Pmcol 344 Flashcards

Midterm 1 + immunopharmacology

1
Q

GIT OTC preparations

A

laxatives
antacids
gas relievers
anti-diarrheals
hemorrhoid tx

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

GIT endocrine system

A

90% of body’s serotonin
50% of body’s dopamine
gastrin
ghrelin
cholecystokinin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

layers of GIT

A

mucosa
submucosa
muscularis
serosa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

mucosa layer

A

epithelium
lamina propria
muscularis mucosae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

submucosa layer

A

submucosal plexus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

muscularis layer

A

circular muscle
myenteric plexus
longitudinal muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

ENS

A

intrinsic neural network within walls of GIT
regulates motility, secretion, local blood flow, and immune responses
integrated with + regulated by ANS; acts independent of CNS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

submucosal plexus

A

innervates epithelia + muscle cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

myenteric plexus

A

regulates gut motility + secretion
lies between muscle layers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

ENS fibres

A

receives vagal psymp ACh = excitatory
- ↑ gut motility, dilate sphincters, ↑ secretions
receives symp NA
- ↓ gut motility = ↑ absorption, constrict sphincters

sensory nerves signaling mech. + chem. stimuli
mediates spinal + brainstem refleces regulating digestive function
provides input to central autonomic circuits that regulate feeding + illness behaviours
transmit both painful + non-painful sensation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

gut motility

A

peristalsis + segmentation
determined by smooth muscle properties and modified by chemical inputs (nerves, hormones, paracrine signals)
different regions = different types of motility due to enzymes and pH differences
- sphincters → tonic contractions
- small intestine → phasic contractions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

peristalsis

A

propulsion of food from mouth to anus (aborally)
involves coordinated contraction + relaxation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

segmentation

A

mixes food to facilitate digestion = more surface area of food for acid + enzymes to act on
mixes contents over a short length of intestine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

small intestine motility

A

rhythmic electrical activity accompanies contractions
phasic activity driven by slow waves
propagation slows with more distal segments
when slow wave reaches threshold, v.g. Ca2+ ch open → influx = APs fire + mm. contract

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

peristaltic reflex

A

triggered by mechanical stretch + mucosal stimulation
activates excitatory motor neurons above bolus (enterochromaffin cells release 5HT → stimulates sensory nn. in myenteric plexus)
activates inhibitory motor neurons below bolus = relaxes gut wall

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

gastrin

A

made in mucosal enteroendocrine cells
stimulates acid secretion by parietal cells in stomach = ↑ digestion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

5HT

A

from enterochromaffin cells
release triggered by food constituents, stretching of gut wall; endogenous mediators ex. gastrin
increases ACh release from ENS nerves

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

motilin

A

from mucosal cells
stimulates contraction of GI smooth muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

cholecystokinin

A

from small intestine enteroendocrine cells
stimulates pancreatic enzyme secretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

glucagon-like peptide 1

A

stimulates insulin release, inhibits glucagon release
promotes satiety

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

enteroendocrine cells

A

respond to nutrients, NTs, local signals to release hormones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

gastric secretions

A

human stomach secretes ~2.5 L gastric juice per day
acidity promotes proteolysis, kills pathogens, aids in iron absorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

peptic Chief cells

A

secrete pro enzymes - prorennin, pepsinogen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

parietal cells

A

secrete 150 mM HCl = pH ~ 1-2

active secretion of Cl- + K+
H+/K+ ATPase pumps K+ into cell and H+ into lumen
carbonic anhydrase generates H+ and HCO3- from H2O and CO2
HCO3- exchanged for Cl- by antiporter

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
protective mechanisms of stomach wall
mucosal cells secrete prostaglandins (PGE2 + PGI2) - mucosal cell EP4 Rs = stimulates mucus secretion + EP1/2 Rs = secretion of bicarbonate ions - ECL cell EP2/3 Rs = inhibit histamine release = ↓ acid release via H2 receptors - vascular EP2/4 Rs = vasodilation → improve blood flow to mucosal layer
26
stimulation of HCl secretion
- histamine (released basally from ECL cells) acts on H2 Rs on parietal cells = activate proton pump via Gs, cAMP - gastrin (secreted by G cells in response to nerve stimulation + stomach contents) acts on CCK2 Rs on ECL cells → ↑ Ca2+ = release of histamine - ACh (released from postG cholinergic neurons) acts on parietal cell M3 Rs to stimulate proton pump via elevation of Ca2+; inhibits somatostatin release from D cells
27
inhibition of HCl secretion
- prostaglandins inhibit histamine production, ↑ HCO3- + mucus secretion, vasodilation - tonic release of somatostatin from D cells in stomach → acts on SST2 Rs on G cells to inhibit gastrin release, ECL cells to inhibit histamine release, parietal cells to inhibit acid secretion
28
ulcer
bleeding, perforation leading to stomach contents entering body cavity and causing peritonitis fatal without intervention
29
GERD
damage to esophageal epithelium can lead to Barrett's esophagus (precancerous)
30
hiatal hernia
similar problem to GERD esophageal acid reflux
31
causes of ulcers
- stress: vagal stimulation = ↑ ACh release = ↑ HCl - COX inhibition by NSAIDs = ↓ production of protective mucosal PGs - H.pylori infection → 80-90% of gastric ulcers, 95% of duodenal ulcers
32
H.pylori infection
primary cause of ulcers corkscrew-shaped, gram (-) bacteria present in ~50% of pop → causes peptic ulcer disease in ~20% gastritis: asymptomatic inflammation of the stomach lining → ulcers → cancer class 1 carcinogen burrow into stomach mucus lining = damage allows acid + pepsin to reach epithelium releases urease → converts urea to HCO3 + ammonia = neutralize acid (forms buffer zone to protect bacteria) produces toxins that kill epithelial cells local infl: immune cells spill destructive agents on stomach lining cells
33
therapeutic goals of ulcer tx
relieve sx allow damaged tissue to heal eliminate cause
34
urea breath test
drink solution containing 13C/14C labelled urea collect exhaled breath → if CO2 is labelled, indicates H.pylori presence quick, cheap, non-invasive
35
tx for ulcers/GERD/hiatal hernia
- antibiotics - ↓ gastric acid secretion - neutralize secreted acid - protect mucosa
36
Triple Therapy
combination of proton pump inhibitor and antibiotics Omeprazole + Clarithromycin + Amoxicillin tx for 2 weeks cure if ulcer caused by H.pylori
37
proton pump inhibitors
↓ acid secretion first line therapy prodrugs → active binds covalently to cysteines in active site of H+/K+ pump inhibits both basal + stimulated acid secretion
38
H2 receptor antagonists
↓ acid secretion selective competitive orthosteric antagonists at H2 receptors on parietal cells = inhibition of histamine + gastrin stimulated acid secretion
39
antacids
neutralize acid basic inorganic salts taken orally to neutralize gastric acid + inhibit peptide enzymes chemical antagonism not absorbed mixture of magnesium + aluminum hydroxide to balance bowel fxn
40
mucosa protectors
enhance endogenous mucosal protective mechanisms / provide physical barrier over ulcer
41
nausea
feeling of impending vomiting harder to control pharmacologically
41
vomiting
emesis forceful evacuation of gastric contents through mouth physiological response to - irritating substances in hut or blood stream - excessive vestibular stimulation - psychological stimuli
42
emetic drugs
induce vomiting to prevent absorption of ingested toxic substances
43
anti-emetic drugs
useful bc vomiting reduces effectiveness of tx, can cause dehydration + nutrient depletion - mAChR antagonists - 5HT3 R antagonists - D2 R antagonists
44
Chemoreceptor trigger zone
BBB is permeable = allows circulating emetogenic mediators to act receives inputs from vestibular nuclei + directly fro GIT activates neurons in vomiting centre D2 + 5-HT3 receptors
45
vomiting centre
receives impulses from CTZ, GIT, + higher cortical centres coordinates physical act of vomiting mACh receptors
46
vestibular nuclei
inputs from labyrinth (inner ear) H1 + mACh receptors
47
diarrhea
frequent passage of liquid feces may be accompanied by abdo cramps + nausea phycl mech to rid gut of irritating substances caused by infection, anxiety, drugs, inflammatory disease primary cause of death in infants in developing countries results from disordered water + electrolyte transport in small intestine = ↑ gut motility, ↑ fluid secretion, ↓ fluid absorption = loss of electrolytes + H2O
48
intestinal fluid balance
consume >2 L of fluid daily saliva + secretions from stomach, pancreas, liver add 7L = 9L enter small intestine H2O + electrolytes are absorbed in intestine + secreted in bowel → ~1L reaches large intestine (exceeds capacity = diarrhea) only 100-200 mL of H2O excreted
49
water absorption
in small intestine (epithelial cells express ion channels, pumps, + transporters on luminal + basolateral membranes) caused by osmotic gradients created when solutes are absorbed from lumen Na+ enters cells → transported into ECF by Na+/K+ ATPase = ↑ ECF osmolarity → H2O follows passively through aquaporins
50
water secretion
in small intestine NaCl is transported from ECF into epit cells Na+ is pumped back into ECF by Na+/K+ pump; Cl- passes into lumen via CFTR channel = osmotic gradient → H2O follows passively via intercellular channels
51
secretory diarrhea
abnormal secretion of H2O + salts into small intestine impaired absorption of Na+; normal/↑ Cl- secretion = net fluid secretion → loss of H2O + salts causes dehydration may be caused by bacteria ex. cholera irreversibly activates CFTR = constant Cl- efflux
52
osmotic diarrhea
due to ingestion of poorly-absorbed, osmotically active substance ex. sorbitol, mannitol H2O moves from ECF into lumen to balance osmolarity = ↑ vol of stool → dehydration due to H2O loss looser coupling bw mvt of NaCl + H2O (H2O loss > NaCl loss = hypernatremia)
53
tx of diarrhea
- oral rehydration (H2O + electrolytes; add glucose to enhance absorption) - anti-infective agents (if bacterial infection) - spasmolytics - adsorbents
54
bacterial infections causing diarrhea
E.coli salmonella shigella campylobacter = treat with anti-biotics
55
spasmolytics
tx of diarrhea: relaxation of gut smooth muscle = ↓ gut motility = ↑ transit time = ↑ time for reabsorption of water - muscarinic antagonists - opioid receptor agonists
56
muscarinic antagonists (tx of diarrhea)
block action of ACh (primary stimulatory NT in gut = contraction, propulsion) = relax gut muscle → ↓ motility side effects significant (widespread action) = rarely used
57
opioid receptor agonists (tx of diarrhea)
activate opiate receptors = affect motility, secretion, + transport of electrolytes + fluid primary site of action: ENS inhibitory preS receptors = ↓ ACh release = ↑ segmentation + ↓ peristalsis ↑ transit time so water absorption ↑ symptomatic relief
58
adsorbents (tx of diarrhea)
bind up H2O in gut = ↓ mvt symptomatic relief (does not ↓ dehydration; efficacy unclear) ex. kaolin, pectin, chalk, charcoal, methylcellulose potentially adsorb pathogens/toxins; coat + protect mucosa
59
constipation
slowed passage of food through GIT 50+% of people affected causes: drugs (opiates, antidepressants, iron supplements); diet, hydration state, exercise; hormones, IBS
60
purgatives
accelerate passage of food through intestine + evacuation of bowels bulk laxatives, osmotic laxatives, stimulating purgatives, stool softeners used to relieve constipation or to clear bowel prior to surgery/examination prolonged use can lead to dependence
61
agents that ↑ GIT motility without purgation
cholinomimetics D2 antagonists 5HT-4 agonists
62
bulk laxatives
ex. methyl cellulose, ispaghula husk polysaccharide polymers that are not broken down by digestion in upper part of GIT form bulky hydrated mass in gut lumen mechanical distension promotes peristaltic reflex slow action but no absorption = no unwanted effects
63
osmotic laxatives
ex. lactulose poorly absorbed = produce osmotic load in lumen → draws water into gut lumen softens stool + stretches gut to promote peristalsis + defecation takes 2-3 days to act but not absorbed = no unwanted effects
64
stimulant laxatives
↑ electrolyte + water secretion / ↑ peristalsis ex. bisacodyl, senna
65
cholinomimetics (tx of constipation)
muscarinic agonists + cholinesterase inhibitors enhance GI contraction but uncoordinated → little/no net propulsive activity
66
dopamine in GIT
GIT produces 50% of body's DA see changes in GI fxn in Parkinson's DA modulates gut motility by mediating neg feedback mechanism → released at same time as ACh during peristalsis to cause muscle relaxation + inhibition of ACh release D2 receptors on preS ACh termines + postS muscle
67
D2 antagonists (tx of constipation)
block effects of endogenous dopamine (no effect on ACh = normal release) inhibit smooth muscle relaxation and enhance ACh release = ↑ contractility + peristalsis actions are negated by anticholinergic drugs (little effect if muscarinic Rs are blocked) → preS D2R effect is more important than postS
68
5HT-4 agonists
tx of constipation preS 5HT-4 Rs in myenteric plexus = ↑ ACh release
69
Irritable bowel syndrome (IBS)
unknown cause, possible: alterations in 5HT reuptake, bacterial infection, hormones not inflammatory bowel disease alterations in GIT motility constipation, diarrhea, mixed, unspecified
70
IBS tx
symptomatic - diet - stress management - laxatives/anti-diarrheals (PG analogues)
71
Ulcerative colitis
inflammatory disorder causing ulcers in lining of rectum + colon inflammation → cell death = ulcer infl in colon = frequent emptying → diarrhea associated w ↑ risk of cancer
72
Crohn's disease
inflammatory disorder infl deeper in intestinal wall; not exclusive to colon → along entire GIT associated w ↑ risk of cancer
73
tx for UC / Crohn's
long term tx with anti-inflammatory / immunosuppressant drugs - anti-diarrheals (treat sx) - glucocorticoids = anti-infl + immunosuppressants - aminosalicylates - biopharmaceuticals (humanized antibodies)
74
hemostasis
dynamic process vascular injury platelet adhesion platelet aggregation fibrin formation (coagulation) fibrinolysis vascular repair
75
thrombosis
pathological formation of hemostatic plug often occurs in absence of bleeding = clot formation
76
white clots
arterial (rapid blood flow) → heart + brain composed of platelets, some fibrin main risk factor = ruptured atherosclerotic plaque clinical presentation = MI, cerebrovascular stroke tx: anti-platelet drugs
77
red clots
venous (relative stasis) → veins, heart chambers composed of fibrin + RBCs, few platelets main risk factor = slow/disturbed blood flow; hypercoagulable state clinical presentation = venous thromboembolism, cardioembolic stroke tx: anticoagulant drugs
78
prevention of blood clots
antiplatelet drugs anticoagulant drugs
79
treatment of blood clots
thrombolytic/fibrinolytic drugs
80
anti-platelet drugs
ASA thienopyridines non-thienopyridine P2Y12 antagonists Dipyridamole GP IIb/IIIa R antagonists prevent platelets from aggregating = prevention of adverse CV events or stroke inhibit platelet aggregation by targeting platelet pathways
81
platelet signaling
platelet activation → release ADP + TXA2 → activate GP IIb/IIIa + fibrinogen binding → aggregation
82
ASA
COX inhibition = prevents TXA2 synthesis
83
Thienopyridines
orally active prodrugs requiring hepatic conversion to active metabolites irreversible non-competitive antagonists of ADP binding to P2Y12 receptors = stop aggregation + secretion of infl. mediators ticlopidine, clopidogrel, prasugrel
83
Non-thienopyridine P2Y12 antagonist
Ticagrelor same target as thienopyridines but different mechanism (reversible allosteric antagonist)
84
Dual Anti-Platelet Therapy
ASA combined with P2Y12 inhibitors (usually Clopidogrel, or Ticagrelor) up to 1 year following MI, 1 month following cerebrovasc stroke
85
Dipyridamole
inhibits phosphodiesterase = prevent breakdown of cAMP/cGMP enhanced inhibition of platelet aggregation
86
glycoprotein IIb/IIIa antagonists
block receptor for fibrinogen or compete for occupancy inhibit final common step leading to platelet aggregation IV administration - prevent thrombosis in pts with acute coronary syndromes or undergoing coronary angioplasty unwanted effects: bleeding, thrombocytopenia abciximab, eptifibatide, tirofiban
87
blood coagulation
secondary hemostasis clotting - blood converted into solid gel (platelets, RBCs, plasma) occurs around platelets, reinforces plug vital hemostatic defense mechanism
88
anticoagulant drugs
warfarin heparins direct oral anticoagulants hirudins
89
Virchow's triad
- circulatory stasis - hypercoagulable state - epithelial cell injury high risk for clots = need for anticoagulation
90
example of circulatory stasis
atrial fibrillation immobilization deep vein thrombosis
91
example of hypercoagulable state
malignancy deficiencies in anti-clotting factors pregnancy
92
example of epithelial cell injury
trauma fractures surgery heart valve replacement
93
coagulation
a proteolytic cascade involving serine proteases complex of Va with negative phospholipids on aggregating platelets provides binding site for Xa + thrombin binding requires Ca2+ + carboxyglutamic acid residues
94
warfarin
vitamin K reductase antagonist enzyme needed to carboxylate factors II, VII, IX, X
95
problems with warfarin
rapid peak plasma concentration after ingestion but delayed pharmacological effect (requires depletion of clotting factors) wide individual variation teratogenic need to monitor clotting time (INR) many drug interactions ↑ bleeding risk - narrow TI unwanted effects: bleeding, liver damage
96
heparins
first extracted from liver IV or subq admin neg charged sulfated glycosaminoglycands rapid onset of action used to bridge warfarin therapy unwanted effects: bleeding, heparin-induced thrombocytopenia/thrombosis mech of action: bind ATIII via unique pentasaccharide sequence induce conformational change in ATIII = ↑ affinity for active site of factors IIa, Xa unfractionated + low molecular weight
97
direct oral anticoagulants
rivaroxaban dabigatran etexilate (prodrug but metabolised rapidly in plasma)
98
hirudins
specific thrombin inhibitor
99
thrombolytic/fibrinolytic drugs
recombinant tPA streptokinase
100
most common cancers
in males: prostate, colorectal in females: breast, colorectal
101
cancer
group of 100+ diseases that develop across time can develop in any of body's tissues both hereditary and environmental factors contribute to its development develops due to loss of growth control in cells as a result of mutations in genes involved in cell cycle control
102
hereditary mutations
inherited/germ-line mutations gene changes that come from a parent and exist in all cells of the body 5-10% of cancers
