Building Trust Through Quality: How QMS Powers Manufacturing Excellence in Life Sciences

16 October 2025 | Thursday | Analysis


From medical devices to cell and gene therapies, a robust Quality Management System (QMS) is redefining manufacturing by uniting compliance, innovation, and patient safety—anchoring a global culture of precision, accountability, and human-centric excellence.

Life sciences sector, a robust Quality Management System (QMS) has become the cornerstone of manufacturing excellence. This is especially true for the medical device and biopharmaceutical industries, where precision and safety are paramount. From advanced medical gadgets to cutting-edge cell and gene therapies, products that transform patients’ lives are born not just from scientific innovation, but from a culture of quality. An effective QMS provides the structured yet flexible framework that helps companies consistently deliver high-quality, safe, and effective products. It does so by embedding quality into every step of the product lifecycle, ensuring compliance with strict regulations while also fostering innovation and efficiency. The result is a win-win: regulators and patients gain confidence in the products, and manufacturers achieve greater operational performance and trust in the marketplace. In an era of personalized medicine and next-generation therapeutics, a human-centric approach to quality – one that keeps the patient’s well-being at the heart of every decision – is more critical than ever.

QMS as the Foundation for Regulatory Compliance

Regulatory compliance is non-negotiable in healthcare manufacturing, and QMS is the backbone that supports it. In highly regulated domains like medical devices and biopharmaceuticals, authorities around the world require manufacturers to implement rigorous quality systems to ensure products meet safety and efficacy standards. A well-implemented QMS aligned with international standards (such as ISO 13485 for medical devices or ICH Q10 for pharmaceuticals) provides a comprehensive framework that keeps companies “audit-ready” and in adherence with global regulations. For example, ISO 13485 is recognised worldwide as the benchmark for medical device QMS, and aligning with it helps manufacturers comply with the U.S. FDA’s Quality System regulations as well as Europe’s Medical Device Regulation (MDR). By establishing documented procedures for design controls, risk management, process validation, and post-market surveillance, a QMS ensures that every product is developed and produced under controlled conditions with full traceability. This greatly reduces the likelihood of defects, failures, or recalls, which is vital for patient safety.

Importantly, a strong QMS also streamlines the path to regulatory approvals across different regions. In the United States and Europe, regulators operate under relatively unified frameworks (e.g. FDA for the U.S., EMA for the EU), whereas the Asia-Pacific landscape is fragmented into multiple national regulatory systems. Here, an overarching QMS can serve as a unifying force – companies can establish core global quality standards and then adapt them to local requirements. In practice, this means maintaining one harmonised quality system that integrates region-specific rules (for instance, Japan’s PMDA guidelines or China’s NMPA regulations) under its umbrella. Such an approach was proposed in a point-of-care cell therapy model, where a central QMS provided general quality standards worldwide, with regional QMS layers to meet local authority requirements, thereby achieving both compliance and global consistency. This strategy requires upfront investment in harmonising quality processes, but it “will result in increased efficiency and cost reduction” in the long run by avoiding duplication of efforts. As international harmonisation efforts progress – exemplified by initiatives of the International Council for Harmonisation (ICH) and programs like the FDA’s CoGenT (Collaboration on Gene Therapies) Global Pilot – companies with mature QMS find it easier to navigate differing regulations and bring therapies to market faster.

