WhatsApp Image 2024-04-15 at 17.49.58

Innovations in Nephrology Care: Exploring the Latest Treatment Options

Introduction

Nephrology emerged as the leading internal medicine subspecialty post-WWI. Kidneys are vital for bodily function, filter waste, regulate fluids and minerals, control blood pressure, and produce urine and erythropoietin.

Moreover, individuals with kidney disease experience impairment in kidney function, often stemming from conditions such as hypertension and diabetes. 

The National Kidney Foundation reports that kidney disease affects roughly 37 million adults, while an additional 80 million are at risk. Additionally, racial minorities have a higher incidence of kidney disease, with African Americans being approximately four times as susceptible.

In this article, let’s learn more about the kidney, kidney diseases, and worldwide research. 

What is Nephrology

Nephrology is a vital medical branch specializing in the comprehensive study, diagnosis, and treatment of kidney-related diseases. This involves a multifaceted approach, employing clinical, laboratory, imaging, and histopathologic techniques to assess kidney function and structure.

On the other hand, Nephrologists are dedicated to preserving kidney health through tailored interventions, including dietary adjustments, medication, and kidney replacement therapy. They adeptly manage various complications such as hypertension, fluid retention, and electrolyte imbalances, ensuring holistic care for their patients. 

Moreover, Nephrologists play a crucial role in addressing chronic conditions like diabetes and hypertension, which significantly impact kidney function, alongside managing acute renal failure cases. Collaborating seamlessly with transplant teams, they extend their expertise to oversee the care of kidney transplant recipients, ultimately striving to enhance patient quality of life and prevent complications.

Areas of Focus in Nephrology

Nephrologists may focus on diagnosing and treating various kidney disorders, catering to specific patient groups, or conducting specialised procedures. Specialised areas within nephrology encompass:

  1. Critical care nephrology
  2. Diabetic kidney disease management
  3. Dialysis oversight
  4. Geriatric nephrology (for age 65+)
  5. Interventional nephrology (including dialysis access and arteriovenous fistula surgery)
  6. Renal oncology (kidney cancer)
  7. Kidney stones treatment
  8. Kidney transplant care
  9. Paediatric nephrology (infants to adolescents)

Latest Treatment Options in Nephrology Care

Research and innovative developments shape treatment paradigms for kidney-related conditions in nephrology care. Some of these innovations are:

Kidney Fibrosis Treatment:

Researchers found increased histone lysine crotonylation (Kcr) in fibrotic kidneys, driven by the ACSS2 enzyme. Histone lysine crotonylation (Kcr) is a new acylation modification discovered in 2011 having important biological significance for gene expression, cell development, and disease treatment. 

TGF-β for Improved CKD Treatment:

In Chronic Kidney Disease (CKD), TGF-β, a transforming growth factor affects kidney cell mitochondria, worsening the disease. However, in diseased conditions, TGF-β loses its anti-proliferative response and becomes an oncogenic factor. 

Moreover, recent research shows blocking TGF-β in mice’s proximal tubules increases mitochondrial damage and inflammation. Similar issues were found in CKD patients’ kidney samples. Hence, this new insight may lead to new CKD treatment approaches targeting TGF-β pathways.

Denosumab in Osteoporosis Patients with Kidney Disease

In a recent innovation, Denosumab, commonly used for osteoporosis in advanced kidney disease patients, raises concerns about severe hypocalcemia. A study of 2804 older females on dialysis reveals a higher risk compared to oral bisphosphonates. Prolia now carries a boxed warning, emphasising intensified monitoring during treatment.

Genetic Solution to Mitigate CKD

New research reveals that certain APOL1 gene variations increase chronic kidney disease (CKD) risk in people of West African descent. However, another mutation, p.N264k, counters this risk. In vitro studies show that p.N264k reduces the harmful effects of high-risk APOL1 variations. This suggests potential drug targets for CKD prevention.

Enhanced Advance Care Planning for Dialysis Patients

A study in 42 dialysis clinics with 430 patients and their decision-makers showed improved patient-surrogate communication through 45-60 minute discussions led by clinic healthcare workers. This approach reduces end-of-life decisional conflicts and increases adherence to care goals among dialysis patients, enhancing their overall care experience.

