Thursday, February 13, 2025
Home Blog Page 103

Clinical Research Job Interview Questions and Answers 2024

0

This blog is written after consulting many interviewers and these are the common questions which are being asked in clinical research job related interviews. You can understand after reading this, that almost all questions are related to basics of clinical research. It is not possible to provide in depth details of each topic so we provide the links to each questions so that you can read in details.

What is informed consent?

Before determining whether to participate, informed consent is the method of discovering crucial information about a clinical trial. The provision of information for the participants is also a continuous process during the study.

The doctors and nurses involved in the trial explain the details of the study to help someone decide whether to participate or not. If the participant’s native language is not English, translation assistance can also be provided.

The research team also offers an informed consent document that contains specifics of the project, such as its purpose, duration, required procedures, and key contacts.

More at: https://lifepronow.com/2020/07/03/informed-consent-defination-and-informed-consent-form-example/

The informed consent statement outlines risks and potential benefits. Then the person decides whether to sign the contract or not. Informed consent is not a contract, and the defendant will withdraw at any time from the trial.

What is Primary and Secondary End points

Primary end point are the specific event that the study is designed to assess the effect of the drugs upon. It answers the most important question of the clinical trial. They are also called primary outcome measure.

Secondary end point are additional events of interest, but which the study is not specifically powered to assess. They are also knows as Secondary outcome measures.

More at: https://lifepronow.com/2020/06/29/primary-endpoint-and-secondary-endpoint/

Orphan Drug Designation in US, EU and Japan
The Drug approval process in EU
Drug Approval Process in India

What is treatment arm?

A group of subjects that receives a specific intervention/treatment, or no intervention in clinical trial. Group of subjects who receive treatment, are called treatment arm.

Active comparator arm:Group of subjects receive standard treatment

Placebo comparator arm: Group of subjects receive standard treatment

Sham comparator arm: group of subjects receive same procedure or device without active component or process. (It is like Placebo comparator arm but for medical devices and procedure)

What is Surrogate end point?

As per National Cancer Institute: “In clinical trials, an indicator or sign used in place of another to tell if a treatment works. Surrogate endpoints include a shrinking tumor or lower biomarker levels.”

Alternatively of stronger metrics such as longer lifespan or better quality of life, they could be used as the test results can be assessed earlier.

Using surrogate endpoints in clinical trials that allow for earlier approval of new drugs for treating severe or life-threatening diseases , such as cancer. Surrogate endpoints are not necessarily accurate markers or measures of how well a treatment operates.
More at: https://lifepronow.com/2020/07/05/surrogate-endpoint-marker-definition-and-examples/

What do you understand by Randomization?

As per the National Cancer Institute “In research, the process by which participants in clinical trials are assigned by chance to separate groups that are given different treatments or other interventions.

Neither the researcher nor the patient chooses what medication or intervention they should receive.

Using chance of assigning people to groups means that the results of the care or intervention obtained by the participants can be more equally compared.

More at: https://lifepronow.com/2020/07/13/randomization-in-clinical-trialsdefinition-and-types/

What do you know about ICH and GCP?

The International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use is the full form of ICH.

Guidelines provided by ICH can be divided into 4 categories: Quality (Q), Safety (S), Efficacy (E) and multidisciplinary guidelines (M).

These guidelines help to harmonize technical guidelines which are being followed by many countries for drug registration.
Read about missing and member countries: https://www.ich.org/

GCP’s full form is good clinical practice. It is provided by ICH.

ICH E6 (R2) segment provides detailed information about GCP. Good clinical practice offers a system of guidelines to ensure the health of participants in the study and the quality and validity of data. GCP contains 13 principles which can be studied here. https://ichgcp.net/2-the-principles-of-ich-gcp-2

Where do the ideas for trials come from?

Ideas typically come from researchers for the clinical trials. After the researchers test new therapies or procedures in the laboratory and in animal studies, the experimental treatments with the most promising laboratory results are moved into clinical trials.

More and more information are gained during a trial about an experimental treatment, its risks and how well it may or may not work. But In general practice, It is decided as organization level.

Organization decides the therapeutic areas in which it wants to develop the molecule. And this is decided based on various factors such as market size, competition, target country, nature of disease etc.

What are the different types of clinical studies?

There are two main type of trials: Observational clinical trial and interventional clinical trial

“Interventional clinical study is performed with the purpose of studying or representing clinical or pharmacological properties of drugs/devices, their side effects and to establish their efficacy or safety.”

Whereas observational studies,are those studies where intervention is not given (treatment, drug, surgery). There are many type of clinical studies and you can read about each study in detail here: (case report or case series, ecologic, cross-sectional (prevalence study), case-control and cohort studies)

More at: https://lifepronow.com/2020/05/12/clinical-trial-introduction-types/

Interviewer can ask about each individual study type as well. so read all the sub-types as well in details.

What are the Phases of clinical trials?

Clinical trials are conducted in four phases. Each Trial have different purpose and help scientists answer different questions:

In Phase I trials, researchers test an experimental drug or treatment in very small group of population (20-80) for the first time in order to evaluate its safety, determine a safe dosage range, and to identify side effects. (Maximum tolerated dose assessment)

In Phase II trials, the experimental study drug or treatment is given to a larger group of population (100-300) to see if it is effective and to further evaluate its safety. (Efficacy)

In Phase III trials, the experimental study drug or treatment is given to large groups of population (1,000-3,000) to validate its effectiveness, monitor side effects, compare it to commonly used treatments, and assemble information that will allow the experimental drug or treatment to be used very safely. (effectiveness)

In Phase IV trials, post-marketing studies describe additional information including the drug’s risks, benefits, and best possible use. (Post Marketing surveillance)

More at: https://lifepronow.com/2020/05/11/phases-of-clinical-trials/

What is “expanded access”?

Expanded access is a means by which manufacturers, under certain circumstances, make investigational new drugs available to treat a patient(s) with a serious illness or condition that can not participate in a controlled clinical trial.

Most human use of the investigational new drugs takes place in controlled clinical trials conducted to measure the safety and efficacy of the new drugs.

Data from these trials are used in order to determine whether a drug is safe and effective, and provide as the basis for the drug marketing application.

Occasionally, patients do not qualify for these controlled trials because of other health problems, age, or other factors, or are or else unable to enroll in such trials (e.g., a patient may not live sufficiently close to the clinical trial site).

For patients who are unable to enroll in a clinical trial of an investigational drug but have a significant disease or disorder that could benefit from the drug therapy, FDA regulations require suppliers of these medications to provide certain patients with access to the medication in such cases, known as “expanded access.

“For example, the drug can’t expose patients to unreasonable risks given the severity of the disease to be treated and the patient does not have any other satisfactory therapeutic options (e.g., an approved the drug that could be used to treat the patient’s disease or condition).

The manufacturer must be willing to make the drug available for the expanded access use. The primary intention of expanded access is to provide treatment for the patient’s disease or condition, rather than to collect data about the study drug.

Many investigational medications are licensed from prescription suppliers for patient use through extended access services listed in ClinicalTrials.gov. If you or a loved one are interested in treating an investigational drug under an expanded access protocol listed in ClinicalTrials.gov, review the eligibility criteria for the protocol and ask at the Contact Information Number.

What do you know about single and double blind studies?

In a single blind study, the subjects in the clinical trial do not know if they are receiving the placebo or the real treatment. where as in double blind clinical trial , both the subjects and the investigator do not know which group got the placebo and which got the experimental treatment.

What are Triple Blind Studies?

In triple blind studies, Subjects, Investigator and individuals who are responsible for analysis the outcomes are blinded. You can find the example of triple blind study here:

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4400707/
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1118396/

You should also read about the benefits and disadvantages of blinding.

What is Open label trials?

Open label trials are those trials where no blinding is done.

Subjects and Principle investigator and outcome analysis team knows about treatment and placebo group.

What is Placebo?

“A placebo is anything that seems to be a “real” medical treatment — but it doesn’t. It might be a pill, a shot, or some other type of “fake” treatment. All placebos have one thing in common is that they do not contain an active substance meant to affect health.

Normally, the person getting a placebo doesn’t know for sure that the treatment isn’t real.

The placebo is often in the form of a “sugar pill,” but it may also be an injection, a liquid, or even a procedure. This is intended to act like a true cure, but it does not affect the disease directly.
https://lifepronow.com/2020/07/08/placebo-is/

What is the difference between IB and Protocol

Clinical trial protocol  describes the objectives, design, methodology, statistical considerations and aspects related to the organization of clinical trials.”

In simple words, We can say clinical trial protocol tells us “how to conduct scientifically sound, reliable and ethically compliance trials”

Investigator Brochure enables investigators (Doctor at hospital) conducting clinical studies to know the risks and benefits associated with an investigational product.”

More at https://lifepronow.com/2020/06/25/difference-between-investigator-brochure-and-clinical-trial-protocol/

What is the approval process by FDA

The regulatory agency of USA is FDA which has two bodies: CBER and CDER.
CBER stands for  Centre for biological evolution and Research and CDER stands for Centre for drug evaluation and research.  CBER agency deals with biological product whereas CDER deals with drugs and biological both. In general the drug (chemical) approval process in US can be divided into four parts, they are: 

What is SAP

SAP (Statistical Analysis Plan) is the document that contains detailed information about the statistical methods and the study objectives to help in the production of the Clinical Study Report (CSR) that includes figures, summary tables, and subject data listings for Protocol.

Documentation of the program variables and algorithms which would be used to produce descriptive statistics and statistical analysis.

What is LOCF?

Pharmaceutical companies spend numerous months to conduct longitudinal studies on the human subject. It is impractical to expect patients to keep timely visit over such a long period of time.

In spite of all the efforts, patient data are not collected for some and these become missing values in a SAS data set soon.

The most recent previously available value for each of the missed visits is replaced for reporting. This is classified as Carried Forward Last Observation (LOCF).

LOCF does not mean the last SAS dataset observation which gets carried forward rather it means last non-missing value carried forward. It is the values of the individual measures which are actually “observations” in this case.

Also if there are multiple variables containing these values then they will be carried forward separately.

Explain the role of clinical research coordinator

In general, clinical research coordinator works as an assistant to Principle investigator (PI). CRC helps in taking the informed consent forms, enter the the in the eDC, followup with the subjects for site visits, respond to data management and clinical research associate queries. They are also involved in budget planning and management of budgets for the study and financial payments.

Details: https://lifepronow.com/2020/05/16/beginners-guide-to-clinical-research-coordinator/

You can also explore the role of CRA here, which is similar question:

What is the difference between Effectiveness and Efficacy

“Efficacy = best possible results for a particular intervention, under perfect conditions;

“Effectiveness = most likely results for a particular intervention under pragmatic or ‘real-life’ conditions (taking into account compliance, dropouts, withdrawals, the learning curve for surgical interventions, etc.) 

more at https://lifepronow.com/2020/05/18/efficacy-vs-effectiveness/

What Are The Benefits And Risks Of Participating In Clinical Trial?

Benefits include: Playing an active role in one’s health care, having access to medications that may not be accessible for a significant period of time, and supporting people by engaging in the study so that the drug can eventually be accepted and made readily available.

