Medical microbiology Flashcards

1
Q

Why do most viruses not infect us?

A

Most of these don’t “infect” us:
They are adapted to non-human hosts
They are excluded by surface barriers
Innate Immunity prevents them establishing
Our adaptive immune response has seen something similar

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

What are the different sites of microbe entry?

A
Conjunctiva 
Respiratory tract 
Alimentary tract 
Urinogenital tract 
Anus 
Skin 
Scratch, injury 
Capillary 
Arthropod
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the general patterns of viral infection?

A

a) Acute infection (a huge spectrum of disease and range outcomes). Resolution by immunity?
Influenza pathogenicity; different strains produce a huge range of outcomes
b) Latent, reactivating infection (chronic infection)
Human Herpes Viruses
Herpes simplex virus, first appears as primary gingivostomatitis, and then stays latent until it is seen as a cold sore.
HHV-3 causes chicken pox then reappears as shingles. Also called varicella virus Persistent infection (chronic infection)-has 2 types of patterns

¥ HIV; Virus infects CD4+ cells and weakens immune system
¥ HCV; Virus infects hepatocytes and damages liver
¥ Congenital Rubella; if infected in utero, virus is seen as self, baby is born immunotolerant and virus continues to replicate (and cause damage) in neonatal tissues

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

How does virus infection of a host lead to disease?

A

Many infections are apathogenic or associated with relatively mild symptoms; it is important to realize that from the virus’ point of view these are not always failed or resolved infections – a successful virus is one that replicates well enough to spread to the next host

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

Describe inapparent infections

A

Many infections are apathogenic or associated with relatively mild symptoms; it is important to realize that from the virus’ point of view these are not always failed or resolved infections – a successful virus is one that replicates well enough to spread to the next host

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

How does virus infection of a host lead to disease?

A

Pathogenesis results from cell and tissue damage caused by the viral infection. On most occasions the damage is limited by the host’s immune system
Eg, Ebola targets vascular endothelial cells.
Influenza A virus targets lung epithelia
RSV induces syncytia in lung epithelia. On some occasions the relative limited damage caused by the virus is made worse or even caused by the host’s immune system (= immunopathology)
eg, hepatitis C

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

Describe HCV virus

A

Chronic hepatitis is a disease of severe liver damage and loss of hepatocytes – caused by persistent HCV infection
HCV is non-cytopathic
Hepatitis associated with extensive liver infiltration of leukocytes
Pro-inflammatory cytokine levels very high
Viral clearance and disease is associated with generation and infiltration of CD8+ cells which attack infected cells and destroy them
HCV persistence is associated with the generation of HCV variants that are not recognised by CD8+ cells

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

Describe Dengue fever

A

Dengue virus infection is the most common mosquito-borne infection worldwide – even surpassing malaria
There are 2.5 billion people at risk of dengue due to living in an endemic area. There are an estimated 50–100 million infections per year, and 500,000 hospitalizations due to severe disease
The case fatality rate from severe dengue is 1 - 5%
There are 4 serotypes (1–4), all of which have the same clinical manifestations

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

Describe the immunopathology: Dengue virus

A

Severe dengue, which may include dengue shock syndrome (DSS), and hemorrhage
Greatest risk is a previous infection with a different serotype
Antibodies formed in response to a dengue infection are not cross-protective against other subtypes of the virus. In fact they may result in more severe disease due to a phenomenon known as antibody-dependent enhancement or ADE
Non-neutralizing antibodies coat virus, forming immune complexes which get internalised into mononuclear phagocytes through their Fc receptors; fixation of complement by circulating immune complexes results in release of products of the complement cascade leading to sudden increased vascular permeability, shock and death

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

Influenza virus

A

Influenza People of all ages are infected, usually only a serious problem in the old or children with asthma
Pathology
Mild URTI to severe LRTI
Lower respiratory tract infection causing damage to lung epithelia and viral pneumonia, often secondary pneumonia
Fever, often prolonged
Neurological (headache, malaise)
Myalgia
Infection generates powerful, long-live immunity
Easy to vaccinate against if you know what’s coming

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

Why do we need antivirals?

A

Quick killers e.g. influenza; ebola; MERS; SARS
Slowly, progressive chronic disease leading to cancer
hepatitis B [350,000,000 carriers]
hepatitis C [200,000,000 carriers]
human papilloma viruses
[cervical cancer, second commonest cancer in women]
Human immunodeficiency virus (HIV)
[40 million infected]

Acute imflammatory e.g. herpes

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

What do we use antivirals for?

