Understanding pharmacology in humans: Phase I and Phase II (Data Generation)

https://doi.org/10.1016/j.coph.2011.06.010Get rights and content

The discovery of novel drugs is a complex and highly regulated process organized around a critical moment, that is, when the novel compound is tested in humans. This process encompasses a series of clinical studies, identified as Phase I and Phase II, whose composite outcome should deliver the data needed for an informed decision about progressing or not the compound in full development (Phase III). Over the last 10 years the global delivery of novel treatments from the pharmaceutical industry has plunged to the level of the ‘70ies in spite of a 10-fold larger investment, the differential mostly due to failures in Phase III. There is the need to improve the decision making at the early clinical stage by using innovation and the high-profile achievements of basic science generated in academic and biomedical labs. A specific attention should be paid to applied biotechnologies, in particular nanotechnology and biomedical devices not only for drug deliver but also for biomarker detection. This path, also supported by regulatory agencies, is calling for an important change of perspective about how drug discovery is made, which we believe should start from the full implementation of the paradigm of Translational Medicine.

Highlights

► Regulatory expectations and conservatory attitude. ► Early human studies and the risk of late phase failure. ► Translational Medicine philosophy: Is the compound effective in humans?. ► Innovation in clinical trial methodologies. ► Novel technologies and discoveries that can be ‘game changers’.

Introduction

The early clinical stage of drug discovery consists of a series of studies required by the regulatory agencies and aimed to characterize the basic features of an experimental compound, the Novel Chemical Entity (NCE) [1]. Traditionally these studies are organized as clinical trials subdivided according to their delivery and design into Phase I and Phase II (see Box 1). While these trials are effective in detecting safety-related issues, their capacity to inform about pharmacological effects and clinical efficacy is less than optimal, in particular when the conventional design is implemented. The low efficiency of early clinical trials is compounded by the problem of translating the effects observed in preclinical tests and in animal models into humans, not always a linear process. In fact, specie-specific biologic differences can be so relevant to severely reduce the predictive power of preclinical pharmacological studies, as experienced, for example, in the area of drug discovery for acute ischemic stroke [2]. In this case the incomplete characterization of the NCE during the early clinical stage had catastrophic effects, resulting in failed Phase III trials, with several hundred patients unnecessary exposed to a non-effective treatment. While no simple recipe is available to avoid errors, improvements in the management of the drug discovery process are possible starting exactly from the early clinical stages.

Section snippets

Inefficiency of the drug discovery process: focus on the early clinical stage

The inefficiency of the drug discovery process, become evident during the first years of the millennium [3]. The long delay between discovery of NCE and the low delivery rate of clinically effective assets to the market were associated with a rapid increase of the R&D costs [3•, 4]. The biggest player in driving this cost increase was the late clinical development, mostly due to repeated failure of large and expensive clinical trials: in those years up to 45% of clinical trials in Phase III

Will Translational Medicine rescue drug discovery?

Translation Medicine consists of the process of transferring basic science achievements into practical applications aimed to improve patients’ health. This translation implies: first, the biologic levels, where the disease understanding is driving the use of biomarkers either as diagnostics or prognostics, or as surrogate markers of the therapeutic effects of a drug; and second, the social level, where the benefits of a novel treatment are distributed to the whole population, resulting in an

A biomarker clinical trial: functional neuroimaging for novel anxiolytics

In our experience a biomarker clinical trial should use a detection technology previously tested in the recruitment site with some success, that is, able to deliver an acceptable Effect Size (ES  1). As example we show the case of the NK1 antagonist GW597599 that was in early clinical discovery as novel anxiolytic based on preclinical animal data at GSK in early 2000. Scientists at the University of Uppsala developed a perfusion PET protocol to measure changes of blood flow in the Medial

Conclusion

Notwithstanding the advancements made towards a proper integration of biomarkers into the Translational Medicine paradigm, several pharmaceutical companies are just starting entering this game. Post-action assessments of the decision-making process following a major failure in Phase III are occurring more often than in the past and articles are published discussing the ‘learnings’. For example, the CETP inhibitor torcetrapib, that reliably increases HDL cholesterol in preclinical setting,

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (20)

  • O. Stüve et al.

    Translational research in neurology and neuroscience 2010: multiple sclerosis

    Arch Neurol

    (2010)
  • R.J.Y. Ho et al.

    Drug delivery trends in clinical trials and Translational Medicine: updated analysis of clinicaltrials.gov database

    J Pharm Sci

    (2009)
  • Glossary of Clinical Trial

    (2011)
  • G.Z. Feuerstein et al.

    Missing steps in the STAIR case: a Translational Medicine perspective on the development of NXY-059 for treatment of acute ischemic stroke

    J Cereb Blood Flow Metab

    (2008)
  • I. Kola et al.

    Can the pharmaceutical industry reduce attrition rates?

    Nat Rev Drug Discov

    (2004)
  • FDA: Innovation or Stagnation. Challenge and Opportunity Report, March...
  • K.I. Kaitin

    Deconstructing the drug development process: the new face of innovation

    Clin Pharmacol Therapeut

    (2010)
  • A.D. McCarthy et al.

    Pharmacogenetics in drug development

    Philos Trans R Soc Lond Ser B: Biol Sci

    (2005)
  • J.K. Willmann et al.

    Molecular imaging in drug development

    Nat Rev Drug Discov

    (2008)
  • J.A. Bilello

    The agony and ecstasy of “OMIC” technologies in drug development

    Curr Mol Med

    (2005)
There are more references available in the full text version of this article.

Cited by (5)

  • Functional effects of chronic paroxetine versus placebo on the fear, stress and anxiety brain circuit in Social Anxiety Disorder: Initial validation of an imaging protocol for drug discovery

    2014, European Neuropsychopharmacology
    Citation Excerpt :

    In the present study each subject was also exposed to an off-scanner Public Speaking Test, a procedure that measures distress while the subject is challenged by performing a speech in front of a silent audience. This test is commonly used to study serotonergic anxiolytics when the study population is small, e.g., 20 or less subjects per group and with an effect size of about 1.0 (Graeff et al., 2003; Furmark et al., 2005; Merlo Pich, 2011). In this article the capacity to detect pharmacologic effects of the Public Speaking Test, as well as that of other clinical scales including LSAS, was used as benchmark to indirectly compare the results of various fMRI tasks assessed in this study.

  • A disruptive innovation model for indigenous medicine research: A nigerian perspective

    2013, African Journal of Science, Technology, Innovation and Development
View full text