Pharmacogenomics – an overview | ScienceDirect Topics

Clinical Implementation of Pharmacogenomics

Pharmacogenomics has the potential to influence clinically relevant outcomes in drug dosing, efficacy, and toxicity that can result in subsequent recommendations for testing. For many routinely used drugs, pharmacogenomics has provided inconclusive evidence for such testing. A probable reason could be the involvement of both genetic and nongenetic factors and their extent of contribution that determines the clinical relevance of some drugs. Therefore, identification of genetic markers associated with drug responses does not always link to clinically useful predictors of adverse outcomes, and most of the time require independent replication of genotypephenotype association before pursuing clinical implementation.

Lack of readily available resources, feasibility, utility, level of evidence, provider knowledge, cost effectiveness, and ethical, legal, and social issues further adds to the limitations and challenges to implementing pharmacogenomic testing in clinical practice. In order for a genetic marker to be implicated in clinical practice, an association of a genetic marker to a particular trait requires screening of tissues from several individuals, and corresponding functional studies are needed to establish probable association with the trait/phenotype. However, to overcome these challenges there are some pharmacogenomic tests for drugs currently used in clinical practice that have applied value in predicting ADRs and/or drug efficacy. Table 7.2 lists some of these clinically valuable pharmacogenomics tests. These tests are based on distinct genetic variants that have well-validated reproducible and significant impact on the drug therapy. These tests have a strong causal association between genetic polymorphisms and drug responses: a strong indication for clinical utility and high prognostic value. The tests are available both commercially and in academic settings, with many of these tests having clinical guidelines for dose adjustment and alternative medications (Wei et al., 2012). In addition, various international pharmacogenomic consortia have been developed recently to supervise drug response studies.

Table 7.2. Examples of Clinically Valuable Application of Pharmacogenomics Tests in Predicting Drug Response (Efficacy and Toxicity)

A list of current pharmacogenomic guidelines from these consortiums along with a well-annotated pharmacogenomic database has been consolidated into one curated database known as Pharmacogenomics Knowledge Base (PharmGKB) (Thorn et al., 2010). PharmGKB is available via an online portal where users can search on the website by gene, drug, metabolic pathway, and disease. To boost the clinical application of pharmacogenetics and address the barriers to implementation of pharmacogenetic tests into clinical practice CPIC was formed as a shared project between PharmGKB and the Pharmacogenomics Research Network (https://cpicpgx.org). CPIC provides freely available, peer-reviewed, updatable, and detailed gene/drug clinical practice guidelines that enable the translation of genetic laboratory test results into actionable prescribing decisions for specific drugs. The guidelines can focus on genes (e.g., thiopurine methyltransferase and its implications for thiopurines) or around drugs (e.g., warfarin and CYP2C9 and VKORC1). Efforts like PharmGKB and CPIC can help to overcome the confusion created about various pharmacogenetic tests and can help clinical decision making. In addition, the FDA has created a table of Pharmacogenomic Biomarkers in Drug Labeling that lists FDA-approved drugs with pharmacogenomic information in their labeling (http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm). This biomarker table provides up to date information on genomic markers that have been referred in FDA package inserts for different drugs. Various biomarkers are included in this table, e.g., germ-line or somatic gene variants, functional deficiencies, expression changes, and chromosomal abnormalities as well as selected protein biomarkers that need to be tested before starting treatment in a selected subset of patients. Moreover, with continued integration of pharmacogenomics in clinical trials and drug development, novel important genes and variants that can predict drug efficacy and toxicity will be identified and can be implemented in clinical practice.

Read the original here:
Pharmacogenomics - an overview | ScienceDirect Topics

Pharmacogenomics: What Is It and How Does DNA Testing for …

Pharmacogenomics, or the study of how genetics affect your bodys response to medications, is a relatively new and exciting field of science. Scientists are learning more each day about how genetic testing can be used to select the best medication for patients. Genetic testing can help a doctor determine whether a medication will be effective for a patient and provide dosing guidance. It can also help alert clincians to medications that might be potentially harmful to patients.

Genetic testing has become increasingly popular among doctors who prescribe psychiatric medication, in particular antidepressants. Finding the right mental health medication can sometimes be a slow process full of unpleasant side effects. Roughly 40% of people who take an antidepressant will stop taking the medication within the first three months because of side effects or because they believe the medication is ineffective. People who take antidepressants often complain of unpleasant side effects like nausea, sexual dysfunction, headaches, drowsiness, dry mouth, and increased anxiety. When a person experiences side effects, it is easy for them to become discouraged and assume that no medication will help their condition.

Side effects sometimes occur because people metabolize medications differently depending on their genetic code. For example, some people might metabolize an antidepressant more slowly, and a higher concentration of the medication in their body can cause unpleasant side effects. People who metabolize a medication very quickly might have fewer side effects but might need more of the medication to effectively treat depressive symptoms. Therefore, doctors are increasingly recommending genetic testing for depression medications to find the proper dosage and the right medication which may result in fewer side effects.

Take our 2-minute Depression quiz to see if you may benefit from further diagnosis and treatment.

Genetic testing is designed to be easy and painless. To complete the test, a laboratory collects a small sample of blood or saliva from the patient. The sample is usually sent to a pharmacogenomic testing laboratory to be analyzed. This lab sequences the DNA and analyzes any variations or changes in specific genes that are associated with how you respond to a particular medication. Testing for a specific kind of medication only has to be done once, but you may require additional pharmacogenomic testing if your doctor wants to evaluate you for another type of medication.

Because the field is still in its infancy, there is not pharmacogenomic testing available for every medication. But genetic testing is available for many of the medications that treat psychiatric conditions including anxiety, depression, bipolar disorder, schizophrenia, panic disorder, obsessive compulsive disorder (OCD), and post-traumatic stress disorder (PTSD). Here are just a few of the many psychiatric medications currently available for testing:

It is important to note that genetic testing is not always completely accurate. Because the field is still new, there are only a few studies supporting the claim that patients who undergo genetic testing for medication will have more positive outcomes than patients who do not. There also are limitations to what genetic testing can tell you about how your body will metabolize a medication. There is not one pharmacogenomic test that will provide information about all medications so you may need more than one test if you are taking more multiple meds. And, some medications cant be tested using this method (i.e. aspirin and other over-the-counter pain relievers).

Sometimes genes have a strong influence over how the medication works, and sometimes other factors are more influential. These factors can include gender, age, nutrition, smoking history, and pregnancy. Your other medical conditions and any medications you takeboth prescription and over-the-countercan also affect how medications are metabolized. Your doctor may take all of these factors into consideration when they prescribe you a medication and when they consider whether to recommend that you complete genetic testing.

Insurance coverage for genetic testing may vary depending on your insurance plan and personal history. You or your doctor may be required to submit documentation proving that you have a certain diagnosis or have experienced difficulty finding the right medication. It may be helpful to ask your healthcare provider for the specific procedure and medical billing codes for the lab tests theyd like to order before calling your insurance company about coverage. If your insurance doesnt cover the cost, check with the genetic testing company to see if they offer any payment assistancethese tests can cost a few hundred dollars.

Treating mental illness usually involves a combination of medication, psychotherapy, and psychoeducation. So its important to participate in other kinds of treatment and self-care activities while taking medication. Your diet, sleep, exercise, and other factors can also play a role in your mood and how your body responds to medication. As your body ages and develops, you may also find that medications will need to be adjusted by your doctor.

Talk to your doctor about whether genetic testing could be useful for helping you find the right medication. Patients who are in crisis or who have a history of difficulty in finding the right medication are sometimes more likely to be recommended for genetic testing. Your primary care doctor may also have to refer you to a psychiatrist to provide more specialized knowledge in selecting a medication or to evaluate whether genetic testing could be beneficial.

