Clinical Pharmacology and Therapeutics. Группа авторов

Чтение книги онлайн.

Читать онлайн книгу Clinical Pharmacology and Therapeutics - Группа авторов страница 27

Clinical Pharmacology and Therapeutics - Группа авторов

Скачать книгу

III clinical trials

      During Phase III, the regulatory affairs department within companies will be pulling together the large amount of manufacturing, preclinical and clinical data necessary for making a formal application to the relevant regional and national regulatory authorities for a product licence. Each major regulator requires the data structured in a different way, therefore first priority will usually be given to submissions to the FDA and the EMA. Review times by these regulators vary based on circumstances, but it usually takes approximately 1 year.

      Based on the data submitted, each regulatory authority will produce a factual summary of the preclinical and clinical results, including the key safety information and dosing instructions. This document will also state whether the marketing approval is general or restricted, e.g. hospital use only. The relevant document issued by the EMA is called the summary of product characteristics (SPC) and provides the key information required to aid a decision by the prescriber as to whether the drug is indicated. A second valuable document is the European Public Assessment Report (EPAR), found on the EMA website, which provides a more detailed summary of the Agency's review of the data submitted. Over time, the SPC will be updated by the company as key new information becomes available, but clinical publications and treatment guidelines are also invaluable in providing additional detail which will not be found in the SPC.

      In the UK, each new drug in the British National Formulary has an inverted triangle symbol next to it reminding doctors that it is a new product and that any suspected adverse effects should be reported to the Commission on Human Medicines (CHM) via the yellow card scheme.

      Factors to consider in Phase III trial design

      The main objective of a Phase III trial is to establish the effect of a new drug while controlling for bias and confounding. This is achieved by randomly allocating trial participants to treatment groups, ensuring blinding is performed where possible, having a representative control group and analysing results on an intention to treat basis. Stratified randomisation is a technique that can be used to balance groups with respect to confounding variables such as age, although it is important to avoid over‐stratification as the trial must include an adequate number of patients for each arm and stratum. Blinding study participants, investigators and/or assessors to allocated drugs helps to reduce bias, although this must be balanced with practicality and cost. For example, in a trial of thrombolytic therapy for acute ischaemic stroke, the drug alteplase is being compared with another drug tenecteplase. Alteplase is administered as an intravenous bolus followed by a 1 hour infusion, whereas tenecteplase is administered only as a bolus, which makes blinding of investigators and participants challenging. The options in this setting are to accept the limitation of only blinding final assessors of the trial data or employing a ‘double‐dummy’ approach that utilises a placebo for each intervention, although the latter approach is often less practical and more expensive.

      It is important to carefully select the study population and endpoints to ensure that the population is representative of the target patient group who may benefit from the drug and that the endpoints are of sufficient clinical relevance to justify drug licencing if proven to be effective. The primary endpoint should therefore be clinically relevant, measurable and potentially sensitive to the effects of the new drug. The trial should aim to have a high proportion of subjects with complete data and the results should generally be interpreted on the basis of intention‐to‐treat (all patients who entered the study) rather than per‐protocol (only the patients who completed the protocol). A large dropout rate may indicate a drug that is unlikely to be commercially successful. Additionally, it is also important to ensure that the sample size is large enough for the trial to be sufficiently powered to detect a meaningful difference between the drugs if one exists and avoid a type II error where an effective drug is incorrectly rejected due to the trial being underpowered.

      Newer approaches to Phase III clinical trial design

      An illustration of a magnifying glass placed inside a shaded circle. Clinical scenario

      The new drug compound for neuroprotection has now been tested in Phase III clinical trials and has shown good efficacy with an acceptable side‐effect profile. It gets approved by the regulatory authorities and the company wants to start marketing the drug. Is there a limit to what the company can do in its marketing strategy? How is its safety monitored? What happens if concerns about side effects become apparent?

      Phase IIIb and IV studies

      Phase III trials will generally have included selected groups of patients based on strict inclusion and exclusion criteria that will form the basis of its licenced indications and the product can only be marketed on this basis. For this reason, further studies will often be performed to either widen the licence indications to broader groups of patients such as the elderly (Phase IIIb) or evaluate drug safety and efficacy in a real‐world setting, over the longer term or against comparators, often involving many thousands of patients (Phase IV). It is important to emphasise that even though a drug may obtain a licence, it does not necessarily mean that there will be widespread use in patients, as many countries also mandate a health economic evaluation to ascertain its cost‐effectiveness (see Chapter 3).

      Pharmacovigilance

      There is now a greatly increased obligation on pharmaceutical companies to set up risk‐management

Скачать книгу