PED2001 Flashcards
What are the two processes that determine drug concentrations
- translocation of drug molecules
- chemical transformation
how do drugs move around the body
- bulk flow (in the bloodstream)
- diffusional transfer (molecule by molecule over short distances)
why does diffusional transfer differ with the chemical nature of the drug
- hydrophobic diffusion barrier
- aqueous diffusion
what is a compartmentalised body
- body made of interconnected compartments separated by cell membranes
- the ability of drugs and other chemicals to move between these compartments depends on the selectivity of the membranes and the chemical properties of the drug
what is the vascular endothelium
- acts as a filter (cut off MW 80-100k)
- the gaps between cells are filled with a protein matrix - tight packed - this acts as MW filter
why is the endothelium discontinuous in liver and spleens
- large fenestrations allows drugs to exchange freely between blood and interstitium in the liver
what is the structure of endothelial cell in liver
- endothelium
- basement membrane
- slit junctions
structure of CNS and placenta
- astrocyte foot processes - lipophilic barriers
- tight junctions
- slit junctions
- basement membrane
what is the importance of the structure of the barriers in CNS and placenta
- charged drugs cannot move through
- lipid soluble and carrier mediated transport can be used to move from plasma to CNS
how can drugs be transferred across cell membranes
- diffusing direct through lipid
- diffusing through aqueous pores - most drugs are too big to move through
- combination with a transmembrane carrier protein
- pinocytosis (for macromolecules e.g. insulin
how do drugs diffuse through lipid
- non-polar substances dissolve readily in non-polar solvents - cell membranes are lipid-rich environments
how is permeability coefficient determined
- the number of molecules crossing the membrane per unit area of time (J)
- concentration difference across the membrane (delta C)
- J = P x delta C
what are the physiochemical factors contributing to permeability
- partition coefficient
- diffusion coefficient
- diffusion coefficient is equal to 1/square root MW
what is the relationship between lipid solubility and permeability
- close correlation between lipid solubility and permeability of cell membrane to different substances
- lipid solubility an important determinant of pharmacokinetic characteristics of a drug
- rate of absorption from the gut, penetration into brain and other tissues, and extent of renal elimination can be predictable
how is pH and ionisation involved in absorption
- ionised drug formed are much less able to penetrate cell membranes
- ratio of charged drug/uncharged drug concentrations is determined by the pH of the compartment
how is lipid solubility involved in absorption
- the lipid solubility of uncharged species AH or B depend on the chemical nature of the drug
- for many drugs the uncharged species is sufficiently lipid soluble (except e.g. aminoglycosides0
- bases are neutral and absorbed easily
what does ionisation effect
- drug permeability across membranes
- ionised drugs show reduced permeability
- steady state distribution of drug molecules between aqueous compartments, in the presence of a pH mechanism
what is the pH partition mechanism
- qualitative effects of pH changes in different body compartments on the pharmacokinetic of weak acids and bases
- but its not the main determinants of drug absorption from the GI tract
- small intestine has hugely greater surface area of absorption compared to stomach
why does aspirin absorption vary
- aspirin absorption increased by metoclopramide and decreased by propantheline
- ionised forms are not totally impermeable
- body compartments are rarely at equilibrium in real life
what are the consequences of the pH partition mechanism
- urinary acidification increases excretion of weak bases and decreased that of weak acids
- urinary alkalisation decreases excretion of weak bases and increases that of weak acids
- increases plasma pH (e.g. sodium bicarbonate) causes extraction of weakly acidic drugs from CNS into plasma
- decreases plasma pH (e.g. acetazolamide) causes weakly acidic drugs to accumulate in the CNS
what is the structure of the nephron
- distal tubule collecting duct (control Na and H2O balance)
- glomerulus (renal blood flow filtration
- loop of hence (urinary concentration
how does urinary alkalinisation increase excretion of aspirin
- at normal urinary pH, a proportion of salicylate is unionised and can be absorbed back into the systemic circulation in the nephron
- when urine is alkaline, salicylate is charged, so reabsorption is much reduced
what is carrier mediated transport
- transport of physiologically important molecules in and out of cells (e.g. sugars, amino acids, neurotransmitters and metal ions)
what are the examples of carrier mediated transport
- passive transport - move molecules in direction of electrochemical gradient
- active transport - movement against an electrochemical gradient coupled to an energy source
what are the properties of carrier mediated transport
- saturable
- can be inhibited
why is renal handling of cisplatin important
- cisplatin is very nephrotoxic because it is efficiently taken up into the proximal tubule, but rate of secretion is lower
- higher exposure results in destruction of mitochondria - proximal tubular cell death
- organic solutes lost to urine increased Na loss increases H2O loss - falcon syndrome
what are the carried mediated transport systems
- levodopa - transported by the carrier responsible for phenylalanine
- fluorouracil - transported by carrier for natural pyrimidine - thymine and uracil
- iron - carrier system in jejunum
- calcium - vitamine D dépendent carrier system
where in the body are carrier mediated transporters essential
- the renal tubule
- the biliary tract
- the blood brain barrier
-the GI tracts - p-glycoprotein - transporter responsible for MDRa
where else are carrier mediated transporters present
- renal tubular brush border
- membranes, in bile canaliculi, in astrocyte foot processes in brain micro vessels and in GI tracts
what are the other factors affecting drug pharmacokinetics (distribution and elimination)
- binding to plasma protein
- partition into body fat and other tissues
what are the routes of drug administration
oral
sublingual
rectal
application to other epithelial surface (e.g. skin, cornea vagina and nasal mucosa)
inhalation
injection
what are the types of injection
subcutaneous
intramuscular
intravenous
intrathecal
factors affecting GI absorption of drugs
GI motility
splanchnic blood flow
particle size and formulation
physiochemical factors
what does gastrointestinal motility have an effect on
migraine, diabetic neuropathy
malabsorption states and GI diseases
coeliac disease
drugs
food
how does coeliac disease effect gastrointestinal motility
- thyroxine anddigoxin absorption decreased
- propranolol, cotrimoxazole and cephalexin absorption increased
what are the physiochemical factors that effect drug GI absorption
- tetracycline and calcium ions
- bile acid binding resins (e.g. cholestyramine) interact with warfarin, thyroxin
what are the drugs not absorbed GI
- vancomycine (used for treatment of pseudomembranous colitis caused by clostridium difficile)
- mesalazine (a formulation of 5-aminosalicyclic acid) for treatment of crohns disease)
- olsalazine (a dimer of two 5-aminosalicyclic acid) cleaved by colonic bacteria
what is systemic availability
the amount of drug that reaches systemic circulation intact
what does the rate of systemic availability rely on
- pharmaceutical factors
- gastrointestinal absorption
- pre systemic metabolism relatively unimportant
what does the extent of systemic availability depend on
both the extent of absorption and the extent of pre-systemic metabolism
what is cutaneous drug administration
- skin allows permeation of drugs from topically applied creams and ointments in quantities sufficient to produce systemic action
what are the problems with transdermal drug delivery
- variability in drug administration through skin
- resulting in lack of precision regarding the real dose absorbed
what are the therapeutic objectives of transdermal delivery
- lack of drug dosage a characteristic of topically applied ointment and creams
- ointments containing nitroglycerin require multiple applications per day
- variable amount and duration of drug input
why is there variable amount and duration of transdermal drugs
- differences in the area of skin covered with ointment
- thickness of the ointment layer applied
what are the new topical dosage forms
- delivery drugs to the blood stream at defined rates over prolonged period of time
- design emphasis on control of systemic input residing in the dosage form rather than in the skin
why is transdermal therapy particularly suitable for the rate controlled administration
- as potent, non-irritating, non-allergenic agents with suitable physiochemical properties whose current administration causes some problems
what are some of problems of ointments
- troublesome side effects or unreliable therapeutic action with repetitive dosing with conventional dosage forms
- patient compliance difficulaties
- need for frequent dosing in conventional dosage forms because of the drugs short biological half life
- gastric irritations with oral therapy
what is the scopolamine system
- application to skin
- drug diffusion in the direction of concentration gradient
- energy source (the difference in the drugs chemical potential between the reservoir and the systems exterior)
- constancy of rate delivered assured as long as drug present in excess in reservoir
what is intravenous drug administration
- fastest and most certain route
- single bolus injection with high concentration of drugs
- peak drug concentration reaching tissues is critically dependent on the rate of injection
- steady IV infusion avoids high peak plasma concentrations (e.g. lignocaine, propofol, diazepam)
what is the benefits of subcutaneous and intramuscular drug injections
SC and IM injections usually produce faster effects than oral administration
what does the rate of absorption of SC and IM injections depend on
- the site of injection
- local blood flow
- drug formulations
what are intrathecal injections
injections into subarachnoid space via a lumbar puncture
what are the examples of intrathecal injections
- methotrexate for treatments of childhood leukaemia
- local anaesthetics (e.g. bupivacaine)
- opiate analgesics
- baclofen - for treatments of muscle spasm cause by chronic neurological disease
- some antibiotics (ahminoglycosides) - treatments of nervous system infection
what diseases require drug administration by inhalation
asthma
bronchitis emphysema
lung cancer
respiratory diseases
why are drugs administered by inhalation
rapid action - rapid delivery across mucous membranes of the respiratory tract and pulmonary epithelium
minimise systemic absorption
minimise side effects (beta-agonists, glucocorticoids
what is asthma
- inflammation (swelling)
- mucus production (snot)
- bronchospasm (muscle tightness)
what are the symptoms of asthma
- shortness of breath
- wheezing
- tightness in the chest
- coughing at night or after physical activity
- waking at night with asthma symptoms
what are the indoor triggers of asthma
strong smells
cockroaches
smoke
dusty mites
furry friends
colds
mould
what are the outdoor triggers for asthma
cold/hot weather
car exhaust
pollens
exercise
mowed lawn
air pollution
grilling
How to treat the symptoms of asthma
- adrenergic agonists (bronchodilators) and glucocorticoids (usually by inhalation)
- beta agonists (e.g. pirbuterol, terbutalines, albuterol and salmeterol)
- relax airway smooth muscle directly
- provide relief for 4-6hours
- little stimulation of alpha or beta1 receptors
what is the mechanism of action of glucocorticoids
- inhaled glucocorticoids reduce/eliminate the use of oral glucocorticoids
- no direct effect on airway smooth muscle
- decrease the number and activity of cells involved in airway inflammation
- prolonged inhalation of steroid reduced hyper-responsiveness of the airway smooth muscle
what are the pharmacokinetics of inhaled drugs
- a large fraction deposited in the mouth and pharynx or swallowed
- many of the clinically useful corticosteroids (e.g. beclomethasone, triamcilonone) undergo extensive 1st pass metabolism
- therefore a small amount reaches the systemic circulation which minimises adverse effects
- 10-20% of inhaled dose reaches the airway
what is theophylline
- a potent bronchodilators
- narrow therapeutic window
- overdosing can lead to seizures and cardiac arrhythmias
- interact with many other prescribed drugs
what are the other inhaled drugs for treatment of asthma
- sodium cromoglycate and nedocromil sodium
- effective prophylactic anti-inflammatory agents
- but not useful in managing acute attacks of asthma - not direct bronchodilators
- theophylline
what is the main benefit of prolonged release inhalation formulations
- current formulations have short duration of clinical effects therefore advantageous to prolong pharmacological effect
what factors affect bioavailability of drugs
- the rate of disintegration of the tablet
- the rate of dissolution of the drug particles in the intestinal fluid
what are the physical factors affecting pharmaceutical availability
- tablet compression and excipients - affect the rate of tablet disintegration
- other tablet excipients - affect interaction with aqueous GI juices
- the form of the drug e.g. crystalline or salt form
- particle size - smaller drug particles dissolve more quickly
what is bioequivalence
- two formulations of a drug are bioequivalent if they show comparable bioavailability and similar times to achieve peak plasma concentrations
what is bioinequivalence
- two formulations of a drug with a significant difference in bioavailability are said to be bioinequivalent
what is therapeutic equivalence
two similar drugs are therapeutically equivalent if they have comparable efficacy and safety
what is the importance of alterations in pharmaceutical availability
- important for drugs with narrow therapeutic index
- switch from a formulation of low pharmaceutical availability to a formulation of high pharmaceutical availability
what are the determining factors for route of administration of drug
- therapeutic objective (slow or fast onset of action)
- properties of the drug
how can we control insulin absorption
- physical state - crystalline or non-crystalline
- the zinc or protein content
- the nature and pH of the buffer suspension
what are the types of subcutaneous injections of insulin
- insulin BP - soluble and amorphous - rapid onset and short duration of action
- ultralente insulin (large crystals of insulin and high zinc content - suspended in a solution of sodium acetate/sodium chloride - onset of action approx. 7hrs and duration of action of 36hrs
what are the examples where the vehicle in which the drug is suspended can affect the drug diffusion rate
- the absorption of drug from IM injection site may be retarded by the use of thick oils which slow down diffusion
- vasopressin tannate in oil - diabetes insidious
- fluphenazine decanoate in oil - schizophrenia
what are the examples where plasma drug concentrations differ between IM and oral dosing of the same drug
- phenytoin - plasma drug concentrations after IM injections are half of those after oral dosing
- chloramphenicol - also poorly absorbed after IM injection
what is the relation between diabetes insidious and lack of ADH (vasopressin)
- ADH (vasopressin) inserts water pores into collecting duct membrane to enable re-absorption of solute-free water from the collecting duct into the systemic circulation
- vasopressin binds to membrane receptor
- receptor activates cAMP (second messenger system)
- cell inserts AQP2 water pores into apical membrane
- water is absorbed by osmosis into the blood
why are local injections used
- some formulations of local anaesthetics contain adrenaline
- vasoconstriction at site of injection
- prevent the drug to be carried away by circulation from site of injection
- prolongs the effect of local anaesthetic
why use sublingual, buccal, rectal and transdermal formulations
- avoidance of 1st pass metabolism - resulting in rapid therapeutic effect
- corticosteroids can be given by the rectal route for direct effect on the large bowel
what is the example of the benefit of avoidance of 1st pass metabolism
- glyceryl trinitrate 10x less of dose required for therapeutic effect compared to local dosing
what are the step that allow corticosteroids to be given by the rectal route
- drug administration via transdermal patches
- controlled release of small amount of drug over a period of time
- e.g. glyceryl trinitrate, hyoscine, oestradiol
what are the benefits of controlled release of drug
- reduce the risk of gastric erosions - e.g. quinidine
- ideal for drugs with short duration of action e.g. theophylline and nifedipine
- unconventional formulations are not always useful e.g. beta-antagonists show good duration of effect from conventional formulation
what is the criteria that needs to be met for combination products in oral therapy
- the frequency of administration of the two drugs is the same
- the fixed doses in the combination product are therapeutically and optimally effective
what are the potential advantages of combination formulations
- improved compliance
- ease of administration
- synergistic or additive effect
- decreased adverse effects
What is an example of improve compliance
- antituberculous drugs (rifampicin and isoniazid)
- ferrous sulphate and folate acid (pregnancy)
what is an example of ease of administration
triple vaccine (diphtheria, tetanus, pertussis)
what are the examples of synergistic or additive effect
- e.g. trimethoprim and sulphonamides (e.g. co-trioxazole)
- amoxycillin and clavulanic acid (co-amoxiclav)
- aspirin and codeine (simples analgesia)
- paracetamol and metoclopramide (migraine)
- combines oral contraceptives (oestrogen and progesterone)
what is the example of decreased adverse effects
- e.g. -dopa and decarboxylase inhibitors (Parkinson’s)
- diuretics (potassium-wasting and potassium sparing)
what are the types of special drug delivery systems
- biologically erodible microspheres - loaded with drugs
- pro drugs - cyclophosphamide, levodopa, zidovudine
- antibody-drug conjugates - cancer chemotherapy
- packaging in liposomes
- gene therapy - viral vector for gene delivery
- implantable devices
when are special drug delivery systems used in clinical application
- brentuximab vedotin - directed to the protein CD30, which is expressed in classical Hodgkins lymphoma
- trastuzumab emptansine (Herceptin) - binds to the HER2/neu receptor - treatment of HER2-positive metastatic breast cancer
what is packaging in liposomes
- phospholipids loaded with non-lipid soluble drugs
- reticuloendothelial cells in the liver - also concentrated in malignant tumours - selective delivery
- e.g. amphotericin - treatments of mycosis - less nephrotoxic and better tolerated
- Pfizer SARS cov2 vaccine - mRNA packaged in liposome
what is drug distribution
process by which a drug reversibly leaves the bloodstream and enters the interstitial (extracellular fluid) and/or the cells of the tissues
what does the delivery of a drug from plasma to the interstitial depend on
- blood flow
- capillary permeability
- the degree of binding of the drug to plasma and tissue protein
- the relative hydrophobicity of the drug
what does blood flow to tissue capillaries depend on
blood flow to tissue capillaries varied widely as a consequence of unequal cardiac output to various organs
what is the direction of blood flow to the organs
- blood flow to the brain, liver, kidney –> skeletal muscle –> adipose tissue
what does capillary permeability depend on
- capillary structure
- drug structure
what is the capillary structure
large fenestrations allow drugs to exchange freely between blood and interstitium in the liver
how do drugs interact with blood-brain barrier
- drugs must pass through the endothelial cells of the capillaries of CNS or be actively transported e.g. levodopa
- lipid soluble drugs readily penetrate the CNS
- ionised or polar drugs unable to pass through the endothelial cells of the CNS
how does hydrophobicity affect drug movement
- hydrophobic drugs readily cross cell membranes
- major factor in distribution of hydrophobic drug is blood flow to the area
- hydrophilic drugs must go through slit junction
what are the characteristics of binding drugs to proteins
- reversible binding to plasma proteins sequesters drugs in non-diffusible form
- binding it non-selective as to chemical structure
- binding sites for drugs similar to those for bilirubin
- plasma albumin conjugate –> free drug –> metabolism –> excretion
what is volume of distribution
- Vd is a hypothetical volume of fluid into which the drugs is disseminated
- has no physiological or physical basis
what is the plasma compartment
- drug has a large MW or binds extensively to plasma protein
- too large to move out through slit junctions of capillaries therefore drug trapped within the plasma compartment
- Vd = plasma water
how does the low molecular impact drug movement
- drug has a low MW but is hydrophilic
- moves through slit junctions into interstitial fluid
- unable to cross lipid membrane of cells and enter intracellular fluid
- Vd = sum of plasma water and extracellular fluid volume
what is the calculation for Vd
plasma water + extracellular + intracellular volumes
why are bound drugs pharmacologically inactive
- unable to reach target site and elicit a biological response
- by binding to plasma proteins - the drug is effectively trapped
what is the binding capacity of albumin
- binding of a drug to albumin is reversible
- low capacity - one drug molecular per albumin molecule
- high capacity - several drug molecules binding to a single albumin molecule
what is the binding affinity of drugs to albumin
- albumin has strongest affinity for anionic and hydrophobic drugs
- most hydrophilic and neutral drugs do not bind to albumin
what is the competition for binding between drugs
- drugs with high affinity for albumin can compete for available binding sites
- drugs with high affinity for albumin can be divided into two classes
- dependent on dose of the drug > or < than the binding capacity of albumin
what are class I drugs
- dose of drug < binding capacity of albumin
- dose/capacity ratio is low, therefore binding sites in excess of the available drug
- most drug molecules are bound to albumin and concentration of free drug is low
what are class 2 drugs
- dose of drug > the number of albumin binding sites
- dose/capacity ratio is high, most albumin molecules contain a bound drug; the concentration of free drug is significant
- a relatively high proportion of drug exists in free state
what is the clinical importance of drug displacement
- e.g. interaction between tolbutamide (class I drug: 95% protein bound), and sulphonamide antibiotic (class II drug)
- displacement of class I drug occurs when a class ii drug is administered simultaneously
- rapid increase in free fraction of tolbutamide in plasma
- a new equilibrium will be reached
what is the relationship between drug displacement and Vd
- impact of displacement dependent on both Vd and the therapeutic index
- if Vd is large, then change in free drug concentration is insignificant
- if Vd is small, the newly displaced drug does not move into the tissue as much
- increase in plasma drug concentration is more profound when Vd is small
what is the Vd when high MW and/or protein bound drugs
trapped in plasma, low Vd
what is the Vd with low MW, hydrophilic drugs
Vd = plasma water + interstitial fluid
what is the Vd with low MW, hydrophobic drugs
Vd is high
what is xenobiotic metabolism
process by which foreign compounds are metabolised in the body to facilitate their elimination
what are the sites of metabolism
- liver - main site of metabolism
- GI tract
- kidney
- skin
- lungs
- plasma - hydrolysis
- brain
what is the liver function
- detoxicification
- metabolism of carbohydrates, lipids and proteins
- synthesis of plasma proteins
- storage of glycogen, vitamins and minerals
- bile products
what are the cell types of the liver
- hepatocytes
- cholangiocytes
- Kupffer cells
- stellate cells
- endothelial cells
- fibroblasts
what is the liver structures
liver
liver lobules
