1/58
Flashcards for Exam 4 covering Drug Discovery, Development, Toxicology, Trials, Introduction to Surgery, and Introduction to Anesthesia.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
Define Drug Target.
The specific molecule or structure in the body that a drug is designed to interact with.
Define Lead Compound.
A chemical compound that has pharmacological or biological activity likely to be therapeutically useful, but may still have suboptimal structure that requires modification to fit better to the drug target.
Define Pharmacophore.
The ensemble of steric and electronic features that is necessary to ensure the optimal supramolecular interactions with a specific biological target structure and to trigger (or to block) its biological response.
Define pharmacodynamics.
The study of the biochemical and physiological effects of drugs on the body or on microorganisms or parasites and their mechanisms of action.
Define pharmacokinetics.
The study of the bodily absorption, distribution, metabolism, and excretion of drugs.
Define in vivo.
Experimentation using a whole living organism.
Define in vitro.
Experimentation taking place in a test tube, culture dish, or elsewhere outside a living organism.
Define ex vivo.
Experimentation done on tissue from a living organism in an artificial environment outside the organism with minimum alteration of natural conditions.
Define bioassay.
An analytical method to determine the concentration or potency of a substance by its effect on living cells or tissues.
Define detoxification.
The process of removing toxic substances from the body.
Define toxification.
The process of converting a substance into a more toxic form.
Define mutagenesis.
The process by which the genetic information of an organism is changed, resulting in a mutation.
Define carcinogen.
A substance or agent that is capable of causing cancer.
Define therapeutic index.
A quantitative measurement of the relative safety of a drug. It is a comparison of the amount of a therapeutic agent that causes the therapeutic effect to the amount that causes toxicity.
Define pharmacology.
The study of the effects of drugs on living organisms.
Define toxicology.
The branch of science concerned with the nature, effects, and detection of poisons.
What is the Orphan Drug Act?
Act to stimulate the development of drugs for rare diseases.
List common methods companies use to find lead compounds
High-throughput screening, chemical modification of existing compounds, identifying the mechanism of action of traditional medicines, rational drug design.
Why is selectivity important in drug discovery?
To minimize off-target effects and improve the therapeutic index of a drug.
What is SAR and why is it important?
SAR stands for Structure-Activity Relationship. It is important as it helps to understand the relationship between the chemical structure of a drug and its pharmacological activity.
How does pharmacodynamics and pharmacokinetics assist in determining toxicity and efficacy?
Pharmacodynamics helps understand the drug's effects on the body, while pharmacokinetics helps understand how the body processes the drug. This information is crucial for determining the drug's potential toxicity and efficacy.
What is meant by the ADME of drugs?
ADME stands for Absorption, Distribution, Metabolism, and Excretion, which are the four main processes that determine the pharmacokinetic profile of a drug.
Describe the phases of drug metabolism, and the role of cytochrome P450.
Phase I involves modification reactions often using cytochrome P450. Phase II involves conjugation reactions. Phase III involves elimination. Cytochrome P450 enzymes are crucial in Phase I metabolism.
Name 6 Molecular Mechanisms of Toxicology
Receptor-Mediated Toxicity, Covalent Binding, Oxidative Stress, Disruption of Calcium Homeostasis, Mitochondrial Dysfunction, Interference with DNA/RNA.
Differentiate between type I and type II hypersensitivity reactions.
Type I is an immediate hypersensitivity reaction mediated by IgE. Type II is antibody-dependent cytotoxic hypersensitivity.
Give an example of receptor mediated toxicity
An example of receptor mediated toxicity is a drug directly binds to and overstimulates or inhibits a receptor, leading to toxic effects.
Give an example of a toxin that blocks a biochemical pathway?
Cyanide inhibits cytochrome c oxidase, blocking the electron transport chain.
Define a proto-oncogene vs. a tumor suppressor gene
Proto-oncogenes are genes that promote cell growth and division, while tumor suppressor genes inhibit cell growth and division. Mutations in proto-oncogenes can lead to uncontrolled cell growth, while inactivation of tumor suppressor genes can remove brakes on cell growth.
What two organs are common targets for toxicity?
Liver and Kidneys
Define hepatic necrosis vs. hepatitis vs. cirrhosis
Hepatic necrosis is cell death in the liver. Hepatitis is inflammation of the liver. Cirrhosis is scarring of the liver.
Explain how acetaminophen is toxic.
Acetaminophen is metabolized into a toxic metabolite (NAPQI) that can cause liver damage if glutathione stores are depleted.
What are three ways kidneys are affected by toxins?
Direct tubular damage, glomerular damage, and interstitial nephritis.
What was the thalidomide tragedy about and what important legislature came from this event?
Thalidomide caused birth defects when used during pregnancy. It led to stricter drug regulations and testing requirements.
Compare in vitro vs. in vivo vs. ex vivo
In vitro is testing in a test tube or culture dish. In vivo is testing in a living organism. Ex vivo is testing on living tissue outside the body.
What are the advantages and disadvantages of in vitro vs. in vivo testing?
In vitro: Advantages are faster and cheaper. Disadvantages are less physiologically relevant. In vivo: Advantages are physiologically relevant. Disadvantages are slower and more expensive.
What are three important steps/tests in pre-clinical testing
Toxicity studies, efficacy studies, and pharmacokinetic studies.
What is the LD50?
The dose of a substance required to kill half the members of a tested population after a specified test duration.
What are the functions of the IRB in Clinical Trials
The IRB (Institutional Review Board) is responsible for ensuring the ethical conduct of research involving human subjects. They review research proposals to ensure the safety and rights of participants are protected.
What is informed consent?
Informed consent is the process by which a potential research participant is provided with sufficient information about the study. The participant must adequately understand the research/clinical trial, and provide their agreement to participate.
What are the four phases of clinical trials?
Phase 1: Safety and dosage. Phase 2: Efficacy and side effects. Phase 3: Confirm efficacy, monitor adverse reactions. Phase 4: Post-marketing surveillance.
Define Survival Surgery.
A surgical procedure where the animal recovers and lives after the surgery.
Define Non-Survival Surgery.
A surgical procedure where the anima is euthanized after the surgery, before recovery.
Define Aseptic Technique.
Set of practices used to eliminate contamination from surgical site.
Define Contamination.
The presence of unwanted microorganisms.
Define Sterile Field.
An area prepared to contain sterile instruments and other items needed during a surgery.
Define Antiseptics.
Substances that are applied to living tissue to kill or inhibit the growth of microorganisms.
What is the goal of Aseptic Technique
The goal is to minimize the risk of infection at the surgical site.
List 4 examples of Contamination prevention procedures.
Sterilizing instruments, proper surgical scrubbing, wearing sterile gloves and gowns, using sterile drapes.
What are the criteria for good personnel in a surgical room
Trained in aseptic technique, adherence to protocols, proper attire, and good personal hygiene.
List 3 ways tools are sterilized for surgery
Autoclaving, chemical sterilization, and radiation sterilization.
Define Anesthesia.
A state of temporary induced loss of sensation or awareness.
Define General Anesthetic.
A drug that produces a state of unconsciousness and insensitivity to pain.
Define Local Anesthetic
A drug that causes loss of sensation in a limited area.
Define Analgesia
Relief from pain.
Define Amnesia
Loss of memory.
What are the risks of Anesthesia to Employees?
Exposure to anesthetic gases, which can cause reproductive problems, neurological effects, and other health issues.
What are the components of the Triad of General Anesthesia?
Unconsciousness, analgesia, and muscle relaxation.
List the 4 States of Anesthesia
Induction, Excitement, Surgical Anesthesia, Medullary Depression.
List 5 Reflexes to monitor under Anesthesia
Palpebral reflex, pedal Withdrawal, corneal reflex, laryngeal reflex, and swallowing reflex.