pepgra https://pepgra.com/ Thu, 03 Mar 2022 07:20:15 +0000 en-US hourly 1 https://wordpress.org/?v=6.4.4 https://pepgra.com/wp-content/uploads/2018/02/cropped-Pepgra_Darker_background-32x32.jpg pepgra https://pepgra.com/ 32 32 Regulatory Problems about Biomaterials and Medical Devices https://pepgra.com/blog/medical-devices/regulatory-problems-about-biomaterials-and-medical-devices/ Thu, 03 Mar 2022 07:11:53 +0000 https://pepgra.com/?p=4507 In brief All biomaterials and medical devices must comply with various Pharmaceutical Regulatory standards and rules to receive clearance. It covers a range of processes and […]

The post Regulatory Problems about Biomaterials and Medical Devices appeared first on pepgra.

]]>
In brief

All biomaterials and medical devices must comply with various Pharmaceutical Regulatory standards and rules to receive clearance. It covers a range of processes and regulations, including commercialization, clinical development, good manufacturing practice, and post-market surveillance. Global rules and regulatory challenges relating to biomaterials and medical devices are discussed. Particularly effective for smaller businesses that may not have the funds to hire a full-time vigilance specialist. Risk management, intellectual property protection, marketing authorization, university patent licences, and general good manufacturing practice are covered in the methods and regulations.

Introduction

Medical Device Regulatory Affairs has many advantages for patients, but they also have a lot of risks, especially those meant to be implanted. The purpose of national medical device regulatory authorities is to safeguard patients and the general public from dangerous goods while also allowing for the rapid introduction of novel technologies that can benefit patients and promote public health.

Based on risk assessment, the regulatory framework and standardized test techniques for medical devices, including biomaterials, are now harmonized worldwide. In this broad context, new implanted biomaterials should pass a set of regulatory tests before receiving marketing authorization. Physical and chemical categorizations, biological in vitro and in vivo studies, and clinical investigations require examinations. For traditional biomaterials and implanted devices, this approach is well-established. However, emerging revolutionary goods, such as Nano biomaterials, tissue engineering materials, and 3D-bioprinting tissues and organs, confront regulatory difficulties that act as hurdles to their timely delivery to patients.

The most recent research on the contradictory concerns emerges when evaluating newly created biomaterials and medical devices. First, a background on regulatory procedures in the United States and the European Union is presented, and a discussion of the definition of medical innovation in the medical device regulatory consulting services. The links between medical device legislation and innovation are examined on this basis. Finally, examples of cutting-edge medical technology are examined.

Methods

This open search corrected other original scientific publications and reports relevant to the articles studied in the systematic search from various regulatory bodies, consulting and expert groups, and governmental departments. These publications are especially significant because numerous stakeholders are concerned about the ethical, scientific, and legal aspects of medical device innovation, so these topics are covered in several places. However, not all of the publications evaluated in our study are cited owing to length limitations.

Medical device regulations

The Device Amendments of the United States Food and Drug Administration (FDA) produced the first regulation for devices intended for human use, and the FDA had a unique position among national regulatory bodies all over the globe until recently. The Center for Devices experts in regulatory affairs and Radiological Health (CDRH) is now in charge of premarket approval of medical devices in the United States and post-market surveillance of these devices.

Most Class I medical devices must be registered with the FDA but are not required to undergo a 510(k) premarket review. However, they must adhere to general controls such as manufacturer registration and announcement to the FDA before marketing, good manufacturing performs (GMP), appropriate branding and labelling, and available post-market reporting procedures. To meet regulatory criteria and post-market supervision, Class II medical devices must have particular labelling requirements.

Clinical studies, done in line with Good Clinical Practices (GCP), are required by the FDA for “high-risk” devices to establish safety and effectiveness. These trials can be costly and take years to complete. After receiving FDA investigational device exemption (IDE) approval, the devices are clinically approved. Unless its “substantially equivalence” to a lawfully sold Class I or Class II item can be proven, a device that has not been on the market is deemed a Class III.

Regulations’ Impact on Medical Device Innovation

Regulatory concerns influence the whole innovation cycle, and they must be considered throughout the design and development of medical devices and during pre-clinical and clinical testing, product regulatory testing, manufacturing, and post-marketing surveillance. As a result, the collaboration between medical device developers and national regulatory bodies is crucial for innovation and competitiveness.

 

To show compliance with the “Essential Principles of Safety and Performance of Medical Devices,” quality and risk management systems must be incorporated throughout the life cycle of medical goods. The identification and description of dangers connected with medical devices and their accessories; risk analysis, evaluation, and control; and monitoring the efficacy of that control are all part of risk management.

Conclusion

Strong medical device laws exist to safeguard patients and the rest of society from dangerous items and improve public health by bringing new products to market that can benefit patients. However, the risk of releasing devices that haven’t been thoroughly tested exists, and an effective review procedure is still a source of worry for academics, business, government, and the general public. The categorization of medical goods into pharmaceutical regulatory affairs, biologics, and medical devices is the cornerstone of the world’s most advanced regulatory systems; nevertheless, novel combination products prove problematic to the present product classification scheme. Furthermore, the use of emerging technologies in medical device design, such as nanotechnology, tissue engineering, and 3D printing, emphasizes classifying medical devices into risk classes. Even at the earliest phases of the invention process, the duty for ensuring the safety and efficacy of devices relies not only on national regulatory authorities but largely on researchers, manufacturers, and clinicians.

About Pepgra

Pepgra CRO provides high-quality Clinical and Regulatory Expert Services to its clients. Our regulatory services experience draws heavily on the most recent research to assist you in designing and implementing clinical and regulatory frameworks that meet your needs. Our pharmaceutical and biotechnology industries clients have been very pleased with our regulatory services. Our regulatory affairs professionals from the world’s leading pharmaceutical companies focus on assisting in developing new medical products, the integration of regulatory principles, and authoring and submitting appropriate reports to health authorities.

References

  1. Guerra-Bretaña, Rosa Mayelin, and Andrea Lucía Flórez-Rendón. “Impact of regulations on innovation in the field of medical devices.” Research on biomedical engineering34 (2018): 356-367.
  2. Gurman, Pablo, O. Rabinovitz, and Tim B. Hunter. “Regulatory challenges on biomaterials: focus on medical devices.” Biomater Sci Integr Clin Eng Approach223 (2012): 48.

 

The post Regulatory Problems about Biomaterials and Medical Devices appeared first on pepgra.

]]>
Guidelines for post-market surveillance of medical devices https://pepgra.com/blog/medical-devices/guidelines-for-post-market-surveillance-of-medical-devices/ Wed, 02 Mar 2022 12:08:55 +0000 https://pepgra.com/?p=4502 In brief Manufacturers perform post-market surveillance to gather and assess experience obtained from medical devices that have been placed on the market and determine the need […]

The post Guidelines for post-market surveillance of medical devices appeared first on pepgra.

]]>
In brief

Manufacturers perform post-market surveillance to gather and assess experience obtained from medical devices that have been placed on the market and determine the need for any action. Post-market surveillance is an important tool for ensuring that medical devices remain safe and effective and taking action if the hazard of continuing to use the device outweighs the benefit. The examination of post-market monitoring experiences can also reveal areas where the medical device could be improved.

