Disclaimer

The ideas, views and opinions expressed in here in blog or comments and profile represent my own views and not those of any of my current or previous employer .They are based and taken from regulatory guidance available freely and my interpretations from my experience.

Sunday 22 November 2020

Monitoring safety of drugs:Pregnancy and Breastfeeding

 Pharmacovigilance in terms of monitoring safety of drugs in pregnant and breastfeeding mothers is very critical and also different. 

For knowing safety profile of drug during pregnancy or breastfeeding we are dependent on pharmacologic or chemical class of drug, or animal studies or clinical trials data.



Why critical?

1. Almost all clinical trials for registering new drugs exclude pregnant and breastfeeding women. So data is not available regarding effects of drug on this population easily.

2. Special clinical trials and data collection methods are required to monitor effects of drug on this population.

Hence to know safety profile of drugs during pregnancy, post marketing surveillance is major source of data. This is collected after the drug has been approved and used by pregnant women voluntarily when they know they are pregnant or when they used drugs without knowing in advance they were pregnant. 

Usage of drugs during pregnancy and breastfeeding is different than when used in general population for 2 reasons:

1. Use of drugs has to be thought in terms of its risks and benefits to mother and child both.

2. Risk of disease if left untreated without administering drug has to be weighed for both mother and child.

E.g. if breastfeeding is continued to benefit baby, is the risk greater to infant from receiving milk with drug excreted from mother in it or the mother's disease left untreated pose more risk in terms of quality of milk?

ICSR:

The route of administration in baby case (ICSR) is transplacental/transmammary and event is exposure in utero/Drug exposure via breast milk . The route of administration for the mother case is to be coded in correct fields in parent/child section .The outcome and event coding should correlate to correct patient i.e. baby and parent. 

Some important fields for cases are: Gestational age, Ultrasound results (in narrative), Use of other drugs especially teratogens, Outcome of pregnancy.It is also important to link cases.

It is also critical to know when to create 2 cases i.e. parent and child and when only 1 i.e. parent. The key to remember is usually if there is an adverse event for baby then 2 cases and if no adverse event then on;y 1 i.e. parent case. E.G.

1.  A pregnant women with abortion then only 1 case of mother with event abortion. However if its full term pregnancy and foetal death is outcome then 2 cases.

2. A pregnant women delivered baby with foetal abnormality then 2 cases baby and mother.

Expedited reporting of cases (parent and baby) follows same timelines. Cases with no adverse event or non serious preganancy cases, cases with termination of pregnancy are usually reported in PSURs/PBRER's.

Aggregate reports: 

PBRER/PSUR for product the frequency and template remains same for the drug concerned. The additional data to be included about drug use in pregnancy and breastfeeding should be described in relevant sections. The data sources is the ICSR received from spontaneous source, literature, pregnancy registries, studies etc where an pregnancy or breastfeeding exposure is reported with or without adverse pregnancy outcome.

Another additional section is drug utilisation by age group wherein it should be monitored if reproductive age group patients were exposed to the product to have data regarding quantitative exposure for this group.

The risk/benefit section should describe the cases of pregnancy and breastfeeding exposure and its relevance to safety profile of drug. If pregnancy and breastfeeding exposure is one of the safety concern in RMP then its important to mention the data relating to it in detail especially what is status of risk minimisation measures already implemented or if planned.

It is important to mention pregnancy/breastfeeding exposure as missing information in RMP and to monitor it.



Pregnancy registries: 

Different types of registries are maintained wherein data is collected regarding women who have been exposed to drugs during pregnancy and for drugs with longer half life's before pregnancy, male exposed to such drugs where pregnancy occurs following it, birth registers, birth defect registries etc.

When studied appropriately they provide very useful information in generating signal or at least signal hypothesis.

Studies conducted to study pregnancy and breastfeeding exposure and data collected from it. This study should be planned in structured manner and outcomes to be collected with longer follow ups (6-12 months at least after pregnancy period and include baby follow up too).

References:

https://www.ema.europa.eu/en/documents/scientific-guideline/draft-guideline-good-pharmacovigilance-practices-product-population-specific-considerations-iii_en.pdf

https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/guideline-exposure-medicinal-products-during-pregnancy-need-post-authorisation-data_en.pdf

https://www.ncbi.nlm.nih.gov/books/NBK208605/

https://www.fda.gov/science-research/womens-health-research/list-pregnancy-exposure-registries

Written by:
Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home

Tuesday 17 November 2020

Basics learnings from MDR (Medical Device Regulation)

Recently while discussing with colleague regarding device regulations, I realized much is not know about this topics. And decided to share my learnings via my blog to open a dialogue and learn more.

