throbber
Science & Society
`
`The ethics of global clinical trials
`
`In developing countries, participation in clinical trials is sometimes the only way to access medical
`treatment. What should be done to avoid exploitation of disadvantaged populations?
`
`Katrin Weigmann
`
`C linical research by academic institu-
`
`tions and pharmaceutical companies
`has followed the general
`trend of
`globalization and has moved inexorably
`towards low- and middle-income countries
`(LMIC). This
`trend has
`raised various
`concerns,
`including whether the research
`being conducted is of value to public health
`in these countries or whether economically
`disadvantaged
`populations
`are
`being
`exploited for the benefit of patients in rich
`countries. Nevertheless, clinical trials and
`the research and health care that accompany
`them can directly benefit patients, in particu-
`lar those who would otherwise have no or
`only little access to health care services. It is
`therefore a matter of striking a fine-tuned
`balance between the economic and research
`interests of pharmaceutical companies and
`academia and the needs of patients in LMIC
`to make sure that all sides benefit.
`......................................................
`“. . . clinical research also helps
`to build research and health
`care capacity and can improve
`local infrastructure and boost
`the economy”
`......................................................
`
`Clinical trials are a necessary step in drug
`development and are conducted throughout
`the world, both in developed and in develop-
`ing countries. Trials themselves are thus
`not per se immoral, and there are a variety
`of reasons to conduct responsible clinical
`trials in LMIC. Doing so, for example,
`is
`often the only way to test drugs and
`vaccines for diseases that predominantly
`afflict people in these countries; trying to
`test
`the safety and efficacy of a malaria
`
`vaccine in Europe or North America would
`be relatively futile given the lack of patients.
`Beyond the obvious and direct public health
`benefits—in terms of both new knowledge
`and new treatments—clinical research also
`helps to build research and health care
`capacity and can improve local infrastructure
`and boost
`the economy.
`In fact, many
`developing countries have been actively
`trying to attract clinical research for these
`reasons.
`From the perspective of those conducting
`the trials, a major benefit of LMIC is that it
`is easy to enrol patients who are willing to
`participate, particularly if they are poor. For
`many of these patients, clinical trials are
`their only access to medical care. However,
`it is precisely this unmet medical need that
`poses a risk for exploitation. “You want to
`make sure that you don’t take advantage of
`deprivation and that you don’t see the world
`as a convenient population of sick people
`where you can go and get the data that you
`need to make more robust health systems in
`high income countries”, commented Alex
`John London, Professor of Philosophy and
`Director of The Center for Ethics and Policy
`at Carnegie Mellon University in the USA.
`Indeed, human subjects research in LMIC
`has often been criticized for exporting the
`risk of research to those who will, in the
`end, not be able to afford the resulting medi-
`cal products. Clinical trials can pose real
`health risks to participants, as the safety and
`side effects of the drugs or vaccines being
`tested are not yet fully known.
`
`T he history of clinical trials and their
`
`globalization can be
`subsequent
`to
`the
`thalidomide
`traced back
`scandal
`in Germany in the early 1960s.
`Many children were born with severe
`
`deformations of their extremities; it subse-
`quently became clear that
`thalidomide, a
`sedative developed by the German company
`Gru¨ nenthal, caused birth defects in babies
`whose mothers had been taking the drug
`during pregnancy. Public outrage over the
`devastating effects of the drug and the fact
`that it had not been sufficiently tested for
`safety fuelled discussion within the US Food
`and Drug Administration (FDA) and quickly
`led to legislation to improve the safety test-
`ing of new drugs. The so-called Kefauver-
`Harris Amendments were passed to prevent
`another thalidomide disaster and—although
`not directly related to the drug scandal—
`introduced the requirement for drug manu-
`facturers to prove drug effectiveness in
`addition to safety.
`......................................................
`“From the perspective of those
`conducting the trials, a major
`benefit of LMIC is that it is
`easy to enroll patients who are
`willing to participate,
`particularly if they are poor”
`......................................................