Introduction

Like many other worldwide regulatory frameworks, the WHO Global Model Regulatory Framework for Medical Devices, which includes in vitro diagnostic medical devices, necessitates the deployment of post-market surveillance systems. Receiving and assessing comments are the bare minimums for post-market monitoring, but this may be broadened to include other tasks. The operations of national regulatory authorities (NRAs) acting in response to reports of adverse events received, known as vigilance, are also included in the WHO Global Model Regulatory Framework for Medical Devices. The terms “adverse event” and “incident” may be used interchangeably in various jurisdictions. The word “incident” will be used in this advice to refer to a variety of experiences that may be obtained when using a medical device.

The WHO Global Model Regulatory Framework for Medical Devices defines post-market surveillance as the activity of NRAs. Market monitoring is the term used in this paper to describe the operations carried out by NRAs. The phrase “post-market monitoring” is only used in this text to refer to processes carried out by manufacturers. As a result, post-market surveillance, vigilance, and market surveillance are all interchangeable concepts. Users’ insights about the use of medical devices are shared with manufacturers. Manufacturers notify NRAs of specific instances and inform them of the steps taken. The NRA will look into the manufacturers’ inquiries and any further steps. This has to do with NRAs’ market surveillance obligations. The term “market surveillance” refers to the entire set of operations carried out.

Basic principles of post-market surveillance

Manufacturers of medical devices undertake pre-market evaluations of product quality, safety, and performance before releasing them into the market. Risk management concepts are used to decide risk reduction and residual risk tolerance. However, problems may develop once the medical equipment is on the market.

Manufacturers’ responsibilities

Even though medical devices are conceived, developed, manufactured, and distributed worldwide following extensive pre-market testing, residual concerns regarding safety and performance will persist throughout the product’s lifetime. This is attributable to various reasons, including intrinsic product variability, factors impacting the medical device’s use, environment, human interaction, and medical device failure or overuse. Before a medical device is released on the market, design and development processes guarantee that the remaining risks are acceptable compared to the anticipated benefits.

Actions of users about manufacturers’ post-market surveillance

 

Post-market surveillance mechanisms

The information that can/will be gathered during post-market surveillance is dependent on the information that can/will be collected. The manufacturer must first determine the post-market surveillance objectives for any given medical device or collection of medical devices, and the maker must next decide which sources are required to achieve these goals. Data will be grouped and analysed based on this.

Electronic post-market surveillance system for medical devices

The gathering of information concerning adverse occurrences using medical devices and communicating such information among Health Care Institutions and Manufacturers Competent Authorities is a fundamental problem in the health care industry. An Electronic Post Market Surveillance System that solves the problem above effectively and securely while adhering to international standards. The PMS Server Application is deployed on the premises of the manufacturers/suppliers, while the PMS Client Application is installed at the Health Care Institutions. The PMS programmes are used to manage PMS Reports and Responses, and they include significant characteristics, including user-friendly interfaces, interoperability, and various implementations based on performance and cost requirements. The system was assessed methodically.

Conclusion

The goals and methods for post-market monitoring for medical devices undertaken by manufacturers with the help of their economic operators, market surveillance conducted by regulators, and the participation of other stakeholders in these processes are discussed in this paper. It explains the steps required after a medical device is placed on the market to ensure that it continues to meet the standards for safety, quality, and performance.

About Pepgra

Pepgra has a lot of post-market surveillance (PMS) report writing knowledge. The European Medicines Agency (EMA), the Food and Drug Administration (FDA), and other local nation criteria are followed by our PMS specialists. Pepgra CRO contributes to the advancement of public health by detecting and assessing safety indications from available data sources using evidence-based techniques and then recommending appropriate regulatory actions such as labelling changes, Risk Evaluation and Mitigation Strategies (REMS) communication of relevant safety information. Pepgra CRO, with its extensive understanding of the pharmaceutical business and remarkable in-house skills, provides high-quality and cost-effective post-marketing monitoring services to its clients.

References

  1. Chaudhry, Junaid, et al. “Enabling Technologies for Post Market Surveillance of Medical Devices.” (2018).
  2. Vlachos, I., Dimitris Kalivas, and Ourania Panou-Diamandi. “An electronic post-market surveillance system for medical devices.” Computer methods and programs in biomedicine2 (2003): 129-140.

The post Guidelines for post-market surveillance of medical devices appeared first on pepgra.

]]>
What Is the Healthcare Impact of Data Collection? https://pepgra.com/blog/healthcare/what-is-the-healthcare-impact-of-data-collection/ Tue, 01 Mar 2022 04:26:57 +0000 https://pepgra.com/?p=4487 In brief Regardless of industry, decisions should be founded on facts. The significance of data collecting and analysis based on it The more precise the information […]

The post What Is the Healthcare Impact of Data Collection? appeared first on pepgra.

]]>
In brief

Regardless of industry, decisions should be founded on facts. The significance of data collecting and analysis based on it The more precise the information acquired, the more sensible the judgments made, and the better the results that may be reached, according to data technology. Medicine is one of the industries that Data has had a significant impact on. If you want to learn more about the usefulness of data collecting in health care and the technologies used to gather data and transform it into commercial value, this paper will be helpful.

Introduction

The systematic process of acquiring, evaluating, and interpreting diverse forms of information from multiple sources is known as data collection. In general, data gathering is carried out for research purposes to gain a comprehensive understanding of a topic and build a basis for decision-making.

Data is divided into two types:

  • Quantitative — a numerical form, such as percentages, comparisons, etc.
  • Qualitative — in words, such as a characteristic or look description, etc.

Statista’s graph depicts a variety of information sources, including transactions, system logs, sensors, texts, social media, clickstream data, videos, and photos. As of summer, 64 % of respondents were already analysing transaction data within their firms, with another 28 % planning to do so, demonstrating the importance of data collection in business.

Data collection in health care is crucial

Healthcare quickly uses cutting-edge technology and devices to monitor and analyse data about patients, hospitals, and other medical systems. Data collecting technologies make it easier to acquire a better picture of a patient’s health, organise evidence more efficiently, and share it with other professionals with just one click.

Patient participation and treatment can be helped by effective information collection and management. That is why hiring healthcare software developers and obtaining a customised solution for your company might be one of the most gentle decisions you make.

Here are the main benefits of data collection in healthcare:

Healthcare data storage challenges

The following are the primary concerns about data gathering that may influence whether or not it is implemented:

  • Data of poor quality. The quality of Data is one of the most significant concerns, and some databases are better than others in terms of efficiency.
  • Data security is essential. Information security, particularly when it comes to sensitive data, has always been a problem, not only in the medical industry.
  • Collaboration is proving to be complicated. Doctors enter data about patients and their treatments in various ways: some utilise EDW, while others use Excel spreadsheets.

Data Collection Process and Identification of Summary Measures

At least two authors examined each article to find the essential parts. The reviewers utilised a spreadsheet template to summarise their major observations from each publication. One of the team members consolidated the spreadsheets and shared them with the rest of the group. Reviewers gathered for a final time to discuss their results in a consensus meeting. Patterns were found from this Data, and conclusions were drawn from those trends.

