Blog Layout

Safety Risk Management in Research - Part 2 of 8

Michael Heald • Dec 23, 2019
Risk Mañana (noun): The temptation to leave risk management until is it impossible to ignore.  Invest in an early  dry run and enjoy the benefits.
Risk management is not just something to worry about during development, it lives on through launch and right up until the last product has been used after it has been taken off the market.  Decisions made early in development about risk scoring and evaluation live with the product, and the manufacturer must stand by those decisions the entire time that the product is on the market.  If the risk bar is set too low to expedite development for the sake of a launch date, imagine having to defend it when people get hurt by an under-developed product on the market? Set it too high and costly over-engineering or even project failure may result.

The aim of risk management is to ensure that the risk of using a device is always outweighed by the benefits of using the device – a simple enough concept.  As development marches on, risk assessments are made to ensure that the device remains on the benefit side of the benefit-risk balance, but where is that tipping point for a specific product?  If it is mis-judged because someone had to make some hurried decisions to get the risk management plan out, the consequences of subsequent rework and back-peddling could be very painful.

So, when can you test and refine your risk scoring and evaluation system before committing to it in development?  In the research stage of course.  During concept development, or bought-in platform device selection, it is the perfect time to size scoring systems and test your benefit-risk tipping point.  Apart from providing an opportunity to calibrate your risk management set-up, risk profiling should be central to the concept/buy-in selection strategy anyway.  One of the responsibilities of the device engineering team should be to make sure that bought-in device selection for development is not just device Tinder; it should be the result of objective assessment, including risk profiling.

An interesting parallel to bough-in device selection is when a pharmaceutical generics manufacturer is selecting a branded drug to copy as a generic.  Sometimes the branded drug is marketed with a delivery device, the design of which which may be rather old by the time the drug patent is expired. In order to market the generic version of the drug the manufacturer often has to ensure that the generic drug delivery device looks-like and works-like the branded one.  Here is the dilemma for the device development team, because the risk bar for development of the generic device must be set to reflect current state-of-the-art safety levels.  Consequently, the device team can have an important role to play in any decision to develop a generic version of a branded combination product.