`
`As the standards for safety and efficacy
`developed over the decades, so did research
`ethics principles. Again, it was medical scan-
`dals that spawned today’s human subjects
`regulations in the USA, most notably the so-
`called Tuskegee Syphilis study. In this infa-
`mous experiment, conducted from 1932 to
`1972, US physicians denied penicillin, a
`known treatment for syphilis since the late
`1940s, to a cohort of syphilis-infected Afri-
`can American sharecroppers to study the
`natural course of advanced syphilis. Outrage
`over these practices led to the passage of the
`
`Freelance journalist in Oldenburg, Germany. E-mail: mail@k-weigmann.de
`DOI 10.15252/embr.201540398 | Published online 7 April 2015
`
`566
`
`EMBO reports Vol 16 | No 5 | 2015
`
`Exhibit 1127
`IPR2017-00807
`ARGENTUM
`ª 2015 The Author
`
`000001
`
`

`

`Katrin Weigmann
`
`Clinical trials in developing countries
`
`EMBO reports
`
`National Research Act in 1974 and eventu-
`ally to the Belmont report, which forms the
`basis
`for any human subject
`research
`conducted in the USA. As a result, the regu-
`latory process for drug development and
`testing in the USA has very high standards,
`but at a price: the US Food and Drug Admin-
`istration (FDA) has been regularly criticized
`for being disproportionately strict and for
`requesting too much data. Clinical
`trials
`have thus become increasingly complex and
`time-consuming in the USA, leading to what
`has been termed “drug lag”: prior to the
`Kefauver-Harris Amendments, an average of
`40 new drugs were introduced each year,
`whereas the number dropped to 16 there-
`after.
`
`T he FDA is in a difficult position: being
`
`too strict ensures safety, but it slows
`down the approval process and can
`delay new drugs from benefiting patients. If
`this
`affects
`drugs
`for
`life-threatening
`diseases, delays can cost lives if there are no
`other treatment options. By way of example,
`during the early 1990s, activists in the US
`lobbied the FDA to ease its rules and expe-
`dite approval for the first generation of anti-
`HIV drugs because so many AIDS/HIV
`patients were dying each year. On the other
`hand, not being strict enough can lead to
`public health problems if unsafe drugs enter
`the market. Prominent examples are Rezulin
`(troglitazone) and Vioxx (rofecoxib), both of
`which turned out to have unacceptable side
`effects.
`......................................................
`“Research began to leave the
`USA, first to Scandinavia and
`the UK, then to Eastern Europe
`and Latin America, and now
`to China and India”
`......................................................
`
`Yet, the FDA has an incentive to err on
`the side of caution: lives lost owing to drug
`delays usually results in fewer negative
`headlines than lives lost to dangerous drugs
`approved too soon. In addition, some of the
`FDA’s regulations make clinical research
`more burdensome and costly even without
`improving human subjects protection or
`drug safety. It has been argued, for example,
`that requiring continued annual review by
`Institutional Review Boards (IRBs) even for
`minimal risk studies is simply not necessary.
`
`In addition, multicentre studies are often
`reviewed by multiple local IRBs, a costly but
`redundant procedure. When clinical
`trials
`started to move abroad in the 1980s, strin-
`gent FDA regulations were a major factor,
`and still are today.
`FDA regulation is not the only reason
`that so many clinical trials have moved to
`LMIC [1]. Because many new drugs confer
`only a small benefit over existing treat-
`ments, ever larger trials with increasing
`numbers of human subjects are required to
`measure
`improvements with
`statistical
`significance. This has become particularly
`relevant for so-called me-too drugs that are
`chemically very similar to those already on
`the market. Recruiting a sufficient number
`of patients therefore remains a major bottle-
`neck for medical research, and companies
`have turned to LMIC to conduct
`trials
`because it is easier to find participants there.
`Moreover, operational costs in developing
`countries are low and there are large pools
`of “treatment-naive” patients, whereas in
`traditional research areas, the use of too
`much medication generates the risk of drug–
`drug interactions. Research began to leave
`the USA, first to Scandinavia and the UK,
`then to Eastern Europe and Latin America,
`and now to China and India (Fig 1). A recent
`investigation found that the 20 largest US-
`based
`pharmaceutical
`companies were
`conducting one-third of their clinical trials
`solely at foreign sites, while the majority of
`study sites are outside the USA [2].
`
`W hen research by US companies is
`
`in other countries,
`carried out
`the FDA still has some responsi-
`bilities to ensure that
`these comply with
`ethical standards. But
`there are doubts
`whether all
`the US rules and regulations
`should be exported to other countries. “The
`US regulatory system is far from perfect, it
`has been criticized as being overworked and
`inefficient. Some would argue that it is not
`the best model to bring to lower income
`countries that don’t have the resources we
`have”, said Sandra Alfano, Research Scien-
`tist at Yale School of Medicine and Chair of
`Yale’s Institutional Review Board (IRB).