Leading organizations across the globe have embraced QMS as a strategic tool for compliance. In the U.S., for instance, the FDA has even proposed aligning its device Quality System Regulation with ISO 13485 to better harmonise with international standards, reflecting how critical a robust QMS is for global market access. European manufacturers, under frameworks like EU MDR for devices and EMA’s guidelines for advanced therapies, rely on their QMS to compile the exhaustive technical documentation and audit trails these regulations demand. Meanwhile, in Asia, regulatory agencies are rapidly updating quality guidelines for advanced therapeutics – China’s CDE (Center for Drug Evaluation) released new guidelines in 2023 to expedite cell and gene therapy development, and Japan’s regulators have pioneered risk-based approvals for regenerative medicine. In all cases, companies with solid quality systems have a head start in meeting these diverse requirements. They can confidently demonstrate to regulators that “safety and compliance are built into the process”, easing approval hurdles. The global players in this industry – from medical device giants like Medtronic and Terumo to biopharma leaders like Novartis and Pfizer – invest heavily in QMS not just as a legal checkbox, but as a competitive advantage. It is telling that key success factors in the cell and gene therapy CDMO sector include technological prowess and regulatory compliance capabilities, and top firms like AGC Biologics, Fujifilm Cellular Dynamics, WuXi AppTec and others pride themselves on their rigorous quality systems to attract business across the U.S., Europe, and Asia.

Fostering Innovation and Continuous Improvement

One might think that strict quality systems stifle creativity, but in practice a well-designed QMS supports and even accelerates innovation. By providing clear guidelines and controlled processes, a QMS creates a “safe space” for R&D teams to explore new ideas without compromising on quality. As one industry perspective notes, “a well-structured QMS provides a clear framework that supports innovation by outlining regulatory expectations and quality benchmarks”, allowing developers to innovate within known safe parameters. In other words, quality processes set the guardrails that keep experimentation and product development on track – enabling innovation to happen in a structured way where risks are assessed and mitigated early. The ICH Q10 pharmaceutical quality system guideline explicitly states that implementing QMS across a product’s lifecycle should “facilitate innovation and continual improvement” while strengthening the link between development and manufacturing. We see this in practice with approaches like Quality by Design (QbD), where product and process are designed from the outset with quality targets and critical parameters in mind. QMS-driven tools (e.g. risk assessments, design reviews, change control procedures) ensure that novel ideas are vetted systematically, so that by the time a new medical device or therapy reaches production, it is backed by robust data and understanding.

Continuous improvement is another hallmark of innovation that QMS embeds into an organisation’s culture. Standards like ISO 13485 and ICH Q10 require ongoing monitoring of processes, customer feedback, and internal audits to identify opportunities for improvement. This means companies are constantly learning and adapting – for example, using CAPA (Corrective and Preventive Action) systems to not only fix problems but prevent their recurrence, or analysing production trends to refine processes. Over time, these incremental improvements add up to significant leaps in performance and product capability. A QMS can thus turn a company into a learning organisation, where every deviation or customer complaint is treated as valuable input to make the product better. Many firms also credit their quality system for speeding up innovation cycles: with controlled document systems and defined workflows, they can iterate designs more quickly and transfer knowledge from development to manufacturing smoothly. In fact, an automated or digital QMS can “foster innovation at the speed required to compete on a global scale”, by streamlining quality processes and closing feedback loops faster. Leading medical device companies have found that embracing modern electronic QMS platforms allows them to develop cutting-edge products without getting bogged down in paperwork, thus marrying compliance with agility.

Crucially, a culture of quality encourages teams to be proactive rather than reactive. Engineers and scientists are empowered to ask “How can we make this process more robust or this product safer?” early in development, rather than firefighting problems later. For example, a biopharmaceutical company implementing ICH Q10 principles might integrate its R&D and manufacturing teams under the quality system, so that process scalability and quality controls are considered from day one. This integrated approach was seen in the cell therapy field where developers employed QMS-guided risk assessments and knowledge sharing to transition lab-scale innovations into clinically compliant manufacturing processes. By facilitating knowledge transfer and capturing institutional learning (through documentation of why certain decisions were made and what pitfalls were encountered), the QMS actually spurs smarter innovation. In essence, quality management and innovation go hand in hand – the former provides the foundation of reliability and trust, upon which creative new solutions can safely be built.

Enhancing Operational Efficiency and Manufacturing Excellence

Beyond compliance and innovation, QMS plays a pivotal role in boosting operational efficiency. A common saying in manufacturing is “do it right the first time” – a philosophy that a good QMS instils by reducing variability and waste in processes. When procedures are standardized, employees well-trained, and changes carefully controlled, production lines run smoother with fewer errors or surprises. Continuous improvement principles embedded in quality systems naturally drive organisations to optimise their operations. For instance, ISO 13485 encourages manufacturers to constantly evaluate and streamline their production processes, which can lead to leaner workflows, shorter cycle times, and lower costs. One industry expert noted that these quality principles “enhance efficiency and reduce waste,” translating directly into cost savings and better productivity. In practical terms, this could mean using quality data to identify bottlenecks or high-defect areas and then refining those steps, or implementing real-time monitoring that prevents out-of-spec products before they require costly rework. Over time, a mature QMS helps an organisation operate like a well-oiled machine – not by making it rigid, but by creating a structured continuous improvement loop.

Real-world examples abound of companies gaining a competitive edge through quality-driven efficiency. In one case study, a small medical device manufacturer implemented a new QMS to address inconsistent product quality and process errors. The results were transformative: operational efficiency saw marked improvements, with streamlined processes and standardized procedures reducing inefficiencies, resulting in cost savings and better resource utilization. By eliminating redundant steps and ensuring everyone followed best practices, the company could redirect time and resources from “firefighting” mode to proactive development work. In fact, the firm found that after QMS implementation, they could focus more on innovation and product development rather than chasing quality issues, directly tying quality management to their business growth strategy. Another example can be drawn from large-scale biopharmaceutical manufacturing: companies often track key performance indicators like batch success rates, deviations per batch, and on-time delivery. When a strong QMS is in place, these metrics tend to improve – fewer batch failures and deviations mean less material waste and fewer production delays, which in turn means medicines reach patients faster.

A QMS also improves efficiency by clarifying roles and responsibilities and improving coordination. Clear documentation (SOPs, work instructions) and training under the QMS ensure that each operator knows the correct way to perform tasks, reducing variation between shifts or sites. Additionally, quality systems enforce preventive maintenance and calibration schedules for equipment, which reduces unexpected downtime due to machine failures. In supply chain management too, QMS principles apply: by qualifying and monitoring suppliers, manufacturers can avoid delays or quality issues with raw materials. All these elements contribute to smoother operations. Big players recognize that an automated QMS can give them a competitive edge via enhanced standardisation and a framework for continuous improvement. In effect, quality management has evolved from being a compliance cost centre into a driver of operational excellence. When integrated with approaches like Lean and Six Sigma, the QMS becomes the backbone of a company’s quest for efficiency – minimizing errors, maximizing yield, and ultimately delivering value faster to patients and healthcare providers.

Safeguarding Patient Safety through Quality

At its heart, every quality management effort ultimately comes down to protecting the patient. In healthcare, manufacturing excellence isn’t just about throughput or cost – it’s measured in patient outcomes and safety records. QMS is the guardian of this mission. By enforcing rigorous checks and balances, a QMS ensures that no product reaching a patient is compromised in quality. This starts from design: for a medical device, the QMS-driven design control process will identify potential hazards and incorporate essential safety features; for a cell therapy, robust quality protocols will ensure sterility and correct cell identity for each personalized dose. During production, a QMS mandates environmental controls (to prevent contamination), validated processes (to guarantee consistency), and testing at multiple stages. These measures drastically reduce the likelihood of product failures and subsequent recalls, which not only protects patients from harm but also builds trust in the healthcare system.

Consider the realm of cell and gene therapies – these treatments are often one-time, life-saving interventions for patients with no alternatives. The stakes are incredibly high. A single breach in aseptic technique or a lapse in verifying a gene vector could have serious consequences for a patient’s health. A robust QMS is what stands between a complex manufacturing process and a tragic error. In fact, contamination is a constant threat in cell and gene therapy manufacturing, and only diligent adherence to quality procedures can mitigate it. There have been instances where regulators flagged cell therapy production facilities because standard operating procedures were not properly followed, opening the possibility for contamination. In one FDA inspection report, investigators observed that staff failed to thoroughly review deviations and sometimes didn’t document follow-ups, and that aseptic protocols were not strictly followed – exactly the kind of QMS failures that could put patients at risk. The company had to scramble to address these observations to avoid delaying a critical therapy’s approval. This example underlines how patient safety is directly tied to the strength of the QMS: every batch record signed, every deviation investigated, every piece of equipment sterilised per procedure is a step toward safeguarding the end user – the patient.

Quality management systems also extend their protective umbrella to post-market surveillance. Even after a product is launched, the QMS governs how complaints are handled and how any adverse events are investigated. This ensures that if a risk to patients emerges, the company can react swiftly – whether that means recalling a device, correcting a drug’s label, or modifying a manufacturing process. A strong QMS-backed pharmacovigilance or device vigilance program will aggregate real-world data to catch safety signals early, again closing the feedback loop to manufacturing and design for improvements. In essence, QMS cultivates a company-wide vigilance for patient well-being. Culturally, it makes every employee – from the factory floor to the executive suite – conscious that quality is not abstract compliance, but a very tangible commitment to those who rely on the product. Many organisations instill this by sharing patient stories with their staff, reminding them that “the end customer is a mother, father, or child whose life may depend on our quality.” Thus, a human-centric quality approach emerges: workers take pride in doing things right not just to pass an audit, but because it means delivering a safe device for a surgeon’s use or a potent therapy for a cancer patient. When quality management is lived as a core value, patient safety becomes ingrained in every decision – truly embedding the patient’s welfare at the centre of manufacturing excellence.

Quality Management for Next-Generation Cell and Gene Therapies

Nowhere is the interplay between quality, innovation, and safety more evident than in the burgeoning field of cell and gene therapy production. These advanced therapies – such as CAR-T cell treatments and gene therapies for rare diseases – represent a new frontier with unique manufacturing challenges. Unlike traditional mass-produced pills, many cell and gene therapies are personalised (a “batch of one” for an individual patient) and highly complex to make. This puts tremendous pressure on QMS frameworks to be both rigorous and adaptable. The production of a cell therapy involves handling living cells, intricate genetic modifications, and often a race against the clock to deliver the treatment fresh to the patient. Given this complexity, QMS in this arena must cover aspects like chain of identity and chain of custody (to track each patient’s cells meticulously), cold chain logistics, and rapid lot-release testing, all while complying with Good Manufacturing Practice (GMP) standards. Leading manufacturers and academic facilities have responded by extending their quality systems in novel ways. For example, many cell therapy producers have adopted electronic batch records and digital QMS tools as “critical enablers” to manage the vast amount of data and documentation in real time. When a single patient’s treatment generates hundreds of pages of records from vein-to-vein, a digital QMS ensures nothing is lost and all steps are executed in the correct sequence – this is essential both for compliance and for the patient’s safety.

Regulatory bodies have set high expectations for quality in advanced therapy manufacturing, and QMS helps organisations meet these expectations. In Europe, cell and gene therapies are regulated as Advanced Therapy Medicinal Products (ATMPs), and every batch must be certified by a Qualified Person (QP) under an approved QMS before release. In the U.S., the FDA requires detailed Chemistry, Manufacturing, and Controls (CMC) data for gene therapy approvals, and has issued guidelines emphasizing robust analytical testing and quality oversight given the products’ novelty. Through a strong QMS, companies can provide evidence that even a one-off, patient-specific product was made with the same control and consistency as a commercial batch. We also see creative quality strategies emerging: for instance, the concept of decentralized manufacturing for cell therapies (producing them at hospital-based labs or regional centers) is gaining traction. To manage this without compromising quality, innovators have designed hub-and-spoke quality systems – a “central QMS” sets global standards and oversight, while regional sites operate under its guidance with local adjustments. A recent technical proposal described how a centralised quality unit (a “Control Site”) could oversee multiple point-of-care manufacturing sites, ensuring each one produces therapy doses that are comparably safe and effective. This model relies on harmonised procedures, intensive training, and regular audits – essentially a QMS scaled across geographies – to maintain excellence even when manufacturing is distributed. It demonstrates how flexible and powerful modern QMS thinking has become, enabling new production paradigms like hospital-based cell therapy while still upholding rigorous quality.

