2k Clinical Consulting, Inc.

Inspection vs Audit:  What’s the difference?

Audit and inspections are critical methods in achieving compliance.  Both are systematic,  and documented processes conducted to verify that a certain set of standards, guidelines and procedures are being met; however, they are used interchangeably according to their processes.

As an FDA Investigator, the term “inspection” was used most often; however, as I ventured out into the industry to work for sponsor companies and CROs, I heard both terms “audit” and “inspection” used interchangeably depending on who was conducting them.   At the same time, professional organizations like ASQ (American Society for Quality) primarily used the term “audit”.   

According to the sponsor companies I have worked for in the industry, the term “audit” is used when it is conducted either internally within the organization or externally with a supplier.   The term “inspection” is used when conducted by a regulatory authority such as FDA, EMA, ISO, etc.

So, what’s the difference? 

Inspection vs Audit

Regulatory Authorities such as the USFDA(United States Food & Drug Administration), EMA (European Medicines Agency), etc.  use the term “inspection” which is routinely done to:

  • Determine the safety and protection of subjects
  • Ensure adherence to federal regulations
  • Determine the validity of clinicals trials supporting approval applications

According to professional organizations like ASQ (American Society for Quality), everything is considered an audit broken down into different types (e.g., process, product, system, etc.) and parties such as:

  • First-party audit
  • Second-party audit or
  • Third party audit

First party audits, or internal audits, are conducted within an organization to measure its strengths and weaknesses against its own procedures and processes against external standards. 

Second party audits, or supplier audits, are conducted externally on a supplier by a customer to determine that products are being handled properly according to the customers’ procedures / industry stands and services are done according to the requirements of contractual agreement.

Third party audits are conducted independently from outside organizations or regulatory authorities and can result in certification, registration, recognition, approval, etc.

According to the sponsor companies I have worked for in the industry, the term “audit” is used when it is conducted either internally within the organization or externally with a supplier.   The term “inspection” is used when conducted by a regulatory authority such as FDA, EMA, ISO, etc.

Key Takeaway

The best way to minimize confusion is to consider First and Second party audits conducted internally or externally by clients or consultant companies as AUDITS and Third-party conducted by regulatory authorities as INSPECTIONS both with the ultimate goal to achieve compliance and adherence to set standards, guidelines and procedures.

How are audits and inspections differentiated in your company or department?  Feel free to contact us with your comments, questions or feedback.  Looking forward to hearing your thoughts on this topic!

 

 

Inspection Readiness Checklists: 

The Benefits & How to Utilize Them

Checklists are invaluable tools in Inspection Readiness programs within regulated industry of pharmaceutical, medical device and biotech companies. They provide a structured approach to ensure that all necessary preparations are made for inspections, audits, and regulatory assessments. The importance of checklists in Inspection Readiness cannot be overstated, as they help organizations mitigate risks, and ensure the quality and safety of their products or services.  They also help with:

  1. Standardization: They create a consistent framework for preparing for different types of inspections, promoting a structured approach to compliance.
  2. Accountability: Checklists assign tasks and responsibilities to individuals or teams, reducing the likelihood of oversights or delays.
  3. Training and Awareness: They educate employees about regulatory requirements and steps for inspection readiness, fostering a culture of compliance.
  4. Documentation Management: They ensure that all required documents are up to date, organized, and readily accessible, simplifying document retrieval during inspections.
  5. Continuous Improvement: Regularly updating checklists based on previous inspection experiences and changing regulations promotes a culture of continuous improvement.