`Moreover, not all ethical principles have
`cross-cultural validity. Informed consent, for
`example,
`is a central concept
`for any
`research with human subjects. However, as
`Alfano remarked, “the principle of informed
`consent flows out of a Western philosophy
`of
`individual autonomy. That does not
`
`always translate well to other countries”. An
`Eastern philosophy, she explained, may
`rather rely on community or family decision.
`“Individualism is not a universally embraced
`idea”, she said. She therefore thinks that
`Western researchers “should be strongly
`encouraged to involve the local community
`in setting the agenda—deciding whether this
`is a good project to be done—and then help-
`ing to design the research protocol and
`addressing the question about who is the
`right body to give consent”. This principle is
`referred to as “community-based participa-
`tory research”, or CBPR. “CBPR is really the
`focus on everyone’s mind, but it is very diffi-
`cult to achieve”, Alfano said.
`......................................................
`“. . . the primary goal of
`conducting clinical trials in
`developing countries should be
`to address the health needs of
`the host population”
`......................................................
`
`cultural differences and the
`Despite
`need to collaborate with local communi-
`ties,
`there are guidelines for conducting
`research in international settings, notably
`the Declaration of Helsinki (DoH) by the
`World Medical Association (WMA), and
`the international guidelines for biomedical
`research involving human subjects by the
`International Organisation
`for Medical
`Sciences (CIOMS). They are not
`legally
`binding, but set out
`important standards
`nonetheless. “Many countries refer to the
`DoH in their legislation. But what is more
`important is that it is a document written
`by physicians and for physicians. There is
`a moral obligation for physicians to stick
`to these rules”, said Ramin Parsa-Parsi,
`Head of the Department for International
`Affairs of the German Medical Association.
`The DoH was adopted in 1964 and has
`since undergone
`seven revisions, most
`recently in October 2013. A major revision
`was made in 2000 in response to studies
`performed with US federal funds of mother–
`child transmission of HIV in developing
`countries, which denied effective medication
`to participants in the placebo control arm.
`The DoH, in its 2000 revision, accordingly
`greatly limited the use of placebos. However,
`some argued that this would restrict research
`too much and that long-term benefits would
`justify the use of placebos [3]. The FDA,
`
`ª 2015 The Author
`
`EMBO reports Vol 16 | No 5 | 2015
`
`567
`
`000002
`
`

`

`EMBO reports
`
`Clinical trials in developing countries
`
`Katrin Weigmann
`
`CANADA
`13555
`
`UNITED
`STATES
`83689
`
`EUROPE
`51990
`
`MEXICO
`2309
`
`CENTRAL
`AMERICA
`2163
`
`AFRICA
`4375
`
`NORTH ASIA
`3463
`
`MIDDLE
`EAST
`7775
`
`EAST ASIA
`17010
`
`JAPAN
`3589
`
`SOUTH
`ASIA
`3097
`
`SOUTH
`AMERICA
`6334
`
`SOUTHEAST ASIA
`3835
`
`PACIFICA
`4914
`
`Source: http://ClinicalTrials.gov
`
`Colours indicate number of studies in clinical trials
`with locations in that region
`
`LEAST
`
`MOST
`
`Labels give exact study count
`
`Figure 1. Distribution of study locations of all trials registered with ClinicalTrials.gov.
`
`when regulating studies outside the USA,
`kept referring to older versions of the DoH
`and,
`in 2008, abandoned referring to it
`altogether at the expense of the “Harmoni-
`sation of Technical Requirements for Regis-
`tration of Pharmaceuticals for Human Use
`Good Clinical Practice” (ICH-GCP) guide-
`lines. “The ICH-GCP has been created by
`European, Japanese and US regulators in
`collaboration with
`the
`pharmaceutical
`industry. There are large differences in
`ethical standards between the DoH and
`ICH-GCP, particularly in the
`areas of
`placebo use and post-trial access”, Parsa-
`Parsi explained.
`The newest revision of the DoH, issued
`in late 2013, leaves the placebo paragraph
`essentially unchanged. The ICH-GCP regula-
`tion, on the other hand,
`is much more
`permissive: it explicitly states that the stan-
`dard of care required to be provided to the
`control group depends on the population,
`meaning that control group subjects are only
`entitled to the standard of care they would
`
`otherwise receive locally. This is morally
`ambiguous. According to London, “you
`can’t use the mere fact that people don’t
`have access to an intervention that works to
`say that you don’t have an obligation to
`provide them with something that works”.