Real examples illustrate the importance of QMS in cell and gene therapy success. When Novartis launched one of the first CAR-T cell therapies, Kymriah, it encountered initial manufacturing challenges in achieving consistent output. The company invested in strengthening its manufacturing QMS, partnering with contract manufacturers and instituting tight process controls to improve yield and reliability. Similarly, a biotech in Asia developing CAR-T treatments implemented an intensive quality monitoring program to satisfy both U.S. FDA and China NMPA regulators, effectively running a dual-compliant QMS. These efforts paid off in reliable supply and regulatory approvals. Across Asia, the United States, and Europe, we see leading players building state-of-the-art manufacturing facilities with quality at their core. In Japan, for example, Fujifilm’s cellular therapy labs and Takeda’s gene therapy plant each incorporate isolators and automation to minimize contamination risks, all managed under strict quality protocols. China has rapidly advanced in this field too – by 2023 it approved at least three domestically developed CAR-T therapies, with companies like Legend Biotech and WuXi AppTec providing manufacturing expertise under GMP-compliant quality systems. These companies not only follow international QMS norms but often exceed them, knowing that product safety and efficacy are under intense scrutiny for novel therapies.

What sets the cell and gene therapy sector apart is the human element: every batch is often a patient’s only hope. This reality has driven a very human-centric quality mindset, where the usual walls between quality, manufacturing, and clinical teams are breaking down. Cross-functional teams work closely to ensure compliance from clinical trial stages through to commercial scale, aligning on GxP (Good Practice) requirements collectively. Companies frequently bring in specialised quality experts or consultants with cell/gene therapy experience to bolster their systems and train staff in the unique aspects of these products. There is also a push for knowledge sharing – capturing the tacit know-how of skilled technicians and making it part of the QMS documentation – so that as these organisations grow, they don’t lose the expertise that quality and consistency depend on. All of these efforts underscore a key insight: advanced therapeutics require advanced quality management. By proactively evolving QMS practices, the industry is managing to deliver some of the most innovative treatments ever conceived, without compromising on the standards of excellence that patients and regulators expect.

Global Perspectives: Asia, the United States, and Europe

Quality management practices, while universally vital, do reflect regional flavours and developments. In the United States, the culture of QMS has long been shaped by FDA regulations and industry leadership. American companies were early adopters of formal QMS standards (with many medical device firms certifying to ISO 13485 even before it was mandated globally) and often spearhead the use of digital quality tools. Initiatives like the FDA’s quality metrics program and Emerging Technology Team encourage manufacturers to quantitatively improve quality and adopt innovative manufacturing under QMS oversight. The U.S. also tends to focus on quality culture – exemplified by pharmaceutical giants fostering “right-first-time” and “no-blame” reporting environments to surface issues early. Many breakthrough cell and gene therapies have originated from U.S. companies, and with them, new quality paradigms. Kite Pharma (a Gilead company) and Juno Therapeutics (now part of BMS) are examples of U.S. pioneers in CAR-T therapy that had to rapidly scale their quality operations. They worked closely with the FDA on CMC requirements, and in some cases navigated setbacks – for instance, BMS’s CAR-T therapy Breyanzi saw approval delays partly due to manufacturing facility QA issues that had to be rectified. Such experiences reinforced for U.S. firms that quality systems are not merely about avoiding FDA warning letters, but are strategic assets to ensure supply continuity and product integrity for patients. Today, many American biopharma companies are investing in advanced QMS software, automation (like electronic deviations management), and extensive employee quality training programs to maintain their edge in manufacturing excellence.

