#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Predicting Harms and Benefits in Translational Trials: Ethics,
Evidence, and Uncertainty


article has not abstract


Published in the journal: . PLoS Med 8(3): e32767. doi:10.1371/journal.pmed.1001010
Category: Essay
doi: https://doi.org/10.1371/journal.pmed.1001010

Summary

article has not abstract

Summary Points

  • Ethical judgments about risk, benefit, and patient eligibility in clinical trials hinge on predictions about harm, therapeutic response, and clinical promise.

  • Predictions for novel interventions in preclinical stages of development suffer from two problems: insufficient attention to threats to validity in preclinical research and a reliance on an overly narrow base of evidence that includes only animal and clinical studies of the intervention in question (“evidential conservatism”).

  • To improve ethical and scientific decision-making in early phase studies, decision-makers should explicitly attend to reporting quality and methodological features in preclinical experiments that address threats to internal, construct, and external validity.

  • Decision-makers should also use evidence that sheds light on the reliability of causal claims embedded within a proposed trial. This evidence can be gathered from outcomes of previous trials involving agents targeting related biological pathways (“reference classes”).

Introduction

First-in-human clinical trials represent a critical juncture in the translation of laboratory discoveries. However, because they involve the greatest degree of uncertainty at any point in the drug development process, their initiation is beset by a series of nettlesome ethical questions [1]: has clinical promise been sufficiently demonstrated in animals? Should trial access be restricted to patients with refractory disease? Should trials be viewed as therapeutic? Have researchers adequately minimized risks?

The resolution of such ethical questions inevitably turns on claims about future events like harms, therapeutic response, and clinical translation. Recurrent failures in clinical translation, like Eli Lilly's Alzheimer candidate semagacestat, highlight the severe limitations of current methods of prediction. In this case, patients in the active arm of the placebo-controlled trial had earlier onset of dementia and elevated rates of skin cancer [2].

Various authoritative accounts of human research ethics state that decision-making about risk and benefit should be careful, systematic, and non-arbitrary [3][5]. Yet, these sources provide little guidance about what kinds of evidence stakeholders should use to ensure their estimates of such events ground responsible ethical decisions. In this article, we suggest that investigators, oversight bodies, and sponsors often base their predictions on a flawed and inappropriately narrow preclinical evidence base.

Prediction and Ethical Decision-Making

According to the core tenets of human research ethics, investigators, sponsors, and institutional review boards (IRBs) are obligated to ensure that risks to volunteers are minimized and balanced favorably with anticipated benefits to society and, if applicable, to the volunteers themselves [4],[6]. Accurate prediction plays a critical role in this process. When research teams underestimate the probability of favorable clinical or translational outcomes, they undermine health care systems by impeding clinical translation. When investigators overestimate the probability of favorable outcomes, they potentially expose individuals to unjustified burdens, which may be considerable for phase 1 studies involving unproven drugs. In both cases, misestimation threatens the integrity of the scientific enterprise, because it frustrates prudent allocation of research resources [7].

Naturally, there are limits to the reliability with which forecasts based on experimental evidence predict clinical outcomes. However, in late stages of clinical development, forecasts underwriting ethical and scientific decision-making have proven fairly reliable. Several analyses of cancer randomized controlled trials indicate that new interventions are just as likely to prove more effective than comparator ones as they were to prove inferior [8][10]. Similar findings have been reported for other indications [11]. In the aggregate at least, researchers and review committees neither overestimate nor underestimate the medical benefits of allocating some patients to new interventions and others to standard drugs.

Whether decision-makers utilize evidence as effectively when predicting outcomes in early phase research has not been systematically investigated. Nevertheless, there are grounds for concern such that a systematic investigation is overdue. Highly promising preclinical findings in cancer, stroke, HIV vaccines, and neurodegenerative diseases frequently fail clinical translation. In cancer, only 5% of products entering trials are eventually licensed [12],[13]. In one study, approximately 5% of high impact basic science reports were clinically translated within 10 years [14]. We suggest that these disappointments partly reflect two problems in the way evidence is used in predicting clinical outcomes.

