Friday, March 29, 2019

Pharmacogenetics And Pharmacogenomics In Pharmacy Practice Biology Essay

Pharmaco heritables And Pharmacogenomics In Pharmacy Practice Biology EssayThe learning of the action among familials and healing(p) medicates is dissimilarly c completelyed pharmaco hereditarys or pharmacogenomics. The differences between the two are the initial overture of the sciencePharmaco contractables starts with an unexpected dose reception result and looks for a heritable vitrine.Pharmacogenomics, on the some other hand, begins with expression for genetic differences at bottom a population that explain certain notice answers to a do do drugss or susceptibility to a health line of work (The Australasian genetics Resource Book, 2007).Pharmacogenetics refers to the study of inter-individual specific genetic model (Zika et al. 2006). The term pharmacogenetics is occasion eachy use in a limiting awareness to describe how different gene patterns affect drug-response just now it dope likewise be defined more broadly as the study of the violence of her edity on benignant drug-response (Newton et al. 2007).Factors that shape how an individual responds to medication embroil their external and internal environments and general health, as head as their genetic cook up. The goal of pharmacogenetics is to understand the role that an individuals genetic fundamental law feeds in how well a treat works, as well as what office effect are likely to drop dead in the individuals body.Understanding this rout out economic aid tailor drugs in the future best suited for a specific individual (personalised medicine) or group (The Australian Genetics Resource Book, 2007). The sm bother differences in the genes between different population groups, or some families within a population group, that necessitate built up over the generations can plastered that they react differently to medicines.However, some diseases, notably cancers, develop in cells which have an altered genetic constitution, so that the genetic make-up of the morbi d tissue is no longer the said(prenominal) as that of the person in which it is present. Specific genes present in the diseased tissue whitethorn play a critical role in determining the optimum interposition. To represent this, it provide thitherfore be necessary to identify the genetic make-up of the cancer itself try oning the uncomplaining before a cancer has genuine is of no use, because the genetic changes are solitary(prenominal) present in the cancer cells and not in the normal host tissues (Nuffield Council on Bioethics, 2003).Some authorisation realizes of pharmacogenetics include the undermentionedMore powerful medicines drugs whitethorn be real targeting specific health problems that entrust maximise therapeutic effects but decrease damage to nearby healthy cellsSafer drugs the early time Doctors could have an idea which drug to use found on a genetic profile versus trial and error, decreasing the likelihood of uncomely responsesMore entire methods of determining loony toonss Instead of dosages being based on body tilt and age, it would be based on an individuals genetics. This would decrease the likelihood of an overdose.Better vaccines Vaccines make of genetic material could activate the immune system to have all the benefits of existing vaccines but with reduced risks of infections (The Australasian Genetics Resource Book, 2007).Implications of pharmacogenetics in practice are vast and encompass broad areas much(prenominal) asDrug response the effects of angiotensin converting enzyme inhibitors have been found to be greater in the great unwashed of European or UK ancestry than African-Americans.Pre- interference genetic screening of patients allow eventually enable this knowledge to be utilise in clinical practice. Moreover, play in the genes that code for receptors (drug targets) may mean that some people may produce receptors that do not interact well with the drug. For example, some people have a lack of response to the drug salbutamol, use in the treatment of asthma, due to genetic variation in the gene that codes a receptor on the surface of good-tempered muscle cells lining airways of the lungs.Drug targets Genes may also watch over how umpteen of the receptors are produced on or within cells and genetic variation may mean that some people produce more of these sites than others. The action of the wide apply antipsychotic drug haloperidol (Haldol) depends on its ability to restrain to the dopamine (D2) receptor site. In one study, 63% of patients whose genetic make-up caused a large flesh of these receptor sites to be produced had a response to treatment with haloperidol. About 29% of patients with a smaller number of dopamine (D2) receptor sites responded well to the drug.Drug metamorphosis Pain relief medications much(prenominal) as codeine require an enzyme produced in the liver called CYP2D6 for the drug to be used by the body, break it vanquish and remove it. Variations in t he information contained in the CYP2D6 gene determine how much of this enzyme is produced in the liver (The Australasian Genetics Resource Book, 2007). The implication of variations in genotype on the metabolism of the immunosuppressor azathioprine is also an example. Polymorphisms in the gene encoding for the enzyme thiopurine S-methyl transferase (TPMT) lead to changes in the activity of the enzyme and rate of metabolism of azathioprine. Changes in the activity of the enzyme present clinically as an attachd risk of neutropenia or a decreased chance of responding to azathioprine, at normal dose ranges. A genetic test for the polymorphism can identify individuals who are more likely to develop neutropenia. Thus, the prey of a pharmacogenetic test here is to minimise an uncomely effect, although in other cases, a pharmacogenetic test may be able to predict an rough-and-ready responseto a medicine by correlating an individuals genotype with the observed