Risks include: Participation in the clinical trial may involve some risks that doctor will explain in more detail. These risks may include:

• Side-effects that are known and those that have not yet been identified;
• risks associated with the study procedures;
• the experimental treatment may be not be effective or less effective than the current standard;
• the experimental treatment may not work for all patient.

Additionally, in some clinical trials the patient may not receive the experimental treatment, although the current standard or a placebo.

In addition to the risks listed above, the trial might require all the participant’s time and attention—including trips to the study site, more treatments, hospital stays or complex dosage requirements.

A prospective, randomized, double-blind, controlled clinical trial is the most rigorous clinical trial design, and the one that regulatory agencies mandate must be conducted to demonstrate a medication’s effectiveness and safety.

Those trials are the highest quality evidence on the medication and its activities in a new drug application, which form the basis for approval.

Patients are carefully chosen for inclusion in this research design and are randomly allocated to obtain the experimental treatment or a similar active drug or placebo.

Neither the patient nor the treating physician knows what care was offered, thereby eliminating potential bias.

Individual definitions of the study descriptions are:

Prospective: Looking ahead, starting before therapy has started.

Randomized: Patients are randomly assigned to obtain the alternative or experimental treatment ( e.g., standard of care or placebo)

Double-blind: Neither patients nor research personnel know which participants receive the experimental drug and which participants receive a placebo or normal treatment.

Controlled: One group of the patients will be given an experimental drug or treatment, while a second group is given either the standard treatment for the illness or a placebo.

Who Sponsors A Clinical Trial?

Clinical trials can be sponsored or funded by a variety of the organizations or individuals including physicians, medical institutions, voluntary groups, foundations, and pharmaceutical companies, in addition to the government agencies such as the National Institutes of Health (NIH), the Department of Defence (DOD), Human Health and Services (HHS), and Department of Veteran’s Affairs (VA).

Explore our courses on clinical research


What should people consider before participating in a trial?


People will learn about the clinical trial as best as possible and feel comfortable asking health care team members questions about it, the treatment provided when in a trial, and the cost of the experiment.

The following questions may be of assistance to the participant to discuss with the health care team. Some of the answers to these questions are found in the informed consent document.

  • What is the purpose of study?
  • Who is going to be in study?
  • How do researchers think the experimental treatment being tested can be effective? Is it reviewed in advance?
  • Which kinds of tests and experimental treatments?
  • What does the research equate the potential complications, side effects and benefits with my current treatment?
  • Why does the case affect my daily life?
  • How long is the case going to last?
  • Does it take hospitalisation?
  • What will pay for the experimental treatment?
  • Would I get a refund for other expenses?
  • Which sort of long-term follow-up treatment does this study include?
  • Why do I know the experimental medication is functioning?
  • Will it provide me with the results of the trials?
  • Who will take the responsibility for my care?

What is the scope of clinical research in India?

The Scope of Clinical Research depends on the growth of the Medical / Pharmaceutical Industry. And in India, this growth is immense, and the pace is rising with time, due to:

– an ageing population
– new developments in treatments and diagnosis
– the Indian economy – its privatization, liberalization & globalization.

Drug discovery involves designing new drugs, and after identifying a prospective article, the drug development process takes place. Action, safety, use, formulation, quality control, storage, packaging, and marketing are few of the other stages associated with the drug development process.

The global demand is more than $70 trillion for this knowledge-intensive field. And it is projected that India ‘s contribution is about 15 percent of the global market, which is about $10.5 billion. The business is currently rising at an average annual cumulative growth rate of a

And it is this reach that attracted Pharmaceutical Companies & Contract Research Organizations (CROs) to India to develop their research offices or even forward some of their clinical activities, such as data processing or quality assurance, to other CROs working in India, thereby reducing costs.

Clinical Research is an field of highly exclusive & lucrative job prospects with the potential to increase your salary in less than a decade by 400 per cent.

In order to soar in the evergreen industry, one should train oneself through appropriate courses that allow candidates to be ready for various operations of clinical research.

When you are interested, you will consider these internship-led training programmes that are conducted at MMS University (www.mmsuniversity.com), MMS Holdings’ educational division (www.mmsholdings.com), in fields these as Scientific Education, Pharmacovigilance, Clinical Planning and Case Ethical Disclosures.

MMS Holdings is a Contract Research Organization that fulfils compulsory steps to help Pharmaceutical companies apply for introducing new drugs into the market. Check them out to know more.

What is the difference between clinical research and medical research?

Clinical research is a subcategory of medical research that involves studies with humans or human-related data. Medical research is concentrating on unlocking the basis of medical treatment by using a variety of approaches.

What is Blinding in Clinical Trial?

Blinding is a procedure in which one or more parties in a trial are unaware of which participants in the treatment arms were assigned, that is, what treatment was received.

Blinding is an essential characteristic of any study performed to prevent and eliminate conscious or implicit bias in planning and conducting a clinical trial.

What are the Types of blinding?

A clinical trial is called a single blind if only one group, usually the patients, are blinded. If both the participants and study staff are blinded, it is known as double blind study. Triple blinded experiments also expand the data analysts to blind.

A trial in which no blinding is used and the treatment groups are known to all parties is called open label or unblinded.

TypeDescription
Unblinded or open labelAll parties are aware of the treatment the participant receives
Single blind or single-masked  Only the participant is unaware of the treatment they receive
Double blind or double-maskedThe participant and the clinicians / data collectors are unaware of the treatment the participant receives
Triple blindParticipant, clinicians / data collectors and outcome adjudicators / data analysts are all unaware of the treatment the participant receives


Investigator Meeting: Planning to Success

“An Investigator Meeting is typically the responsibility of a sponsor, but this role can also be assigned to a CRO. For those from the industry, the meeting of investigators is strongly targeted and criticized for overspending.

It is suggested that such meetings are usually held in clinical trials to attract participants, but if we seek to understand the need for these gatherings, they are much more productive than it is believed.

What is an investigator meeting?

It is a group meeting conducted on behalf of the sponsor/CROs to train investigators and their lead clinical trial staff on trial related activities, standard operating procedures and to talk about the applicable regulatory picture.

The content of an investigator meeting is typically trial specific, however a common agenda, on SOP’s, Adverse Event Reporting, Source Documentation etc.

Why Investigator meetings necessary?

Investigators’ meetings are necessary for the success of a clinical trial. Although face-to – face meetings cost more than web-based meetings, they are highly desirable to allow investigators and research coordinators to network and learn from each other

The meeting has several sessions on every aspect of the clinical trial, addressing the pros and cons of the activities related to the trials. The topic is also expanded to illustrate the potential issues that researchers can face, and how to resolve them.

How to Plan Investigator Meeting?

Following tips helps any event planner navigate successfully to an event:

1. How does an event planner for an investigator meeting even start?

You will need to learn as much detail as possible from your service provider: all who, what, where, where, and how for the meeting; details such as who the necessary attendees are, what the meeting agenda includes, where this meeting will take place, when the meeting will take place, and how much budget is there for the entire meeting.

2.The attendants are now starting to arrive, now what?

A group email announcement to the participants should be sent well in advance of their arrival time / day, outlining the key details that they will need for their arrival.

This will allow the attendees enough time to receive and review the information before boarding a flight, and is an immense chance to send out the overall agenda to the attendees as well! As a personal, yet important touch, send an email to each attendee with their exact travel information, including flight and hotel confirmation numbers, and suggest transportation services that are available once they reach your destination.

Be sure to let them know who to contact upon arrival.

3. Will those last-minute changes ever end?

This can feel like an uphill struggle to get the final copies of meeting materials. Creating and finalizing the meeting materials typically rests with the client, but the event planner may need to know that all the materials are completed and whether copies will be made available to the meeting attendees either electronically (preferably) or hardcopying.

Set a strict deadline for when materials need to be finalized in order to ensure that they get sent to attendees ahead of the meeting and are printed (if needed).

4. There’s no I in team…

Delegation is important at any case, since one person can not be in multiple places at once.

If having a secondary coordinator on site with you is not an option and you need to find out how to be in two positions at once, alert your customer to this and see if they can ask someone on their team to step in for immediate support.

5. Always room for improvement, right?

Print-outs at a meeting can be a bit of an issue. Often the printouts are discarded immediately after the meeting, ending up in the trash can.

If attendants need full access to all meeting materials, look for ways to reduce paper waste by urging participants to use electronic methods first.

Not only is this a significant cost saving as it costs time to plan printed documents at the last minute, but it is also an field of event management that has a tremendous effect on the environment.

Choosing electronic methods over paper printouts is an opportunity to reduce not only costs but also post meeting waste.

An investigator meeting ensures to:

Ensure that all investigators understand how to perform the trial in full compliance with the protocol, SOP’s, instructions and relevant regulations.

• To notify the CRF investigators.
• Register and train the roles of all participating state attorney and their staff before commencing the case.
• In-depth description of the research protocol.
• Facilitate communication between investigators.

The ICH Guideline for GCP states that investigators “will ensure accuracy, completeness, legibility and timeliness of the data reported to the sponsor in the CRFs”.

It is the Investigator Meeting which gives a CRO a primary opportunity to introduce a sponsor to the team of clinical trials. Hence successful execution of IM is seen as representing the team’s capabilities.”

References:

1) ICH – International Conference on Harmonisation Good Clinical Practices, Jan 21st 2012, http://ichgcp.net/4-investigator

https://www.meetingsnet.com/pharmaceutical-meetings/investigating-investigator-meetings

https://link.springer.com/chapter/10.1007/978-3-319-53877-8_37

FDA Ok’s TREMFYA® as First Selective IL-23 Inhibitor for Psoriatic Arthritis

0

July 14, 2020: “The Janssen Pharmaceutical Companies of
Johnson & Johnson today announced that the U.S. FDA has approved TREMFYA® (guselkumab) for adult patients with active psoriatic
arthritis (PsA), a chronic progressive disease characterized by painful joints and skin inflammation.

TREMFYA is the first treatment approved for active PsA that selectively inhibits interleukin (IL)-23, a naturally occurring cytokine that is involved in normal inflammatory and immune responses associated with the symptoms of PsA

The safety and efficacy of TREMFYA in PsA have been demonstrated in two pivotal Phase 3 clinical trials.

TREMFYA is administered as a 100 mg subcutaneous injection every eight weeks, following two starter doses at weeks 0 and 4. TREMFYA can be used alone or in combination with a conventional Disease-Modifying Anti-Rheumatic Drug or DMARD (e.g., methotrexate).

The Unmet Needs in PsA
Psoriatic arthritis affects about 1.5 million Americans. Studies show that up to 30 percent of the more than eight million Americans living with psoriasis will also develop PsA.

There is currently no cure for the disease and, despite available treatments,
many people living with PsA are still searching for more options that can help alleviate their symptoms and provide some relief.

Psoriatic arthritis is a chronic, progressive, immune-mediated disease characterized by joint inflammation, enthesitis (inflammation where the bone, tendon and ligament meet), dactylitis (severe inflammation of the digits of the hands and feet), axial disease (pain in the axial skeleton, primarily in the spine, hips and shoulders) and the skin lesions associated with psoriasis.


