midterm 1 Flashcards
functions of clinical laboratory
diagnosis or confirm diagnosis of infectious disease
guidance of treatment
outbreak detection
support for infection control
collect and collate data/info: trends in resistance, antimicrobial susceptibility summaries
epidemiological studies
what do clinical microbiologists provide advice on?
appropriate specimen collection and transport
interpret test results
patient management - recommendations on antimicrobials
why is it important to understand how the clinical lab works?
need to give the lab the appropriate specimens
decrease frustration
cost effective use - choose test wisely
want physicians to provide clinically relevant information such as allergies or the type of wound/body part/symptoms the sample was collected from
general workflow of lab
collection transport/storage accessioning processing interpretation/identification susceptibility testing/ molecular testing reporting/ documentation
requisition form
must have physicians name and contact info
patients name, birthday, unique identifier such as health card number or passport number
specimen type
method of collection - how it is collected determines how it is processed
time/date of collection
required analysis
relevant clinical info - allergies
things to consider for specimen collection
- will the specimen provide useful information - if you’re not prepared to change the patients management based on the results then don’t do it
- choice of the actual specimen to take
- instructions for collection by patient if required
- transport time to lab - may need transport media
- quality of the specimen
- risk of false positives/negatives
- specimens that required being cultured are never put in formalin
Accessioning of the sample
process of identifying the specimen
is the specimen properly labeled? does the requisition info match the specimen label?
was it properly collected
time/date of collection
is it being processed or rejected
is it STAT/life threatening
received specimens must be coded and entered into the LIS system
criteria for specimen rejection
missing information
info on specimen does not match requisition
specimen is too old- improper transport
swab or media is expired
inappropriate specimen
poor quality specimen
specimen is leaking
duplicate specimens
cant be from physician or their family member
things to consider for transport of specimens
transport time to the lab
type of specimen (swab, urine, blood, stool, fluids, scrapings)
transport media: maintain viability but inhibit growth, Cary Blair Transport Media, SAF (parasites)
incubation/refrigeration/storage
processing of specimens
types of specimen: swab, fluid, stool, blood
what tests are required: culture, molecular point of care (POC), serology, microscope (gram stain, AFB stain)
additional processing such ad decontamination or centrifugation if required
what media
incubation conditions: CO2. low O2 high O2, 37 degrees, 42, etc.
why are molecular diagnostics not great for bacterial identification?
good or viral diagnosis but huge cost for bacterial diagnosis and not super effective because the culture is often more sensitive than the actual PCR itself
molecular diagnostics also does not provide susceptibility information
sputum/specimen grading
Q0- very poor quality: oropharyngeal contamination determined via microscope
Q1- poor quality: oropharyngeal contamination but specimen is still processed - results to be interpreted with caution
Q2- good quality
Q3- very good quality
interpretation/identification and turn around times
microscopy: 30mins-same day
Point of Care (POC): rapid streptococcal antigen test in an hour or less
direct MALDI-TOF: same day
Culture: 24hr to 3 weeks
Serological: same day/ week
molecular: same next day for diagnosis but same/next week for epidemiological studies
susceptibility testing: 24-72 hours or longer for mycobacterium
antimicrobial chemotherapy
use of drugs to combat infectious agents including antivirals, antibiotics, antifungals, and antiparasitic
most are derived from naturally occurring compounds some may be semi-synthetic or synthetic
differential toxicity
drug is more toxic to the infecting organism than to the host
spectrum of activity
broad vs narrow
broad kills a lot of different organisms
narrow kills a select group - try to use these if possible
minimum inhibitory concentration (MIC)
minimum concentration of the antibiotic required to inhibit the growth of the organism
minimum bactericidal concentration (MBC)
minimum concentration required to kill the organism
bacteriostatic vs bactericidal drugs
bacteriostatic drugs inhibit the organism - MBC is higher than MIC
bactericidal drugs - kill, MIC and MBC are the same
time dependent killing vs concentration dependent killing
time dependent killing: goal is to maximize exposure of the drug to the bacteria - dont care how high the concentration is just want to maintain the MIC for as long as possible - dosed more frequently bc want to keep it stable
Concentration dependent killing: goal is to maximize the concentration of the drug - only need to dose one or two times per day