1.       What is medical device?

It is any instrument which is used in diagnosis, prevention, monitoring, prediction, prognosis, treatment, alleviation of disease ,injury or disability. Primarily it is an agent which does not alter or work on  pharmacological, immunological or metabolic systems of human body, but which assists this systems in its function.

Let's look at key points mentioned in EU-MDR for monitoring safety of medical devices (European Union's Medical Device Regulation document).

What is combination product?

21 CFR 3.2 (e) Definitions

(1) A product comprised of two or more regulated components, i.e., drug/device, biologic/device, drug/biologic, or drug/device/biologic, that are physically, chemically, or otherwise combined or mixed and produced as a single entity

(2) Two or more separate products packaged together in a single package or as a unit and comprised of drug and device, device and biological, or biological and drug product. [single entity]

(3) A drug, device, or biological product packaged separately that according to its investigational plan or proposed labeling is intended for use only with an approved individually specified drug, device, or biological product where both are required to achieve the intended use. [cross-labeled ]


·        1.  Reporting post market surveillance of serious incidents and field safety corrective actions through the electronic system:            

  •  any serious incident involving devices made available in EU, except expected side-effects which are clearly documented in the product information and quantified in the technical documentation
  •  any field safety corrective action in respect of devices made available in EU (includes actions from third country if the reason for the field safety corrective action is not limited to the device made available in the third country).
  • All serious related incidents no later than 15 days after MAH (Marketing Authorisation Holder*) become aware of the incident.
  • All serious related incident which pose serious public threat no later than 2 days after MAH become aware of the incident.
  • Serious related death incident, no later than 10 days after MAH become aware of the incident.
  • Inform field safety corrective actions in same timeframe as above before implementing them except in case of urgency when it can be notified after implementing.
  • Serious incidents and field safety corrective action which are identified and documented or a implemented and are common they may be submitted as periodic summary reports instead of individual serious incident reports, after consultation with the competent authorities.
HiHighlight:       

      MDRs are required when a manufacturer (or representative) becomes aware of information that reasonably suggests that their marketed medical device has or may have caused or contributed to a death, serious injury, or has malfunctioned and likely to cause or contribute to a death or serious injury if malfunction were to recur.

MDR Report Requirements:

5-Day MDR Report, described in 21 CFR 803.53(a), which requires a MDR to be filed no later than five business days following the occurrence of an event that necessitates remedial action to prevent an unreasonable risk of substantial harm to the public health

30-Day Device Malfunction Report, described in 21 CFR 803.20(b)(3)(ii), which must be filed no later than 30 days after receipt of a report of the malfunction of a device; this requirement applies only to certain permanently implantable, life-supporting or life-sustaining  medical devices


 


2.  Recording and reporting of adverse events that occur during clinical investigations:

             The sponsor shall fully record all of the following:

  • adverse event of a type identified in the clinical investigation plan as being critical to the evaluation of the results of that clinical investigation;
  • any serious adverse event;
  • any device deficiency that might have led to a serious adverse event if appropriate action had not been taken, intervention had not occurred, or circumstances had been less fortunate;
  •  any new findings in relation to any event referred above.

3. Other documents required:

  • Post market surveillance plan (PMCF) is necessary except for custom-made device and is part of technical documentation.
  • Clinical evaluation report is required
  • PSUR required for class IIa, class IIb and class III devices. For class IIb and class III devices update the PSUR at least annually. For class IIa devices update the PSUR when necessary and at least every two years. For class III devices or implantable devices, submit PSURs as specificed with other assessment documentation during registration. For devices other than mentioned, PSURs should be made available upon request of competent authorities (CA).
  • Trend analysis is required: any statistically significant increase in frequency or severity of non serious or listed side effects that can alter benefit-risk analysis. The MAH should inform such to competent authorities and also mention how to manage it.
  •   Field safety notices: Report containing UDI,SRN and field safety corrective action prepared by MAH and submitted to HCP and users after approval form CA.
  • RMP ( risk management plan) for each device is required.
  • For implantable devices and for class III devices, other than custom-made or investigational devices, the manufacturer shall draw up a summary of safety and clinical performance (equivalent to SmPC) to the public via Eudamed. This document should have UDI-DI and SRN if available other content remains same as of SmPC for drugs. This document should be made available on Eudamed (European database on medical devices).
  • .   * Also include manufacturer/distributors.
4. Terminologies and requirements:
  •   Having a Unique Device Identification (UDI) system in place which allows to trace the device throughout the system.The UDI shall be used for reporting serious incidents and field safety corrective actions .
  •    SRN (Single Registration Number) generated from electronic database by Competent Authority  while registering device for MAH
  • Eudamed is used for maintaining registration, manufacturer details,clinical investigation,certificates,pharmaocivgilance etc by Member States, notified bodies, economic operators and sponsor.
  • ‘incident’ means any malfunction or deterioration in the characteristics or performance of a device made available on the market, including use-error due to ergonomic features, as well as any inadequacy in the information supplied by the manufacturer and any undesirable side-effect
  • ‘serious incident’ means any incident that directly or indirectly led, might have led or might lead to any of the following:

  (a)the death of a patient, user or other person,

 (b)the temporary or permanent serious deterioration of a patient's, user's or other person's state of health,

(c)a serious public health threat;

(d) any of the above


5.   Regulatory representative:

  •   a diploma, certificate or other evidence of formal qualification in law, medicine, pharmacy, engineering or another relevant scientific discipline, and at least one year of professional experience in regulatory affairs or in quality management systems relating to medical devices;
  •   four years of professional experience in regulatory affairs or in quality management systems relating to medical devices.
  •   at least two years of professional experience within a relevant field of manufacturing.
  •   Micro and small companies do need to have this person within their organisation but have a person on contract permanently and continuously at their disposal.

References:

https://eur-lex.europa.eu/legal-content/EN/TXT/HTML/?uri=CELEX:32017R0745&from=IT#d1e1058-1-1

Written by:
Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home




Sunday 25 October 2020

Vaccine Pharmacovigilance Part- 3


In my last 2 blogs, we covered what are vaccines, challenges, benefits and key differences in vaccine pharmacovigilance. This is last blog on vaccines where we will see some interesting facts about vaccines.




  1.  We have only approx. 30 vaccines meaning only 30 diseases can be prevented as opposed to more than 20 thousand drugs.
  2.  WHO Recommends 15 vaccines currently for routine vaccination and 10 for selected usage.
  3.   UIP of India has  11 vaccines  in the program such as BCG,OPV,HEP B, Pentavalent vaccine (dipt,tetanus,pertussis,Hib and hep b),rotavirus, Pneumococcal Conjugate Vaccine, Fractional Inactivated Poliomyelitis Vaccine, Measles/ MR vaccine, Japanese encephalitis vaccine, DPT booster and Tetanus toxoid.
  4.    First vaccine used via inoculation since 1000 CE in China,India,Africs and Turkey but first vaccine discovered by Edward Jenner for small pox in 1796 and six years later was given to a 3-year old girl in Mumbai, first Indian person to receive the vaccine.
  5.  Case-In 2011, WHO and a group of partners developed a strategic document on vaccine safety called the Global Vaccine Safety Blueprint. GVSI, was set up to implement the Blueprint strategy that aims for enhancing global vaccine safety activities especially vaccine pharmacovigilance in all low- and middle-income countries.
  6. Vaccination is when a vaccine is administered to you (usually by injection). Immunisation is what happens in your body after you have the vaccination.
  7.  MenAfriVac, a revolutionary 1st vaccine developed in 2012 in collaboration with Serum Institute of India through the WHO and PATH Meningitis Vaccine Project, gained approval for use outside the cold chain – for as long as 4 days without refrigeration and at temperatures of up to 40°
  8.    Encouragingly, a recent report from the Pharmaceutical Research and Manufacturers of America listed 145 new vaccines undergoing clinical trials testing, with many targeting infections for which there is no current vaccine (PhRMA, 2010).

 Some sources to train or learn more on vaccines:

World Health Organization Global Training Network.https://www.who.int/ith/vaccines/en/

 

References:

1.      https://www.microbiologyresearch.org/docserver/fulltext/jmm/61/7/889_jmm039180.pdf?expires=1563520785&id=id&accname=guest&checksum=CFDFF62D1140C568A15423186A717EFC

2.       World Health Organization Global Training Network. "Similarities and differences between vaccines and medicines," In: WHO GlobalTraining Network: Adverse events following immunization (AEFI), Geneva: WHO, 2009

3.       http://medind.nic.in/iby/t14/i4/ibyt14i4p491.pdf

4.      https://www.microbiologyresearch.org/docserver/fulltext/jmm/61/7/889_jmm039180.pdf?expires=1563520785&id=id&accname=guest&checksum=CFDFF62D1140C568A15423186A717EFC

5.       https://pediatrics.aappublications.org/content/138/3/e20162146

6.       https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/guideline-conduct-pharmacovigilance-vaccines-pre-post-exposure-prophylaxis-against-infectious_en.pdf