A

Treatment of acute infection
Influenza ; Chickenpox; herpes infections -(aciclovir)

Treatment of chronic infection:
HCV, HBV, HIV (numerous different agents)

Post-exposure prophylaxis and preventing infection:
HIV (PEP)

Pre-exposure prophylaxis: HIV (PrEP)

Prophylaxis for reactivated infection: e.g. in transplantation
CMV (ganciclovir, foscarnet)

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

Describe the principle of antivirals as therapeutic agents

A

Selective toxicity Due to the differences in structure and metabolic pathways between host and pathogen
Harm microorganisms, not the host
Target in microbe, not host (if possible)
Difficult for viruses (intracellular), fungi and parasites
Variation between microbes

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

Why is it so difficult to develop effective, non toxic anti-viral drugs

A

Viruses enter cells using cellular receptors which may have other functions
Viruses must replicate inside cells – obligate intracellular parasites
Viruses take over the host cell replicative machinery
Virsues have high mutation rate - quasispecies
Anti-virals must be selective in their toxicity
i.e. exert their action only on infected cells
Some viruses are able to remain in a latent state e.g. herpes, HPV
Some viruses are able to integrate their genetic material into host cells
e.g. HIV

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

What are the considerations in developing safe anti-viral agents?
-Can stages of infection be targeted?

A

Cellular receptor may have other important function

Viral enzymes may be very similar to host

Blocking cellular enzyme may kill cell

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

Describe the virus life cycle

A
  1. Recognition
  2. Attachment
  3. Penetration
  4. Uncoating
  5. Transcription
  6. Protein synthesis
  7. Replication
  8. Assembly, and envelope
  9. Lysis and release, or budding and release.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Describe the modes of action of selected anti-virals

A

Preventing virus adsorption onto host cell
Preventing penetration
Preventing viral nucleic acid replication (nucleoside analogues)
Preventing maturation of virus
Preventing virus release

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

gIve example of some antivirals

A
Amantadine 
Acyclovir, Ganciclovir, Ribavarin, 
AZT
Interferons, 
HIV protease inhibitors 
RSV -guanosine analogue 
Zanamivir-influenza release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Give examples of selective toxicity viral targets?

A

Discovery of virally encoded enzymes sufficiently different from human counterparts
e.g.

Thymidine kinase and HSV/VZV/CMV
Protease of HIV
Reverse transcriptase of HIV
DNA polymerases
Neuraminidase of influenza virus

Act as selective targets with minimal effect on host enzymes or processes

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

What virus can cause muco-cutaneous lesions?

A

Herpes simplex type 1

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

What are the 4 types of herpes viruses

A
Herpes viruses include: 
Herpes simplex (HSV), 
Varicella Zoster Virus (VZV)
Cytomegalovirus (CMV)
Epstein-Barr virus (EBV)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are the different antivirals we can use for herpes virus

A

aciclovir
IV/oral/topical
For HSV, VZV treatment/prophylaxis

CMV/EBV prophylaxis

ganciclovir
IV/oral
For CMV
Foscarnet
IV/local application
For CMV	
cidofovir
IV  for CMV
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What virus causes chickenpox, and then reactivates to cause shingles

A

Varicella Zoster virus

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

Describe the selective toxicity of aciclovir

A

Aciclovir is activated to active drug
Substantially more in infected cells. Requires 2 viral enzymes
= selectively activate ACV
= selectively inhibited

Accounts for low toxicity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Why is aciclovir so effective and safe?

A

HSV thymidine kinase (TK) has 100x the affinity for ACV compared with cellular phosphokinases

Aciclovir triphosphate has 30x the affinity for HSV DNA polymerase compared with cellular DNA polymerase

Aciclovir triphosphate is a highly polar compound - difficult to leave or enter cells (but aciclovir is easily taken into cells prior to phosphorylation)

DNA chain terminator

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

what can aciclovir be used as a treatment for?