To prepare for your appointment, you may want to prepare notes about the following information:

Genetic testing isnt a magic solution to a persons mental health challenges, but it can sometimes provide your doctor with important information about how your body will metabolize certain medications. Many people, however, find the right medication for their symptoms without the use of genetic testing. So dont be discouraged if it is not an option or not covered by your insurance. It never hurts, however, to start a conversation with your doctor about your options. The testing may not inform him of the perfect medication, but it can alert a physician as to what medications are likely to cause adverse effects or be ineffective. So dont hesitate to talk to your healthcare provider about whether you might benefit from pharmacogenomic testing.

Last Updated: Jul 31, 2018

Go here to see the original:
Pharmacogenomics: What Is It and How Does DNA Testing for ...

Pharmacogenomics Jobs, Employment | Indeed.com

Filter results by: Sortby: relevance - date

more

Myriad Neuroscience

Mason, OH 45040

ISR will undergo continued pharmacogenomic and product sales training. Communicate with healthcare providers and their staff about pharmacogenomic testing and

Translational Medicine group at AstraZeneca is looking for a highly motivated scientist with experience in development of molecular assays and expertise in

Lighthouse Lab Services

Albany, NY

$55,000 - $65,000 a year

Lighthouse Lab Services is representing a laboratory billing and RCM management company, based out of Albany NY, that is looking to hire two account managers to

(medicinal chemistry, pharmacology, toxicology, pharmacogenomics and biopharmaceutics) for Pharm.D. The assistant or associate professor is a full-time, tenure

In these internships, you will work closely with an experienced pharmaceutical industry statistician to perform statistical analysis of data from and/or

The company currently offers advanced genome-based diagnostics for reproductive health, oncology, and pharmacogenomics, and is building predictive models of

Discovery and Clinical Pharmacogenomics (Kenilworth, NJ; Our company's Genetics and Pharmacogenomics team is currently recruiting for summer interns.

Translational Medicine group at AstraZeneca is looking for a highly motivated scientist with experience in development of molecular assays and expertise in

The company currently offers advanced genome-based diagnostics for reproductive health, oncology, and pharmacogenomics, and is building predictive models of

Early focus is on pharmacogenomics, test guidance, rare diagnosis, risk prediction, and screening. Beacon Oncology Information System is built for the

Ethical, Legal, Social Issues in Genomics and Pharmacogenomics. Epigenetics and pharmacogenomics and their role in todays healthcare. Each term is eight weeks.

One study will recruit 1800 adults of African ancestry with hypertension to evaluate the impact of returning high-risk APOL1 genetic test results to patients

ISR will undergo continued pharmacogenomic and product sales training. Communicate with healthcare providers and their staff about pharmacogenomic testing and

Read more here:
Pharmacogenomics Jobs, Employment | Indeed.com

Pharmacogenomics and Personalized Medicine | NorthShore

The right drug, at the right dose, at the right time. This is the power of pharmacogenomics.

When it comes to medications, a drug that works well for one person may not work well for another. A standard dosage may achieve the desired treatment outcome in most patients yet could result in side effects or no therapeutic benefit at all in others.

We know that 98% of patients who take the MedClueRx test have at least one result that may impact their care. Pharmacogenomics testing is particularly valuable before you have tried several medications for a condition. It is also useful if you feel your medications are not working or have experienced adverse side effects.

*Do not change or stop taking any medicine based on a genetic test report without consulting your healthcare provider. This test is not intended to inform you about your current state of health, including whether or not you should take a medication or how much you should take. This test does not diagnose any health conditions and is not a substitute for visiting your health care provider. Discuss the results of the genetic test with your healthcare provider, including whether the medication label includes information on how to use genetic information to determine dosage. Medicine should always be taken as prescribed by your healthcare provider.

See the article here:
Pharmacogenomics and Personalized Medicine | NorthShore

What is pharmacogenomics? – Genetics Home Reference – NIH

Pharmacogenomics is the study of how genes affect a persons response to drugs. This relatively new field combines pharmacology (the science of drugs) and genomics (the study of genes and their functions) to develop effective, safe medications and doses that will be tailored to a persons genetic makeup.

Many drugs that are currently available are one size fits all, but they don't work the same way for everyone. It can be difficult to predict who will benefit from a medication, who will not respond at all, and who will experience negative side effects (called adverse drug reactions). Adverse drug reactions are a significant cause of hospitalizations and deaths in the United States. With the knowledge gained from the Human Genome Project, researchers are learning how inherited differences in genes affect the bodys response to medications. These genetic differences will be used to predict whether a medication will be effective for a particular person and to help prevent adverse drug reactions.

The field of pharmacogenomics is still in its infancy. Its use is currently quite 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.

Read the original post:
What is pharmacogenomics? - Genetics Home Reference - NIH

MediMap – pharmacogenomic testing for adults and children …

MediMap is a one-time Pharmacogenomics (or "PGx") test that may indicate how a person will respond to specific prescription medications. Your MediMap test results can help guide your healthcare providers to ensure better medication choices and doses based on your genetic makeup... leading to more effective illness management and improved health. The future of medicine is about putting into practice the knowledge that with prescription medicines and dosage amounts, one size does NOT fit all.

The MediMap test is available for all ages; everyone in the family including newborns can benefit from pharmacogenomics testing.

Please note: some studies may overlap; for example, a MediMap multi-drug panel may include the specific drug(s) you were looking to be tested for within a condition-specific panel. A genetic counselor will go over your best options during your consult.

What is pharmacogenomics?+

Pharmacogenomics is the study of how people's genes affect their response to medications.The term pharmacogenomics (also called PGx) is a combination of pharmacology (the study of medications) and genomics (the study of how peoples genetic information can influence their health).PGx may be used to help choose the best medications and doses for you.

What is MediMap?+

MediMap is a pharmacogenomic test that helps guide you and your healthcare providers to better medication choices and doses for you. Until recently, most medicines have been developed and given to patients in a "one size fits all" approach.However, people respond differently to medications due, in part, to their genetic makeup. While some people will find a medication helpful, others may not respond to that medication at all. Some people will need a dose that is higher or lower than what is usually prescribed. Even others may have negative side effects (called adverse drug reactions).MediMap looks at specific genetic changes (called variants) that influence a persons response to many medications.

Does MediMap cover all prescription medications?+

While the number of personalized medicines increases steadily every year, not all medications can yet be connected with genetic changes that influence a persons response to medications.Also, there are other medications (besides those included in our tests) that have been reported to be affected by pharmacogenomic variants. However, at this time, there is not enough evidence available to include them on our reports. To help you better understand differences between evidence levels, we differentiate between "actionable" and "informative" information on our reports."Actionable" means there are expert recommendations to help guide clinical treatment; and "informative" means there is insufficient evidence, at this time, and clinical use of this information is optional.You can see which prescription medications are covered in each MediMap tests on our MediMap test comparison sheet.

Which MediMap test is best for me?+

At your appointment, you will meet with a genetic counselor to discuss available testing options. Factors to consider may include: medications you are currently taking, those you might be prescribed in the future, and a history of medication side effects or ineffectiveness. Our goal is for you to have a good understanding of how pharmacogenomic testing may help you, and allow you and your physician to make the best decisions for you.

Are my results confidential?+

Yes. The results will be entered into the secure Inova electronic medical record, and a copy of the report will be mailed to your home. You are responsible for sharing your test results with your healthcare providers.

What will happen to my DNA sample?+

Your DNA sample will be kept for at least 90 days after the MediMap report has been sent to you. At that time, the sample will be disposed or de-identified (name and other identifiers are removed) depending on your selection at the time you authorize the test. If your sample is de-identified, it may be used for quality control purposes, to develop new tests, or for educational activities. No clinical tests other than MediMap will be performed using your DNA sample.

Read the original:
MediMap - pharmacogenomic testing for adults and children ...

Genes Advice | Pharmacogenomics

Have you or a loved one ever had a bad reaction or unusual side effects when taking a medicine? Reading the long list of allergies, warnings, interactions and other possible complications on any prescription drug leaflet can be scary. Why is it that some drugs work great for some people but dont work well for others?

With 1,222 new drugs approved by the Food and Drug Administration (FDA) between 1950 and 2008, there are many options on the market (Munos, 2009, Nature Reviews Drug Discovery).With so many options, finding the right medication can be a drawn-out process of trial and error for the patient and the provider. As a result, complications are becoming alarmingly common. In 2015, the FDA received nearly 1.3 million reports ofdrug interactions, a rate that more than doubled in six years. Deaths from severe allergic reactions are also on the rise. *How your body reacts to any given medicine will be different from that of your spouse, your cousin, your best friend, your next-door neighbor, and your favorite grocery checkout clerk. (Reactions may even differ with the over-the-counter-drugs like aspirin or dietary supplements that the very same checkout clerkrang up for you!)

Read the original:
Genes Advice | Pharmacogenomics

MediMap genomics test – precision medicine at Inova …

Go to Inova Translational Medicine Institute

Pharmacogenomics, also called PGx, combines the science of how medications work (pharmacology) with the science of how genetic differences can influence health (genomics).Inova is pleased to offer the MediMap PGx test to adults and children, as well asto newbornsdelivered atInova Womens Hospital.

MediMap is part of the standard package of services offered to all babies born at Inova Womens Hospital located at Inova Fairfax Medical Campus, and it is therefore performed at no additional cost. Inova is the only health system in the U.S. that provides this optional pharmacogenomics test to newborns as part of our standard package of care. More info about MediMap for newborns

Inova is pleased to announce that we will be offering theMediMap test to adults and children in the near future.More info about MediMap for adults and children

Until recently, most medicines have been developed and prescribed to patients in a one size fits all approach. PGx testing informs your physician about your, or your childs, genetic makeup to help determine which medications to use or the amount prescribed. PGx testing may also reduce side-effects.

MediMap is a one-time genetic test that may indicate how a person will respond to some prescription medications. The test helps guide healthcare providers to better medication choices and doses for their patients. MediMap testing provides information to more effectively manage illnesses and improve their health.

Cant find what you are looking for? Please call us at 1-844-GENOME-4U (1-844-436-6634) or email us at geneinfo@inova.org.

Link:
MediMap genomics test - precision medicine at Inova ...

PHG Foundation – Interactive Tutorial: Pharmacogenomics …

Pharmacogenetics refers to the study of genetic influences on an individuals response to drugs. In pharmacogenetics, the analysis of a specific gene, or group of genes, may be used to predict responses to a specific drug or class of drugs.

Pharmacogenomics refers collectively to all the genes that influence drug responses, and how genome-wide analysis may be used to identify such genes in the search for novel drug targets and/or key determinants of drug reactions.

The effects of a specific dose of a specific drug will differ between individual recipients. A drug that is effective in one person may have no discernible therapeutic effect in another, whilst a third might show a partial response; in some, there may be undesirable side-effects.

There are multiple contributory factors to such variation in drug response, such as gender, age, body mass, diet, the presence of other drugs or of particular disease states and exposure to certain chemicals or toxins, such as cigarette smoke. In addition to these, genetic factors also influence drug response.

To view this interactive tutorial, you need the latest Flash player.

Read more:
PHG Foundation - Interactive Tutorial: Pharmacogenomics ...

Pharmacogenomics – ncpanet.org

General Description, Overview, and Opportunities

Pharmacogenomics has increasingly become an area of interest to clinicians because of the potential to tailor pharmacotherapy based on genetic variations in patients. Pharmacogenomics is one of the key aspects of personalized medicine, focusing on how an individual's DNA affects the way they respond to medications. All individuals have different genetic make-up so they respond differently to the same medication. Based on this insight, pharmacogenomics allows customized treatment for a wide range of health problems including; cardiovascular disease, Alzheimer's disease, cancer, HIV/AIDS, and asthma. Often, drug choice and dosage require experimentation (trial and error) in order to find the best treatment option. With pharmacogenomics testing, the need for this experimentation is decreased. As a result, the process becomes faster and more cost-effective and the possibility of adverse events caused by the wrong drug choice or dosage is significantly reduced.

One avenue for implementing pharmacogenomic is through medication therapy management (MTM), where pharmacists assess and evaluate a patient's complete medication therapy regimen. By gathering key pieces of information, e.g. which medications and supplements a patient is currently taking, pharmacists can assess current treatment and suggest alternative therapies.

As medication experts and POC service providers, pharmacists can educate physicians and patients and perform the actual sample collection to be utilized for genetic testing. The broad application of pharmacogenomics to personalized medicine will improve patient outcomes and lower healthcare costs.

Test Features

Pharmacies require a lab partner to provide clinically relevant data and interpret results for physicians. Most tests screens all well-established pharmacogenomics genes in a single, cost-effective test. Results are delivered quickly via intuitive, clinically relevant, medically actionable report. The data provides lifetime utility of data, thereby decreasing the need for future testing.

Community pharmacists routinely perform point of care services and can assist patients by:

Performing a buccal swab in minutes

Send the collected DNA to the lab

Interpret results and discuss with physicians

Contact the patient to explain the results and any changes in therapy

Companies

Pharmacist Resources and Training

Here is the original post:
Pharmacogenomics - ncpanet.org

Pharmacogenetics – Wikipedia

Pharmacogenetics is the study of inherited genetic differences in drug metabolic pathways which can affect individual responses to drugs, both in terms of therapeutic effect as well as adverse effects.[1] The term pharmacogenetics is often used interchangeably with the term pharmacogenomics which also investigates the role of acquired and inherited genetic differences in relation to drug response and drug behavior through a systematic examination of genes, gene products, and inter- and intra-individual variation in gene expression and function.[2]

In oncology, pharmacogenetics historically is the study of germline mutations (e.g., single-nucleotide polymorphisms affecting genes coding for liver enzymes responsible for drug deposition and pharmacokinetics), whereas pharmacogenomics refers to somatic mutations in tumoral DNA leading to alteration in drug response (e.g., KRAS mutations in patients treated with anti-Her1 biologics).[3]

Much of current clinical interest is at the level of pharmacogenetics, involving variation in genes involved in drug metabolism with a particular emphasis on improving drug safety. The wider use of pharmacogenetic testing is viewed by many as an outstanding opportunity to improve prescribing safety and efficacy. Driving this trend are the 106,000 deaths and 2.2 Million serious events caused by adverse drug reactions in the US each year.[4][unreliable medical source?] As such ADRs are responsible for 5-7% of hospital admissions in the US and Europe, lead to the withdrawal of 4% of new medicines, and cost society an amount equal to the costs of drug treatment.[5]

Comparisons of the list of drugs most commonly implicated in adverse drug reactions with the list of metabolizing enzymes with known polymorphisms found that drugs commonly involved in adverse drug reactions were also those that were metabolized by enzymes with known polymorphisms (see Phillips, 2001).

Scientists and doctors are using this new technology for a variety of things, one being improving the efficacy of drugs. In psychology, we can predict quite accurately which anti-depressant a patient will best respond to by simply looking into their genetic code.[citation needed][dubious discuss] This is a huge step from the previous practice of adjusting and experimenting with different medications to get the best response. Antidepressants also have a large percentage of unresponsive patients and poor prediction rate of ADRs (adverse drug reactions). In depressed patients, 30% are not helped by antidepressants. In psychopharmacological therapy, a patient must be on a drug for 2 weeks before the effects can be fully examined and evaluated. For a patient in that 30%, this could mean months of trying medications to find an antidote to their pain. Any assistance in predicting a patients drug reaction to psychopharmacological therapy should be taken advantage of. Pharmacogenetics is a very useful and important tool in predicting which drugs will be effective in various patients.[6] The drug Plavix blocks platelet reception and is the second best selling prescription drug in the world, however, it is known to warrant different responses among patients.[7]GWAS studies have linked the gene CYP2C19 to those who cannot normally metabolize Plavix. Plavix is given to patients after receiving a stent in the coronary artery to prevent clotting.

Stent clots almost always result in heart attack or sudden death, fortunately it only occurs in 1 or 2% of the population. That 1 or 2% are those with the CYP2C19 SNP.[8] This finding has been applied in at least two hospitals, Scripps and Vanderbilt University, where patients who are candidates for heart stents are screened for the CYP2C19 variants.[9]

Another newfound use of pharmacogenetics involves the use of Vitamin E. The Technion Israel Institute of Technology observed that vitamin E can be used to in certain genotypes to lower the risk of cardiovascular disease in patients with diabetes, but in the same patients with another genotype, vitamin E can raise the risk of cardiovascular disease. A study was carried out, showing vitamin E is able to increase the function of HDL in those with the genotype haptoglobin 2-2 who suffer from diabetes. HDL is a lipoprotein that removes cholesterol from the blood and is associated with a reduced risk of atherosclerosis and heart disease. However, if you have the misfortune to possess the genotype haptoglobin 2-1, the study shows that this same treatment can drastically decrease your HDL function and cause cardiovascular disease.[10]

Pharmacogenetics is a rising concern in clinical oncology, because the therapeutic window of most anticancer drugs is narrow and patients with impaired ability to detoxify drugs will undergo life-threatening toxicities. In particular, genetic deregulations affecting genes coding for DPD, UGT1A1, TPMT, CDA and CYP2D6 are now considered as critical issues for patients treated with 5-FU/capecitabine, irinotecan, mercaptopurine/azathioprine, gemcitabine/capecitabine/AraC and tamoxifen, respectively. The decision to use pharmacogenetic techniques is influenced by the relative costs of genotyping technologies and the cost of providing a treatment to a patient with an incompatible genotype. When available, phenotype-based approaches proved their usefulness while being cost-effective.[11]

In the search for informative correlates of psychotropic drug response, pharmacogenetics has several advantages:[12]

The first observations of genetic variation in drug response date from the 1950s, involving the muscle relaxant suxamethonium chloride, and drugs metabolized by N-acetyltransferase. One in 3500 Caucasians has less efficient variant of the enzyme (butyrylcholinesterase) that metabolizes suxamethonium chloride.[13] As a consequence, the drugs effect is prolonged, with slower recovery from surgical paralysis. Variation in the N-acetyltransferase gene divides people into "slow acetylators" and "fast acetylators", with very different half-lives and blood concentrations of such important drugs as isoniazid (antituberculosis) and procainamide (antiarrhythmic). As part of the inborn system for clearing the body of xenobiotics, the cytochrome P450 oxidases (CYPs) are heavily involved in drug metabolism, and genetic variations in CYPs affect large populations. One member of the CYP superfamily, CYP2D6, now has over 75 known allelic variations, some of which lead to no activity, and some to enhanced activity. An estimated 29% of people in parts of East Africa may have multiple copies of the gene, and will therefore not be adequately treated with standard doses of drugs such as the painkiller codeine (which is activated by the enzyme). The first study using Genome-wide association studies (GWAS) linked age-related macular degeneration (AMD) with a SNP located on chromosome 1 that increased ones risk of AMD. AMD is the most common cause of blindness, affecting more than seven million Americans. Until this study in 2005, we only knew about the inflammation of the retinal tissue causing AMD, not the genes responsible.[9]

One of the earliest tests for a genetic variation resulting in a clinically important consequence was on the enzyme thiopurine methyltransferase (TPMT). TPMT metabolizes 6-mercaptopurine and azathioprine, two thiopurine drugs used in a range of indications, from childhood leukemia to autoimmune diseases. In people with a deficiency in TPMT activity, thiopurine metabolism must proceed by other pathways, one of which leads to the active thiopurine metabolite that is toxic to the bone marrow at high concentrations. Deficiency of TPMT affects a small proportion of people, though seriously. One in 300 people have two variant alleles and lack TPMT activity; these people need only 6-10% of the standard dose of the drug, and, if treated with the full dose, are at risk of severe bone marrow suppression. For them, genotype predicts clinical outcome, a prerequisite for an effective pharmacogenetic test. In 85-90% of affected people, this deficiency results from one of three common variant alleles.[14] Around 10% of people are heterozygous - they carry one variant allele - and produce a reduced quantity of functional enzyme. Overall, they are at greater risk of adverse effects, although as individuals their genotype is not necessarily predictive of their clinical outcome, which makes the interpretation of a clinical test difficult. Recent research suggests that patients who are heterozygous may have a better response to treatment, which raises whether people who have two wild-type alleles could tolerate a higher therapeutic dose.[15] The US Food and Drug Administration (FDA) have recently deliberated the inclusion of a recommendation for testing for TPMT deficiency to the prescribing information for 6-mercaptopurine and azathioprine. The information previously carried the warning that inherited deficiency of the enzyme could increase the risk of severe bone marrow suppression. It now carries the recommendation that people who develop bone marrow suppression while receiving 6-mercaptopurine or azathioprine be tested for TPMT deficiency.[citation needed]

A polymorphism near a human interferon gene is predictive of the effectiveness of an artificial interferon treatment for Hepatitis C. For genotype 1 hepatitis C treated with Pegylated interferon-alpha-2a or Pegylated interferon-alpha-2b (brand names Pegasys or PEG-Intron) combined with ribavirin, it has been shown that genetic polymorphisms near the human IL28B gene, encoding interferon lambda 3, are associated with significant differences in response to the treatment.[16] Genotype 1 hepatitis C patients carrying certain genetic variant alleles near the IL28B gene are more probable to achieve sustained virological response after the treatment than others, and demonstrated that the same genetic variants are also associated with the natural clearance of the genotype 1 hepatitis C virus.[17]

Despite the many successes, most drugs are not tested using GWAS. However, it is estimated that over 25% of common medication have some type of genetic information that could be used in the medical field.[18] If the use of personalized medicine is widely adopted and used, it will make medical trials more efficient. This will lower the costs that come about due to adverse drug side effects and prescription of drugs that have been proven ineffective in certain genotypes. It is very costly when a clinical trial is put to a stop by licensing authorities because of the small population who experiences adverse drug reactions. With the new push for pharmacogenetics, it is possible to develop and license a drug specifically intended for those who are the small population genetically at risk for adverse side effects. [19]

The ability to test and analyze an individuals DNA to determine if the body can break down certain drugs through the biochemical pathways has application in all fields of medicine. Pharmacogenetics gives those in the health care industry a potential solution to help prevent the significant amount of deaths that occur each year due to drug reactions and side effects. The companies or laboratories that perform this testing can do so acrossed all categories or drugs whether it be for high blood pressure, gastrointestinal, urological, psychotropic or anti-anxiety drugs. Results can be presented showing which drugs the body is capable of breaking down normally versus the drugs the body cannot break down normally. This test only needs to be done once and can provide valuable information such as a summary of an individuals genetic polymorphisms, which could help in a situation such as being a patient in the emergency room.[20]

As the cost per genetic test decreases, the development of personalized drug therapies will increase.[21] Technology now allows for genetic analysis of hundreds of target genes involved in medication metabolism and response in less than 24 hours for under $1,000. This a huge step towards bringing pharmacogenetic technology into everyday medical decisions. Likewise, companies like deCODE genetics, Navigenics and 23andMe offer genome scans. The companies use the same genotyping chips that are used in GWAS studies and provide customers with a write-up of individual risk for various traits and diseases and testing for 500,000 known SNPs. Costs range from $995 to $2500 and include updates with new data from studies as they become available. The more expensive packages even included a telephone session with a genetics counselor to discuss the results.[9]

Pharmacogenetics has become a controversial issue in the area of bioethics. It's a new topic to the medical field, as well as the public. This new technique will have a huge impact on society, influencing the treatment of both common and rare diseases. As a new topic in the medical field the ethics behind it are still not clear. However, ethical issues and their possible solutions are already being addressed.

