You are on page 1of 8

11/29/2019 Compensation for injury in drug trials in India

11 29 2019 Last update Sat, 22 Jul 2017 12pm SEARCH

Public health
HOME AGENDA SPECIAL SECTIONS

You are here: Home | Public health | Public health | Analysis | Compensation for injury in drug trials in India

Compensation for injury in drug trials in India SPECIAL SECTIONS

Children

Can the new rules deliver justice, asks Sandhya Srinivasan


Corporate responsibility
In the last decade, there have been many reports of unethical and illegal clinical trials in India. It is only recently
that the government framed rules to monitor their conduct. In 2014, rules were devised on compensation to be Digital inequality
paid to participants or their families in cases of injury and death in clinical trials. However, there are challenges to
be met much before compensation amounts can be determined, starting from the manner in which trials are
Disabilities
conducted extending to the procedure for assessing whether an injury is related to the trial. Some of these
Malnutrition
problems may be reduced with the planned improvements in regulation. However, trial sponsors are likely to resist
regulation, participants remain vulnerable, and regulatory agencies have a poor track record in protecting people’s Disasters
rights.
Education
Following the January 2005 amendments to Schedule Y of the Drugs and Cosmetics Act (GoI 2005) regulating
clinical trialsi for approval of new drugs, the multinational pharmaceutical industry rushed to India, which the
government was promoting for its lower costs, available infrastructure, trained healthcare personnel, and a large Environment
population on whom trials could be conducted (Sengupta 2009). As of June 30, 2010, 666 of the 1,078 trials
registered on the Indian registry for clinical trials were for drugs, the overwhelming majority sponsored by the Food security
private pharmaceutical industry (Nikarge 2010).

Globalisation
However, there were no regulatory mechanisms in place to protect the rights and health of the people on whom
new drugs were being tested. In the absence of such rules the office of the Central Drug Standards Control Medical technology:
Organisation (CDSCO) – the government regulatory body whose work includes governing the conduct of clinical Ethics Governance
trials towards approval of new drugs – was grossly negligent in its duties, even functioning as a “facilitator for
industry” (Jesani 2013: 76). HIV/AIDS

Though Schedule Y required that trials follow the Indian good clinical practice (GCP) guidelines for clinical
research (CDSCO 2001), neither document contained detailed rules on the requirements of institutional ethics Human rights
committees (IECs) which review and monitor trials; on the responsibilities of investigators who conduct trials; on
how injuries and deaths in trials were to be reported, investigated, and compensated, and so on. Industry exploited Livelihoods
these loopholes, with more than a little help from the government. A government report on the functioning of the
CDSCO found that it was substantially understaffed, and needed a drastic upgrade in technology, infrastructure
Media
and training to meet the job of monitoring the influx of trials. Most important, it noted “a collusive nexus between
drug manufacturers, some functionaries of the CDSCO and some medical experts” (GoI 2012: para 7.36). This
nexus has certainly been apparent in the reports of the last decade. Population
HIV/AIDS: Big questions

The right to compensation for trial-related injury Poverty

In India, the subject of compensation for injury or death in trial has been discussed in the course of the campaign
against unethical trials and poor regulation. However, this right has nothing to do with the ethics of a trialii. Trial Public health
subjects are injured in even the best run trials. Pharmaceutical companies conduct clinical trials to gather
information on a drug’s safety and efficacy, which they use to get marketing approval. People who participate in Technology
the trial take risks to provide this information. It goes without saying that participants who are injured or die in
these trials have a right to medical treatment as well as compensation for any harm caused by taking this riskiii.
Trade and development
Jesani has pointed out that all trial participants, whether on the experimental drug or a standard treatment, “are
experimented upon and take risks in drug trials, and so have some right to compensation when they suffer injury
or death”. (Jesani 2013: 78) Urban India
Access to Healthcare
The right to compensation for injury or death in a clinical trial is enshrined in the guidelines of international bodies Water resources
such as the World Medical Association’s Declaration of Helsinki that lays down ethical principles for medical
research (WMA 2013). The right to compensation is also stated in Indian guidelines, though the imprecise HEALTHCARE
Women
language has worked to the benefit of pharmaceutical companies. MARKETS AND YOU
The Wild West of stem cell
According to Indian GCP guidelines, subjects who suffer injury or death as a result of trial participation are entitled procedures
to be compensated “equitably for any temporary or permanent impairment or disability subject to confirmation from
IEC.” (CDSCO 2001: 2.4.7). The guidelines also assert that this applies to all trials, whether conducted by “a The corrupt doctor
pharmaceutical company, a government, or an institution” (CDSCO 2001: 2.4.71). The sponsor should agree,
before the research begins, “to provide compensation for any serious sphysical [sic] or mental injury for which Pirates of traditional
subjects are entitled to compensation or agree to provide insurance coverage for an unforeseen injury whenever knowledge
possible.” (CDSCO 2001: 2.4.7.1) Prospective trial subjects should be informed of their right to compensation.
Before a clinical trial starts, the researcher must provide evidence that compensation for trial-related injury will be A vaccine for every
available. The trial sponsor is responsible for ensuring that the investigator promptly reports injuries and deaths to ailment
the IEC and other competent authorities, and submits all medical and related documents.
Why does a drug company
However, the GCP guidelines do not specify details such as who determines whether or not the injury is related to sponsor research?
the trial, how this is determined, or what an “equitable” compensation is. They only state that compensation is
“subject to confirmation from IEC”.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 1/8
11/29/2019 Compensation for injury in drug trials in India
Similarly, the guidelines of the Indian Council of Medical Research (ICMR) state: “The sponsor whether a
pharmaceutical company, a government, or an institution, should agree, before the research begins, in the a priori
agreement to provide compensation for any physical or psychological injury for which participants are entitled or
agree to provide insurance coverage for an unforeseen injury whenever possible.” (ICMR 2006: 29). The research
institution “could” set up an arbitration committee “where such a step is feasible”. (ICMR 2006: 29) However, they
give no further details on the roles required to be played by the sponsor, the investigator, the IEC, and the
regulator. Until recently there were no rules spelling out these requirements.

