We use cookies to distinguish you from other users and to provide you with a better experience on our websites. Close this message to accept cookies or find out how to manage your cookie settings.
To save content items to your account,
please confirm that you agree to abide by our usage policies.
If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account.
Find out more about saving content to .
To save content items to your Kindle, first ensure [email protected]
is added to your Approved Personal Document E-mail List under your Personal Document Settings
on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part
of your Kindle email address below.
Find out more about saving to your Kindle.
Note you can select to save to either the @free.kindle.com or @kindle.com variations.
‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi.
‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.
The life expectancy for people with intellectual disability is increasing due to advances in medical treatment and social care. However, significant discrepancies in life expectancy between people with intellectual disability and the general population remain, and there continues to be scope to close the inequality gap. This was confirmed in the recent 2021 Learning Disability Mortality Review (LeDeR) report. The standardised mortality ratio for people with intellectual disability ranges from 2–5, which draws a comparison against the general population. Those with additional comorbidities such as epilepsy, genetic syndromes, and functional impairments have a lower age of death. The leading causes of death in older adults (at or over 65 years of age) between 2018 and 2021 were reviewed in the 2021 LeDeR report. In comparison to the general population, a higher proportion of deaths in older intellectual disability adults were due to COVID-19 (coronavirus disease), cancers, and influenza or pneumonia. Unsurprisingly, dementia (in particular Alzheimer’s disease), cerebrovascular disease, chronic lower respiratory tract infections, and diseases of the urinary system were more common causes of death in older intellectual disability compared to that reported in their younger counterparts. This chapter explores the various issues associated with medicating older people.
Many paediatric studies report that patients must be established on aspirin therapy for a minimum of 5 days to achieve adequate response. This is not always practical especially in critical settings. Prospective identification of patients that are unresponsive to aspirin sooner could potentially prevent thrombotic events.
Aims:
The aim of this study was to investigate prospectively if the first dose of aspirin is effective in decreasing platelet aggregation, and thromboxane formation and if this can be measured after 2 hours in paediatric cardiology patients. A secondary aim was to identify a cut-off for a novel marker of aspirin responsiveness the maximum amplitude with arachidonic acid, which could potentially dramatically reduce the blood volume required. Third, we aimed to prospectively identify potentially non-responsive patients by spiking a sample of their blood ex vivo with aspirin.
Results:
The majority (92.3%) of patients were responsive, when measured 2 hours post first dose of aspirin. Non-response or inadequate response (7.7%) can also be identified at 2 hours after taking the first dose of aspirin. Additionally, we have shown a novel way to reduce blood sample volume requirements by measurement of the maximum amplitude with arachidonic acid as a marker of response, particularly for monitoring.
Conclusions:
These findings of rapid efficacy in the majority of patients offer assurance in a sound, practical way to attending clinicians, patients, and families.
The absorption and distribution of radiocarbon-labeled urea at the ultratrace level were investigated with a 14C-AMS biotracer method. The radiopharmaceutical concentrations in the plasma, heart, liver, spleen, lung, kidney, stomach, brain, bladder, muscle, testis, and fat of rats after oral administration of 14C urea at ultratrace doses were determined by AMS, and the concentration-time curves in plasma and tissues and pharmacokinetic distribution data were obtained. This study provides an analytical method for the pharmacokinetic parameters and tissue distribution of exogenous urea in rats at ultratrace doses and explores the feasibility of evaluation and long-term tracking of ultratrace doses of drugs with AMS.
The ongoing pandemic, COVID-19 (SARS-CoV-2), has afflicted millions of people around the world, necessitating that the scientific community work, diligently and promptly, on suitable medicaments. Although vaccination programs have been run globally, the new variants of COVID-19 make it difficult to restrict the spread of the virus by vaccination alone. The combination of vaccination with anti-viral drug formulation is an ideal strategy for tackling the current pandemic situation. Drugs approved by the United States Food and Drug Administration (FDA), such as Remdesivir, have been found to be of little or no benefit. On the other hand, re-purposing of FDA-approved drugs, such as niclosamide (NIC), has offered promise but its applicability is limited due to its poor aqueous solubility and, therefore, low bioavailability. With advanced nano-pharmaceutical approaches, re-purposing this drug in a suitable drug-carrier for a better outcome may be possible. In the current study, an attempt was made to explore the loading of NIC into exfoliated layered double hydroxide nanoparticles (X-LDH NPs); prepared NIC-X-LDH NPs were further modified with eudragit S100 (ES100), an enteric coating polymer, to make the final product, ES100-NIC-X-LDH NPs, to improve absorption by the gastro/intestinal tract (GIT). Furthermore, Tween 60 was added as a coating on ES100-NIC-X-LDH NPs, not just to enhance its in vitro and in vivo stability, but also to enhance its mucoadhesive property, and to obtain, ultimately, better in vivo pharmacokinetic (PK) parameters upon oral administration. Release of NIC from Tween 60-ES100-NIC-X-LDH NPs was found to be greater under gastro/intestinal solution within a shorter period of time than the uncoated samples. The in vivo analysis revealed that Tween 60-ES100-NIC-X-LDH NPs were able to maintain a therapeutically relevant NIC plasma concentration in terms of PK parameters compared to the commercially available Yomesan®, proving that the new formulation might prove to be an effective oral drug-delivery system to deal with the SARS-CoV-2 viral infections. Further studies are required to ensure their safety and anti-viral efficacy.
Edited by
Xiuzhen Huang, Cedars-Sinai Medical Center, Los Angeles,Jason H. Moore, Cedars-Sinai Medical Center, Los Angeles,Yu Zhang, Trinity University, Texas
Pharmacogenomics is the study of genetic factors that influence drug response. Pharmacogenomics combines pharmacology and genomics to identify genetic predictors of variability in drug response that can be used to maximize drug efficacy while minimizing drug toxicity in order to tailor drug therapy for patients, thus improving patient care and reducing healthcare costs. In this chapter we review the field of pharmacogenomics in its current state and clinical practice. Recent research, methods, and resources for pharmacogenomics are reviewed in detail. We discuss the advantages and challenges in pharmacogenomic studies. We elaborate on the barriers to clinical translation of pharmacogenetic discoveries and the efforts of various institutions and consortia to mitigate these barriers. We also discuss applications and clinical translation of pharmacogenomic research moving forward, along with social, ethical, and economic issues that require attention. We conclude by previewing the use of big data, multi-omics data, advanced computing technology, and statistical methods by scientists across disciplinary boundaries along with the efforts of government organizations, clinicians, and patients that could lead to successful and clinically translatable pharmacogenomic discoveries, ushering in an era of precision medicine.
Edited by
Deepak Cyril D'Souza, Staff Psychiatrist, VA Connecticut Healthcare System; Professor of Psychiatry, Yale University School of Medicine,David Castle, University of Tasmania, Australia,Sir Robin Murray, Honorary Consultant Psychiatrist, Psychosis Service at the South London and Maudsley NHS Trust; Professor of Psychiatric Research at the Institute of Psychiatry
Cannabis produces its characteristic intoxicating effect through its actions with specific receptors in the brain. This chapter will explore what we know how about cannabis produces these effects. Since cannabis produces its effects by interacting with the endocannabinoid system, we will start with a brief consideration of the endocannabinoid system relevant to cannabis’ actions. This will be followed by a discussion of the primary psychoactive components of cannabis, their routes of administration, and pharmacokinetics. The next section will focus on how tetrahydrocannabinol interacts with CB1 cannabinoid receptors and how these interactions differ from endocannabinoid interactions with CB1 receptors. Finally, these results will be synthesized in a potential explanation on how cannabis works in the brain.