The disease commonly appears between the ages of 30 and 50 but can develop at any time. Though the exact cause of PsA is unknown, genes, the immune system and environmental factors are all believed to play a role in the onset of the disease.

Without early recognition, diagnosis and treatment, the disease can continue to progress.

“Psoriatic arthritis is a complex multi-faceted disease and, for many patients, additional biologic options are very much needed,” said Philip J. Measei, M.D.,DISCOVER-2 Lead Study Investigator, Director of Rheumatology Research at theSwedish Medical Center/Providence St. Joseph Health and Clinical Professor at the University of Washington School of Medicine in Seattle, WA.

“The two Phase 3 pivotal trials evaluating the safety and efficacy of TREMFYA, an IL-23 inhibitor, for the treatment of adults with active psoriatic arthritis provided insight into how it can improve joint symptoms.

Today’s approval is exciting for both patients and their physicians, as there is now a new approach available to help manage the symptoms.”
https://johnsonandjohnson.gcs-web.com/static-files/4f9074e1-268a-4a89-a6bf-88d8bf812d2a

Coronavirus (COVID-19) Update: Daily Roundup July 14, 2020

0

July 14, 2020: “The U.S. Food and Drug Administration announced the following actions taken in its ongoing response effort to the COVID-19 pandemic:

  • The FDA has posted an updated Coronavirus Treatment Acceleration Program (CTAP) webpage.

    The webpage provides information about CTAP’s purpose, strategy and operations.

    Specifically, this revised webpage includes a dashboard showing crucial statistics related to the development of potential COVID-19 therapeutics.

    Also included are key guidance and links for therapeutic developers, researchers, patients and consumers.

    Today, the FDA also released a new FDA Voices, titled An Update and Behind the Scenes: FDA’s Coronavirus Treatment Acceleration Program, in which agency leaders discuss efforts to leverage cross-agency scientific resources and expertise on COVID-19 therapeutic development and review.

    There are now more than 510 drug development programs in planning stages for potential therapies for COVID-19, and as of today, the agency has reviewed more than 230 trials of potential therapies for COVID-19.
  • The FDA encourages consumers to test their knowledge about the appropriate use of hand sanitizer with this hand sanitizer quiz, posted today.

    The quiz provides questions and answers to some of the most frequently asked questions related to hand sanitizer. This quiz can help consumers learn how to correctly use hand sanitizer.
  • As part of the FDA’s effort to protect consumers, the agency issued warning letters to the following two firms for selling unapproved products in the United States with false or misleading claims that their products can mitigate, prevent, treat, diagnose or cure COVID-19 in people:
  • Today, the FDA added dexamethasone sodium phosphate to the lists of drugs for temporary compounding by outsourcing facilities and pharmacy compounders during the COVID-19 public health emergency.

    These updates help address shortages and access concerns affecting some drugs urgently needed for hospitalized COVID-19 patients.

    The updated lists are referenced in the following guidances:
    • Temporary Policy for Compounding of Certain Drugs for Hospitalized Patients by Outsourcing Facilities During the COVID-19 Public Health Emergency
    • Temporary Policy for Compounding of Certain Drugs for Hospitalized Patients by Pharmacy Compounders not Registered as Outsourcing Facilities During the COVID-19 Public Health Emergency Guidance for Industry
  • Testing updates:
    • To date, the FDA has currently authorized 179 tests under EUAs; these include 148 molecular tests, 29 antibody tests, and 2 antigen tests.

The FDA, an agency within the U.S. Department of Health and Human Services, protects the public health by assuring the safety, effectiveness, and security of human and veterinary drugs, vaccines and other biological products for human use, and medical devices.

The agency also is responsible for the safety and security of our nation’s food supply, cosmetics, dietary supplements, products that give off electronic radiation, and for regulating tobacco products.”
https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-daily-roundup-july-14-2020

Randomization in Clinical Trials: Definition and types

0

Randomization

“As per the National Cancer Institute “In research, the process by which participants in clinical trials are assigned by chance to separate groups that are given different treatments or other interventions.

Neither the researcher nor the participant chooses which treatment or intervention the participant will receive.

Using chance to assign people to groups means that the effects of the treatment or intervention that participants receive can be compared more fairly.”

Randomization is the process of making something random; in various contexts this involves, for example:

  • generating a random permutation of the sequence (such as when shuffling cards);
  • selecting a random sample of the population (important in statistical sampling);
  • allocating experimental units via random assignment to the treatment or control condition;
  • generating random numbers (see Random number generation); or
  • transforming data stream (such as when using a scrambler in telecommunications).

What are the Reasons for Randomization?

Researchers in life science research demand randomization for a number of reasons.

First, topics in different groups do not vary in any systematic way. Within a clinical research, if patient groups are significantly different, research results will be biased.

Suppose that subjects are assigned to control and treatment groups in a study investigating the effectiveness of a surgical intervention.

If a greater proportion of the older subjects are assigned to the treatment group, then the result of the surgical intervention may be influenced by this imbalance.

The effects of the treatment would be identical from the influence of the imbalance of covariates, thereby requiring the researcher to control for the covariates in the analysis in order to obtain an unbiased result.

Second, proper randomization ensures no former knowledge of group assignment (i.e., allocation concealment). That is, researchers, subject or patients or participants, and others should not know to which group the subject will be assigned.

Knowledge of the group assignment creates a layer of potential selection bias that may taint the data.

Schul and Grimes stated that trials with insufficient or unclear randomization tended to overestimate treatment effects up to 40% compared with those that used proper randomization. The outcome of the research can be negatively influenced by this insufficient randomization.

Statistical techniques such as analysis of covariance (ANCOVA), multivariate ANCOVA, or both, are often used to adjust for covariate imbalance in the analysis stage of the clinical research.

On the other hand, the interpretation of this post adjustment approach is often difficult because imbalance of the covariates regularly leads to unanticipated interaction effects, such as unequal slopes among subgroups of covariates.

One of the critical assumptions in ANCOVA is that the slopes of the regression lines are the same for each group of the covariates.

The adjustment needed for each covariate group may vary, which is difficult because ANCOVA uses the average slope across the groups to adjust the outcome variable.

Thus, the ideal way of balancing covariates among groups is to apply sound randomization in the design stage of the clinical research (before the adjustment procedure) as an alternative of the post data collection.

In such instances, random assignment is essential and guarantees validity for the statistical tests of significance that are used to compare treatments.

What the types of Randomization?

Many procedures have been proposed for the random assignment of the participants to treatment groups in clinical trials.

Common randomization techniques includes, simple randomization, block randomization, stratified randomization, and covariate adaptive randomization, are reviewed.

Each method is described along with its advantages and disadvantages. It is very important to select a method that will produce interpretable and valid results for your study.

Use of online software to generate randomization code using block randomization procedure will be presented.

Simple randomization

Randomization based on a single sequence of the random assignments is known as simple randomization.

This technique maintains complete randomness of the assignment of a subject to a particular group.

The most common and basic method of the simple randomization is flipping a coin.

For example, with two treatment groups (control versus treatment), the side of the coin (i.e., heads – control, tails – treatment) determines the assignment of each subject.

Other methods include using shuffled deck of cards (e.g., even – control, odd – treatment) or throwing a dice (e.g., below and equal to 3 – control, over 3 – treatment).

A random number table found in a statistics book or computer-generated random numbers can also be used for the simple randomization of subjects.”

image:Statistics How-To

Block randomization

“The block randomization method is designed in order to randomize subjects into groups that result in equal sample sizes.

This method is used to make sure a balance in sample size across groups over time. Blocks are small and balanced with predetermined group assignments, which keeps the numbers of the subjects in each group similar at all times.

The block size is determined by the researcher and should be a multiple of the number of groups (i.e., with two treatment groups, block size of either 4, 6, or 8). Blocks are best used in smaller increments as researchers can more easily control balance.

After block size has been determined, all possible balanced combinations of assignment within the block (i.e., equal number for all groups within the block) must be calculated. Blocks are then randomly chosen to determine the patients’ assignment into the groups.”

image:Statistics How-To

Stratified randomization

“The stratified randomization method addresses the need to control and balance the influence of the covariates.

This method can be used to attain balance among groups in terms of subjects’ baseline characteristics (covariates).

Specific covariates must be identified by the researcher who understands the potential influence each covariate has on the dependent variable.

Stratified randomization can attained by generating a separate block for each combination of covariates, and subjects are assigned to the appropriate block of covariates.

 After all subjects have been identified and assigned into blocks, simple randomization is performed within each block in order to assign subjects to one of the groups.

image:Statistics How-To

Covariate adaptive randomization

One possible issue with clinical research of small to moderate size is that simple randomization (with or without taking into account the stratification of prognostic variables) can contribute to the imbalance of important covariates between treatment groups.

Covariate imbalances are significant because of their potential to influence the interpretation of a research finding.

Several researchers have proposed covariate adaptive randomisation as a legitimate alternative form of randomization for clinical research.

In covariate adaptive randomization, the sequential assignment of a new participant to a specific treatment group takes into account the specific covariates and previous assignments of participants.

Covariate adaptive randomization uses the minimisation approach by measuring the sample size difference of multiple covariates.

image:Statistics How-To

Permuted block randomization

Sometimes, just choosing participants randomly isn’t enough. You might want to balance your participants into groups, or blocks.

Permuted block randomization is a way to randomly allocate a participant to a treatment group, while keeping a balance across treatment groups. Each “block” has a specified number of randomly ordered treatment assignments.”

Eg..    AAABBBBAAABBB
          BAABBAAABBAAA

Importance of randomized studies in Health Economics Outcomes Research (HEOR)

Randomized studies are an essential tool in Health Economics and Outcomes Research (HEOR) for evaluating the effectiveness, safety, and cost-effectiveness of healthcare interventions. Read full blog on https://digitalho.com/blog/heor/

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3136079/

https://www.theisn.org/isn-act-toolkit/study-stage-1-design-and-development/randomization

http://www.randomization.com/

https://www.frontiersin.org/articles/10.3389/fnins.2018.00355/full

https://digitalho.com/blog/heor/

What is the electronic Common Technical Document (eCTD)?

0

Introduction:

The standard format for submitting submissions, modifications, supplements, and reports to the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER) of the FDA is known as electronic Common Technical Document (eCTD).

Understanding the eCTD requirements and applying them effectively to your submissions is critical.

Such specifications are not always sensitive, however, and headaches that occur when documents do not fit seamlessly into the eCTD structure that is permitted.

If that happens, sponsors will face last-minute changes, putting deadlines in trouble. Worse yet, the FDA, which is known as technical rejection, could bounce back the entire submission.

eCTD Structure

In the past, paper submissions were a mode of life for anyone submitting an application to the FDA. Unfortunately, paper submissions are bulky, hard to store, and inefficient to assemble, update, and review. In order to eliminate these issues and speed the review process, the FDA now requires electronic submissions for most of the applications.

Since May 5, 2018, all submissions for the new drug applications (NDAs), Abbreviated NDAs (ANDAs), Biologics License Applications (BLAs), Commercial Investigational New Drug Applications (INDs), and Master Files are required to follow eCTD specifications.