Trial sponsors call the shots

It is not surprising that sponsors of clinical trials drafted policies on compensation for injury to their own benefit.
Monitoring bodies such as ethics review committees have not insisted on participants’ right to compensation for
injury or death. A study interviewing investigators, members of ethics committees, and sponsors, and reviewing
informed consent documents, found that “47% of investigators were either unaware of, or had not understood, the
legal requirements and depended on sponsors to manage these issues.” One in four (26%) ethics committee
members were unaware that compensation was required. 40% of investigators, 30% of ethics review committees
and all the sponsors interviewed reported that their institutions had policies on compensation. However, these
policies were almost exclusively for provision of limited medical care, or reimbursement of medical expenses.
These policies did not cover compensation for time and wages lost, injury or death, though this was required by
law (Thatte et al 2009)iv.

Increasing reports of unethical trials

The consequences of this lax regulatory framework soon became apparent. Reports emerged of unethical
recruitment practices and unethical research: both government and private doctors, motivated by incentives from
drug companies, recruiting patients for drug trials without their voluntary, informed consent; research ethics review
committees approving unethical trial designs that put participants at risk of harm; particularly vulnerable groups
being recruited without scientific justification for their inclusion, and without ensuring that they were able to
consent; and poor patients entering trials on their physician’s advice, to obtain free treatment or better treatment
(Srinivasan 2009). Reports, many of them based on investigations by advocacy organisations, also emerged of
patients suffering injuries or dying in trials. It is not an accident that participants in clinical trials come from
particularly vulnerable and marginalised sections of society. A few examples are cited below.

In August 2008, it was learned that 4,142 paediatric patients in the government’s All India Institute of Medical
Sciences (AIIMS) – a hospital used mostly by poor patients -- had been part of clinical trialsv and 49 children died
in these trials. The government disclosed this in reply to an application by Rahul Verma of Uday Foundation under
the Right to Information (RTI) Act. An AIIMS representative is quoted as saying: "All the deaths can't be attributed
to the trials. Some patients were suffering from conditions in which mortality was the normal outcome and we were
trying to see whether a drug could improve the situation (Sinha 2008). According to the RTI reply, all deaths in
clinical trials in the hospital were discussed in review meetings and data safety monitoring board meetings, notified
to the trial sponsor, and reported in publications (AIIMS 2008). However, the government did not reveal the details
of any investigation into these deaths: whether they had in fact been investigated and what the conclusions were.
Though the government announced an inquiry in response to the public outcry following news reports, the results
do not seem to have been made public.

In 2010, it was learned that over 25,000 girls had been administered a vaccine against Human Papilloma Virus
(HPV) infection, in what a Parliamentary Committee report confirmed was a clinical trial (GoI 2013) vi. The trial
was conducted by the international NGO PATH in collaboration with government organisations, in tribal regions of
Gujarat and Andhra Pradesh, using vaccines provided by Merck, Sharp & Dohme and Glaxo SmithKline and with
financial support of the Bill and Melinda Gates Foundation. At least seven girls who had been administered the
vaccine died. A report published by the advocacy group SAMA Resource Group for Women and Health identified
serious malpractices at many levels: government organisations collaborated with an international NGO to further
the interests of private pharmaceutical companies; minors in marginalised communities were targeted for the trials;
the informed consent documents were misleading and signatures on the forms were forged; there was no proper
follow-up of the girls who had been vaccinated; and injuries and deaths following immunisation were not
investigated, nor medical treatment provided, and so on (Sarojini et al 2010).

In 2010, it became known that the Bhopal Memorial Hospital & Research Centre (BMHRC), set up specifically to
provide free treatment to survivors of the 1984 gas leak, had conducted drug trials for foreign pharmaceutical
companies vii between 2004 and 2008. 215 of the 279 trial participants were gas survivors, though none of the
drugs tested were for conditions specific to gas-affected people. At least 14 died.

Participants (or the next of kin of those who had died) traced by the advocacy group Bhopal Group for Information
and Action (BGIA) were unaware that they had been part of a drug trial (Lakhani 2011). Even otherwise, as people
dependent on free treatment, the gas survivors would have found it very difficult to refuse to be in the trial. The
critically ill would have found it even harder to refuse. For example, patients in the trial of Organon/ Sanofi’s –blood
thinner Fondaparinux were recruited even as they were experiencing a heart attackviii. Moreover, at least one of
the trials violated the rules in place at the time: the study of Fondaparinux received DCGI approval in July 2004
and started recruiting patients in September 2004, four months before the law permitted multinational
pharmaceutical trials in India.