A

Treatment of encephalitis
Treatment of genital infection
suppressive therapy for recurrent genital herpes

Varicella zoster virus
Treatment of chickenpox
Treatment of shingles
Prophylaxis of chickenpox

CMV/EBV
Prophylaxis only

Shingles (zoster)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe ganciclovir

A

Active for CMV
- reactivated infection or prophylaxis in organ transplant recipients
congenital infection in newborn
retinitis in immunosuppressed
Structurally similar to aciclovir
CMV does not encode TK but has UL97 kinase
Inhibits CMV DNA polymerase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

what virus does aciclovir not work on?

A

CMV, because it doesnt have the tyrosine kinase needed to activate the drug in the first step.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Describe other anti-herpes virus agents

A

Foscarnet:
Selectively inhibits viral DNA/RNA polymerases and RTs
No reactivation required
Binds pyrophosphate binding site – a structural mimic
used for CMV infection in the immunocompromised
e.g. pneumonia in solid organ and bone marrow transplants.
May be used because of ganciclovir resistance (TK mutants)

Cidofovir
Chain terminator - targets DNA polymerase
Competes with dCTP
Monophosphate nucleotide analog
Prodrug – phosphorylated by cellular kinases to di-phosphate
drug active against CMV; but MUCH MORE nephrotoxic
Treatment of retinitis in HIV disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Describe the resistance to antivirals in herpes viruses

A

Two main mechanisms

Thymidine Kinase mutants
DNA polymerase mutants

If occurs in TK, drugs not needing phosphorylation are still effective (e.g. foscarnet, cidofovir)

If occurs in DNA polymerase, all drugs rendered less effective

VERY RARE in immune competent patients (low viral load)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Describe the structure of HIV

A

Envelope protein, gp120 with transmembrane gp41
Membrane associated matrix protein Gag 17
Nucleocapsid protein Gag p24
Reverse transcriptase
Viral envelope
ds RNA genome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Name the 7 steps in the life cycle of HIV

A
  1. Attachment with binding of viral gp120 via CD4 and CCRX
  2. Reverse transcription of RNA into ds DNA
  3. Integration into host chromosome of proviral
  4. Transcription of viral genes
  5. Translation of viral mRNA into viral proteins
  6. Virus assembly and release by budding
  7. Maturation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Describe anti-HIV drugs

A
  1. Anti-reverse transcriptase inhibitors
    nukes -nucleoside/nucleotide RT inhibitors
    non-nukes -non-nucleotide RT inhibitors (allosteric)
  2. Protease inhibitors - multiple types
  3. Integrase inhibitors – POL gene - protease, reverse transcriptase and integrase (IN)
    with the 3´end encoding for IN (polynucleotidyl transferase)
  4. Fusion inhibitors – gp120/41 - biomimetic lipopeptide

Treatment-HAART
(combination of drugs to avoid resistance)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Describe nucleoside reverse transcriptase (RT) inhibitors

A

Nukes
AZT - Zidovudine

Synthetic analogue of nucleoside thymidine –
when converted to tri-nucleotide by cell enzymes, it blocks RT by
- competing for natural nucleotide substrate dTTP
incorporation into DNA causing chain termination
Others ddI, ddC, d4T, and 3TC (2′,3′-dideoxy-3′-thiacytidine

35
Q

Describe non nucleoside reverse transcriptase inhibitors ‘Non-nukes’

A

Non-competitive inhibitor of HIV-1 RT

Synergistic with NRTI’s such as AZT because of different mechanism

36
Q

Describe pre and post exposure prophylaxis for HIV

A

PEP – within 72 hours post exposure - take for 28 days.
2x NRTIs + integrase inhibitor

PrEP – pre-exposure - blocks transmission  
2x NRTIs (Truvada) 
two tablets 2 – 24 hours before sex, one 24 hours after sex and a further tablet 48 hours after sex -  called ‘on-demand’ or ‘event based’ dosing
2 x NRTIs = 
Combination of Nucleoside RTIs
emtricitabine (guanosine analog) 
\+ 
tenofovir (adenosine analog)
37
Q

Describe the resistance to anti-virals

A

Use of single agents leads to rapid development of resistance

The drug binding site is altered in structure by as few as one amino acid substitution

Mutation rate - high
Viral load – high
resistance

38
Q

describe HIV resistance to antivirals

A

Selection pressure and mutation frequency
Increased mutation rate seen in HIV.
They form a quasispecies within an individual patient:-
A viral swarm

The error rate in copying viral genome by reverse transcriptase enzyme is 1 base per 10 4-5 incorporations; lacks proof reading capacity. So, for HIV with 10 9-10 viruses produced every day, ALL possible viral variants would be produced
Hence use of combinations of antivirals
e.g. HAART

39
Q

Antivirals for the influenza virus

A

Amantadine
Inhibit virus uncoating by blocking the influenza encoded M2 protein when inside cells and assembly of haemagglutinin
Now rarely used

Zanamivir and Oseltamivir (Tamiflu)
Inhibits virus release from infected cells via inhibition of neuraminidase
Oseltamivir –oral
Zanamivir- inhaled or IV - less likely for resistance to develop

40
Q

Describe action of relenza (zanamivir) and Tamiflu (oseltamivir)

A

target and inhibit NA at highly conserved site (reduce chances of resistance via mutation)
prevent release of sialic acid residues from the cell receptor
preventing virus budding and release and spread to adjacent cells.

Neuraminidase inhibitors

41
Q

Describe the influenza resistance to antivirals

A

Resistance sometimes only requires a single amino acid change - seen recently with swine flu (H1N1) and Tamiflu (oseltamivir)
Point mutation (H275Y; tyrosine replacing histidine)
Seen in immunocompromised patients; shed virus for weeks/months
Likely to be selected from among quasispcies during treatment
Transmissible and virulent
Remains sensitive to zanamivir;

42
Q

Describe postexposure prophylaxis to viruses

-Emergency and require rapid treatment

A

Hep B
specific Hep B immunoglobulin (passive immunity)
+ vaccination
within 48 hours (HBV treatment includes antivirals 3TC/NRTIs)

Hep C
interferon- + ribavarin (anti-viral) for 6 months
within first 2 months of exposure
90% cure rate - now direct acting antivirals

HIV
	80% protection i.e. no sero-conversion
	must be FAST – hours
	antiviral drug treatment – 28 days
	2xNRTI + protease or integrase inhibitor
43
Q

Describe the Hepatitis C virus

A

9.6 Kb RNA virus, enveloped; Flaviviridae family; identified in 1989
transmitted via blood – infectious (mother to baby)
increasingly common – high risk groups – drug users 20% +ve; – needles (sex?)
major cause of chronic liver disease
estimated 170 million people infected worldwide
occupational risk groups – healthcare workers
needle-stick risk – 3% to sero-conversion; chronic carriage almost certain (85%)
long incubation – 1 - 6 months
vaccination NOT available
prevalence in UK - ~6000 per year ( 95% are i/v drug users)
early treatment facilitates resolution

44
Q

Ribavirin

A

Block RNA synthesis by inhibiting inosine 5’-monophosphate (IMP) dehydrogenase –
this blocks the conversion of IMP to XMP (xanthosine 5’-monophosphate)
and thereby stops GTP synthesis and, consequently, RNA synthesis

Treat: RSV and HepC (in combination with pegylated interferon)

45
Q

Describe Hepatitis C virus and direct acting antivirals

A

relatively new class of medication

acts to target specific steps in the HCV viral life cycle

shorten the length of therapy, minimize side effects, target the virus itself, improve sustained virologic response (SVR) rate.

structural and non-structural proteins - replicate and assemble new virions

HCV - first chronic viral infection to be cured without IFN or ribavirin.
All the major HCV-induced enzymes - NS2-3 and NS3-4A proteases, NS3 helicase and NS5B RNA-dependent RNA polymerase (RdRp) are essential for HCV replication and are potential drug targets.
DAA with different viral targets, are synergistic in combinations

46
Q

Why are some vial infections untreatable?

A
Because they are self limiting. Can give vaccinations for them. rabies
dengue
Common cold viruses
Ebola
HPV
Arbovirsues
47
Q

What antibiotics are cell wall inibitors?

A

B-lactams (penicillins, cephalosporins) and vancomycin.

48
Q

Principle of antibiotic resistance

A

Bacteria acquire genes, and if you select with antibiotics for these clones of bacteria that have acquired the genes, they then spread throughout the population.