Health data collection methods

The first and most serious step is to choose a method that suits your particular enterprise’s needs and goals. Here are some questions that may help:

  • Management of customer relationships (CRM). These systems allow you to collect and manage data from various sources and store it in one location. They also evaluate data and keep it safely within a business. There are CRMs for operational, strategic, analytical, and collaborative purposes.
  • Electronic medical records (EMR) (EHR). CRM technology is related to HER technology. It helps collect, analyse, and share data between hospitals and other organisations. Medical history, allergies, immunisations, radiological pictures, and personal data may all be found in EHRs. Over 94 % of hospitals in the United States and over 60 % of private medical providers now use EHR systems.
  • Apps for mobile devices. Patients and hospitals can be connected via smartphone and tablet applications, collecting data and storing it in databases. They reduce time by creating a quick and secure environment between a doctor and a patient.

Impact of Data Collection in Healthcare

The best examples of how analysis and data tracking improve the world may be found in the healthcare industry. The need to solve local organisational concerns, such as lowering workloads and increasing profitability for a medical agency, and global challenges, such as anticipating epidemics and battling current diseases more effectively, drives the use of Big Data in medicine. Health systems may use data collecting to generate holistic perspectives of patients, personalise therapies, advance treatment procedures, increase communication between physicians and patients, and improve health outcomes.

Conclusion

Because data analysis has the potential to improve people’s lives, the range of data applications in medicine should be systematically expanded. Information technology allows for the diagnosis of individual diseases and the prediction of the health of entire social groupings. As a result, incorporating data collecting into healthcare is critical for creating preventative strategies and saving lives.

About Pepgra

The vast majority of Data is available in doctor’s notes, computerised medical records, prescriptions, and other related information. Despite the golden potential of big data in healthcare, good insights are difficult to come by due to complicated, noisy (unstructured), longitudinal, and enormous data. We collect data from various sources and do semantic annotation at Pepgra. We gather information depending on the research questions that you want to solve.

References

  1. Kruse, Clemens Scott, et al. “Challenges and opportunities of big data in health care: a systematic review.” JMIR medical informatics4 (2016): e5359.
  2. Lavallee, Danielle C., et al. “Incorporating patient-reported outcomes into health care to engage patients and enhance care.” Health Affairs4 (2016): 575-582.

The post What Is the Healthcare Impact of Data Collection? appeared first on pepgra.

]]>
Evaluating the results of the literature search and screening https://pepgra.com/blog/literature/evaluating-the-results-of-the-literature-search-and-screening/ Mon, 28 Feb 2022 09:37:57 +0000 https://pepgra.com/?p=4478 In brief Literature searching is generally accepted as an essential part of the literature review process. It entails a study search in the literature to produce […]

The post Evaluating the results of the literature search and screening appeared first on pepgra.

]]>
In brief

Literature searching is generally accepted as an essential part of the literature review process. It entails a study search in the literature to produce a transparent report of study identification that informs readers about how studies were identified and how the review’s results fit into the relevant evidence. The goal of this study is whether a standard model of the Global and Local Literature Search Screening Services can be found in literature review guidance materials, and if so, how this process is represented in the advice and supported by research.

Introduction

A literature review.

The literature was divided into two categories: advice and published research. The Cochrane and Campbell Handbooks were among the nine guideline materials found. ‘Pearl growth,’ citation chasing, a PubMed search utilising the systematic and literature review regulatory services techniques filter, and the authors’ topic expertise were used to find published research.

After that, the essential portions of each guideline document were reviewed and re-read to identify crucial methodological phases. The steps of the methodology were recognised and specified. This information was analysed to find areas of common ground and regions where scientific writing services’ guide manuals differed. The selection of ‘important stages’ in literature searching was based on consensus across numerous guideline publications.

Identifying supporting studies

The authors wanted to create an evidence foundation of supporting research (henceforth referred to as “studies”) that contribute to current pharmacovigilance literature search service practice in addition to offering guidelines. The authors chose articles initially based on their expertise of the issue area and then by meticulous citation hunting of relevant studies found at each stage of the search procedure.

Table 1. The critical stages of literature search guidance as identified from nine key texts

 

Extracting the data

The relevant portions (chapters) on literature searching were reviewed and re-read to find critical methodological phases to disclose the implicit process of medical literature review searching inside each guiding document. A significant methodological stage was described as a discrete step in the entire process in which specific guidance is reported, and action is done, resulting in a finished literature monitoring service.

Search Strategies

We conducted two PubMed searches to find studies relevant to our research. The following is a description of how these searches were organised.

Search #1: The following search technique (no date restrictions, search date October 22, 2010) was used to find publications discussing approaches of priority setting:

(“Research”[Mesh] OR “Health Services Research”[Mesh]) AND (exercise[title/abstract] OR tool[title/abstract] OR tools[title/abstract] OR model[title/abstract] OR models[title/abstract] OR method[title/abstract] OR methods[title/abstract] OR “models, theoretical”[MeSH Terms] OR “costs and cost analysis”[MeSH Terms] OR “resource allocation”[MeSH Terms] OR “investments/economics”[Mesh Terms]) AND (“health priorities”[MeSH Terms] OR “priority setting”[title/abstract] OR “research priorities”[title/abstract] OR “research priority”[title/abstract])

Search #2: The following search method (no date limitations, search date December 1, 2010) was used to find publications particularly addressing VOI:

“value of information”[title/abstract] AND ((“Decision Making”[Mesh] OR “Decision Theory”[Mesh]) OR (“Research”[Mesh] OR “Health Services Research”[Mesh]) OR research[title/abstract])”

Limitations

The concentration on recommendations provided in Europe and Australia may be a possible restriction of these local literature review services. In the section “Identifying guidance,” we decided to choose nine of the nine advice documents analysed in this literature analysis. In summary, these nine guidance documents were selected as the most significant healthcare guidance that informs systematic and literature-reviewing practice, given its prominence in the science of health-information retrieval.

Conclusion

The presence of a shared model of the literature searching process in systematic reviews appears to be demonstrated by this literature review. This methodology is referred to as “the typical approach” since it appears prevalent in nine separate guidance manuals. The data described above show eight essential steps in conducting a systematic reviews literature searches, and these key stages are consistently documented in guidance papers, implying that the key scenes are well-understood.

About Pepgra

Pepgra offers pharmacovigilance Literature review search services as part of the drug safety and efficacy services. Our medical regulatory staff has extensive experience in searching for published articles from multiple databases and can comprehensively cost-effectively manage your literature screening requirement in conjunction with writing periodic safety reports. We perform the literature search for aggregate reports, benefit-risk analyses, signal evaluation, or ongoing screening as required by local and regional requirements. We can conduct literature screening as per your requirement in local territories.

References

  1. Cooper, C., Booth, A., Varley-Campbell, J. et al.Defining the process to literature searching in systematic reviews: a literature review of guidance and supporting studies. BMC Med Res Methodol 18, 85 (2018). https://doi.org/10.1186/s12874-018-0545-3
  2. Myers E, Sanders GD, Ravi D, et al. Evaluating the Potential Use of Modeling and Value-of-Information Analysis for Future Research Prioritization Within the Evidence-Based Practice Center Program [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2011 Jun.

 

The post Evaluating the results of the literature search and screening appeared first on pepgra.