The campaign against unethical trials

In 2012, the Indore-based organisation Swasthya Adhikar Manch and Anand Rai filed separate cases in the
Supreme Court of India, calling for an investigation of clinical trials in India and appropriate action against
unethical and illegal trials. They asked for information on the number and type of trials being conducted in India,
the number of people who had been recruited into these trials, the number of injuries and deaths, and the medical
treatment and compensation given (WP [civil] 33/2012 and WP [civil] 79/2012). The Supreme Court clubbed the
two cases along with an intervention by groups in Bhopal. Public interest litigation was also filed regarding the
unethical trials of the HPV vaccine (Kalpana Mehta and others in WP [civil] 558/2012 and SAMA and others in WP
[civil] 921/2013).

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 2/8
11/29/2019 Compensation for injury in drug trials in India
Government reports submitted to the Supreme Court and Parliament confirmed that regulatory authorities were
grossly derelict in their duty to monitor clinical trials and protect trial participants. The pharmaceutical industry, not
the government, decided whether each serious adverse effect (SAEix), including death, was related to the drug,
whether to pay a compensation and how much. Just 3% of deaths reported in clinical trials were attributed to the
drug (Jesani 2013). Compensation amounts -- when paid -- varied according to the sponsor and were as low as
Rs 50,000 (GoI 2014).There is no record of money paid to people who suffered “non-death” SAEs in a trial (Jesani
2013).

Finally, rules for the conduct of clinical trials

The PILs, along with investigations by advocacy groups, media reports and questions in parliament, have been
instrumental in strengthening the regulatory framework for clinical trials in India. The new regulations (CDSCO
2014a) on the conduct of drug trials in India include those for registration of clinical trials; registration, composition
and functioning of ethics review committees which reviewed and approved trials; registration of clinical research
organisations; standard operating practices for inspection of clinical trial sites, audiovisual recording of informed
consent, and so on.

In July 2014, the CDSCO announced a proposed online database with comprehensive information on each clinical
trial, to be uploaded before applying for permission to conduct the trial, and updated daily by sponsors,
researchers and IECs. This would include details of the drug being tested, the informed consent documents, IEC
decisions, all participants, any SAEs, their investigation and compensation claimed and paid, if applicable
(CDSCO 2014b). It is not clear how much of this information, if any, would be in the public domain. One major
demand of advocacy organisations has been for the government to make such details public.

Rules on compensation for trial-related injury

In January 2013, the CDSCO introduced draft rules on compensation for serious adverse events including death in
clinical trials. These rules represent a major advance from regulations elsewhere in the world. The sponsor of the
trial is required to pay compensation, and not only for SAEs caused by the investigational drug, but also for those
that occurred due to misconduct by the investigator or sponsor; failure of investigational drug to provide the
intended therapeutic effect; use of placebo in a placebo-controlled trial; other medication (other than standard
care) necessitated by the trial protocol; injury to the child in-utero, and any clinical trial procedures in the study.
That is, compensation would not be limited to cases in which the SAEs were attributable to the experimental drug
alone; it would be awarded in all cases in which the SAE was attributable to trial participation itself. The new rules
also required that the trial sponsors provide free medical management for all injuries or illnesses suffered in these
trials, whether or not they were related to the experimental drug, for as long as is required.

The finalised rules (CDSCO 2014c) were amended apparently because of pressure from the pharmaceutical
industry: compensation for failure of the investigational drug, or due to the use of a placebo, can be granted only if
standard treatment was available but not provided. Moreover, free medical care is to be provided only until it is
established whether the injury is, or is not, related to the clinical trial x.

Timelines have been laid down for all steps in the reporting and review of SAEs, decisions on compensation, and
its disbursal. The trial’s principal investigator (PI) is required to report SAEs, in a specified format, to the Drugs
Controller General of India (DCGI) of the CDSCO, the institutional ethics committee (IEC) which reviewed the
protocol and monitors the trial, and the trial sponsor; the sponsor is required to send a report to the DCGI. The IEC
is required to review the PI’s report and submit its assessment to an expert committee, appointed in 2013 to
review all cases of death in clinical trials. The expert committee receives the reports of the PI, the IEC and the
sponsor to make a final assessment on whether or not the SAE is related to the trial, and sends it to the DCGI.
The DCGI informs the trial sponsor if compensation is to be paid, and if so how much. The sponsor must make the
payment within a given time. Formulae have also been issued for calculating the quantum of compensation for
death and for serious injury but have met with widespread criticism (Ramanathan et al 2013; Ambhore et al 2014)

Need more than just a law

However, it will take more than such rules to ensure that all people injured in drug trials are even awarded this
compensation, let alone receive it. There is a need to improve the conduct of trials themselves, their review and
monitoring in order for the new rules to have a significant impact. Regulatory mechanisms must also compensate
for the enormous disparity in the situations of sponsors and participants – their differential access to money,
information, legal representation and influential ‘contacts’. The clinical trials industry is powerful and will resist
regulation. The trial participant at the centre of the industry is invisible and has no voice. This situation, along with
the inherent complexity of ascribing medical causality, may make it very difficult for the participants to obtain
compensation even under the new rules.

Some of these problems are discussed using CDSCO’s inspection reports of three trials in Bhopal, obtained by
BGIA; the report of a Parliamentary Committee on the HPV vaccine trials, and interviews with expertsxi. These
reports followed exposes by advocacy organisations. The malpractices described in these reports will be just the
tip of the iceberg. The workings of the clinical trials industry remain opaque, shielded by the government in the
name of protecting trade secrets (Sengupta et al 2011) and participants’ confidentiality.