There are three main ethical issues that have risen from pharmacogenetics. First, would there be a type equity at both drug development and the accessibility to tests.[22] The concern of accessibility to the test is whether it is going to be available directly to patients via the internet, or over the counter. The second concern regards the confidentiality of storage and usage of genetic information.[23] Thirdly, would patients have the control over being tested.

One concern that has risen is the ethical decision health providers must take with respect to educating the patient of the risks and benefits of medicine developed by this new technology. Pharmacogenetics is a new process that may increase the benefits of medicine while decreasing the risk. However clinicians have been unsuccessful in educating patients regarding the concept of benefits over risk. The Nuffield Council reported that patients and health professionals have adequate information about pharmacogenetics tests and medicine.[23] Health care providers will also encounter an ethical decision in deciding to tell their patients that only certain individuals will benefit from the new medicine due to their genetic make-up.[22] Another ethical concern is that patients who have not taken the test be able to have access to this type of medicine. If access is given by the doctor the medicine could negatively impact the patient's health. The ethical issues behind pharmacogenetics tests, as well as medicine, are still a concern and policies will need to be implemented in the future.

Here is the original post:
Pharmacogenetics - Wikipedia

Internships Internship Search and Intern Jobs …

Internships.com is the worlds largest internship marketplace bringing students, employers andhigher education institutionstogether in one centralized location. We specialize in helpingstudentsand young professionals find the right internship to kick start their career.

Its no secret that internships are the most effective way for students to gain work experience before graduation. In fact, studies show that 7 out of 10 internships turn into full-time jobs. Luckily,starting your internship searchis easy. Simply tell us yourcollege majorandpreferred locationand you can connect with thousands ofcompaniesthat are hiring interns now.

Whether youre looking for work experience, want toreceive college creditsor just need some extra spending money, you can use Internships.com to findpaid internships,summer jobs or entry level jobs.Students can even use ourInternship Predictorto help find the right job. Best of all, searching for internships is completely free.

Are you anemployerlooking to find interns for your business? Employers canpost internshipsand student jobs for free on Internships.com. Once you post your job listing, you canfind studentsusing our extensive resume database. Employers can access extensiveemployment resourcesto help ensure your business has thebest summer internship program.

Start your internship searchnow by browsing opportunities by major, location, or company name. Employers,create an accountto post internships for free.

Excerpt from:
Internships Internship Search and Intern Jobs ...

Genomics|Update|Non Communicable Diseases

FDA: The steps were taking to push precision medicine forward, by Meghana Keshavan, Med City News, September 23, 2015

Dr. Henry T. Lynch Symposium: Advances in hereditary cancer summary by G, I have Lynch Syndrome, September 22, 2015

Who knows what it means, Genome Web, September 21, 2015 [by subscription only]

87-year-old Creighton University doctor finds motivation in tracking cancer families, by Rick Ruggles, Omaha.com, September 20, 2015

Genomics will redraw the rare disease map: The "Rarity Catch-22", CVID, Primary Immune and Rare Disease Blog, September 19, 2015

NIH moves forward on genetic database while hoping for funding, iHealthBeat, September 18, 2015

Enthusiasm for personalized cancer drugs runs ahead of the science, by Asher Mullard, Nature News, September 17, 2015

For some children with cancer, genomic information may help guide treatment decisions, Cancer Currents Blog, NCI, September 17, 2015

Is fish oil good for you? Depends on your DNA, by Elizabeth Pennisi, Science News, September 17, 2015

Cure for sickle cell in adults validated, Science Daily, September 16, 2015

Pres. Obama's precision medicine initiative, the human genome project, and your individualized genetic data, by Samantha Olson, Medical Daily, September 16, 2015

Here is the original post:
Genomics|Update|Non Communicable Diseases

Vanderbilt Pharmacogenomics

Welcome

Clinicians and patients recognize that not every person responds to drugs in the same way. Some drugs carry a risk of adverse reactions that often seem to occur by chance. Even drugs that are well-tolerated may be highly effective at low doses in some patients, and minimally effective at high doses in others.

The Human Genome Project has established the initial sequence of all human DNA. In doing so, the Genome Project enabled study of how variations among patient genomes affects why disease develops in some patients and not in others. Pharmacogenetics is the study of how individual DNA variations affect drug responses, and the term pharmacogenomics is often used to describe how many variations in an individual patient, or in large groups of patients, affect the outcome of drug therapy.

Vanderbilt University is a center of excellence in the study of mechanisms underlying individual variability in response to drug therapy. This work reaches from basic science to clinical medicine, and includes studies of metabolism and transport of many drugs, as well as, specific studies of drug therapies in diverse clinical settings such as arrhythmias, hypertension, autonomic dysfunction, psychiatric disease, cancer, HIV infection, and recovery from anesthesia.

Research Centers with a special focus on pharmacogenetics and pharmacogenomics include the Division of Clinical Pharmacology, the Vanderbilt-Ingram Cancer Center, the Center for Molecular Neuroscience, the Vanderbilt-Meharry Center for AIDS Research, the Division of Genetic Medicine, the General Clinical Research Center, the Center for Human Genetics Research, and the Center for Genetics and Health Policy. Studies of arrhythmia therapies are supported by Vanderbilt's participation in the NIH-sponsored Pharmacogenetics Research Network.

More:
Vanderbilt Pharmacogenomics

Pharmacogenomics – PubMed Central (PMC)

BMJ. 1999 Nov 13; 319(7220): 1286.

Departments of Biopharmaceutical Sciences and Pharmaceutical Chemistry, University of California, San Francisco, CA94143-0446, USA

We all differ in our response to drug treatmentoccasionally with dramatic effects. The era of one drug fits all patients is about to give way to individualised therapy matching the patient's unique genetic make up with an optimally effective drug.1 Pharmacogenetics and pharmacogenomics are the emerging disciplines that are leading the way towards individualised medicine.2,3 Initially, researchers focused their attention on pharmacogeneticsvariations in single candidate genes responsible for variable drug response. Subsequently, studies involving the entire human genome broadened the scope of investigation, giving rise to pharmacogenomics as one of the hottest fields in biotechnology today.

Response to drug treatment can vary greatly between patients; genetic factors have a major role in treatment outcome

Pharmacogenetics and pharmacogenomics are emerging disciplines that focus on genetic determinants of drug response at the levels of single genes or the entire human genome respectively

Technologies involving gene chip arrays can determine thousands of variations in DNA sequences for individual patients; most variants are single nucleotide polymorphisms

Pharmacogenomics aims at establishing a signature of DNA sequence variants that are characteristic of individual patients to assess disease susceptibility and select the optimal drug treatment

This approach has the potential to revolutionise prevention and treatment of diseases

Unexpected drug reactions have been noted for some time, but the systematic study of hereditary origins began only in the 1950s. A few patients developed prolonged respiratory muscular paralysis after being given succinylcholine (suxamethonium), a short acting muscle relaxant widely used in surgery and electroshock treatment. In the 1970s, a trial with the antihypertensive agent debrisoquine resulted in a precipitous drop of blood pressure and collapse in nearly 10% of volunteers. Furthermore, isoniazid therapy for tuberculosis caused peripheral neuropathies in patients who were sensitive to the neurotoxic effects of the drug. Ground breaking genetic and biochemical studies by Werner Kalow and others showed that these adverse effects result from polymorphisms in genes encoding the drug metabolising enzymes serum cholinesterase,4 cytochrome P-450,5 and N-acetyltransferase.6 These observations laid the foundation for pharmacogenetics.