7.      https://cdsco.gov.in/opencms/export/sites/CDSCO_WEB/Pdfdocuments/biologicals/3GuidanceBioloGicalProducts.pdf

8.       https://jamanetwork.com/journals/jama/fullarticle/2275444?resultClick=1

9.       https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf

10.   https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf

11.  http://www.wpro.who.int/topics/immunization_safety/ImmunizationSafetySurveillance.pdf

12.  https://jmm.microbiologyresearch.org/content/journal/jmm/10.1099/jmm.0.039180-0;jsessionid=UqYxJJOAuK-VvmyvN7g9HPSv.x-sgm-live-03#tab2

13.  http://www.searo.who.int/india/topics/routine_immunization/AEFI_standard_operating_procedures_SOPs_2010.pdf


Written by:
Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home

Tuesday 13 October 2020

Vaccine Pharmacovigilance-Part 2

In my last blog we talked about what are vaccines and what are unique challenges in vaccine development and benefits of vaccine.

 In this blog lets cover what are the key differences in vaccine pharmacovigilance.

1.Manufacturing or testing:

Testing of every batch is not done for other drug products. The lot release system is perhaps the greatest difference between the NRA vaccine functions and NRA functions for other medicines 



2. Collection of adverse events:

  • ·         Pre-licensure studies (mostly Randomized Controlled Trialss) often identify common and acute negative reactions that occur with a frequency greater than 1 in 10,000 vaccinations, depending on total sample size of the study. Detection of uncommon or rare adverse events, or delayed onset AEs is, however, low in trials.
  • ·         Post-licensure monitoring of vaccine safety is needed to identify and evaluate such adverse events. Post-licensure clinical trials and phase IV surveillance studies are almost mandatory for vaccine safety.
  • ·         Passive surveillance systems (or spontaneous reporting systems) is cornerstone for monitoring vaccine surveillance.
  • ·         AEFI programs are mainstay in terms of ensuring vaccine safety e.g.The Vaccine Adverse Event Reporting System (VAERS) system in U.S and AEFI program in India that collects and analyze vaccination related AE’s separately.

3. Processing of adverse events:

  • ·         Causality- Similar as drug safety but important addition would be, clinical or laboratory proof that the vaccine caused the event. Causality assessment has to be performed for eligible AE’s to ensure to establish if it is vaccine related,immunization errors or coincidental event.
  • ·         Time to onset- Delayed onset reactions due to immunological factors
  • ·         Action taken- This includes risk prevention and risk minimization measures such as withdrawal of the lot, changing manufacturing specifications or quality control, SmPc/PIL update,change in logistics for supplying vaccine,change in procedures at the health facility, training of health workers and intensified supervision.
  • ·         Dechallenge and Rechallenge- This information is not always available or possible
  • ·         Follow up - HCPs (Healthcare Professionals) may hide administration or storage errors,lot number is must which many times is missed.
  • ·         Risk-Benefit assessment –This is done at an community level and not limited to individual level.
  • ·         Safety Reporting forms - This are different for vaccines, e.g.VAERS Form, AEFI form.

 

4. Reportable AEFI's :

 Adverse Event Following Immunization (AEFI)- This are the AEs for which there is mandatory requirement for reporting to MAH (Marketing Authorization Holder) or HA (Health Authority).

  • ·         serious AEFIs,
  • ·         signals and events associated with a newly introduced vaccine
  • ·         AEFIs that may have been caused by an immunization error-related reaction
  • ·         significant events of unexplained cause occurring within 30 days after vaccination
  • ·         events causing significant parental or community concern
  • ·         Swelling, redness, soreness at the injection site IF it lasts for more than 3 days or swelling extends beyond nearest joint

 

So this are the key differences we need to keep in mind when processing vaccine AEs. There are of course more, this is short summary.

In my next blog, lets discuss with examples some interesting facts about various vaccines.

References:

  1. World Health Organization Global Training Network. "Similarities and differences between vaccines and medicines," In: WHO GlobalTraining Network: Adverse events following immunization (AEFI), Geneva: WHO, 2009
  2. http://medind.nic.in/iby/t14/i4/ibyt14i4p491.pdf
  3. https://www.microbiologyresearch.org/docserver/fulltext/jmm/61/7/889_jmm039180.pdf?expires=1563520785&id=id&accname=guest&checksum=CFDFF62D1140C568A15423186A717EFC
  4. https://pediatrics.aappublications.org/content/138/3/e20162146
  5. https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/guideline-conduct-pharmacovigilance-vaccines-pre-post-exposure-prophylaxis-against-infectious_en.pdf
  6. https://cdsco.gov.in/opencms/export/sites/CDSCO_WEB/Pdfdocuments/biologicals/3GuidanceBioloGicalProducts.pdf
  7. https://jamanetwork.com/journals/jama/fullarticle/2275444?resultClick=1
  8. https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf
  9. https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf
  10.  http://www.wpro.who.int/topics/immunization_safety/ImmunizationSafetySurveillance.pdf
  11. https://jmm.microbiologyresearch.org/content/journal/jmm/10.1099/jmm.0.039180-0;jsessionid=UqYxJJOAuK-VvmyvN7g9HPSv.x-sgm-live-03#tab2
  12. http://www.searo.who.int/india/topics/routine_immunization/AEFI_standard_operating_procedures_SOPs_2010.pdf
  13. http://vaccine-safety-training.org/vaccine-procurement-and-lot-release.html

Written by:
Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home

Wednesday 23 September 2020

Vaccine Pharmacovigilance-Part 1

 In current situation, all of us are desperately waiting for only 1 thing and that is Corona vaccine. We need an effective vaccine which will help us have our pre corona life back, right? Agreed, but as safety physician i also keep thinking about safety of such vaccine when we finally get it.

I started my career in pharmacovigilance with vaccines. I was fortunate to learn about vaccine pharmacovigilance at start of my career, which gave me peek into how vaccine pharmacovigilance and drug pharmacovigilance are different.

What are the key differences in vaccine pharmacovigilance? But before we jump to that, lets cover some basics.



What is a vaccine? 

A vaccine is a biological preparation containing an agent (disease causing microbe or its part) which stimulates the body's immune system to recognize the agent as foreign,destroy it, and "remember" it, so that the immune system can more easily recognize and destroy any of these microorganisms that it later encounters.

How does it differ from a drug and what are challenges of vaccine?

We have lower risk tolerance to adverse event associated with vaccines as they are given to prevent disease, unlike drugs which are given to treat disease.

Vaccines teach immune system of our bodies to fight infection, however we all know how immune system is complicated and every persons immune system can have different reactions. Hence its very critical to monitor vaccine reactions and gather data regarding same to know such reactions post vaccination and if possible be prepared to prevent them. This takes time and lot of research and costs money.

Scientific challenges in developing vaccines:

  1. Lack of knowledge about immunological pathways causing serious safety concerns
  2. Antigenic variation requiring frequent updates
  3. Inadequate preclinical data which does not provide enough information with immunity correlation results in clinical trial failures
  4. Lack of data regarding infectious exposure of intended vaccine recipients
  5. Product related issues-storage, administration e.g cold storage
  6. Vaccines contain different components to make them effective. However, each component in a vaccine adds a potential risk of an adverse reaction.

Reading about such vaccine reactions, one might think why not let body's immune system learn to fight by directly getting a disease, why administer vaccine to healthy people?

But we must remember below benefits of vaccines:

1. the very low risk of an adverse event caused by a vaccine greatly outweighs the risk of illness and complications caused by natural infection.

2.Vaccines have wide reach-protect individuals, communities and societies

3.Vaccines have rapid impact- Immediate reduction in morbidity and mortality

4.Vaccines save lives and costs.

In a nutshell, it takes time,efforts,money to bring a vaccine to market, however they very effective in controlling diseases.

In my next blog, we will talk about differences in vaccine pharmacovigilance ...


References:

  1. World Health Organization Global Training Network. "Similarities and differences between vaccines and medicines," In: WHO GlobalTraining Network: Adverse events following immunization (AEFI), Geneva: WHO, 2009
  2. http://medind.nic.in/iby/t14/i4/ibyt14i4p491.pdf
  3. https://www.microbiologyresearch.org/docserver/fulltext/jmm/61/7/889_jmm039180.pdf?expires=1563520785&id=id&accname=guest&checksum=CFDFF62D1140C568A15423186A717EFC
  4. https://pediatrics.aappublications.org/content/138/3/e20162146
  5. https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/guideline-conduct-pharmacovigilance-vaccines-pre-post-exposure-prophylaxis-against-infectious_en.pdf
  6. https://cdsco.gov.in/opencms/export/sites/CDSCO_WEB/Pdfdocuments/biologicals/3GuidanceBioloGicalProducts.pdf
  7. https://jamanetwork.com/journals/jama/fullarticle/2275444?resultClick=1
  8. https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf
  9. https://cioms.ch/wp-content/uploads/2017/01/report_working_group_on_vaccine_LR.pdf
  10.  http://www.wpro.who.int/topics/immunization_safety/ImmunizationSafetySurveillance.pdf
  11. https://jmm.microbiologyresearch.org/content/journal/jmm/10.1099/jmm.0.039180-0;jsessionid=UqYxJJOAuK-VvmyvN7g9HPSv.x-sgm-live-03#tab2
  12. http://www.searo.who.int/india/topics/routine_immunization/AEFI_standard_operating_procedures_SOPs_2010.pdf

Written by:
Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home


Sunday 5 July 2020

Narrative writing in pharmacovigilance

In pharmacovigilance(PV), narrative writing is an art of describing a single ICSR /  or multiple ICSR in aggregate manner but with scientific and medical sense.