prophylaxis
antimicrobial agents are given prevent an infection - do this before a surgery for example
treatment
antimicrobial agents are administered to treat an existing infection
therapeutic index
+ examples of drugs with low therapeutic index
therapeutic dose/ effective dose
drugs with a low therapeutic dose may require therapeutic drug monitoring to ensure drug levels are both effective at treating the infection and not killing the patient
examples:
aminoglyosides
vancomycin
the ideal antibiotic
no/low toxicity to the host
low probability of having resistance mechanisms
does not induce hypersensitivities in the host eg penicillin
rapid and extensive distribution to the tissues
relatively long half life but not too long
free of interactions with other drugs
convient for administration
cheap
empiric therapy
some infectious disease require immediate treatment - need to prescribe a drug before getting test results back
make a prescription based on:
- epidemiology (most probable diseases and etiologies)
- severity of the disease
- local rates of resistance
once the organism is identified the treatment should be adjusted - narrowed is possible or a new drug given if the first guess was wrong
advantages and disadvantages of combination therapy
advantages:
- treating polymicrobial infections
- initial empiric treatment
- synergy - 1 +1 = 4
- may prevent the emergence of resistance (this is the case with TB)
disadvantages:
- may be antagonistic - 2+2=1
- cost
- increased risk of side effects and drug-drug interactions
- usually not required for maximum efficacy
what influences you choice of antibiotics
activity against isolated or suspected organism
acute vs chronic disease
antibiotic history of the patient - can’t give the same one more than once in 3 months
site of infection
mode of administration/toxicity/cost
metabolism and excretion
duration of treatment / frequency of dose
local rates of resistance
concomitant medications that may react with antibiotic
re-infection vs reoccurrence
reinfection = same infection by different organism
reoccurrence = same infection by same organism - could indicate drug resistance
antimicrobial resistance
resistance = the inability to kill or inhibit the organism with clinically achievable drug concentrations
resistance can be innate such as gram negatives are resistant to vancomycin because it only works on gram positives - don’t really care about this stuff
resistance may be acquired through: mutation and acquisition of DNA
this results in: up/down regulation of things such as efflux pumps or OMPs, expanded spectrum of enzymatic activity (beta lactamases), and target site modification
antimicrobial selection of resistance
the use of antibiotics doesn’t create resistance it selects for it
have a colony of bacteria (billions of mutations happening in the genome) and one of them mutates to modify something in the cell that will resist the effect of antibiotics
the treatment with the antibiotics selects for that mutation
then have a resistant population develop
mechanisms of resistance gene transfer
transduction
transformation
conjugation
causes for the spread of antimicrobial resistance pathogens
global travel - can travel the world in 12 hours
don’t know you’re infected until you get there and then arrive with a new bacteria
factors that accelerate the development of resistance
main reason: overuse and misuse of antibiotics
other reasons:
- inadequate levels of antibiotics at the site of infection
- duration of treatment too short/long
- overwhelming numbers or organisms
- poor quality counterfeits
- over the counter/ no prescription needed
- animal husbandry
- frequent exposure to the same class
inappropriate use of antibiotics
63% of adults with upper respiratory tract infections received antibiotics even though most of them just had a cold
how you use antibiotics in your own health care system
france over presrcibed antibiotics and resulted in more resistant bacteria strains in the country
germany did not over prescribe and resulted in much less resistance
what happened when fluroquinolones were used more than once in 3 months
the chances of being infected with resistant S. pneumoniae increased dramatically compared to the chances when using other drugs
implications of resistance
treatment failure - patient death mortality rate of 42% with resistant strain infection
forced to use more expensive or more toxic alternatives
longer hospital stays = increased health care cost
possibility of not alternative agents - much less drugs are being made
impact of lacking drug development and antibiotic resistance
too expensive to make antibiotics now because there is not a wide enough market to make the money back suing the patent given the time it takes to go through the trials
the threat of antimicrobial resistance
carbapenemase resistance enterobacteriaceae (CRE)
kills up to half of the people it infects - resulted from the colistan resistance gene being passed on through china feeding their animals antibiotics
antimicrobial stewardship
limiting the use of antibiotics to those patients who absolutely require them:
- right patient
- right drug
- right time
- right dose
- right route
cellular targets of antibiotics
cell wall synthesis
nucleic acid synthesis
protein synthesis
cell membrane
general mechanism of resistance
altered permeability - OMP alteration in gram negatives inactivation/destruction of antibiotic novel binding sites efflux mechanisms bypass of metabolic pathways
cell wall synthesis inhibitors
beta lactams
glycopeptides
fosfomycin
beta lactam antibiotics
penicillins cephalosporins: as you go up generations you get more gram negative activity - 1st gen - 2nd gen - 3rd gen - 4th gen - 5th gen carbapenem
structure/mechanism of beta lactam drugs
have a beta-lactam ring that is a substrate analogue of D-Ala-D-Ala
therefore the target of beta-lactams are transpeptidases (penicillin binding proteins) that are involved in the crosslinking of the PG
beta lactams work through competitive inhibition
beta lactam resistance
beta lactamases (most common method) - inactivate the drug by opening up the beta lactam ring
altered PBP (S. pneumoniae)
novel PBP (MRSA)
altered permeability
beta lactam/beta lactamase inhibitors
inhibits the beta lactamase which prevents the destruction of the drug
examples of these include:
- piperacillin-tazobactam
- amoxicillin - clavulanic acid
- ceftolozane - tazobactam
allergies to beta lactams
only about one in every 25 000-40 000 patients
the allergy is caused by beta lactam R groups
rash occurs in 5% of patients develop a rash but this is not an allergic reaction
cell wall active agents and how they work: glycopeptides
glycopeptides such as vancomycin and teicoplanin
these are gram positive agents only
mechanism of action: they bind to the terminal D-Ala of the cell wall peptide, preventing crosslinking to make PG
glycopeptide (Vancomycin) resistance
this primarily happens/is concerning for enterococcus and staph. aureus
the D-Ala-D-Ala target is altered - subsitutes D-lac instead
prevents vancomycin from being able to bind
vancomycin specturm of activity
gram positive cocci:
- MRSA, coagulase positive staph
- pen resistant S.pnemoniae and enterocuccos
gram positive rods:
- C. jeikeium - multidrug resistant
- C. difficile (vancomycin must be given orally bc it needs to be in the gut)
fosfomycin
cell wall synthesis inhibitor
good drug bc you dont have to worry about renal function problems - good for old people
one 3 gram satchel (like a tea bag)
in Canada mainly used for uncomplicated cystitis (multidrug resistant UTI) caused by E.coli or E. faecalis
fosfomycin mechanism of action
inhibits the synthesis of cell wall building blocks in the cytoplasm
does this by inactivating the enzyme enol-pyruvyl transferase
this blocks the condenesation of UDP-NAG with p-enolpyruvate
evolution of fluoroquinolones
1st gen: nalidixic acid
2nd gen: ciprofloxicin - broader spectrum and anti psuedomonal (can be given orally or through IV)
levofloxacin
3rd gen: moxifloxacin - enhanced gram positive and (+/-) anaerobic activity
fluoroquinolones
dna synthesis inhibitors
concentration dependent and highly bactericidal
very good oral bioavailablilty (don’t need an IV)
fluoroquinolones mechanism of action
binds DNA gyrase and DNA complex - allows gyrase to cut and unwind the DNA but does not allow it to re-anneal = toxic to the cell
also does this with topoisomerase 4
development of fluoroquinolones resistance
spontaneous mutations in gyrA and parC (most common method)
- this results in amino acid substitution causing reduced affinity
- requires both mutations to occur
over expression or up-regulation of efflux pumps
- pumps drugs out - when combined with the other mutations it is really bad
- pmrA (Gm+)/norA (Gm-)
- can also have down regulation of porin channels in gram negatives
also have acquired resistance but not as common
adverse events of fluoroquinolones
hyper and hypoglycemia
can have cartilage toxicity - causes tendons to rupture - restrict paediatric usage
what are fluroquinolones used to treat?
gram negative/atypical infections
- oral step down from serious infections
- pseudomonas (cipro/levofloxacin only)
inhibitors of protein synthesis
macrolides, lincosamides, streptogramins (MLS)
tetracyclines
aminoglycosides
linezolid
the macrolides
examples: erythromycin (not nice), clarithromycin, azithromycin
binds to the 50S subunit of bacterial ribosomes - inhibiting protein synthesis
macrolides mechanism of resistance
M phenotype: efflux pump - only effects macrolides
MLS phenotype: target site modification - effects macrolides, lincosamides, streptogramin
adverse events of macrolides
GI upset
infusion related phlebitis
ventricular arrhythmia
cyto P-450 interactions
clindamycin
this antibiotic is most commonly associated with C. difficile colitis
a lincosamide drug
also treats anaerobic infections (usually with a gram negative agent)
gram positive infections such as necrotizing fasciitis and staph infections
also used to treat C. perfringens in penicillin allergy cases
tetracyclines
examples: tetracycline, doxycycline, minocycline
bind reversibly to the 30S ribosomal subunit