Today, many examples of genetic variability in drug response and toxicity are known (table). In a few cases, genetic tests are beginning to find their way into clinical practice. In cancer chemotherapy with tioguanine, severe toxicity or even death can result if a patient is unable to inactivate the drug. Functional assays of thiopurine methyltransferase in red blood cells or genotyping can identify those patients who are at risk and must be given a much lower dose of tioguanine.7,8 This is particularly critical for the 1 in 300 patients who is homozygous for null alleles (non-functional) of the gene encoding thiopurine methyltransferase which converts the drug to its inactive methylated form. Therefore, genotyping or functional analysis has become standard practice in major cancer treatment centres such as the Mayo Clinic in Rochester, Minneapolis, and St Jude Children's Research Hospital in Memphis, Tennessee.

The large family of cytochrome P-450 genes has been most intensely studied because it contains the main drug metabolising enzymes encoded by numerous genes.2 Among the cytochrome P-450 subtypes, CYP2D6 and CYP2C19 play a critical part in determining the response to several drugs. This is particularly important for lipophilic drugssuch as drugs that act on the central nervous system and penetrate the lipophilic blood-brain barrierbecause renal excretion is minimal and cytochrome P-450 metabolism provides the only means of effective drug elimination. Thus, homozygous carriers of CYP2D6 null alleles and cannot readily degrade and excrete many drugs, including debrisoquine, metoprolol, nortriptyline, and propafenone.9 These patients are termed poor metabolisers for CYP2D6 selective drugs. Because of this they are exquisitely sensitive to these drugs. The incidence of poor metabolisers varies greatly among ethnic groups, ranging from 1% in Japanese people to 15% in Nigerians. Similarly, patients with defective CYP2C19 subtypes are highly sensitive to methoin (mephenytoin), hexobarbital (hexobarbitone), and other drugs selectively metabolised by this P-450 isoform.

The principal molecular defect in poor metabolisers is a single base pair mutation (AG) in exon 5 of CYP2C19.10 Gene chips designed to test for polymorphisms of the main subtypes of cytochrome P-450 are now commercially available, but not yet in general clinical use. Cytochrome P-450 polymorphisms also affect the inactivation or, in some cases, activation or toxification of xenobiotics, and thus affect an individual's susceptibility to environmental toxins. This is studied in a field of research called toxicogenetics. Launched recently by the US National Institute of Environmental Health Sciences, the environmental genome project aims at understanding genetic factors in individual responses to the environment and parallels the study of genetic variability in drug response.11

As a scientific discipline, pharmacogenetics has made steady progress, but the human genome project has shattered any complacency as it has revealed profound gaps in our knowledge. By broadening the search for genetic polymorphisms that determine drug responses, the new field of pharmacogenomics begins to supersede the candidate gene approach typical of earlier pharmacogenetic studies. Initially hailed by pharmaceutical biotechnology as the latest trend in biotechnology, pharmacogenomics is now taken seriously everywhere. While genomic techniques serve to identify new gene targets for drug research, and some might refer to this as pharmacogenomics, the broader consensus is that pharmacogenomics deals specifically with genetic variability in drug response. The distinction between pharmacogenetics and pharmacogenomics remains blurred, but here are some of the new ideas typical of pharmacogenomics.

Each drug is likely to interact in the body with numerous proteins, such as carrier proteins, transporters, metabolising enzymes, and multiple types of receptors.1 These proteins determine the absorption, distribution, excretion, targeting to the site of action, and pharmacological response of drugs. As a result, multiple polymorphisms in many genes could affect the drug response, requiring a genome-wide search for the responsible genes. We now know that that there are thousands of receptor genes in the human genome, many of which are closely related to each other because they have evolved by gene duplications. Therefore, we must anticipate that a drug rarely binds just to a single receptor but rather interacts promiscuously with several receptor types. Chlorpromazine, for example, is known to engage several dopaminergic, adrenergic, and serotonergic receptors. As a result, polymorphisms in multiple genes can affect the drug response.

Polymorphisms are generally defined as variations of DNA sequence that are present in more than 1% of the population. Most polymorphisms are single nucleotide polymorphisms (referred to as snips). As the human genome contains three billion nucleotides, and variations between individuals occur in 1/300 base pairs, around 10 million single nucleotide polymorphisms probably exist. Only 1% of these may have any functional consequence at all, and thus individuals differ from each other genetically by roughly 100000 polymorphic sites, providing for near infinite variety. As only a small fraction of these single nucleotide polymorphisms will prove relevant to drug response, our goal will be to identify the most important variants.

Novel technology in the form of microarray chips enables us to scan the entire human genome for relevant polymorphisms.12,13 We can determine simultaneously many thousands of polymorphisms in a patient. At present, these single nucleotide polymorphisms are selected merely as markers evenly distributed throughout the genome, in the hope that functionally relevant polymorphisms can be associated with specific markers by virtue of their proximity on the chromosome. Such genome-wide association studies are already being used in the discovery of susceptibility genes for diseases such as asthma and prostate cancer, but they are equally suitable for determining the genes involved in drug response. Genome-wide scanning can identify these genes even if we do not know the mechanisms by which the drug acts in the body. The French genomics company, Genset, currently uses gene chips with 60000 single nucleotide polymorphism markerssufficient for a complete genomic scanapplied to clinical drug trials in partnership with major pharmaceutical companies. Expanding the number of single nucleotide polymorphisms and selecting functionally relevant single nucleotide polymorphisms in coding or promoter/enhancer regions of genes is quite feasible with current technology and would greatly enhance the power of genome-wide scanning. Herein lies the main incentive for the current rush in the pharmaceutical industry to patent single nucleotide polymorphism markers. It might also be possible to salvage useful experimental drugs that would have failed with standard clinical trials, because of an unacceptable incidence of toxicity in a poorly defined patient population. Stratifying patient populations in relation to genetic criteria emerges as a major challenge to the pharmaceutical industry. Undoubtedly, the insights expected to emerge from such an approach are staggering, but they cannot be gauged accurately at present.

Microarrays can further serve to determine the expression pattern of genes in a target tissue. This shows the mechanisms of drug action in a genomic context. It can also clarify interindividual differences in drug response that are downstream of immediate drug effects in the body by shear force of the massive amount of information emanating from chip technology. Analysing the entire transcriptional programme of a tissuefor example, fibroblasts in response to serum stimulation14provides unprecedented details of a complex system and leads to new insights in pathophysiology and biological drug response. Tissue transcript profiling is especially appropriate in cancers because mRNA can be extracted from biopsy specimens or surgical samples. Altered gene expression in the tumour can serve as a guide for selecting effective drug therapy or avoiding unnecessary exposure to toxic but ineffective drugsfor example, the overexpression of drug resistance genes encoding transporters (table).

These advances are the harbinger of profound changes in treatment. What then do we expect to gain from pharmacogenomics? In the near future, genotyping can help avert severe drug toxicity that is genetically determined but occurs only rarely. Alternatively, drugs may be designed a priori so that they are not subject to extreme variations that result from a few well defined polymorphisms. Drug structures under development are already being selected so that they do not interact with cytochrome P-450 subtype CYP2D6 to avoid unwarranted toxicity in people who metabolise this poorly.

Looking further ahead, and on a much broader scale, we could improve drug efficacy by distinguishing between people who respond well to a drug and those who respond poorly. Often, an effective drug response is found in a few patients treated, while most benefit little or not at all. Much could be gained if we could select the optimal drug for the individual patient before treatment begins. Perhaps a gene chip that establishes a single nucleotide polymorphism signature involving multiple genes relevant to therapeutic outcome for each individual will be developed. This signature could offer insights into an individual's susceptibility to disease and responsiveness to drugs, enabling optimal drug selection by genetic criteria. For example, cure rates with combined surgical and drug treatment of advanced colorectal carcinoma range from 20% to 40%, while the remainder of the patients experience little gain or even severe toxicity from chemotherapy. If we could predict which patients respond best to a particular drugor better, which drug will yield optimal effects for a given patientmuch will be gained. The success of this approach will depend critically on the selection of single nucleotide polymorphisms tested by the gene chip. Single nucleotide polymorphisms must be informative and many must be tested to scan the entire genome. This task is by no means complete and constitutes a major goal of those companies which are focusing on genomics.