What to Include in an Inspection Readiness Checklist

The following areas should be part of checklists that encompass your Inspection Readiness program:

  1. Regulatory Requirements: List and be aware of specific regulations, guidelines, or standards applicable to your industry or organization (ie. FDA, EMA, Health Canada, etc).
  2. Document Review: Verify the accuracy and completeness of essential documents, such as SOPs and regulatory records.
  3. Training and Competency: Document employee training and certifications to ensure competence and qualification.
  4. Facility and Equipment maintenance (if applicable): Regularly inspect and maintain infrastructure and equipment to meet regulatory standards.
  5. Quality Control and Assurance: Evaluate quality control and assurance processes to prevent deviations and non-conformances.
  6. Risk Assessment: Identify potential risks within processes and develop strategies to mitigate them.
  7. Corrective and Preventive Actions (CAPA): Track and address open CAPAs to demonstrate a commitment to improvement.
  8. Internal/Mock Inspections:  This will include internal and mock inspection schedules, simulations and documentation to help identify gaps and improve readiness.
  9. Communication Plan: This helps to outline how your organization will communicate with regulatory agencies and inspectors during the inspection.
  10. Emergency Response Plan: Prepare for unexpected situations with an emergency response plan.

In conclusion, checklists are indispensable. An effective checklist should encompass all relevant aspects of the organization’s operations, from regulatory compliance to documentation management, to guarantee a successful Inspection Readiness program. 

If you are seeking alignment within your team but are struggling with where to start in regard to creating an Inspection Readiness checklist for your company, Contact us! We’d love to hear from you to discuss strategies!

 

Pharmacovigilance and Drug Safety

Recently approved medications and those already on the market can benefit from pharmacovigilance to help ensure patient safety. While pharmacovigilance practices are highly regulated in the context of clinical trials, post-marketing incidence reporting is not properly adopted or enforced. As a result, it is anticipated that adverse events related to medications that are already in the market are underreported by about 90%. Therefore, it can be difficult to identify drug safety problems in individuals who have numerous comorbid conditions and complex disorders.

What is Pharmacovigilance?

The research and practices involved in the identification, evaluation, comprehension, and avoidance of side effects or any other issue involving medications is known as pharmacovigilance.

Before a treatment is approved for use, the only available proof of its efficacy and safety comes from clinical trials, in which patients are carefully chosen and monitored under carefully controlled settings. This indicates that a treatment has only undergone brief testing in a very small number of carefully chosen patients at the time of its authorization.

Following approval, the medication may be administered to a large number of patients, continuously, and in conjunction with other medications. In such cases, some adverse effects could manifest. Therefore, it is crucial to continuously assess the safety of all medications used in medical procedures.

How is Drug Safety Ensured?

For the evaluation of prescription medications, there are regulatory frameworks. First, the FDA examines the efficacy and safety of new pharmaceuticals that manufacturers want to market in the United States during the premarket approval process. The FDA’s examination of the studies in which people use the investigational new medicine in meticulously controlled, typically randomized trials such as Phase 1, 2 and 3 trials.

Second, the FDA takes action through its post market regulatory processes once a maker has sufficiently shown a drug or device safety and effectiveness for a specific population and set of conditions. Clinicians and patients may also report any significant or unusual adverse effects to the FDA.

According to the law, the FDA is authorized to take only limited action if it determines that a product’s post-approval use entails a higher risk of a negative outcome. However, many argue that in addition to needing a wider variety of enforcement measures, FDA also has to make better use of the authority it already possesses.

Six areas are discussed, including the FDA budget, the industry’s role, the opportunity to leverage the drug approval process to improve post market operations, the paucity of post-market data, and the limited public access to data obtained. The FDA is able to execute some of the recommended adjustments. Others would necessitate legislative action.

Conclusion

It is important to understand that no medication is 100% risk-free. A prescription drug is actually one that has “toxicity or other potentiality for harmful effect, or the method of its use, or the collateral measures necessary to its use, is not safe for use except under the supervision of a practitioner licensed by law to administer such drug,” according to the Federal Food, Drug, and Cosmetic Act.

Therefore, it is important to ensure that the drug safety plan is working in all areas of the healthcare sector to ensure utmost safety. Regulatory authorities and pharmaceutical medicine pupils must adhere to the fundamental principle and primary objective of pharmacovigilance.