]]>
Literature Screening and Risk Management for Biosimilars – Challenges and Preventive Measures https://pepgra.com/blog/literature/literature-screening-and-risk-management-for-biosimilars-challenges-and-preventive-measures/ Fri, 26 Feb 2021 04:25:23 +0000 https://pepgra.com/?p=4148 In-Brief Biosimilars signify a new class of medical products that will significantly impact the clinical practice of pharmacovigilance literature search. They are the same on an […]

The post Literature Screening and Risk Management for Biosimilars – Challenges and Preventive Measures appeared first on pepgra.

]]>
In-Brief
  • Biosimilars signify a new class of medical products that will significantly impact the clinical practice of pharmacovigilance literature search.
  • They are the same on an amino acid sequence level to present reference biopharmaceutical products. However, they may show differences on a protein level.
  • Pepgra blog provides a brief overview of biosimilar development. It describes the preventive measures and challenges that should be considered during bio similars’ admission into the clinic using literature surveillance in pharmacovigilance and provides pharmacovigilance literature search services.

Introduction

The US FDA characterizes a biosimilar as “a natural product that is profoundly like a US-authorized reference organic product despite minor contrasts in clinically idle parts. For which there are no clinically significant contrasts between the organic product and the reference product regarding the security, virtue, and intensity of the product.” The European Medicine Agency definition is practically identical in literature screening.

The World Health Organization characterizes pharmacovigilance as the science and exercises identifying with the recognition, evaluation, comprehension, and anticipation of unfavourable impacts or other medication-related issues.

Assembling the two has yielded a complex administrative scene with wide varieties and irregularities across nations and markets.

What’s reasonable is that it is adequately troublesome to assemble and keep a powerful PV program to meet administrative prerequisites for little particle drugs – but such projects won’t fulfil the necessities for biosimilars.

Organizations creating biosimilars, regardless of whether from their trendsetter biologic or another that has gone off-patent, should know about a few significant questions as for biosimilars that will affect their pharmacovigilance literature screening services.

Challenges in Literature screening and risk management for biosimilars:

Assembling strategies

The assembling cycle for biopharmaceuticals is more perplexing than for conventional little particle drugs. Little contrasts between assembling strategies can altogether affect a biosimilar’s natural properties, perfection and clinical action. Consequently, there is no assurance that the subsequent biosimilar will be equivalent to its reference product.

Product names

Fifty particular biosimilars are presently being developed—but since their names are not unmistakable, this groundswell is probably going to bring about visibility issues in case of an ADR, at any rate temporarily. That is because biosimilars in the EU can have a similar International Non-proprietary Name (INN) as the trendsetter biologic. The FDA plans to assign a non-proprietary name that incorporates a postfix made out of four lowercase letters. Nonetheless, in reality, patients and clinicians may keep on alluding to a biosimilar by its reference image name, even in ADRs from scientific literature search services.

Generics and brand name

Products can be recommended reciprocally by and large. Biosimilars—albeit tantamount to the trendsetter medications—can’t. “Programmed” compatibility would require information showing that a biosimilar produces an identical clinical outcome in some random person. The FDA still can’t uncover how it will deal with biosimilars’ compatibility, though the EMA leaves the individual party states’ choice.

Post endorsement observation for immunogenicity

Post endorsement observation for immunogenicity and uncommon unfavourable occasions might be required or potentially needed over the long-term when a biosimilar is available. Such observing is commanded in the EU, although the FDA presently can’t explicitly address this issue.

Advancing rules

As rules for biosimilar endorsements and PV advance, particularly in the US, drug organizations should remain cautious with the goal that their PV projects can quickly and effectively adjust to developing administrative measures.

Preventive measures

  • Maintaining a vault of data on natural products accessible in the district will help in right recognizable proof of the product elaborate when a negative response is accounted for in the PV framework.
  • Developing unique contents that would take into consideration the assortment of point by point data of the product connected with the unfriendly response in the underlying or follow up correspondence
  • Ensuring cautious clinical assessment of all speculated immunogenicity reports with comprehension
  • Implementing continuous total survey of wellbeing information and examination with the security profile of the reference product to comprehend the distinctions in danger profiles
  • Designing an RMP with extra measures to distinguish or assess obscure security issues, including immunogenicity and uncommon occasions, yet
  • Setting up unique product/tolerant vaults for associate occasion checking
  • Conducting controlled post-endorsement adequacy and wellbeing concentrates signs and target populaces altogether sufficiently
  • A product name with viability and security data was identified with both the reference product and biosimilar recognized by source.

Conclusion

End-of-patent exclusiveness and developments in biotechnology, enabling their producers, have created substantial opportunities for follow-on biologics or Biosimilars to arrive at the market and serve patients’ requirements all over the world in a cost-effective manner. However, Biosimilars’ PV and risk management present many unique and special challenges. Pepgra also listed the preventive measures to control the risks in clinical sectors and provides pharmacovigilance literature screening services.

References

  1. Casadevall, N., Edwards, I. R., Felix, T., Graze, P. R., Litten, J. B., Strober, B. E., & Warnock, D. G. (2013). Pharmacovigilance and biosimilars: considerations, needs and challenges. Expert Opinion on Biological Therapy13(7), 1039-1047.
  2. Scavone, C., Rafaniello, C., Berrino, L., Rossi, F., & Capuano, A. (2017). Strengths, weaknesses and future challenges of biosimilars’ development. An opinion on how to improve the knowledge and use of biosimilars in clinical practice. Pharmacological research126, 138-142.
  3. Zuñiga, L., & Calvo, B. (2010). Biosimilars: pharmacovigilance and risk management. Pharmacoepidemiology and drug safety19(7), 661-669.

 

 

 

 

The post Literature Screening and Risk Management for Biosimilars – Challenges and Preventive Measures appeared first on pepgra.

]]>
Uses and Implementation process of Electronic Data Capture (EDC) in Clinical Trials https://pepgra.com/blog/clinical-trail-design/uses-and-implementation-process-of-electronic-data-capture-edc-in-clinical-trials/ Tue, 23 Feb 2021 11:49:07 +0000 https://pepgra.com/?p=4144 In-Brief: Clinical Trial Management Systems (CTMS) are an essential part of every clinical trial. Choosing the right CTMS helps address inadequacies on the operational side of […]

The post Uses and Implementation process of Electronic Data Capture (EDC) in Clinical Trials appeared first on pepgra.

]]>
In-Brief:
  • Clinical Trial Management Systems (CTMS) are an essential part of every clinical trial. Choosing the right CTMS helps address inadequacies on the operational side of research, such as clinical trial preparation, planning, performance and writing for clinical trial monitoring.
  • As more pharma and biopharma promoters start to identify the potential breaks with EDC-CTMS integration, there is a growing requirement to address the complicated electronic data capture (EDC) execution procedure.
  • Pepgra explains the EDC benefits and implementation process in this blog and also offers clinical trial monitoring services

Introduction:

An Electronic Data Capture (EDC) framework is programming that stores persistent information gathered in clinical trials to lay it out plainly. Information is ordinarily first recorded on paper and is then translated into the framework and saved in an electronic case report format (CRF). An ever-increasing number of clinical trials are taking EDC programming and supplanting paper records with electronic records. Supporters, contract research organization (CROs), and locales have embraced EDC frameworks to do both straightforward and complex trials taking all things together periods of research. While there are numerous EDC merchants, a few associations assemble their framework in-house using clinical trial services. Most EDC frameworks offer somewhat various highlights, yet all EDC programming is intended to smooth out information assortment.