There are several variations to the eCTD, but in its simplest terms, the eCTD is a standardized document structure that enables details to be presented reliably and comprehensively within a request.

The eCTD submissions comprise 5 sections, called modules, each of which contains a specific type of information.

  • Module 1 : (not technically part of the CTD): region-specific administrative information
  • Module 2: manufacturing, nonclinical, and clinical overviews and summaries
  • Module 3: detailed manufacturing information
  • Module 4: nonclinical study reports
  • Module 5: clinical study reports

Chemistry, Manufacturing, and Controls (CMC)

As anyone who has written an IND or marketing application knows, the CMC modules (module 3 and summaries in module 2) are generous and can be tedious and time-consuming to complete.

Because more documents usually mean more “cooks in the kitchen,” it’s tempting to combine allowed parts in an attempt to streamline writers and versions management.

Nevertheless, the granularity of the CMC parts should be written with all of the submission’s life cycle in mind.

For instance, if you decide to consolidate module 2.3 (Quality Overall Summary), any updates to the drug substance sections will require the resubmission of this complete module rather than just the pertinent information.

As a general rule, a more granular structure should be measured in order to avoid issues during the lifecycle of the application.

The “M4 Organization of the Common Technical Document for the Registration of Pharmaceuticals for Human Use” can be used as a reference to determine the permitted granularity.”

M4 : The Common Technical Document

“The agreement to assemble all the Quality, Safety and Efficacy information in a common format (called CTD – Common Technical Document ) has revolutionised the regulatory review processes, led to harmonised electronic submission that, in turn, enabled implementation of good review practices.

For industries, it has eliminated the need to reformat the information for submission to the different ICH regulatory authorities.

The CTD is organised into five modules. Module 1 is region specific and Modules 2, 3, 4 and 5 are intended to be common for all regions. In July 2003, the CTD became the mandatory format for new drug applications in the EU and Japan, and the strongly recommended format of choice for NDAs submitted to FDA, United States.

More information: An electronic version of the Common Technical Document (eCTD) can be produced using the information developed by the eCTD Implementation Working Group.”


Clinical Study Reports

“Even the experienced teams can meet problems while determining the best way to structure a clinical study report (CSR) within the eCTD. Should it be submitted as a single document? If splitting it up, what is the proper way to group these sections?

The eCTD structure for Module 5 follows the levels outlined in “The Comprehensive Table of Contents Headings and Hierarchy.” even though legacy CSRs can be submitted as one document, the current recommendation is to divide CSRs into sections: synopsis, report body, and individual appendices (i.e., Section 16 of CSR separated to 3rd level headings).

Preparing a CSR in this format allows reviewers to more simply navigate the large amount of the information, leading to more proficient reviews. Additionally, by dividing the report into sections, changes can be more easily tracked, as the whole report does not need to be replaced if only one section is updated.

Report Numbering

Each study report, both non-clinical and clinical, must have an ID and title of the study inserted into the submission.

As such, it is important to ensure that all reports have an unique id associated with them before they are sent to your publishing team. It usually becomes a concern as data from the study extends over several studies, such as an initial clinical trial and related population PK analysis.

If a population PK analysis is being conducted for a definite clinical study, the population PK report should receive a unique study ID to ensure that the analysis is different from the general clinical study report.

This also helps to ensure that any cross-references in the summary sections are clear as to the content being referenced.

Annual Reports

Back in paper days, the annual report was frequently written as a single document; on the other hand, the current granularity in “The Comprehensive Table of Contents Headings and Hierarchy” outlines multiple sections for annual reports.

While it may be tempting for the sponsors to continue authoring the annual reports as one document, the structure of the eCTD does not readily allow for this approach and can lead to validation errors, putting the application at risk of the technical rejection.

When published as a single text, publishers can seek to avoid validity issues by putting the annual report within a single node that corresponds to one of the parts of the annual report (e.g., Nonclinical Studies Summary).

This method causes ambiguity during the analysis, however, as the content does not fit properly with the definition of the nodes.

Hyperlinking

One of the most popular problems faced by publishers is trying to hyperlink to a section and finding that the section either does not exist or can not be linked to.

Part of this is due to typographic errors during document authoring; however, other instances arise due to the writers’ lack of comprehension about what material could actually be connected to?

As a rule, hyperlinks can only be made to individual documents and not to section folders.

For example, a common mistake when hyperlinking is referencing a section that is of a higher level of granularity than that to which the documents are written.

 Some examples include:

  • Referencing the module for the single dose toxicity studies (i.e., 4.2.3.1) rather than the specific study
  • Referencing module 3.2.P.3 as a whole rather than specific relevant section(s) (e.g., 3.2.P.3.1)

The “Comprehensive Table of Contents Headings and Hierarchy” and “M4 Organization of the Common Technical Document for the Registration of the Pharmaceuticals for Human Use” can be used to determine permitted levels within an application.

In order to make the publishing process as competent as possible, it is important for the writers to ensure that each document has undergone adequate quality control review to verify that all cross-referenced sections exist previous to sending them to the publishing team.”

https://admin.ich.org/sites/default/files/2019-05/CTD_triangle.pdf
https://www.ich.org/page/ctd
https://www.nuventra.com/resources/blog/regulatory-submissions-ectd/
https://www.trilogywriting.com/common-technical-document-ctd/

Free Course on Clinical Data Management (self explanatory)

You must remember one thing: To get a job in clinical data management, you need to have basic knowledge of clinical research and clinical data management. Needless to mention other things such as good communication skills and Soft skills ie. Microsoft Excel, PPT and Word are “must-have skills” to get closer to your first job.

The below blog is written and tailored to fulfil the need of those freshers who are willing to make their career in the clinical data manager. The language is kept as simple as possible so it is kind of self-explanatory course. Any doubt, you can ask in the comment section or write to us at [email protected], Let’s start it.

Introduction

The primary goal of Clinical Data Management (CDM) is to ensure timely delivery of high-quality clinical data needed to satisfy both the  good clinical practice (GCP) guidelines and the criteria for statistical analysis and regulatory reporting.

It means clinical data managers are caretakers of clinical data. They arrange the things for proper collection of clinical data, make sure that clinical data is clean (minimum error) and ready for statistical analysis.

CDM team members participate actively in all stages of clinical trials right from start to finish

CDM is the process by which subject data are processed, cleaned, and handled in compliance with regulatory standards. We will discuss later on this blog, about  all the regulatory requirements in CDM.

 First let’s understand what CDM is in actuality.

Clinical Data Management Process

The whole process of clinical data management  is divided in three parts across the globe.

  • Set up or start-up.
  • Conduct
  • Closeout
Key activities in each Phase of Clinical Data Management

Set up Phase:

We have divided all the Set Up Process in 11 Steps and and we will explain each steps in details. Please read each steps and ref the below flow chart to have a better understanding.

Step 1: Reading the finalized protocol

As we know that Clinical trials are run based on clinical trial protocol.

Once the Protocol is finalized. CDM activities start. It may start before also, if the sponsor is ready to start activities based on drafted protocol and simultaneously sponsors can work on protocol to finalize it.

So here, I am not talking about special cases (drafted protocol) but cases which are quite common (finalized protocol). So forgot about what will happen if the protocol is not finalized.

After reading the complete protocol, CDM has to start working on to below two activities

Now the question comes,

  • What is case report form? and
  • What are the documents need to be drafted by Clinical Data Management.

Lets learn about Case Report Form

Case Report Form and eDC

As per ICH E6 (R2),

“A printed, optical, or electronic document designed to record all of the protocol required information to be reported to the sponsor on each trial subject”.

 The forms which are used to capture the information as per the protocol is called case report form. It may be in the paper format or electronic format. eDC systems use electronic forms to capture clinical trial data.

You would have filled many forms while schooling or in college life and these forms are the same with only one difference and that is: Information is  asked and provided as per clinical trial protocols.

Paper CRFs are not common nowadays so we will focus on electronic case report form.

A well-designed CRF will reflect the essential contents of the research protocol, and when the research protocol is finalized, CRF is designed.

to read more about case report forms and example:
Basics of case report form

Now we know about CRF, lets learn about eDC.

eDC (electroninc data capture)

The method to capture clinical data is decided at the organization level. 

Trial requirements, tool specifications and budget are the deciding factors to select a particular method to capture data. Organizations can use paper forms to capture the clinical trial-related data or can consider to go with electronic one.

An electronic tool or system to capture clinical data is called eDC (electron data capture). It is also called a clinical database (from data management point of view)

 There are many tools available in the markets. Most popular tools are Medidata RAVE, Inform and OC-RDC.

As per FDA, “Electronic data must meet the same fundamental elements of data quality (e.g. attributable, legible, contemporaneous, original, & accurate) and integrity (complete and consistent) expected of paper records “

Case report forms are designed electronically with the help of programming in these eDC.

Now you understand the link between CRF and eDC.

Data Manager reviews the protocol and identifies the key information to be captured.

Information is captured in the Case report form.

  • Subject number
  • Gender/Sex
  • Age
  • Demography
  • Inclusion-Exclusion Criteria
  • Informed consent form
  • Visit schedule eg visit 1, Visit 2 etc
  • Lab tests to be done at each visit as given below
  • Chemistry, Hematology, coagulation, Thyroid, cytokines, Virology, cytokines etc.
  • ECG
  • Biomarkers test
  • RECIST check if oncology study
  • Dose administration record (if intervention trial)
  • Sample collection time point
  • Treatment arm
  • informed consent from the subjects

    These are general Case report forms that can vary study to study based on protocol
    Recommended further reading : Difference between Investigator Brochure and Clinical Trial Protocol

Step 2: Search in standard CRF  library to identify desired case report forms

Clinical data managers choose the case report forms based on protocol requirements. Every Sponsor keeps a commonly used CRF (also called standard CRF)  in their library (like shared drive) and the Data manager chooses the appropriate CRF from this library.

recommended further reading: Basics of case report form

Step 3: All the case report form is identified  and the standard specification document (SSD) is ready

Study specific documents mentiones the clinical database structure (eDC)

It mentions all the visit schedules (flow of visits at site) and all the applicable case report forms. It also mentioned all the variables of each case report forms and access conditions of each role. Below example of the SSD is quite simple and the actual SSD is quite big and complicated. SSD is the skeleton of the database and it is drafted in excel format.

Now the question comes how. Let understand it with below table

Visits Condition metFormsRestriction
ScreeningIf subject sign the informed consent formHematologyData manager-read access, CRC-Entry access
 Screening If subject sign the informed consent formChemistryData manager-read access, CRC-Entry access
Screening  If subject sign the informed consent formVirologyData manager-read acces, CRC-Entry access
 Screening  If subject sign the informed consent formInclusion criteriaData manager-read acces, CRC-Entry access
V1If subject met all the inclusion criteria, V1 should open in the data baseHematologyData manager-read acces, CRC-Entry access
 V1 If subject met all the inclusion criteria, V1 should open in the data baseChemistryData manager-read access, CRC-Entry access
 V1 If subject met all the inclusion criteria, V1 should open in the data baseVirology Data manager-read access, CRC-Entry access
V2if subject status is “continue” at V1, then V2 should open Hematology Data manager-read access, CRC-Entry access
A small segment of SSD

Step 4 Request to the programming team to program the Case report forms in the eDC

Till step 3, we have identified the case report forms and SSD is ready. Now we have to share this (CRF plus SSD) with programmers so that they can design the database (eDC).