References

Jones, R. (2022, April 16). Pharmacovigilance – a complete guide to Pharmacovigilance and Drug Safety – Clinical Research Certification. CCRPS Clinical Research Taininrg. Retrieved September 24, 2022, from https://ccrps.org/clinical-research-blog/pharmacovigilance-training

The theory and definitions of drug safety — pharmacovigilance. (2019). Cobert’s Manual of Drug Safety and Pharmacovigilance, 1–7. https://doi.org/10.1142/9789813278851_0001

Yehia, D. H. (2022, June 24). Introduction to pharmacovigilance: How drug safety is enforced by pharmacovigilance – pharmacovigilance foundations. Pharmacovigilance Foundations – Simplifying Healthcare Professionals Workspace. Retrieved September 24, 2022, from https://www.pharmacovigilancefoundations.com/pharmacovigilance-introduction/

The Benefits of Storyboarding in Clinical Research Trials

As part of a sponsor’s inspection readiness activities, storyboarding is a crucial step that provides a solid framework for addressing challenging or important areas of noncompliance via corrective and preventive actions. With the aid of organizational and risk assessments, sponsor companies should be able to pinpoint issues and utilize storyboards to systematically discuss the implementation of action plans with inspectors as requested during an inspection.

Storyboard Benefits

Utilizing storyboards has various benefits.  The top five benefits include the following:

  1. They offer a structure for formulating concise content that the inspection team can convey reliably.
  2. They are particularly beneficial in circumstances where an organization has gone through a transition or considerable change, for closing gaps that have already existed, and in other exceptional cases of self-identified compliance.
  3. They point out the corrective measures taken to guarantee patient safety and maintain data integrity.
  4. They assist in removing ambiguity, potential disagreement, and pressured decision-making while responding to inspection questions about challenging and perplexing clinical trial components.
  5. They lessen the possibility of issues occurring during an inspection as a result of several facilitators or SMEs accidentally giving conflicting accounts of what happened, and the steps taken to address associated problems or gaps.

Storyboard Tips

Being proactive and starting development when a problem or situation emerges can save a ton of time and work later on when attempting to put together a summary later. Therefore, it is a recommended practice to generate storyboards as early as possible.

The following are useful tips regarding a storyboard:

  • A storyboard should be utilized as a tool to assist the facilitators in making sure that important information is accurately and succinctly communicated, and that those informing inspectors are on the same page.
  • The location of pertinent supporting paperwork should be listed on a storyboard so that facilitators can respond to inspection demand swiftly.
  • Storyboards should not be lengthy things of the past with background or unnecessary material, but rather brief and to the point. They are designed to contain minimum amount of information necessary to properly respond to an inspection request.
  • A storyboard’s content can be discussed verbally with an inspector but storyboards themselves should not be shared or mentioned because they are not meant to be discussed in full during an inspection.
  • Storyboards shouldn’t include statements that unintentionally encourage facilitators to disclose information that an inspector might not have otherwise asked, as this could raise further questions and create confusion or ambiguity.

Conclusion

In conclusion, storyboards can offer the foundation required to communicate important information in a clear and simple manner with assurance and in synchronization across all departments of the organization, making the inspection readiness process considerably simpler for all parties involved.

References

Gwizdak , S., & Marshall, M. (2020, December 10). 4 ways to Adapt Your Inspection Readiness Framework in a post Covid-19 World. Halloran Consulting Group. Retrieved August 1, 2022, from https://www.hallorancg.com/2020/06/19/4-ways-to-adapt-your-inspection-readiness-framework-in-a-post-covid-19-world/#:~:text=Storyboarding%20is%20an%20important%20activity,critical%20aspects%20of%20a%20study.

HealthResearchBC. (2020, October 27). Regulatory update: Clinical trial storyboards. Clinical Trials BC. Retrieved August 1, 2022, from https://www.clinicaltrialsbc.ca/regulatory-update-clinical-trial-storyboards/

LifeScienceLeader. (2020). 09.28.20 — bringing clarity to Regulatory Inspection Readiness & Facilitation. 09.28.20 — Bringing Clarity To Regulatory Inspection Readiness & Facilitation. Retrieved August 1, 2022, from https://www.lifescienceleader.com/doc/bringing-clarity-to-regulatory-inspection-readiness-facilitation-0001

The Importance of Documenting and Reporting Adverse Events

One of the primary goals of every clinical trial is to evaluate an intervention’s safety and effectiveness to that of a control or other care. As a result, all studies expose participants to risk, and these are risks that may be similar to those they encounter in ordinary clinical practice in many circumstances. It is critical to document instances of injury or poor outcomes that occur during the study to ensure that both expected and unforeseen hazards are identified.