Uses of an EDC system in clinical trials

Many research associations understand EDC’s upsides over different techniques and utilize new medical monitor responsibilities in a clinical trial. An EDC framework can assist you with making progress in the accompanying manners:

Quicker Access to Data

An EDC framework can save a lot of time with continuous admittance to information and less time spent on inquiry the board. It additionally saves time toward the finish of an investigation, permitting faster accessibility of the data for examination. While it can require some investment to at first figure out how to utilize a particular framework, some are spontaneous to such an extent that a couple of long periods of preparing is required for clinical research monitoring.

Data Security 

An EDC framework is facilitated online with an information section finished on an electronic interface in the clinical trial audit. Given the idea of the information gathered in an EDC framework, programming sellers ensure the information is secured and sponsored up. Since every client account has assigned consents, most activities must be completed by specific jobs.

Accuracy

EDC frameworks improve information quality. There are choices to include limitations in a format that keep erroneous or strange rates from being entered. Utilizing a modernized framework empowers decipherable passages and programmed counts for cleaner information using clinical trial monitoring services.

Organization

The utilization of an EDC framework expands clinical trial management proficiency because of its easy to understand the route. Search choices permit you to discover and channel precisely what you need handily and store everything in one area with more prominent while utilizing less paper.

Cost-Effectiveness

Monetarily talking, the expense of an EDC framework goes from free to costly. Valuing shifts, and a few sellers charge for extra help and different costs. Buying an EDC framework can appear to be a huge venture. However, it should set aside cash over the long haul.

Compliance

An EDC should be consistent with administrative prerequisites. The product ought to have technical controls set up to guarantee information honesty. To appropriately keep an EDC framework, standard operation procedure (SOPs) are fundamental to ensuring administrative and hierarchical approaches are met.

Performing EDC in clinical trials

Making the transition to EDC

The appearance of EDC advancements is molding the clinical preliminaries information the executive’s scene, offering numerous advantages for the business. EDC’s appropriation instead of paper-based strategies was initially passive; anyway, clients can get things done with EDC that they can’t do with standard information bases, making a solid case for the venture. The most recent years have seen an extraordinary move in EDC’s take-up with paper CRFs currently just being considered for specific prerequisites.  Advanced layouts are handily changed to suit each new investigation; saving time spent planning and delivering paper CRFs. On average, it has been demonstrated that EDC cuts 41% of pre-study planning time. Utilizing EDC, information is gathered and gone into an information assortment device just a single time, with a paper framework; information should be entered first and foremost into a case report structure then into an electronic framework by an information section gathering. It expands handling time as well as influences record honesty.

EDC likewise permits information cleaning to happen promptly and doesn’t need escalated hands-on work from information the executive’s bunch for preparing. The info the executive’s bunch executes the rationale checks against data gathered weeks or months back as paper contemplates.  EDC framework rationale checks are conducted when the site enters and submits information, permitting it to be cleaned progressively.  At last, following appropriate framework determination and advancement, just as excellent examination the executives, EDC allows clients brisk admittance to clean information with low operational expenses.

Making a complete EDC data set plan

There is a requirement for interest as expected and assets to guarantee that every preliminary information base is exhaustive. If organizations don’t make this venture, there is a danger that changes, or increments, will be required later, which will be exorbitant and could have more extensive ramifications on tasks.

Most EDC frameworks accompany a standard set-up of reports; nonetheless, extra pieces might be needed for study oversight and the board. Most great EDC merchants can give additional custom announcing. Likewise, with the information base turn of events, the plan of reports must be very much thought of and indicated toward the start of the investigation.

Inside the plan of an EDC framework, it is likewise imperative to consider the configuration in which information is being gathered to be genuinely deciphered or customized in another outsider programming once it leaves the EDC framework. Executing a predictable information assortment strategy incorporates normalizing the definitions for the information gathered across numerous destinations.

EDC frameworks should have a choice to observe Clinical Data Interchange Standards Consortium (CDISC) principles, and for datasets to be following Clinical Data Acquisition Standards Harmonization (CDASH) rules, so it is prepared for factual examination later on. It can save time and efficiencies as non-CDASH information should be re-attempted to satisfy a CDISC guideline upon administrative accommodation.

The utilization of a more prominent number of standard plans during study constructs can assist with proficiency and the nature of revealing. Yet, it altogether lessens the time it takes for information bases to be assembled.

Conclusion:

As more pharma and biopharma promoters start to identify the potential breaks with EDC-CTMS integration, there is a growing requirement to address the complex electronic data capture (EDC) execution procedure and its applications. Pepgra explains the EDC benefits and implementation process in this blog and also offers clinical monitoring services.

References:

  1. Sahoo, U., & Bhatt, A. (2004). Electronic data capture (EDC)–a new mantra for clinical trials. Quality Assurance10(3-4), 117-121.
  2. El Emam, K., Jonker, E., Sampson, M., Krleža-Jerić, K., & Neisa, A. (2009). The use of electronic data capture tools in clinical trials: Web-survey of 259 Canadian trials. Journal of medical Internet research11(1), e8.

The post Uses and Implementation process of Electronic Data Capture (EDC) in Clinical Trials appeared first on pepgra.

]]>
Challenges faced during FDA Device Approval Process https://pepgra.com/blog/food-and-drug/challenges-faced-during-fda-device-approval-process/ Thu, 18 Feb 2021 10:22:17 +0000 https://pepgra.com/?p=4139 In-Brief: Effective planning is fundamental to success for any business but an even greater degree in the medical device manufacturing industry. Pepgra covers the top 5 […]

The post Challenges faced during FDA Device Approval Process appeared first on pepgra.

]]>
In-Brief:
  • Effective planning is fundamental to success for any business but an even greater degree in the medical device manufacturing industry.
  • Pepgra covers the top 5 challenges the medical device manufacturing industry faces and how effective requirements planning allows organizations to position themselves to meet these unique challenges proactively to get FDA approval from a pharmacovigilance literature search

Introduction:

As we plan, medical device makers face another world that is brimming with favorable circumstances. Nonetheless, the vulnerability lies ahead too. New rules and guidelines are arising, repayment rules are getting more perplexing, medical care elements are advancing, and organizations face an undeniably difficult situation. This challenging climate necessitates that medical devices organizations decrease costs, smooth out their activities, and enhance them more rapidly says a literature screening. Moreover, medical device producers are set to observe development openings in developing business sectors with the expanding significance of medical devices and careful hardware in current medical care using literature surveillance in pharmacovigilance.

FDA Device Approval

Worldwide, the medical device makers are moving from an exchange based way to deal with a methodology. It includes zeroing in on making an incentive for suppliers, experts, payers, and patients by giving careful instruments and medical apparatuses that are exceptionally cost-proficient, inventive, and say a lot about the product quality. The medical device fabricating industry is required to develop significantly inferable from the rising maturing populace, expanding wellbeing concerns, and soaring medical services costs from a medical device literature review.

