A Comprehensive Guide to Clinical Trial Protocol Design & Development
A clinical trial protocol encompasses study objectives, methodologies, and participant selection while complying with ethical standards and regulatory requirements. A well-designed protocol also increases a trial’s chance of successful completion, including planning for patient recruitment and retention and anticipating challenges.
The science of drug and medical device development is expanding rapidly, opening up new possibilities to manage or cure many heretofore untreatable diseases and conditions. Likewise, protocols have advanced, with new technologies and methodologies offering exciting opportunities but adding complications. Designing a protocol is an intricate endeavor, and this comprehensive guide explains key aspects of protocol development.
Table Of Contents:
- Key Components Of A Clinical Trial Protocol
- The Protocol Development Process
- Statistical Methods Used In Clinical Trial Protocols
- Tools And Technologies For Protocol Design
- Challenges In Clinical Trial Protocol Design
- Best Practices For Protocol Optimization
- Case Studies And Real-World Examples
- Current And Future Trends In Clinical Trial Protocol Design
- Conclusion
- Frequently Asked Questions (FAQs)
Key Components Of A Clinical Trial Protocol
A clinical trial methodically and rigorously tests scientific hypotheses to achieve clear, actionable objectives. However, because human subjects are involved, the protocol must also follow strict ethical standards in compliance with regulatory bodies. The key components of the protocol include study objectives and hypotheses, design and methodologies, and participant selection/ eligibility.
Study Objectives And Hypotheses
First, researchers define the study’s objectives and formulate testable hypotheses.
Defining Study Objectives
Study objectives should conform to SMART (specific, measurable, achievable, relevant, time-bound) principles. For example, the vague goal “evaluate safety and efficacy of cardiovascular drug X” would be better transformed into the SMART goal “measure the effect of cardiovascular drug X on Y population over Z period and compare results to the current standard of care.”
Primary objectives focus on the central research question, such as evaluating a treatment’s efficacy or safety. Secondary objectives offer additional questions, like exploring subgroup effects or long-term outcomes.
Formulating Hypotheses
To meet the primary and secondary objectives, researchers formulate hypotheses to test during the study. These hypotheses should be based on prior research, address knowledge gaps, and demonstrate that they are biologically plausible. Hypotheses reflect relationships between variables using constructs such as:
- If/then format: “If patients receive drug X, then arrhythmia episodes will decrease by 20% compared to placebo.”
- Correlational format: “There is a negative correlation between drug X dosage and arrhythmia frequency.”
- Null hypothesis (H₀) states no effect (e.g., “drug X has no impact on arrhythmia”), while the alternative hypothesis (H₁) represents the expected outcome.
Aligning Objectives And Hypotheses
Objectives and hypotheses must logically align, with primary hypotheses testing primary objectives and secondary hypotheses testing secondary objectives. For example, if a primary objective is to test efficacy, the hypothesis could predict a specific treatment’s effectiveness. Likewise, secondary hypotheses could test ancillary outcomes. All hypotheses should be operationalized with defined variables and statistical endpoints.
Study Design And Methodologies
A study’s protocol must suit the specific study type and methodology to create a robust, efficient clinical trial that meets the primary objectives while maintaining scientific integrity and protecting participant safety. Researchers can choose from several trial methodologies depending on the study's objectives.
Single-Arm/ Non-Randomized Trial
In a single-arm, non-randomized trial, all participants receive the treatment being tested with no control group. This method is frequently used in Phases 1 and 2 to collect preliminary efficacy and safety data. To design a protocol for this type of trial, researchers:
- Identify a clear objective, such as measuring drug X’s safety and efficacy.
- List precise endpoints and outcome measures.
- Develop detailed participant inclusion and exclusion criteria.
- Outline the intervention, dosing, and follow-up schedule.
- Define data collection methods and a statistical analysis plan.
- Address potential biases due to the lack of a control group.