49
Q

Describe the use of antibacterials

A

Widely used and misused drugs
20-50% questionable use

In hospitals - 30% of drug budget
~25% of patients have received antibiotics within the previous 24h
In ITU, 50% of patients are on antibiotics

50 million prescriptions per year
80% of human use is in the community
50% - respiratory infections
15% - urinary tract infections

50
Q

What are antibiotics

A

Natural products of fungi and bacteria - soil dwellers

- natural antagonism and selective advantage
 	- kill or inhibit the growth of other microorganisms

most derived from natural products by fermentation,
then modified chemically :increase in pharmacological properties
Increase in antimicrobial effect
Some totally synthetic - e.g. sulphonamides

51
Q

Describe how we discovered Penicillin

A

Alexander Fleming
Diffusion of penicillin into agar caused
lysis of S.aureus. The plate had been left
in a room for several weeks at a temperature
that allowed growth of mould
without overgrowth of the bacteria.

52
Q

Describe the principles of antibiotics as therapeutic agents

A

Selective Toxicity
Due to the differences in structure and metabolic pathways between host and pathogen
Harm microorganisms, not the host
Target in microbe, not host (if possible)
Difficult for viruses (intracellular), fungi and parasites
Variation between microbes

53
Q

What is the therapeutic margin

A

active dose (MIC) versus toxic effect
narrow for toxic drugs - e.g. aminoglycosides, vancomycin
ototoxic, nephrotoxic

-the dose between when a drug can stop becoming therapeutic, and start causing host damage.

54
Q

Describe microbial antagonism

A

Maintains flora - complex interactions
Competition between flora

Limits growth of competitors and PATHOGENS

55
Q

how can we get a loss of flora?

A

bacterial or pathogen overgrowth

e.g. Antibiotic Associated Colitis :
(clindamycin, broad-spectrum lactams, fluoroquinolones)
- pseudomembranous colitis

	Clostridium difficile  (part of normal flora of 3% of population), causes pseudomembranous colitis of colon, so colon cannot absorb water, causing diarrhoea. 

Ulcerations-inflammation
Severe diarrhoea
Serious hospital cross infection risks.

56
Q

What do is equally as important as antibiotics to result in bacteiral clearance?

A
Immunity 
Antibiotics and immunity of individual result in bacteria  clearance. Immunosuppression
e.g. 
cancer chemotherapy, transplantations,  myeloma, leukaemias, 
HIV with low CD4
Neutropenics, asplenics, renal disease, 
diabetes, alcoholics,
Babies,  elderly., ………..
57
Q

What can antibiotics be classified by?

A

Classified by:-

Type of activity

Structure

Target site for activity

58
Q

Bactericidal vs Bacteriostatic

A

Bactericidal:
Kill bacteria
Used when the host defense mechanisms are impaired
Required in endocarditis, kidney infection

Bacteriostatic:
Inhibit bacteria
Used when the host defense mechanisms are intact
Used in many infectious diseases

Varies for drug and species and concentration

59
Q

Types of activity of bacteria

Spectrum of activity

A

Broad Spectrum Antibiotics:
Effective against many types
Example: Cefotaxime

Narrow Spectrum Antibiotics:
Effective against very few types
Example: Penicillin G

60
Q

Describe the refinement of antibiotic activity

A

Antbiotics can be altered pharmacologically to have enhanced effect on some bacteria. Go from being first generation, to second generation, to third generations.

61
Q

Molecular structure of antibiotics classificatin

A

Duff antibiotics have diff structures.
Structural mimics of natural substrates for enzymes

B-lactams have a beta lactam ring, mimics the active structure in penicillin.

62
Q

Describe the structure of bacteria

A

Infolding of plasma membrane, capsule, cell wall, DNA coiled into nucleoid, basal body, ribosomes, cytoplasm, plasma membrane, pili, cytoplasmic inclusion,

63
Q

Describe the different bacterial targets for current antibiotics used in the clinic

A
  1. Cell wall synthesis, eg, cycloserine, vancomycin, teichoplanin, penicillin.
  2. Folic acid metabolism, trimethoprim, sulfonamides.
  3. Cell membrane, colistin, daptomycin.
  4. Protein synthesis
    50S inhibitors, erythromycin, clindamycin
    30S inhibitors, tetracycline, streptomycin, getamicin.
  5. DNA and RNA processing
    Quinolones,
    -DNA gyrase, DNA directed RNA polymerase.
  6. Forming free radicals, Metroonidazole, nitrodurantoin.
64
Q

Difference between gram positive and gram negative bacteria

A

Gram positive

  • Peptidoglycan on outside of membrane
  • Peptidoglycan makes majority of cell wall, but also contains lipoteichoic acids, and integral proteins.

Gram negative
-peptidoglycan found in periplasmic space between outer and inner membrane. To get to it, you need porins, and selective transport mechanisms, found in outer membrane.

-Different structure of cell wall between them both.

65
Q

How do bacterial cell wall inhibitors work?

A

To make peptidoglycan, there are two dimers, one with 5 peptide amino acids attatched. This contains to D-ALA, D-ALA terminal amino acids.
An inhibitor like cycloserine inhibits reactions involved in incorporation of alanine into cell wall precursor.

An enzyme in the bacterial cell breaks off the terminal D-ALA, and attaches the remaining D-ALA to adjacent chain, causing the formation of peptidoglycan. Vancomycin inhibitor binds to the terminal D-ALA D-ala residues, and prevents incorporation of sub unit into growing peptido glycan chain.
Bacitracin prevents dephosphorylation of phospholipid carrier which prevents regeneration of carrier necessary for synthesis to continue.

Penicillins and cephalosporins (beta-lactams) bind to and inhibit enzymes which catalyse this link. Peniclilin forms covalent enzyme-inhibitor complex with transpeptidase, as it is a structural mimic of the 5 amino peptide chain.

Antibiotics are often structural mimics of natural substrates for bacterial enzymes.

66
Q

Describe folic acid synthesis inhibitors

A

bacteria can make their own folic acid. So we can inhibit this process, sulfonamides and dapsone block the first enzyme.
Trimethoprim antibiotic blocks the enzyme dihydrofolate reductase.

Gives the bacteria selective toxicity.

67
Q

When do we use antibiotics

A

Treatment of bacterial infections

Prophylaxis - close contacts of transmissible infections
carriage rates ( ~80% in outbreaks)
e.g. meningitis
- prevention of infection e.g. tuberculosis

	- peri-operative cover for gut surgery
	- people with  susceptibility to infection. 

Inappropriate use - viral sore throats - patient pressure

68
Q

Dose of antibacterial MIC

A

This will depend upon the age, weight, renal and liver function of the patient and the severity of infection
Depend on the susceptibility of the organism
Will also depend upon properties of the antibiotic i.e. enough to give a concentration higher than the MIC (minimum inhibitory concentration)
! at the site of infection

69
Q

Antibiotic combinations

A

BEFORE an organism identified in life-threatening infections
e.g. endocarditis, septicaemia

Polymicrobial infections e.g. abscess, G.I. perforation
anaerobes and aerobes

Less toxic doses of an individual drug possible

Synergy e.g. penicillin and gentamicin
Co-trimoxazole (sulphonamides + trimethoprim)

reduce antibiotic resistance e.g. Tuberculosis

70
Q

Beta lactams : Penicillins

A

Basic penicillins e.g. benzylpenicillin(PenG), penicillin V
Active against streptococci, pneumococci, meningococci, treopnemes.
Most strains of Staphylococcus aureus are resistant.

Anti-staphylococcal penicillins e.g. flucloxacillin
narrow spectrum, G+ves, beta-lactamase resistant, less potent that PenG
Not MRSA

Pen G benzlypenicillin (G= gold standard);
not acid stable i/v or i/m good for some G-ves as well as G+ves

penV phenoxymethlypenicillin
oral (more acid stable than penG)
less active v G-ves, but same activity v G+ves as PenG

Broader spectrum penicillins e.g. ampicillin
Spectrum of activity is similar to basic penicillins but also includes some Gram-negative organsims and also enterococci

Anti-pseudomonal penicillins e.g. piperacillin
extended spectrum beta-lactam antibiotic
also G+ve, G-ve, anaerobes

Beta-lactam/beta-lactamase inhibitor combinations
e.g. co-amoxiclav (Augmentin)
Spectrum like amoxicillin plus activity against some Gram-negatives and Staph aureus

71
Q

Why is antibiotic resistance a global concern?