pharmacological actions of medicines (phenotype) (Clemerson et al. 2006).Drug development Excluding from clinical trials those people whose genetic makeup would make the drug being tested harmful or ineffective for them leave alone increase the chance that a drug will show itself useful to a particular population group. This would increase the chance that the same drug will make it into the marketplace. Undertaking pre-genetic screening of those patients taking part in a clinical trial should also make the clinical trials smaller, faster, and therefore slight expensive. For example, as seen in clinical trials for developing drugs for Alzheimer disease and other forms of madness (The Australasian Genetics Resource Book, 2007).The application of pharmacogenetics has two main aspects improvements in the arctic and force of medicines. In improving estimablety, pharmacogenetics works in the following waysPharmacogenetic tests reveal genetic variations already known to be associated with adverse reaction s, allowing physicians to avoid exposing patients to medicines that would put them at risk. The majority of adverse reactions are caused because of an magnified effect of a medicine in the body. Less much, an adverse reaction may be an idiosyncratic response to the medicine.Adverse reactions to medicines have meaning(a) cost, in both human and monetary terms. However, it is difficult to ascertain the preserve of genetic variation in response to medicines because data concerning adverse reactions often include problems caused by errors in prescription, and because information about other causes much(prenominal) as interaction between different medicines may be non-existent.Results from pharmacogenetic tests may also inform physicians in strikeing the medicine nigh likely to benefit a particular patient. Many medicines are effective in only a proportion of patients treated. Sometimes, for a medicine to be effective, different doses are required for different patients. In the ab sence of a pharmacogenetic test for efficacy, the most get hold of medicine or dose is conventionally found by trial and error, although in some cases, tests of renal function may be used to predict the take into account dose. It has been suggested that a trial and error approach to prescription may reduce compliance for medicines that do work, since patients acquire a general aversion to taking medicines because of the unpleasant side-effects which they skill experience. This therefore helps in improving efficacy of medicines (Nuffield Council on Bioethics, 2003).A potential barrier to the development of pharmacogenetic tests concerns the application of intellectual property rights. Pharmacogenetic tests may be developed in a number of ways. The pharmaceutical company which is developing the medicine may also develop the pharmacogenetic test. Alternatively, a third party, such as other company or researchers from the public sector may develop the test independently. stillmore, while the effect of pharmacogenetics may be to reduce some of the costs of developing new medicines, it would be imprudent to infer from this that the cost of buying medicines will necessarily fall (Nuffield Council on Bioethics, 2003).PharmacogenomicsPharmacogenomics is the study of genetic variations that influence individual response to drugs. Knowing whether a patient carries any of these genetic variations can help prescribers individualise drug therapy, decrease the chance for adverse drug events, and increase the effectiveness of drugs (AMA, 2013). Pharmacogenomics holds the promise that drugs might be tailor-make for individuals and adapted to each persons own genetic makeup. Environment, diet, age, lifestyle, and state of health all can influence a persons response to medicines, but understanding an individuals genetic makeup is thought to be the key to creating personalised drugs with greater efficacy and safety.Pharmacogenomics combines traditional pharmaceutical scienc es such as biochemistry with annotated knowledge of genes, proteins, and single base polymorphisms (Human Genome vomit Information, 2011).The field of pharmacogenomics is still in its infancy. Its use is currently sort of limited, but new approaches are under study in clinical trials. In the future, pharmacogenomics will allow the development of tailored drugs to treat a wide range of health problems, including cardiovascular disease, Alzheimer disease, cancer, HIV/AIDS, and asthma (Genetics house Reference, 2013). The cytochrome P450 (CYP) family of liver enzymes is responsible for breaking down more than 30 different classes of drugs. DNA variations in genes that code for these enzymes can influence their ability to metabolise some drugs. Less active or smooth forms of CYP enzymes that are unavailing to break down and properly eliminate drugs from the body can cause drug overdose in patients. Clinical trials researchers use genetic tests for variations in cytochrome P450 ge nes to screen and monitor patients. In addition, many pharmaceutical companies screen their chemical substance compounds to see how well they are broken down by variant forms of CYP enzymes.Another enzyme called TPMT (thiopurine methyltransferase) plays an important role in the chemotherapy treatment of common childishness leukemia by breaking down a class of therapeutic compounds called thiopurines. A small percentage of Caucasians have genetic variants that prevent them from producing an active form of this protein. As a result, thiopurines elevate to toxic levels in the patient because the inactive form of TMPT is unable to break down the drug. Today, doctors can use a genetic test to screen patients for this deficiency, and the TMPT activity is monitored to determine appropriate thiopurine dosage levels (Human Genome Project Information, 2011).Similarly to pharmacogenetics, pharmacogenomicshas the potential toprovide tailored drug therapy based on genetically