Several phrases are frequently misconstrued as synonyms, including adverse events, adverse drug reactions, adverse effects, serious adverse events, serious adverse effects, side-effects, complications, and damages. When it comes to drug safety, though, both terms are often used interchangeably.

The following are examples of adverse events (AEs):

  • A physiological occurrence, such as a rash.
  • A psychological occurrence, such as altered cognition.
  • An abnormality in the laboratory, such as a high creatinine level.
  • Increased severity of a pre-existing ailment, such as uncontrolled blood glucose levels

Documenting and reporting adverse events; especially those events related to the study intervention helps in:

  1. Informing health authorities, clinical investigators, and others of new and important information about events that occur on a clinical trial
  2. Contributing to the summary of adverse experiences related to the development the drug, device or regimen toxicity profile

Documenting Adverse Events

All adverse occurrences must be recorded in the medical record of the patient. The study team must first grasp how AEs should be collected before documenting them. To avoid bias in AE collection, patients should not be questioned about specific occurrences that may be expected while on the trial.

AEs should be reported or elicited from a person at each study visit in the following situations:

  • during open-ended inquiries
  • during examinations
  • during evaluations

The collection of AE data begins when the study intervention (drug/procedure) is started. The AE data should be collected from the commencement of a placebo lead-in period or other observational period intended to establish the patient’s baseline status. The AEs should be observed until they resolve or stabilize. All AEs that necessitate interruption or termination of the study drug, or those that are present at the end of study treatment, must be followed up on.

Reporting Adverse Events

Clinical trials can be conducted in a single or multi-center setting. Multicenter studies include a larger number and a wider range of research participants, making it more difficult to track adverse events, and certain incomplete safety reports from these multi centers may reduce the overall understanding of the adverse event.

The regulatory authorities must be able to analyze the safety information based on accurate documentation. The severity, study intervention relationship, action taken about the study intervention, adverse event outcomes, and if it was serious should all be documented.

All observed adverse events, as well as all adverse events reported by study participants, must be documented by the investigator. Regardless of the seriousness of the information presented, it should be recorded and reported. This data is saved in the safety database for the medicine or device under evaluation.

Conclusion

Each adverse event must be understood, as well as its relevance and significance to the drug or device being tested. Patient safety is increased as a result of the recording and reporting of these incidents.

Need inspection readiness assistance in your safety department or specialized training in the area of adverse events? Contact us! We’d love to hear from you to discuss strategies!

 

References

ClinicalTrails.gov. ClinicalTrials.gov. (n.d.). Retrieved June 11, 2022, from https://clinicaltrials.gov/ct2/help/adverse_events_desc#:~:text=Adverse%20Events%20are%20unfavorable%20changes,specified%20period%20following%20the%20trial.

MB;, G. R. E. D. N. A. L. (2014). Registries for evaluating patient outcomes: A user’s guide [internet]. National Center for Biotechnology Information. Retrieved June 11, 2022, from https://pubmed.ncbi.nlm.nih.gov/24945055/

What are ‘adverse events’ and why is it necessary to record and report them? students 4 best evidence tutorials and fundamentals. Students 4 Best Evidence. (2021, March 26). Retrieved June 11, 2022, from https://s4be.cochrane.org/blog/2021/03/26/what-are-adverse-events-and-why-is-it-necessary-to-record-and-report-them/

 

3 Ways to Close the Diversity Gap in Clinical Trials

The selection of the appropriate research participants is critical to the success of any clinical trial. Regrettably, the majority of clinical trials done are severely lacking in diversity. All too frequently, cultural and genetic factors unique to Asians, African-Americans, Latino-Americans, and other ethnic communities can contribute to differences in treatment responses and risks of adverse events.