103
acquired mutations
sporadic/somatic mutations environmental influences such as exposure to radiation or toxins, not passed on to offspring
104
multiple-hit hypothesis
Knudson hypothesis normally, the body can compensate for one mutation → second mutation is the cause of effects if hereditary: only one additional mutation is required
105
proto-oncogenes
genes that normally control how often cell divides and the degree to which it differentiates
106
oncogenes
mutated proto-oncogenes = cause normal cells to grow out of control and become cancer cells permanently turned on or activated = continuous division
107
classes of oncogenes
growth factors growth factor receptors signal transducers transcription factors / nuclear transducers programmed cell death regulators
108
tumor suppressor genes
normal genes that slow down cell division, repair DNA mistakes, and tell cells when to die ~30: p53, BRCA1, BRCA2, APC, RB1 genes that control cell division (RB1) genes that repair DNA (HNPCC) cell "suicide" genes (p53)
109
cancer development
cell with genetic mutation → uncontrolled proliferation = hyperplasia → loss of function = dysplasia → dedifferentiation = in situ cancer → invasiveness = invasive cancer → metastasis
110
invasive cancer - other mutations
- production of metalloproteinases to break down extracellular matrix - telomerase expression = ↑ cell division - factors to cause growth of local blood vessels
111
tumor directed angiogenesis
triggered when solid tumors exceed 2 mm3 and become hypoxic secrete factors to attract blood vessel growth = transition from hyperplasia to neoplasia
112
fundamental issues of chemotherapy
1. cancer is often far advanced by the time of detection 2. they are your own cells 3. cannot leave any cancer cells behind after treatment ends 4. most cancer chemotherapy drugs target proliferating cells 5. chemo drugs target rapidly dividing cells 6. solid tumor heterogeneity 7. development of drug resistance
113
1. cancer is often far advanced by the time of detection
~30 cell doublings = cell mass with a diameter of 2 cm → within limits of diagnostics but might be unnoticed another 10 doublings would be ~20 cm = lethal tumor mass neoplasm is silent for first 3/4+ of its existence
114
2. they are your own cells
there can be little reliance on the host immunological defense mechanisms in ridding the body of the cancer cells anything that kills cancer cells is likely to also affect normal cells
115
3. cannot leave any cancer cells behind after treatment ends
a given therapeutic dose of a cytotoxic drug destroys a constant fraction of the malignant cells a dose that kills 99.99% of cells used to treat a tumor with 10^11 cells will still leave 10 million viable malignant cells
116
4. most cancer chemotherapy drugs target proliferating cells
cells need to be actively proliferating for effective tx most sensitive in S phase
117
5. chemo drugs target rapidly dividing cells
any rapidly dividing cell in the body will be impacted by the agents - bone marrow toxicity - impaired wound healing - hair loss - GI epithelium damage - depression of growth in children - sterility - teratogenicity
118
6. solid tumor heterogeneity
3 layers: A = outside, C = inner mass A - dividing cells, continuously in cell cycle B - resting cells = G0 C - cells are no longer able to divide, contribute to tumor vol
119
7. development of drug resistance
1. ↓ drug uptake due to ↓ expression of transporter 2. ↑ drug efflux due to upreg of efflux pumps 3. change in drug metabolism (↓ activation of prodrug) 4. ↑ DNA damage repair 5. ↑ resistance to apoptosis 6. change in drug target or target levels
120
cell cycle
G0: rest G1: growth S: synthesis, replication of DNA G2: growth M: division
121
primary drug resistance
present when the drug is first given
122
acquired drug resistance
result from either adaptation of the tumor cells or mutation i. mutant is not affected by tx → continues to grow ii. persister cells after tx have innate resistance → more sensitive to mutations after tx
123
alkylating agents
originated from mustard gas and nitrogen mustard alkylation: transfer of an alkyl group from one molecule to another form highly reactive, positively charged (electrophilic) intermediates alkyl groups bind covalently to DNA bifunctional = cross link DNA *myelosuppression nitrogen mustards, nitrosoureas, cisplatin
124
myelosuppression
↓ production of RBCs, WBCs, platelets
125
nitrogen mustards
cannot cross BBB only targets cells in S phase substitutions alter PK → different drug distribution *myelosuppression, immunosuppression, hemorrhagic cystitis mechlorethamine, chlorambucil, melphalan cyclophosphamide
126
CMF or IMF
treatment of breast cancer after surgery cyclophosphamide/ifosfamide, methotrexate, fluorouracil
127
nitrosoureas
require activation in the liver to reactive intermediates lipid soluble = can penetrate CNS → brain tumors *myelosuppression, leukomogenic, hepatotoxicity carmustine, lomustine
128
alkyl sulfonate