determined variati on in effectiveness and side effects (AMA, 2013). This will meanMore powerful medicines Pharmaceutical companies will be able to produce therapies more targeted to specific diseases, maximising therapeutic effects while decreasing damage to nearby healthy cells.Better, safer drugs the first time Recovery time will go down and safety will go up as the likelihood of adverse reactions goes down or is eliminated altogether. Improvements in drug discovery, design, and development are obvious applications for pharmacogenomics. A deeper understanding of the genetic factors which cause variance in drug metabolism can aid in the design of drugs with improved potency, reduced toxicity, and fewer side effects. For example, pharmacogenomics can identify potential drug targets (targets are typically enzymes or other proteins), and determine which targets are least prone to genetic variance. By selecting drug targets which are not prone to genetic variance, drug designers can create drugs which a re more likely to have standard, expected, and safe reactions in people who take it.More accurate methods of determining appropriate drug dosages Current methods of basing dosages on weight and age will be replaced with dosages based on a persons genetics how well the body exercisees the medicine and the time it takes to metabolise it.Pharmacogenomics can also be useful in clinical trials for drugs which have passed through the approval process sufficiently that human trials are possible.Using this approach, a technique called genostratification can be used in selecting participants for clinical trials. This means that clinicians use genetic typing to select participants who are genetically more likely to react positively to the treatment which is under study.This can potentially allow for an improved level of treatment success, and means that proof of concept can be achieved sooner. This technique can also allow for a reduction in the required smack size for the trial, or shorte ned trial duration. Ultimately, a drug which may help save or improve lives can be used in the general public more quickly than otherwise would be possible.Economic issues from molecule to marketplace Pharmacogenomics eventually can lead to an overall decrease in the cost of health care because of decreases inthe number of adverse drug reactions,the number of failed drug trials,the time it takes to get a drug approved,the length of time patients are on medication,the number of medications patients essential take to find an effective therapy, andthe effects of a disease on the body (through early detection).Applying pharmacogenomics to patient treatment can help word individualised treatment regimes, to ensure that patients receive the drugs which are most appropriate for their genetic makeup.In particular, this approach has significant potential in treating cancer, because there is a great degree of variance in the way people react to chemotherapy drugs. Tumors themselves are hig hly variable in genetic terms, and this part accounts for the variance in drug responses. Using an approach which individualizes treatment regimes, to match for this variance could improve cancer treatments significantly.Pharmacogenomics is useful in general for patient treatment because it has the potential to identify on an individual basis the drugs which might cause adverse reactions. A person who might experience such a reaction can then be prescribed an substitute(a) drug (Lloyd, 2008).However, there are several potential barriers to pharmacogenomics which have to be overcome before the above discussed benefits of pharmacogenomics can be realised (Human Genome Project Information, 2011). These include the followingComplexity of finding gene variations that affect drug response Single nucleotide polymorphisms (SNPs) are DNA sequence variations that occur when a single nucleotide (A,T,C,or G) in the genome sequence is altered. SNPs occur all(prenominal) 100 to 300 bases alon g the 3-billion-base human genome, therefore millions of SNPs must be identified and analyzed to determine their involvement (if any) in drug response. Further complicating the process is our limited knowledge of which genes are involved with each drug response. Since many genes are likely to influence responses, obtaining the big picture on the impact of gene variations is highly time-consuming and complicated.Limited drug alternatives tho one or two approved drugs may be purchasable for treatment of a particular condition. If patients have gene variations that prevent them utilise these drugs, they may be left without any alternatives for treatment.Disincentives for drug companies to make sextuple pharmacogenomic products Most pharmaceutical companies have been successful with their one size fits all approach to drug development. Since it costs hundreds of millions of dollars to bring a drug to market, will these companies be willing to develop alternative drugs that serve onl y a small portion of the population?Educating healthcare providers Introducing multiple pharmacogenomic products to treat the same condition for different population subsets undoubtedly will complicate the process of prescribing and dispensing drugs. Physicians must execute an extra diagnostic step to determine which drug is best suited to each patient. To interpret the diagnostic accurately and press the best course of treatment for each patient, all prescribing physicians, regardless of specialty, will need a better understanding of genetics.ConclusionDespite the various potential barriers to both pharmacogenetics and pharmacogenomics, these fields are rapidly evolving with the promise that someday a simple and rapid DNA test will determine potential risks of adverse effects with a certain drug, and thus bout to another drug which would be more suitable for the patient.

No comments:

Post a Comment

Note: Only a member of this blog may post a comment.