There are also formidable obstacles that we are unlikely to overcome in the near future. The dynamic complexity of the human genome, involvement of multiple genes in drug responses, and racial differences in the prevalence of gene variants impede effective genome-wide scanning and progress towards practical clinical applications. Furthermore, the drug response is probably affected by multiple genes, each gene with multiple polymorphisms distributed in the general population. For example, the anticancer drug 5-fluorouracil used in the treatment of colorectal cancer is activated and inactivated by nearly 40 different enzymes. Each of these is currently being scanned for relevant polymorphisms at the biotech company Variagenics. Dihydropyrimidine dehydrogenase is a likely candidate in 5-fluorouracil inactivation (table). However, whether extensive genotyping will provide useful predictors of clinical response remains to be seen.

Racial differences add further confounding factors. Drug response might be predicted from a certain pattern of polymorphisms rather than only a single polymorphism, yet these patterns probably differ between ethnic groups. This could prevent us from making predictions about drug responses across the general patient population, and it emphasises the need to stratify clinical pharmacogenomics studies.

Genomic technologies are still evolving rapidly, at an exponential pace similar to the development of computer technology over the past 20 years. We are not certain where genomic technologies will be 10 years from now.

Ethical issues also need to be resolved. Holding sensitive information on someone's genetic make up raises questions of privacy and security and ethical dilemmas in disease prognosis and treatment choices. After all, polymorphisms relevant to drug response may overlap with disease susceptibility, and divulging such information could jeopardise an individual. On the other hand, legal issues may force the inclusion of pharmacogenomics into clinical practice. Once the genetic component of a severe adverse drug effect is documented, doctors may be obliged to order the genetic test to avoid malpractice litigation.

Pharmacogenomics will have a profound impact on the way drug treatment is conducted. We can include here bioengineered proteins as drugs, or even gene therapy designed to deliver proteins to target tissues. These treatments are also subject to constraints and complexities engendered by individual variability. A case in point is the treatment of breast cancer with trastuzumab (Herceptin; Genentech, USA) a humanised monoclonal antibody against the HER2 receptor. Overexpression of HER2 may occur as a somatic genetic change in breast cancer and other tumours. This correlates with poor clinical prognosis and serves as a marker for effective therapy with trastuzumab, either alone or in combination with chemotherapy.15,16

Whether we will see broad use of gene chips in clinical practice within 10 years is questionable, but the mere knowledge of the principles underlying genetic variability will prove valuable in optimising drug therapy. Pharmacogenomics will lead us towards individualised therapy, but it will also help us understand limitations inherent in treating disease in a broad patient population

Incyte's microarray service allows researchers to analyse differential expression in normal and diseased cells

Examples of inherited or acquired variations in enzymes and receptors that affect the drug response23

Competing interests: None declared.

2. Weber WW. Pharmacogenetics. New York: Oxford University Press; 1997.

13. Sinclair B. Everything's great when it sits on a chip: a bright future for DNA arrays. Scientist. 1999;13:1820.

View post:
Pharmacogenomics - PubMed Central (PMC)

New Genetic Tools Learn Genetics

HOME

Pharmacogenomics

Your Doctor's New Genetic Tools

When physicians are given the tools to evaluate a patient's genetic make-up, they will be able to make more accurate diagnoses, and prescribe more efficient drug therapies with fewer adverse side effects.

Differences between people extend beyond our outer physical features. How individuals respond to drugs for the treatment of cancer or other illnesses differs based on the activity and function of enzymes in the body. This information is available in each individual's genetic profile. Even today, genetics is being integrated into individuals' medical treatment plans.

JavaScript and the Flash Player are required to view this content.

APA format: Genetic Science Learning Center (2014, June 22) Your Doctor's New Genetic Tools. Learn.Genetics. Retrieved July 10, 2015, from http://learn.genetics.utah.edu/content/pharma/intro/ MLA format: Genetic Science Learning Center. "Your Doctor's New Genetic Tools." Learn.Genetics 10 July 2015 <http://learn.genetics.utah.edu/content/pharma/intro/> Chicago format: Genetic Science Learning Center, "Your Doctor's New Genetic Tools," Learn.Genetics, 22 June 2014, <http://learn.genetics.utah.edu/content/pharma/intro/> (10 July 2015)

Original post:
New Genetic Tools Learn Genetics

Genomics|Update|Current

CDC authors are indicated in bold

Distinct pathological phenotypes of Creutzfeldt-Jakob disease in recipients of prion-contaminated growth hormone. Cali I, Miller CJ, Parisi JE, Geschwind MD, Gambetti P, Schonberger LB. Acta Neuropathol Commun. 2015 Jun 25;3(1):37.

Bacterial factors associated with lethal outcome of enteropathogenic Escherichia coli infection: genomic case-control studies Donnenberg MS, Hazen TH, Farag TH, Panchalingam S, Antonio M, Hossain A, Mandomando I, Ochieng JB, Ramamurthy T, Tamboura B, Zaidi A, Levine MM, Kotloff K, Rasko DA, Nataro JP. PLoS Negl Trop Dis. 2015 May;9(5):e0003791.

Specificity and Strain-Typing Capabilities of Nanorod Array-Surface Enhanced Raman Spectroscopy for Mycoplasma pneumoniae Detection. Henderson KC, Benitez AJ, Ratliff AE, Crabb DM, Sheppard ES, Winchell JM, Dluhy RA, Waites KB, Atkinson TP, Krause DC. PLoS One. 2015 Jun 29;10(6):e0131831

Identification of influenza A/PR/8/34 donor viruses imparting high hemagglutinin yields to candidate vaccine viruses in eggs Johnson A, Chen LM, Winne E, Santana W, Metcalfe MG, Mateu-Petit G, Ridenour C, Hossain MJ, Villanueva J, Zaki SR, Williams TL, Cox NJ, Barr JR, Donis RO. PLoS One. 2015 ;10(6):e0128982.

A novel botulinum toxin, previously reported as serotype H, has a hybrid structure of known serotypes A and F that is neutralized with serotype A antitoxin Maslanka SE, Luquez C, Dykes JK, Tepp WH, Pier CL, Pellett S, Raphael BH, Kalb SR, Barr JR, Rao A, Johnson EA. J Infect Dis. 2015 Jun 10.

Effects of laser printer-emitted engineered nanoparticles on cytotoxicity, chemokine expression, reactive oxygen species, DNA methylation, and DNA damage: a comprehensive analysis in human small airway epithelial cells, macrophages, and lymphoblasts Pirela SV, Miousse IR, Lu X, Castranova V, Thomas T, Qian Y, Bello D, Kobzik L, Koturbash I, Demokritou P. Environ Health Perspect. 2015 Jun 16.

Pathway-Focused Genetic Evaluation of Immune and Inflammation Related Genes with Chronic Fatigue Syndrome. Rajeevan MS, Dimulescu I, Murray J, Falkenberg VR, Unger ER. Hum Immunol. 2015 Jun 24. pii: S0198-8859(15)00180-9.

View previous CDC-authored publications

Read more:
Genomics|Update|Current

Pharmacogenomic Testing Services | Personalized … – DNA stat

Welcome to DNA Stat. We specialize in personalized medicine services, specifically in the pain management and pharmacogenomics arena. We take pride in both our research and unsurpassed customer service, providing clients with genetic & pharmacogenomics testing which is the fastest growing field in the medical industry today.

Pain management and pharmacogenomics is vitally important as we progress into the 21st century as it is a realization and acknowledgement that one size does not fit all when it comes to medications. What might work for one individual flawlessly could mean an adverse reaction and a trip to the emergency room for another. Genetic Testing is the tool used to determine the difference before the medication is ingested. In this way, we are spearheading and defining personalized medicine services and enabling people to recover and maintain their illnesses and conditions worry-free. By eliminating the guess work, patients can recover more fully and quicker than ever before.

We know that the medical industry can be daunting to most people. Fortunately, the genetic & pharmacogenomics testing at DNA Stat comes down to a simple Buccal swab of the cheek. No needles involved, no fear, no blood no problem. Within three weeks, the patients doctor will have in his or her hands a Pharm D Report which is the roadmap to prescribing better medications and better treatments for their patient. DNA Stat, the leader in genetic& pharmacogenomics testing, is changing the way the world sees medicine one patient at a time.

Go here to see the original:
Pharmacogenomic Testing Services | Personalized ... - DNA stat

What is pharmacogenomics? – Genetics Home Reference

Pharmacogenomics is the study of how genes affect a persons response to drugs. This relatively new field combines pharmacology (the science of drugs) and genomics (the study of genes and their functions) to develop effective, safe medications and doses that will be tailored to a persons genetic makeup.

Many drugs that are currently available are one size fits all, but they dont work the same way for everyone. It can be difficult to predict who will benefit from a medication, who will not respond at all, and who will experience negative side effects (called adverse drug reactions). Adverse drug reactions are a significant cause of hospitalizations and deaths in the United States. With the knowledge gained from the Human Genome Project, researchers are learning how inherited differences in genes affect the bodys response to medications. These genetic differences will be used to predict whether a medication will be effective for a particular person and to help prevent adverse drug reactions.

The field of pharmacogenomics is still in its infancy. Its use is currently quite 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.

The National Institute of General Medical Sciences offers a list of Frequently Asked Questions about Pharmacogenomics.

A list of Frequently Asked Questions about Pharmacogenomics is also offered by the National Human Genome Research Institute.

Additional information about pharmacogenetics is available from the Centre for Genetics Education as well as Genes In Life.

The Smithsonian National Museum of Natural Historys exhibit Genome: Unlocking Lifes Code discusses the utility of pharmacogenomics.

The Genetic Science Learning Center at the University of Utah offers an interactive introduction to pharmacogenomics. Another interactive tutorial is available from the PHG Foundation.

The American Medical Association explains what pharmacogenomics is and provides a list of practical applications.

The National Genetics and Genomics Education Centre of the National Health Service (UK) provides information about predicting the effects of drugs based on a persons genes.

See the original post here:
What is pharmacogenomics? - Genetics Home Reference

Pharmacogenomics – Wikipedia, the free encyclopedia

Pharmacogenomics (a portmanteau of pharmacology and genomics) is the study of the role of genetics in drug response. It deals with the influence of acquired and inherited genetic variation on drug response in patients by correlating gene expression or single-nucleotide polymorphisms with drug absorption, distribution, metabolism and elimination, as well as drug receptor target effects.[1][2][3] The term pharmacogenomics is often used interchangeably with pharmacogenetics. Although both terms relate to drug response based on genetic influences, pharmacogenetics focuses on single drug-gene interactions, while pharmacogenomics encompasses a more genome-wide association approach, incorporating genomics and epigenetics while dealing with the effects of multiple genes on drug response.[4][5][6]

Pharmacogenomics aims to develop rational means to optimize drug therapy, with respect to the patients' genotype, to ensure maximum efficacy with minimal adverse effects.[7] Through the utilization of pharmacogenomics, it is hoped that drug treatments can deviate from what is dubbed as the one-dose-fits-all approach. It attempts to eliminate the trial-and-error method of prescribing, allowing physicians to take into consideration their patients genes, the functionality of these genes, and how this may affect the efficacy of the patients current and/or future treatments (and where applicable, provide an explanation for the failure of past treatments).[4] Such approaches promise the advent of "personalized medicine"; in which drugs and drug combinations are optimized for each individual's unique genetic makeup.[8][9] Whether used to explain a patients response or lack thereof to a treatment, or act as a predictive tool, it hopes to achieve better treatment outcomes, greater efficacy, minimization of the occurrence of drug toxicities and adverse drug reactions (ADRs). For patients who have lack of therapeutic response to a treatment, alternative therapies can be prescribed that would best suit their requirements. In order to provide pharmacogenomic-based recommendations for a given drug, two possible types of input can be used: genotyping or exome or whole genome sequencing.[10] Sequencing provides many more data points, including detection of mutations that prematurely terminate the synthesized protein (early stop codon).[10]

Pharmacogenomics was first recognized by Pythagoras around 510 BC when he made a connection between the dangers of fava bean ingestion with hemolytic anemia and oxidative stress. Interestingly, this identification was later validated and attributed to deficiency of G6PD in the 1950s and called favism.[11][12] Although the first official publication dates back to 1961,[13] circa 1950s marked the unofficial beginnings of this science. Reports of prolonged paralysis and fatal reactions linked to genetic variants in patients who lacked butyryl-cholinesterase (pseudocholinesterase) following administration of succinylcholine injection during anesthesia were first reported in 1956.[1][14] The term pharmacogenetic was first coined in 1959 by Friedrich Vogel of Heidelberg, Germany (although some papers suggest it was 1957). In the late 1960s, twin studies supported the inference of genetic involvement in drug metabolism, with identical twins sharing remarkable similarities to drug response compared to fraternity twins.[15] The term pharmacogenomics first began appearing around the 1990s.[11]

There are several known genes which are largely responsible for variances in drug metabolism and response. The focus of this article will remain on the genes that are more widely accepted and utilized clinically for brevity.

The most prevalent drug-metabolizing enzymes (DME) are the Cytochrome P450 (CYP) enzymes. The term Cytochrome P450 was coined by Omura and Sato in 1962 to describe the membrane-bound, heme-containing protein characterized by 450nm spectral peak when complexed with carbon monoxide.[16] The human CYP family consists of 57 genes, with 18 families and 44 subfamilies. CYP proteins are conveniently arranged into these families and subfamilies on the basis of similarities identified between the amino acid sequences. Enzymes that share 35-40% identity are assigned to the same family by an Arabic numeral, and those that share 55-70% make up a particular subfamily with a designated letter.[17] For example, CYP2D6 refers to family 2, subfamily D, and gene number 6.

From a clinical perspective, the most commonly tested CYPs include: CYP2D6, CYP2C19, CYP2C9, CYP3A4 and CYP3A5. These genes account for the metabolism of approximately 80-90% of currently available prescription drugs.[18][19] The table below provides a summary for some of the medications that take these pathways.

Also known as debrisoquine hydroxylase (named after the drug that led to its discovery), CYP2D6 is the most well-known and extensively studied CYP gene.[22] It is a gene of great interest also due to its highly polymorphic nature, and involvement in a high number of medication metabolisms (both as a major and minor pathway). More than 100 CYP2D6 genetic variants have been identified.[21]

Discovered in the early 1980s, CYP2C19 is the second most extensively studied and well understood gene in pharmacogenomics.[20] Over 28 genetic variants have been identified for CYP2C19,[23] of which affects the metabolism of several classes of drugs, such as antidepressants and proton pump inhibitors.[24]

CYP2C9 constitutes the majority of the CYP2C subfamily, representing approximately 20% of the liver content. It is involved in the metabolism of approximately 10% of all drugs, which include medications with narrow therapeutic windows such as warfarin and tolbutamide.[24][25] There are approximately 57 genetic variants associated with CYP2C9.[23]

The CYP3A family is the most abundantly found in the liver, with CYP3A4 accounting for 29% of the liver content.[20] These enzymes also cover between 40-50% of the current prescription drugs, with the CYP3A4 accounting for 40-45% of these medications.[12]CYP3A5 has over 11 genetic variants identified at the time of this publication.[23]

Excerpt from:
Pharmacogenomics - Wikipedia, the free encyclopedia