Medicinal Options in Nephrology Care

Medicinal Options in Nephrology Care delves into the diverse pharmacological interventions available for managing kidney-related conditions.

Renaglob

Renagold Tablet is frequently prescribed as a nutritional supplement for individuals suffering from chronic kidney failure and uremia. Its primary function is to inhibit the elevation of urea levels in the bloodstream from consuming non-essential amino acids among kidney failure patients.

Uriglob/Uriglob D

Uriglob Tablet effectively relaxes muscles in the bladder and prostate to alleviate symptoms associated with an enlarged prostate. This relaxation enables easier urination, providing rapid relief from urinary difficulties.

Trientine HCL Capsules

Trientine Hydrochloride is prescribed for Wilson’s disease, functioning as a copper-chelating agent. Its mechanism involves binding surplus copper in the body’s tissues and facilitating its elimination through the kidneys in the urine.

Selaglob

Trientine Hydrochloride reduces high blood phosphorus levels in dialysis patients. Selaglob Tablets stop phosphate absorption in the intestine, reducing blood phosphate levels.

Kalara

Calcium Polystyrene Sulfonate reduces high blood potassium levels, particularly in kidney conditions such as anuria, severe oliguria, and chronic kidney disease. It’s also utilised to lower potassium levels in patients undergoing regular dialysis.

Febuglob

Febuglob Tablet treats gout by lowering uric acid levels. It’s for patients unresponsive to allopurinol. Also used for hyperuricemia in adults with hematologic malignancies at medium to high TLS risk during chemotherapy.

Deferglob

Deferglob Tablet is prescribed for managing chronic iron overload resulting from recurrent blood transfusions. Its function involves eliminating surplus iron from the body and lowering the likelihood of organ damage induced by iron accumulation.

Conclusion

Innovations in nephrology care are revolutionising treatment approaches for kidney-related conditions, addressing diverse challenges with promising solutions.

The landscape of nephrology is evolving, from groundbreaking genetic insights offering potential preventive strategies for chronic kidney disease to enhanced communication practices improving end-of-life care for dialysis patients. Furthermore, advancements in medicinal options provide tailored interventions, such as Renagold for nutritional support and Trientine HCL for Wilson’s disease.

These developments underscore nephrology’s commitment to optimising patient outcomes and enhancing the quality of life, ushering in a new era of personalised and effective kidney care.

 Globela Pharma offers high quality and affordable nephrology solutions across 50+ countries. 

blog image 2 (1)

Collaboration-A need of hour to Speed up Access to New Vaccines

Introduction:

The fatal wave of COVID-19 made everyone aware of the importance of vaccines to prevent and control tremendously dangerous and infectious diseases. The whole world during the COVID-19 pandemic needed an efficient vaccine to kill the virus and save them from a lethal contagious disease. Scientists and researchers globally were focused on creating an efficient and reliable vaccine to protect the world from such a dangerous virus. 

During the pandemic crisis, this situation not only demonstrated the importance of vaccines and medicines in our lives but also proved how important it is to have a well-organized and synchronised process for vaccine manufacturing.  To combat all the challenges that the world faced during the time of pandemic a new tool of “regulatory collaboration” came to light. 

In this article, we will learn the importance of global regulatory collaboration and how this global regulatory collaboration helps in achieving a single goal i.e., speed up the access to vaccines.

What does Global Regulatory Collaboration mean?

A global regulatory collaboration simply means the collaboration or working together by regulatory bodies from various countries. It works in different ways to discuss, develop, manage and achieve the same goal. 

The Global Regulatory Collaboration came out as an emerging tool to have more access to vaccines and medications. Also, it helps to manage its proper manufacturing as well as distribution across the globe. 

What is the importance of Global Regulatory Collaboration?

The traditional regulatory process for the validation and approval of vaccines involves a series of rigorous trials and preclinical examinations along with an assessment of manufacturing processes. 

Global regulatory collaboration also plays a vital role in ensuring the rapid development, approval and distribution of vaccines worldwide. 