Precision medicine is an approach to maximise the effectiveness of disease treatment and prevention and minimise harm from medications by considering relevant demographic, clinical, genomic and environmental factors in making treatment decisions. Precision medicine is complex, even for decisions about single drugs for single diseases, as it requires expert consideration of multiple measurable factors that affect pharmacokinetics and pharmacodynamics, and many patient-specific variables. Given the increasing number of patients with multiple conditions and medications, there is a need to apply lessons learned from precision medicine in monotherapy and single disease management to optimise polypharmacy. However, precision medicine for optimisation of polypharmacy is particularly challenging because of the vast number of interacting factors that influence drug use and response. In this narrative review, we aim to provide and apply the latest research findings to achieve precision medicine in the context of polypharmacy. Specifically, this review aims to (1) summarise challenges in achieving precision medicine specific to polypharmacy; (2) synthesise the current approaches to precision medicine in polypharmacy; (3) provide a summary of the literature in the field of prediction of unknown drug–drug interactions (DDI) and (4) propose a novel approach to provide precision medicine for patients with polypharmacy. For our proposed model to be implemented in routine clinical practice, a comprehensive intervention bundle needs to be integrated into the electronic medical record using bioinformatic approaches on a wide range of data to predict the effects of polypharmacy regimens on an individual. In addition, clinicians need to be trained to interpret the results of data from sources including pharmacogenomic testing, DDI prediction and physiological-pharmacokinetic-pharmacodynamic modelling to inform their medication reviews. Future studies are needed to evaluate the efficacy of this model and to test generalisability so that it can be implemented at scale, aiming to improve outcomes in people with polypharmacy.
Understanding the transfer of polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) as well as polychlorinated biphenyls (PCBs) from oral exposure into cow’s milk is not purely an experimental endeavour, as it has produced a large corpus of theoretical work. This work consists of a variety of predictive toxicokinetic models in the realms of health and environmental risk assessment and risk management. Their purpose is to provide mathematical predictive tools to organise and integrate knowledge on the absorption, distribution, metabolism and excretion processes. Toxicokinetic models are based on more than 50 years of transfer studies summarised in part I of this review series. Here in part II, several of these models are described and systematically classified with a focus on their applicability to risk analysis as well as their limitations. This part of the review highlights the opportunities and challenges along the way towards accurate, congener-specific predictive models applicable to changing animal breeds and husbandry conditions.
Fisetin, a polyphenol found in several fruits and vegetables, has shown potential health benefits in many pre-clinical studies for neuroprotection, cardioprotection, chemoprevention, diabetes, inflammation and oxidative stress. However, the clinical effectiveness of fisetin may be limited by its poor bioavailability when ingested. Using a novel green technology of Hybrid-FENUMAT™, a food-grade fisetin formulation (FF-20) was developed through encapsulation of fisetin micelles into fenugreek galactomannan (FG) hydrogel scaffold to improve its physical characteristics and bioavailability. This is the first human pharmacokinetic study of fisetin following a single-dose, comparative, double-blinded, cross-over protocol, supplementing with FF-20 and unformulated fisetin (UF). Fifteen healthy volunteers were given a single dose of 1000 mg UF or 1000 mg FF-20 (delivering 192 mg fisetin) with a 10-d washout period between each dose. Blood samples were taken at 0⋅5, 1, 2, 3, 5, 8 and 12 h after both days of supplementation to quantify fisetin and geraldol, an active metabolite. The plasma concentration of fisetin when individuals consumed FF-20 was 26⋅9-fold greater than UF as determined by the area under the curve over 12 h [AUC0–12 h (FF-20) = 341⋅4 v. AUC0–12 h (UF) = 12⋅67]. The maximum plasma concentration (Cmax) was also more than twenty-three times higher when supplemented with FF-20 (238⋅2 ng/ml) compared to UF (9⋅97 ng/ml). The encapsulation also reduced the amount of conversion of fisetin to geraldol. No adverse events were reported during the study. Therefore, the encapsulation of fisetin into FG dietary fibre hydrogel scaffold could improve its delivery and bioavailability in human subjects.
Safe and effective medication use in the elderly requires heightened awareness in comparison to other patient populations. The issues of increased sensitivity to drug effects, use of many medications to treat comorbidities, and a high incidence of medication nonadherence increase the need for due diligence in prescribing and monitoring drug therapy in this population. The employment of an interprofessional approach to patient care as well as the use of known methods for improving medication adherence are key to long-term success in the medical management of the elderly patient.