In Europe, there is a strong tradition of quality and safety ingrained through regulations and standards. The European Medicines Agency and national authorities enforce strict GMP and ISO requirements, and European industry has been a champion of international harmonisation (for example, Europe was instrumental in developing ICH Q10). European medical device makers had to upgrade their quality systems significantly to meet the new EU MDR/IVDR, which demand robust post-market surveillance and risk management – all facets of an effective QMS. This has arguably pushed European device companies to adopt more integrated and risk-based quality systems in recent years. Europe is also home to many leading biopharmaceutical manufacturers (from Roche and Novartis to Sanofi and BioNTech), who operate extensive networks of manufacturing sites. To coordinate quality across multiple countries, these firms use centralized QMS frameworks that ensure every site – whether in Germany, Ireland, or Italy – adheres to the same global standards while complying with local laws. The concept of a global QMS with local adaptability is something European multinationals have mastered, giving them flexibility to transfer production or scale up capacity quickly without quality drift. Additionally, Europe’s emphasis on Qualified Persons (QPs) means there is a high bar for batch release – QPs serve as an extra layer of quality oversight, personally verifying that each batch meets all requirements. This has arguably made European releases very reliable. In the realm of cell and gene therapy, Europe has been progressive with initiatives like the Catapult centre in the UK, which helps small companies build GMP manufacturing and quality capabilities, and the Hospital Exemption pathway in the EU that still mandates hospital production under a quality system for bespoke ATMPs. European consortia and regulators are actively sharing best practices on QMS for these advanced therapies, knowing that public confidence is key for such innovations to thrive.

In Asia, we see diverse stages of QMS adoption and some of the most dynamic growth. Japan, with its long history of manufacturing excellence (think of concepts like “kaizen” continuous improvement, which align perfectly with QMS principles), has a very mature quality culture. Japanese pharmaceutical companies and medtech firms are typically ISO-certified and many are part of PIC/S (an international GMP cooperation), reflecting high compliance standards. Japan’s PMDA has embraced some flexibility for regenerative medicines, allowing conditional early approvals, but that comes with stringent post-market quality monitoring – all under the QMS of the sponsor. China, on the other hand, has undergone a rapid revolution in quality standards over the past decade. Where once Chinese manufacturers were seen as less stringent, today many have upgraded to world-class QMS to compete globally and satisfy their National Medical Products Administration (NMPA). The NMPA has introduced guidelines mirroring ICH Q8-Q10, joined ICH itself, and routinely inspects facilities for GMP compliance. The result is that Chinese biologics and cell therapy producers like WuXi, Legend, and others are earning international regulatory approvals, a testament to their quality system improvements. A telling sign is that China approved its first CAR-T therapies in 2021-2023, and the facilities producing them had to meet the same quality benchmarks as in the West, showcasing Asia’s leap in QMS implementation. Other parts of Asia-Pacific, such as South Korea, Singapore, and India, are also elevating quality practices. South Korea mandates GMP for cell therapies and has globally accredited production sites; Singapore has attracted advanced therapy manufacturing by offering strong IP protection and quality infrastructure; India is increasingly aligning its medical device rules with ISO 13485 to boost its medtech sector quality. However, Asia-Pacific’s challenge is the patchwork of regulations – unlike the FDA or EMA, each country has its own nuances. Companies operating in APAC thus rely on their internal QMS to navigate this complexity, often maintaining a base certification (like ISO 13485 or ISO 9001) and then adding modules to address country-specific requirements. As an IQVIA analysis pointed out, the lack of unified frameworks in APAC makes multi-country trials and manufacturing complex, but international harmonisation efforts are underway. Asian regulators are increasingly collaborating through forums and convergence projects to raise the regional quality baseline, which in turn encourages companies to invest in stronger quality systems. The emergence of Asian CDMOs serving global clients (e.g. Japan’s AGC Biologics expanding into Europe and the U.S., or India’s Serum Institute supplying worldwide) also means Asian players are meeting international QMS expectations at a greater scale than ever before.