Preclinical Reporting and Validity

First, decision-makers may not be adequately responsive to problems in preclinical research practice [15]. Systematic reviews repeatedly demonstrate that many animal studies do not enable reliable causal inference and clinical generalization because they do not address important threats to internal, construct, and external validity. With respect to the first, one recent analysis of animal studies showed that only 12% used random allocation and 14% used blinded outcome assessment [16]. Construct validity concerns the relationship between clinical implementation of an intervention and implementations evaluated in preclinical studies. A recent review found that clinical studies of cardiac arrest interventions applied treatment significantly sooner after cardiac events than in preclinical studies [17]. In the case of Astra Zeneca's failed stroke drug NXY-059, use of normotensive rodents in preclinical development may have led to spurious predictions of clinical activity [18]. Preclinical studies do not always test the extent to which cause and effect relationships hold up under varied conditions (external validity). In a systematic review of neuroprotective agents in phase 2 and 3 trials, only two of ten agents were tested in both rodents and higher order species [19]. Finally, deficiencies in reporting and aggregation of preclinical evidence deprive decision-makers of crucial evidence. In one recent analysis, publication bias in preclinical stroke studies led to a 30% overestimation of treatment effect size [20]. Clearly, preclinical researchers should endeavor to follow reporting guidelines [21] such as the recently proposed Animals In Research: Reporting In Vivo Experiments Guidelines (ARRIVE; http://www.nc3rs.org.uk/page.asp?id=1357) [22], and clinical predictions following from animal studies should take into account deficiencies in design and reporting.

In the case of semegacestat, it has been over 5 years since the drug was first tested in human beings, and preclinical studies have yet to be published. However, narrative reviews by Eli Lilly scientists indicate trials were launched on the basis of molecular, rather than behavioral, endpoints [23]. Although the absence of publication makes difficult any assessment of animal study quality, the use of molecular endpoints raises questions about the construct validity of clinical generalizations drawn from preclinical experiments.

Evidential Conservatism

A second concern about forecasting outcomes in translational trials relates to a tendency to base clinical inferences on a relatively narrow class of evidence: those preclinical studies that involve the particular agent. We call this “evidential conservatism.” Such evidential conservatism is reflected in various policies. For example, the American Society of Clinical Oncology states that “the decision to move an agent into phase I evaluation is based… central[ly on]… the observation of sufficient preclinical antitumor activity, such that a therapeutic effect in human cancer is anticipated” [24],[25]. International Council on Harmonization policy requires investigators to furnish ethics review committees with only a narrow type of preclinical evidence [26]. Similarly, some commentators argue that risk-benefit decisions in early phase trials should be driven by mechanistic evidence about an agent [27].

Evidential conservatism, however, fails to address the higher-order question of the reliability of forecasts made from such a narrow evidence base. This higher-order question is of special relevance for early phase research because agents that do not enjoy the support of promising preclinical results will not be plausible candidates for translation. Yet when agents are supported by equally promising preclinical results they may be differentiated by the maturity of the knowledge surrounding a nexus of variables concerning the relationship between test and target populations.

For instance, although neuroprotective stroke treatments have moved to translation on the basis of very encouraging preclinical studies, they have consistently failed randomized trials. Estimates of the risks and benefits of any particular neuroprotective compound that are based solely on preclinical evaluation of that compound will be less reliable than those that incorporate information about the relative success of neuroprotective compounds as a class. In part, this is because the success or failure of other interventions in this reference class provides evidence about the degree to which clinical development is guided by a reliable working knowledge of relevant disease processes.

Our claim that decision-makers need to use a broader base of evidence for evaluating early phase research is consistent with a recent call for incorporating whole research program outcomes into systematic reviews of particular agents [28].

Assessing Relevant Evidence

How might researchers depart from evidential conservatisim in a way that is open to scrutiny and amenable to assessment, revision, and improvement? Decision-makers who make forecasts about agent activity in early phase research must identify reference classes that are relevant to the decision at hand. Delimiting the reference class of relevant evidence poses a challenge in that interventions possess limitless characteristics. A drug might be classed within neuroprotective compounds, stroke drugs, and drugs beginning with the letter “n.” Decision-makers thus confront the timeless problem of selecting those characteristics most salient for prediction.

There are no simple formulas here. In some cases, choice of reference classes will be straightforward (e.g., a new, small molecule HMG-CoA reductase inhibitor); in other cases, consensus may be elusive. Nevertheless, we suggest that the very act of attending to reference class identity would be a departure from evidential conservatism. As a starting place, decision-makers should identify reference classes that index the maturity of knowledge regarding central causal premises embedded within a protocol. In an era in which basic science heavily informs product development, drug developers themselves often class their agents according to explicit ambitions about causal pathways. Asserting that a drug targets a particular pathophysiologic process should prompt us to look at how other drugs that target the same process performed in clinical translation. We can then base our estimates of the maturity of knowledge about these causal premises on the success or failure of past attempts at redeeming these ambitions. Decision-makers should therefore adjust their confidence in clinical generalizations on the basis of outcomes with previous interventions that addressed the same pathological processes.

Semagacestat was screened and designed to target amyloid-β production, which is believed to be a key step in dementia onset. Eight other anti-amyloid drugs have either failed randomized trials or been abandoned due to toxicity (Table 1) [29],[30]. Although a variant of this approach may eventually succeed, promising preclinical evidence supporting semagecestat should have been tempered by the accumulation of data about outcomes in the same reference class.

Tab. 1.

Practical Implications

To illustrate how our suggestions interface with ethical decision-making, consider recent proposals to reinitiate trials of fetal-derived tissues for Parkinson's disease [31]. Previous trials involved treatment-refractory patients, but investigators are now proposing trials involving patients with recent onset. The rationale is that fetal-derived tissues can only protect dopaminergic neurons to the extent that the latter remain intact. However, the risk-benefit balance is contentious, because the trial will expose patients who can manage symptoms with standard treatments to the risks of neurosurgery, immunosuppression, and cell transplantation.

According to evidential conservatism, investigators and ethics bodies should evaluate the risk-benefit balance by consulting preclinical studies and the biological rationale for patient-subject selection. One commentator notes that, on the basis of preclinical studies showing the intervention is designed to address early disease processes, performing studies in patients with advanced disease would be unethical [27]. We think this way of using evidence in ethical evaluation is misguided.

Our proposal directs decision-makers to make risk-benefit decisions in light of two additional factors. First, to what degree do the preclinical studies incorporate design elements that support reliable inferences about clinical activity? This directs stakeholders to attend to those methodological features of the preclinical studies that support credible claims of internal, construct, and external validities in preclinical studies. As these preclinical studies are presently underway, researchers have an opportunity to overcome past limitations in addressing validity threats in Parkinson's disease models [32].

Second, our proposal directs stakeholders to consider evidence that sheds light on the maturity of the knowledge relating to key causal claims presupposed by therapeutic predictions. As investigators propose to intervene in degenerative processes, a claim of therapeutic action would need to be evaluated in light of outcomes in previous Parkinson's trials involving surgically delivered neuroprotective agents and/or transplanted tissues. No such strategies have produced positive randomized trials (Table 2). Accordingly, even with carefully collected preclinical evidence, decision-makers should approach new trials with modest therapeutic expectations.

Tab. 2.

Thoughtful commentators have argued that, before initiating cell-based dopamine replacement, strategies should be “clinically competitive” with standard of care [33]. However, this may present an unworkable standard [34]. Previous unsuccessful attempts at translation betray profound uncertainty concerning risks and benefits for research volunteers. Given the preliminary nature of such interventions, the ethical justification for their administration in early phase trials should not hinge on the prospect of benefit for volunteers. It should rest instead on a compelling claim of knowledge value and on the reduction of avoidable risks. The latter entails pursuing trials in patients less likely to suffer opportunity costs from study participation, and maintaining a background of medical management that does not fall below standard of care. Rather than being told that the approach is comparable to standard of care, the consent process should emphasize that clinical benefit is unlikely.

Conclusion

Systematic study of preclinical research has centered on stroke and practices focused on internal validity. Our proposal makes clear the need to broaden the scope of this research agenda to cover a wider range of preclinical research, and to expand its focus to include issues of construct and external validity. A key component of this process will involve creating databases for aggregating translational outcomes according to relevant reference classes.

Some may worry that such an analysis might produce less optimistic predictions, and hence stymie product development. However, we do not see how medicine is advanced by forging ahead on the basis of predictions of dubious reliability. Moreover, there are many productive ways in which stakeholders may respond to less optimistic projections. For instance, review of relevant information may prompt researchers to test certain hypotheses before moving ahead with human trials. Investigators might adjust the design of translational studies to align the risk profile with ethical judgments. Or, investigators might decide that moving forward with a protocol represents the best way to advance a particular scientific initiative, but that risks can only be justified by appealing to the value of the knowledge sought, rather than the product's therapeutic activity.

Stakeholders might already adjust their predictions in light of intuitions about validity or experiences with success or failure for similar agents. If so, they do so on the basis of private beliefs, and often without the data needed to make these adjustments systematically. Our approach provides a more publicly accessible basis for making and adjudicating risk-benefit predictions. We suggest that this would better cohere with a sage prescription offered by the National Commission: “there should first be a determination of the validity of the presuppositions of the research…. The method of ascertaining risks should be explicit… It should also be determined whether an investigator's estimates of the probability of harm or benefits are reasonable, as judged by known facts or other available studies ” [3].


Zdroje

1. KimmelmanJ

2010

Gene transfer and the ethics of first-in-human research: lost in

translation.

Cambridge

Cambridge University Press

2. ExtanceA

2010

Alzheimer's failure raises questions about disease-modifying

strategies.

Nat Rev Drug Discov

9

749

751

3. The National Commission for the Protection of Human Subjects of

Biomedical and Behavioural Research

1979

The Belmont report: ethical principles and guidelines for the

protection of human subjects of research.

Bethesda

Department of Health Education and Welfare

4. World Medical Association

1964

Declaration of Helsinki.

Helsinki

18th World Medical Assembly

5. MannH

2010

ASSERT: a standard for the review and monitoring of randomized

clinical trials.

Available: http://www.assert-statement.org/. Accessed 31 January

2011

6. Department of Health and Human Services

2005

Protection of human subjects: criteria for IRB approval of

research.

Title 45 CFR 46.111(a)(1)

1

12

7. LondonAJKimmelmanJEmborgME

2010

Research ethics. Beyond access vs. protection in trials of

innovative therapies.

Science

328

829

830

8. DjulbegovicBKumarASoaresHPHozoIBeplerG

2008

Treatment success in cancer: new cancer treatment successes

identified in phase 3 randomized controlled trials conducted by the National

Cancer Institute-sponsored cooperative oncology groups, 1955 to

2006.

Arch Intern Med

168

632

642

9. KumarASoaresHWellsRClarkeMHozoI

2005

Are experimental treatments for cancer in children superior to

established treatments? Observational study of randomised controlled trials

by the Children's Oncology Group.

BMJ

331

1295

10. SoaresHPKumarADanielsSSwannSCantorA

2005

Evaluation of new treatments in radiation oncology: are they

better than standard treatments?

JAMA

293

970

978

11. GrossCPKrumholzHMVan WyeGEmanuelEJWendlerD

2006

Does random treatment assignment cause harm to research

participants?

PLoS Med

3

e188

doi:10.1371/journal.pmed.0030188

12. KolaILandisJ

2004

Can the pharmaceutical industry reduce attrition

rates?

Nat Rev Drug Discov

3

711

715

13. PangalosMNSchechterLEHurkoO

2007

Drug development for CNS disorders: strategies for balancing risk

and reducing attrition.

Nat Rev Drug Discov

6

521

532

14. Contopoulos-IoannidisDGNtzaniEIoannidisJP

2003

Translation of highly promising basic science research into

clinical applications.

Am J Med

114

477

484

15. van der WorpHBHowellsDWSenaESPorrittMJRewellS

2010

Can animal models of disease reliably inform human

studies?

PLoS Med

7

e1000245

doi:10.1371/journal.pmed.1000245

16. KilkennyCParsonsNKadyszewskiEFestingMFCuthillIC

2009

Survey of the quality of experimental design, statistical

analysis and reporting of research using animals.

PLoS ONE

4

e7824

doi:10.1371/journal.pone.0007824

17. ReynoldsJCRittenbergerJCMenegazziJJ

2007

Drug administration in animal studies of cardiac arrest does not

reflect human clinical experience.

Resuscitation

74

13

26

18. BathPMGrayLJBathAJBuchanAMiyataT

2009

Effects of NXY-059 in experimental stroke: an individual animal

meta-analysis.

Br J Pharmacol

157

1157

1171

19. PhilipMBenatarMFisherMSavitzSI

2009

Methodological quality of animal studies of neuroprotective

agents currently in phase II/III acute ischemic stroke

trials.

Stroke

40

577

581

20. SenaESvan der WorpHBBathPMHowellsDWMacleodM

2010

Publication bias in reports of animal stroke studies leads to

major overstatement of efficacy.

PLoS Biol

8

e1000344

doi:10.1371/journal.pbio.1000344

21. MacleodMRFisherMO'CollinsVESenaESDirnaglU

2009

Good laboratory practice. Preventing introduction of bias at the

bench.

Stroke

40

e50

e52

22. KilkennyCBrowneWCuthillIEmersonMAltmanD

2010

Improving bioscience research reporting: the ARRIVE guidelines

for reporting animal research.

PLoS Biol

8

e1000412

doi:10.1371/journal.pbio.1000412

23. HenleyDBMayPCDeanRASiemersER

2009

Development of semagacestat (LY450139), a functional

gamma-secretase inhibitor, for the treatment of Alzheimer's

disease.

Expert Opin Pharmacother

10

1657

1664

24. American Society of Clinical Oncology

1997

Critical role of phase I clinical trials in cancer

treatment.

J Clin Oncol

15

853

859

25. ChristianMShoemakerD

2002

The investigator's handbook: a manual for participants in

clinical trials of investigational agents sponsored by DCTD,

NCI.

Bethesda

Cancer Therapy Evaluation Program

26. International Conference on Harmonisation of Technical Requirements for

Registration of Pharmaceuticals for Human Use (ICH)

1996

ICH Harmonized Tripartite Guideline.

Guideline for Good Clinical Practice E6(R1)

27. LowensteinPR

2008

A call for physiopathological ethics.

Mol Ther

16

1771

1772

28. IoannidisJPKarassaFB

2010

The need to consider the wider agenda in systematic reviews and

meta-analyses: breadth, timing, and depth of the evidence.

BMJ

341

c4875

29. MangialascheFSolomonAWinbladBMecocciPKivipeltoM

2010

Alzheimer's disease: clinical trials and drug

development.

Lancet Neurol

9

702

716

30. CummingsJ

2010

What can be inferred from the interruption of the semagacestat

trial for treatment of Alzheimer's disease?

Biol Psychiatry

68

876

878

31. HoldenC

2009

Neuroscience. Fetal cells again?

Science

326

358

359

32. KimmelmanJLondonAJRavinaBRamsayTBernsteinM

2009

Launching invasive, first-in-human trials against

Parkinson's disease: ethical considerations.