Inadequate clinical trial representations of all populations can therefore leave underrepresented groups vulnerable due to the lack of subgroup-specific data. This disparity in diversity can lead to poor drug development and increase minority health inequities; however, there are ways to close this diversity gap.

Three Ways to Close the Diversity Gap

1. Educate Staff About Importance of Diversity in Trials

The importance of diversity in clinical trials guarantees that they are being conducted properly and strategically. It’s important to learn more about the significant unmet needs of patients who could benefit from a therapy or medicine. If we know, for example, that a health problem would affect a diverse patient community in terms of “race” and ethnicity, we should plan ahead of time to ensure that those patients are equally included in the recruiting and selection process.

2. Build Partnerships

Engaging patients is a mission in which we are not only collaborating with various groups, advocacy organizations, and patients, but also building a method in which patients feel like they are a part of our scientific study from start to finish. A clinical researcher’s job entails analyzing real-world and secondary data  to better understand disease and treatment trends, track patients’ healthcare journeys, and find strategies to demonstrate its worth in improving health outcomes. The idea is to have a varied group of people at the table in order to come up with solid and relevant solutions.

You learn to value patient, advocate, and expert collaboration. You will learn how to form relationships and engage in discourse from a variety of viewpoints. So that when it comes to performing research at various stages of the drug development process, you already have those partnerships in place and can use them to improve your knowledge and influence.

3. Start at a Micro-Level

Using more community-based routes that are congruent with how people may be familiar with acquiring information, such as barber shops, beauty parlors, or community centers, to contact and deliver information to possible study volunteers is an important tactic. Non-traditional outreach tactics not only help create valued relationships, it also provides essential information about the concerns of the community as it relates to participation in clinical trials.  Many in the community have difficulty participating in trials because of the historic and unethical Tuskegee Syphilis Study so of course, building trust in these communities is an important factor as well.

Conclusion

Minority involvement in clinical trials should be a primary focus for the entire health-care system. Participating in the aforementioned measures will help us develop medications more effectively, reduce minority health disparities, and improve overall public health. However, in order to reduce the diversity gap, more businesses and government agencies must support programs like these and be more empathic to the concerns of the minority communities.

 

References

Barron, D. (2015). Bridging the diversity gap in clinical trials: Reuters events: Pharma. Bridging the Diversity Gap in Clinical Trials | Reuters Events | Pharma. Retrieved May 23, 2022, from https://www.reutersevents.com/pharma/clinical/bridging-diversity-gap-clinical-trials

Bodicoat, D. H., Routen, A. C., Willis, A., Ekezie, W., Gillies, C., Lawson, C., Yates, T., Zaccardi, F., Davies, M. J., & Khunti, K. (2021). Promoting inclusion in clinical trials—a rapid review of the literature and Recommendations for action. Trials, 22(1). https://doi.org/10.1186/s13063-021-05849-7

Buckman, P. (2022, May 11). Council post: Bridging the gap: Why clinical trials have a diversity problem and how to fix it. Forbes. Retrieved May 23, 2022, from https://www.forbes.com/sites/forbesbusinessdevelopmentcouncil/2022/05/10/bridging-the-gap-why-clinical-trials-have-a-diversity-problem-and-how-to-fix-it/?sh=2b074b4a5078

Janssen Oncology. (2021, December 16). Working together to close the diversity gap in clinical trials: Industry and researcher perspectives. STAT. Retrieved May 23, 2022, from https://www.statnews.com/sponsor/2021/12/06/working-together-to-close-the-diversity-gap-in-clinical-trials-industry-and-researcher-perspectives/

Strategies Needed to Maintain Compliance in Clinical Research

We are constantly working towards the highest level of compliance possible.” – Mike Davidson

In clinical research, compliance is essential to ensuring that the research trial is conducted safely and accurately. The ability to maintain the highest level of compliance requires an organized and streamlined process throughout the trial.  If a clinical site or sponsor company is unable to maintain compliance, the site staff and trial team will continuously struggle to be inspection-ready throughout every phase of the trial from start-up to closeout.