Part 3 – What is Acceptable Risk?
By Michael Heald 18 Oct, 2023
While the FDA’s Office of Combination Products is working on a formal definition of Essential Performance Requirements behind closed doors, Industry is busy second-guessing. There are some clues to be had, and this is how I think it can work…
By Michael Heald 23 Dec, 2019
It’s hard to argue with safety as a reason for doing, or not doing, something; unacceptable risk is, well, unacceptable. This is the last article on risk management from me for a while – thanks for reading.
By Michael Heald 23 Dec, 2019
To misquote Edwin R. Fisher, “In god we trust, all others must provide documented evidence”. Just like a list of new year’s resolutions, a list full of risk controls does not mean anything unless they are acted upon. Fortunately, we don’t have to provide documented evidence that we have stuck to our new year’s resolutions.
By Michael Heald 23 Dec, 2019
Keep your friends close and your enemies closer. While all aspects of design and manufacture of a medical device should be carefully controlled, special attention should be given to those elements that have the potential to cause the most harm.
By Michael Heald 23 Dec, 2019
Pressing deeper into safety risk management we look at two tools for performing risk assessment and evaluation: the ubiquitous FMEA and the exotic FTA.
By Michael Heald 23 Dec, 2019
Imagine a band performing a piece in which every musician played a different tune. In my fourth article I’m looking at how to coordinate the disparate groups that have to work under one risk management plan. How can they be helped to assess risk consistently?
By Michael Heald 23 Dec, 2019
I admire the achievements of free solo climbers, but I do question their notion of acceptable risk. Modern office health and safety culture operates at the other end of the scale; with carefully sealed hot drinks containers and the mandatory use of handrails on stairs. So, what level of risk is going to be acceptable to the users of your medical device?
By Michael Heald 23 Dec, 2019
This is the first of a short series of articles written to share my insights into the bit of medical device development that is always last to get picked for the team.
By Mike Heald 03 Dec, 2019
Medical devices designed to inject sterile liquid typically contain a cartridge or pre-filled syringe, usually made from glass, and its job is to contain and protect the sterile drug solution. These glass containers, or primary packs, also have plungers and seals made from rubbery elastomeric materials. As long as the glass and rubbery bits form a hermetically sealed environment, nothing nasty can get in, and nothing precious can escape. For those in the industry this simple concept is lovingly referred to as Container Closure Integrity, or CCI. Consider the life of a primary pack. Once it has been proven that a filling process is reliably producing sterile primary packs, they can be sent to be assembled into their host drug delivery devices. Device assembly is no pampering session for the primary packs; they are slid down tracks, bump into each other, get gripped and handled by machinery, pushed into some sort of holder, and then they get squeezed and bumped some more during other assembly processes. After all this, are they still safe to use? Then there is the shipping process as the devices are moved around the world. Happily, they are not usually subject to the professional ministrations of ordinary airport baggage handlers, nonetheless temperature changes, pressure changes and impact forces must all be endured en-route to their new home. Still safe? Eventually the devices will be stashed somewhere, during which time the primary packs age gracefully inside their host device and packaging. Even now they can fall victim to environmental conditions, or even long term stresses placed on them by the device. Filling and sterility testing is but a distant memory. It’s pretty obvious that we need to look after CCI right up until the patient gets their jab, and there is some common sense regulation on the subject. With device assembly, shipping and ageing in mind, try these for size: 21cfr211.87 “… drug product containers, and closures shall be retested … after exposure to … conditions that might adversely affect the … drug product container, or closure.” 21cfr211.94 (b) “Container closure systems shall provide adequate protection against foreseeable external factors in storage and use that can cause deterioration or contamination of the drug product.” FDA Guidance - Submission Documentation for Sterilization Process Validation and Applications for Human and Veterinary Drug Products: “Study designs should simulate the stresses of the sterilization process, handling, and storage of the drug and their effects on the container closure system.” Glass can form micro-cracks, or worse, and rubbery bits can fail to seal properly. So the big question is; when manufacturing thousands, or even millions, of drug delivery devices, how do we demonstrate that each and every primary pack will continue to keep the bad stuff out and the good stuff in until it’s used? The answer to our big question lies in a specialist field of testing called Container Closure Integrity Testing (CCIT). The old probabilistic dye ingress techniques are, quite frankly, a bit rubbish. Luckily, a growing number of clever quantitative techniques are available to prove reliable container closure integrity for a range of different applications. Don’t think for a moment that device design is not involved in CCIT. Firstly, the device simply gets in the way and prevents us from checking CCI (think Schrödinger's cat). Secondly, if the device is dismantled to check CCI, the act of dismantling may itself cause a leak path to develop. So, to all those engaged in the design and development of drug delivery devices; don’t design a CCIT nightmare, use Design Controls and Risk Management to both improve CCI performance and pave the way for CCIT friendly device design.
By Michael Heald 02 Dec, 2019
When devices are being developed for the delivery of pharmaceutical drug, two worlds collide. Pharmaceutical regulation meets medical device regulation, and what is at ground zero? The primary pack*. The bit of the device that holds and protects the drug substance. Often the device itself is referred to as secondary packaging, which hardly does justice to the amazing engineering that can go into it. Then there are more ordinal layers of packaging with trays, cartons, sleeves, wraps, multipack boxes etc. Beyond Primary Pack there seem to be no hard and fast rules for packaging nomenclature, so it is a good idea for everyone to agree on packaging terminology early in a project. But I digress. Going back to the Primary Pack, I should point out that containers designed to do no more than hold and protect a drug substance are not regulated as devices, and therefore are not subject to medical device regulation. As soon as a container plays a part in controlling the delivery of a drug, it is moving into medical device territory, and the medical device regulations are invoked. There are, of course, grey areas. Eye dropper bottle design seems to be one. From a regulatory point of view, and the Americans are particularly clear on this, you need to run both drug and device quality systems for drug delivery devices. There are drug constituent parts, and there are device constituent parts. And then there is the Primary Pack, which is both. It is the common interface between device and drug. For the pharma development team, the Primary Pack has to hold the right amount of drug, and it needs to prevent deterioration or undesirable exchange between the drug, primary pack and environment, all for the entirety of its shelf life. The contents may also need to be kept sterile. For the device development team, the Primary Pack has to allow accurate, often metered, delivery of the drug. To do this the Primary Pack becomes an integral part of a precision mechanism, which necessitates tight control over dimensional tolerances and operating forces. There are also shared interests, such as drug visibility, labelling, filling requirements and container closure integrity. Shared interests further extend to compatibility with certain manufacturing processes, so someone from Manufacturing needs to get involved too. Put simply; the pharma development team and the device development team need to collaborate closely on the design of the Primary Pack from the start. It is surprising, especially in parenterals, how often project risk is unwittingly invited by one-sided Primary Pack decision making. So which team should have custody of the Primary Pack specification? I think that the right answer to this question is that it doesn’t matter. As long as the controlling team and the governing processes ensure that collaboration is effective, and that both drug and device regulations are met, then ownership is irrelevant. Traditionally the pharma guys have it. One particularly tricky aspect of developing devices for sterile injection is container closure integrity testing (CCIT). Not very long ago CCIT was subject to a pharmacopeia re-boot with USP <1207> and the industry is upping its game by abandoning old probabilistic methods and moving to deterministic ones. This field of testing is evolving quickly, which is why went along to the Container Closure Integrity Testing workshop run by PDA Europe in March. If you have read this far, maybe you would like to take a look at the CCIT blog that resulted. * Apart from the Primary Pack, other parts of the drug delivery device can demand a similar mode of co-operation. Typically, these are other components that are in direct drug contact, though usually contact duration is transient instead of being measured in months or years.
Share by: