• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/195

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

195 Cards in this Set

  • Front
  • Back
A term often used for an array of substances normally present in the body or formed there. Usually these active substances have a brief lifetime and act near their sites of synthesis. They are not neurotransmitters or hormones, but are sometimes called local hormones. It is a term sometimes used to refer to inflammatory mediators.
Autacoids
Antihistamines and leukotriene modifiers
target _______ mediator(s)
a single
Anti-inflammatory steroids, NSAIDs target _______ mediator(s)
multiple
_______ inhibitors stop mediator production
Synthesis
Antagonists or inverse agonists ______ mediator action
stop
What mediator has these actions?
Vasodilation
Increased vascular permeability
Airway constriction
Not chemotaxis
HIstamine
What mediator has these actions?
vasodilate
increase vascular permeability cause pain
PGE and PGI
Which mediators cause bronchoconstriction?
PGD and TX
Which mediator causes platelet aggregation? Which opposes platelet aggregation?
TX
PGI
Which leukotriene is chemotactic (PMNs) and reduces pain threshold?
LTB4
Which leukotrienes cause airway constriction, increased vascular permeability, chemotaxis (eosinophils)
LTC4, LTD4, LT3E4
Which mediator does everyting, especially vasodilation with hypotension... but not a major chemotactic agent
Kinins
Histamine, PGE2, PGI2, and Kinins all cause what symptom?
Redness (vasodilation)
Histamine, Peptidoleukotrienes, and kinins all cause what symptom?
Swelling - increased vascular permeability
PGE, PGI, LTB4, and Kinins all cause what side effect?
Pain - causes pain or reduces the pain threshold
LTB4 and Peptidoleukotrienes cause what symptom?
Chemotactic - directed migration of WBC.
LTB4 - neutrophils, etc
Peptidoleukotrienes
Which mediators induce fever?
PGEs
Histamine, Peptidoleukotrienes, Kinins, and PGD2 all cause what symptom?
Airway constriciton - bronchoconstriction (relevant for type 1 hypersensitivity)
Kinins and Histamines cause what symptom?
Hypotension = decreased bp (relevant in shock)
________ is a little, active, endogenous amine found in high concentrations in skin, lung, and stomach (portals of entry)
Histamine
There are two pools of histamine:
1) in ______ and _____ cells, within granules, bound by _____ bonds to heparin protein complex
2) Non mast cell histamine - rapid turnover, in CNS cells, epidermis, tissues undergoing rapid growth or repair
basophils and mast cells
ionic bonds
Histidine is coverted to Histamine in what kind of cells? Done by enzyme L-Histidine decarboxylase which does what action?
Basophils and mast cells
Takes off a carboxyl group
Histamine metabolites have strong/moderate/weak pharmacological activity. Metabolism enzymes widely distributed, including histamine specific enzymes
weak - little or no pharmacological activity
If you eat histamine, large doses don't even cause effects because it's inactivated by histaminase in the _____ or intestinal wall. Intestinal ________ also convert it to N-acetylhistamine
liver
bacteria
When you inject histamine into your skin you get _____ and pain first, then the triple response which includes localized _____, flare, and localized _____ (wheal formation)
itching
redness
edema
____ formation occurs in 1-2 min in the same area as the localized redness from histamine. Increased permeability with leakage of the postcapillary venules
wheal
Intranasal histamine causes intense itching, 2 reflexes?, and nasal blockage
sneezing and hypersecretion
IV administration of histamine leads to blood pressure ________
decrease -
vasodilation, increased capillary permeability and fluid loss. Sometimes there’s a secondary increase in blood pressure due to histamine-induced release of catecholamines from the adrenal medulla.
IV administration leads to ______ heart rate.
Increased - reflex response elicited by the decrease in blood pressure
Bronchoconstriction, flushing of face, headache, wheal and flare, mucus and gastric acid secretion- are all from IV administration of what?
histamine
Antigen interaction with IgE antibody on mast cells and basophils caused enogenously released ________
histamine (either into bloodstream of locally)
Clinical uses of histamine are limited - inhale to assess _______ reactivity, use intradermally toa ssess integrity of ______ neurons
bronchial
sensory
Antihistamines are ______ agonists, also known as ________ antagonists
Inverse
competitive
Antihistamines shift the dose-response curve to the ____ because they are a _______ antagonist
right
competitive
Stimulation of the __ receptor causes bronchoconstriction, contraction of __ smooth muscle, wheal formation, itch (pruritis), release of catecholamines from the ____ ______
H1
GI
adrenal medulla
Stimulation of the __ receptor causes gastric acid secretion, inhibition of IgE mediated basophil histamine release (feeds back on self), inhibition of t-cell cytotoxicity, suppression th2 cells and cytokines
H2
receptors __ and __ have no clinically useful agents. __ may be important for nerve cells and __ for immune cells
H3 and H4
H3
H4
Mixed H1 and H2 receptor cardiac effects:
_______ HR
_______ force of contraction
_______ arrhythmias
_______ AV conduction
increased
increased
increased
slows
Vasodilator effects:
__ - rapid, short dilator response
__ - slower and more sustained dilation
H1
H2
Triple response caused by these histamine receptors:
Vasodilation (__ and __)
_____ H1 (prob H2)
Wheal - __ primarily
H1 and H2
Flare
H1
Nasal symptoms - stimulant action on nerve endings are generally __
H1
nasal blockage has some H2, mucus production H2
Classical, first generation antihistamines block __, muscarinic, alpha adrenergic and _____ receptors.
H1 receptors have 45% homology with _______ receptors
H1
serotonin
muscarinic
The second generation antihistamines have minimal _______ properties and are non-sedating and no drying of secretions
anticholinergic
First generation antihistamines are well absorbed orally, distributed widely (even CNS), used as ophthalmic solution, but shouldn't be used _______ due to allergic response development
topically
First generation antihistamines stay in the CNS because they aren't recognized by _-________ efflux pumps on _______ cells
P-glycoprotein
endothelial
Adverse effects of classical antihistamines:
Sedation - potentiate effect of CNS depressants
__ Disturbances
Drying of secretions - due to _______ properties
G.I.
Anticholinergic
Classic antihistamine poisoning leads to atropine-like symptoms (what are they?), excitiations, hallucinations, incoordination, convulsions, coma, cardioresp collapse
fixed-dilated pupils, flushed face, fever with dry mouth
Diphenydramine/Tripelennamine/Chlorpheniramine
Low incidence of GI side effects, sedation
Diphenhydramine
Diphenhydramine, Tripelennamine, and Chlorpheniramine are all what?
Older antihistamines (first generation)
Diphenydramine/Tripelennamine/Chlorpheniramine
GI side effects common, feeble central effects, fewer anticholinergic effects than others
Tripelennamine
Diphenydramine/Tripelennamine/Chlorpheniramine
Most suitable for daytime use
Chlorpheniramine
Newer antihistamines (second generation) have minimal _______ properties, do not cause _______ or drying of secretions
anticholinergic
sedation
Non-sedating Antihistamines penetrate the _____. Have affinity for the P-glycoprotein efflux pump. Do not potentiate the effects of CNS ______
CNS
depressants
What class are these drugs: Cetirizine, Fexofenadine, Loratadine
Non-sedating antihistamines (second generation)
These are not cardiotoxic unlike the first non-sedating antihistamines made
H1 antihistamines (new and old) are good against allergies: allergic rhinitis (relieve sneezing, itching, rhinorrhea), Urticaria (____), atopic dermatitis (poison ivy), NOT for ______
hives
asthma NO
H1 histamines are good against allergies and ____ ______
motion sickness - older antihistamines
this is the antimuscarinic effect
The only reason H1 antihistamines might help a cold would be what?
Older antihistamines dry secretions so could reduce rhinorrhea of common cold
What class are these?
Cimetidine, Famotidine, Ranitidine, Nizatidine
They are inverse agonists
H2 antihistamines
__ antihistamines inhibit histamine, _____ ___ secretion from decreasing muscarinic agonism and gastrin and pentagastrin-induction
H2, gastric acid
What are H2 antihistamines used for?
Ulcers and gastric hypersecretory states
What are Prostanoids?
Prostaglandins and Thromboxane - derivatives of prostanoic acid, a 20 C fatty acid containing a cyclopentane ring
What is the common precursor for Leukotrienes, Prostanoids, and Epoxygenase products?