Shoddy research practices

Incomplete and substandard documentation, poor follow-up and delayed reporting of SAEs were among the major
irregularities identified by all the experts interviewed. These were also apparent in the Bhopal and HPV trials.

The investigating team of the trial is supposed to maintain complete documentation of all aspects of the study,
monitor all subjects on a regular basis and document these details, follow up outpatient as well as in-patient
participants when they are discharged, and ensure that they can contact the investigator at all times of the day and
night if they fall ill, or have any questions. The trial sponsor is responsible for ensuring that the investigating team
complies with these requirements, by scrutinising the data submitted and conducting site visits.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 3/8
11/29/2019 Compensation for injury in drug trials in India
However, PIs may be reluctant to report serious adverse events as they could be due to negligence. Moreover,
since serious adverse event reports related to the investigational drug reduce its value to the sponsor, PIs may
view such reporting as contrary to the interests of the sponsor.

Another reason for these delays is that out-patient trial subjects living far from the trial centre will find it difficult to
travel to the trial centre for treatment, especially if their conveyance expenses are not compensated forxii.
Treatment given at small health centres is likely to be poorly recorded, and the details will probably not be reported
to the trial centre. Researchers who do not follow up with their subjects (or do not provide them with telephone
numbers where they can be reached at all times of the day) will not learn about their illness or death until the next
visit is due -- if in fact another visit is due. By the time they do get this information, if the subject has died, the body
could have been cremated without a post mortem. Researchers who are not meticulous in following up trial
participants may also just lose all contact with those who have moved to another town or city. Injuries or deaths of
such subjects will not even be recorded in the trial data, resulting in an under-estimation of the number of SAEs in
the trial.

In the HPV vaccine trial, researchers actually chose not to follow up the girls once they administered the vaccine.
They relied, instead, on a routine surveillance – known to be grossly inadequate – for reports of any injuries or
deaths, thereby violating the legal obligation to report all adverse events and deaths in a clinical trial within a
specified period (GoI 2013).

The Parliamentary Committee report on the HPV vaccine trial noted that the deaths were reported to the
authorities much later than required by government guidelines, “action on investigations into the causes of deaths
took an unacceptably long time” and there were many “discrepancies and gaps in the investigations of the deaths”
(GoI 2013: para 6.19). The medical records were inadequate, and the staff were not prepared to deal with the
SAEs. The committee concluded that the possibility that the deaths were related to the vaccine could not be
completely ruled out “mainly because the alternate cause of death as listed can not be fully substantiated on the
basis of medical records in all the cases.” (GoI 2013: 38). The committee also questioned the accuracy of the SAE
reporting; there may have been more deaths in the trial that were not reported.

The CDSCO inspections of trials in the BMHRC hospital in Bhopal found delayed reporting in all three trials
(CDSCO 2010, 2011a, 2011b). In the trial of Wyeth’s antibiotic Tigecycline (conducted on hospitalised patients with
serious infections), SAE reports were delayed by between 40 days and five months. Three deaths were recorded
in the Wyeth trial, but there was no follow-up of a patient who suffered an SAE and sought a discharge against
medical advice. So “no information was available to the inspection team for the current status with respect to
condition of health of the above mentioned subject”. In the trial of Theravance’s antibiotic Telavancin, one of the
deaths was reported to the IEC more than eight months after it occurred. In the trial of Organon/Sanofi’s
Fondaparinux, in which six people died, at least one SAE was reported three weeks after it occurred.

Negligent ethics review committees

Schedule Y requires the IEC to review and approve the clinical trial protocol and monitor the conduct of the trial to
protect the health and rights of trial subjects. The IEC has the power to halt a trial if it is felt necessary (GoI 2005)
and is empowered to pay site visits (ICMR 2006). Starting in 2014, the IEC is also charged with assessing the
SAE reports submitted by the PI and forwarding this assessment to the EC and the DCGI, and with awarding
compensation for non-death SAEs.

It is well known that many members of IECs are investigators in industry-funded research and will have conflicts of
interest when reviewing trials funded by the same company -- or even of competitor companies. It is also known
that such conflicts of interest are not always disclosed (Campbell et al 2006). IEC members from within the
institution may tend to give in to the views of senior colleagues and it is not uncommon for the director of the
institution to be a member of the IEC.

The CDSCO inspection teams in the Bhopal trials (CDSCO 2010, 2011a, 2011b) found that none of the IECs
followed standard practices on membership, terms of reference, review and decision-making procedures, review
of SAEs, etc. The IEC minutes in the Fondaparinux trial indicated that the committee approved the protocol
“without proper scrutiny of safety aspects”. And though IECs are required to meet after each SAE is reported, to
consider whether the trial should be halted, there was no record of the IECs in the Fondaparinux and Tigecycline
trials having reviewed the trial after deaths were reported.

The IEC reviewing the Fondaparinux trial would have been aware that the secretary of the IEC is married to the PI.
Yet it approved an informed consent form that carries the PI’s telephone number to report injuries, and the IEC
secretary’s telephone number for information on the participant’s rights: aggrieved participants would have to
complain to the PI’s spouse. This obvious ethical violation seems to have been missed by the IEC.