forms G-G and G-A intra-strand crosslinks selective effects on blood forming cells (myelosuppression) used in high doses for bone marrow transplantation busulfan
129
triazines
used for glioblastomas + melanomas N-7 or O-6 positions of G dacarbazine, temozolomide
130
ethylenimines
lymphoma + cancers of breast, ovary, bladder used to treat malignant effusions thiotepa
131
resistance to alkylating agents
↓ transport across cell membrane ↑ [thiol]i (↓ efficacy of alk. agents) ↑ enz detoxification of reactive intermediates ↑ DNA repair
132
MGMT
DNA repair enzyme if MGMT promoter region is methylated, cells no longer produce MGMT = more responsive to alkylating agents methylation of MGMT promoter in gliomas = predictor of responsiveness to triazines
133
platinum complexes
bifunctional passive diffusion or copper transporter to enter cell hydrolyzed due to ↓ [Cl-]i = trapped reacts with DNA to form intrastrand cross links = stops replication
134
cisplatin
cell cycle non-specific renal toxicity due to generation of ROS in kidney by metabolism nausea + vomiting resistance: ↓ uptake + retention, ↑ DNA repair, ↑ production of cellular thiols
135
PEB
cisplatin, etoposide, bleomycin treatment of testicular cancer: >90% cures
136
antimetabolites
mimic natural metabolites interfere with biosynthetic pathways inhibit critical steps in nucleic acid synthesis cell cycle specific = S phase toxicity due to effects on proliferating cells: myelosuppression, GI toxicity, hair loss
137
why are antimetabolites less carcinogenic than alkylating agents
administration continuous IV infusion = constant level → lower peak concentrations
138
folate cycle
formation of thymidine nucleotide DHF (FH2) → THF (FH4) → DHF = dUMP → dTMP → dTTP interfere with cycle = alter ratio of nucleotides → affect DNA synthesis inside cells, folate + MTX converted to polyglutamates
139
osteogenic sarcoma
HDMTX + leucovorin rescue * oral + GI ulceration, myelosuppression, pulmonary infiltrates/fibrosis
140
selectivity of leucovorin rescue
tumor cells upregulate efflux pumps = remove leucovorin from cells accumulates in normal cells = conversion of FH4
141
mechanisms of resistance to MTX
↓ uptake via folate transporters ↑ MTX efflux altered target enzymes ↓ polyglutamation ↑ thymidine uptake (competes w effect of MTX)
142
pyrimidine analogues
antimetabolites that mimic structure of metabolic pyrimidines interfere with enzymatic interaction
143
suicide substrate
compound that binds enzyme + triggers conformational change to intermediate state = permanent block of enzymatic activity FdUMP → inhibits TS
144
specific thymidylate synthase inhibitors
Raltitrexed: transported into cells via folate transporters, fully active after polyglutamation potent inhibitor of TS (100x higher than 5-FU)
145
cytadine analogues
pyrimidine nucleosides → modification of ribose Cytarabine + Gemcitabine prodrugs → requires phosphorylation to activate incorporation into DNA = inhibit polymerization
146
resistance to araC
↓ uptake via nucleoside transporters ↑ activity of cytidine deaminase ↓ activity of dCK change in activity of DNA polymerase = less affected
147
dFdC action
phosphorylated to dfdCDP → irreversible inhibition of ribonucleotide reducase dFdCTP is incorporated into DNA → inhibits DNA polymerase
148
metabolism of thiopurines
6-MP enters cell through ENBT1 either inactivated or converted: - inhibition of de novo purine synthesis (TPMT) - incorporation into DNA → apoptosis 6-TG → incorporated into DNA = apoptosis
149
TPMT
thiopurine methyltransferase pharmacogenomics = high individual variability → ↓ or absent enz activity (5%: normal dose is toxic) ↑ TGN metabolites = higher myelosuppression screen prior to admin of 6-MP
150
DNA topoisomerases
unwind helix for DNA replication + RNA transcription present at elevated levels in tumors
151
type I topoisomerase
change degree of supercoiling of DNA ss breaks + religation topI
152
type II topoisomerase
ds breaks topIIa, topIIb
153
topoisomerase inhibitors
1. inhibit religation step = locks enzyme in cleavage complex (↑ rate of cleavage) 2. competitive inhibition of ATP binding site in type II - prevents ATP-hydrolysis to drive enz action 3. inhibition of DNA topoisomerase binding 4. inhibit ATP hydrolysis and DNA release at last step
154
FOLFIRI
5-FU, leucovorin, Irinotecan tx of colon cancer
155
irinotecan (CPT-11) pharmacogenomics
SN-38 inactivated by UGT1A1 - 10% of caucasians have ↓ fxn variant = poor metabolism, toxicity - ↑ expression = resistance CPT-11 inactivated by CYP3A4 - ↑ expression = resistance
156
anthracyclins
inhibition of topII = DNA strand breakage → apoptosis intercalative binding to DNA, partial unwinding *free radical formation by generation of OH and O2- → cell membrane damage, large CV effects cell cycle non specific
157
toxicity of anthracyclins
myelosuppression GI irritation cardiac toxicity - delayed = related to cumulative lifetime dose
158
resistance to anthracyclins
1. ↑ DNA repair 2. ↑ efflux 3. inactivation (type II metabolism) 4. ↓ apoptosis (↓ apoptotic proteins + ↑ antiapoptotic proteins)