Narratives are part of various reports in PV such as clinical study reports, IND (Investigational New Drug) safety letter, PBRER(Periodic Benefit Risk Evaluation Report), Signal management reports etc. But for the ease of understanding and presentation, we will talk about narratives for ICSR (Individual Case Safety Report).

Each MAH (Marketing Authorization Holder) has their own internal SOPs (Standard Operating Procedures)  as guidance and specific format of narratives. The guidelines available from the Health Authorities (HA) describes the elements that are expected by them without template, hence each MAH has to decide own template keeping HA guidance in mind.

In this blog we will cover narratives for different types of ICSR , sample format and brief summary of 3 HA guidelines on narrative writing.

A. Clinical trial (CT) cases/ICSR:

1. Relevant narrative only:
 Usually this ICSR have lot of information and data because the reporter (Investigator) is mandated to provide it. Hence, writing clinical case narratives should be focused more on event and investigational drug while applying clinical knowledge to exclude large portions of  data that has no relevance to event or drug (e.g. hospital discharge summaries, lab data unrelated to event etc).

2. Specific format:
 For this ICSR specific format should be followed. The narrative should include information such name of clinical trial, patient details (age, gender), medical history (most relevant to reported event and/or to disease under investigation), concomitant medications (special attention to any prohibited medications, drug-drug interactions etc.)

3. Blinding information:
Generally, the technical setting in PV database should be checked as narratives are reflected on *CIOMS. Hence, refrain from adding comments that disclose blinding information (e.g. placebo or investigational drug).  Reason being, IND safety letter and/or as part of site TMF (trial master file), CIOMS are forwarded to site and this will be violation of GCP (Good Clinical Practice) in case of double blinded study.

4. Missing information or follow up attempts:
It is a good practice to mention regarding missing critical information required for assessing causality of the ICSR. Apart from being it a good documentation practice, it is helpful for a reader assessing this ICSR later, to understand follow up attempts were made regarding the ICSR for critical information necessary for complete assessment of ICSR.

5. Reporting as it is:
Refrain from making own conclusions in ICSR’s especially in CT cases, as these are solicited cases and clarification can always be sought from Investigator (Medical doctor) instead of making own conclusions.


B. Spontaneous cases:

These are the cases that are reported by patients/consumers, which has very minimum information or vague information. Many times, the source document is also not readable. The events, drug or patient details are also bare minimum. So narrative writing becomes very tedious. The focus should be to make a clincial story that makes sense without confusing readers with lot of redundant sentences just to follow the narrative template of MAH.

1. Missing information - There is lot of missing information, writing redundant sentences such as “medical history not available, comments not available” etc should be avoided. This should be summarised in company comment. Even if MAH do not write company comments it is still fine to avoid writing it. As it is not mandatory to have company comment, is a good practise to write follow up sentence in narrative, mentioning that “medical history, comments etc” missing information was sought and awaited or will not be available. Including lost to follow up sentence is very important which comes handy, when undergoing audit or inspection, where we can document that all follow up attempts were made and no data was still reported.

2. Language – As these are unsolicited ICSR which are reported by consumers/patients the language is non-medical which makes very less sense. Additionally, if the reporters first language is not english then writing narrative is  more tedious. In such cases, only coherent and relevant data should be picked up from source document by using own clinical judgement and reproduced in narrative in a sensible clinical story pattern.

3. Template – Lot of MAH have auto narrative feature in PV database, but it makes no sense in unsolicited cases when lot of data is missing and events as reported terms (these are exact terms used by reporter to describe events which are very vague) are populated in narrative. Hence PV professional should edit it again to make it coherent. Necessary template should be used as guidance and set criteria for writing narrative. The goal should be to make sense of what happened to patient (event and drug) and then present it in meaningful way.

C. Flow:

As an example, here is the flow that is usually followed by most of the MAH.