Randomized Control Trial
A randomized control trial (RCT) randomly assigns participants to either an experimental or a control group, allowing researchers to better evaluate treatment efficacy than during a single-arm trial. Developing an RCT trial entails the following steps:
- Formulate a primary hypothesis and any secondary hypotheses.
- Design the randomization process.
- Establish the sample size based on statistical power calculations.
- Define primary and secondary outcome measures.
- Develop a detailed timeline for recruitment, intervention, and follow-up.
- Codify procedures for blinding (if applicable) and data management.
Master Protocol Designs
Master protocol designs, such as basket, umbrella, or platform trials, are complex protocols that test multiple diseases or treatments.
Basket trials examine a single therapy with a common molecular alteration across multiple diseases. Protocols define the common biomarker or genetic alteration of interest, create separate cohorts for each disease type, and develop statistical methods to analyze multiple cohorts simultaneously.
Umbrella trials focus on a single disease and test multiple targeted therapies, with participants stratified into subgroups based on molecular alterations. Umbrella trial protocols define the disease and its molecular subtypes, outline separate treatment arms based on subtype, and detail how subjects will be assigned the appropriate arm.
Platform trials leverage a perpetual study structure wherein treatment arms can be added or removed over time. The protocol sets the criteria for introducing new treatment arms or discontinuing ineffective ones, describes adaptive randomization procedures if applicable, and creates methods for controlling type I errors across multiple comparisons.
Medical Device Trials
Medical device trials fall into two categories: investigational device exemption (IDE) studies for high-risk devices and non-significant risk (NSR) trials for lower-risk devices. For either type, protocols clearly describe the device and its intended use and outline safety monitoring procedures. The protocols also define what training investigators and participants need to operate the device and outline device accountability and handling procedures. IDE trial protocols also include risk analysis, while NSR trials must justify their NSR designation.
Participant Selection And Eligibility
Protocols determine who is or is not eligible to participate in the clinical trial through inclusion and exclusion (I/E) criteria. These criteria are based on scientific, ethical, and regulatory considerations, balancing real-world applicability with study goals. They minimize confounding variables, enhance reproducibility, and maintain fairness by avoiding unnecessary exclusions.
Inclusion Criteria
Inclusion criteria like age, gender, medical condition, disease stage, or treatment history determine participant eligibility. Further considerations like geographic location or the ability to comply with study requirements may be added. These criteria create a targeted study population for reliable, generalizable results.
Exclusion Criteria
Exclusion criteria are characteristics that disqualify individuals from participation, such as conflicting medical conditions, medications that interact with the intervention, or high-risk factors. These criteria protect participants from unnecessary harm and ensure the trial's safety and integrity.
The Protocol Development Process
Developing a protocol entails several stages: planning, writing, submitting to regulatory authorities, developing operational details, and ensuring quality.
Planning And Collaboration With Stakeholders
In the planning and collaboration stage, researchers define clear, measurable primary and secondary objectives that align with the research question. Researchers conduct a thorough literature review to uncover knowledge gaps and justify the study’s rationale before designing the study, which is based on intervention type, target population, and available resources.
During this process, researchers collaborate with various stakeholders, including clinicians, statisticians, and patient advocates, to refine the study design and ensure feasibility. Throughout, ethical considerations and regulatory guidelines must be considered.
Writing And Reviewing The Protocol
Researchers draft the protocol document, commonly using a standardized template provided by regulatory bodies or institutions, such as:
- ICH M11 Clinical Electronic Structured Harmonised Protocol Template, which the FDA recommends for structured protocol development.
- NIH templates for different types of research, including Phase 2 and 3 clinical trials requiring FDA IND/IDE applications and behavioral/social science studies.
- The National Cancer Institute's Cancer Therapy Evaluation Program protocol templates for Phase 1 and 2 trials, organ dysfunction studies, and informed consent documents.
These templates include background information, objectives, study design, participant selection criteria, treatment details, and statistical considerations. Once drafted, the protocol undergoes a rigorous peer review process to identify and address potential issues. After review, the document is revised based on feedback and finalized.