excellent for treating atypical bacteria such as RTI and chlamydia
good activity against most gram positives, many enterobacteriaceae and community acquired MRSA
also treat animal born pathogens such as yersinia pestis, burcella, B. burgdoferi, rickettsiae
adverse events of tetracyclines
discolouration of teeth (only tetracycline not doxycycline), photosensitivity, depression of skeletal growth, esophageal ulceration
mechanisms of resistance against tetracyclines
energy dependent efflux
enzymatic inactivation
ribosomal protection
aminoglycosides
natural or semi-synthetic antibiotics - streptomycin (1944)
excellent gram negative activity including pseudomonas
good gram positive activity
bactericidal and concentraatoin dependent
examples = gentamicin, tobramicin, amikacin
how aminoglycosides work
enter through the inner membrane via an energy dependent transport system
- this step is rate limiting and blocked by divalent cations and anaerobiosis
the aminoglycosides irreversibly bind to the 30S ribosomal subunit
do not work in anaerobic environments because they require the ETC to enter the cell (this requires O2 as final e acceptor)
aminoglycosides mechanism of resistance
enzymatic modification is the most common
more than 70 enzymes
different substrate species
plasmid mediated
less common methods:
- altered ribosomal binding sites (only for streptomycin) and reduced uptake or decreased permeability
adverse events of aminoglycosides
have a narrow therapuetic index - therefore long term use required therapuetic drug monitoring
at too high of a concentration they can interfere with mammalian protein synthesis
ototoxicity - causes deafness due to cochlear and vestibular damage
nephrotoxicity - proximal tubule damage
aminoglycosides: monitoring therapy
once daily - trough levels only
six hours before the next dose should be less than 1mg/L
inhibitors of metabolic pathways
trimethoprim/sulfamethoxazole (septra, TMP/SMX)
good gram negative activity and some gram positive
blocks folic acid synthesis at two different points
TMP and SMX act additively
TMP-SMX mechanism of action
sulfonamides block tetrahydropteroic acid synthetase which converts PABA to dihydrofolic acid
- first step in converting PABA to purines
trimethoprfim block dihydrofolate reductase which converts dihydrofolic acid to tetrahydrofolic acid
- second step in converting PABA to purines
TMP-SMX mechanisms of resistance
chromosomal (less common):
metabolic bypass
overexpression of DHFR (dihydrofolate reducatse)
Plasmid (most common):
drug resistant variants of DHFR/DHPS (synthetase enzyme)
antibiotics that target bacterial membranes
colistin -gram negative agent (some gram positive activity)
daptomycin - gram postive agents
colistin
displaces divalent cations from phosphate groups of membrane lipids - disrpting the outter membrane
useful for treating multidrug resistant gram negative infections
can cause renal or neurotoxicity
daptomycin
cant be used to treat RTI because lung surfactant inhibits the drug
works by inserting into the cell membrane - causes rapid depolarization and K+ ion flux
bactericidal and concentration dependent
useful for treating MRSA
metronidazole
go to for anaerobic infections
90% bioavailability
very little resistance
cheap
metronidazole mechanism of action
little/no activity against aerobic bacteria
produces short lived toxic intermediates or free radicals under anaerobic (reducing ) conditions
-this inhibits nucleic acid synthesis by damaging or disrupting DNA
metronidazole mechanism of resistance
reduced drug activation
reduced permeability/efflux
altered DNA repair
adverse effects of metronidazole
GI intolerance antabuse effect - alcohol consumption will cause power puking peripheral neurotoxicity metallic tase black/brown discolouration of urine
why do we do susceptibility testing
- as a guide for treatment
- as an epidemiological tool
- can track the emergence of resistant strains
- real time tracking of local susceptibility - can recommend the optimal empiric treatment
why do we need to keep doing susceptibility testing
susceptibility pattern of many pathogens is not always predictable
susceptibility patterns of some pathogens evolve over time
components of susceptibility testing
- identification of the organism
- site of infection
- selection of anitbiotics
- selection of appropriate test method
- interpretation: requires indepth knowledge of resistance mechanisms, some organisms behave differently in vitro vs in vivo
- selective reporting
- quality control
susceptibility testing: identification of the organism- things to consider
is it predictably susceptible?
beta haemolytic strep are all suscetible to beta lactams
does it have intrinsic resistance?
gram negative bacteria to vancomycin
enterococcus to cephalosporins
does it have inducible resistance?
when you put the patient on these drugs (look susceptible in lab) their resistance is activated
hetero-resistance phenotypes
only about 3-4 cells are resistant but we need to find these cells
- eg oxacillin and MRSA
why is the site of infection is important for susceptibility testing?