This article will define compliance and discuss what three (3) strategies can be utilized to maintain compliance when conducting a clinical trial.

Compliance Defined

Compliance refers to the act of adhering to a set of regulations, processes, or rules without raising any objections or raising concerns. This process takes place during all stages of clinical research. Regulatory compliance is critical in clinical research since it helps to ensure that all research is done safely and appropriately.  Deviations from compliance and quality standards can quickly affect clinical sites and sponsor companies and ultimately, the integrity of the product and clinical data.

How to Maintain Compliance

Three strategies that can be utilized in maintaining compliance are:

  1. Internal audits – conducting regular internal audits and document reviews ensures adherence to the protocol requirements and study procedures such as the consenting process, visit schedules and source documentation. In addition, internal audits and document reviews address prompt reporting and resolution of issues such as protocol deviations, temperature excursions and safety-related issues. 
  2. Ongoing staff training– clinical staff and trial team members should receive continuous training throughout every phase of the trial(s) on how to maintain compliance with the protocol, trial procedures and applicable requirements from regulatory authorities.
  3. Automated processes – automating processes of creating, reviewing and approving quality and clinical trial documents such as Standard Operating Procedures (SOPs), protocol deviations, Case Report Forms (CRFs) and training records, reduces error and increases compliance amongst clinical staff and trial teams.

Conclusion

Maintaining compliance in clinical research is essential to ensuring that the trial is conducted safely and accurately. Maintaining compliance in clinical research may be achieved by conducting internal audits and document reviews, continually training staff and automating clinical trial process. Implementing these strategies are essential to achieving inspection readiness and being successful in the conduct of clinical trials overall.

Struggling to maintain compliance at your clinical staff or clinical department?  Contact us! We’d love to hear from you to discuss strategies!

Source Documentation: “May the SOURCE Be with You…”

Lack of consistent, clear, and adequate source documentation is one of the most typical inspection results in investigator site inspections. This is also the most discovered flaw during sponsor audits. To guarantee that the study results are established on the foundation of trustworthy and legitimate data, investigator sites must be reminded of the need for good documentation practices. 

The origins of excellent documentation standards can be found in the (International Conference of Harmonizaton – Good Clinical Practice) ICH-GCP, which defines source data and source documents. 

 ICH E6 1.51 source data 

This includes all original documents and authenticated copies of original records of clinical results, observations, or other activities in a clinical trial that are required for the trial’s restoration and assessment. The documents containing source data are referred to as sources. 

ICH E6 1.52 source documents 

This includes original documents, data, and records such as hospital records, clinical and office charts.  The ALCOA-C acronym (attributable, legible, contemporaneous, original, accurate and complete) was coined by the US Food and Drug Administration to describe key characteristics of acceptable documentation. The World Health Organization has also adopted these. These requirements have changed over time. More ‘letters’ have been added by the EMA to indicate features of good source documentation, particularly for computerized systems. 

 Common Findings Associated with Source Documentation 

Following are the common findings associated with source documentation: 

  1.  It was not possible to validate the eligibility criteria. 
  2. Because there were multiple entries for the same data point, it was impossible to tell which one was the correct source record. 
  3. Inconsistencies in records were used to corroborate the study’s primary effectiveness outcome. 
  4. Abnormal lab values were not indicated on lab reports nor any contradicting material gathered in source documentation have been shown clinical importance. 
  5. Missing pieces of information from subject interview scales, a slew of unexplained revisions months after the initial entries, and contradictory data; erroneous subject identification, inaccurate date were identified. 
  6. Factually inaccurate paperwork regarding drug disposition—dates, amount, and subject use were found.  

How Can Documentation Be Improved? 

Documentation can be improved by the following ways:

  • Give PI the responsibility to assign tasks to the trained staff. 
  • Commitment and continuous presence must be acknowledged by the Principal Investigator (PI) throughout the study. 
  • Sites conducting the study must prepare SOPs which ought to be shared with the Contract Research Organization (CRO) or study sponsor. 
  • All technical aspects must be clear of confusions and errors prior to starting work on any study.
  • The sponsor and/or the CRO ensure PI’s commitment to the study.  