A

~ 25,000 people die every year across Europe because of infections related to Antimicrobial Resistance (AMR)

In USA, MDR infections cost the health-care system ~20 billion US$ annually and generate more than 8 million additional hospital days

Antibiotic resistance:
Increases mortality
challenges control of infectious diseases
threatens a return to the pre-antibiotic era
increases the costs of health care
jeopardizes health-care gains to society

72
Q

Describe the ‘Superbug’

A

Drug resistant bacteria
are NOT MORE pathogenic

We just have fewer antibiotic options
for treatment

73
Q

Describe the mechanisms of antibiotic resistance

A
  1. Altered or new target
    e.g.
    Ribosome
    Porin
    PBPs –
    peptidoglycan synthesis
    DNA gyrase
    RNA polymerase
    Mcr1 & colistin
  2. Drug inactivation
    e. g. beta-lactamase
  3. Metabolic by-pass
    e.g. vancomycin
    D-ala-D-lac
  4. Efflux pump
  5. Overproduction of target
    - trimethoprim
  6. Intrinsic impermeability

Mechanisms of resistance
Natural resistance

Genetic Mechanisms - acquired

Non-Genetic Mechanisms (growth phases)

74
Q

Describe the antibacterial paths to resistance

A

Directed at antibiotic itself -
Degrading the drug
Modifying the drug

New or Altered target
antibiotic no longer binds
e.g. PBPs - PBP2a in MRSA

Altered transport
Actively pumping drug out - efflux pump
porins no longer influx drug

Metabolic by-pass
metabolic change D-ala-D-lac and vancomycin

75
Q

Describe the difference between natural resistance as opposed to acquired.

A

Drug must reach target - natural barriers, porins, export pump

G+ve peptidoglycan - highly porus - no barrier to diffusion
G-ves outer membrane - barrier resistance advantage

Porins single mutation - multiple resistance

76
Q

Genetic mechanisms

A
Chromosome-mediated
Due to spontaneous mutation: 
in the target molecule 
in the drug uptake system
Mutants are SELECTED ; they  are NOT induced

Plasmid-mediated
Common in Gram-negative rods
Transferred via conjugation
Multidrug resistance

The mutant is out there-we select it. As bacteria are constantly mutating.

Genetic mechanisms of resistance Production of drug-inactivating enzymes

Altered target structures – new or mutations

Alteration of membrane permeability

Altered influx or efflux - new pumps or mutations or altered gene expression

77
Q

Gene transfer in bacteria summary

A

Mechanism for genetic heterogeneity and evolution
Rapid, cross-species
Virulence (toxins), drug resistance, antigens (immune evasion)

  1. Transformation
    - fragment of DNA from another bacterial cell enters bacterial chromosome.
  2. Transduction
    - Fragment of DNA from another bacterial cell (former phage host, is transmitted into bactera)
  3. Conjugation
    - Sex pili form, allow the transfer of bacterial DNA.
78
Q

Describe resistance to Beta-lactams by gram positive and gram negative bacteria

A

Gram + ve

ß-lactamase (Penicillinase), destroys beta lactam ring

Alteration of the transpeptidase enzyme
(PBP)

Gram - ve

ß -Lactamase (Penicillinase), destroys beta lactam ring.

Alteration of porins

Augmentin/co-amoxiclav

  • combination of both
  • binds to and inactivates beta lactamases. No antibacterial activity of its own.
79
Q

Describe the different ways of beta lactam resistance in gram negative bacteria

A
  1. Porin mutates or new porin type
    Multi-resistant, so beta lactam cannot enter bacteria
  2. PBP - mutates or bacteria
    acquires a new PBP (penicillin binding protien)
  3. Bacteria acquires a beta lactamase enzyme.
80
Q

Mechanisms by which bacteria become resistance to penicillin

A

Produce penicillinases / beta lactamases that cleave the beta lactam ring
- penicillin is inactivated

Acquire alternative forms of / or mutations in penicillin binding proteins (PBPs)
- penicillin can’t bind

Acquire alternative forms of / mutations in porins,
- penicillin cannot get into cell

Acquire alternative forms of / mutations in efflux pumps
- penicillins are pumped out faster

81
Q

What is the only effective treatment of MRSA

A

vancomycin

82
Q

Describe vancomycin resistance

A

Acquisition of van operon by transposition

-Makes D-ala, D-lactate, prevents vancomycin binding.

83
Q

Non genetic mechanisms of resistance

A

Inaccessibility to drugs
(e.g., abscess, TB lesion)

Stationary phase/vegetations and biofilms
(non-susceptible to inhibitors of cell wall synthesis)

84
Q

How to prevent/overcome antibiotic resistance

A
  1. Control use not in animal feeds
    complete course [DOTS for TB]
    appropriate prescribing
  2. New or modified drugs few in past 25 years
  3. Combination therapy different targets
    overcome mutation rates
  4. Infection control individual - ward - society

Re-establish susceptible fora?