`
`T he ongoing debate shows just how
`
`difficult it is to define ethical stan-
`dards for research in international
`settings, to walk the narrow line between
`protecting research subjects and enabling
`research that contributes to the social good.
`According to London,
`the question of
`placebo-controlled trials should be viewed in
`the context of the research question studied.
`In his view, the primary goal of conducting
`clinical trials in developing countries should
`be to address the health needs of the host
`population. “In general, we should provide a
`high standard of care”, he said, but main-
`tained that there could be rare exceptions.
`Such exceptions might be in cases where
`placebo-controlled trials are particularly
`
`important to developing countries: for exam-
`ple, when testing a new intervention that is
`similar to but cheaper than existing drugs, or
`one that has some other characteristic that
`conveys particular advantages in LMIC. One
`such feature would be heat resistance, which
`would make it easier to transport medication
`to remote locations in tropical areas. In such
`cases,
`it would be reasonable to use a
`placebo control
`in the trial, as it will be
`difficult to compare the efficiency of the new
`product to the existing one.
`A crucial aspect in the DoH and CIOMS
`guidelines concerns research on vulnerable
`groups. If people in developing countries
`belong to severely socio-economically disad-
`vantaged populations, this would impact on
`their
`freedom to consent, making them
`susceptible to undue influence and there-
`fore vulnerable. According to the DoH,
`research in vulnerable groups is only justi-
`fied, “if the research is responsive to the
`health needs or priorities of this group and
`the research cannot be carried out
`in a
`
`568
`
`EMBO reports Vol 16 | No 5 | 2015
`
`ª 2015 The Author
`
`000003
`
`

`

`Katrin Weigmann
`
`Clinical trials in developing countries
`
`EMBO reports
`
`this
`In addition,
`non-vulnerable group.
`group should stand to benefit
`from the
`knowledge, practices or interventions that
`result from the research”.
`......................................................
`“. . . taking advantage of
`people’s deprivation to impose
`on them the risk of health
`research can be exploitive if it
`is not carefully planned and
`regulated”
`......................................................
`
`“We have discussed including, in addition
`to the current requirements which reflect
`the ‘reasonable availability’ approach, a
`wording on fair benefits”, Parsa-Parsi said.
`Indeed, “fair benefits” is a hotly debated
`topic. Whereas the DoH and CIOMS base
`ethical acceptability mainly on responsive-
`ness to health needs of the host commu-
`nity, others maintain that
`research is
`justified as long as the host community
`receives “fair benefits” in a more broadly
`defined sense. Such benefits would not
`have to be directly related to the research
`projects and might include employment and
`training for community members or the
`building of
`infrastructure [4]. However,
`after long debates and consultations with
`developing countries, a fair benefits option
`was not included in the current version of
`the DoH. “The possibility of exploitation—
`that you buy consent with money or other
`benefits—was considered too high by the
`WMA”, Parsa-Parsi explained. London takes
`a similar view: “Ancillary benefits are
`important, but I don’t think they can make
`up for lack of relevance of the science to
`local health needs”, he said. For him,
`addressing the local health needs is the key
`issue. But ethical commitment should not
`stop there. “Just because I know the answer
`doesn’t make you better off. That informa-
`tion then has to be translated into some-
`thing from which you benefit concretely”,
`he said. “You start by studying the health
`needs, you build up an information base
`needed to discover interventions that work
`and then you close the cycle of translation
`by making those interventions and that
`knowledge available to their communities.”
`Availability of
`the drugs developed
`remains another major issue. Medication is
`often far too costly people in LMIC to afford,
`but with time, that too can change. “People
`
`were worried that HIV studies carried out in
`LMIC were not relevant to local health needs
`because the drugs were too expensive”,
`London recalled. “Ten years later, many
`more people have access than what was
`originally expected.” According to the WHO,
`the number of patients taking antiretroviral
`therapy has increased rapidly in LMIC since
`2003 from just 400,000 to 11.7 million by
`the end of 2013 (http://www.who.int/gho/
`hiv/epidemic_response/ART_text/en/).
`
`I n the meantime, other diseases are taking a
`
`terrible toll in LMIC, and it is not clear
`when new drugs will be cheap enough for
`wide use by their citizens. Hepatitis C, an
`infectious disease that can lead to liver cirrho-
`sis, is on the rise globally, and developing
`countries carry the highest burden with infec-
`tion rates as high as 11% in Egypt, 4.8% in
`Pakistan and 3.2% in China. During the past
`decade, research has led to the development of
`new drugs that work more effectively, in less
`time and with fewer side effects. Moreover, in
`contrast to the previous treatment regimens,
`these all-oral medications do not require regu-
`lar interferon injections, making them ideal for
`use in remote settings. The drugs are just start-
`ing to come to market, with more in the pipe-
`line, but their cost at the outset puts them out
`of reach for most people in developing coun-
`tries. It is likely that, as in the case of AIDS
`medication, it will take long until they become
`available to most LMIC.