 Conclusion 

The ALCOA-C and other properties indicated by regulatory agencies and GCP should be demonstrated in source documents. During regular audits, the most frequently stated findings are those linked to source documentation. The PI’s dedication to the trial and participation in it makes a tremendous difference. Efforts to educate sites, understand their practices from the pre-study visit onwards, and to monitor and train them on a regular basis will all aid in increasing and sustaining the quality of site source documentation procedures. 

 

References 

“ALCOA”: Elements of good documentation. (n.d.). Retrieved October 24, 2021, from https://blink.ucsd.edu/research/policies-compliance-ethics/compliance/ALCOA-Standards-210304.pdf. 

Bargaje, C. (2011, April). Good documentation practice in clinical research. Perspectives in clinical research. Retrieved October 24, 2021, from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3121265/. 

Commissioner, O. of the. (n.d.). List – ich guidance documents. U.S. Food and Drug Administration. Retrieved October 24, 2021, from https://www.fda.gov/science-research/clinical-trials-and-human-subject-protection/ich-guidance-documents. 

GLP documentation. (2007). Good Laboratory Practice Regulations, 241–258. https://doi.org/10.3109/9780849375842-11  

 

 

FDA vs EMA in Terms of GCP Inspections

The national and global regulations for conducting clinical trials involving human participants are known as Good Clinical Practice (ICH-GCP). They include not only quality criteria, but also regulatory guidelines to ensure that all newly created pharmaceuticals and medical devices have been clinically shown to benefit the health of the public.  The FDA and the EMA are two of the most important regulatory authorities involved in ensuring patient safety and data integrity, and here is some information about both.

FDA vs EMA

The United States Food and Drug Administration (USFDA) is a division of the United States Department of Health and Human Services. All investigative product and approved products  (drugs and devices) sold in the United States are reviewed, approved, and regulated by the FDA both domestically and internationally. The European Medicines Agency (EMA), on the other hand, controls the drug development process for all European Union member countries.

How do the FDA and EMA work differently?

Inspection Focus:

FDA Investigators will spend some time looking at generic processes, but their main focus will be on research activities. The overall approach will be to follow the Bioresearch Monitoring Program guidelines and check conformity on each study. While the EMA will analyze study details in their trial master file (TMF) review, their Subject Matter Expert (SME) interview will focus mostly on general processes.

Trial Master Files (TMF):

There is no particular FDA mandate for organizations to develop a trial master file in the United States, but if the regulatory body wants ICH GCP to be followed, then a trial master file must be created and maintained.

Inspectors from the EMA, on the other hand, will conduct a thorough and comprehensive assessment of the TMF and, with rare exceptions, will prepare to browse without assistance. TMF review will normally take up major time during the inspection. Moreover, these organizations anticipate that the majority of study documents will be accessible directly within the TMF and will be recorded in a timely manner. If a TMF is ready for an EMA inspection, it is probably ready for any other significant agency as well.

Document Review:

According to the EMA’s inaugural documents, the agency’s main goal was to recognize the importance of improving patient-reported health-related quality of life (HRQOL). The EMA’s patient-reported outcomes (PRO) advice focuses on numerous domains for generalized HRQOL assessment, whereas the FDA’s focus is on symptom-specific measurements. This distinction can be seen in the pazopanib approval documentation. While the EMA included HRQOL data from pazopanib phase III studies in its assessment, the FDA statement makes no mention of this objective.

Conclusion

The two most influential regulatory agencies, USFDA and EMA, assure us that we can trust the industry as their respective accomplishments become more transparent in improving current processes and safeguarding patients and the clinical industry’s future.

 

References

CTA. (2019, January 11). Observations from GCP sponsor inspections. Clinical trials arena. Retrieved October 11, 2021, from https://www.clinicaltrialsarena.com/comment/how-to-prepare-for-gcp-sponsor-inspections.