lobule
what is in the liver lobule
- hepatocytes
- central vein - feeds into hepatic vein
- sinusoid
- branch of hepatic artery
- branch of portal vein
- bile ductile
what is liver regeneration
- damaged liver cells can be replaced by liver regeneration
- limited process - repeated damage and exhaustion of regeneration leads to fibrosis and cirrhosis
what causes the need for liver regeneration
exposure to xenobiotics can stress and damage the liver - more likely to be damaged buy reactive metabolites
- hepatocyte proliferation
- liver stem/progenitor cell
where are microsomal enzymes found
- smooth ER is the major site of drug metabolising enzymes
- smooth ER are high in abundance in hepatocytes
where are non-microsomal enzymes found
cytosol
mitochondria
what reaction are included phase 1 of drug metabolism
most mediated by cytochrome P450 enzymes
oxidation and reduction reactions
what reaction are included in phase 2 drug metabolism
conjugation reactions
what happens during phase 1 metabolism
functionalisation reactions where a functional group is introduced normally producing a more polar excitable molecule
which molecules require phase 2 metabolism before they can be excreted
esterases
oxidases
epoxide hydrolyses
dehydrogenases
what is the structure of cytochrome P450
haem proteins - 57 Human CYPs
distinct but overlapping substrates
what reactions are cytochrome P450s involved in
genetic polymorphism
genotype-phenotype relationship
subject to induction and inhibition
drug-drug interactions
what is included in cytochrome P450 induction and inhibition
- CYP3A4 - responsible for metabolising 50% of drugs
- wide substrate specificity
- most abundant in liver and gut
what is an examples of induction of cytochrome P450
St johns wort - induction of CYP3A4 and P-gp
what is an example of CYP450 inhibition
grapefruit juice - irreversible inhibition of CYP3A4
what happens during phase 2 metabolism
conjugation reactions which detoxify compounds and prepare them for excretion
why do some drugs not require phase 1 metabolsism before phase 2
when suitable functional groups for conjugation are already present on the molecules
what are the examples of types of drugs that don’t require phase 1 metabolism before phase 2
glucuronyl
sulphase
methyl
acetyl
what are the enzymes involved in phase 2 metabolism
UDP-glucuronosyltransferases
sulphontransferases
glutathione S-transferases
N-acetyltransferases
methyltransferases
amino acid conjugating enzymes (e.g. glycine and glutamic acid)
what is the relationship between first pass metabolism and bioavailability
first pass metabolism in the liver and gut wall reduces the bioavailability of drugs given PO
What are the factors that can affect first pass metabolism
- genetic variations between individuals in the liver and GI
- variations between individuals in the liver and GI blood flow
- gut mitochondria
what are the characteristics of prodrugs
increase solubility
high first pass metabolism
instability
poor absorption
target drug to specific sites
taste
pain at site of administration
improve PK
reduce toxicity
how are prodrugs administered
administered as an inactive form and require metabolism to become active
what are the examples of pro drugs
diacetyl-morphine
codeine
cyclophosphamide
what are cytochrome P450 enzymes
- most important family in phase 1 metabolism
- contain single haem molecule as prosthetic group
where are cytochrome P450 enzymes found
endoplasmic reticulum
mitochordria
what is the function of cytochrome P450 enzymes
forms complex that shows maximum absorbance at 450nm when reduced and CO added
what is the cytochrome P450 nomenclature
use root CYP followed by family, subfamily and form number
sometimes include allelic variants
what are the functions of cytochromes P450
- xenobiotic metabolism
- steroid, fatty acid and vitamin oxidation
- steroid biosynthesis
where does xenobiotic metabolism occur
occurs in endoplasmic reticulum
where are the cytochromes P450 for steroid biosynthesis found
mitochondria
what are the biochemical properties of cytochromes P450
- monomeric proteins of molecular weight 40 000 to 50 000
- all contain haem as a prosthetic group
- regions of the protein concerned with haem and oxygen binding tend to be conserved but area important for substrate binding vary more in sequence
- crystal structures of most human P450s relevant to xenobiotic metabolism not available
what is the cytochrome P450 reaction
DH + NADPH + O2 –> DOH + NADP + H2O
What is the function of NADPH in the cytochrome P450 reaction
- NADPH supplies 2 protons and 2 electrons in an electron transfer process
- interacts with cytochrome P450 enzyme by electrostatic interaction involving carboxyl groups on the reductase and amino groups on the cytochrome P450
what is the name of the protein used for electron transfer
flavoprotein containing a prosthetic group flavin adenine dinucleotide and flavin mononucleotide