Arachidonic acid made from Phospholipase A2 and phospholipids in the cell membrane
The availability of ________ is a control step in the production of PG and TX
arachidonic acid
PG1's (1 double bond) precursor is 8,11,14-eicosatrienoic acid.
PG2's (2 double bonds) precursor is ?
arachidonic acid
ARACHIDONATE METABOLISM
Cyclooxygenase pathway makes:
Lipoxygenase pathway makes?
PG and TX
Leukotrienes
Are PG stable/unstable, long/short half life, local/distal actions
short, unstable, local actions
_______ is the key enzyme for the two step synthesis of PGH2 in the cell
cyclooxygenase. Two isozymes COX-1 and COX-2
COX-_ is found in platelets and is constitutively expressed in most cells. Thought to protect gastric mucosa
COX-1
COX-_ not found in platelets, expressed constitutively in the brain and kidney and can be induced by some things at inflammation site. More important isozyme in the production of PG and TX in inflammation
COX-2
COX products degrade quickly via spontaneous chemical ______. Uptake into cells by transport protein and subsequent enzymatic degradation
hydrolysis
Sources of COX:
Numerous cell types and different stimuli
___ and ___ are synthesized on demand and liberated from cells
PG and TX
prostaglandins and thromboxane
Cells commonly produce more than one type of prostaglandin. The type of PG produced by particular cell type is influenced by expression pattern of ______.
enzymes
Platelets Prostacyclin
Endothelium PGD2
Mast Cells Thromboxane

Which cell type goes with which prostaglandin?
Platelets - Thromboxane (vaxoconstrictor)
Endothelium - Postacyclin (vasodilator)
Mast cells - PGD2 (bronchoconstrictor)
Action of COX product in a tissue determined by which _____ receptor. There are _ major types, they are 7 transmembrane G protein coupled receptors
Prostanoid
5
Match receptor with prostaglandin for which they have greatest affinity
DP PGI2
FP PGE
IP PGF
TP PGD
EP (4 subtypes) TXA2
DP - PGD
EP - PGE (4 subtypes)
FP - PGF
IP - PGI
TP - TXA2
Each receptor has a different G protein and therefore a different ____ _______
second messanger
PG and TX receptors are characterized in part by their ability to mediate ______ aggregation or _____ ______ constriction or dilation
platelet
smooth muscle
Prostaglandins can induce fever with ___ being the most potent
IL-1 -> PG's -> fever
PGEs
Increased vascular permeability and vasodilation due to what 2 prostaglandins?
PGEs and PGI2's
PGEs cause pain
PGEs and ___ lower pain threshold or sensitize pain receptors
PGI2
Cytokines, Bradykinin, Other mediators cause _______ production and result in pain
Prostaglandin (PGE)
Therapeutic relevance of drugs that inhibit COX (cyclooxygenase)
Inflammation (redness, swelling, heat, pain), Fever, CV disease (PG and TX very important in balance of platelet aggregation)
NSAIDs, COX inhibitors, aspirin-like drugs, analgesic, anti-inflammatory, anti-pyretic drugs all have what action?
Inhibit cyclooxygenase
Drugs that inhibit COX 1 and 2:
Analgesia, Antipyretics, Anti _______
inflammatory
The analgesia of COX-1 and 2 inhibitors leads to pain of low intensity with little ____ _____ and no addiction compared to opioids. ___ lower pain threshold, so blocking them restores it
side effects
PG's
The antipyretic effect of COX-1 and 2 can reduce body temp in febrile states
Inflammation -> IL-_ -> PGE2 -> Hypothalamus -> fever

The action of ______ in hypothalamus results in elevated body temp
IL-1
PGE2
What do Aspirin, Ibuprofen, Naproxen Diflunisal, Ketoprofen, Indomethacin, Sulindac, Piroxicam all have in common?
What makes Aspirin different form the other NSAIDs?
They all inhibit cyclooxygenase (COX 1 and 2)

Aspirin irreversibly acetylates COX (need to produce new platelets before it's reversed)
Aspirin ________ acetylates COX
irreversibly
In what respect is Ibuprofen better than Aspirin?
Ibuprofen has fewer GI side effects than aspirin
Recognize the following names as what?