There were two ethics committees charged with monitoring the HPV vaccine trials. The Parliamentary Committee
report found both to be grossly negligent, noting that they “existed only as a formality and they did not play the role
they were designated for” (GoI 2013: para 6.24). Though ethics committees are required to meet and monitor the
progress of the trials and review SAE reports, “Only after reports of deaths appeared in the media, the meetings of
these committees were held... This is a clear dereliction of duty on the part of the Ethics Committees.” (GoI 2013:
para 6.23).

Regulators who do a whitewash job

Such serious lapses should lead regulators to at least question trial sponsors’ conclusions as to whether the death
of a trial subject is related to the trial. However, when inspections are motivated entirely by public pressure, as was
the case in the Bhopal and the HPV vaccine trials, the report may identify numerous violations only to absolve the
investigator and others responsible.

Indeed, the CDSCO inspections in Bhopal identified lapses in all three trials, including violations by the PIs (such
as late reporting, improper documentation and “improper” causality assessments) that could have affected the
proper assessment of the SAEs reported. Despite noting these lapses, the investigators’ assessments – that none

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 4/8
11/29/2019 Compensation for injury in drug trials in India
of the deaths were related to the trials – were accepted. At the very least, the inspection teams should have
demanded a re-investigation into the SAEs. Interestingly, the reports contain many identical passages, in some
cases contradicting other portions of the report, evidence of a careless copy-paste job. Clearly the inspections
were a hurriedly conducted whitewash.

In the case of the HPV vaccine trials, the Parliamentary Committee report is a scathing criticism of the role of
government institutions at every level: the state departments of health, the Indian Council for Medical Research,
the DCGI, and even the government’s inquiry committee, whose members identified many ethical and legal
violations but absolved all parties of responsibility. The Parliamentary Committee report also notes that at least two
members participated in the inquiry without reporting major conflicts of interest (GoI 2013).

Difficulties assessing ‘relatedness’ of the injury

Starting in 2013, the government’s expert committee scrutinises each SAE report to assess its “relatedness” to the
trial. But the assessment method is acknowledged to be imperfect and in many cases, opinions can differ on
whether the death was related to the trial. In such situations, there is scope for decisions to be biased against the
participant.

It was in the course of the SC case (33/2012) that the government disclosed that 3,458 people had died in clinical
trials between January 2005 and December 2012, as reported by drug companies. Only 89 of these deaths were
attributed to the trial, according to reports submitted by the companies. Another 14,320 “non-death SAEs” were
reported, of which 506 were attributed to the trials (Legal correspondent 2014).

A detailed review of the 679 deaths reported in 2010 found that only 25 were attributed to the drug. 92 were of
‘indeterminate causality’ – ‘possible’, ‘probable’, ‘unlikely’, ‘implied’, ‘suspected’, ‘suspected to standard therapy’,
‘dubiously suspected’, ‘not suspected’, and ‘doubtful’. In an additional 30 deaths, there was no information on
causality (Jesani 2013).

No explanation was given of how the sponsors made any of these assessments, why they decided that deaths
with ‘indeterminate causality’ should not be compensated, and why they had not conducted causality assessments
for 30 deaths. Obviously sponsors were not required to justify their assessments to the regulators.

The collective wisdom of experts

It is only in 2013 that the CDSCO started conducting its own causality assessment of deaths in clinical trials. In
December 2014, it was learned that the expert committeehad examined 220 of the 370 deaths reported by trial
investigators and their sponsors from January 2013. Compensation had been awarded in 21cases, based on a
formula worked out by the government (Pulla 2014). Nowhere is it mentioned how the committee made its
causality assessment.

At present, Schedule Y does not specify the method by which causality assessments are made. An assessment
protocol is reportedly in the process of being finalised. The expert committee currently uses a method that has
been described as “global introspection” or “the application of collective wisdom of experts”xiii.

Each SAE report along with all documents submitted by the PI, and the assessment made by the IEC, is
scrutinised by a team from the panel of experts on the expert committee. This team usually (but not always) has a
clinical pharmacologist and a subject expert (for example, in a trial of a cardiac drug, a cardiologist). It may call on
another expert for an opinion, and the PI of the trial if needed. The team considers questions such as: is there a
temporal relation between the drug and the SAE? Is the drug known to cause this SAE? Is the drug’s mechanism
such that it might have caused the SAE? Are there other causes of this SAE? Could it be due to the natural history
of the illness? Could it be a drug-drug interaction? The assessment process does not consider non-medical
causes, so, for example, it would not dismiss the case of a woman in a contraceptive trial who suffers domestic
violence because her husband learns of her contraceptive use, though the injury itself might be reported as an
adverse event.

The team must decide if an injury is ‘related’ or ‘not related’; it may not use any other categories, even if it is
unable to be completely sure if the death is – or is not – related to the trial. It must arrive at a decision by
consensus.

No place for the trial participant

Neither the expert committee nor the IEC hears from trial subjects or their relatives. They will certainly have
important information that the PI was not aware of or did not supply. They could have information about the injury
or death itself or about possible negligence by the investigator or sponsor. While the IEC may call the participants
or relatives if it is felt necessary, this is not required. The participant, who is at the centre of the clinical trials
industry, who personally accepts the risks of research that is meant to generate knowledge beneficial for others, is
invisible in the entire process.

Johari and others have written a detailed critique of the draft rules for compensation in which they have called for
all SAEs to be investigated by an independent expert committee. This committee would, in addition to the
documents provided by the investigator and sponsor, hear from participants as well as make its own inquiries.
They have also criticised the narrow definition of SAE as physical harm that occurs during the trial, and called for a
comprehensive understanding that includes psychological harm, social harm, loss of livelihood, and harm that
becomes evident much after the trial ends xiv (Johari et al 2014).

Consensus built by influence

All the experts interviewed asserted that causality or relatedness assessment is a complex, technical issue. Very
often, it is not possible to conclude with certainty whether or not a participant’s death was due to the trial. However,
the expert committee does not have guidelines on how to make decisions in such cases.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 5/8
11/29/2019 Compensation for injury in drug trials in India
Each decision in a difficult case will be influenced by the views of the experts reviewing that case. Experts can
have conflicts of interest if they are also investigators for industry-sponsored trials. Their judgment is likely to be
influenced by their views on the question: does the injured participant deserve the benefit of doubt? Some experts
hold that unless one can say with certainty that the injury was not caused by the trial, it should be treated as
caused.

If there is an unresolvable disagreement within the team of experts, it would be correct to keep the case open until
the team receives additional information or expert opinion that allows it to make a final decision. However, this is,
in the end, a matter of judgment, and some experts concede that the judgment of the senior-most expert could
prevail.

Experts say that their most common problem is that PIs submit insufficient information. In such a situation, an
expert says, “The EC is at liberty to call for more information and to keep the case on hold until it is satisfied.”
What if the EC does not call for this information? What if more information is just not available? Should the EC
make an assessment nevertheless? There are no explicit guidelines for these situations.

In such situations, it would be unfair to rule against the participant because the researcher and sponsor have not
done their job. “In a clinical trial, documentation is a key prerequisite,” says an expert. “Subjects are supposed to
be closely followed up. If this has not happened, it is a failure of the PI and the sponsor.”

Surely, when there is not enough information to decide on relatedness, the benefit of doubt should go to their next
of kin.

“My personal view is that the subject should be entitled to compensation as soon as screening (for eligibility, such
as a stress test) starts,” says one expert. “If the patient’s death could have been delayed if s/he had not
considered participation in the trial, if s/he agreed to participate and his/her death was preponed, s/he should be
entitled to compensation.” However, this view may not be shared by all the experts assessing a particular death

In conclusion, there are times when it is just not possible to state with certainty if a person who died in a clinical
trial died due to participation in the trial. As there is no well-defined protocol for causality assessment, with
instructions on how to resolve differences of opinion, or to decide with insufficient information, these decisions will
depend on the views of individual experts, and the assertiveness or persuasiveness of the experts on the team.
The deliberations of the EC are not made public.

A partial victory

The new rules on compensation for injury and death could be viewed as a victory for those who have been
campaigning against unethical practices in the clinical trials industry. However, in the present circumstances, it is a
very partial and conditional victory. The acid test is whether they help protect trial participants and deliver justice to
those who have suffered. In fact, many trial participants or their families who deserve compensation for cases of
death or injury will not receive compensation, for reasons spelled out above.

This commentary highlights some problems in the process leading up to establishing a trial subject’s right to
compensation: the quality of documentation in a trial, reporting of injuries and deaths, functioning of monitoring
mechanisms and difficulties in assessing causality.

The current discussion must continue to be located in the issues raised by advocacy organisations: the unethical
and illegal practices of many trial sponsors which provoked the campaigns for regulation and transparency in
clinical trials, and the wide disparity in resources and access to the law between the trial sponsors and the
participants. This disparity can only be reduced if the State takes an active regulatory role in the entire process. In
the case of injury or death, it must use a definition of injury that goes beyond immediate physical harm. It must
also establish clear guidelines for causality assessment that include the views of trial participants and their
families, and give the benefit of doubt to trial participants and their families, rather than to trial sponsors. It is
essential that all aspects of this process be made public. Unfortunately, the actual record of State and
administrative conduct, discussed above, does not inspire much confidence.

Acknowledgements: This comment is based on work funded by Wemos Foundation, Amsterdam, The
Netherlands. I thank Rachna Dhingra and Hazra Bee of the Bhopal Group for Information and Action for
making available documents obtained related to the trials in Bhopal.

References

All India Institute of Medical Sciences (2008). Department of Paediatrics. Information under Right to Information
Act 2005. Application of Shri Rahul Verma. Received in MS Office on 21.06. 2008 vide Diary No 3466.

Ambhore, V, Sarojini, N, Srinivasan, S, Jesani, A and V Johari (2014). Comments and suggestions on the draft
formula to determine quantum of compensation in case of clinical trial related injury (other than death). 16 May.

Campbell, EG, Weissman, JS, Vogeli, C et al (2006). Financial relationships between institutional review board
members and industry. New England Journal of Medicine. 355: 2321-9.

Central Drug Standards Control Organisation (2001). Good Clinical Practice for Clinical Research in India.
Viewed on 5 March 2015 (http://cdsco.nic.in/html/GCP.htm )

Central Drug Standards Control Organisation (2010). DCGI letter regarding Protocol Title “A Phase 3
Randomized Double Blind Parallel Group Multinational Trial of

Intravenous Telavancin Versus Vancomycin for Treatment of Hospital Acquired Pneumonia with a Focus on
Patients with Infections Due to Methicilin Resistant Staphoyloccus aureus” at Bhopal Memorial Hospital and
Research Centre. 28 September.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 6/8
11/29/2019 Compensation for injury in drug trials in India
Central Drug Standards Control Organisation (2011a). Inspection Report. Clinical Trial Site: Bhopal Memorial
Hospital and Research Centre. Protocol Title: “A Multicenter, Open-Label, Randomized Comparative study of
Tigecycline vs Ceftriaxone Sodium Plus Metronidazonle for the treatment of Hospitalized Subjects with
Complicated Intraabdominal infection.”