SSD helps them to understand interaction/flow of various case report form in eDC. For example, Visit 2 should come after visit 1, hematology case report should come at V1 but should not come at V2 etc.

Step 5 Once all the case report forms are programmed, User acceptance testing is performed and it is called UAT-screen.

User Acceptance Testing (UAT): It is a critical component of using Electronic Data Capture (EDC) to record data from clinical trials. The sponsor / CRO must conduct UAT before using EDC to gather data in compliance with a protocol.

Once programmers complete the programming of case report forms in eDC system as per our request, we start UAT.

Generally, Data Manager creates a subject (dummy) in eDC system and thoroughly test every e-CRF. 

Data Manager has to raise a request for UAT environment access. it is also called test environment access.

All the Case report forms in eDC systems are checked to ensure that all the variables (questions or data points), the flow of information are programmed correctly and the eDC system is showing the data as intended.

All the finding needs to be documented and is shared with the Programmer to correct. Once Programmer corrects the programming errors, it is considered is “UAT Pass”

UAT is performed whenever there is a change in the database (eDC System). For example, there is a protocol amendment and new CRFs need to be added in the database. The data manager drafts those CRFs related requests and programmer programmes those CRFs in Database. Now the data manager has to perform UAT in the Database to ensure that all the new changes are appearing as intended.

Step 6 All the issues identified in the UAT is resolved then database is shown to other associate teams such as Bio statistics, clinical, Medical etc

Once Screen UAT is passed in step 5, the skeleton of Database is ready and it is shown to all the concerned teams.

It is called online screening 1

You can consider it as a trailer launch of a movie. All the concerns raised by other teams are addressed. It may require to reprogram certain things.

Step 7: Now edit specification document is drafted and once it is ready, it is shared with the programming team to program edit checks

After step 6, a new document needs to be drafted, it is called edit check specification

Edit check Specification

Edit check document mentions all the manual and programmable checks.

“Edit check programs are written i order to identify the discrepancies in the entered data, which are embedded in the database, to ensure data validity”

Edit checks consist of manual and computer checks, which need to be performed on the clinical data to ensure the data is accurate and consistent. The first stage consists of producing an Edit Check Specifications (ECS) document and implementation stage involves the programming and testing of the checks.

With the help of edit check specification document, programming team program the edit checks in the DC. Lets learn more about edit checks

Edit Cheks:

As per CDISC;

“An auditable process, usually automated, of assessing the content of a data field against its expected logical, format, range, or other properties that is intended to reduce error.

NOTE: Time-of-entry edit checks are a type of the edit check that is run (executed) at the time data are first captured or transcribed to an electronic device at the time entry is completed of each field and/or group of fields on a form. Back-end edit checks are a type that is run against data that has been entered or captured electronically and has also been received by the centralized data store.”

Electronic edit Checks allow us to use the computer’s power to review illogical, incomplete or inconsistent data in clinical trials quite efficiently.

Uni-variate edit checks (include range checks)

These are the edit checks which only apply to a single field or variable. For example, we can set up an edit check for subject weight to ensure that the extreme or improbable value is not entered.

Let’s say we set up a range check if data entry is smaller than 40 kg or greater than 90 kg for the subject’s weight. 

We can set up an edit check for predicted FEV1 to be no less than 20 per cent for lung function testing because it is less likely to have someone with predicted FEV1 < 20 per cent

Multivariate edit checks (also called aggregate edit checks):

To ensure that the data is logical and consistent, these edit checks cross-check the entries through more than one fields/variables.

For example, if the Gender field entry is ‘ Male, ‘ there should be no data for the result field of the pregnancy test.

If a subject dies at some time point of the study, then there should be no visit or lab test entries for that subject after that death date

One misconception is to think that the implementation of the edit checks will fix all data issues. Edit check is just one step in the process of cleaning up results. Unnecessary edit checks on non-critical fields can be very irritating for the sites.

Step 8 Once all the edit checks are programmed, a UAT-Edit check is performed.  This is the second UAT in the set up phase.

Now edit specification document is drafted and once it is ready, it is shared with the programming team to program edit checks. Once all the edit checks are programmed, a UAT-Edit check is performed.  This is the second UAT in the setup phase.

Step 9: Once all the issues identified in the UAT-II are resolved, the database is ready for screening 2 with all other teams. They can suggest if any modification is needed.


Step 10: Other things like set up of any third-party systems, or any other protocol-specific requirement can be programmed here.

Step 11: Now clinical database (eDC) is live and site (the hospital where the trial is being conducted) can enter the data

There are another two important documents which are drafted by Data Managers in set up,  they are

Data Management Plan
CRF completion guidelines

Data Management Plan

Each clinical trial should have a perspective plan for how to capture, process and store data. It is called DMP, Data Handling Plan (DHP) and Data Handling Protocols.

It answers the following questions:

  • Which data to be captured
  • How to Capture the Data
  • What to review and how to handle the different type of Data
  • Which tools are used in the study
  • Team members in the study
  • Handling of Serious adverse event

ICH E6 R2 states  “trial sponsors should “implement a system to manage quality throughout all stages of the trial process” and goes on to specify that quality management includes tools and procedures for data collection and processing (ICH E6 R2 2018, SS 5.0).”

CRF completion guidelines:

CRF completion guidelines provide the instruction to site personnel while adding the data into the database. 

DMP and CRF completion guideline should be ready before the database go-live.

You should note one thing that the sponsor provides the template for each document. You just need to tailor it according to your project/study

Conduct Phase

conduct phase

Once the database is released into production, the conduct phase of CDM starts.

In this Phase, site enters the data and CDM has to review it. If CDM finds any inconsistency, discrepancy or mistake, then he/she can raise the query in the database. Generally, the clinical research coordinator enters the data and they will respond to all queries raised by DM to site.

There is another role in clinical research. It is called Clinical research associate. CRA works as a liaison between Sponsor and site. CRA performs the source data verification. It means they check the original document and its entries in the database. There is much data which is entered first on primary documents such as local laboratory data, then CRC enters all the data into a database. CRA can look into this data and check whether data present in the primary document and entered data is the same or not.

Data manager has to perform cross check for the data for example, subject age is died on 19-Jan-2020 but  sample collection date is mentioned as 21-Jan-2020. He has to be quite good at excel.

Reconciliation of external data: 

All the data do not flow directly into the database as many vendors are associated with many lab assessments and they send the data separately.  This is quite an important task performed in the conduct phase of the study. 

For instance,  Advsere event is captured in two different database. Clinical database (the one, Data manager designs and having CRFs) and Safety database. Argus is the most commonly used software to capture safety data. Both data are reconciled as per standard operating procedure and issues are communicated to the safety team. So this data is verified with existing data in the database.

For example, if Lab A needs to perform at all visits, then external data should have lab data for all the visits, the subject has performed to the site.  Once all the data is reconciled, it is sent to the vendor or site for correction/clarification.

There are many types of reviews performed during the conduct phase. For example, few subjects’ data is analysed to check if it is good to go with study. Interim analysis is performed to check the efficacy and safety profile of the drug.

Close out

Close out
Database-lock activities

When all the data is entered and all the validation check is done and no discrepancy is observed, data is considered ready for lock. Pre-lock checklist is singed and permission from all the stakeholder is taken prior locking the database

Database lock is considered a state of database where no change is permitted. It denotes that all the relevant information is collected, reviewed and it is free of any discrepancy.

It is performed once all the stakeholder such as CDM, Biostatistics, Safety and Medical team has reviewed the data and pre-lock checklist is signed.

There are two types of lock:

Soft lock

 It is done before hard lock. It refers to the process where access is quite limited and CDM personnel confirms the suitability of data for final analysis.

Data can be unlocked at this stage if required by requesting privileged user.

Final lock

it is also called freeze or hard lock. All the access to the database is removed and no further changes are allowed.

Data is extracted once the database lock is done. Statistical analysis is done on extracted data. Results are documents in well-strcutered reports and submitted to the regulatory authority.

Archiving

All the essential documents such as clinical trial protocols, IB, UAT related documents, ECS and other database related documents are archived in sponsor specific archival systems. All the stakeholder has to archive the data as per their work.

Recommended: What is Clinical Trial Master File?

Regulatory Guidelines

CDM has certain guidelines and standards that must be met

  • Code of Federal Regulations (CFR), 21 CFR Part 11.
  • Good Clinical Data Management Practices
  • ICH E6- GCP
  • Clinical Data Interchange Standards Consortium

Electronic records must comply with a Code of Federal Regulations (CFR), 21 CFR Part 11.

“This regulation is applicable to records in an electronic format that are created, modified, maintained, archived, retrieved, or transmitted. This demands the use of validated systems to ensure accuracy, reliability, and consistency of data with the use of secure, computer-generated, time-stamped audit trails to independently record the date and time of operator entries and actions that create, modify, or delete electronic records”

Good Clinical Data Management Practices (GCDMP) guidelines are published by Society for Clinical Data Management (SCDM) .

As per SCDM:

“The Good Clinical Data Management Practices (GCDMP©) standard provides a reference to clinical data managers in their implementation of high-quality Clinical Data Management processes and is used as a guidance tool for clinical data managers when preparing for CDM training and education.”

They are drafting a new GCDMP in 2020.

ICH E6- GCP guideline is also applicable to CDM as it is part of core clinical trial activity. Other guidelines also refer to ICH guidelines.

https://scdm.org/gcdmp/
https://www.ich.org/page/ich-guidelines

Clinical Data Interchange Standards Consortium:

Study Data Tabulation Model Implementation Guide for Human Clinical Trials (SDTMIG) and the Clinical Data Acquisition Standards Harmonization (CDASH) standards are the most important standards given by CDISC

SDTMIG : It provides model and standard terminologies for the data. 

CDASH: It defines the standards to be followed for collection of data and provides a list of basic information which is needed from a clinical, scientific and regulatory point of view.

Summery

Clinical data management is a quite important aspect of clinical research which is responsible for data collection, cleaning and reliable and statistically sound data generation. There are three Phases of clinical data management. Set up, conduct and close.

Setup phase is divided into 11 steps. It starts from reading of finalized protocol and CRF selection. Database developer program these selected CRF in eDC.
UAT-Screen is performed to check if the database is performed as expected.
Now Edit checs are programmed and UAT-Edit check is done to check if edit checks are firing as expected or not. Edit checks are responsible for firing autoqueries in the database whenever inconsistent or inaccurate data is entered.

Once UAT-edit check is passed, database (eDC) is moved to production (database go live).
Now the conduct phase of the CDM starts. In this phase site personnel enter the data into the database and clinical data management review this data and raise the queries  whenever there is  any inaccurate information (Discrepancy management and data cleaning). Reconciliation of various data point also takes place in conduct phase. 
Once all the subjects visit ends, CDM has to review all the data and need to make sure that all data entries are done and it is cleaned and ready for lock.
Once all the  cleaning and data entry is completed, it is time to perform database lock. Database lock activities are close out activities. Now data can be extracted for further statistical analysis.