Regulatory Submission And Approvals
Gaining regulatory approval for clinical trials often involves multiple agencies and regulations to protect participant safety and data integrity. For instance, clinical trials conducted in the U.S. must meet FDA and HIPAA standards, while studies performed in the EU follow EMA and GDPR guidelines. The protocol document includes plans for complying with these local and regional regulatory requirements.
FDA
Drug trials require an IND application with a detailed protocol, preclinical data, and manufacturing information. The FDA has 30 days to review and approve the applications. Once approved, the study can begin. If the trial is successful, sponsors submit an NDA.
Medical device trials follow a similar path. First, researchers submit their application for an IDE or NSR trial. For Class III high-risk devices, the FDA requires a pre-market approval application.
All trials must follow the FDA's data standards for case report forms (CRFs) and data analysis, and companies must provide post-marketing surveillance or pharmacovigilance. The FDA has also released draft guidance covering decentralized elements and real-world data (RWD).
HIPAA
HIPAA safeguards protected health information (PHI) and guides its use in research. Participants must authorize using their PHI, and data must be deidentified to protect privacy. Sponsors need approval from an institutional review board (IRB) or privacy board for the use/disclosure of PHI in research. Additionally, companies must conduct routine risk assessments and provide staff training.
EMA
Drug trials require a clinical trial application submitted through the clinical trial information system. This application includes Part I (scientific and medicinal product documentation) and Part II (ethical and operational considerations) and is reviewed by EU member states.
Medical device trials are subject to medical device regulation policies that underwent significant changes in 2024-2025. The new guidelines carry stricter requirements for quality management, technical documentation, and clinical evaluations. Devices must be registered in the EUDAMED database, and the EMA has expanded its role in combination product approval. After January 2025, high-risk devices undergo joint clinical assessments under the new Health Technology Assessment Regulation.
GDPR (General Data Protection Regulation)
GDPR standards protect participant privacy and establish data-management guidelines. First, companies must conduct a data-protection impact assessment to identify and mitigate privacy risks. Participants must grant their informed, explicit consent, including understanding how their data will be used and transferred and any associated risks.
For data management, companies must protect personal data throughout the trial, follow data-minimization and purpose-limitation principles, and establish robust safeguards for transferring data outside the EU.
Developing Operational Details
Operational details are another necessary component of the clinical trial protocol to ensure the trial is conducted systematically, ethically, and in compliance with regulatory standards in the following areas:
Participant Management
- Recruitment and enrollment strategies identify and enroll eligible participants.
- Informed consent procedures educate participants and obtain documented consent.
- Inclusion/exclusion criteria ensure a consistent study population.
Study Procedures
- A schedule of activities is the timeline for tests, procedures, medications, and follow-ups.
- Treatment administration includes dosage, route, and frequency of investigational products or interventions.
- Stopping rules are criteria for discontinuing treatment for individual participants or the entire trial.
Data Collection And Management
- CRFs record participant data.
- Data-handling procedures cover data entry, storage, and confidentiality.
- Source data verification (SDV) cross-checks data against original records.
Safety Monitoring
- Adverse-event reporting methods identify, document, and report adverse events.
- Risk-mitigation plans protect participants.
Quality Control
- SOPs for all trial activities ensure consistency across sites.
- Training requirements for investigators and staff must be specific to the protocol.
Statistical Considerations
- Sample-size calculations justify the number of participants.
- Analysis plans describe methods for evaluating primary and secondary outcomes.
Ethical And Regulatory Compliance
- Ethics approval includes submission to IRBs or ethics committees.
- Data privacy compliance follows regulations like HIPAA or GDPR.
Trial Oversight
- Monitoring plans for on-site or remote monitoring safeguard protocol adherence.
- Audit trails document protocol amendments and deviations.
Statistical Methods Used In Clinical Trial Protocols
QA in protocol development begins with following good clinical practice (GCP) guidelines and meeting regulatory requirements. Protocols must also demonstrate their commitment to participant safety, data integrity, and ethical standards. Additionally, QA procedures ensure the protocol is consistent with the study’s objectives and statistical analysis plan and that any amendments are adequately documented. QA personnel provide an independent review of the proposed protocol, compare CRFs to protocol objectives, and implement QC plans for each operational step in the study.