Across all these regions, one clear trend is that leading players treat quality as a strategic priority. Whether it’s an American biotech pioneering a gene therapy, a European medtech firm launching a novel implant, or an Asian pharma scaling up vaccine production, the common thread is the understanding that without quality, there is no sustainable success. They exchange best practices at global forums, benchmark against each other, and even partner (such as Western firms collaborating with Japanese or Korean manufacturers) to leverage each region’s strengths – with QMS being the lingua franca that ensures they operate in sync. This global perspective on QMS enriches the whole industry: patients worldwide benefit when a device made in one country or a therapy made in another is produced under equally stringent and excellent quality systems.

Case Studies and Best Practices

To illustrate how QMS drives manufacturing excellence in practice, it’s worth highlighting a few concrete examples and best practices observed in the field:

  • Small Manufacturer’s Transformation: A small UK-based medical device company developing diagnostic tools discovered that inconsistent processes were hurting its product quality and compliance record. By implementing a formal QMS (aligned to ISO 13485) and investing in training, the company standardised its work instructions and greatly improved its documentation. Within a year, they saw defect rates plunge and customer complaints drop sharply. They also sailed through their next regulatory audit with minimal findings – a stark contrast to earlier audits. Crucially, the QMS didn’t just fix problems; it changed the company’s mindset. Employees began proactively suggesting process tweaks and felt ownership of quality outcomes. Management noted that meetings shifted from blame games to data-driven discussions on improvement. This cultural shift, fostered by the QMS’s structured approach, not only saved costs (by reducing rework and scrap) but also accelerated their development timelines as teams spent less time troubleshooting and more time innovating. This case demonstrates how even resource-constrained organisations can achieve excellence by embracing quality systems – turning compliance from a burden into a catalyst for performance.
  • Cell Therapy Scale-up: In a real-world example from a cell therapy manufacturer in the United States, the company faced a daunting task: scaling production of an autologous T-cell therapy from a few dozen treatments a year to several hundred, to meet patient demand post-approval. Early on, they encountered variability in product potency and some manufacturing failures. The company undertook a comprehensive QMS enhancement project. They introduced electronic batch records to reduce manual errors, tightened their change control process to carefully evaluate any process tweaks, and implemented weekly cross-department quality review meetings. As a result, consistency improved markedly – within six months, the batch success rate climbed significantly and the release testing deviations dropped. One key best practice they reported was the use of “quality agreements” and close communication with their contract testing labs and suppliers. By setting clear quality expectations with partners (e.g. how quickly a contract lab must report an out-of-spec result, or how a supplier should inform them of any raw material issues), they created an extended QMS that covered the whole supply chain. This end-to-end quality oversight proved invaluable when a critical reagent was found to be subpar – the supplier’s early alert (per the agreement) allowed the cell therapy maker to switch lots and avoid a batch failure. The lesson here is that QMS thinking must encompass not only one’s own operations but also all external inputs; quality is truly a team sport in modern manufacturing.
  • Cross-Continental Compliance: A European biopharmaceutical firm working on a gene therapy partnered with a manufacturing site in Asia to produce viral vectors. Despite distance and cultural differences, they maintained a unified QMS. They achieved this by deploying the same electronic QMS platform in both locations, complete with shared SOPs and real-time audit of each other’s data. Auditors from the European team would remotely review batch records in Asia via the system, and vice versa, creating a mutual trust and accountability. This best practice of leveraging technology for a “single-source-of-truth” quality system meant that both sites could pass FDA and EMA inspections confidently, each knowing that the other was in full compliance as well. It also sped up technology transfer: when a process change was made to improve yield in the Asian site, the European site received the change request through the QMS and could implement it in sync after proper evaluation, keeping both facilities aligned. The success of this collaboration highlights how global operations can excel by treating QMS as a living, shared framework rather than siloed local manuals. Especially for companies operating in multiple regulatory jurisdictions, having an integrated quality system prevents errors and misunderstandings that often occur when juggling different standards.
  • Human-Centric Quality Culture: A large multinational device company (with significant presence in Europe and the U.S.) provides a heartening example of human-centric quality in action. They instituted a program where any employee could halt a production line if they spotted a potential quality issue – no penalties or blame, only thanks for preventing a possible defect from reaching a patient. This empowerment, part of their QMS policy, led to a notable decrease in finished device returns. In one instance, a line worker noticed a subtle irregularity in a component’s moulding. Trusting the QMS ethos, she stopped the line and triggered an investigation. It was found that a machine calibration drift had begun to introduce a minor flaw. It was fixed within hours, and only a small batch was scrapped, avoiding what could have grown into a larger problem. Management publicly recognised the employee’s action, reinforcing that quality is everyone’s responsibility. This case underscores that while processes and technology are critical, the people operating within the QMS are the true heroes of quality. By cultivating a culture where adherence to quality procedures is seen as a source of pride and where everyone feels accountable for patient safety, companies can achieve extraordinary levels of excellence and reliability.