References and recommended sources



What is Clinical Trial Master File?

0

Introduction:

“A trial master file is a collection of the important content for clinical trials that are overseen by the regulatory agency.

To meet government regulatory requirements for clinical trials, each company involved in clinical trials must preserve and store all clinical trial-related records, images, and content. This information can be contained in the trial master file or TMF, which today takes the form of an electronic trial master file (eTMF), depending on the regulatory jurisdiction.

In 1996, the International Conference on Harmonization (ICH) released a consolidated industry guidelines on good clinical practice with the goal of establishing a common framework for the European Union, Japan and the United States of America to promote the universal recognition by regulatory authorities of clinical data in those jurisdictions.

This guidance document set out the requirement for all ICH regions to create trial master files containing important documents that individually and collectively enable assessment of the conduct of a trial and the quality of the generated data.

There is no clear provision for a court master file in some jurisdictions, for example the US. However, if the regulatory authority requires ICH GCP to be followed, then there is consequently a requirement to create and maintain a trial master file.

A TMF is the compilation of important documentation used by promoters, CROs and investigators / institutions to manage the trial, and by supervisors, auditors and inspectors to review and check whether the trial was performed by the sponsor and the investigators / institutions in compliance with the relevant legal criteria and the GCP principles and standards.

Therefore, all basic requirements are the same for both formats or when used in combination as a hybrid TMF.

The TMF should provide for document recognition, version history, search and retrieval; as specified in both Directive 2005/28 / EC (Article 17) and Regulation (Articles 57 and 58), it should also be archived in such a way as to ensure that it is readily available and accessible directly to the competent authorities of the Member States upon request.

Article 47 of the Regulation states that sponsors and investigator/institution shall take appropriate account of the ICH GCP guideline and shall conduct the trial in accordance with GCP principles, two of which are:

· “All clinical trial information have to be recorded, handled, and stored in a way that allows its accurate reporting, interpretation and verification”  

· “Systems with procedures that assure the quality of every aspect of the trial must be implemented.”
Aspects of the trial that are essential to ensure human subject protection and reliability of the trial results should be the focus of such systems.”

Trial master file contents

Essential documents: “Essential documents are documents that enable evaluation of the conduct of a trial and the quality of the data generated individually and collectively.” Essential documents allow the investigator / institution and sponsor to conduct the trial effectively and show compliance to the GCP guidelines and all other regulatory criteria that apply.

The TMF maintained by the investigator / institution and the one maintained by the sponsor have a different content, due to the different nature of the investigator / institution’s and sponsor’s responsibilities,6 as specified in the ICH GCP guideline. Article 57 of the Regulation declares the TMF necessary

Article 57 of the Regulation states that the quality of the necessary TMF documents shall take into account ‘all the characteristics of the clinical trial, including, in particular, whether the clinical trial is a clinical trial with low involvement.’

Therefore, some documentation specified in the ICH GCP guideline may not be necessary due to the implementation of a risk proportionate approach7. The justification for reducing documentation should be documented in the TMF.

The documentation specified in section 8 of the ICH GCP Guideline specifies the documents that are considered necessary (as applicable to the trial) and the documents should be submitted to the TMF investigator / institution or sponsor, or both; however, this list should not be used as a final checklist for TMF material.

It is not a complete list. Therefore, the sponsor and/or investigator / institution will provide any documentation that enables the reconstruction and review of the conduct of the trial as part of the TMF, depending on the activities being carried out. Unnecessary duplication of the documentation in the TMF should be avoided.

Examples of documents that are considered important when generated for a specific trial, but not listed in section 8 of the ICH GCP guideline include:

  • Completed forms, checklists and reports etc. related to the case, produced by the sponsor, investigator or any third party performing testing activities on their behalf from the following quality system procedures;
  • qualified person certification of the IMP;
  • assay method validation report for the analysis of IMP or metabolite(s) in clinical samples;
  • advanced therapy investigational medicinal product (ATIMP) traceability documents;
  • Documentation showing validity of computer system test-specific constructs (e.g. electronic case report form (eCRF) and interactive response technologies (IRT) and electronic patient-reported outcomes);
  • data management documentation, e.g. data management plan, data validation plan and data-review meeting minutes;
  • statistics documentation, e.g. SAS program validation, statistical analysis plan and sample size estimations;
  • Delegation log as part of TMF researcher / institution. Documentation showing software validation can be maintained by a CRO after the sponsor has contracted the operation, but the sponsor should ensure continuous access for the required archiving duration to such documentation in the contractual agreements with the CRO.

Documents relating to the trial-specific software configuration are part of the TMF and should be determined whether these are maintained/archived by the sponsor or CRO providing this service.

Some records from good manufacturing practice activities should also be defined as part of the TMF, for example, when these relate to the assembly and packaging of the investigational medicinal product (IMP) and confirm, as applicable, compliance with the randomisation schedule and blinding of the trial

Superseded documents

Throughout the development of a document ( e.g., creation and release of a clinical trial protocol), the procedures of the sponsor / CRO that require input and review by various roles. The documentation to demonstrate that the process was followed should be retained.

Reconstruction of the trial includes superseded copies of the final documents and should therefore be maintained in the TMF.

Superseded versions of the documents generated by the sponsor (e.g. trial protocol, IB and eCRF) should be available at the TMF investigator / institution in such a way as to allow reconstruction without access to the TMF sponsor, with proof of the date of receipt, review and/or approval (if necessary) and date of implementation by the investigator/institution.

Correspondence

Relevant correspondence that is essential for reconstruction of key trial conduct activities and decisions should be retained.

This includes correspondence with the ethics committees, data safety monitoring committee and regulatory authorities (confirming sponsor approval of processes, documents and decisions and the communication concerning issues that arise in the trial conduct and how they are dealt with).

In the same way, electronic correspondence (e-mails and associated attachments between CROs, sponsor departments, investigator/institution) should be readily accessible and may be retained electronically. It should be ensured that both sent and received correspondence is filed in the TMF.

One or more separate central repository may be used (e.g. for e-mail), as long as they are evidently defined as being  the part of TMF.

Care should be taken regarding e-mail ‘chains’ and attachments to ensure that relevant strands of conversations and their associated documents are maintained.

Contemporariness of trial master file

The requirement “at all times” within Article 57 of the Regulation means that the TMF must have all documentation added in a timely manner during the trial, as this greatly assists the successful 8 management of a trial by the investigator/institution, the sponsor and CROs to whom the sponsor has delegated its duties.

The deadlines for submitting and filing all documents to the TMF should be specified in the procedural documents or TMF plans. This is particularly important for more complicated TMF relationships involving multiple parties.

Quality of trial master file

Article 57 of the Regulation states “The clinical trial master file shall at all times contain the essential documents”. The sponsor and/or investigator/institution should implement risk-based quality checks (QC) or review processes to ensure the TMF is being maintained up-to-date and that all essential documents are appropriately filed in the TMF. Areas to consider during QC and review include the following:

  • all necessary documents generated available in the TMF;
  • documents filed in the suitable locations;
  •  documents added to the TMF on time;
  • documents correctly indexed;
  • documents only reachable according to the assigned roles and permissions;
  • review of the audit trail (for eTMF).

The sponsor should also undertake routine QA measures, e.g. system audits of the TMF-management processes. Additionally, the sponsor should ensure the TMF is readily available and directly accessible to the competent authority, e.g. for inspection purposes.

Security and control of trial master file

Access to trial master file

To ensure completeness and to avoid accidental or premature loss, unauthorized modification or destruction of records, the TMF should be handled safely at all times.

Access to the TMF should be dependent upon a summary of the position and permission specified by the sponsor and/or investigator / institution.

The TMF sponsor and investigator/institution TMF that provide some details that may unblind personnel who need to remain blind during the conduct of the trial.

This should be properly managed, for example, by storing the documents in another system or archive and/or by a summary of the function and authorization that is specified by the sponsor and/or investigator/institution.

Storage areas for the trial master file

The TMF documents storage area (such as paper or electronic media archives and server rooms) will also be appropriate to maintain the records in such a way that they remain accurate and legible throughout the trial conduct and the required retention period and can be made accessible to Member States’ competent authorities upon request.

sufficient and suitable space should be provided for the storage of all the essential documents from completed studies. The facilities should be protected, with appropriate environmental controls and adequate protection from physical damage.

The factors to be considered when assessing a appropriate storage facility, should take into consideration certain factors such as security, location (e.g. environmental risk factors) and size.

Sponsors should make a documented assessment of the conditions at the investigator site/institution for storage of the investigator/institution TMF during the clinical trial and for archiving. The sponsor should be notified, if the agreed arrangements are changed (e.g. sub-contracting of storage).

Sponsor/CRO electronic trial master file

Electronic TMFs should enable suitable security and reliability, ensuring that no loss, alteration or corruption of data and documents occur. The primary eTMF is a system for managing documents that should hold the controls listed below:

  • user accounts;
  • secure passwords for users;
  • a system in place locking/protecting individual documents or the entire eTMF (e.g. at time of archiving) to prevent changes to documents;
  • regular backup;
  • periodic test retrieval or restores to confirm the on-going availability and integrity of the data;  
  • an audit trail in the place to identify date/time/user details for creation and/or uploading deletion of and changes to a document (explanation of the deletion or modification, if necessary);
  • role-based permissions for activities being undertaken, such as restricted access to the files/documents (e.g. randomisation codes and unblinded adverse event data);
  • the appropriateness of the system for archiving purposes should be appropriate.”

https://www.ema.europa.eu/en/documents/scientific-guideline/reflection-paper-good-clinical-practice-compliance-relation-trial-master-files-paper/electronic-management-audit-inspection-clinical-trials_en.pdf

https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-content-management-archiving-clinical-trial-master-file-paper/electronic_en.pdf

http://www.ct-toolkit.ac.uk/routemap/trial-master-file/

https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-content-management-archiving-clinical-trial-master-file-paper/electronic_en.pdf

https://www.syneoshealth.com/solutions/clinical-development/trial-master-file-tmf-operations

https://khpcto.co.uk/SOPs/05_TMF.php



Coronavirus (COVID-19) Update: Daily Roundup July 8, 2020

July 08, 2020: “The U.S. Food and Drug Administration announced the following actions taken in its ongoing response effort to the COVID-19 pandemic:

  • As part of the FDA’s effort to protect consumers, the agency issued a warning letter to one company for selling fraudulent COVID-19-related products.

    The company, Ionogen, LLC, offers “Ionopure Skin & Hands” for sale in the United States with misleading claims that the products can mitigate, prevent, treat, diagnose, or cure COVID-19 in people.

    There are currently no FDA-approved products to prevent or treat COVID-19. Consumers concerned about COVID-19 should consult with their health care provider.

    Related News: https://lifepronow.com/blog/2020/07/07/coronavirus-covid-19-update-daily-roundup-july-6-2020/
  • Today, federal prosecutors in Miami charged four Florida residents who allegedly marketed “Miracle Mineral Solution (MMS),” a toxic bleach, as a cure for COVID-19, with conspiracy to defraud the United States, conspiracy to violate the Federal Food, Drug, and Cosmetic Act, and criminal contempt.