Phase 1 clinical trials are the entrance to the further clinical development of new compounds. The chapter describes the regulatory background and highlights most important issues about selection of the maximum recommended starting dose, dose escalation steps, and definition of maximum tolerated dose, or maximum applied dose in a study considering actual guidelines. There is an overview about selection of subject populations and frequently used trial designs. The principles of single-ascending-dose and multiple-ascending-dose tolerance studies are described with a few examples of studies in Alzheimer’s disease (AD). The safety assessment is important in clinical practice, as AD drugs will be used over many years, so excellent tolerability is a must! In Phase 1, a careful assessment of pharmacokinetic (PK) properties of a new compound forms the basis for dose selection in Phase 2 and 3 studies and supports the decision on the treatment regimen. The importance of inclusion of different biomarkers in these studies to allow assessment of pharmacodynamic and PK relationship and to potentially identify first signals in human studies indicating therapeutic usefulness in the indication.
There is a tremendous need for disease-modifying treatments for Alzheimer’s disease (AD). The pharmaceutical sector has expended considerable resources on AD drug discovery, yet to date have obtained regulatory approval for only one agent that slows AD progression. This has led to increased interest in identifying new AD drug targets and disease mechanisms. Academic laboratories can play a meaningful role in the validation of AD drug targets and the identification of molecular probes that modulate these targets. We discuss here how academic researchers can contribute to the AD drug discovery process. This includes examples of assays that have been used for AD small molecule screens within academic laboratories, and discussions on assay optimization for compound screening, the selection of molecular libraries, and the iterative process of compound optimization to identify molecules suitable for advancement to in vivo pharmacokinetic, safety, and efficacy testing. Finally, we outline how academic researchers might work with pharmaceutical partners in AD drug discovery, and note the pros and cons of such collaborations.
There are many challenges of proper drug dosing with nanodelivery systems. The first part of this discussion concerns how experts think about drug dosing with conventional drugs. In the second part, we need to consider the differences between nanodevices and traditional drug delivery, and pharmacokinetics using nanodrug delivery. Drug dosing uses scale-up methods from animal model data before testing on humans. New organ-on-a-chip and human-on-a-chip technologies may someday replace animal data.
Multi-compartment models described by systems of linear ordinary differential equations are considered. Catenary models are a particular class where the compartments are arranged in a chain. A unified methodology based on the Laplace transform is utilised to solve direct and inverse problems for multi-compartment models. Explicit formulas for the parameters in a catenary model are obtained in terms of the roots of elementary symmetric polynomials. A method to estimate parameters for a general multi-compartment model is also provided. Results of numerical simulations are presented to illustrate the effectiveness of the approach.
Coenzyme Q10 (CoQ10), a lipid involved in ATP synthesis, exhibits very limited oral absorption, and its endogenous production decreases with ageing and with the occurrence of oxidative stress. Our group previously showed that monoglycerides omega-3 (MAG-OM3) increase OM3 plasma concentrations. Since CoQ10 is liposoluble, we hypothesised that its 48 h pharmacokinetics is higher when provided with MAG-OM3 compared to CoQ10 alone (in powder form) or added to rice oil (a neutral triacylglycerol oil). A randomised triple-blind crossover study was performed with fifteen men and fifteen women consuming the three supplements providing 200 mg of CoQ10 in a random order. Blood samples were collected before (t = 0) and 1, 3, 5, 6, 7, 8, 10, 11, 24 and 48 h after the supplement intake. Plasma total CoQ10 concentrations were analysed on ultrahigh-performance liquid chromatography coupled to a tandem mass spectrometer (UPLC-MS/MS). Participants were 26⋅1 ± 4⋅8 years old. When CoQ10 was provided with rice or MAG-OM3 oils, the 48 h area under the curve (AUC 0–48 h) was approximately two times higher compared to when provided without an oil. The delta max concentration (ΔCmax) of plasma CoQ10 was, respectively, 2 (MAG-OM3) and 2⋅5 (rice oil) times higher compared to CoQ10 alone. There was a significant sex by treatment interaction (P = 0⋅0250) for the AUC 0–6 h supporting that in postprandial, men and women do not respond the same way to the different supplement. Women had a higher CoQ10 concentration 48 h after the single-dose intake compared to men. We conclude that CoQ10 supplements must be provided with lipids, and their kinetics is different between men and women.