159
podophyllotoxin derivatives
semi-synthetic glycoside derivatives non-intercalating inhibitor of topII forms stabilized complex with topII-DNA single + double strand breaks late S/early G2
160
etoposide resistance
↑ efflux ↓ binding to topII ↑ glutathione conjugation
161
microtubules
play a role in cellular structural support (component of cytoskeleton) important roles in formation + function of mitotic spindles, axonal transport of organelles dynamic structure: add and remove subunits
162
antimicrotubule agents
interfere with formation and function of microtubules stabilizers = blocks dissolution (-) destabilizers = blocks assembly (+) side effects: nausea + abdominal discomfort
163
vinca alkaloids
bind to tubulin at the end of microtubules and to tubulin dimers destabilizers = dissolution of mitotic spindles (no cell division), inhibit organelle transport cell cycle specific: G1/S phase
164
vinca alkaloid resistance
↑ efflux through ABC transporters
165
taxanes
stabilizers: bind to tubulin = enhance polymerization = discrete bundles of stable microtubules, inhibit cell replication cell cycle specific: G2/M phase
166
hormonal therapy
tx that adds, blocks, or removes hormones tissues like breast, prostate, endometrial rely on hormones (androgens + estrogens) for proliferation non cytotoxic drugs → slow down cancer growth, used in combination with other chemo effects mediated through ER and PR
167
targeting ER action in breast cancer
i. aromatase inhibitors = block ligand ii. antiestrogens, SERMs = block R activation iii. small molecule inhibitors = block R interaction with DNA iv. peptides + small molecules block coactivators
168
Tamoxifen + SSRIs
SSRIs used to treat hot flashes (side effect of tamoxifen) anti-CYP2D6 activity = drug competition → ↓ tamoxifen efficacy
169
hormonal therapy for prostate cancer
DHT modulates prostate growth → binds to cytoplasmic androgen Rs → activation, transport to nucleus → binds HRE in gene promoters 80% is AR+ = androgen-sensitive
170
androgen withdrawal therapies
↓ androgen production - bilateral orechidectomy - medical castration - GnRH antagonists - androgen synthesis inhibitors androgen R antagonism
171
GnRH antagonist
↓ activity of system rapid castration (96h) no flare 8.9% PSA failure 40% local injection reaction monthly administration
172
GnRH agonist
↓ activity of system slow castration (3-4 weeks) flare → stim of system = worsens first 14.1% PSA failure 1% local injection reaction administration every 3 months
173
molecularly targeted therapeutics
interfere with specific molecular target that plays role in tumor growth or progression more selective than previous therapies = ↓ side effects
174
epidermal growth factor receptors
(receptor tyrosine kinases) activation enhances proliferative capabilities of cancer cells: - self sufficiency in growth signals - insensitivity to anti-growth signals - evasion from apoptosis - limitless replication potential - sustained angiogenesis - tissue invasion and metastasis
175
inhibition of RTK
1. block ligand binding to receptor 2. block receptor dimerization 3. induce receptor endocytosis and degradation via ubiquitination 4. block tyrosine kinase activity
176
HER2 receptors
EGRF family of RTKs amplification of gene = ↑ proliferative capacity ↑ HER2 = poor prognosis target for anti-cancer therapeutics: - MCAB against extracellular domain - TK inhibitors of intracellular enz activity of R
177
problems with Ab approach to HER2 R
dose limiting systemic toxicities (some EGFR is necessary for normal fxn) skin rashes + diarrhea high molecular weight = slow + incomplete distribution requires IV elicits immune response targeting one specific R = too specific
178
tyrosine kinase inhibitors
solution to over-specificity block phosphorylation step
179
chronic myeloid leukemia
Philadelphia chromosome: chromosomal translocation = genetic recombination to combine BCR-ABL tyrosine kinase activates mediators of cell cycle regulation = growth factor is constitutively active → high level of proliferation
180
imatinib resistance
BCR-ABL overexpression BCR-ABL kinase mutation (↓ binding affinity) BCR-ABL independent mechanisms
181
TKI vs Abs
TKI - orally admin - low MW = rapid distribution - broader spectrum of kinases (cross react) - lack immune responses - severe side effects Abs - IV admin - specific - downregulate Rs = dual activity
182
angiogenesis inhibitors
prevent metastasis monoclonal Abs against vascular endothelial growth factor A and R = starves tumor cell of oxygen/nutrients
183
proteasomes
degradation of unneeded or damaged proteins tag protein by linkage to ubiquitin critical for cell cycle inhibition action blocked by proteasome inhibitors
184
immunopharmacology
1. drugs acting on the immune system 2. pharmacological actions of substances derived from the immune system
185
immune system
collection of cells, tissues, and molecules that protect the body from pathogens, toxins, and cancerous cells
186