End Notes:

Quality Management Systems are far more than bureaucratic requirements – they are the driving force behind excellence in modern medical manufacturing. In both the medical device and biopharmaceutical realms, and most strikingly in breakthrough areas like cell and gene therapy, a robust QMS is what translates cutting-edge science into safe, reliable, and accessible treatments for patients. By supporting regulatory compliance across a patchwork of global requirements, QMS gives companies the passport to operate and innovate internationally. By embedding continuous improvement and innovation, it turns compliance into a platform for creative progress rather than a hindrance. Through gains in operational efficiency, it demonstrates that doing things right is also the fastest and most cost-effective way. And by an unwavering focus on patient safety, it aligns every procedure and decision with the ultimate mission of healthcare – improving lives.

The comparative landscape across Asia, the US, and Europe shows that while regulations and market conditions may differ, the commitment to quality unites the industry. Each region contributes its own best practices and learnings: from the holistic quality culture of Japanese manufacturing, to the technological and regulatory rigor of American firms, to the harmonisation and patient-centric approaches seen in Europe. Collaboration and knowledge exchange on quality strategies have made the global industry stronger, ensuring that a therapy developed on one continent can be produced with equal excellence on another. As we move into the future, trends like digital QMS, real-time analytics for quality data, and artificial intelligence in process control are poised to further enhance how we manage quality – but the fundamental principles will remain the same. Quality management is, and will always be, a human-centric endeavor at its core: it’s about scientists, engineers, doctors, and regulators coming together around a shared belief that every patient deserves the best, most reliable product we can make.

Inspiring, strategic, and deeply human – a Quality Management System embodies all these attributes when implemented with care. It inspires confidence and accountability in teams, provides strategic direction and consistency in business operations, and ultimately safeguards the humans at the end of the supply chain: the patients whose health and hopes are in our hands. In the quest for manufacturing excellence in life-saving therapies and devices, the QMS stands out as the quiet, powerful engine driving progress. By continuing to invest in and elevate our quality systems, we are not only complying with rules or improving metrics – we are building a future where innovation and quality walk hand in hand, delivering better outcomes and brighter possibilities for patients around the world.

News

Stay Connected

Sign up to our free newsletter and get the latest news sent direct to your inbox

Show

Forgot your password?

Show

Show

Lost your password? Please enter your email address. You will receive a link to create a new password.

Back to log-in

Close