitted to the authority for review and accepted in 2015.

Evolving regulations Example 2 Untrained user data In the pre-submission meeting about a year later where the HF validation results were supposed to be discussed it was revealed that now data from untrained users would be required as well. Ultimately another, supplemental study was required. Roche will collect data on untrained users in future HF studies but maintains that this is to be considered (and evaluated as) a foreseeable misuse scenario, if training is defined in the intended use of the product.

Evolving regulations Strategies to deal with it Engaging with health authorities early on and frequently will help to avoid (most) surprises. Taking ownership of the entire risk management file will enable swift, consistent and transparent response to risk profile changes.

RELIANCE ON SUPPLIERS Why can it be challenging to rely on a vast network of external suppliers and service providers?

Reliance on suppliers Background Even in many large Pharma companies a significant part of the device development work is outsourced: Mechanical engineering Prototyping Process development Usability testing Strategy consulting Etc. Customers will have to rely on program management, technical expertise, documentation and compliance.

Reliance on suppliers Example Raw material changes Case study: Change of raw material lead to an increase of insertion torque for a plunger rod. Several potential root causes were identified and correction measures explored: process parameters adjusted lubricant added to resin mold polishing applied Gap between stopper and rod Ultimately the use of CT scan enabled identification of thread dimensions of the plunger in its compressed state.

Reliance on suppliers Example Raw material changes Based upon these CT scans the best matching thread design of the plunger rod was identified. Thread design was fine tuned by samples from inexpensive trial tools. By use of a plunger rod surrogate a fast first check of functionality could be performed. Supposedly minimal changes may require a lot of and very close collaboration between supplier and pharmaceutical company.

Reliance on suppliers Strategies to deal with it Enough internal expertise is required, to be able to make informed sourcing decisions to assess the adequacy of the services provided to build a mutually trustful relation

Summary Is it really worth the effort? Development of combination products presents many challenges. Build awareness Dynamic development programs collide with a very sequential Design Controls methodology. Evolving regulationsdemand constant adaptation and make it difficult to establish consistent and effective processes. Reliance on suppliers requires an indepth understanding of device technologies and associated regulatory principles. Health authority interaction Standardize components and documentation

Opportunities Minimize medication errors Improve adherence Provide users with a better experience

Acknowledgements Ulla Grauschopf Stephen Hyland Markus Hemminger Sherri Biondi Chin-Wei Soo

Doing now what patients need next