EMA. (2021, August 10). European Medicines Agency. Retrieved October 11, 2021, from https://www.ema.europa.eu/en.

NCBI. (n.d.). FDA in PMC. National Center for Biotechnology Information. Retrieved October 11, 2021, from https://www.ncbi.nlm.nih.gov/pmc/funder/fda/#:~:text=FDA%20is%20responsible%20for%20protecting,manufacturing%2C%20marketing%2C%20and%20distribution%20of.

NIRH. (n.d.). Trial Master File. Trial master file. Retrieved October 11, 2021, from https://www.ct-toolkit.ac.uk/routemap/trial-master-file/.

Shalby, M. (2018, August 3). Good clinical practice: FDA vs. Ema. LinkedIn. Retrieved October 11, 2021, from https://www.linkedin.com/pulse/good-clinical-practice-fda-vs-ema-michaela-shalby/.

Getting to the Core of the CAPA System – The Root Cause Analysis

You have just undergone an audit and discovered a gap in your process.  What’s the next step?  The key to inspection readiness is having an effective CAPA system that not only correct the issues but also prevent them from happening again. Only by identifying the root cause of the problem will you be able to prevent it from happening again.

When it comes to diagnosing the source of an issue in a fast-paced industry, speed is important. As a result, many departments rely on the tried-and-tested procedures of Root Cause Analysis (RCA) and Corrective Action Planning (CAPA) to identify and prevent problems. Here’s a closer look at Root Cause Analysis.

What Is a Root Cause Analysis?

Root Cause Analysis (RCA) is a technique for determining what, how, and why an event occurred so that preventative measures can be adopted. Data collection, root cause identification and execution are all part of it. To put it another way, RCA is a set of procedures that allows you to delve behind the surface of a problem to uncover causal pathways that lead to the problem’s underlying root causes.

What Are the Root Causes?

To comprehend fundamental causes, we must first comprehend what the issue is in the first place. A problem could be a divergence from customer specifications or another type of non – compliance at its most basic level.  The root causes of these issues are the precise, root factors that can be properly identified, are within the company’s authority to address, and result in effective solutions to prevent relapses.

How Are RCA and CAPA Connected?

The CAPA as discussed before,  is the action phrase, whereas if RCA is the subject. The root cause is what is causing the problem, and the CAPA is what will be done to fix it and keep it from occurring again.

The 5 Why’s in RCA

The 5 Whys is a simple yet powerful cause-and-effect method for determining the fundamental cause of a problem. You’ll begin by identifying the problem (RCA input), then query why each issue is happening until you find the root cause. Keep in mind that you don’t have to stop at five; in some circumstances, six or seven repeats may be necessary.

The Action Plan

The team must build suitable countermeasures or remedial activities after determining the root cause.  The team should also devise a strategy for putting the solutions into effect. The counter-measures can be divided into two categories:

  • Short-term Action Plan: Countermeasures that can be implemented quickly, usually in less than a week
  • Long-term Action Plan: Long-term or lasting solutions are usually more difficult to implement and may necessitate additional resources. All “long-term” action plans should be completed in less than one month. If not, they should be sent to the Continuous Improvement (CI) team for review.

Conclusion

By discovering the underlying cause and taking action to prevent it from reoccurring, the establishment of a comprehensive, well-planned Root Cause Analysis (RCA) methodology can be extremely beneficial to a department in terms of inspection readiness. Many of the lessons learned during a successful RCA can be applied to similar designs or processes.

Need to strengthen the Root Cause Analysis of your CAPA System? Contact us! We’d love to hear from you to discuss strategies!

 

References

  • Buchholz, V. (2019). What Went Wrong and How To Fix It.
  • Quality-One. (2021). Root cause Analysis (RCA). Quality. Retrieved September 10, 2021, from https://quality-one.com/rca/.
  • Wikimedia Foundation. (2021, July 13). Five whys. Wikipedia. Retrieved September 10, 2021, from https://en.wikipedia.org/wiki/Five_whys.