Statistical Methods Used In Clinical Trial Protocols
Several statistical methods are used in clinical trial protocols to prove the validity and reliability of the study’s results and inform clinical decision-making. These methods are used throughout the trial’s design, conduct, and analysis stages.
Sample-Size Calculation Methods
The correct sample size demonstrates the trial has adequate statistical power to detect meaningful treatment effects. The sample size is determined based on factors such as study design, type of outcome measure, expected effect size or clinically meaningful difference, desired statistical power, significance level, expected data variability, and the anticipated dropout rate. Approaches to determining sample size include:
- Power analysis estimates the necessary sample size to detect a specified effect size with a given confidence level, balancing false positive and false negative risks.
- Precision-based approach determines the desired precision level when estimating the treatment effect, commonly used in pilot or feasibility studies.
- Resource-based approach calculates the maximum possible sample size based on available resources to gauge the detectable effect size.
- Adaptive designs allow sample sizes to be re-estimated during the trial based on interim results to conserve resources.
Statistical Analysis Plan Development
A statistical analysis plan (SAP) details how the trial data will be analyzed and is often required as part of the approval process. The plan is developed early in the trial process as a collaborative effort between statisticians, study team members, investigators, data managers, and regulatory experts. The SAP outlines specific statistical methods, defines analysis populations, identifies endpoints and tests, and specifies data handling procedures. The SAP should be finalized before the data is unblinded to prevent bias.
Handling Missing Data
Missing data can damage the study’s statistical power and introduce bias; thus, the SAP must list procedures for handling missing data. Ideally, precise data-collection procedures minimize missing information. However, if data are missing, several imputation methods help researchers track them and mitigate their effects, such as:
- Last observation carried forward
- Multiple imputation
- Complete case analysis
- Sensitivity analyses to assess the impact of missing data on results
- Mixed models or other statistical techniques that can handle incomplete datasets.
Modeling And Biostatistical Methods
Advanced statistical methods enhance data analysis, but they should be listed in the SAP and align with the study’s objectives and design. Approaches include:
- Survival analysis for time-to-event data
- Mixed-effects models for longitudinal data
- Bayesian methods for adaptive trial designs
- Propensity-score matching for observational studies
- Non-inferiority and equivalence testing methods.
Tools And Technologies For Protocol Design
Significant advances in protocol-design technologies support complex trials and improve data collection and analysis. As these technologies evolve, they promise to make clinical trials more efficient, patient-centric, and data-rich.
Protocol Design Software And Tools
Specialized design software streamlines protocol creation and management. Features like customizable templates for different study types, collaboration capabilities, version control, and regulatory guidance improve the design stage. Frequently, these tools integrate with other clinical trial management systems to improve trial execution.
AI And Machine Learning Applications
AI and machine learning (ML) can optimize protocol design and risk assessment by detecting design flaws or inconsistencies, predicting recruitment hurdles, and optimizing endpoint selection and measurement. Examples of these techniques include:
- Clinical trial risk tools use natural language processing to analyze protocols and estimate the risk of unsuccessful trial completion.
- Predictive modeling identifies the best sites for trials, which can cut recruitment times and costs.
- Protocol optimization using AI analyzes historical trial data to improve study design and mitigate regulatory risks.
Digital Health Technologies
Digital health technologies (DHTs) encompass a wide range of devices, apps, and platforms that enhance data collection and participant engagement. DHTs increase the amount of objective and RWD collected during the trial, expand participant access, and engender novel clinical endpoints.
Wearable devices like smartwatches continuously or periodically measure clinical features for automatic, remote data collection. Mobile health apps allow participants to report data remotely, and telemedicine platforms provide remote participant monitoring.
Challenges In Clinical Trial Protocol Design
Designing a clinical trial protocol entails negotiating several competing factors, such as scientific rigor, feasibility, and patient recruitment.
Balancing Scientific Rigor With Practical Feasibility
Protocols must be scientifically sound to generate valid results, but overly complex designs can be costly or impossible. For example, a protocol requiring frequent, complex assessments may provide excellent data, but participant dropout and increased site burden could hamper the trial.
Early collaboration with clinical operations, data management, and biostatistics teams improves protocol development, and cross-functional input identifies potential operational challenges. Analyzing data from similar trials can also inform protocol design decisions, and business intelligence tools can be used to assess historical site or regional performance.
Focusing on patient-centricity also improves feasibility. Every procedure should be evaluated based on its contribution to key objectives to avoid unnecessary complexity. Participant burden should be considered, including the number of site visits, invasive procedures, and time commitments. Patient focus groups and disease advocacy groups can provide valuable feedback on protocol design to improve recruitment and retention.
Likewise, sites commonly juggle multiple trials, so site capabilities, resources, and workload should be factored into the protocol design. Feasibility studies with potential investigator sites can reveal potential problems with the protocol before the design is finalized.
Recruitment And Retention Issues
Protocol development includes planning for patient recruitment and enrollment, which can make or break a clinical trial. Globally, 55% of trials fail due to insufficient enrollment, and in the U.S., over 80% fail to meet enrollment goals, with an average attrition rate of 30%.
Several factors contribute to low enrollment, including overly stringent I/C criteria, competition from similar trials, geographical limitations, and lack of awareness. To combat these, researchers develop a subject recruitment and retention plan (SRRP). The SRRP creates strategies for identifying potential participants, performing community outreach, collaborating with patient advocacy groups, and leveraging digital platforms like social media.
Best Practices For Protocol Optimization
Applying best practices helps researchers develop effective, patient-centric protocols that meet global standards.
Enhancing Protocol Readability
Clinical trial protocols should follow plain-language guidelines to explain clinical outcome assessments to researchers and participants, avoiding technical jargon or complex terminology. Documents should be well-structured with clear headings and subheadings and use visual aids. During development, documents should be subjected to readability assessments and patient feedback and revised to reach lower reading levels.
Aligning With Global Standards
Protocols should follow global standards for ethics and scientific rigor like ICH E6(R2) GCP Section 6, which outlines 15 key protocol elements. These elements cover general background information, trial objectives, design, subject selection, treatment, efficacy and safety assessments, statistics, quality control, ethics, data handling, and publication policy. Additionally, the ICH M11 guideline provides a harmonized electronic protocol template.
The ethical principles of the Declaration of Helsinki and relevant regulatory requirements from agencies such as the FDA and EMA must also be followed. Finally, the WHO Trial Registration Data Set should be included in the protocol.
Leveraging Real-World Evidence
Real-world evidence (RWE) analyzes RWD from numerous sources to provide actionable insights that improve protocol design. RWE informs feasibility assessments, optimizes inclusion/exclusion criteria, identifies potential participants, and enhances site selection. RWE can also help researchers identify possible issues with the protocol during the development stage.
Creating Patient-Centric And Diversity-Focused Approaches
Recruitment is the first piece of the participant engagement puzzle, but retention strategies must also be in place to minimize attrition rates. Additionally, today's clinical trials are expected to recruit diverse populations, which requires targeted strategies.
Patient Engagement And Retention Strategies
Protocol designs should minimize the burden placed on participants as much as possible like minimizing study visits and procedures; planning flexible scheduling options; providing support services for patients and caregivers, such as transportation assistance and reminders; and offering ethically appropriate incentives.
Retention strategies should be developed beforehand. They should include regularly checking in with participants, personalizing follow-ups, and sharing interim results and study updates. Internally, investigators should monitor attrition rates and reasons to adjust strategies, plus flag participants at risk of dropping out.
Decentralized And Hybrid Trials
Hybrid and decentralized trials (DCTs) offer participants greater flexibility by reducing or eliminating the need for on-site visits. Telemedicine, home health visits, direct-to-patient drug shipments, wearable devices, and mobile apps can minimize patient burden and improve retention.