    In prior warning statements, the FDA has strongly urged consumers not to purchase or use MMS, explaining that drinking MMS is the same as drinking bleach and can cause dangerous side effects, including severe vomiting, diarrhoea, and life-threatening low blood pressure.

    As alleged in the criminal complaint, the defendants sold this dangerous product under the guise of Genesis II Church of Health and Healing (“Genesis”), an entity they allegedly created in an attempt to avoid government regulation of MMS.

    The FDA’s Office of Criminal Investigations conducted this investigation, with assistance from the Office of Chief Counsel.

    “The Genesis II Church of Health and Healing has actively and deliberately placed consumers at risk with their fraudulent Miracle Mineral Solution and Americans expect and deserve medical treatments that have been scientifically proven to be safe and effective,” said Catherine Hermsen, Assistant Commissioner of the FDA’s Office of Criminal Investigations.

    “We commend the efforts of our law enforcement partners for vigorously investigating this matter. The FDA will continue our efforts to make sure these and other like-minded sellers do not jeopardize the health of Americans during this pandemic and in the future.”
  • FDA is alerting consumers and health care professionals that additional hand sanitizer products have been added to the agency’s list of products with confirmed and potential methanol contamination.

    Consumers should check their hand sanitizer products to determine if a product is on this list.

    Methanol is not an acceptable ingredient for hand sanitizer products and can be toxic when absorbed through the skin as well as life-threatening when ingested. FDA is aware of reports of adverse events associated with hand sanitizer products, including those contaminated with methanol.

    FDA encourages health care professionals, consumers, and patients to report adverse events or quality problems experienced with the use of hand sanitizers to the FDA’s MedWatch Adverse Event Reporting program.

    (When reporting such events, please provide the agency with as much information as possible to identify the product).
  • Testing updates:
    • To date, the FDA has currently authorized 169 tests under EUAs; these include 141 molecular tests, 26 antibody tests, and 2 antigen tests.

      The FDA, an agency within the U.S. Department of Health and Human Services, protects the public health by assuring the safety, effectiveness, and security of human and veterinary drugs, vaccines and other biological products for human use, and medical devices.

      The agency also is responsible for the safety and security of our nation’s food supply, cosmetics, dietary supplements, products that give off electronic radiation, and for regulating tobacco products.”
      https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-daily-roundup-july-8-2020

Lynparza approved in the EU for pancreatic cancer

July 08, 2020: “AstraZeneca and MSD announced that Lynparza (olaparib) has been approved in the European Union (EU) for patients with germline BRCA-mutated (gBRCAm) metastatic pancreatic cancer.

Pancreatic cancer is a rare, life-threatening disease with the lowest survival rate among the most common cancers.

Approximately 5-7% of patients with metastatic pancreatic cancer have a germline BRCA mutation.2

The approval by the European Commission was based on results from the Phase III POLO trial, which were published in The New England Journal of Medicine. 

It follows the recommendation for approval by the Committee for Medicinal Products for Human Use of the European Medicines Agency.

Hedy L. Kindler, Co-Principal Investigator of the POLO trial and Professor of Medicine, University of Chicago Medicine, said: “Today’s approval opens the door to a new era of biomarker-led care for patients with metastatic pancreatic cancer in the EU, which has the highest incidence of any region globally. Lynparza now provides clinicians with a targeted, well-tolerated treatment option for patients with germline BRCA-mutated metastatic pancreatic cancer.”

Related News: Lynparza recommended for approval in EU by CHMP

FDA OK’s for Lynparza for metastatic castration-resistant prostate cancer

Lynparza approved in the US as 1st-line maintenance treatment with bevacizumab for HRD-positive advanced ovarian cancer

Dave Fredrickson, Executive Vice President, Oncology Business Unit, said: “Patients with metastatic pancreatic cancer historically have faced poor outcomes due to the aggressive nature of the disease and few treatment advances have been made over the last few decades.

In the POLO trial, Lynparza nearly doubled median progression-free survival versus placebo after 1st-line chemotherapy for patients with germline BRCA-mutated metastatic pancreatic cancer.

This approval underscores the importance of testing all patients for germline BRCA mutations at the time of diagnosis, as it will help inform personalised treatment options for patients in the EU.”

Roy Baynes, Senior Vice President and Head of Global Clinical Development, Chief Medical Officer, MSD Research Laboratories, said: “MSD and AstraZeneca are committed to advancing research into the treatment of patients with challenging types of cancer, including those with metastatic pancreatic cancer.

 Lynparza is now the only approved PARP inhibitor in biomarker-selected patients with metastatic pancreatic cancer. We look forward to making this targeted treatment option available for patients across the EU as quickly as possible.”

The POLO trial demonstrated that Lynparza nearly doubled the time patients with gBRCAm metastatic pancreatic cancer lived without disease progression or death to a median of 7.4 months versus 3.8 months on placebo. The safety and tolerability profile of Lynparza in the trial was consistent with previous trials.

Lynparza is indicated as monotherapy for the maintenance treatment of adult patients with germline BRCA1/2 mutations who have metastatic adenocarcinoma of the pancreas and have not progressed after a minimum of 16 weeks of platinum treatment within a 1st-line chemotherapy regimen.

Lynparza is approved in the US and several other countries as a 1st-line maintenance treatment for patients with gBRCAm metastatic pancreatic cancer based on the Phase III POLO trial, with ongoing regulatory reviews in other regions.”
https://www.astrazeneca.com/media-centre/press-releases/2020/lynparza-approved-in-the-eu-for-brca-mutated-metastatic-pancreatic-cancer.html

ViiV Healthcare announces superior efficacy cabotegravir for HIV-PrEP

“ViiV Healthcare announces superior efficacy of investigational, long-acting injectable formulation of cabotegravir dosed every two months over daily oral PrEP

07 July 2020: ViiV Healthcare announced that data presented from the HIV Prevention Trials Network (HPTN) 083 study demonstrated the superior efficacy of investigational, long-acting, injectable cabotegravir administered every two months when compared to daily oral emtricitabine/tenofovir disoproxil fumarate 200 mg and 300 mg (FTC/TDF) tablets for HIV prevention.

HPTN 083 is a Phase IIb/III randomised, multicentre, double-blind, clinical trial that is evaluating the safety and efficacy of long-acting, injectable cabotegravir for HIV pre-exposure prophylaxis (PrEP) among men who have sex with men (MSM) and transgender women who have sex with men.

The blinded phase of the study was stopped early in May 2020 following a pre-planned independent Data and Safety Monitoring Board review, which showed that long-acting cabotegravir was highly effective at preventing HIV in the study population.

Final analysis has since confirmed the superiority of long-acting cabotegravir, which was 66% more effective at preventing HIV when compared to daily oral FTC/TDF tablets. Study results were announced today at a press conference at the virtual 23rd International AIDS Conference (AIDS 2020) and will be presented at the conference on 8 July.

Related News: U.S FDA issues complete response letter to ViiV Healthcare for the use of investigational cabotegravir and rilpivirine long-acting regimen for the treatment of HIV

ViiV Healthcare to present new data on long-acting regimens for HIV prevention and treatment

Kimberly Smith, M.D., MPH, Head of Research & Development at ViiV Healthcare, said: “These data are truly ground-breaking, demonstrating that long-acting injectable cabotegravir dosed every two months is superior to daily oral FTC/TDF at preventing HIV in at-risk men and transgender women who have sex with men.

This advancement has the potential to be a game-changer for HIV prevention, offering an option with very high rates of effectiveness and the convenience of reduced dosing from daily to just six times per year.

We are thrilled with the results not only because of the high effectiveness of cabotegravir but also because this study adequately represents some of the populations most disproportionately impacted by HIV — black MSM in the US, young MSM globally and transgender women.”

In the final data analysis, 52 documented cases of HIV were observed in the HPTN 083 trial, with 13 cases occurring in the long-acting cabotegravir arm and 39 cases occurring in the daily, oral FTC/TDF arm.

This translated to an HIV incidence rate of 0.41% in the cabotegravir group (95% confidence interval [CI] 0.22%-0.69%) and 1.22% in the FTC/TDF group (95% CI 0.87%-1.67%).

Preliminary assessment of adherence to oral FTC/TDF was high, based on a random subset of 372 FTC/TDF participants that measured any detectable (> 0.31 ng/ml) tenofovir in 87% of participants and concentrations consistent with daily dosing levels (> 40 ng/ml) in 75% of all samples tested.

Despite this high level of adherence to oral therapy, long-acting cabotegravir demonstrated superiority in the primary efficacy endpoint of documented HIV incident infections, and was 66% (95% CI 38%-82%) more effective than FTC/TDF in preventing HIV acquisition in the study population. 

Long-acting cabotegravir and FTC/TDF tablets were both well tolerated throughout the study, with most adverse events being mild or moderate in nature and balanced between both treatment arms.

Injection site reactions, pyrexia, and hypertension were more common in the cabotegravir arm while nausea was more common in the FTC/TDF arm.

Most participants in the cabotegravir group (80%) reported pain or tenderness at the injection site, compared to 31% of those in the FTC/TDF arm, who received placebo injections. 

Discontinuation due to injection site reactions or injection intolerance in the cabotegravir arm of the study was 2.2% and there were no discontinuations due to ISRs in the FTC/TDF arm.

The HPTN 083 study enrolled HIV-negative men and transgender women who have sex with men, participants considered at increased risk for HIV acquisition. Two-thirds of study participants were under 30 years of age (median age of 26 years), and 12% were transgender women (n=567). Half of the participants in the United States identified as Black or African American (n=844).

Myron S. Cohen, M.D., Co-Principal Investigator of the HPTN and the Yeargan-Bate Distinguished Professor of Medicine, Microbiology and Immunology and Epidemiology at the University of North Carolina (UNC) at Chapel Hill, said: “Medicines that help prevent new HIV incidence are essential to our ongoing global fight to end the HIV epidemic.

It’s exciting to discover that with injectable, long-acting cabotegravir, we now have compelling clinical evidence of another effective PrEP option that could play a critical role in helping to reduce HIV transmission that will ultimately save lives.”

HPTN 083 was jointly funded by the U.S. NIAID, part of the NIH, and ViiV Healthcare, and was conducted by the HPTN. Study product was provided by ViiV Healthcare and Gilead Sciences.

In addition to the findings of HPTN 083, an additional study evaluating the safety and efficacy of long-acting cabotegravir for HIV prevention is being conducted in sexually active women (HPTN 084).

To date, more than 3,000 sexually active women in seven African countries have enrolled in HPTN 084, which is co-funded by NIAID, ViiV Healthcare and the Bill & Melinda Gates Foundation.

ViiV Healthcare plans to use the data from HPTN 083 for future regulatory submissions. Cabotegravir has not yet been approved for the treatment or prevention of HIV as a single agent by regulatory authorities anywhere in the world.

HPTN 083 (NCT02720094)
The HPTN 083 study is a phase IIb/III double blind study designed to evaluate the safety and efficacy of long-acting injectable cabotegravir for HIV prevention administered every eight weeks compared to daily oral FTC/TDF tablets (200 mg/300 mg).