1. Paragraph 1:

1.1 Solicited or unsolicited case (spontaneous, CT, literature, legal, compassionate use, pregnancy registry, HA cases) .
1.2 CT case – Study ID and CT title and EUDRA-CT number.
1.3 Patient identifiers: Pay attention to local country specific HA requirement . (e.g. GDPR).

2. Paragraph 2: Event and suspect drug details.

3. Paragraph 3: Medical history,surgical history, historical drugs and concomitant medications.

4. Paragraph 4: Description of event/s in relation to suspect drug/s, when patient started taking suspect drug/s, after how much time he/she had event/s, seriousness of event/s, lab investigations to diagnose event/s, treatment given, action taken with suspect drug/s, dechallenge/rechallenge and outcome of event/s.

5. Paragraph 5: Reporter comments (Investigator/Reporter/Author) – reporters assessment of seriousness and causality.

6. Paragraph 6: Company comment: This is not always written by all MAH but expected for CT cases. This should be written for CT cases by HCPs (HealthCare Professionals). For other cases, written by HCP/PV professionals. It describes company’s stand regarding causality and listedness/expectedness of event/s.

7. Paragraph 7: As a good practice many MAH’s include a sentence mentioning follow up attempts made to sought missing information to reporter. The usual industry practice is 3 attempts for serious cases and 2 attempts for non-serious cases after which a close of follow up attempt sentence is added.


D. Health Authority Guidelines for narrative writing:

Usually HA guidelines provide outline regarding what information should be added for a good narrative but do not give very strict specific format. As a brief note, below information from EMA, FDA and Health Canada guidelines can be checked.

1. EMA and ICH guidelines: ICH-E2D and  GVP Annex IV: These talk about presenting information in a logical time sequence meaning how the patient started experiencing event i.e. appearance of first symptoms after taking suspect drug and then following the chronology in terms of clinical course of event, lab, treatment measures, outcome and follow-up information awaited or received. In case of death ICSR, relevant autopsy or post-mortem data should be added. With respect to the patient identifiers, information should be provided in accordance with local data protection laws. Case narratives should not include information that could lead to the identification of the patient, his/her hospital, HCP, treatment center etc details.

2. FDA guideline :  As per FDA, a good case reports include the following elements:
Description of the adverse events or disease experience and it’s clinical course including time to onset of signs or symptoms;  Suspected and concomitant product therapy details, Patient details, including medical history;  Documentation of the diagnosis of the events, patient outcomes (e.g., hospitalization or death); Relevant therapeutic measures and laboratory data at baseline, during and post recovery of event, during therapy, dechallenge and rechallenge; and any other relevant information

3. Health Canada: As per this guidelines, the narrative should describe the following: the nature and intensity of event, its clinical course leading up to event, with an indication of timing relevant to test drug/investigational product administration; relevant laboratory measurements, whether the drug was stopped, and when; countermeasures; post mortem findings; investigator's opinion on causality, and sponsor's opinion on causality, if appropriate. In addition, the following information should be included:Patient identifier with medical history, relevant concomitant/previous medication with details of dosage, Test drug/investigational product administered, drug dose, if this varied among patients, and length of time administered.
*CIOMS~Also includes other reportable formats required by local HA's.



References:

1. FDA: IV - B. Characteristics of a Good Case Report https://www.fda.gov/files/drugs/published/Good-Pharmacovigilance-Practices-and-Pharmacoepidemiologic-Assessment-March-2005.pdf


2. EMA: VI.C.6.2.2.4. Case narrative, comments and causality assessment https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/guideline-good-pharmacovigilance-practices-gvp-module-vi-collection-management-submission-reports_en.pdf

3. Health Canada: 12.3.2 Narratives of Deaths, Other Serious Adverse Events and Certain Other Significant Adverse Events https://www.canada.ca/en/health-canada/services/drugs-health-products/drug-products/applications-submissions/guidance-documents/international-conference-harmonisation/efficacy/structure-content-clinical-study-reports-topic-health-canada-1996.html#a12.3.2

4. FDA: https://www.fda.gov/media/73593/download


Written by:

Dr.shraddha Bhange.
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me: ThinkSharp Foundation http://thinksharpfoundation.org/#home
Special Thanks for co content creation- Dr.Suresh PVVD.


Tuesday 16 June 2020

Timelines for adverse drug reports in Pharmacovigilance


Have you wondered why PV professionals seem to be under stress regarding meeting timelines? Its friday evening and you have plans but then the last minute SUSAR comes in inbox and your friday is gone.We all have some or the other time faced this as PV professionals.

What are the timelines in PV and why they are so urgent and important?

We can divide this topic in 2 sections for ease of understanding.