Addressing Diversity And Inclusion
Recruiting diverse participants who reflect the target population is necessary to develop safe and effective therapeutics and meet FDA guidelines. Historically, women and minorities have been underrepresented in clinical research, but regulatory bodies and researchers are working to improve diversity and inclusion in clinical trials. Protocol design should include strategies for recruiting participants from diverse populations, consider cultural factors hindering participation, and encourage collaboration with community leaders to build trust.
Case Studies And Real-World Examples
Several case studies demonstrate that when companies apply best practices to protocol development, they can improve participant recruitment and retention, leverage new technologies, and effectively apply RWE.
Successful Clinical Trial Protocols
Salford Lung Studies
This GSK-sponsored study evaluated COPD treatment using a real-world setting with minimal exclusion criteria. 2,802 patients were seen by their existing GPs, and the trial enjoyed a 93% retention rate and conclusive clinical results.
Avelumab Approval
RWE helped Avelumab achieve approval for metastatic Merkel cell carcinoma. Electronic health record data provided a benchmark for chemotherapy efficacy, and Avelumab's performance was compared to the RWD.
ADAPTABLE Trial
Over 15,000 patients participated in this massive DCT to determine optimal aspirin dosage for preventing heart attacks and strokes. The trial used ticketed invites for recruitment as well as eConsent and eSource for remote data capture and patient-reported adverse events.
LIFE-BTK Trial
Abbott's peripheral artery disease (PAD) treatment trial recruited investigators and sites with access to underserved communities. In particular, the trial focused on African American, Hispanic, and Native American patients who are disproportionately affected by PAD.
Current And Future Trends In Clinical Trial Protocol Design
Clinical trial protocols have become increasingly complex to create innovative therapeutics. In particular, adaptive and master protocols offer flexibility and increased efficiency, especially for oncology and rare disease research.
Adaptive And Master Protocols
Adaptive clinical trial designs allow adjustments to the protocol during the trial based on interim data analysis. Sample sizes, treatment arms, and endpoints can all be modified as the trial progresses. Adaptive trials can accelerate drug development timelines and conserve resources and are increasingly accepted by regulatory bodies, particularly for exploratory studies.
Trends in adaptive protocols include incorporating multiple study populations into a single protocol, using adaptive randomization to optimize treatment allocation, and seamless Phase 2/3 designs that shorten development time.
Master protocols use an overarching framework to evaluate multiple therapies or diseases in a single study. They are categorized into three main types: basket trials, umbrella trials, and platform trials.
Master protocols are increasingly used for oncology research, especially biomarker-driven studies, and are a growing trend in rare disease research because they improve recruitment.
Adaptive and master protocols enhance RWE integration, rely on advanced technologies like AI/ML, and refine statistical methods to address complex trial designs. Finally, these protocols require close collaboration among stakeholders to streamline implementation and approval processes.
Conclusion
Clinical trial protocols are the backbone of therapeutic and medical device research. A fully optimized protocol creates a feasible, scientifically valid, ethical clinical trial or study that meets regulatory requirements. As the science of drug development advances to include complex, flexible trial designs and incorporate new technologies, protocols are rapidly evolving to become more efficient and patient-centric.
Frequently Asked Questions (FAQs)
Below are FAQs regarding clinical trial protocol design:
1. What is a clinical trial protocol?
A clinical trial protocol is a document that describes how a clinical trial will be conducted, including its objectives, design, methodology, statistical considerations, and organization. It ensures participant safety and data integrity.
2. What are the key components of a clinical trial protocol?
Key components include background information, objectives, study design, subject selection criteria, treatment details, efficacy and safety assessments, statistical methods, ethical considerations, and data handling procedures.
3. What is RWE, and how does it impact clinical trial design?
RWE evaluates a medical product's safety and efficacy using RWD sources like electronic health records and claims data. RWE optimizes trial design, compares treatment effectiveness, and supports ongoing safety assessments.