innate immunity
rapid provides immediate protection against infection first line of defense primitive, does not require priming initiated by engagement of PRRs → nonspecific to the invading pathogen soluble factors + cellular components
187
adaptive immunity
slow characterized by antigenic specificity and memory consists of T lymphocytes, B cells, and antibodies
188
innate immune system
physical barriers: mucosal epithelia and skin → produce mucins and antimicrobial peptides phagocytes (neutrophils + macrophages) NK cells eosinophils, basophils, mast cells complement system
189
dendritic cells
traffic cops of innate immune system → surveil for infection present at all epithelial barriers use PRRs to monitor environment for pathogens direct T cells and B cells to their targets transition between innate and adaptive immunity
190
macrophages
effector cells of the innate immune system that phagocytose bacteria and secrete both pro-inflammatory and antimicrobial mediators
191
mast cells and basophils
produce histamine that helps immune system attack allergens degranulation + cytokines
192
neutrophils
short-lived cells first line of defense against infection NETs, cytokines, ROS
193
inflammation
splinter breaches the physical barrier of skin + transfers bacteria into wound damaged cells release chemical signals = ↑ blood flow → redness, heat, swelling, + recruitment of innate immune cells inflammation: direct effect of accumulation of immune cells + fluid at site of injury macrophages + neutrophils dendritic cells might carry pathogen antigens to activate acquired immunity
194
complement system
variety of secreted enzymes and proteins that function in the circulation and tissue fluids; activated in cascading fashion chemotaxis, opsonization, bacteria lysis (MAC)
195
chemotaxis
recruit phagocytes through complement components with chemotactic activity
196
opsonization
C3 fragments attach to pathogens + enhance recognization of bacteria to macrophages
197
membrane-attack complex
complement proteins attach to wall of bacteria + form pore → release of water, salt, proteins = direct killing of pathogens → bacteria lysis
198
PAMPs
pathogen-associated molecular patterns products essential characteristic of microbes (absent in mammalian tissues) → allows distinction between self and non self indistinguishable between microorganisms of a given class ex. lipopolysaccharide, nucleic acids, oligosaccharides
199
DAMPs
damage-associated molecular patterns host or environmental molecules that can initiate an inflammatory response; produced or released by damaged/dying cells promote sterile inflammation can lead to inflammatory disease recognized by PRRs and non-PRR DAMP receptors
200
PRRs
pattern recognition receptors proteins capable of recognizing PAMPs or DAMPs ex. TLR (toll-like receptors)
201
transition from innate to adaptive immunity
establish an association between PAMPs and DAMPs recognized by PRRs and the antigens recognized by lymphocytes mature dendritic cells migrate to lymph nodes degradation of pathogen antigen + loaded into MHC = antigen presentation complex
202
MHCs
major histocompatibility complex trans membrane proteins with extracellular domains that bind peptide fragments from proteins which were degraded in the cell
203
MHC class 1
bind peptides derived from intracellular proteins ex. virus or tumor antigen expressed on various cell types including epithelial cells recognized by cytotoxic CD8+ T cells
204
MHC class 2
bind peptides derived from endocytosed extracellular proteins expressed only on professional APCs recognized by CD4+ T cells
205
lymphocytes
cells of adaptive immunity T cells + B cells each group has specific surface markers, location in lymphoid organs, and function
206
CD3
T cell antigen part of TCR complex involved in antigen recognition, cell signaling, and proliferation signal transduction
207
CD4
T cell antigen functions as a co-receptor of the TCR in T helper and T reg cells active when antigen is presented by MHC class II participates in HIV infection
208
CD28
T cell antigen most effective co-stimulatory molecule → necessary for synthesis and secretion of IL2 expressed by naive and activated T cells interacts with B7 expressed by mature APCs
209
CTLA-4
T cell antigen inhibitory molecule homologous to CD28 expressed only on activated T cells binds to same ligand as CD28 but with higher affinity
210
CD8+ cells
two subsets 1. short lived cytotoxic T lymphocytes: rapidly die after pathogen clearance migrate to peripheral sites of infection, induce cytotoxicity against infected target cells, produce effector cytokines like IFN-y and TNF-a 2. memory precursor effector cells: mediate long term protection against secondary challenge
211
collaboration between CD4+ and CD8+ T cells
recognition of specific antigens activates CD4+ and CD8+ T cells CD4+ cells produce cytokines that help CD8+ cells to differentiate into CTLs → kill target cells by cytotoxic activity in an antigen-specific and cell contact-dependent manner
212