CNS/Brain infections- need a drug that will cross the blood-brain barrier
UTI- need a drug that will be excreted by the kidneys into the urine
RTIs - daptomycin is inactivated by lung surfactant
do we report all antibiotics when we do susceptibility testing?
no
don’t report:
predictably susceptible organisms
organisms with inducible resistance mechanisms
inappropriate bug-drug combinations
known contraindications
don’t report 2nd or 3rd line drugs if 1st line works
quality control in susceptibility testing
reference strains (ATCC): MIC ranges/zone diameters
media: must be standardized to ensure the diffusion zone will be comparable to reference strains
antibiotics: shelf life and potency
incubation: temp, time, atmosphere, amount of inoculum
test methods
kirby bauer - disk diffusion
broth/agar microdilution
E-test
automated systems (vitek/microscan)
screening methods: agar based and nitrocephin discs
establishment of antibiotic breakpoints for susceptibility testing
interpretive criteria that establish the categories of susceptible, intermediate, or resistant:
MIC distributions
pharmacokinetics - absorption, distribution, accumulation, elimination (all in vivo measurements)
pharmacodynamics: %time/MIC, AUC/MIC(Cmax/MIC)
clinical/ bacteriological response
nitrocephin disks
quick screen for beta lactamase production
drop disk (with cephlosporins in it) on plate- if there is beta lactamase the disk changes colour
disk diffusion
also known as kirby bauer method
filter disc impregnated with antibiotic is place on a plate with a lawn of bacteria
measure the zone of inhibition around the disc
qualitative test
compare the diameters with standard tables - each drug-bug combination has a specific value for susceptible, resistant, intermediate
factors that affect size of zone inhibition
inoculum density - inoculum too light = larger zones
timing of disc application - kept on plate too long = small zones
temperature of incubation - temp less that 35 = larger zones
incubation time - should be 16-20 hours
depth of the medium - too thin = excessive zone size
potency of antibiotic disc - deterioration = reduced size
composition of medium
acidic or alkaline pH of medium
reading o zones - subjective errors
how to determine macrolide resistance phenotypes
do this via double diffusion disc testing - one disk contains clindamycin and the other contains macrolide - placement of these disks at a specific distance is ESSENTIAL
the M phenotype will show a zone of inhibition around the clindamycin disc but not the macrolide disk - therefore can only be efflux mechanism
if there are no zones of inhibition around the disks then the bacteria have the MLS phenotype
if the there is not zone of inhibition around the macrolide disk + there is a skewed zone around the clindamycin disc then the bacteria have an inducible MLS phenotype
- this zone will be smaller on the side closest to the macrolide disk
E-test
strip has a gradient of antibiotics
have a plate with a lawn of bacteria on it
place strip on plate
the MIC is equal to the point on the plate where the zones intersect
dilution method
broth or agar methods
give quantitative results:
- indicated the amount of drug needed to inhibit or kill a bacteria being tested
the MIC is the lowest concentration of the drug that inhibits the growth or multiplication of bacteria
the MBC is the lowest concentration that leaves less than 0.1% of the inoculum population alive
broth dilution method
90% of this testing is done using mueller-hinton broth
prepared in doubling dilutions (0.5,1,2,4,8, etc)
inoculation of bacteria is incubated overnight
- controls + no antibiotic and no bacteria
turbidity visualization = MIC
subculture non-turbid tubes overnight
growth (bacterial count) - MBC
typically if the MIC is determined to be 16 mg/L then the MBC will be 32mg/L - need to go one dilution step further to ensure that there are no bacteria still alive
agar dilution method
agar plates with doubling dilutions of antibiotics
one concentration per plate but can have multiple different strains on one plate bc you can use a replicator
automated systems (vitek)
based on broth dilution
limited break points
useful for most common pathogens
not reliable fo fastidious pathogens (hard to grow)
what must bacterial identification begin with in the clinical setting?
understanding of epidemiology, patho-physiology, and infectious disease
general steps on classifying using taxonomy
gram negative or positive?
cocci of bacilli?
anaerobic or aerobic?
classification of aerobic gram positive cocci
need to determine if the arrangement is in clusters or pairs and chains
clusters indicates staph species
pairs and chains indicates streptococcus or enterococcus
general properties of staphylococcus
gram positive
facultative anaerobes
catalase positive except for one exception
commonly found on human skin and mucous membranes
20+ species
4 species associated with disease: S. aueus, S. lugdunensis, S. epidermidis, and S. saprophyticus
there is also S. psuedintermedius which is an animal pathogen that was been seen more in humans now
catalase test
determines if the organism produces catalase
catalase breaks hydrogen peroxide into water and oxygen
- this allows organisms to break down harmful metabolic products that result from aerobic respiration
this test is positive for staph nut negative for strep and enterococcus
epidemiology of S.aureus
10-15% of people carry it around in their nose or other mucous membranes (this number is higher for people in hospital settings)
gram positive
most common type of hospital acquired infection
typically infections are endogenous (spread form person to person)
MRSA is a concern in hospitals and community
slide and tube coagulase postive