4. How are DCTs and hybrid trials changing protocol design?
DCTs and hybrid trials reduce clinical site visits, improving accessibility and retention. They allow for more flexible protocols, enhanced data collection through wearables/apps, and increased inclusion of diverse populations.
5. How are clinical trials addressing the need for increased diversity?
Efforts to improve diversity include engaging diverse communities, selecting trial sites in underrepresented areas, providing culturally appropriate materials, and using digital tools to increase participation flexibility.
6. What role does AI play in clinical trial protocol design?
AI analyzes vast amounts of medical literature and patient data, refines research questions, identifies optimal endpoints, and simulates trial scenarios to predict outcomes.
7. How does the focus on patient-centricity affect protocol design?
Patient-centric approaches involve gathering patient input during protocol development, enhancing participant engagement, and designing flexible protocols that accommodate patients' lifestyles and reduce participation burden.
8. What is an adaptive design in clinical trials?
An adaptive design allows for pre-planned modifications to trial parameters based on interim data analysis. This can include changes to sample size, treatment arms, or endpoints, offering greater flexibility and efficiency in drug development.
9. What are the benefits of using adaptive designs in clinical trials?
Benefits include improved statistical efficiency, the ability to address ethical considerations more quickly, enhanced understanding of treatment effects, and increased flexibility in completing the trial.
10. How do regulatory agencies view adaptive design clinical trials?
Regulatory agencies, like the FDA, have guided adaptive designs and generally support their use when properly implemented. However, they emphasize the importance of maintaining trial integrity and controlling type I error rates.
EXPERT INSIGHTS ON PROTOCOL DEVELOPMENT
-
Automation: From Protocol Definition To Submission
Three members of Merck's global clinical data integration team discuss how automating various parts of the clinical trial process can improve data integrity.
-
If Sponsors Invite Patients To See The Final Draft Protocol, It's Already Too Late
Consultant Kamila Novak dives into the importance of early patient input — and why sponsors have a hard time getting it (and implementing it).
-
Digitizing The Clinical Protocol: Small Steps For Seismic Change
A proliferation of clinical trial platform systems has meant increased friction for data exchange. Discover how TransCelerate is tackling this problem with small steps that yield big results.
-
Improving Study Efficiencies With Protocol Templates For Nonclinical Studies
Clinical research organizations (CROs) are often required to adhere to a variety of sponsor-specific protocol and/or reporting templates, often presenting the same information in various places or in different ways, depending on sponsor-specific requests. Maintaining multiple templates requires a significant amount of time and resources.
-
3 Protocol Design Strategies To Enhance Patient Recruitment
How many of the clinical trials that your company has conducted have met the initial recruitment timelines? How often have you seen recruitment issues increase costs or cause delays in your company’s operational objectives? Nearly 80 percent of clinical trials experience delays or even early termination due to recruitment issues.1 These delays not only negatively affect the sponsor company but also keep patients from getting potentially life-saving treatments in a timely manner (if at all).
EDITORIAL PERSPECTIVES ON PROTOCOL DEVELOPMENT
-
The Impact Of Protocol Design On An IRT System
DTP and home treatment studies can get expensive — and be a hassle to patients — if the IRT element isn’t well planned for during trial design. In their SCOPE Summit presentation, Irina Grishina and Kelsey Kern of CSL Behring plan to share their personal anecdotes and lessons learned from both types of trials as they pertain to IRT.
-
Master Trial Protocols: Better For Pharma, Better For Sites
Over the last few years there have been increased discussions in the industry around a master protocol approach to clinical trials. Those discussions have really accelerated over the past year, spurred on by the FDA, which in September 2018 released draft guidelines on this topic for the industry.
-
Bayer Overhauls Its Clinical Trial Planning Process
In 2015 Bayer decided to take a hard look at how it selected patients and sites, and how it prepared protocols and conducted planning for trials. A transformation program was put in place by senior management to oversee those changes and bring insights to the internal development teams.