This chapter provides a concise overview of the key principles of pharmacology that prescribers should be familiar with in their day-to-day prescribing practice. It includes sections on pharmacokinetics, pharmacodynamics, drug interactions and adverse drug reactions. This third edition includes updated examples and reference to deprescribing in relation to applied pharmacology, as well as information on biosimilars and bioequivalence
1. Poor anti-microbial stewardship and selection pressure encourage antibiotic resistance.
2. Pharmacokinetics may be altered significantly and unpredictably by critical illness.
3. Optimal dosing of antibiotics is difficult. Many patients are likely under-dosed.
4. Different antibiotics require different approaches to achieving optimal antibiotic exposure.
5. Therapeutic drug monitoring is standard for aminoglycosides and glycopeptides, but may be helpful for β-lactams, linezolid and some ‘azole’ anti-fungal agents.
1. Understanding how the patient will handle a drug (pharmacokinetics) and how the drug will affect the patient (pharmacodynamics) is crucial to prescribing any drug safely.
2. It is important to consider both how the drugs may interact with other drugs being co-administered and how their effect/handling may be affected by the patient’s critical illness and associated organ dysfunction.
3. The loading dose, repeat doses and/or dosing interval may need to be adjusted, and in some cases monitoring of drug levels may be possible/necessary.
4. Critical illness is a rapidly changing dynamic state. Daily medication chart review, considering if drugs are still indicated and/or if their dosing needs reviewing, depending on the changing clinical condition of the patient, is essential.
5. Prescribing in critical illness can be a complex area; however, there are comprehensive resources available to guide practice. Seeking support and advice from your pharmacist when in doubt is a very useful and sensible approach.
There are significant differences between men and women in the efficacy and tolerability of antipsychotic drugs. Here, we provide a comprehensive overview of what is currently known about the pharmacokinetics and pharmacodynamics of antipsychotics in women with schizophrenia spectrum disorders (SSDs) and translate these insights into considerations for clinical practice. Slower drug absorption, metabolism and excretion in women all lead to higher plasma levels, which increase the risk for side-effects. Moreover, women reach higher dopamine receptor occupancy compared to men at similar serum levels, since oestrogens increase dopamine sensitivity. As current treatment guidelines are based on studies predominantly conducted in men, women are likely to be overmedicated by default. The risk of overmedicating generally increases when sex hormone levels are high (e.g. during ovulation and gestation), whereas higher doses may be required during low-hormonal phases (e.g. during menstruation and menopause). For premenopausal women, with the exceptions of quetiapine and lurasidone, doses of antipsychotics should be lower with largest adjustments required for olanzapine. Clinicians should be wary of side-effects that are particularly harmful in women, such as hyperprolactinaemia which can cause oestrogen deficiency and metabolic symptoms that may cause cardiovascular diseases. Given the protective effects of oestrogens on the course of SSD, oestrogen replacement therapy should be considered for postmenopausal patients, who are more vulnerable to side-effects and yet require higher dosages of most antipsychotics to reach similar efficacy. In conclusion, there is a need for tailored, female-specific prescription guidelines, which take into account adjustments required across different phases of life.
Smoking is highly prevalent in the psychiatric population, and hospital admittance usually results in partial or complete smoking cessation. Tobacco use is known to affect the metabolism of certain psychoactive drugs, but whether smoking influences the plasma concentration of tricyclic antidepressants (TCAs) remains unclear. This article investigates the possible effect of smoking on the plasma concentration of TCAs. A systematic review of the literature available on PubMed and EMBASE as of October 2020 was carried out using PRISMA guidelines. Studies reporting plasma concentrations of any TCA in both a smoking and a non-smoking group were included and compared. Ten eligible studies were identified and included. In the eight studies investigating the effect of smoking on amitriptyline and/or nortriptyline, five studies found no significant effect. Two studies investigating the effect of smoking on imipramine found a significant effect, and one study investigating the effect of smoking on doxepin found no significant effect. The majority of studies included in this review were influenced by small study populations and other methodical issues. The effect of smoking on the plasma concentration of TCAs is still not entirely clear. There is a possibility that smoking affects the distribution of TCA metabolites, but this is probably not of clinical importance.