Central Drug Standards Control Organisation (2011b). Inspection Report. Clinical Trial Site: Bhopal Memorial
Hospital and Research Centre. Protocol Title: “An international randomized study evaluating the efficacy and
safety of Fondaparinux sodium versus control therapy and Glucose-insulin-potassium infusion versus control in a
broad range of patients with ST segment elevation acute myocardial infarction.”

Central Drug Standards Control Organisation (2014a). Major achievements of CDSCO (January, 2012 to
February, 2014) viewed on 5 March 2015 (
http://www.cdsco.nic.in/writereaddata/Majorachievements_CDSCO_01032014_-_final.pdf )

Central Drug Standards Control Organisation (2014b). Notice dated 28 July 2014. Subject: Stakeholders
suggestions/comments on Clinical Trials conductance in India. Viewed on 5 March 2015 (

http://www.cdsco.nic.in/writereaddata/PROPOSAL%20FOR%20CREATION%20OF%20IT%20ENABLED%20SYSTEM
)

Central Drug Standards Control Organisation (2014c). Rule 122DAB in the Drugs and Cosmetics Rules, 2005.
Given in Gazette Notifications GSR 53E (2013 Jan 30), GSR 292E (2014 Apr 24), GSR 889E (2014 Dec 12).
Viewed on 5 March 2015 (http://cdsco.nic.in )

Government of India (2005). Ministry of health and family welfare. The Drugs and Cosmetics Act and Rules. As
amended up to the 30th of June 2005. GoI: New Delhi. Viewed on 5 March 2015
http://cdsco.nic.in/writereaddata/Drugs&CosmeticAct.pdf

Government of India (2012). Department Related Parliamentary Standing Committee on Health and Family
Welfare. 59th Report on the functioning of the Central Drugs Standard Control Organisation (CDSCO). New Delhi:
Rajya Sabha Secretariat.

Government of India (2013). Department Related Parliamentary Standing Committee on Health and Family
Welfare. 72nd report on the alleged irregularities in the conduct of studies using Human Papilloma Virus (HPV)
Vaccine by PATH in India (Department of Health Research, Ministry of Health and Family Welfare). GoI: New
Delhi.

Government of India (2014). Ministry of Health and Family Welfare. Press Information Bureau. Compensation for
Clinical Trial Subjects. (viewed on 6 March 2015 http://pib.nic.in/newsite/PrintRelease.aspx?relid=104153 )

Indian Council of Medical Research (2006). Ethical guidelines for biomedical research on human participants.
(New Delhi: ICMR) viewed on 5 March 2015 ( http://icmr.nic.in/ethical_guidelines.pdf )

Jesani, A (2013). New regulations on compensation for injury and death in drug trials. Indian Journal of Medical
Ethics. 10: 76-9.

Johari, V, Jesani, A, Srinivasan, S and Sarojini N (2014). Letter to Shri Arun K Panda re: comments on the draft
of the Drugs and Cosmetics (Third Amendment) Rules, 2014 to amend Rule 122DAB at GSR 292 (E), Gazetted
Notification dated April 24, 2014, Ministry of Health and Family Welfare.18 June.

Lakhani, N (2011). From tragedy to travesty: drugs tested on survivors of Bhopal. The Independent. 15 November.
Viewed on 6 March 2015. (http://www.independent.co.uk/news/world/asia/from-tragedy-to-travesty-drugs-tested-
on-survivors-of-bhopal-6262412.html)

Legal Correspondent (2014). SC shocked at clinical trial deaths. The Telegraph. 11 March. Viewed on 6 March
2015 ( http://www.telegraphindia.com/1140311/jsp/nation/story_18067977.jsp#.VNNDOZ2UeQw )

Nikarge, S (2010). Clinical Trials Watch. Indian Journal of Medical Ethics. 7: 259-62.

Pulla, P (2014).Campaigners criticise India's new system for compensating clinical trial deaths. BMJ. 29
December. Viewed on 6 March 2015 (http://www.bmj.com/content/349/bmj.g7806 )

Ramanathan, M, Sankara Sarma P and U S Mishra (2012). Compensation for trial related injury: does simplicity
compromise fairness? Indian Journal of Medical Ethics. 9: 232-5

Sarojini NB, Anjali, S and S Ashalata (2010): “Preliminary Findings of Visit to Bhadrachalam: HPV Vaccine
‘Demonstration Project’ Site in Andhra Pradesh” (New Delhi: SAMA).

Sengupta, A (2009). Fatal trials: clinical trials are killing people. Indian Journal of Medical Ethics. 6: 118-9.

Sengupta, A, Shenoi, A, Sarojini, NB and Madhavi Y (2011). Human Papillomma Vaccine Trials in India. The
Lancet. 9767: 719.

Sinha, K (2008). 49 babies die during clinical trials at AIIMS. The Times of India. 18 August. Viewed on 6 March
2015 (http://timesofindia.indiatimes.com/india/49-babies-die-during-clinical-trials-at-
AIIMS/pmredirectshow/3374492.cms?curpg=1 )

Srinivasan, S (2009). Background of clinical trials in India. Economic & Political Weekly. 29 August: 29-33.