`In many cases, therefore, studying local
`health needs will not necessarily generate
`immediate benefits for poorer communities.
`Nonetheless, studies that are responsive to
`local health needs will generate information
`that is necessary to improve public health in
`the long run. “When you ensure that the
`information is relevant to the local popula-
`tion, you build the foundation of knowledge
`necessary to generate beneficial
`interven-
`tions and policies, and this is a kind of bene-
`fit
`itself”, London commented. Making
`important new treatments available in LMIC
`should be considered a health priority, but
`this might take years or even decades at the
`current rate of progress. For participants in
`clinical trials, it will therefore be important
`to help them to bridge the gap between the
`end of
`the trial and the time the drug
`becomes available in their country.
`
`B oth the CIOMS and the DoH require
`
`to
`groups
`studies on vulnerable
`address their health needs. But does
`
`research in LMIC address local health needs
`in reality? According to the 2014 report by
`the Access to Medicine Foundation, an inter-
`national not-for-profit organization that
`ranks the top 20 research-based pharmaceu-
`tical companies with respect to their efforts
`to improve global access
`to medicine
`(http://www.accesstomedicineindex.org/),
`there is some promising development, but
`there is still a long way to go. “There is
`evidence of sustained commitment to R&D
`for relevant diseases, with most companies
`having an R&D strategy in place that explic-
`itly takes patients in developing countries
`into account”,
`the
`report
`says. More
`precisely,
`the index investigated research
`activities on 47 diseases that were regarded
`as most relevant to LMIC according to the
`WHO. Although a number of studies tackle
`these diseases, the research activities can
`mainly be attributed to only five of the 20
`companies, and they concentrate on only
`five of the 47 diseases: diabetes, lower respi-
`ratory infections, hepatitis, HIV/AIDS and
`malaria. Neglected tropical diseases, as the
`report shows, remain neglected. In addition,
`the report criticises companies’ lack of effort
`to make their products available to people in
`LMIC: “On average, products are registered
`in only [. . .] 8% of low-income countries
`covered by the Index.”
`Conducting clinical trials in LMIC is not
`intrinsically immoral. Indeed, trials some-
`times represent the only medical treatment
`that patients in deprived settings can hope
`to receive, and can also provide other
`advantages at the personal, community and
`national levels. Nonetheless, taking advan-
`tage of people’s deprivation to impose on
`them the risk of health research can be
`exploitive if it is not carefully planned and
`regulated. At the moment, there is such a
`variety of
`legislation that
`the conduct of
`trials differs widely, often depending on why
`a company decided to use a developing
`country in the first place. Trials that address
`locally relevant diseases and that will deliver
`affordable drugs and care for the local popu-
`lation are certainly to be lauded. Ones that
`do not provide immediate or obvious bene-
`fits locally are less laudable and must be
`assessed on a case-by-case basis.
`
`References
`1. Petryna A (2009) When Experiments Travel:
`Clinical Trials and the Global Search for Human
`Subjects. Princeton, NJ: Princeton University
`Press
`
`ª 2015 The Author
`
`EMBO reports Vol 16 | No 5 | 2015
`
`569
`
`000004
`
`

`

`EMBO reports
`
`Clinical trials in developing countries
`
`Katrin Weigmann
`
`2. Glickman SW, McHutchison JG, Peterson ED,
`Cairns CB, Harrington RA, Califf RM, Schulman
`KA (2009) Ethical and scientific implications of
`the globalization of clinical research. N Engl J
`Med 360: 816 – 823
`
`3. Wolinsky H (2006) The battle of Helsinki:
`two troublesome paragraphs in the Declara-
`tion of Helsinki are causing a furore over
`medical research ethics. EMBO Rep 7:
`670 – 672
`
`4.
`
`Schulz-Baldes A1, Vayena E, Biller-Andorno N
`(2007) Sharing benefits in international health
`research. Research-capacity building as an
`example of an indirect collective benefit.
`EMBO Rep 8: 8 – 13
`
`570
`
`EMBO reports Vol 16 | No 5 | 2015
`
`ª 2015 The Author
`
`000005
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket