Search This Blog

Thursday, March 28, 2019

Environmental impact of pharmaceuticals and personal care products

From Wikipedia, the free encyclopedia

A vervet monkey with a stolen box of aspirin that was not securely stored.
 
The environmental effect of pharmaceuticals and personal care products (PPCPs) is currently being widely investigated. PPCPs include substances used by individuals for personal health or cosmetic reasons and the products used by agribusiness to boost growth or health of livestock. More than twenty million tons of PPCPs are produced every year. PPCPs have been detected in water bodies throughout the world. The effects of these chemicals on humans and the environment are not yet known, but to date there is no scientific evidence that they affect human health.

Further research is needed to evaluate the risks of toxicity, persistence, and bioaccumulation. PPCPs encompass environmental persistent pharmaceutical pollutants (EPPPs) and are one type of persistent organic pollutants. They are not removed from wastewater by conventional methods. The European Union has declared pharmaceutical residues with the potential of contamination of water and soil to be "priority substances".

Overview

Since the 1990s, water contamination by pharmaceuticals has been an environmental issue of concern. In addition, it is important to note that many public health professionals in the United States began writing reports of pharmaceutical contamination in waterways in the 1970s.” Most pharmaceuticals are deposited in the environment through human consumption and excretion, and are often filtered ineffectively by municipal sewage treatment plants which are not designed to manage them. Once in the water, they can have diverse, subtle effects on organisms, although research is still limited. Pharmaceuticals may also be deposited in the environment through improper disposal, runoff from sludge fertilizer and reclaimed wastewater irrigation, and leaky sewer pipes. In 2009, an investigative report by Associated Press concluded that U.S. manufacturers had legally released 271 million pounds of compounds used as drugs into the environment, 92% of which was the industrial chemicals phenol and hydrogen peroxide, which are also used as antiseptics. It could not distinguish between drugs released by manufacturers as opposed to the pharmaceutical industry. It also found that an estimated 250 million pounds of pharmaceuticals and contaminated packaging were discarded by hospitals and long-term care facilities. The series of articles led to a hearing conducted by the U.S. Senate Subcommittee on Transportation Safety, Infrastructure Security, and Water Quality. This hearing was designed to address the levels of pharmaceutical contaminants in U.S. drinking water. This was the first time that pharmaceutical companies were questioned about their waste disposal methods. "No federal regulations or laws were created as a result of the hearing." "Between the years of 1970-2018 more than 3000 pharmaceutical chemicals were manufactured, but only 17 are screened or tested for in waterways." Alternately, "There are no studies designed to examine the effects of pharmaceutical contaminated drinking water on human health.” In parallel, the European Union is the second biggest consumer in the world (24% of the world total) after the USA and in the majority of EU Member States, around 50% of unused human medicinal products is not collected to be disposed of properly. In the EU, between 30 and 90% of the orally administered doses are estimated to be excreted as the active substances in the urine.

The term environmental persistent pharmaceutical pollutants (EPPP) was suggested in the 2010 nomination of pharmaceuticals and environment as an emerging issue to Strategic Approach to International Chemicals Management (SAICM) by the International Society of Doctors for the Environment (ISDE).

Safe disposal

Depending on the sources and ingredients, there are various ways in which the public can dispose of pharmaceutical and personal care products in acceptable ways. The most environmentally safe disposal method is to take advantage of a community drug take-back programs that collect drugs at a central location for proper disposal. Several local public health departments in the United States have initiated these programs. In addition, the United States Drug Enforcement Administration (DEA) periodically promotes local take-back programs, as well as the National Take Back Initiative.

Currently, take-back programs are funded by state or local health departments or are volunteer programs through pharmacies or health care providers. In recent years, the proposition that pharmaceutical companies should be responsible for their products “from the cradle to the grave,” has been gaining traction. This philosophy suggests that the manufacturers should fund the proper disposal of pharmaceutical products. Take-back programs should exist in every community, and, if further information is required on this, city officials should be contacted. If no such program exists locally, the Environmental Protection Agency and the Office of National Drug Control Policy suggest that consumers do the following:
  1. take the prescription drugs out of their original containers;
  2. mix drugs with cat litter or used coffee grounds;
  3. place the mixture into a disposable container with a lid, such as a sealable bag;
  4. cover up any personal identification with a black marker that is on the original pill containers;
  5. place these containers in the bag with the mixture, seal them, and place them in the trash.
This will hopefully keep these chemicals separated from the open environment and especially water bodies long enough for them to naturally break down. 

When these substances find their way into water, it is much more difficult to deal with them. Water treatment facilities use different processes in order to minimize or fully eliminate these pollutants. This is done by using sorption where suspended solids are removed by sedimentation. Another method used is biodegradation, and through this method microorganisms, such as bacteria and fungi, feed on or break down these pollutants thus eliminating them from the contaminated media.

Types

Illicit drugs such as ecstasy (above) can be found in waterways.
 
Pharmaceuticals, or prescription and over-the-counter medications made for human use or veterinary or agribusiness purposes, are common PPCPs found in the environment. There are nine classes of pharmaceuticals included in PPCPs: hormones, antibiotics, lipid regulators, nonsteroidal anti-inflammatory drugs, beta-blockers, antidepressants, anticonvulsants, antineoplastics, and diagnostic contrast media.

Personal care products have four classes: fragrances, preservatives, disinfectants, and sunscreen agents. These products may be found in cosmetics, perfumes, menstrual care products, lotions, shampoos, soaps, toothpastes, and sunscreen. These products typically enter the environment when passed through or washed off the body and into the ground or sewer lines, or when disposed of in the trash, septic tank, or sewage system.

Traces of illicit drugs can be found in waterways and may even be carried by money.

Routes into the environment

More attention has been devoted of late to PPCPs in the environment. Two causes may contribute to this: PPCPs are actually increasing in the environment due to widespread use and/or analytical technology is better able to detect PPCPs in the environment. These substances enter the environment directly or indirectly. Direct methods include contamination of surface water by hospitals, households, industries, or wastewater treatment plants. Direct contamination can also affect the sediment and soil.

It is generally assumed (albeit hardly verified) that the production of pharmaceuticals in industrialised countries is well controlled and unharmful to the environment, due to the local legal restrictions usually required to permit production. However, a substantial fraction of the global production of pharmaceuticals takes place in low-cost production countries like India and China. Recent reports from India demonstrate that such production sites may emit very large quantities of e.g. antibiotics, yielding levels of the drugs in local surface waters higher than those found in the blood of patients under treatment.

The major route for pharmaceutical residues to reach the aquatic environment is most probably by excretion from patients undergoing pharma treatment. Since many pharmaceutical substances are not metabolized in the body they may be excreted in biologically active form, usually via the urine. Furthermore, many pharmaceutical substances are not fully taken up from the intestine (following oral administration in patients) into their blood stream. The fraction not taken up into the blood stream will remain in the gut and eventually be excreted via the faeces. Hence, both urine and faeces from treated patients contain pharmaceutical residues. Between 30 and 90% of the orally administered dose is generally excreted as active substance in the urine.

An additional source to environmental pollution with pharmaceuticals is improper disposal of unused or expired drug residues. In European countries take-back systems for such residues are usually in place (although not always utilized to full extent) while in e.g. the US only voluntary initiatives on a local basis exist. Though most of the waste goes to incineration and people are asked to throw unused or expired pharmaceuticals into the household waste investigations in Germany showed that up to 24% of liquid pharmaceuticals and 7% of tablets or ointments are disposed always or at least “rarely” via the toilet or sink.

Proper destruction of pharma residues should yield rest products without any pharmaceutical or ecotoxic activity. Furthermore, the residues should not act as components in the environmental formation of new such products. Incineration at a high temperature (more than 1000 degrees Celsius) is considered to fulfill the requirements, but even following such incineration residual ashes from the incineration should be properly taken care of.

Pharmaceuticals used in veterinary medicine, or as additives to animal food, pose a different problem, since they are excreted into soil or possibly open surface waters. It is well known that such excretions may affect terrestrial organisms directly, leading to extinction of exposed species (e.g. dung-beetles). Lipid-soluble pharma residues from veterinary use may bind strongly to soil particles, with little tendency to leak out to ground water or to local surface waters. More water-soluble residues may be washed out with rain or melting snow and reach both ground water and surface water streams.

Presence in the environment

Methods of PPCP entry into the environment from residential homes via septic and sewage systems.
 
The use of pharmaceuticals and personal care products (PPCPs) is on the rise with an estimated increase from 2 billion to 3.9 billion annual prescriptions between 1999 and 2009 in the United States alone. PPCPs enter into the environment through individual human activity and as residues from manufacturing, agribusiness, veterinary use, and hospital and community use. In Europe, the input of pharmaceutical residues via domestic waste water is estimated to be around 80% whereas 20% is coming from hospitals. Individuals may add PPCPs to the environment through waste excretion and bathing as well as by directly disposing of unused medications to septic tanks, sewers, or trash. Because PPCPs tend to dissolve relatively easily and do not evaporate at normal temperatures, they often end up in soil and water bodies. 

Some PPCPs are broken down or processed easily by a human or animal body and/or degrade quickly in the environment . However, others do not break down or degrade easily. The likelihood or ease with which an individual substance will break down depends on its chemical makeup and the metabolic pathway of the compound.

A 2002 study by the U.S. Geological Survey found detectable quantities of one or more chemicals in 80 percent of a sampling of 139 susceptible streams in 30 states. The most common pharmaceuticals detected were nonprescription drugs; detergents, fire retardants, pesticides, natural and synthetic hormones, and an assortment of antibiotics and prescription medications were also found.

A 2006 study found detectable concentrations of 28 pharmaceutical compounds in sewage treatment plant effluents, surface water, and sediment. The therapeutic classes included antibiotics, analgesics and anti-inflammatories, lipid regulators, beta-blockers, anti-convulsant, and steroid hormones. Although most chemical concentrations were detected at low levels (nano-grams/Liter (ng/L)), there are uncertainties that remain regarding the levels at which toxicity occurs and the risks of bioaccumulation of these pharmaceutical compounds.

A study published in late 2014 reported a spike in the levels of ecstasy, ketamine, caffeine and acetaminophen in nearby rivers coinciding with a Taiwanese youth event attended by around 600,000 people. In 2018, shellfish in Puget Sound, waters that receive treated sewage from the Seattle area, tested positive for oxycodone.

Besides the identified input from human medicine there appears diffuse pollution for example from pharmaceuticals used in agriculture, too. Investigations in Germany, France and Scotland showed traces of PPCPs upstream of waste water treatment plant effluents to rivers, too.

Effects

Human

The scope of human exposure to pharmaceuticals and personal care products from the environment is a complex function of many factors. These factors include the concentrations, types, and distribution of pharmaceuticals in the environment; the pharmacokinetics of each drug; the structural transformation of the chemical compounds either through metabolism or natural degradation processes; and the potential bioaccumulation of the drugs. More research is needed to determine the effects on humans of long-term exposure to low levels of PPCPs. The full effects of mixtures of low concentrations of different PPCPs is also unknown.

"The U.S. EPA risk assessment states that the acceptable daily intake (ADI) of pharmaceuticals is around 0.0027 mg/kg‐day." Due to the lack of research of toxicity guidelines and their effects on human health it is difficult to determine a healthy dosage for water contaminated by pharmaceuticals. "The pharmaceutical sample size tested does not give a full representation of human exposure. Only 17 out of 3000 prescriptions are screened for in drinking water."

In addition, “The EPA and FDA regulations state that a drug or chemical is not considered harmful until clear evidence shows that a substance causes harm". This means that we are not testing or screening for thousands of contaminants in our drinking water. Health risk assessments have not been conducted to provide concrete evidence to link pharmaceutical contamination and adverse human health effects. 

"However adverse health outcomes are displayed in aquatic organisms. Fish living near water treatment plants have been reported to be feminized." "Some male fish started to develop ovaries and other feminized characteristic due to pharmaceutical pollution some species have decreased in population due to exposure of EE2 and other hormonal ECD substances."

Although research has shown that PPCPs are present in water bodies throughout the world, no studies have shown a direct effect on human health. However, the absence of empirical data cannot rule out the possibility of adverse outcomes due to interactions or long-term exposures to these substances. Because the amounts of these chemicals in the water supply may be in the parts per trillion or parts per billion, it is difficult to chemically determine the exact amounts present. Many studies have therefore been focused to determining if the concentrations of these pharmaceuticals exist at or above the accepted daily intake (ADI) at which the designed biological outcomes can occur.

PPCPS: shelves with tampons, women's sanitary towels, tooth brushes, health and body care products
 
In addition to the growing concerns about human health risks from pharmaceutical drugs via environmental exposures, many researchers have speculated about the potential for inducing an antibiotic resistance. One study found 10 different antibiotics in sewage treatment effluents, surface water, and sediments. Some microbiologists believe that if antibiotic concentrations are higher than the minimum inhibitory concentrations (MICs) of a species of pathogenic bacteria, a selective pressure would be exerted and, as a result, antibiotic resistance would be selectively promoted. It has also been proven that at even sub-inhibitory concentrations (e.g., one-fourth of the MIC), several antibiotics are able to have an effect on gene expression (e.g., as shown for the modulation of expression of toxin-encoding genes in Staphylococcus aureus).

For reference the MIC of erythromycin that is effective against 90 percent of lab grown Campylobacter bacteria, the most common food-borne pathogen in the United States, is 60 ng/mL. One study found that the average concentration of erythromycin, a commonly prescribed antibiotic, was 0.09 ng/mL in water treatment plant effluents,. Additionally, transfer of genetic elements among bacteria has been observed under natural conditions in wastewater treatment plants, and selection of resistant bacteria has been documented in sewers receiving wastewaters from pharmaceutical plants. Moreover, antibiotic resistant bacteria may also remain in sewage sludge and enter the food chain if the sludge is not incinerated but used as fertilizer on agricultural land.

The relationship between risk perception and behavior is multifaceted. Risk management is most effective once the motivation behind the behavior of disposing unused pharmaceuticals is understood. There was little correlation found between the perception of risk and knowledge regarding pharmaceutical waste according to a study conducted by Cook and Bellis in 2001. This study cautioned against the effectiveness of attempting to change the public’s behavior on these health issues by warning them of the risks associated with their actions.

It is advised to take careful measures to inform the public in a way that does not impart guilt but rather public awareness. For example, a study carried out by Norlund and Garvill in Sweden (2003) that found that some people may make a personal sacrifice in terms of comfort because they feel that it would be helpful to reduce further environmental damage caused by the use of cars. Awareness of air pollution problems was a factor in their decision to take action on a more environmentally favorable choice of transportation. Thus, the goal of Bound’s project encapsulates whether the perception of risk associated with pharmaceuticals has an effect on the way in which medication is commonly disposed. 

In order to conduct this study, the pharmaceuticals were grouped by their therapeutic action in order to help participants identify them. The eight therapeutic groups are listed below: antibacterials, antidepressants, antihistamines, antiepileptics, hormone treatments, and lipid regulators. Next, a survey was created to examine the disposal patterns of the participants and their perception of the existing risk or threat against the environment. Respondents were asked the following questions in part one of the survey: 1. When and how they disposed of pharmaceuticals. 2. How they perceive the risk to the environment posed by pharmaceuticals. 3. To differentiate between the risks associated with different classed of pharmaceuticals. Part two of the survey involved each of the eight pharmaceutical groups described above individually. Finally, the third part asked information about the age, sex, profession, postcode, and education of participants. The sample size of participants was precise in comparison to the actual distribution of males and females in the UK: Sample- 54.8 percent were female and 45.2 percent male vs. Actual- the UK of 51.3 percent female to 48.7 percent male. Results showed that when a medication must be discarded, 63.2 percent of participants throw them in a bin, 21.8 percent return them to a pharmacist, and 11.5 percent dispose of them via the toilet/sink, while the remaining 3.5 percent keep them. Only half of the respondents felt like pharmaceuticals could potentially be harmful to the environment. Upon examination of factors relevant to risk perception, there was no definite link found between perception and education or income.

Dr. Bound noted that participation in altruistic activities such as Environmental Conservation groups may provide members with the ability to better grasp the effects of their actions in the environment. In regards to the aquatic environment, it is hard for one to perceive the favorable effects of properly disposing medication. There also exists the plausibility that a person’s behavior will only be affected if there is a severe risk to themselves or humans as opposed to an environmental threat. Even though there are serious threats of pharmaceutical pollution resulting in the feminization of certain fish, they have a lower priority because they are not easily understood or experienced by the general public. In Jonathan P. Bound’s opinion, the provision of information about exactly how to go about disposing unused medication properly in conjunction with risk education may have a more positive and forceful effect.

Recommendations

Several recommendations and initiatives have been made to prevent pharmaceutical pollution in the environment. Important practices include:
  • Educating patients on the importance of proper unused drug disposal,
  • Educating physicians and patients of proper drug disposal,
  • Encouraging pharmaceutical industries to implement strategies for proper disposal of drugs or recycling strategies, and
  • Requiring hospitals to implement better management practices for disposing pharmaceutical waste.
First, it is imperative that patients become educated on pharmaceutical pollution and its hazardous effects on humans, animals, and the overall environment. By educating patients on proper disposal of unused drugs, steps are being taken to further prevent pharmaceutical waste in the environment. Consumers should take precautions before tossing out drugs in the trash or flushing them down the toilet. Community take-back programs have been set up for consumers to bring back unused drugs for proper disposal. Another initiative is for pharmacies to serve as a take-back site for proper drug disposal such as implementing recycling bins for customers to bring back unused or expired medicines while they’re shopping. In addition, medical foundations could receive these medicines to administer them to people who need them, while destroying those that are in excess or expired. Furthermore, educating physicians and patients on the importance of proper drug disposal and the environmental concern will help further reduce pharmaceutical waste. 

Also, implementing initiatives for hospitals to focus on better practices for hazardous waste disposal may prove to be beneficial. The US EPA encourages hospitals to develop efficient pharmaceutical disposal practices by giving them grants. This incentive may be very beneficial to other hospitals worldwide. 

Additionally, “It is critical for us to develop an analytical method of identifying, testing, and regulating the amount of pharmaceuticals in the water systems”. Data must be collected in order to accurately measure the prevalence of pharmaceuticals in drinking water. “Multiple Health risk assessments should be conducted to understand the effects of prolonged exposure to pharmaceuticals in drinking water”.

Community-based programs should be developed to monitor exposure and health outcomes. We should encourage the pharmaceutical industry to develop technology that extracts pharmaceuticals from waterways. “Extensive research must be conducted to determine the amount of pharmaceutical contamination in the environment and its effects on animals and marine life”.

We must remember that many pharmaceuticals pass through the human body unchanged, so it is best for human excrement to not go into waterways, even after conventional treatment, which can also not remove these chemicals. It is therefore preferable for human feces and urine to go into fertile soil, where they will receive more effective treatment by numerous microbes found there, over longer amounts of time, and stay away from waterways, via the use of Urine-diverting Dry Toilets, Composting Toilets, and ArborLoos. As mentioned below, constructed wetlands are efficient at removing these chemicals, but it is better for them to not go into water in the first place.

Environmental

While the full effects of most PPCPs on the environment are not understood, there is concern about the potential they have for harm because they may act unpredictably when mixed with other chemicals from the environment or concentrate in the food chain. Additionally, some PPCPs are active at very low concentrations, and are often released continuously in large or widespread quantities. 

A class of antidepressants may be found in frogs and can significantly slow their development
 
Because of the high solubility of most PPCPs, aquatic organisms are especially vulnerable to their effects. Researchers have found that a class of antidepressants may be found in frogs and can significantly slow their development. The increased presence of estrogen and other synthetic hormones in waste water due to birth control and hormonal therapies has been linked to increased feminization of exposed fish and other aquatic organisms. The chemicals within these PPCP products could either affect the feminization or masculinization of different fishes, therefore affecting their reproductive rates.

In addition to being found only in waterways, the ingredients of some PPCPs can also be found in the soil. Since some of these substances take a long time or cannot be degraded biologically, they make their way up the food chain. Information pertaining to the transport and fate of these hormones and their metabolites in dairy waste disposal is still being investigated, yet research suggest that the land application of solid wastes is likely linked with more hormone contamination problems. Not only does the pollution from PPCPs affect marine ecosystems, but also those habitats that depend on this polluted water. 

There are various concerns about the effects of pharmaceuticals found in surface waters and specifically the threats against rainbow trout exposed to treated sewage effluents. Analysis of these pharmaceuticals in the blood plasma of fish compared to human therapeutic plasma levels have yielded vital information providing a means of assessing risk associated with medication waste in water. In a study by Dr. Jerker Fick from Umeå University rainbow trout were exposed to undiluted, treated sewage water at three different sits in Sweden. They were exposed for a total of 14 days while 25 pharmaceuticals were measured in the blood plasma at different levels for analysis. The progestin Levonorgestrel was detected in fish blood plasma at concentrations between 8.5 and 12 ng mL-1 which exceed the human therapeutic plasma level. Studies show that the measured effluent level of Levonorgestrel in the three areas was shown to reduce the fertility of the rainbow trout.

The three sites chosen for field exposures were in located in Stockholm, Gothenburg, and Umeå. They were chosen according to their varying degrees of treatment technologies, geographic locations, and size. The effluent treatment includes active sludge treatment, nitrogen and phosphorus removal (except in Umeå), primary clarification, and secondary clarification. Juvenile rainbow trout were procured from Antens fiskodling AB, Sweden and Umlax AB, Sweden. The fish were exposed to aerated, undiluted, treated effluent. Since all of the sites underwent sludge treatment, it can be inferred that they are not representative of the low end of treatment efficacy. Of the 21 pharmaceuticals that were detected in the water samples, 18 were identified in the effluent, 17 in the plasma portion, and 14 pharmaceuticals were found in both effluent and plasma.

Current research

There are traces pharmaceuticals in waterways that have an adverse effect on the environment.
 
Starting in the mid-1960s, ecologists and toxicologists began to express concern about the potential adverse effects of pharmaceuticals in the water supply, but it wasn’t until a decade later that the presence of pharmaceuticals in water was well documented. Studies in 1975 and 1977 found clofibric acids and salicylic acids at trace concentrations in treated water. Widespread concern about and research into the effect of PPCPs largely started in the early 1990s. Until this time, PPCPs were largely ignored because of their relative solubility and containment in waterways compared to more familiar pollutants like agrochemicals, industrial chemicals, and industrial waste and byproducts.

Since then, a great deal of attention has been directed to the ecological and physiological risk associated with pharmaceutical compounds and their metabolites in water and the environment. In the last decade, most research in this area has focused on steroid hormones and antibiotics. There is concern that steroid hormones may act as endocrine disruptors. Some research suggests that concentrations of ethinylestradiol, an estrogen used in oral contraceptive medications and one of the most commonly prescribed pharmaceuticals, can cause endocrine disruption in aquatic and amphibian wildlife in concentrations as low as 1 ng/L.

Current research on PPCPs aims to answer these questions:
  • What is the effect of exposure to low levels of PPCPs over time?
  • What is the effect of exposure to mixtures of chemicals?
  • Are the effects acute (short-term) or chronic (long-term)?
  • Are certain populations, such as the elderly, very young, or immuno-compromised, more vulnerable to the effects of these compounds?
  • Jaipur cows eating trash
  • Jaipur cows eating trash, which may contain medicines and supplements that will pass through their system and enter the environment
    What is the effect of PPCPs on bacterial, fungal, and aquatic life?
  • Are the levels of antibiotics in the aquatic environment sufficient to promote antibiotic resistance?
  • What is the effect of exposure to steroid hormones on animal and human populations?

Pharmacoenvironmentology

Pharmacoenvironmentology is an extension of pharmacovigilance as it deals specifically with the environmental and ecological effects of drugs given at therapeutic doses. Pharmacologists with this particular expertise (known as a pharmacoenvironmentologist) become a necessary component of any team assessing different aspects of drug safety in the environment. We must look at the effects of drugs not only in medical practice, but also at its environmental effects. Any good clinical trial should look at the impact of particular drugs on the environment. Things we need to address in pharmacoenvironmentology are drugs and their exact concentration in different parts of the environment.

Pharmacoenvironmentology is a specific domain of pharmacology and not of environmental studies.This is because it deals with drugs entering through living organisms through elimination.

Ecopharmacovigilance

Pharmacovigilance is a new branch of science, which was born in 1960 after the incidence of the thalidomide disaster. Thalidomide is a teratogen and caused horrific birth abnormalities. The thalidomide disaster lead to the present day approach to drug safety and adverse event reporting.

According to the EPA, pharamacovigilance is science aiming to capture any adverse effects of pharmaceuticals in humans after use. However, ecopharmacovigilance is the science, and activities concerning detection, assessment, understanding, and prevention of adverse effects of pharmaceuticals in the environment which affect humans and other animal species. There has been a growing focus among scientists about the impact of drugs on the environment. In recent years, we have been able to see human pharmaceuticals that are being detected in the environment which most are typically found on surface water.

The importance of ecopharmacovigilance is to monitor adverse effects of pharmaceuticals on humans through environmental exposure. Due to this relatively new field of science, researchers are continuously developing and understanding the impacts of pharmaceuticals in the environment and its risk on human and animal exposure. Environmental risk assessment is a regulatory requirement in the launch of any new drug. This precaution has become a necessary step towards the understanding and prevention of adverse effects of pharmaceutical residue in the environment. It is important to note that pharmaceuticals enter the environment from the excretion of drugs after human use, hospitals, and improper disposal of unused drugs from patients.

Ecopharmacology

Ecopharmacology concerns the entry of chemicals or drugs into the environment through any route and at any concentration disturbing the balance of ecology (ecosystem), as a consequence. Ecopharmacology is a broad term that includes studies of “PPCPs” irrespective of doses and route of entry into environment.

The geology of a karst aquifer area assists with the movement of PPCPs from the surface to the ground water. Relatively soluble bedrock creates sinkholes, caves and sinking streams into which surface water easily flows, with minimal filtering. Since 25% of the population get their drinking water from karst aquifers, this affects a large number of people. A 2016 study of karst aquifers in southwest Illinois found that 89% of water samples had one or more PPCP measured. Triclocarban (an antimicrobial) was the most frequently detected PPCP, with gemfibrozil (a cardiovascular drug) the second most frequently detected. Other PPCPs detected were trimethoprim, naproxen, carbamazepine, caffeine, sulfamethoxazole, and fluoxetine. The data suggests that septic tank effluent is a probable source of PPCPs.

Fate of pharmaceuticals in sewage treatment plants

Sewage treatment plants use physical, chemical, and biological processes to remove nutrients and contaminants from waste water.
 
Sewage treatment plants (STP) work with physical, chemical, and biological processes to remove nutrients and contaminants from waste water. Usually the STP is equipped with an initial mechanical separation of solid particles (cotton buds, cloth, hygiene articles etc.) appearing in the incoming water. Following this there may be filters separating finer particles either occurring in the incoming water or developing as a consequence of chemical treatment of the water with flocculating agents.

Many STPs also include one or several steps of biological treatment. By stimulating the activity of various strains of microorganisms physically their activity may be promoted to degrade the organic content of the sewage by up to 90% or more. In certain cases more advanced techniques are used as well. The today most commonly used advanced treatment steps especially in terms of micropollutants are:
PPCPs are difficult to remove from wastewater with conventional methods. Some research shows the concentration of such substances is even higher in water leaving the plant than water entering the plant. Many factors including environmental pH, seasonal variation, and biological properties affect the ability of a STP to remove PPCPs.

A 2013 study of a drinking water treatment plant, found that of 30 PPCPs measured at both the source water and the drinking water locations, 76% of PPCPs were removed, on average, in the water treatment plant. Ozonation was found to be an efficient treatment process for the removal of many PPCPs. However, there are some PPCPs that were not removed, such as DEET used as mosquito spray, nonylphenol which is a surfactant used in detergents, the antibiotic erythromycin, and the herbicide atrazine.

Several research projects are running to optimize the use of advanced sewage treatment techniques under different conditions. The advanced techniques will increase the costs for the sewage treatment substantially. In a European cooperation project between 2008 and 2012 in comparison 4 hospital waste water treatment facilities were developed in Switzerland, Germany, The Netherlands and Luxembourg to investigate the elimination rates of concentrated waste water with pharmaceutical “cocktails” by using different and combined advanced treatment technologies. Especially the German STP at Marienhospital Gelsenkirchen showed the effects of a combination of membranes, ozone, powdered activated carbon and sand filtration. But even a maximum of installed technologies could not eliminate 100% of all substances and especially radiocontrast agents are nearly impossible to eliminate. The investigations showed that depending on the installed technologies the treatment costs for such a hospital treatment facility may be up to 5.50 € per m2. Other studies and comparisons expect the treatment costs to increase up to 10%, mainly due to energy demand. It is therefore important to define best available technique before extensive infrastructure investments are introduced on a wide basis. 

The fate of incoming pharmaceutical residues in the STP is unpredictable. Some substances seem to be more or less completely eliminated, while others pass the different steps in the STP unaffected. There is no systematic knowledge at hand to predict how and why this happens. 

Pharmaceutical residues that have been conjugated (bound to a bile acid) before being excreted from the patients may undergo de-conjugation in the STP, yielding higher levels of free pharmaceutical substance in the outlet from the STP than in its incoming water. Some pharmaceuticals with large sales volumes have not been detected in the incoming water to the STP, indicating that complete metabolism and degradation must have occurred already in the patient or during the transport of sewage from the household to the STP.

Regulation

In the United States, “There are no federal regulations limiting the levels of pharmaceuticals in wastewater or drinking water”, according to EPA. Three birth controls substances and one antibiotic were added to the pharmaceuticals contaminant candidate list (CCL 3). EPA states that they are 8 pharmaceuticals that are classified as hazardous waste. Out of 3000 pharmaceuticals EPA has only evaluated 100 of them leaving numerous of unscreened compounds in the water.” In 2008, EPA proposed an amendment to the Universal Waste Rule to address pharmaceutical wastes.” However, no action on the Rule has occurred since the hearing in the U.S. senate in 2009”.

The Clean Water Act (CWA), Safe Drinking Water Act (SDWA), and the Universal Waste Regulations under the Resource Conservation and Recovery Act (RCRA) do little to protect the American population from pharmaceutical contamination in waterways. The EPA has the ability to implement governmental regulations to insure that the public has access to water that is free of pharmaceutical pollution however, no laws have been enacted even though “ the U.S. Government Accountability Office (GAO) reports that some research has demonstrated potential impact on human health from exposure to pharmaceuticals found in drinking water, such as antibiotics and EE2 substances that interfere with human hormone development”.

The EPA and other governmental agencies are not focused on this issue. They are not demanding any changes or regulations for Pharmaceutical pollution. They did not review the 3000 Pharmaceutical substances to determine their effect on human health. There are no long term studies or health assessments created to examine how this affects us or the environment.

Treatment methods

PPCPs are emerging pollutants, which pose potential risks on the environment and health. They are the result of many sectors, such as household consumption, industrial, farming, medicine, and aquaculture. However, because of the toxic and intractable characteristics, these pollutants cannot be removed by a traditional wastewater treatment plant in an effective way. Hence, they are becoming more and more universal. The fact that conventional wastewater treatment processes could not eradicate PPCPs completely has been highlighted. In final effluents, high concentrations of PPCPs can be detected, and these contaminants can accumulate in rivers, sludge, soil, and biosolids. A study pointed out that organic pollutants, including PPCPs, appeared in 80% of 139 U.S. streams from 1999 to 2000. As such, environmental pollution caused by PPCPs are becoming more and more serious. Therefore, the removal of PPCPs is an urgent topic in the treatment of wastewater. Up to now, the methods used for the removal of PPCPs can be roughly divided into three categories, including physical, chemical, and biological methods.

Physical adsorption processes

The most common physical process is adsorption. It is usually used to remove traceable organic pollutants in water. Generally, carbon-based materials, which include biochar, activated carbon, graphene and graphene oxide, and carbon nanotubes, are applied to adsorb PPCPs from aqueous solution. Different materials have a different adsorption capacity for PPCPs. As for these materials, carbon nanotubes have the highest adsorption capacity, while the adsorption capacity adjusted with the surface chemistry and properties of carbon nanotubes. By comparison, activated carbon as a traditional adsorbent has a wide application in adsorption of PPCPs from wastewater. Although physical adsorption processes are regarded as an advanced technology, and developing fast in the field of treatment methods, there are still some problems that should be addressed in future research, like the production of carbon nanotubes, the high cost of graphene, and the absorption of macromolecular substances.

Biological degradation processes

Microbial degradation is regarded as an essential method for the removal of organic pollutants. The processes have numerous advantages, like economical input costs, and mild operational conditions. In these processes, microorganisms can make use of their metabolic functions to degrade the contaminants and in some cases several microbes cooperate with each other to achieve this goal. Pure cultures, mixed cultures, and activated sludge processes can be used in these processes, while an activated sludge process is widely used as a biological treatment in the conventional wastewater treatment plants. Constructed wetlands show great promise and are discussed below.

Chemical advanced oxidation processes

Advanced chemical oxidation processes are effective in removing PPCPs from wastewater. These include ozonation, Fenton oxidation, ionizing radiation, and UV treatment, are efficient in the degradation of recalcitrant organic pollutants in aqueous solution. Among these, ozonation is the most widely used oxidation method for the removal of PPCPs.

Combined chemical and biological methods

Because some PPCPs are highly resistant and toxic to microorganisms, it can be difficult for biological methods to eliminate them on their own. On the other hand, advanced chemical oxidation processes behave well in removing persistent pollutants, but the intermediates produced during the treatment process are sometimes more resistant to oxidation. Thus, if using one of these two methods to treat pollutants, the process requires long times and high energy inputs, resulting in a high cost. In order to overcome the shortfalls of using a single method, combinations of biological and chemical processes have increasingly been applied in recent years. In the first stage, advanced chemical oxidation processes are used for pre-treatment to transform persistent pollutants into the biodegradable intermediates. Then, biological methods are applied to degrade them. Combined systems are being shown to be efficient and cost effective.

Examples

Blister packs

80% of pills in the world are packed with blister packaging, which is the most convenient type for several reasons. Blister packs have two main components, the “lid” and the “blister” (cavity). Lid is mainly manufactured with aluminum (Al) and paper. The Cavity consists of polyvinyl chloride (PVC), polypropylene (PP), polyester (PET) or aluminum (Al). If users employ proper disposal methods, all these materials can be recycled and the harmful effects to the environment can be minimized. However, a problem arises with the improper disposal either by burning or disposing as normal household waste. 

Burning of blister packs directly causes air pollution by the combustion products of polypropylene ([C3H6]n), polyester ([C10H8O4]n), and polyvinyl chloride ([CH2CHCl]n). The combustion reactions and products of these chemicals are mentioned below. 

The basic configuration of blister packaging
[C3H6]n + 9n/2 O2 → 3n CO2 +3n H2O
[C10H8O4]n + 10n O2 → 10n CO2 +4n H2O
[CH2CHCl]n + 2n O2 → n CO2 + n H2O + n HCl + n CO 

Even though polypropylene and polyester is harmful to the environment, the most toxic effect is due to the combustion of polyvinyl chloride since it produces hydrochloric acid (HCl) which is an irritant in the lower and upper respiratory tract that can cause adverse to human beings.

The disposal of blister packs as normal waste, will forbid recycling process and eventually accumulate in soil or water, which will result soil and water pollution since bio-degradation processes of compounds like PVC, PP and PET are very slow. As a result, ecologically damaging effects like disturbances of the habitats and movements can be seen. Ingestion by the animals, affect the secretion of gastric enzymes and steroid hormones that can decrease the feeding stimuli and may also cause problems in reproduction. At low pH, aluminum can increase its solubility according to the following equation. As a result, the negative effects of both aquatic and terrestrial ecosystems can be generated.

2Al(s)+ 6H+ → 2Al3+ (aq) + 3H2 (g) 

By employing proper disposal methods, all manufacturing materials of blister packs like PP, PE, PVC and Al can be recycled and the adverse effects to the environment can be minimized. Even though, the synthesis of these polymers relatively simple, the recycling process can be very complex since the blister packs contain metals and polymers together.

As the first step of recycling, separation of Al and Polymers using the hydrometallurgical method which uses hydrochloric acid (HCl)  can be incorporated. Then PVC can be recycled by using mechanical or chemical methods. The most recent trend is to use biodegradable, eco-friendly “bio plastics” which are also called as biopolymers such as derivatives of starch, cellulose, protein, chitin and xylan for pharmaceutical packaging, to reduce the hostile effects to the environment.

Nail polish remover

Nail polish remover has the ability to enter bodies of water and soil after entering landfills or by precipitation, such as rain or snow. However, due to acetone's high volatility, most of it that enters the bodies of water and soil will evaporate again and re-enter the atmosphere. Not all of the acetone molecules will evaporate again, and so, when acetone remains in the bodies of water or soil, a reaction will occur. Nail polish remover evaporates easily because acetone's intermolecular forces are weak. An acetone molecule can't attract other acetone molecules easily because its hydrogens are not slightly positive. The only force that holds acetone molecules together is its permanent dipoles which are weaker than hydrogen bonds.

Nail polish remover contains acetone.
 
Since nail polish remover is a solvent, it will dissolve in water. When acetone dissolves in water, it hydrogen bonds with water. The more nail polish remover that enters the hydrosphere will increase the concentration of acetone and then increase the concentration of the solution created when acetone and water bonds. If enough nail polish remover is disposed, it can reach the lethal dose level for aquatic life. 

Nail polish remover can also enter the lithosphere by landfills and by precipitation. However, it will not bind to the soil. Microorganisms in the soil will decompose acetone. The consequence of microorganisms decomposing acetone is the risk it has to cause oxygen depletion in bodies of water. The more acetone readily available for microorganism decomposition leads to more microorganisms reproduced and thus oxygen depletion because more microorganisms use up the available oxygen. 

When nail polish remover evaporates, acetone enters the atmosphere in the gaseous phase. In the gaseous phase, acetone can undergo photolysis and breakdown into carbon monoxide, methane, and ethane. When temperatures are between 100 - 350 degrees Celsius, the following mechanism occurs:

(CH3)2CO + hv → CH3 + CH3CO
CH3CO → CH3+ CO
CH3+ (CH3)2CO → CH4 + CH2COCH3
2CH3 → C2H6

A second pathway that nail polish remover can enter in the atmosphere is reacting with hydroxyl radicals. When acetone reacts with hydroxyl radicals, its main product is methylglyoxal. Methylglyoxal is an organic compound that is a by-product of many metabolic pathways. It is an intermediate precursor for many advanced glycation end-products, that are formed for diseases such as diabetes or neurodegenerative diseases. The following reaction occurs: 

(CH3)2CO + ·OH → CH3C(O)OH + ·CH3
CH3C(O)OH + ·CH3→ CH3C(O)COH + 3H+

Case study

The Location of the Pearl River Delta

Pearl River, South China

It is well known that releasing antibiotics into the water can lead to antibiotic resistance. In the recent decade, the Pearl River has been altered by antibiotics. A study found that concentrations of antibiotics were lower than those in the estuary, and decreased from the east side to the west side. A series of phenomena showed that the primary sources of antibiotics came from river tributaries and terrigenous sources and there were frequent human activities and hydraulic conditions along the west banks of the river. Seasonal variations happened in most of the detected antibiotics in water as well. Additionally, the TOC content of sediments, and the pH of water affected the performance of antibiotics. The highest concentrations of antibiotics were found downstream of Guangdong Province, which was affected severely by nearby densely populated megacities, like Shenzhen, Hong Kong, and Macau. These have an adverse impact on people there.

The coast of Guangdong, South China

The coast of Guangdong is a place polluted by another representative contaminant, plastic debris. After an extended period of observation, scientists found that the beach was a place that would help the fragmentation of plastic debrisinto highly fragmented plastic particles that can attach to small organisms. Through the process of washback, the plastic debris may be transported into the water, and make an adverse impact on a wide range of marine life at different trophic levels. It is noticeable that polystyrene (PS) foams and fragments, and microplastics account for a great percentage of plastic debris on the beach. Most of the polystyrenes degrade within a year on average, and could not be observed in deep beach sediment. Thus, coastal beaches store highly fragmented and degraded micro-plastics. However, they may be active and return to the water.

An alternative best management practice for PPCPs treatment – the application of constructed wetlands

Structures of constructed wetlands: (a) surface flow (SF-CW); (b) horizontal subsurface flow (HSSF-CW); and (c) vertical subsurface flow (VSSF-CW).
 
In recent years, scientists have been trying to find a way to handle PPCPs, but most of the advanced treatment processes are too expensive to apply in large scale. As such, a low-cost alternative approach for PPCPs treatment is of great significance for the environment. Based on this purpose, increasing concerns have been raised on constructed wetlands, which are cheaper in construction, operation, maintenance and monitoring. Actually, constructed wetlands have been proved to be effective for removal of conventional contaminants in wastewater treatment. However, applying constructed wetlands to deal with these issues is a totally new application in the field.

Important components of constructed wetlands

Substrate

Based on several researches and further analysis for various constructed wetlands, scientists found that gravel is the most common substrate materials for the removal of PPCPs in constructed wetlands. The gravel substrate is efficient for some certain PPCPs which are difficult to be biodegraded but with relatively high hydrophobicity. On the other hand, according to a set of experiments, it is reported that light expanded clay aggregate as a sorbent in constructed wetlands performs well for the treatment of the acidic compounds and alkaline with positive charge.

Vegetation

Vegetation plays an important part in directly taking up several organic pollutants from wastewater in constructed wetlands. With the function of diffusion, the organic pollutants like PPCPs can be transported within plants. That means, theses pollutants cannot move into the plant tissues through the plant roots and some specific transporters. Generally, the physico-chemical characteristics of PPCPs, such as hydrophobicity, water solubility, and concentration, affect the diffusion of the compounds. After being soaked up by plants, the degradation may happen in metabolism processes. Due to the activities of microbial populations, there are numerous biological processes occurring in the rhizosphere. However, the toxicity of PPCPs to plants is a nonnegligible issue.

Microbes

The processes of transformation and mineralization of nutrients and organic pollutants are primarily dependent on microbes in constructed wetlands, and the microbial degradation processes can occur in both the aerobic and the anaerobic environments with the participation of diverse microorganisms, like fungi, specific protozoa, heterotrophic bacteria, and autotrophic bacteria. The chemical structures of organic pollutants strongly affect the degradation processes. Some contaminations with simple structures owning high water solubility and low absorptivity, so degradation via microorganisms will be easier. However, for PPCPs, which are xenobiotic organic pollutants with totally different compounds, they are degraded relatively slowly. Even though there are non-specific enzymes, the rate of degradation is still slower.

Pending questions

  • Is there a temperature at which PPCPs are burned and destroyed? Would they thus be eliminated when materials are made into biochar?
  • Are there artificial colorings that degrade under similar conditions to PPCPs and could be used as proxies in low-tech experiments of how to eliminate PPCPs?
  • Ultraviolet light is known to degrade PPCPs. How long would urine need to lay in the sun in transparent bottles to destroy the PPCPs before its use as fertilizer?
  • Do soil microbes develop or evolve the ability to break down PPCPs over time? If a person who consumes a pharmaceutical uses a Urine-diverting Dry Toilet, in which the urine is dispersed into fertile soil among plants, would the microbes eventually decompose this chemical entirely? After how much time? Which types of pharmaceuticals would break down faster and which slower?
  • Are there types of PPCPs that cannot enter into the roots of plants because their molecules are simply too large?
  • When essential oils are extracted from plants, would PPCPs pass into them, stay in the cauldron, or be destroyed by the heat?

Pharmacovigilance

From Wikipedia, the free encyclopedia

Pharmacovigilance (PV or PhV), also known as drug safety, is the pharmacological science relating to the collection, detection, assessment, monitoring, and prevention of adverse effects with pharmaceutical products. The etymological roots for the word "pharmacovigilance" are: pharmakon (Greek for drug) and vigilare (Latin for to keep watch). As such, pharmacovigilance heavily focuses on adverse drug reactions, or ADRs, which are defined as any response to a drug which is noxious and unintended, including lack of efficacy (the condition that this definition only applies with the doses normally used for the prophylaxis, diagnosis or therapy of disease, or for the modification of physiological disorder function was excluded with the latest amendment of the applicable legislation). Medication errors such as overdose, and misuse and abuse of a drug as well as drug exposure during pregnancy and breastfeeding, are also of interest, even without an adverse event, because they may result in an adverse drug reaction.

Information received from patients and healthcare providers via pharmacovigilance agreements (PVAs), as well as other sources such as the medical literature, plays a critical role in providing the data necessary for pharmacovigilance to take place. In fact, in order to market or to test a pharmaceutical product in most countries, adverse event data received by the license holder (usually a pharmaceutical company) must be submitted to the local drug regulatory authority.

Ultimately, pharmacovigilance is concerned with identifying the hazards associated with pharmaceutical products and with minimizing the risk of any harm that may come to patients. Companies must conduct a comprehensive drug safety and pharmacovigilance audit to assess their compliance with worldwide laws, regulations, and guidance.

Terms commonly used in drug safety

Pharmacovigilance has its own unique terminology that is important to understand. Most of the following terms are used within this article and are peculiar to drug safety, although some are used by other disciplines within the pharmaceutical sciences as well.
  • Adverse drug reaction is a side effect (non intended reaction to the drug) occurring with a drug where a positive (direct) causal relationship between the event and the drug is thought, or has been proven, to exist.
  • Adverse event (AE) is a side effect occurring with a drug. By definition, the causal relationship between the AE and the drug is unknown.
  • Benefits are commonly expressed as the proven therapeutic good of a product but should also include the patient's subjective assessment of its effects.
  • Causal relationship is said to exist when a drug is thought to have caused or contributed to the occurrence of an adverse drug reaction.
  • Clinical trial (or study) refers to an organised program to determine the safety and/or efficacy of a drug (or drugs) in patients. The design of a clinical trial will depend on the drug and the phase of its development.
  • Control group is a group (or cohort) of individual patients that is used as a standard of comparison within a clinical trial. The control group may be taking a placebo (where no active drug is given) or where a different active drug is given as a comparator.
  • Dechallenge and rechallenge refer to a drug being stopped and restarted in a patient, respectively. A positive dechallenge has occurred, for example, when an adverse event abates or resolves completely following the drug's discontinuation. A positive rechallenge has occurred when the adverse event re-occurs after the drug is restarted. Dechallenge and rechallenge play an important role in determining whether a causal relationship between an event and a drug exists.
  • Effectiveness is the extent to which a drug works under real world circumstances, i.e., clinical practice.
  • Efficacy is the extent to which a drug works under ideal circumstances, i.e., in clinical trials.
  • Event refers to an adverse event (AE).
  • Harm is the nature and extent of the actual damage that could be or has been caused.
  • Implied causality refers to spontaneously reported AE cases where the causality is always presumed to be positive unless the reporter states otherwise.
  • Individual Case Safety Report (ICSR) is an adverse event report for an individual patient.
  • Life-threatening refers to an adverse event that places a patient at the immediate risk of death.
  • Phase refers to the four phases of clinical research and development: I – small safety trials early on in a drug's development; II – medium-sized trials for both safety and efficacy; III – large trials, which includes key (or so-called "pivotal") trials; IV – large, post-marketing trials, typically for safety reasons. There are also intermediate phases designated by an "a" or "b", e.g. Phase IIb.
  • Risk is the probability of harm being caused, usually expressed as a percent or ratio of the treated population.
  • Risk factor is an attribute of a patient that may predispose, or increase the risk, of that patient developing an event that may or may not be drug-related. For instance, obesity is considered a risk factor for a number of different diseases and, potentially, ADRs. Others would be high blood pressure, diabetes, possessing a specific mutated gene, for example, mutations in the BRCA1 and BRCA2 genes increase propensity to develop breast cancer.
  • Signal is a new safety finding within safety data that requires further investigation. There are three categories of signals: confirmed signals where the data indicate that there is a causal relationship between the drug and the AE; refuted (or false) signals where after investigation the data indicate that no causal relationship exists; and unconfirmed signals which require further investigation (more data) such as the conducting of a post-marketing trial to study the issue.
  • Temporal relationship is said to exist when an adverse event occurs when a patient is taking a given drug. Although a temporal relationship is absolutely necessary in order to establish a causal relationship between the drug and the AE, a temporal relationship does not necessarily in and of itself prove that the event was caused by the drug.
  • Triage refers to the process of placing a potential adverse event report into one of three categories: 1) non-serious case; 2) serious case; or 3) no case (minimum criteria for an AE case are not fulfilled).

Adverse event reporting

The activity that is most commonly associated with pharmacovigilance (PV), and which consumes a significant amount of resources for drug regulatory authorities (or similar government agencies) and drug safety departments in pharmaceutical companies, is that of adverse event reporting. Adverse event (AE) reporting involves the receipt, triage, data entering, assessment, distribution, reporting (if appropriate), and archiving of AE data and documentation. The source of AE reports may include: spontaneous reports from healthcare professionals or patients (or other intermediaries); solicited reports from patient support programs; reports from clinical or post-marketing studies; reports from literature sources; reports from the media (including social media and websites); and reports reported to drug regulatory authorities themselves. For pharmaceutical companies, AE reporting is a regulatory requirement in most countries. AE reporting also provides data to these companies and drug regulatory authorities that play a key role in assessing the risk-benefit profile of a given drug. The following are several facets of AE reporting:

Individual Case Safety Report (ICSR)

If one or more of these four elements is missing, the case is not a valid ICSR. Although there are no exceptions to this rule there may be circumstances that may require a judgment call. For example, the term "identifiable" may not always be clear-cut. If a physician reports that he/she has a patient X taking drug Y who experienced Z (an AE), but refuses to provide any specifics about patient X, the report is still a valid case even though the patient is not specifically identified. This is because the reporter has first-hand information about the patient and is identifiable (i.e. a real person) to the physician. Identifiability is important so as not only to prevent duplicate reporting of the same case, but also to permit follow-up for additional information. 

The concept of identifiability also applies to the other three elements. Although uncommon, it is not unheard of for fictitious adverse event "cases" to be reported to a company by an anonymous individual (or on behalf of an anonymous patient, disgruntled employee, or former employee) trying to damage the company's reputation or a company's product. In these and all other situations, the source of the report should be ascertained (if possible). But anonymous reporting is also important, as whistle blower protection is not granted in all countries. In general, the drug must also be specifically named. Note that in different countries and regions of the world, drugs are sold under various tradenames. In addition, there are a large number of generics which may be mistaken for the trade product. Finally, there is the problem of counterfeit drugs producing adverse events. If at all possible, it is best to try to obtain the sample which induced the adverse event, and send it to either the EMA, FDA or other government agency responsible for investigating AE reports. 

If a reporter can't recall the name of the drug they were taking when they experienced an adverse event, this would not be a valid case. This concept also applies to adverse events. If a patient states that they experienced "symptoms", but cannot be more specific, such a report might technically be considered valid, but will be of very limited value to the pharmacovigilance department of the company or to drug regulatory authorities.

Coding of adverse events

Adverse event coding is the process by which information from an AE reporter, called the "verbatim", is coded using standardized terminology from a medical coding dictionary, such as MedDRA (the most commonly used medical coding dictionary). The purpose of medical coding is to convert adverse event information into terminology that can be readily identified and analyzed. For instance, Patient 1 may report that they had experienced "a very bad headache that felt like their head was being hit by a hammer" [Verbatim 1] when taking Drug X. Or, Patient 2 may report that they had experienced a "slight, throbbing headache that occurred daily at about two in the afternoon" [Verbatim 2] while taking Drug Y. Neither Verbatim 1 nor Verbatim 2 will exactly match a code in the MedDRA coding dictionary. However, both quotes describe different manifestations of a headache. As a result, in this example both quotes would be coded as PT Headache (PT = Preferred Term in MedDRA).

Seriousness determination

Although somewhat intuitive, there are a set of criteria within pharmacovigilance that are used to distinguish a serious adverse event from a non-serious one. An adverse event is considered serious if it meets one or more of the following criteria:
  1. Results in death, or is life-threatening;
  2. Requires inpatient hospitalization or prolongation of existing hospitalization;
  3. Results in persistent or significant disability or incapacity;
  4. Results in a congenital anomaly (birth defect); or
  5. Is otherwise "medically significant" (i.e., that it does not meet preceding criteria, but is considered serious because treatment/intervention would be required to prevent one of the preceding criteria.)
Aside from death, each of these categories is subject to some interpretation. Life-threatening, as it used in the drug safety world, specifically refers to an adverse event that places the patient at an immediate risk of death, such as cardiac or respiratory arrest. By this definition, events such as myocardial infarction, which would be hypothetically life-threatening, would not be considered life-threatening unless the patient went into cardiac arrest following the MI. Defining what constitutes hospitalization can be problematic as well. Although typically straightforward, it's possible for a hospitalization to occur even if the events being treated are not serious. By the same token, serious events may be treated without hospitalization, such as the treatment of anaphylaxis may be successfully performed with epinephrine. Significant disability and incapacity, as a concept, is also subject to debate. While permanent disability following a stroke would no doubt be serious, would "complete blindness for 30 seconds" be considered "significant disability"? For birth defects, the seriousness of the event is usually not in dispute so much as the attribution of the event to the drug. Finally, "medically significant events" is a category that includes events that may be always serious, or sometimes serious, but will not fulfill any of the other criteria. Events such as cancer might always be considered serious, whereas liver disease, depending on its CTCAE (Common Terminology Criteria for Adverse Events) grade—Grades 1 or 2 are generally considered non-serious and Grades 3-5 serious—may be considered non-serious.

Clinical trial reporting

Also known as SAE (serious adverse event) reporting from clinical trials, safety information from clinical studies is used to establish a drug's safety profile in humans and is a key component that drug regulatory authorities consider in the decision-making as to whether to grant or deny market authorization (market approval) for a drug. SAE reporting occurs as a result of study patients (subjects) who experience serious adverse events during the conducting of clinical trials. (Non-serious adverse events are also captured separately.) SAE information, which may also include relevant information from the patient's medical background, are reviewed and assessed for causality by the study investigator. This information is forwarded to a sponsoring entity (typically a pharmaceutical company) that is responsible for the reporting of this information, as appropriate, to drug regulatory authorities.

Spontaneous reporting

Spontaneous reports are termed spontaneous as they take place during the clinician's normal diagnostic appraisal of a patient, when the clinician is drawing the conclusion that the drug may be implicated in the causality of the event. Spontaneous reporting system relies on vigilant physicians and other healthcare professionals who not only generate a suspicion of an ADR, but also report it. It is an important source of regulatory actions such as taking a drug off the market or a label change due to safety problems. Spontaneous reporting is the core data-generating system of international pharmacovigilance, relying on healthcare professionals (and in some countries consumers) to identify and report any adverse events to their national pharmacovigilance center, health authority (such as EMA or FDA), or to the drug manufacturer itself. Spontaneous reports are, by definition, submitted voluntarily although under certain circumstances these reports may be encouraged, or "stimulated", by media reports or articles published in medical or scientific publications, or by product lawsuits. In many parts of the world adverse event reports are submitted electronically using a defined message standard.

One of the major weaknesses of spontaneous reporting is that of under-reporting, where, unlike in clinical trials, less than 100% of those adverse events occurring are reported. Further complicating the assessment of adverse events, AE reporting behavior varies greatly between countries and in relation to the seriousness of the events, but in general probably less than 10% (some studies suggest less than 5%) of all adverse events that occur are actually reported. The rule-of-thumb is that on a scale of 0 to 10, with 0 being least likely to be reported and 10 being the most likely to be reported, an uncomplicated non-serious event such as a mild headache will be closer to a "0" on this scale, whereas a life-threatening or fatal event will be closer to a "10" in terms of its likelihood of being reported. In view of this, medical personnel may not always see AE reporting as a priority, especially if the symptoms are not serious. And even if the symptoms are serious, the symptoms may not be recognized as a possible side effect of a particular drug or combination thereof. In addition, medical personnel may not feel compelled to report events that are viewed as expected. This is why reports from patients themselves are of high value. The confirmation of these events by a healthcare professional is typically considered to increase the value of these reports. Hence it is important not only for the patient to report the AE to his health care provider (who may neglect to report the AE), but also report the AE to both the biopharmaceutical company and the FDA, EMA, ... This is especially important when one has obtained one's pharmaceutical from a compounding pharmacy. 

As such, spontaneous reports are a crucial element in the worldwide enterprise of pharmacovigilance and form the core of the World Health Organization Database, which includes around 4.6 million reports (January 2009), growing annually by about 250,000.

Aggregate reporting

Aggregate reporting, also known as periodic reporting, plays a key role in the safety assessment of drugs. Aggregate reporting involves the compilation of safety data for a drug over a prolonged period of time (months or years), as opposed to single-case reporting which, by definition, involves only individual AE reports. The advantage of aggregate reporting is that it provides a broader view of the safety profile of a drug. Worldwide, the most important aggregate report is the Periodic Safety Update Report (PSUR) and Development Safety Update Report (DSUR). This is a document that is submitted to drug regulatory agencies in Europe, the US and Japan (ICH countries), as well as other countries around the world. The PSUR was updated in 2012 and is now referred to in many countries as the Periodic Benefit Risk Evaluation report (PBRER). As the title suggests, the PBRER's focus is on the benefit-risk profile of the drug, which includes a review of relevant safety data compiled for a drug product since its development.

Other reporting methods

Some countries legally oblige spontaneous reporting by physicians. In most countries, manufacturers are required to submit, through its Qualified Person for Pharmacovigilance (QPPV), all of the reports they receive from healthcare providers to the national authority. Others have intensive, focused programmes concentrating on new drugs, or on controversial drugs, or on the prescribing habits of groups of doctors, or involving pharmacists in reporting. All of these generate potentially useful information. Such intensive schemes, however, tend to be the exception. A number of countries have reporting requirements or reporting systems specific to vaccine-related events.

Risk management

Risk management is the discipline within pharmacovigilance that is responsible for signal detection and the monitoring of the risk-benefit profile of drugs. Other key activities within the area of risk management are that of the compilation of risk management plans (RMPs) and aggregate reports such as the Periodic Safety Update Report (PSUR), Periodic Benefit-Risk Evaluation Report (PBRER), and the Development Safety Update Report (DSUR).

Causality assessment

One of the most important, and challenging, problems in pharmacovigilance is that of the determination of causality. Causality refers to the relationship of a given adverse event to a specific drug. Causality determination (or assessment) is often difficult because of the lack of clear-cut or reliable data. While one may assume that a positive temporal relationship might "prove" a positive causal relationship, this is not always the case. Indeed, a "bee sting" AE—where the AE can clearly be attributed to a specific cause—is by far the exception rather than the rule. This is due to the complexity of human physiology as well as that of disease and illnesses. By this reckoning, in order to determine causality between an adverse event and a drug, one must first exclude the possibility that there were other possible causes or contributing factors. If the patient is on a number of medications, it may be the combination of these drugs which causes the AE, and not any one individually. There have been a number of recent high-profile cases where the AE led to the death of an individual. The individual(s) were not overdosed with any one of the many medications they were taking, but the combination there appeared to cause the AE. Hence it is important to include in your/one's AE report, not only the drug being reported, but also all other drugs the patient was also taking. 

For instance, if a patient were to start Drug X and then three days later were to develop an AE, one might be tempted to attribute blame Drug X. However, before that can be done, the patient's medical history would need to be reviewed to look for possible risk factors for the AE. In other words, did the AE occur with the drug or because of the drug? This is because a patient on any drug may develop or be diagnosed with a condition that could not have possibly been caused by the drug. This is especially true for diseases, such as cancer, which develop over an extended period of time, being diagnosed in a patient who has been taken a drug for a relatively short period of time. On the other hand, certain adverse events, such as blood clots (thrombosis), can occur with certain drugs with only short-term exposure. Nevertheless, the determination of risk factors is an important step of confirming or ruling-out a causal relationship between an event and a drug. 

Often the only way to confirm the existence of a causal relationship of an event to a drug is to conduct an observational study where the incidence of the event in a patient population taking the drug is compared to a control group. This may be necessary to determine if the background incidence of an event is less than that found in a group taking a drug. If the incidence of an event is statistically significantly higher in the "active" group versus the placebo group (or other control group), it is possible that a causal relationship may exist to a drug, unless other confounding factors may exist.

Signal detection

Signal detection (SD) involves a range of techniques (CIOMS VIII). The WHO defines a safety signal as: "Reported information on a possible causal relationship between an adverse event and a drug, the relationship being unknown or incompletely documented previously". Usually more than a single report is required to generate a signal, depending upon the event and quality of the information available. 

Data mining pharmacovigilance databases is one approach that has become increasingly popular with the availability of extensive data sources and inexpensive computing resources. The data sources (databases) may be owned by a pharmaceutical company, a drug regulatory authority, or a large healthcare provider. Individual Case Safety Reports (ICSRs) in these databases are retrieved and converted into structured format, and statistical methods (usually a mathematical algorithm) are applied to calculate statistical measures of association. If the statistical measure crosses an arbitrarily set threshold, a signal is declared for a given drug associated with a given adverse event. All signals deemed worthy of investigation, require further analysis using all available data in an attempt to confirm or refute the signal. If the analysis is inconclusive, additional data may be needed such as a post-marketing observational trial. 

SD is an essential part of drug use and safety surveillance. Ideally, the goal of SD is to identify ADRs that were previously considered unexpected and to be able to provide guidance in the product's labeling as to how to minimize the risk of using the drug in a given patient population.

Risk management plans

A risk management plan (RMP) is a documented plan that describes the risks (adverse drug reactions and potential adverse reactions) associated with the use of a drug and how they are being handled (warning on drug label or on packet inserts of possible side effects which if observed should cause the patient to inform/see his physician and/or pharmacist and/or the manufacturer of the drug and/or the FDA, EMA)). The overall goal of an RMP is to assure a positive risk-benefit profile once the drug is (has been) marketed. The document is required to be submitted, in a specified format, with all new market authorization requests within the European Union (EU). Although not necessarily required, RMPs may also be submitted in countries outside the EU. The risks described in an RMP fall into one of three categories: identified risks, potential risks, and unknown risks. Also described within an RMP are the measures that the Market Authorization Holder, usually a pharmaceutical company, will undertake to minimize the risks associated with the use of the drug. These measures are usually focused on the product's labeling and healthcare professionals. Indeed, the risks that are documented in a pre-authorization RMP will inevitably become part of the product's post-marketing labeling. Since a drug, once authorized, may be used in ways not originally studied in clinical trials, this potential "off-label use", and its associated risks, is also described within the RMP. RMPs can be very lengthy documents, running in some cases hundreds of pages and, in rare instances, up to a thousand pages long. 

In the US, under certain circumstances, the FDA may require a company to submit a document called a Risk Evaluation and Mitigation Strategies (REMS) for a drug that has a specific risk that FDA believes requires mitigation. While not as comprehensive as an RMP, a REMS can require a sponsor to perform certain activities or to follow a protocol, referred to as Elements to Assure Safe Use (ETASU), to assure that a positive risk-benefit profile for the drug is maintained for the circumstances under which the product is marketed.

Risk/benefit profile of drugs

Pharmaceutical companies are required by law in most countries to perform clinical trials, testing new drugs on people before they are made generally available. This occurs after a drug has been pre-screened for toxicity, sometimes using animals for testing. The manufacturers or their agents usually select a representative sample of patients for whom the drug is designed – at most a few thousand – along with a comparable control group. The control group may receive a placebo and/or another drug, often a so-called "gold standard" that is "best" drug marketed for the disease. 

The purpose of clinical trials is to determine:
  • If a drug works and how well it works
  • If it has any harmful effects, and
  • If it does more good than harm, and how much more? If it has a potential for harm, how probable and how serious is the harm?
Clinical trials do, in general, tell a good deal about how well a drug works. They provide information that should be reliable for larger populations with the same characteristics as the trial group – age, gender, state of health, ethnic origin, and so on though target clinical populations are typically very different from trial populations with respect to such characteristics
.
The variables in a clinical trial are specified and controlled, but a clinical trial can never tell you the whole story of the effects of a drug in all situations. In fact, nothing could tell you the whole story, but a clinical trial must tell you enough; "enough" being determined by legislation and by contemporary judgements about the acceptable balance of benefit and harm. Ultimately, when a drug is marketed it may be used in patient populations that were not studied during clinical trials (children, the elderly, pregnant women, patients with co-morbidities not found in the clinical trial population, etc.) and a different set of warnings, precautions or contraindications (where the drug should not be used at all) for the product's labeling may be necessary in order to maintain a positive risk/benefit profile in all known populations using the drug.

Pharmacogenetics and pharmacogenomics

Although often used interchangeably, there are subtle differences between the two disciplines. Pharmacogenetics is generally regarded as the study or clinical testing of genetic variation that gives rise to differing responses to drugs, including adverse drug reactions. It is hoped that pharmacogenetics will eventually provide information as to which genetic profiles in patients will place those patients at greatest risk, or provide the greatest benefit, for using a particular drug or drugs. Pharmacogenomics, on the other hand, is the broader application of genomic technologies to new drug discovery and further characterization of older drugs.

International collaboration

The following organizations play a key collaborative role in the global oversight of pharmacovigilance.

The World Health Organization (WHO)

The principle of international collaboration in the field of pharmacovigilance is the basis for the WHO Programme for International Drug Monitoring, through which over 150 member nations have systems in place that encourage healthcare personnel to record and report adverse effects of drugs in their patients. These reports are assessed locally and may lead to action within the country. Since 1978, the programme has been managed by the Uppsala Monitoring Centre to which member countries send their reports to be processed, evaluated and entered into an international database called VigiBase. Membership in the WHO Programme enables a country to know if similar reports are being made elsewhere. When there are several reports of adverse reactions to a particular drug, this process may lead to the detection of a signal, and an alert about a possible hazard communicated to members countries after detailed evaluation and expert review.

The International Council for Harmonisation (ICH)

The ICH is a global organization with members from the European Union, the United States and Japan; its goal is to recommend global standards for drug companies and drug regulatory authorities around the world, with the ICH Steering Committee (SC) overseeing harmonization activities. Established in 1990, each of its six co-sponsors—the EU, the European Federation of Pharmaceutical Industries and Associations (EFPIA), Japan's Ministry of Health, Labor and Welfare (MHLW), the Japanese Pharmaceutical Manufacturers Association (JPMA), the U.S. Food and Drug Administration (FDA), and the Pharmaceutical Research and Manufacturers of America (PhRMA)—have two seats on the SC. Other parties have a significant interest in ICH and have been invited to nominate Observers to the SC; three current observers are the WHO, Health Canada, and the European Free Trade Association (EFTA), with the International Federation of Pharmaceutical Manufacturers Association (IFPMA) participating as a non-voting member of the SC.

The Council for International Organizations of Medical Science (CIOMS)

The CIOMS, a part of the WHO, is a globally oriented think tank that provides guidance on drug safety related topics through its Working Groups. The CIOMS prepares reports that are used as a reference for developing future drug regulatory policy and procedures, and over the years, many of CIOMS' proposed policies have been adopted. Examples of topics these reports have covered include: Current Challenges in Pharmacovigilance: Pragmatic Approaches (CIOMS V); Management of Safety Information from Clinical Trials (CIOMS VI); the Development Safety Update Report (DSUR): Harmonizing the Format and Content for Periodic Safety Reporting During Clinical Trials (CIOMS VII); and Practical Aspects of Signal Detection in Pharmacovigilance: Report of CIOMS Working Group (CIOMS VIII).

The International Society of Pharmacovigilance (ISoP)

The ISoP is an international non-profit scientific organization, which aims to foster pharmacovigilance both scientifically and educationally, and enhance all aspects of the safe and proper use of medicines, in all countries. It was established in 1992 as the European Society of Pharmacovigilance.

Society of Pharmacovigilance, India, also established in 1992, is partner member of ISoP. Local societies include the Boston Society of Pharmacovigilance Physicians.

Regulatory authorities

Drug regulatory authorities play a key role in national or regional oversight of pharmacovigilance. Some of the agencies involved are listed below (in order of 2011 spending on pharmaceuticals, from the IMS Institute for Healthcare Informatics).

United States

In the U.S., with about a third of all global 2011 pharmaceutical expenditures, the drug industry is regulated by the FDA, the largest national drug regulatory authority in the world. FDA authority is exercised through enforcement of regulations derived from legislation, as published in the U.S. Code of Federal Regulations (CFR); the principal drug safety regulations are found in 21 CFR Part 312 (IND regulations) and 21 CFR Part 314 (NDA regulations). While those regulatory efforts address pre-marketing concerns, pharmaceutical manufacturers and academic/non-profit organizations such as RADAR and Public Citizen do play a role in pharmacovigilance in the US. The post-legislative rule-making process of the U.S. federal government provides for significant input from both the legislative and executive branches, which also play specific, distinct roles in determining FDA policy.

Emerging economies (including Latin America)

The "pharmerging", or emerging pharmaceutical market economies, which include Brazil, India, Russia, Argentina, Egypt, Indonesia, Mexico, Pakistan, Poland, Romania, South Africa, Thailand, Turkey, Ukraine and Vietnam, accrued one fifth of global 2011 pharmaceutical expenditures; in future, aggregated data for this set will include China as well.

China's economy is anticipated to pass Japan to become second in the ranking of individual countries' in pharmaceutical purchases by 2015, and so its PV regulation will become increasing important; China's regulation of PV is through its National Center for Adverse Drug Reaction (ADR) Monitoring, under China's Ministry of Health.

As JE Sackman notes, as of April 2013 "there is no Latin American equivalent of the European Medicines Agency—no common body with the power to facilitate greater consistency across countries". For simplicity, and per sources, 17 smaller economies are discussed alongside the 4 pharmemerging large economies of Argentina, Brazil, Mexico and Venzuala—Bolivia, Chile, Colombia, Costa Rica, Cuba, Dominican Republic, Ecuador, El Salvador, Guatemala, Haiti, Honduras, Nicaragua, Panama, Paraguay, Peru, Suriname, and Uruguay. As of June 2012, 16 of this total of 21 countries have systems for immediate reporting and 9 have systems for periodic reporting of adverse events for on-market agents, while 10 and 8, respectively, have systems for immediate and periodic reporting of adverse events during clinical trials; most of these have PV requirements that rank as "high or medium...in line with international standards" (ibid.). The WHO's Pan American Network for Drug Regulatory Harmonization seeks to assist Latin American countries in develop harmonized PV regulations.

Some further PV regulatory examples from the pharmerging nations are as follows. In India, the PV regulatory authority is the Indian Pharmacopoeia Commission, with a National Coordination Centre under the Pharmacovigilance Program of India, in the Ministry of Health and Family Welfare. Scientists working on pharmacovigilance share their experiences, findings, innovative ideas and researches during the annual meeting of Society of Pharmacovigilance, India. In Egypt, PV is regulated by the Egyptian Pharmacovigilance Center of the Egyptian Ministry of Health.

European Union

The EU5 (France, Germany, Italy, Spain, United Kingdom) accrued ~17% of global 2011 pharmaceutical expenditures. PV efforts in the EU are coordinated by the European Medicines Agency (EMA) and are conducted by the national competent authorities (NCAs). The main responsibility of the EMA is to maintain and develop the pharmacovigilance database consisting of all suspected serious adverse reactions to medicines observed in the European Community; the data processing network and management system is called EudraVigilance and contains separate but similar databases of human and veterinary reactions. The EMA requires the individual marketing authorization holders to submit all received adverse reactions in electronic form, except in exceptional circumstances; the reporting obligations of the various stakeholders are defined by EEC legislation, namely Regulation (EC) No 726/2004, and for human medicines, European Union Directive 2001/83/EC as amended and Directive 2001/20/EC. In 2002, Heads of Medicines Agencies agreed on a mandate for an ad hoc Working Group on establishing a European risk management strategy; the Working Group considered the conduct of a high level survey of EU pharmacovigilance resources to promote the utilization of expertise and encourage collaborative working.

In conjunction with this oversight, individual countries maintain their distinct regulatory agencies with PV responsibility. For instance, in Spain, PV is regulated by the Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), a legal entity that retains the right to suspend or withdraw the authorization of pharmaceuticals already on-market if the evidence shows that safety (or quality or efficacy) of an agent are unsatisfactory.

Rest of Europe, including non-EU

The remaining EU and non-EU countries outside the EU5 accrued ~7% of global 2011 pharmaceutical expenditures. Regulation of those outside the EU being managed by specific governmental agencies. For instance, in Switzerland, PV "inspections" for clinical trials of medicinal products are conducted by the Swiss Agency for Therapeutic Products.

Japan

In Japan, with ~12% of all global 2011 pharmaceutical expenditures, PV matters are regulated by the Pharmaceuticals and Medical Devices Agency (PMDA) and the Ministry of Health, Labour, and Welfare MHLW.

Canada

In Canada, with ~2% of all global 2006 and 2011 pharmaceutical expenditures,PV is regulated by the Marketed Health Products Directorate of the Health Products and Food Branch(MHPD). Canada was second, following the United States, in holding the highest total prescription drug expenditures per capita in 2011 at around 750 US dollars per person. Canada also pays such a large amount for pharmaceuticals that it was second, next to Switzerland, for the amount of money spent for a certain amount of prescription drugs (around 130 US dollars). It was also accessed that Canada was one of the top countries that increased its yearly average per capita growth on pharmaceutical expenditures the most from 2000-2010 with 4 percent a year (with taking inflation into account)  The MHPD mainly collects adverse drug reaction reports through a network of reporting centers to analyze and issue possible warnings to the public, and currently utilizes newsletters, advisories, adverse reaction centers, as well as electronic mailing lists. However, it does not currently maintain a database or list of drugs removed from Canada as a result of safety concerns.

In August 2017, there was a government controversy in which a bill, known as “Vanessa’s Law”, to protect patients from potentially dangerous prescription drugs was not being fully realized by hospitals; Health Canada only required hospitals to report “unexpected” negative reactions to prescription drugs, rather than any and all adverse reactions, with the justification of managing “administrative overload”.

South Korea

The Republic of Korea, with ~1% of all global 2011 pharmaceutical expenditures, PV matters are regulated in South Korea by the Ministry Of Food And Drug Safety (MFDS)

Africa

Kenya

In Kenya, PV is regulated by the Pharmacy and Poisons Board.The Pharmacy and Poisons Board provides a Pharmacovigilance Electronic Reporting System which allows for the online reporting of suspected adverse drug reactions as well as suspected poor quality of medicinal products. The Pharmacovigilance activities in Kenya are supported by the School of Pharmacy, University of Nairobi through its Master of Pharmacy in Pharmacoepidemiology & Pharmacovigilance program offered by the Department of Pharmacology and Pharmacognosy.

In Uganda, PV is regulated by the National Drug Authority.

Rest of world (ROW)

ROW accrued ~7% of global 2011 pharmaceutical expenditures. Some examples of PV regulatory agencies in ROW are as follows. In Iraq, PV is regulated by the Iraqi Pharmacovigilance Center of the Iraqi Ministry of Health.

Pharmacoenvironmentology; (Ecopharmacovigilance [EPV])

Despite attention from the FDA and regulatory agencies of the European Union, procedures for monitoring drug concentrations and adverse effects in the environment are lacking. Pharmaceuticals, their metabolites, and related substances may enter the environment after patient excretion, after direct release to waste streams during manufacturing or administration, or via terrestrial deposits (e.g., from waste sludges or leachates). A concept combining pharmacovigilance and environmental pharmacology, intended to focus attention on this area, was introduced first as pharmacoenvironmentology in 2006 by Syed Ziaur Rahman and later as ecopharmacology with further concurrent and later terms for the same concept (ecopharmacovigilance [EPV], environmental pharmacology, ecopharmacostewardship).

The first of these routes to the environment, elimination through living organisms subsequent to pharmacotherapy, is suggested as the principal source of environmental contamination (apart from cases where norms for treatment of manufacturing and other wastes are violated), and EPV is intended to deal specifically with this impact of pharmacological agents on the environment.

Activities of EPV have been suggested to include:
  • Increasing, generally, the availability of environmental data on medicinal products;
  • Tracking emerging data on environmental exposure, effects and risks after product launch;
  • Using Environmental Risk Management Plans (ERMPs) to manage risk throughout a drug's life cycle;
  • Following risk identification, promoting further research and environmental monitoring, and
  • In general, promoting a global perspective on EPV issues.

Related to medical devices

A medical device is an instrument, apparatus, implant, in vitro reagent, or similar or related article that is used to diagnose, prevent, or treat disease or other conditions, and does not achieve its purposes through chemical action within or on the body (which would make it a drug). Whereas medicinal products (also called pharmaceuticals) achieve their principal action by pharmacological, metabolic or immunological means, medical devices act by physical, mechanical, or thermal means. Medical devices vary greatly in complexity and application. Examples range from simple devices such as tongue depressors, medical thermometers, and disposable gloves to advanced devices such as medical robots, cardiac pacemakers, and neuroprosthetics. This modern concept of monitoring and safety of medical devices which is known materiovigilance was quite documented in Unani System of medicine.

Given the inherent difference between medicinal products and medical products, the vigilance of medical devices is also different from that of medicinal products. To reflect this difference, a classification system has been adopted in some countries to stratify the risk of failure with the different classes of devices. The classes of devices typically run on a 1-3 or 1-4 scale, with Class 1 being the least likely to cause significant harm with device failure versus Classes 3 or 4 being the most likely to cause significant harm with device failure. An example of a device in the "low risk" category would be contact lenses. An example of a device in the "high risk" category would be cardiac pacemakers.

Medical device reporting (MDR), which is the reporting of adverse events with medical devices, is similar to that with medicinal products, although there are differences. For instance, in the US user-facilities such as hospitals and nursing homes are legally required to report suspected medical device-related deaths to both FDA and the manufacturer, if known, and serious injuries to the manufacturer or to FDA, if the manufacturer is unknown. This is in contrast to the voluntary reporting of AEs with medicinal products.

For herbal medicines

The safety of herbal medicines has become a major concern to both national health authorities and the general public. The use of herbs as traditional medicines continues to expand rapidly across the world; many people now take herbal medicines or herbal products for their health care in different national health-care settings. However, mass media reports of adverse events with herbal medicines can be incomplete and therefore misleading. Moreover, it can be difficult to identify the causes of herbal medicine-associated adverse events since the amount of data on each event is generally less than for pharmaceuticals formally regulated as drugs (since the requirements for adverse event reporting are either non-existent or are less stringent for herbal supplements and medications).

Education reform

From Wikipedia, the free encyclopedia https://en.wikipedia.org/wiki/Education_reform ...