Piroxicam, Sulindac, Indomethacin, Ketoprofen, Diflunisal, Naproxen and Ibuprofen (both OTC)
Nonselective COX inhibitors - traditional NSAIDs
What is unique about Celecoxib? (Celebrex)
Selective COX2 inhibitors or "coxibs" (10-20x more selective for COX2)
Which common over the counter drug is NOT an NSAID, and is analgesic, antipyretic, but NOT anti-inflammatory?
Acetaminophen - weak inhibitor of COX. Inhibits COX in the brain but not at sites of inflammation
Hepatic injury with large doses (always associate tylenol with that!)
Gastric or intestinal ulceration, prolonged gestation, renal function, hepatic function, and increased bleeding time are side effects of what drugs?
Drugs that inhibit cyclooxygenase
Gastric/intestinal ulceration from NSAIDs related to inhibition of ______ production by COX-1 which is thought to protect gastric mucosa. Sometimes secondary anemia from resultant blood loss. Reduced with _____ inhibitors
Prostaglandin
COX-2 inhibitors
________ via COX-1 and -2 play a role in initiating labor
Prostaglandins
Being on a large dose of NSAIDs for years or abusing them can cause ______ problems.
kidney
What condition has been reported with sulindac, indomethacin, ibuprofen, and naproxen.
Hepatitis
Inhibition of COX 1 in the platelet prevents platelet aggregation and __ formation. This increases bleeding time.
TX (thromboxane)
Aspirin hypersensitivty is present in 3-10% of patients with what condition? Symptoms are rhinitis, urticaria, ______, and laryngeal edema
asthma asthma
Aspirin Hypersensitivity Mechanisms:
-Maybe block of COX shifts AA to lipoxygenase pathway -> _________ -> hypersensitivity
-Inhibition of COX1 results in decreased PGE2 -> increased ________ -> hypersensitivity
leukotriene production
leukotrienes
COX-2 inhibitors are better than nonselective COX inhibitors because they are less likely to cause gastric ________, don't inhibit ________ function, and are less likely to cause ______ hypersensitivity
ulceration
platelet
aspirin
COX1 in ________ is not inhibited by COX2 inhibitors. Thus, platelets continue to make thromboxane (platelet aggregator). COX2 inhibitors reduce the production of _________(inhibits platelet aggregation) by endothelial cells.
platelets
prostacyclin
_____ toxicity associated with Reye syndrome (encephalopathy and fatty liver follow viral infection with kids)
Aspirin
Anti-inflammatory doses of _____ are close to toxic doses
Aspirin
______ irreversibly inactivates platelet cyclooxygenase
Aspirin
Cyclooxygenase pathway makes ________ and ________
Lipoxygenase pathway makes ________
Both from Arachidonic Acid
Prostaglandins and Thromboxane
Leukotrienes
Limitation of the leukotriene pathway is availability of _______ _____
Arachidonic Acid
HETEs have ________ activity
chemotactic
Addition of ________ to peptidoleukotrienes (LTC4, LTD4, LTE4) which cause bronchoconstriction and asthma. "slow-reacting substance of ________"
glutathione
anaphylaxis
5-lipoxygenase (5-LO) is a _______ enzyme which is translocated to membranes by binding to the protein 5-lipoxygenase activating protein (FLAP). In cell types of myelomonocytic origin.
cytosolic
Degradation of Lipoxygenase Products:
LTA4- short half life
LTB4- oxidized by enzymes in ____ to inactive
LTE4- low potency, excreted by urine or acetylated and excreted in the _____
PMN's
bile
AA from PMN, Mast Cells, Basophils is transformed into ____ via 5-lipoxygenase
LTA4
LTA4 hydrolase transforms LTA4 into what?
LTB4
LTC4 formaiton can happen via:
1) LTC4 synthase in what kind of cells?
2) Glutathione S transferase in what kind of cells?
1) mast cell or basophil
2) endothelial cell or smooth muscle cell
LTA4 travels to other cell types where it can be converted to:
LTC4 in endothelial or _____ ____ cells
LTB4 in platelets or _____
smooth muscle
RBC
Receptor for which leukotriene is distinct from the other receptors?