Thatte, UM, Kulkarni-Munshi R and SA Kalekar (2009). Review of policies for injuries to research participants in
India. Journal of Medical Ethics. 35: 133-9.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 7/8
11/29/2019 Compensation for injury in drug trials in India
World Medical Association (2013). Declaration of Helsinki. Ethical principles for medical research involving
human subjects. Adopted by the WMA General Assembly, Helsinki, Finland, June 1964 and last amended at the
64th WMA General Assembly, Fortaleza, Brazil, October 2013. Viewed on 6 March 2015
(http://www.wma.net/en/30publications/10policies/b3/ )

i Phase I trials collect information on the drug, including its safety and adverse reactions. They are usually
conducted on a small number of healthy volunteers. Phase II trials evaluate the effectiveness and safety of a drug
on a small number of patients. Phase III trials are conducted on larger numbers of people to confirm the evidence
from earlier phase trials towards obtaining marketing approval of the drug. Until January 2005, India required a
‘phase lag’: phase 2 trials could be conducted in India only after the phase 3 trial had been conducted abroad.
This ruled out India as a site of a multi-country trial. Multi-country trials of the same phase were permitted only for
drugs with special relevance to India, such as, for example, new antibiotics for tuberculosis. In January 2005, the
law was changed to permit trials of drugs being developed abroad to be tested in the same phase in India as they
were abroad.

ii Compensation for research-related injury is separate from punitive compensation for the harm done because of
illegal or unethical practices.

iii Unfortunately, this right is not recognised universally. In some countries, injured participants receive
compensation without having to prove that the injury was caused by the trial. On the other hand, in the US, trial
sponsors are not required to pay compensation for injury; they are not even required to provide medical treatment.

iv The sponsor, not the regulatory authorities, also decided on whether compensation should be paid, if one goes
by the informed consent form for Organon/Sanofi’s trial of the drug Fondaparinux in Bhopal: “In the event that
injury occurs as a direct result of administration of the investigational drug under study, necessary medical care will
be available. However, neither free medical treatment nor any financial compensation for any illness or injury will
be provided unless the study sponsor, Organon, in its discretion, approves payment for such medical care.”

v The top sponsors were the ICMR, DBT, WHO, AIIMS and Johns Hopkins. The drugs tested included
Rituximab, Olmesartan, Valsartan provided by Novartis, Gene activated human glucocerebrosidase from Shire
Human Genetics Therapies, USA.

vi The Parliamentary Committee report stated: “the demonstration project is a study of a pharmaceutical product
carried out on humans and since the primary objectives include the study of serious adverse effects, it is clear that
clinical trial rules and guidelines should apply” (GoI: 2.3).

vii Astra Zeneca, Wyeth, Pfizer, Theravance, Schering Plough, GSK and Sanofi (earlier Organon), in the
departments of cardiology, pulmonary medicine, gastrointestinal surgery, anaesthesia and psychiatry. Many of
these were conducted by the contract research organisation Quintiles.

viii The informed consent form of the Fondaparinux trial states: “You are being asked to participate in this trial
because you are experiencing clinical signs and symptoms of a heart attack.”

ix The CDSCO defines a Serious Adverse Event (SAE) as any untoward medical occurrence that results in
death, or requires inpatient hospitalisation or prolongation of existing hospitalisation or results in persistent or
significant disability/incapacity, or is a congenital anomaly/birth defect.

x The rule as finalised in December 2014 is not clear: “free medical management shall be given as long as
required or till such time it is established that the injury is not related to the clinical trial, whichever is earlier;”. This
does not indicate that treatment will be provided free even once it is established that the injury is related to the
trial.

xi The following documents provided by activists in Bhopal are referred to: CDSCO’s inspection reports of two
trials in Bhopal: Tigecylcline (Wyeth Pharmaceuticals) and Fondaparinux (Organon later transferred to Sanofi
Synthelabo and then GSK Ltd); the CDSCO’s warning letter for a trial in Bhopal on Telavancin (TeravanceInc); and
an informed consent document template for the Fondaparinux trial. Interviews were also conducted with various
experts including some members of the newly formed expert committee entrusted with assessing SAEs.

xii Both Indian GCP and the ICMR guidelines state that participants must be compensated for travel and related
expenses. None of the three trials in Bhopal inspected by the DCGI compensated participants for travel and
related expenses.

xiii There are alternatives to the expert opinion method, such as the causality assessment system developed by
the WHO-Upsalla Monitoring Centre or the Naranjo Scale, both of which have been discussed by CDSCO experts
for modification. Both scales can help arrive at judgments of ‘certain’, ‘probable’, ‘possible’ and ‘unlikely’. They
would have to be converted into a ‘binary’ system to arrive at a yes or a no, or ‘related’ or ‘unrelated’, in order to
make decisions on compensation. It is generally acknowledged that none of the algorithms and scales used for
causality or relatedness assessment is perfect.

xiv Both the GCP and the ICMR guidelines include mental or psychological harm within the definition of injury for
which compensation must be paid, but the CDSCO’s definition of SAEeligible for compensation does not mention
mental harm.

Infochange News & Features, August 2015

About Us Disclaimer Contact Us Top

INFOCHANGE INDIA © 2012 InfoChange India News & Features development news India
Developed By Tekdi Web Solutions.

https://infochangeindia.org/public-health/112-public-health/analysis/9467-compensation-for-injury-in-drug-trials-in-india 8/8

You might also like