Mov Disord

24

1893

1901

33. LindvallOKokaiaZ

2010

Stem cells in human neurodegenerative disorders—time for

clinical translation?

J Clin Invest

120

29

40

34. AndersonJAKimmelmanJ

2010

Extending clinical equipoise to phase 1 trials involving

patients: unresolved problems.

Kennedy Inst Ethics J

20

75

98

35. GilmanSKollerMBlackRSJenkinsLGriffithSG

2005

Clinical effects of Abeta immunization (AN1792) in patients with

AD in an interrupted trial.

Neurology

64

1553

1562

36. FeldmanHHDoodyRSKivipeltoMSparksDLWatersDD

2010

Randomized controlled trial of atorvastatin in mild to moderate

Alzheimer disease: LEADe.

Neurology

74

956

964

37. Elan Corporation

2010

Elan and Transition Therapeutics announce topline summary results

of Phase 2 study and plans for Phase 3 for ELND005 (Scyllo-inositol)

[press release]

38. SallowaySSperlingRGilmanSFoxNCBlennowK

2009

A phase 2 multiple ascending dose trial of bapineuzumab in mild

to moderate Alzheimer disease.

Neurology

73

2061

2070

39. WinbladBGiacobiniEFrolichLFriedhoffLTBruinsmaG

2010

Phenserine efficacy in Alzheimer's disease.

J Alzheimers Dis

22

1201

1208

40. GoldMAldertonCZvartau-HindMEggintonSSaundersAM

2010

Rosiglitazone monotherapy in mild-to-moderate alzheimer's

disease: results from a randomized, double-blind, placebo-controlled phase

III study.

Dement Geriatr Cogn Disord

30

131

146

41. GreenRCSchneiderLSAmatoDABeelenAPWilcockG

2009

Effect of tarenflurbil on cognitive decline and activities of

daily living in patients with mild Alzheimer disease: a randomized

controlled trial.

JAMA

302

2557

2564

42. Bellus Health Inc

2008

Neurochem announces results from Tramiprosate (ALZHEMED(TM))

North American Phase III clinical trial

43. MarksWJJrBartusRTSiffertJDavisCSLozanoA

2010

Gene delivery of AAV2-neurturin for Parkinson's disease: a

double-blind, randomised, controlled trial.

Lancet Neurol

9

1164

1172

44. NuttJBurchielKJComellaCLJankovicJLangAE

2003

Randomized, double-blind trial of glial cell line-derived

neurotrophic factor (GDNF) in PD.

Neurology

60

69

73

45. LangAEGillSPatelNKLozanoANuttJG

2006

Randomized controlled trial of intraputamenal glial cell

line-derived neurotrophic factor infusion in Parkinson

disease.

Ann Neurol

59

459

466

46. FreedCRGreenePEBreezeRETsaiWYDuMouchelW

2001

Transplantation of embryonic dopamine neurons for severe

Parkinson's disease.

N Engl J Med

344

710

719

47. OlanowCWGoetzCGKordowerJHStoesslAJSossiV

2003

A double-blind controlled trial of bilateral fetal nigral

transplantation in Parkinson's disease.

Ann Neurol

54

403

414

48. WattsRLFreemanTBHauserRABakayRAEElliasSA

2001

A double-blind, randomised, controlled, multicenter clinical

trial of the safety and efficacy of stereotaxic intrastriatal implantation

of fetal porcine ventral mesencephalic tissue (Neurocelli-PD) vs. imitation

surgery in patients with Parkinson's disease (PD).

Parkinsonism Relat Disord

7

Suppl

S87

49. OlanowCWSternMBSethiK

2009

The scientific and clinical basis for the treatment of Parkinson

disease.

Neurology

72

21 Suppl 4S1

S136

Štítky
Interní lékařství

Článek vyšel v časopise

PLOS Medicine


2011 Číslo 3
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#