LTB4
The CysLTR1 receptor is also known as the _____ receptor because it interacts preferentially with that leukotriene
LTD4
CysLTR2 is also known as the ______ receptor. It interacts with both ____ and LTD4
LTC4
LTC4
LTB4 through interaction with the LTB4 receptor can cause
- ________ of white cells
- leukocyte adhesion, enzyme release, and production of ____
- hyperalgesia or reduction of pain threshold
chemotaxis
ROS
Peptidoleukotrienes interact wit the ______ to cause:
-bronchoconstriction
-Eosinophil chemotaxis and cytokine secretion
-increased vascular perm and mucous production
-dendritic cell maturation and migration
-smooth muscle proliferation
CysLTR1
Peptidoleukotrienes interact with the _____ to cause:
-endothelial cell and macrophage activation
-fibrosis
CysLTR2
Which leukotrienes are imporant in ashtma?
Which leukotriene is found in the synovial fluid of pts with arthritis and gout?
LTC4/LTD4
LTB4
Leukotriene inhibitors are used in the treatment of what?
Bronchial asthma

Used for chronic asthma but not acute
Zileuton, Zafirlukast and Montelukast are all what type of drug?
Leukotriene Inhibitors
Zileuton inhibits 5-lipoxygenase and thus prevents synthesis of _______
leukotrienes - most importantly LTB4
Zileuton is metabolized by ________ ___ and may cause drug interactions
_________ inhibits that enzyme and may also cause significant drug interactions
cytochrome P450
Zafirlukast
Zafirlukast and Mantelukast are leukotriene receptor antagonists for what receptor?
LTD4 receptor, CysLTR1
Which leukotriene inhibitor requires monitoring for hepatic toxicity?
Zileuton
______ are synthesized extracellularly in blood or interstitial fluid, NOT in cell
Kinins
_______ and kallidin are kinins that cause:
Hypotension
Pain
Edema
Important in BP control and ______
Bradykinin
inflammation
Kallikrein inhibitors and kinin receptor antagonists being used in __ Inhibitor Deficiency (Hereditary angiodema)
C1 (complement - first step)
Activated Hageman Factor
Kallikrein
Plasmin
C1 esterase
These 4 enzymes are key in what?
Kinin, Complement, Coagulation and Fibrinolytic Pathways (which are all interrelated)
Bradykinin is just _____ with one less amino acid
Kallidin
C1 inhibitor inhibits C1 esterase, kallikrein, plasmin, Factor XIa and XIIa. Lacking in HAE. Critical event is lack of control of _____ by missing C1 inhibitor
kallikrein
If you don'ot have it, you have excessive production of kinins and swelling
Kininase II = ________ converting enzyme (ACE) or Dipeptide hydrolase
Angiotensin
Kininase I - Carboxypeptidase N or anaphylatoxin inactivator removes the carboxy terminal _______.
arginine
Bradykinin (_ aa) and Kallidin (_aa) are mediators with inflammatory activities
9
10
B1 receptor - chronic inflammation, vasodilation, pain, ________ production, _____ recruitment
cytokine
WBC
B2 receptor - Vasodilation, pain, _______ EXCRETION
sodium
Actions of Kinins via B1 and B2 receptor - Kallidin and Bradykinin are more active than when they have a terminal ____ attached
arginine
When Kallidin and Bradykinin bind the B2 receptor -
hyper or hypotension?
edema
algesic - cause pain
contract smooth muscle
release catecholamines from the ____ _____
release PGE
HYPOTENSION
adrenal medulla
When bradykinin and kallidin bind B1 receptor:
Chronic ______ effects
Induced after ______
hypotension and pain
inflammatory
trauma
Deficiency of HMW kininogen or Prekallikrein results in clotting & fibrinolytic defects with decreased ____ formation
kinin
Kallikrein inhibitors and kinin receptor antagonists are useful in __ _______ deficiency
C1 inhibitor
Immunosuppressive drugs are used to dampen the immune response in organ _________, ________ disease, and hypersensitivity.
transplantation
autoimmune
The goal of immunosuppression treatment is to avoid immune mediated ______ damage from the immune response and the ______ response
tissue
inflammatory
The immune response is more likely to be inhibited if therapy is begun when?
After exposure to the immunogen
Are primary or secondary immune responses more effectively suppressed?
Primary
Limitations of immunosuppressive therapy:
Increased risk of ________ of all types
Increased risk of ________ and related malignancies
infections
lymphomas
Major classes of immunosuppresants:
- Glucocorticoids - anti-inflammatory _____
- Cancineurin _________
-Antiproliferative/antimetabolic drugs
-Anti_____
steroids
inhibitors
bodies
Primary clinical uses of immunosuppression:
________ disease
Transplantation
________ anemia of newborn
Autoimmune
Hemolytic
Corticosteroids (21 carbons) and Androgens (19 carbons - body building) are synthesized by what?
Adrenal Cortex
The corticosteroids have 2 different types of activity
________ - carbohydrate metabolism regulating
mineralcorticoid - ________ balancing activity
Glucocorticoid
electrolyte
Corticosteroids are secreted by the adrenal gland in response to stimulation by ____. Endogenous corticosteroids have varying degrees of mineral/glucocorticoid activity
ACTH (adrenocorticotrophic hormone)
In humans, ________ (cortisol) is the main glucocorticoid and _________ is the main mineralocorticoid
hydrocortisone
aldosterone
Sodium retention is the ability of the steroid to reduce sodium excretion by the kidney in an adrenalectomized animal. Does aldosterone or cortisol exhibit this?
aldosterone (mineralcorticoid)
Cortisol is able to do liver glycogen deposition and ____ _______ activity
anti-inflammatory

Liver glycogen deposition, anti-inflammatory activity and involution of lymphoid tissue parallel one another.
The HPA axis is made up of what?
What steroid results?
Hypothalamus acting on Anterior Pituitary acting on Adrenal Cortex -> produces cortisol which is anti-inflammatory on the immune system
The hypothalamus communicates with the anterior pituitary with what hormone?
Corticotropin releasing hormone
The anterior pituitary acts on the adrenal cortex with what hormone?
What does the adrenal cortex then produce, which feeds back to turn off immune system?
ACTH
Cortisol - negative feedback on endogenous inflammation
When the immune system gets too active, we add exogenous __________ to turn it off because our endogenous can’t handle the job
We want to minimize drug action on the __________ receptor
glucocorticoids
mineralocorticoid
What is an anti-inflammatory, endogenous steroid that we have to know?
Cortisol
What do all of these drugs have in common?
Betamethasone, Dexamethasone, Methylprednisolone, Prednisone
They are all synthetic steroids used as anti-inflammatory drugs
Glucocorticoids are administered how?
Where are they metabolized?
Where are they excreted?
Administered orally, parenterally, topically
Metabolized in the liver
Excreted in the kidney
Steroids act on ________ receptors. The glucocorticoid receptor binds and promotes _______ of proteins that inhibit the immune response. Can also inhibit proteins that help the response (NF-kB and AP-1)
intracellular
transcription
Anti-inflammatory steroids cause more circulating _________, their release from bone marrow accelerated, their half time in circ increased, and blockage of their migration into inflammatory sites
Neutrophils
Anti-inflammatory steroids cause a profound, transient ________ in which the cells are not lysed, but move to extravascular compartments
lymphopenia (lack of lymphocytes)
Anti-inflammatory steroids cause increased/decreased monocytes and eosinophils in peripheral blood
decreased!
Steroids reduce the expression of COX_, inhibit release of _______ ____ from phospholipids - thus affecting PGE and LK production, inhibit degran of mast/basophils, inhibit synthesis and release of TNF, IL-_ and _, and IFN
COX2
arachidonic acid
IL-1 and IL-2
Using glucocorticoids in anti-inflammatory therapy helps by reducing inflammation BUT
the underlying cause of the disease remains
After systemic administration - ____ _____ are common and life threatening.
Also, host ______ to microbial and fungal infection is lowered
side effects
resistance
How is dosing done for steroids? Does one dose or a few days of therapy have any bad effects? Does prolonged therapy have any bad effects? What happens if you just stop taking them after prolonged treatment?