Investigates of medical device administrative frameworks ordinarily present one of two clashing perspectives from a pharmacovigilance literature review: exhausting administrative prerequisites postpone or forestall helpful device accessibility. Less specific administrative necessities put residents in danger presented to dangerous or incapable devices. Regularly, the evaluations of device guideline are upheld by examinations contrasting the US Food and Drug Administration (FDA) approaches to the European Union administrative cycle. It appears to recognize the better framework and recommend a redesign of the framework to reflect the other.

 

Challenges faced during FDA Device Approval Process

Different regulatory approval pathways

Medical device mixes include segments that various parts of the FDA would generally supervise. Therefore, every blend product presents exceptional and testing administrative contemplations.

The task depends on the assurance of the product’s PMOA. If the PMOA is inferable from the medical development, the centre liable for the premarket survey of that drug product would have actual locale for the mixed product. For this situation, it is the FDA’s Center for Drug and Evaluation Research (CDER).

Additionally, if the PMOA of a device drug mix product is inferable from the device, the centre liable for a premarket survey of that device using global and local literature search screening would have essential purview for the blended product. For this situation, the lead place is the Center for Devices and Radiological Health (CDRH).

The correct focus will lead the pack to investigate the application and talk with different directions regarding the medical device mix’s signature piece.

The test is that each centre unexpectedly handles matters. The evidentiary norms for medicals and devices are relatively unique. Like this, each centre has its way of thinking on directing the segment under their ward, and they have their thought about what information is protected and powerful enough to help endorsement. It can here and there prompt misalignment inside the survey group, making knocks in the Sponsor Street.

Expertise

Typically, Sponsors building up a medical device mix product have tremendous skill in one of the two constituent territories. They are either too experienced with the medical part or are exceptionally knowledgeable about the device segment. It can represent a test because, paying little mind to the endorsement pathway, the FDA expects them to give ample proof in the two territories. Accordingly, the Sponsor should figure out how to acquire the vital aptitude before presenting their clinical trials’ application for pharmacovigilance.

Cost and time

The FDA necessitates that every constituent piece of a mix product is tried freely just as together. It implies much more proof that should be submitted to the Agency to get a blend of product endorsement. Consequently, the general expense and time for advancement can be a lot more noteworthy than for a solitary substance.

Risk of interaction

The FDA anticipates that Sponsors should take a gander at all the dangers related to connections between the parts. The entirety of the components is in contact with one another, either genuinely or procedurally, which means there might be associations representing a danger to the client. It implies testing zeroed in on moderating those dangers should be directed.

Human factors

The FDA frequently requires a human components assessment for drug-device blend products, particularly for those that are to be utilized by the patient or parental figure. When cross-marking the device and medical constituent parts, there should be sufficient alerts and directions for every segment’s utilization. Those warnings and guidelines are predictable across every aspect. These necessities add additional layers of intricacy, cost, and time to the administrative cycle.

Conclusion:

Here are the top challenges Pepgra explains for the device manufacturers to get FDA approval after many regulatory submissions and safety measures. The entire device manufacturer should follow FDA regulatory measures to get approval easily. Pepgra also offers pharmacovigilance service and pharmacovigilance service providers.

References:

  1. Van Norman, G. A. (2016). Drugs and devices: comparison of European and US approval processes. JACC: Basic to Translational Science1(5), 399-412.
  2. Sorenson, C., & Drummond, M. (2014). Improving medical device regulation: the United States and Europe in perspective. The Milbank Quarterly92(1), 114-150.

 

 

The post Challenges faced during FDA Device Approval Process appeared first on pepgra.

]]>
How to manage risks in clinical studies? Top 5 tips https://pepgra.com/blog/clinical-writing/how-to-manage-risks-in-clinical-studies-top-5-tips/ Wed, 17 Feb 2021 07:12:06 +0000 https://pepgra.com/?p=4133 In-Brief: Fronting ever-increasing costs of running a clinical trial, sponsors must guarantee they are correctly directing their financial plan and resolving the highest risk areas while […]

The post How to manage risks in clinical studies? Top 5 tips appeared first on pepgra.

]]>
In-Brief:
  • Fronting ever-increasing costs of running a clinical trial, sponsors must guarantee they are correctly directing their financial plan and resolving the highest risk areas while preserving patient safety and data reliability in Patient recruitment for clinical trials.
  • How can sponsors implement a vigorous process to allow earlier documentation of emerging risks during a trial?
  • Pepgra blog covers five tips for significant risk levels, categorizing risk and maintaining oversight to confirm that risks and responses are correctly identified, documented, tracked, and achieved throughout the patient recruitment companies’ life cycle and offers patient recruitment clinical trials.

Introduction:

Risk management includes a series of activities or processes undertaken through a clinical trial’s life cycle to recognize, evaluate, monitor, switch, prevent, moderate, communicate, and analyze any factor that threatens the test’s quality. It pertains to participants’ risks and all other steps related to the prosecution, especially the trial data’s quality, consistency, and integrity. Risk management must start at the trial opening so that risk justification can be a part of the protocol and additional essential forms and clinical research patient recruitment process.

Top 5 tips for managing risks in clinical studies

Outlining your levels of risks

Risk is a natural incidence in any trial at the program, study level, site level and working level. Defining it is the first step to attaining control. At a high level, the full risk of a study can be assessed. For eg, a Phase II oncology study would specify a higher risk that needs a more rigorous monitoring strategy than a low-risk Phase IV observational work.

Study risks can also differ based on known, high-performing spots versus new sites with less knowledge of clinical trial recruitment companies‘ helpful area. Finally, operational risk can be projected based on real-time patient acceptance data to compare actual presentation to the forecast.

Evaluating and categorizing risk

The distinct levels of risk at the study, site and working levels, and overall risk valuation can be produced for a protocol and across a program. The Risk Assessment Categorization Tool, One module of the platform, relates an algorithm to generate an overall category score based on the chance, impact and detectability of the risks, permitting sponsors to make a data-driven decision about the most suitable intervention levels.

Concentrating on essential areas of risk

After risks are considered, they can also be riddled through Monitoring’s user interface to highlight those with the most significant impact on a study, enabling sponsors to redirect resources appropriately. With Risk and Issue Management, all study team members can create, view, and manage real-time issues from a single interface using patient recruitment services.

For instance, if the framework recognizes key risks as inordinate underreporting and patient maintenance, the support and CRO can cooperate to guarantee they are checking and controlling these regions for the examination duration. This cycle empowers early usage of preventive activities and can help limit quality disappointments.

Observing and controlling risks

While observing the risks defined and categorized, it’s essential to monitor the changes’ status throughout a study’s life. With automatic metrics, the system makes recommendations to escalation, reduction, or maintenance Monitoring at a site using essential risk indicator scoring for clinical trials patient recruitment. It helps the trial’s project team take action and allows sponsors and CROs to prioritize and target particular areas. The automated process also helps manage growth paths and fulfils regulatory guidance surrounding adapted and triggered site monitoring.

Estimating the efficiency of risk management

As risks are identified, categorized and achieved over time, sponsors and their supportive CRO can view the increasing actions taken month over month, assessing their level of success and determining if the activities accomplished helped bring a site back to a lower risk level in clinical study recruitment.

Ideally, sponsors should see that a more significant proportion of sites are moving into the standard and low-risk categories over time, with an overall decrease in the high-risk types. This transparency level helps with continuous improvement practices and demonstrates full control and compliance with regulatory agencies.

Conclusion

With today’s extensive global trials and virtual project teams using several systems acting in separation, sponsors need an effective method to quicken decision making and close the gaps in trial error. Unifying quality and risk supervision across a single study or a portfolio of studies support revealing signals before they become general issues that disrupt a trial.

References:

  1. Sundar, S., & Olliaro, P. L. (2007). Miltefosine in the treatment of leishmaniasis: clinical evidence for informed clinical risk management. Therapeutics and clinical risk management3(5), 733.
  2. Vincent, C., Taylor-Adams, S., Chapman, E. J., Hewett, D., Prior, S., Strange, P., & Tizzard, A. (2000). How to investigate and analyze clinical incidents: clinical risk unit and association of litigation and risk management protocol. Bmj320(7237), 777-781.
  3. Hall, J. A., Salgado, R., Lively, T., Sweep, F., & Schuh, A. (2014). A risk-management approach for effective integration of biomarkers in clinical trials: perspectives of an NCI, NCRI, and EORTC working group. The Lancet Oncology15(4), e184-e193.

The post How to manage risks in clinical studies? Top 5 tips appeared first on pepgra.

]]>
Significances and Difficulties of E-labelling in the Medical Device Industries https://pepgra.com/blog/medical-devices/significances-and-difficulties-of-e-labelling-in-the-medical-device-industries/ Thu, 11 Feb 2021 12:29:10 +0000 https://pepgra.com/?p=4127 In-Brief: E-labelling is not new evidence; nor is its use partial to the life sciences business. Creating the latest product data available online adds value across […]

The post Significances and Difficulties of E-labelling in the Medical Device Industries appeared first on pepgra.

]]>
In-Brief:
  • E-labelling is not new evidence; nor is its use partial to the life sciences business.
  • Creating the latest product data available online adds value across various industries, particularly in healthcare analytics solutions.
  • Pepgra gives you the challenges and uses of e-labelling (electronic labelling) in the healthcare data analytics companies.

Introduction:

E-labelling (electronic labelling) plays a severe role in the medical devices industry, as it confirms to improve operational efficacy, maintain brand consistency, obtain obedience and support business growth. Hence, the healthcare data analytics solutions move towards adopting end-to-end e-labelling life cycle procedures, starting from label content creation to review and approvals. How are they beneficial and what kind of challenges one would face while adopting them? Let us know in detail.

E-labelling for Clinical Devices – Beneficial factors

As a piece of the EU’s new Clinical Device guidelines, any device producer with a site should distribute client data to it in electronic structure. Other worldwide wards have comparative prerequisites, which put a new accentuation on e-naming as a control: the electronic distributing of naming substance, including Instructions/directions for Use (IFU/DFU).

Better Utility and Data

  • Simple admittance to data, upgraded comprehensibility and more justifiable for clients so they can get all the item data
  • Customization for country-explicit labels and plans
  • Expanded proficiency and utility through online evaluation by a controller and other related Healthcare Data Analysis.

Environment Friendly and Time Saving 

  • Diminished utilization of materials for labels and decreased natural effect
  • The general season of the assembling interaction is abbreviated
  • The big data analytics in healthcare on e-labels can be refreshed distantly for a product

Practicality and Upgraded Understanding Safety

  • The e-labels save cost, paper and space and dispense with the danger of IFU (Instructions for Use) getting lost or isolated from their related devices for healthcare data analytics
  • They are tough and are not liable to blurring or pollution
  • They upgrade device security and improve data trade by making successive updates to the electronic IFUs

Utilizing QR Codes:

  • The QR codes are generally speedy and straightforward and help to associate the controllers and purchasers to the site facilitating consistency and with different prerequisites
  • Given the expansion of cell phone access using free applications, the QR codes are utilized in a broad scope of settings and permit the client to examine the QR codes and access the critical data

E-labelling for Clinical Devices – Difficulties

Multilingual Labelling

  • The labels should be securely and precisely tweaked in proper area dialects to get to a few topographies and hold fast to the country-explicit naming necessities, plans and Administrative prerequisites
  • Any labelling mistakes can make the danger of brand picture harm, particularly during item reviews. Nonetheless, mark Lifecycle Management Solutions (LMS) have become a powerful weapon in the language the board stockpile and a demonstrated, approved naming programming framework will eliminate the apparent complexities of locality language detailing same time meeting country-explicit prerequisites.

Overseeing Complexities of Extension

  • With steady consolidations, acquisitions and developments, the labelling tasks of clinical devices are altogether influenced.
  • A labelling programming framework will improve the name trustworthiness and consistency and help deal with the naming information. All the more successfully, aiding each part of the by and large naming interaction, including adherence to country-explicit necessities and decreasing the requirement for physically observing the naming cycle.

Controlling Mark Deformities

  • Expanding labelling mistakes lead to visit item reviews, other than affecting operational proficiency and benefit.
  • Robotized vision frameworks are acquainted with guarantee more powerful, proficient, and secure methods of zero-imperfection naming.

Absence of Gear and Specialized Capacities

  • Network issues are experienced while checking e-labels with handheld devices for healthcare analytics
  • To guarantee network, versatile, or Wi-Fi internet providers can be utilized. Additionally, scanners, that can peruse, and store labels disconnected can be used, and the labels can be checked later when the device interfaces with an organization

Conclusion:

More or less, e-labelling will decrease the manual intercession and give collected quality data, improve quiet security, consistency, and brand respectability, diminish the natural effect, and in particular, it will be financially savvy. The solitary inquiry device makers should ask themselves now: “would we say we are e-labelling prepared”? Do a complete examination with an Administrative labelling master for clinical devices. Stay educated. Stay consistent. Pepgra gives you the challenges and uses of e-labelling (electronic labelling) in the healthcare analytics companies.

References:

  1. Ângelo, A., Barata, J., da Cunha, P. R., & Almeida, V. (2017, September). Digital transformation in the pharmaceutical compounds supply chain: Design of a service ecosystem with e-labelling. In European, Mediterranean, and Middle Eastern conference on information systems(pp. 307-323). Springer, Cham.
  2. Bolislis, W. R. R., Mortazavi, C., Riccioni, R., Schaeffer, P. E., & Kühler, T. C. (2019). From Print to Screen: Regulatory Considerations to Adopting Innovative Approaches for Patient Information and Safety. Therapeutic innovation & regulatory science, 1-8.
  3. Gassner, U. M. (2007). E-labelling & the Comitology Procedure–Outcomes of the MDD Review Process.

 

The post Significances and Difficulties of E-labelling in the Medical Device Industries appeared first on pepgra.

]]>
Top tips for a medical writer to prepare a successful NDA https://pepgra.com/blog/medical-literature/top-tips-for-a-medical-writer-to-prepare-a-successful-nda/ Tue, 09 Feb 2021 05:09:33 +0000 https://pepgra.com/?p=4120 In-Brief: Medical writers must be subject matter experts of the regulatory control inside and out and know the drug under study in clinical research. To prepare a […]

The post Top tips for a medical writer to prepare a successful NDA appeared first on pepgra.

]]>
In-Brief:
  • Medical writers must be subject matter experts of the regulatory control inside and out and know the drug under study in clinical research.
  • To prepare a successful NDA(New drug application), a medical writer should know all the regulatory submissions basics.
  • Pepgra provides you with essential tips for a medical writer to prepare a successful NDA and provides regulatory writing services.

Introduction:

Medical writing includes writing scientific documents of different types, with regulatory and research-related papers, disease or drug-related and promotional literature, publication articles like journals, manuscripts and abstracts, essential content for healthcare sites, health-related publications or news articles. The scientific data in these documents need to be accessible to suit the target audience’s level of understanding, namely, patients or the general public, physicians or the regulators. Medical writers require an understanding of the medical concepts and terminology, knowledge of relevant guidelines regarding the structure and contents of authentic documents, and good writing skills. They also need to be familiar with searching medical literature, understanding and presenting research data, document reviewing, and editing and publishing requirements. Many resources are now accessible for medical writers to regularly get vital training in medical writing science and art and regularly upgrade their knowledge and skills. The demand for the medical report is growing gradually in pharmaceutical and healthcare communication marketplace. Medical writers can work individually or active as full-time professionals.

Tips for a medical writer to prepare a successful NDA

It’s never too early to start your NDA preparation.

It would help if you began to think about your eventual NDA( New Drug Application) from the moment you start planning for your Pre-IND meeting and IND submission, and well before you have dosed a single patient. For instance:

  • What is the most suitable regulatory path?
  • What are clinical trials needed for the NDA and product label to be supported?
  • Do you plan to seek expedited growth and expedited review?
  • What is production information needed for the NDA?
  • To help your application, do you need long-term toxicology/carcinogenicity studies?
  • What needs to be outsourced?

All these questions should be front and centre as early as possible in your mind. Even though your NDA submission seems far off shortly, early preparation will make the difference between smooth sailing and rough seas.

 

Start assembling your NDA

Far too often, sponsors wait to start compiling their NDA until late in production, which can place undue pressure on resources and timelines. Note, each piece of the NDA you can finish and “put on the shelf” is one bit less that you have to think about later. You can relax a little better as the submission date approaches by knowing what elements can be accomplished early and then proactively completing those assignments during medical writing in clinical trials.

In most cases, by the time the drug reaches the clinic, a significant portion of the nonclinical program will be completed. All of this information can be explicitly pulled into the NDA’s related modules and incorporated as other studies become final. On the clinical side, as soon as you can, you should start reviewing your data and preparing your clinical research reports (CSRs). Before the research is complete, you can even start on the report shell. You can then collect the rest of the components and collect all the required approval signatures until final data is available. Analyses have been carried out using regulatory medical writing in clinical trials. When the time comes to colonize the NDA clinical overview pages, completing these activities would also encourage performance.

Know the related laws and regulatory guidelines

The FDA review division must manage your application, and for NDAs in general, to understand the applicable laws and regulatory guidelines that relate to your specific program. Although rules are legally binding, documents of advice are not in regulatory writing. However, guidance documents include the regulators’ latest thinking, reviewing the NDA (and hopefully approve it). Therefore, it should be done with caution, reason, and consultation with the FDA’s relevant review division to deviate from these guidelines.

Use standardized templates

When you start writing your IND, make sure you use uniform models and that a standard style guide regulates all authorship. To put together a masterful submission, you don’t have to have a fancy eCTD template kit, but you can make sure your submission documents conform to a standard format and style wherever possible. The same grammatical, punctuation, numbering, abbreviation, and text conventions should be practical by all writers working on the submission and should understand how to preserve the dignity of the underlying document’s formatting and styles.

Because of all the data and scientific weight in the submission, this may seem like a minor detail. Still, a polished, well-written, regularly formatted application package that conforms to current regulatory document requirements goes a long way to the success of a submission.

Enter your study data

To get all your early-phase data entered, queried, cleaned, and locked, do not wait until Phase 3. It will save many headaches, later on, to get an early start on data management and free up time to emphasis on other tasks as the NDA submission date approaches.  If the data is locked, you can start producing study-specific tables, listings, and figures to help study reports and summaries inside the NDA.

Request a Pre-NDA meeting

Your primary late-stage opportunity to get feedback from the FDA on your request is the Pre-NDA meeting. As such, you must put a lot of thought into the topics of discussion you want to raise and ensure that the questions you pose are straightforward, well-supported, and placed to allow the FDA to make a meaningful response. Suppose any particular elements of your program or submission may affect successful filing or review and have not been addressed through previous interactions. In that case, it is now time for the FDA to discuss them. Prepare your claims and your “Plan B” well in advance and consider the possible impacts of both of these on your submission’s content and deadlines from medical writing solutions.

Establish realistic timelines for every aspect

Communicate deadlines and obligations, and follow-up often, to all relevant stakeholders and participants. The overwhelming amount of information that needs to be obtained, processed, interpreted, and shared inside the NDA should be careful in the timelines. Establishing an NDA “tracker” is one of the easiest ways to do this. The most important thing if the tracker is part of a sophisticated software package specifically designed for regulatory submissions, a simple spreadsheet, must contain some crucial and actionable submission information. It requires listing constituent documents with sufficient granularity, the parties involved, each document’s current status, and the due dates.

Allow at least a month for publishing your submission

Publishing timelines are often looking at as something that can be dense if “more critical” tasks such as authoring or scientific review take longer than planned. However, it can sometimes cut to publish timelines, mainly if study reports and different NDA parts are being firm up as you go. It is important not to underrate the vast amount of effort and time involved in setting up a proper submission. Even with leading-edge technologies, publishing needs a considerable amount of time, not to mention quality control activities and final validation of the request, to ensure consistent document formatting, hyperlinking, bookmarking, and arrangement within the eCTD system. Publishing timelines should be compressed with care, prospectively wherever possible, and in close contact with the publishing team at all times.

Take the big picture to guarantee a good submission

Take a minute to consider the picture of what it takes to confirm a good proposal, whether this is your first NDA or just one of many that you’ve done. Pulling an NDA together is a vast undertaking that takes considerable time and effort to complete an investment. Consider your organization or institution’s resources and skills and the internal ability to execute the necessary tasks in your desired timelines, and then make a reasonable assessment of whether additional support is required.

Conclusion:

Pepgra offers several scalable services to help our clients yield high-quality, compliant NDA submissions. Suppose you are thinking of outsourcing your whole NDA(New Drug Application). In that case, you need a partner to assist with publishing and submitting. Even if you have just one or two papers that may benefit from an accomplished clinical pharmacology expert’s examination, Pepgra will help regardless of your need along with Regulatory Writing Service in Clinical Research.

References:

  1. Lies, R. L. (1984). Preparing the Nda Summary: How to, How Not to and why. Clinical Research Practices and Drug Regulatory Affairs2(3), 259-271.
  2. Schwarz, S. W., Dick, D., VanBrocklin, H. F., & Hoffman, J. M. (2014). Regulatory requirements for PET drug production. Journal of Nuclear Medicine55(7), 1132-1137.

The post Top tips for a medical writer to prepare a successful NDA appeared first on pepgra.

]]>