Each participant was to receive a maximum of three years of blinded study medication.

The study opened to enrolment in November 2016. HPTN 083 was conducted in approximately 4,566 men who have sex with men and transgender women who have sex with men at research centres in Argentina, Brazil, Peru, United States, South Africa, Thailand and Vietnam.”

https://www.gsk.com/en-gb/media/press-releases/viiv-healthcare-announces-superior-efficacy-of-investigational-long-acting-injectable-formulation-of-cabotegravir-dosed-every-two-months-over-daily-oral-prep/

Placebo is?

1

“A placebo is anything that seems to be a “real” medical treatment — but it doesn’t.”

What is Placebo?

“A placebo is anything that seems to be a “real” medical treatment — but it doesn’t. It might be a pill, a shot, or some other type of “fake” treatment. All placebos have one thing in common is that they do not contain an active substance meant to affect health.

Normally, the person getting a placebo doesn’t know for sure that the treatment isn’t real.

The placebo is often in the form of a “sugar pill,” but it may also be an injection, a liquid, or even a procedure. This is intended to act like a true cure, but it does not affect the disease directly.

How Are Placebos Used?

During studies, researchers use placebos to help them understand what effect a new drug or some other treatment could have on a particular condition.

For example, a new drug may be given to some people in a study to lower the cholesterol. Others will get placebo. None of the study’s people would know if they got the real medication or placebo.

Researchers then compare medication and placebo effects on study participants. This way, they will assess the new drug ‘s efficacy and test for side effects.

Advantages of Using a Placebo

The major advantage of using a placebo when evaluating a new drug is that it weakens or eliminates the effect that expectations can have on the outcome.

If researchers expect a certain result, they may unknowingly give clues to participants about how they should behave. This can affect the results of the study.

To minimize this, researchers sometimes conduct what is known as a double-blind study. In this type of study, neither the study participants nor the researchers know who is getting the placebo and who is getting the real treatment.

By minimizing the risk of these subtle biases influencing the study, researchers are better able to look at the effects of the drug and the placebo.

A pain relief is one of the most observed and powerful placebo effects. According to some estimates, approximately 30% to 60% of people will feel that their pain has diminished after taking a placebo pill.

For example, imagine that a participant has volunteered for a study to determine the effectiveness of a new headache drug.

She notices that her headache dissipates soon after taking the medication, and feels much better. She later discovers, however, that she was in the placebo category, and that the medication she was given was simply a sugar pill.

How Placebos Are Used in Medical Research

In medical research, a placebo may be given to some individuals in a study, while others are being tested for the new treatment.

The purpose of doing this is to determine whether the new treatment is effective. If participants taking the actual drug show considerable improvement over those taking the placebo, the study can help support the claim for the effectiveness of the drug.

While a placebo has no effect on illness, it can have a real effect on how some people feel. The potency of this effect depends on a variety of factors. Some things that can influence the placebo effect include:

  • The nature of the sickness
  • How strongly the patient believes the treatment will work
  • The type of the response the patient expects to see
  • The type of the positive messages a doctor conveys about the treatment’s effectiveness
  • Genes may also manipulate how people respond to placebo treatments

Some people are genetically inclined to respond more to placebos. One study found that people with a gene variant that codes for the higher levels of the brain chemical dopamine are more prone to the placebo effect than those with the low-dopamine version.

People with the high-dopamine version of this gene also be inclined to have higher levels of pain perception and reward-seeking.

When testing new medications or therapies, scientists want to know if the new treatment works and if it’s better than what already exists.

Throughout their research, they learn the sort of side effects the new treatment might produce, which patients may benefit the most, and if the potential benefits outweigh the risks.

By comparing the effects of the treatment to a placebo, researchers hope to be able to determine if the effects of the medicine are due to the treatment itself or caused by some other variable.

What Is the Placebo Effect?

The placebo effect is defined as a phenomenon in which some people experience a benefit after an inactive “look-alike” substance or treatment is administered.

There’s no established medical benefit of this drug or placebo. The placebo is sometimes in the form of a pill (sugar pill) but can also be an injection (saline solution).

How do people undergo actual improvements arising from fake therapies? The patient’s beliefs will play a large part in the placebo effect.

The more a person expects treatment to work, the greater the likelihood that they will exhibit a placebo response.

In most cases, the person does not know that the treatment they are receiving is actually a placebo. Instead, they believe that they are the recipient of the real treatment.

The placebo is designed to seem exactly like the real treatment, whether it is a pill, injection, or consumable liquid, yet the substance has no actual effect on the condition it purports to treat.

It is important to note that a “placebo” and the “placebo effect” are the different things. The term placebo refers to the inactive substance itself, while the term placebo effect refers to any effects of taking a medicine that cannot be attributed to the treatment itself.

How does the placebo effect work?

In the past, some researchers have asked if there’s compelling evidence that the placebo effect is a real influence.

But there are studies that show that the placebo effect is true in some circumstances. For example, in response to a placebo, scientists have documented brain activity.

Since several experiments have shown a placebo effect, we know for certain that mind and body are connected in one way.

Some scientific evidence suggests that the placebo effect on pain may be due in part to endorphin releases in the brain. Endorphins are the ultimate killers of pain in the body. But it appears there’s more to it than this.

The expectation effect

Many believe the placebo effect is due to the patient believing in the substance, treatment, or doctor. The thoughts and emotions of the patient often induce physical changes in the brain or body at short notice.

The patient wants to feel better, and for some time he or she is feeling better.

But even if a person feels better after taking a placebo, this does not mean that the illness or symptoms of the individual were not real. The person can feel less nervous, for example, so the hormones of stress decrease.

For Example, taking a placebo can change their perception – a person may reinterpret a sharp pain as uncomfortable tingling.

The conditioning effect

One group had a real pain medicine in one study that looked at the placebo effect in pain relief, and the other did not. Both groups were given a placebo in the following days which looked like the real pain medicine.

Anyone who had administered the true pain medication could handle more discomfort than someone who had not previously provided pain medicines.

This helped to separate the power of the researcher telling them they would have an effect from the learned experience of having the effect in the past.

This type of learned response after the personal experience is called the conditioning effect. It seems to be part of what we call the placebo effect.

The nocebo effect

The nocebo effect, in which a person after a placebo has more symptoms or side effects, is still being studied. Researchers believe a substance in the body that sends signals through the nerves can explain this in part.

For example, when a person is concerned the substance is activated and the person feels more pain than a person who is not anxious.

The nocebo effect can be seen in the brain: brain-imaging studies have shown that pain is more intense when a person expects more pain than when they don’t. This is linked to changes in certain brain regions on the imaging studies.

The mind-body connection

Even though we may not know all the ways it could work, the idea that the mind can affect the body has been around for thousands of years and is well proven for some situations.

Most ancient civilizations relied on the relations between mind and body to manage the illness. Shamans or men in medicine would not have regarded their efforts as placebos.

But their healing powers may have worked partly through the patient’s strong belief that the shaman’s treatments would restore health.

Or it could be that a sick person was going to get better anyway, but recovery was thought to be because of the treatment – which might have really done nothing for the illness.

Because placebos frequently have an effect, even if the effect doesn’t last long, some people think that the placebo produced a cure.

But placebos do not cure. And in studies where doctors are looking at whether a tumor shrinks, placebos have very little, if any, effect.

Even, placebos can obviously help to alleviate other symptoms in certain people, such as discomfort, anxiety, and sleeping problems. In earlier times, doctors sometimes administered placebos out of frustration or desperation, because nothing else was available or seemed to work.

A 2008 study found that almost half of the doctors polled said that they used a placebo when they felt that it might help the patient feel better.

Some scientists believe that the effects of many alternative and complementary therapies can simply be a placebo effect. If the patient believes in the treatment and wants it to work, it may, at least for a while, appear to do so.

If the placebo worked on a disease that would not usually get better on its own, and it lasted, it would be considered a true cure, not a placebo effect.

Other things that can add to or be confused with the placebo effect

Certain other factors can affect study outcomes, and may be confused with the placebo effect. These are effects that can cause a study to report that a placebo (or even a treatment that doesn’t actually work) had some effect when it really didn’t.

Timing of unrelated events

Along with the placebo or nocebo effect, incidental events (unrelated effects that might have happened without the placebo) might also be linked to the placebo because of their timing.

For example, a headache or rash that happens soon after taking a placebo could be caused by something else entirely, but the person might think the placebo caused it. The same can be said for good outcomes: if a person happened to start feeling better after taking a placebo, it might be thought to be due to the placebo.

Healing or changes in symptoms

A health problem that improves on its own (many do) can sometimes add to what’s thought of as the placebo effect. Even in serious conditions such as cancer, some types appear to get better and worse on their own, although they continue to spread and worsen over time. This is part of the effect of timing, noted above.

A person who was taking a placebo when symptoms started improving on their own is very likely to believe that the placebo had some effect. And a self-limited illness that goes away completely on its own at such a time might have the placebo-taker convinced it was a miracle drug.

Timing can have another effect when symptoms cycle through getting better and worse on their own.

The patient is more likely to sign up for or be accepted into a study when their symptoms are very bad. But in cyclic illnesses, the worst symptoms are usually followed by a period when they start getting better – no matter what’s done.

This can make the test method seem like it works, or it can look like the placebo effect if the person ends up in the placebo group.

An advantage of having a control group that’s very much like the test group is that both groups should have about the same numbers of people who’ll get better with or without treatment, so they balance each other out.

Patient selection and sign-up

Patients who already have some faith in a particular method are more likely to sign up for a study about that method. For example, people who have faith in their medical doctors’ methods are more likely to sign up for a study on medical treatment.

People who believe that acupuncture works are likely to volunteer for a study of acupuncture. Those who don’t believe a method works probably won’t sign up to test it.

This means that the group who volunteers for the study already has some expectation effect before the study starts.

Many who sign up are likely to report at least a brief improvement in symptoms with the treatment (or even with a placebo, if one is used) because they expect to be helped by the treatment.

Patient reporting

Lack of blinding can affect reporting of these kinds of coincidental timing effects and belief in what caused them.

For example, people who know they’re getting placebos will know that their headache is not caused by the medicine, and they probably won’t report it.

Those who know or believe that they are getting the real medicine are more likely to believe that the medicine is causing the headache, and are more likely to report it.

Blinding and having groups that are very much alike help to balance these incidental timing effects.

Limits of the placebo effect

A 2010 study carefully evaluated over 200 trials that included a placebo group along with a group named “no treatment.” The reviewers found that placebo treatments have no major effects on disease but appear to improve patient-reported outcomes in some settings – particularly pain and nausea.

The placebo effects on pain varied quite widely, from nearly none to substantial relief. These variations in placebo effect were partially explained by differences in how experiments were performed, and what was said to patients.

The reviewers noted that it is hard to tell if patient-reported results were actually due to placebo effects or whether patient responses were simply biased.

In the end, placebos don’t cure. We can also make people feel better but they can make them feel bad as well.

And placebo effects (along with other factors that may affect outcomes of the study) often result in clinical trials that are not carefully designed with falsely positive outcomes. This is one of many reasons why good study design, like having control groups that are set up to be very similar to the test groups, are so important in human studies.”