1. ICSR timelines (Individual Case Safety Reports)/Adverse drug event reports.*
2. Aggregate report timelines
3. Others

In this blog i wanted to cover ICSR timelines and Aggregate report consisting of ICSRs, so as to keep the length of blog short and easy for comprehension.

Every Health Authority (HA) has there own timelines as per regulations, i will highlight four main HA timelines i.e. EU, US, India and Japan.

1.  Europe

a) Spontaneous ICSRs:

Submit the valid ICSR (EEA) (European Economic Area) and non-EEA serious (non life threatning and non -fatal) within 15 days from initial receipt of the information, and EEA non-serious ICSR within 90 days from initial receipt of the information to EudraVigilance (EV)).

 Non-serious non-EEA ICSRs should not be submitted to EV.

We should remember it says within 15 days, most of the HA mean it as soon as possible but within 15 days, this is important to understand and make sure to write it this way in internal SOPs. 
Also, serious (fatal and life threatening) ICSR the timeline is within 7 days for EEA and Non-EEA.

b) Clinical ICSRs:

SUSARs (suspected serious unexpected adverse reactions) that are fatal or life-threatening are submitted as soon as possible but within 07 days and relevant follow-up information within an additional 08 days from initial receipt of the information .
In addition to reporting to EU, this needs to be submitted to Ethics Committee and Investigators. 

All other suspected serious unexpected adverse reactions  (non life threatening and non -fatal) are submitted to the HAs concerned and to the Ethics Committee concerned as soon as possible but within 15 days from initial receipt of the information

2. United States

a) Spontaneous ICSRs:

ICSRs that are serious and unexpected, whether foreign or domestic, are to be submitted as soon as possible but within 15 calendar days from initial receipt of the information and must submit follow-up reports within 15 calendar days of receipt of new information to FDA.

The non-SUSAR cases timelines are covered in aggregate reporting timelines.

b) Clinical ICSRs

Submit unexpected fatal or life-threatening suspected adverse reaction (SUSARs) as soon as possible but within 7 calendar days from initial receipt of the information to FDA.

In addition to FDA, they are to be submitted to all participating investigators  in an IND safety report  as soon as possible, but within 15 calendar days from receipt of information and to Ethics committee as applicable.

3. INDIA

a) Spontaneous ICSRs:


All serious AEs/ADR (adverse events (AE) and adverse drug reactions (ADR)) must be reported to regulatory authority CDSCO/PvPI, IPC within 15 days from receipt of information .

All non-serious AEs/ADR must be reported to CDSCO/NCA-PvPI, IPC within 30 days from receipt of information.


b) Clinical ICSRs: All serious adverse events must be submitted to DCGI within 14 calendar days receipt of information.

In addition to DCGI, it needs to be submitted to ethics committee (local and national) and Investigators. 

One important thing to note here is, for India, it says licensing authority and regulatory authority. Licensing authority can be DCGI or state FDA and regulatory authority is CDSCO but submission to  PVPI is needed too. This is my understanding. There is email (xml file + DCGI format) and paper submission.

4. Japan

 ICSRs:

ICSRs that are serious and unexpected (unpredictable), are to be submitted as soon as possible but within 15  days from initial receipt of the information to PMDA.

ICSRs that are serious and expected (predictable) within 30 days. This excludes serious death expected ICSR which were caused by new drug  within 2 years of approval and ICSR from drug which is under EPPV program (Early phase post marketing vigilance) which are to be submitted within 15 days.

Non serious unexpected (unpredictable) ICSRs are submitted annually and non serious listed (predicatble) not submitted. 

EPPV-Early phase post marketing vigilance - This  is for newly approved drugs when 2 years have not passed from the date of approval for drugs


Aggregate reports:

1. US:

ICSRs that are serious listed, non-serious unlisted and listed are submitted at quarterly intervals, for 3 years from the date of approval of the application, and then at annual intervals.Each quarterly report is submitted within 30 days of the close of the quarter and each annual report within 60 days of the anniversary date of approval of the application. Follow-up information to adverse drug experiences submitted in a periodic report may be submitted in the next periodic report.

*for the sake of ease i have considered ICSR and Adverse drug reaction report and event as equivalent, however they are not and standard definitions can be found in HA guidance documents.


References:




 Written by:


Dr.shraddha Bhange.

Content co-developed by:

Dr.Sridhar Yeshamaina
Connect with me Via comments below or on linkedin. (I do not respond to Facebook messages)
Support the cause of better rural education with me:ThinkSharp Foundation http://thinksharpfoundation.org/#home


Disclaimer

Disclaimer