Trial and Error
Virtually no harmful effects if one day, unlikely if a few days
Prolonged therapy increases incidence of lethal effects
Risk of adrenal insufficiency with abrupt cessation of prolonged, high dose
Uses for Steroids in Nonendocrine diseases: arthritis, systemic lupus (collagen diseases), allergic diseases (not acute ______), bronchial asthma, eczema, malignancies, diseases of the ____, used to mask progression of _____ disesases
anaphylaxis
liver
ocular
Prolonged large doses of steroids administered systemically can lead to immunosuppression, peptric _______, behavioral disturbances, cataracts, osteoporosis, inihibiton of ______
ulcers
growth
Withdrawl/discontinuation of long term use of ______ can cause fever, myalgia, athralgia, malaise, death with ___tension and shock
steroids
hypotension
One major class of immunosuppressive drugs are ______ inhibitors, including cyclosporine and tacrolimus
Calcineurin
Inhibition of calcineurin activity blocks the _____________ events critical for cytokine gene expression and __ cell activation
dephosphorylation
T cell
Both cyclosporine and tacrolimus bind to cytoplasmic receptor proteins to inhibit calcineurin activity.
Cyclosporine binds to ________
Tacrolimus binds to ____
cyclophilin
FKBP (FK506 binding protein)
Cyclosporine is metabolized extensively in the ____. Potential for numerous drug interactions. Used for long term therapy for _________. BIG side effect is what?
liver
transplantation
Nephrotoxicity - happens in as many as 75% of patients
Tacrolimus is 100x more potent than _______. Its BIG side effect is what?
Also Nephrotoxicity
Antiproliferative and Antimetabolic drugs prevent the clonal expansion of _ and _ lymphocytes.
T and B lymphocytes
Sirolimus and Mycophenolate Mofetil are both what class of drugs?
Antiproliferative and Antimetabolic
Sirolimus (from Antiproliferative) and Tacrolimus (from Calcineurin inhibitors) both bind to what receptor?
FKBP receptor
Siromilus is used in combo therapy for organ transplant rejection. It binds to ____ receptor to inhibit a key enzyme in the cell cycle progression from __ to _ phase
FKBP
G1 to S phase
What drug has these side effects?
Dose dependent increase in cholesterol and triglycerides, ________ when combined with cyclosporine, increased risk of lymphomas and infections, potential drug interactions because CYP3A4 is a substrate
Sirolimus

combined with cyclosporine
Mycophenolate mofetil is used for organ _______. A metabolite inhibits inosine monophosphate dehydrogenase which is used for guanine nucleotide synthesis. Which cells are dependent on this pathway for proliferation while other cell types can use salvage pathways??
B and T cells
Toxicity from mycophenolate mofetil includes hematologic - _____penia, and gastrointestional _____ and vomiting
leukopenia
diarrhea
There is an overlap between drugs used for immunosuppression and those used for treatment of _____. Immune cells proliferate when there is an antigen while ____ cells proliferate unstimulated
Immune cell proliferation is partially synchronized while ____ cells are not.
cancer x3
What is the difference in drug administration for immunosuppression versus cancer chemotherapy
Immuno - low dose daily - continuous blockade
Chemo - intermittently in high dose pulses to kill cells and allow immune system to rebound inbetween
Antibodies have 2 functions:
Eliminate _______, done by anti-thymocyte globulin
Affect _ cell function, done by Muromonab-CD3, Daclizumab, Basiliximab
lymphocytes
T cell
Daclizumab or Basiliximab – antibodies to ___ receptor so cytokine can’t bind
IL-2
Anti-thymocyte globulin binds to thymocytes in circulation resulting in ____penia and impaired _ cell immune responses
Toxicity due to Ig being recognized as foreign resulting in sickness and ____itis
lymphopenia
T cell
nephritis
Muromonab-CD3 is used to prevent acute rejection of kidney, liver and ____ transplants. Mouse monoclonal Ab that binds to E chain of CD3 glycoprotein (part of _ ___ receptor) -> complex internalized preventing _____ recognition
heart
T-cell
antigen
______ release syndrome results from binding of muromonab CD3 to CD3 as well as crosslinking of Fc receptor. Administration of ______ prior to Muromonab reduces symptoms considerably. Working on humanized, non-Fc version
Cytokine
glucocorticoids
Daclizumab or basiliximab bind to the __-_ receptor present on activated/resting T cells and blocks what?
What unique side effect does this drug have compared to anti-thymocyte globulin?
IL-2
activated
IL-2 mediated T cell activation events
CAN have anaphylaxis