The era of COVID-19 demanded the healthcare system’s urgency of efficient and useful vaccines along with acceleratory regulatory pathways without compromising on scientific rigour. 

Thus, the Global Regulatory collaboration helps in acknowledging the need for expeditious decision-making along with ensuring excellent standards and quality in vaccine development and efficient distribution. 

How does the Global Regulatory Collaboration aim to speed access to new vaccines?

There are various benefits of Global Regulatory Collaboration as it aims to speed the access to new vaccines.

Global Regulatory collaborations work while promoting the sharing of information, harmonizing standards and promoting mutual recognition of regulatory decisions it enables the availability of access to safe and efficient vaccines. 

So let’s understand it’s working as follows:

Enhancing Information Sharing and Cooperation:

A very crucial aspect of global regulatory collaboration is the exchange of information and cooperation among regulatory authorities. Collaboration enables regulatory agencies to share their data from every event. 

It starts from clinical trials to post-marketing surveillance along with no confidentiality, but total transparency. By enabling access to the expertise and resources of various collaborated regulatory bodies the companies can minimize the approval timelines and other efforts too. 

It also can help companies to facilitate the global availability of safe and quality vaccines throughout the globe. 

Harmonization of Standards:

Various countries have various wide range of rules. Yes, different countries have different and unique regulatory requirements and approval processes which eventually make the consumers suffer for their needs.

Additionally, these processes are time-consuming and require various recognitions. Thus, it takes very long for these vaccines to reach across borders. 

In this, Global regulatory collaboration attempts to harmonize standards and requirements among different agencies to hasten the approval process. 

By ensuring proper synchronisation and utilising recognition companies can prevent the duplication of efforts. Also, it aids the regulatory bodies to speed up access to vaccines without compromising their safety and efficacy.

Mutual Recognition of Regulatory Decisions:

Mutual recognition agreements (MRAs) are the key tools that play a major role in global regulatory collaboration. It allows regulatory bodies to accept the decision and verdict established by another authentic regulatory body. 

This process not only saves time by avoiding fake evaluations but also helps in enhancing trust and confidence in the safety and functioning of approved vaccines.

MRAs’ functioning is so effective and incredible that they make countries rely on the expertise of authentic regulatory agencies and also enable quick authorisation of vaccines within their jurisdictions.

International Consortia and Collaborative Platforms:

International consortia and collaborative platforms have established themselves as the leaders of global regulatory collaboration. Huge and reputed agencies like the World Health Organization (WHO), The International Coalition of Medicines Regulatory Authorities, and the Coalition for Epidemic Preparedness Innovations have also played a major part in cherishing cooperation among regulatory bodies. 

These platforms focus on sharing scientific information, harmonization standards and other relevant information required to help maintain a coordinated global development and distribution of vaccines throughout the globe. The international consortia and collaborative platforms also help by optimising resources and coordinating efforts to make vaccines. 

Impact on Access and Equity:

Global regulatory collaboration has profound implications for access and equity in vaccine distribution. Accelerating the regulatory process ensures the access of vaccines to developing countries as well, as that to the advanced ones. 

For instance, WHO’s very well-known COVAX and other Emergency Use Listing (EUL) had an equal distribution of the vaccines by regulatory collaborating bodies. They also encouraged the sharing of the doses with other countries in need. 

By developing and distributing vaccines across the world the regulatory collaboration not only bridged the gap in accessing vaccines to individuals but also saved thousands of lives reducing health disparities.

Conclusion:

In a nutshell, global regulatory collaborations have proved themselves to be a pillar in saving lives by speeding up access to vaccines during times of crisis. The COVID-19 pandemic has reinforced the need for a synchronised and coordinated regulatory body to ensure the timely availability of safe and effective vaccines. 

As the world continues to battle with an ongoing pandemic and prepares itself for future global health crises, it is now a mandatory part to promote and accept global regulatory collaboration as a smart strategy to accelerate access to life-saving vaccines.

This same goes for the medicines as well. Collaboration is also a need in the manufacturing of medicine as well. In this, Globela Pharma with collaboration services provides rigid cooperation while maintaining the transparency in manufacturing of medicines.

Accreditations

Achieving Trust and Credibility: The Role of Approvals and Accreditations in the Pharma Industry

Introduction 

The Pharma industry is a highly regulated sector responsible for developing and producing drugs that are safe, effective, and of high quality. Although, achieving trust and credibility within this industry is crucial for companies to succeed, and one of the key ways to do this is through obtaining approvals and accreditations. 

Here ahead in this article, we will let you know how these approvals and accreditations are necessary for any pharmaceutical company.

Approvals in Pharmaceutical Industry

Approvals refer to the process by which regulatory agencies, such as the US Food and Drug Administration (FDA), evaluate new drugs and medical devices to determine whether they are safe and effective for their intended use. The approval process involves extensive clinical trials and rigorous testing to ensure that the product meets the highest standard of safety and efficacy. After approval, it is time to launch a medicine on the market and sell it to patients and healthcare providers. 

Moreover, Approvals provide a competitive advantage they give a strong reputation for the quality and safety of the products. Approvals also provide access to effective and affordable healthcare by manufacturing generic drugs. Because of the approval, Globela Pharma reaches over 30 countries, including developing nations like India.

Accreditations in Pharmaceutical Industry

Accreditations are certifications that demonstrate compliance with specific standards and guidelines. These standards relate to quality management, environmental sustainability, or other aspects of the company’s operations. Furthermore, Accreditation bodies, such as the International Organization for Standardization (ISO), evaluate companies against these standards and issue certifications to those that meet the criteria.

Additionally, Accreditations provide a framework for benchmarking performance against industry standards and best practices. It helps companies to identify improvements and stays ahead of the competition.Globela Pharma has fulfilled all the majorly accreditations. It is an established ISO, FDCA – India and WHO cGMP accredited company with multiple therapeutic areas. 

Roles of approvals and accreditations in achieving trust and credibility in the Pharma industry

By obtaining approvals and accreditations, pharmaceutical companies can demonstrate their commitment to producing safe and effective drugs that meet industry standards. All this helps to build trust and credibility with patients, healthcare providers, and regulatory agencies to establish credibility.

1. Ensuring Safety and Efficacy

The primary goal of approvals is to ensure that drugs are safe and effective for their intended use. It includes rigorous and repeated testing and assessments to identify potential risks or side effects.

Testings that help to check the efficiency and safety of drugs are:

2. Preclinical testing: In this testing phase, the drug is tested in laboratory settings and on animal models to evaluate its safety and efficacy. This phase includes pharmacological and toxicological tests.

3. Clinical trials: In this phase, the drug is tested on human volunteers in a controlled and monitored environment. Clinical trials are conducted in different phases to evaluate the drug’s safety, efficacy, and side effects. In this process, Pharma companies can demonstrate their commitment to patient safety and earn the faith of healthcare providers and patients at their best which gives them approval and accreditation. 

4. Demonstrating Compliance with Regulations

The Pharma industry is highly regulated, with strict guidelines and requirements for everything from clinical trials to manufacturing processes. By obtaining approvals and accreditations, companies can demonstrate compliance with these regulations and show that they are committed to ethical and responsible business practices. It helps stakeholders to build trust and credibility.

Globela Pharma has implemented a quality management system compliant with ISO, an internationally recognized standard for quality management. This certification demonstrates the Pharmaceutical company’s dedication to producing high-quality products and following ethical and responsible business practices.

5. Improving Quality and Efficiency

Implementing quality management systems and continuous improvement processes helps identify areas of improvement to increase efficiency. Moreover, it can also deteriorate costs and increase productivity, leading to improved profitability and competitiveness in the market. With improved quality and efficiency, companies reduce the risk of product recalls and other quality-related issues. 

6. Enhancing Transparency and Accountability

The Pharma industry has faced criticism in the past for a lack of transparency and accountability. Approvals and accreditations demonstrate a commitment to transparency and susceptibility, which helps to build trust with collaborators. It can also help the company to improve its reputation. Such as Globela Pharma’s commitment to continuous improvement, transparency, and accountability are the reasons that make it an authentic and reliable firm. 

7. Building Reputation and Brand Value

Ultimately, Approvals and Accreditations help to build a company’s reputation and brand value. Companies known for producing safe, effective, and high-quality products are more likely to be authorized by patients and healthcare providers, leading to increased sales and market share. This can help to build the company’s brand value.

Additionally, this not only helps the company to build its name in the world of pharma companies. But, ensures its users a safe and reliable brand to trust for their health.

Conclusion 

Approvals and accreditations are necessary for a pharma company to ensure fair and satisfactory quality medicines and healthcare products. Also, it provides the sections of improvement that keep a company ahead of others. Globela Pharma is also one of the recognised companies with access to effective and affordable healthcare by manufacturing generic drugs with accreditations and approvals.

Overall, approvals and accreditations play a critical role in the success of a pharmaceutical company. They provide assurance, build trust, and can provide a competitive advantage in a highly regulated and competitive industry. 

R&D blog-min

Role of Research and Development in Modern Pharmaceutical Industry

Introduction

In a life cycle of a drug from its discovery till launch, a series of crucial steps are involved in order to comply with regulatory requirements as per respective local regulatory authority. These steps from discovering a new drug to its launch in the market contributes to research and development in the pharmaceutical industry. The process is time consuming and may take several years for completion.

Steps involved in research and development in the modern pharmaceutical industry are as follows, i) early drug discovery, ii) preclinical studies, iii)clinical development, iv) review and approval by applicable regulatory bodies, v) post marketing surveillance.

Identifying a potential target-

Early drug discovery involves target identification and validation, hit discovery, assay development and screening, high throughput screening, hit to lead and lead optimization. Target identification begins with identifying the function of potential therapeutic agents and its role in the disease. It can be approached by direct biochemical methods, genetic interactions or computational interface. However, a combined approach may be required to fully characterize on-target and off-target interactions in order to understand molecular action mechanisms. Main motive of hit discovery is to identify molecules with potential interactions with drug targets.

Assay development-

Different types of assays can be used for assay development and compound screening, ranging from biochemical to cell-based assays. The choice of the assay depends on the biology of the drug target protein, scale of the compound screen, the equipment infrastructure, etc. Factors required for assay development are; i) Pharmacological importance of the assay– ability to identify compounds with the desired mechanism of action, ii) Reproducibility– is readily reproducible across assay plates, screen days and the length of the drug discovery programme, iii) Quality– pharmacology of the standard compounds falls within predefined limits, iv) Effects of compounds in the assay– should not be sensitive to the concentrations of solvents used in the assay.

Screening methods-

High throughput screening, (HTS) involves screening of the entire compound library against the drug target. Knowledge-based screening is a method of selecting from the chemical library smaller subsets of molecules with potential activity at the target protein. Fragment screening is making very small molecular weight compound libraries which are screened at high concentrations. Physiological screening is a tissue-based approach with the response more in direction with the desired in vivo effect.

Lead optimization-

Drug-like molecules must go through different phases to identify the hit lead molecule and optimization with a potency of 100nM – 5mM at the drug target. The refinement process involves generating dose-response curves in primary assay for each hit. Followed by examining the surviving hits in a secondary assay. Generation of rudimentary structure-activity relationship, SAR data and identifying the essential elements in the structure linked with the activity. Lastly, in vitro assays providing significant data with regards to absorption, distribution, metabolism and excretion (ADME) properties as well as physicochemical and pharmacokinetic (PK) measurements. Overall, the aim is to achieve a lead compound optimized with desirable effects on the target that can provide therapeutic benefits within an acceptable safety window. Average time required for this step is 2-6 months.

A glance at preclinical trials-

Preclinical studies or non clinical studies, carries out testing on animals to accurately model the desired biological effect of a drug in order to predict treatment outcomes in patients determining its efficacy, and to identify all toxicities associated with the drug to predict adverse effects for safety assessment. There are two types of preclinical studies, i) in vitro, ii) in vivo, iii) ex vivo assay and iv) in silico. In compliance with good laboratory practices, GLP, in vitro studies are carried out outside of living organisms in a test tube, glass or petri dish. On the other hand, in vivo studies are those which involve living organisms, including animal studies and human clinical trials. Ex vivo assay refers to a medical procedure in which an organ, cell or tissue are taken from the living body for treatment testing such as skin biopsies or isolated samples from tumor biopsy. In silico studies refers to using computer simulations to predict the reaction of a compound with specific proteins or pathogens. 

Goal of preclinical studies involve determination of pharmacokinetics, proof of concept, formulation, optimization & bioavailability, establishing safe dose, therapeutic dose, lethal dose and maximum tolerated dose. The compound from drug discovery is modified through preclinical studies and becomes Investigational New Drug, IND. IND application is filed for review and approval as per guidelines and standards of local and national regulatory authority. On an average the time required for this phase is approximately ranging from 1-6 years.

A complete overview of Clinical trials-

Clinical development of drug discovery begins after approval of IND for further testing. Clinical trials are conducted for testing of the new drug classified into several phases.

Phase 0 and Phase I-  Phase 0 is known as human micro dosing studies, which involves 10-15 individuals who are administered with small amounts of sub therapeutic dose mainly to determine pharmacokinetics, oral bioavailability and half-life of the drug. Phase 0 trials are often skipped to direct Phase I trials unless some of the data is inconsistent from previously conducted preclinical studies. Phase I studies are conducted amongst healthy volunteers to test the safety, tolerability, pharmacokinetics & pharmacodynamics, side effects & adverse effects, optimum dose, half-life and formulation method for the drug. In circumstances when testing for diseases like cancer or HIV, the treatment for which is likely to make healthy individuals ill, clinical patients are selected as an exception. Phase I trials are not randomized and hence are vulnerable to selection bias. This phase involves 20-100 individuals. Phase I trials can be further divided into, i) Single ascending dose, Phase I (a) in which a small number of participants are entered sequentially at a particular dose while monitoring them for a period of time to confirm safety. If no adverse effects are noted, then dose is escalated for newer groups. It is continued until pre-calculated pharmacokinetic safety levels are achieved or intolerable side effects are noted, it is the point where drug reaches at maximum tolerated dose, MTD; ii) Multiple ascending dose, Phase I (b) in which group of participants receives multiple low doses of the drug, which is subsequently escalated for further group of participants up to a predetermined level. It helps in determining pharmacokinetics and pharmacodynamics of multiple doses of the drug along with its safety and tolerability.

Phase II- Phase II trials are performed on larger groups (50–300) and are designed to assess biological activity and effect of the drug. Trial design of Phase II trials are either as case series, which demonstrates safety and efficacy in a selected group of participants, or as randomized controlled trials ,RCT, where some participants receive the drug/device and others receive placebo/standard treatment. Phase II studies are divided into Phase II (a) and Phase II (b). Phase II (a) studies are pilot studies designed to demonstrate clinical efficacy or biological activity of the drug. Phase II (b) studies determine the optimal dose at which the drug shows biological activity with minimal side-effects. It is also known as maximum effective dose, MaxED.

Phase III- Phase III trials are conducted in a large patient population of 300-3000 individuals determining the efficacy of the new drug in comparison to existing standard treatment. They are time consuming and expensive with complicated trial designs such as Randomized controlled multicentre trials with single, double or triple blinded factors in order to avoid bias and clean results. Phase III (a) studies are trial designed and executed to obtain statistically significant data for new drug approval by regulatory authority. Phase III trials that continue while awaiting regulatory approval in order to provide life-saving drugs to patients until the drugs are available in the market are categorized as Phase III (b) studies. Label expansion studies by the sponsor also fall under this category.

Phase IV- If the new drug successfully passes through Phase I, II, and III, with desirable outcomes, the manufacturing, preclinical and clinical data is then submitted as a new drug application, NDA, for review and marketing approval by national applicable regulatory authority. Post approval the new drug is marketed and Phase IV trials begin, which is post marketing surveillance of the new drug and lasts for up to 5 years. The entire process from developing a drug from preclinical research till marketing can take approximately 12-18 years. A Phase IV trial is a drug monitoring trial to assure long-term safety and effectiveness of the drug, vaccine, device or diagnostic test. These trials involve the safety surveillance, i.e, pharmacovigilance and ongoing technical support of a drug after it receives regulatory approval to be sold. Phase IV studies may be required by regulatory authorities or may be undertaken by the sponsoring company for competitive reasons, such as finding a new market for the drug, or other reasons, for example, the drug may not have been tested for interactions with other drugs, or on certain population groups such as pregnant women, who are unlikely to subject themselves to trials. The safety surveillance is designed to detect any rare or long-term adverse effects over a much larger patient population and longer time period than was possible during the Phase I-III clinical trials. Harmful effects discovered by Phase IV trials may result in a drug being withdrawn from the market or restricted to certain uses; examples include cerivastatin (brand names Baycol and Lipobay), troglitazone (Rezulin) and rofecoxib (Vioxx).

Conclusion

Thus Research & Development is essential when it comes to the pharmaceutical industry, since R&D services not only generate income for the companies involved in the research but it often saves lives. Reliable Pharmaceutical R&D services allow for companies to have technical and manufacturing procedures, quality control measures and production scope aspects as per required standards.

Drug Discovery Blog-min

How we test safety and efficacy of new drugs.

Introduction

The concepts of efficacy and safety have been with mankind since ages. In native sense, an efficacious and safe medical intervention is one that works and causes no undue harm. A drug should be used only when it will benefit a patient. Benefit takes into account both the drug’s ability to produce the desired result (efficacy) and the likelihood of adverse effects (safety). For a major portion of the history of medicine, efficacy and safety were measured by that native standard, which till today lies at the heart of medical practice, but the meaning and measurement of those concepts have evolved with increased sophistication and advancement of scientific methods in medicine. This article introduces the concepts of efficacy, effectiveness and adverse effects. It also throws light on patient oriented and surrogate outcomes along with their comparison and correlation in safety assessment of new drugs. Lastly concluding with the importance of long term monitoring and assessment of benefit to risk ratio for new drugs.

How does efficacy & effectiveness differ from eachother?

Efficacy is the capacity to produce an effect (e.g., lower blood pressure, lower or control high blood sugar). Efficacy can be assessed accurately only in ideal conditions (i.e., when patients are selected by proper criteria and strictly adhere to the dosing schedule). Thus, efficacy is measured under expert supervision in a group of patients most likely to have a response to a drug, such as in a controlled clinical trial.

Effectiveness differs from efficacy because the former takes into account the overall performance of the drug to real world use. Often, a drug that is efficacious in clinical trials is not very effective in actual use. For example, a drug may have high efficacy in lowering blood pressure but may have low effectiveness because it causes undesirable adverse effects which makes it difficult for patients to adhere to it. Effectiveness also may be lower than efficacy if clinicians inadvertently prescribe the drug inappropriately (e.g., giving a fibrinolytic drug to a patient thought to have an ischemic stroke, but who had an unrecognized cerebral haemorrhage on CT scan). Thus, effectiveness tends to be lower than efficacy.

Patient-oriented outcomes & Surrogate outcomes-

Patient-oriented outcomes should be used rather than surrogate or intermediate outcomes to judge efficacy and effectiveness. Patient-oriented outcomes are those that affect a patient’s well-being. They involve prolongation and better quality of life, improve function or prevent disability, and provide relief from symptoms. Surrogate or intermediate outcomes are factors that do not directly involve the patient’s well-being. They are features such as physiologic parameters (e.g., blood pressure) or test results (e.g., concentrations of glucose or cholesterol, tumour size on CT scan) that are thought to predict actual patient-oriented outcomes. For example, clinicians typically presume that lowering blood pressure will prevent the patient-oriented outcome of uncontrolled hypertension (e.g., death resulting from myocardial infarction or stroke). However, it is conceivable that a drug could lower blood pressure but not decrease mortality, perhaps because it has fatal adverse effects. Also, if the surrogate is merely a marker of disease (e.g., HbA1C) rather than a cause of disease (e.g., elevated blood pressure), an intervention might lower the marker by means that do not affect the underlying disorder. Thus, surrogate outcomes are less desirable measures of efficacy than patient-oriented outcomes.

On the contrary, surrogate outcomes are more feasible to use, for example, when patient-oriented outcomes take a long time to appear (e.g., kidney failure resulting from uncontrolled hypertension) or are rare. In such cases, clinical trials would need to run for a long time unless a surrogate outcome (e.g., lowered blood pressure) is used. In addition, the main patient-oriented outcomes, death and disability, are binary (i.e., yes/no), whereas surrogate outcomes are often continuous, numerical variables (e.g., blood pressure, blood glucose). Numerical variables, unlike binary outcomes, may indicate the magnitude of an effect. Thus, use of surrogate outcomes can often provide much more data for analysis than can patient-oriented outcomes, allowing clinical trials to be done using limited patients for a certain amount of time.

Correlation of patient-oriented and surrogate outcomes- 

However, surrogate outcomes should ideally be proved to correlate with patient-oriented outcomes. There are many studies in which such correlation appeared reasonable but was not actually present. For example, lowering blood glucose to near-normal concentrations in patients with diabetes in the intensive care unit resulted in higher mortality and morbidity (possibly by triggering episodes of hypoglycaemia) than lowering blood glucose to a slightly higher level. Some oral antihyperglycemic drugs lower blood glucose, including HbA1C concentrations, but do not decrease risk of cardiac events. Some antihypertensive drugs decrease blood pressure but do not decrease risk of stroke.

Factors that help in assessing safety index of drugs-

Adverse Effects are clinically relevant undesirable effects that are patient-oriented outcomes, such as, death, disability or discomfort. Surrogate adverse effects (e.g., alteration of concentrations of serum markers) are often used but, as with surrogate efficacy outcomes, should ideally correlate with patient-oriented adverse effects. Clinical trials that are carefully designed to prove efficacy, can still have difficulty identifying adverse effects, if the time needed to develop an adverse effect is longer than the time needed for benefit to occur or if the adverse effect is rare. For example, cyclooxygenase-2, COX-2 inhibitors relieve pain quickly, and thus their efficacy can be shown in a comparatively brief study. However, the increased incidence of myocardial infarction caused by some COX-2 inhibitors, such as Rofecoxib marketed as Vioxx, occurred over a longer period of time that was not apparent in shorter, smaller trials. Hence, clinical trials may exclude certain subgroups and high-risk patients, adverse effects may not be fully known until a drug has been in widespread clinical use for several years. Post marketing surveillance or pharmacovigilance is one way to test for safety of new drugs in long term.

Another factor that ensures safety and efficacy of new drugs is benefit to risk ratio. Known or expected benefits vs. unknown or unexpected risks is when the efficacy of a new drug is tested, a specific type of benefit is expected. Other benefits are usually additional to the outcome sought. While assessing risk, the negative outcomes are often unknown or unexpected, and, unlike the additional benefits, the significance of these adverse effects must be considered to the extent practicable before the drug is considered for acceptable risk. When thalidomide was tested as a sleeping pill, no major negative effects were discovered. Its effects upon the fetus were not tested, and thalidomide was marketed as a safe drug. The birth defects that resulted, vividly demonstrates the need to consider risks from many perspectives. 

Conclusion-

The drug safety concept has earned a lot of attention during the last century due to the fact it plays a direct role in a patient’s health. Recent regulatory laws, stresses that drug safety should be included in the process of new medication’s approval and continued conduct of post-marketing drug evaluations, i.e., pharmacovigilance. Benefit–risk assessment should be considered by all health care professionals when they prescribe specific drugs to specific groups of patients. Hence, drugs with a high risk profile should be avoided unless their benefit outweighs the risk. Drug safety has gone through different stages from the last century till now, with several unfortunate tragedies that incline us to protect our patients from all aspects. All patients should be protected; however, specific groups of patients demand paramount care, such as pregnant women, children, and the elderly, since they are identified as vulnerable populations.