Abstract

Application of clinical pharmacology best practices is essential to the efficient and rational development of drugs. In general, knowledge gained about exposure–response relationships in preclinical models aids drug and dose selection in human studies, and biomarkers and pharmacokinetic (PK) data one collects in early to middle drug development can be used to predict the dose and treatment response of promising therapeutics in definitive phase 3 trials. The essentiality of sound clinical pharmacology in tuberculosis (TB) drug and regimen development is heightened by unique challenges in assessing drugs for this disease—aspects of the organism’s biology, the variability in lung pathology, uncertainties about how to link treatment outcomes seen in preclinical models with those seen in humans (which thwarts preclinical–clinical translational work), the lack of predictive early clinical biomarkers, and the high variability in treatment response across patients and populations (Fig 1). In TB disease, Mycobacterium tuberculosis (M.tb) bacilli are detected in necrotic granulomas, large cavities with liquefied contents, and intracellularly within macrophages. We believe that drugs must access each of these compartments to achieve cure in patients [1]. We also believe that TB drugs and regimens must kill bacilli in different metabolic states, from actively multiplying to semidormant [2,3]. Both in vitro and in vivo preclinical models are leveraged to assess the clinical utility of new TB drugs and drug combinations. These models vary both in their ability to assess efficacy relative to the shifting metabolic states of M.tb infection and in their ability to recapitulate human disease. Still, two models are proving to be highly informative. The mouse model of infection has been invaluable in selecting rank-ordered drug combinations, whereas the now-validated in vitro pharmacodynamic (PD) system (IVPDS, or “hollow fiber model” for TB) has significantly improved our understanding of the PK drivers of treatment response in various growth and physiologic states [4,5]. In the IVPDS, an elaborate system of dialysis-like tubing allows the investigator to reproduce human-like concentration–time curves and see how different PK profiles affect killing of bacilli that are living in the system. Dose-fractionation studies, for example, can be carried out, and one can determine whether a drug’s activity is time dependent or, rather, concentration dependent. Or one can test a drug’s activity when the organism is nutrient starved, in log-phase growth, or intracellular. Whereas these models are informative, there remain gaps in our ability to bridge preclinical and clinical data using modern translational quantitative modeling [6]. There are also gaps in our ability to link surrogate end points in early-phase clinical trials (namely, longitudinally collected sputum cultures) and clinically relevant end points of treatment failure, relapse, and death in later-phase trials [7,8]. The identification of accurate tools that identify those patients who are unlikely to achieve cure with shortened regimens (specifically, patients with a disease phenotype that is “hard to treat”) would have immense value to both clinical trialists and TB clinicians [9]. The TB clinical pharmacology field has the opportunity to apply state-of-the-art quantitative pharmacology tools to bridge preclinical and clinical data more effectively and to enhance learning across the continuum of clinical development [9–13]. In this paper, based on discussions occurring at a WHO workshop held in March 2018, we describe our views on best practices for incorporating translational, PK-PD, and microbiologic assessments into drug development [14]. Open in a separate window Fig 1 Schema of preclinical and clinical pharmacology studies important for TB drug and regimen development. By phase of development, in green are the questions to be addressed, in blue are the tools to use to answer the questions, and in red are the outputs. ADME, absorption, distribution, metabolism, excretion; DDI, drug–drug interaction; Dz, disease; MIC, minimum inhibitory concentration; PBPK, physiologically based PK; PD, pharmacodynamic; Ph2A, phase 2A; Ph2B/C, phase 2B and C; Ph3, phase 3; PK, pharmacokinetic; TB, tuberculosis; y.o., year-olds.

Highlights

  • Application of clinical pharmacology best practices is essential to the efficient and rational development of drugs

  • The essentiality of sound clinical pharmacology in tuberculosis (TB) drug and regimen development is heightened by unique challenges in assessing drugs for this disease—aspects of the organism’s biology, the variability in lung pathology, uncertainties about how to link treatment outcomes seen in preclinical models with those seen in humans, the lack of predictive early clinical biomarkers, and the high variability in treatment response across patients and populations (Fig 1)

  • In this paper, based on discussions occurring at a WHO workshop held in March 2018, we describe our views on best practices for incorporating translational, PK-PD, and microbiologic assessments into drug development [14]

Read more

Summary

Introduction

Application of clinical pharmacology best practices is essential to the efficient and rational development of drugs. Knowledge gained about exposure–response relationships in preclinical models aids drug and dose selection in human studies, and biomarkers and pharmacokinetic (PK) data one collects in early to middle drug development can be used to predict the dose and treatment response of promising therapeutics in definitive phase 3 trials.

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call