Chemotherapy Flashcards

1
Q

What is the concept of an antibiotic

A

typically antibacterial or antifungal drugs, interfering with some structure or process that is essential to growth or survival of these organisms without harm to the eukaryotic host harbouring the infecting cells.

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

How have most of the antibiotics in the last 60 years come about

A

natural products, elaborated by one microorganism in a particular habitat and set of environmental conditions to affect neighbouring microorganisms, either to regulate their growth or to trigger their elimination.

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

What are the 3 main classes of synthetic antibiotic

Give an example of each and their introduction date

A

sulfa drugs (such as sulfamethoxazole), introduced in the 1930s,

fluoroquinolones (such as ciprofloxacin), introduced in the 1960s,

oxazolidinone (linezolid), approved in the USA in 2000.

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

What dichotomy does the existence and clinical development of both synthetic and natural product antibiotics reflect in the 20th century

A

At one end of the spectrum was the medicinal chemistry view and the
classical “magic bullet” approach, initiated by Ehrlich, that pure
compounds could be made with therapeutic specificity and utility

At the other end of the spectrum, and from a separate track, came the isolation of penicillin (by Fleming in 1929), a natural product, as a potent antibacterial agent

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

Which type of antibiotic was a triumph for Ehrlich’s ‘magic bullet’ approach

A

sulfa drugs

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

Why was the isolation of penicillin important

A

led to the recognition of the paradigm that microbes wage war against each other with antibiotics

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

Name 8 antibiotic drug classes which stemmed from the realization that microbes wage war on each other

A

penicillins and cephalosporins,
tetracyclins,
streptomycins
and later generations of aminoglycosides,
chloramphenicol, rifamycins,
glycopeptides
the erythromycin class of macrolide antibiotics

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

What does broad or narrow range mean?

A

those that kill or inhibit a wide range of Gram-positive and Gram-negative bacteria are said to be broad spectrum.

If effective mainly against Gram-positive or Gramnegative bacteria, they are narrow spectrum.

If effective against a single organism or disease, they are referred to as limited spectrum.

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

When did the 2 strands of antibiotic discovery converge

A

with the introduction of later generations of semisynthetic variants of b-lactam antibiotics and macrolides, in which chemistry is used to engineer some desired new property, such as oral bioavailability, increased stability, broader spectrum of activity (referred to as extended spectrum), or efficacy against resistant microorganisms.

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

From 2000 to 2010, how did the use of last resort antibiotics change

A

increased consumption of carbapenems (45%) and polymixins (13%)

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

Antibiotic use increased by 36% from 2000 to 2010, where was this mainly accounted for? How can we stop this?

A

Brazil, Russia, India, China, and South Africa accounted for 76% of this increase

programmes that promote rational use through coordinated efforts by the international community should be a priority

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

bactericidal vs bacteriostatic

A

antibiotics that stop bacteria or fungi from growing are bacteriostatic, exemplified by chloramphenicol.

Antibiotics that cause cell death are
bactericidal; they lower the cell count of the infecting organism, as shown for penicillin

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

When would you use bactericidal drugs over bacteriostatic antibiotics?

A

when the immune system is compromised as bacteriostatic antibiotics require the bacteria to be eliminated by the immune system

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

Can a drug be both bacteriostatic and bactericidal

A

Some antibiotics can display bacteriostatic activity in some circumstances and bactericidal activity in others, where sufficient damage to one or more metabolic pathways or cellular structures occurs such that a net bactericidal response is triggered

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

What is the MIC and MBC for antibiotics

A

minimal inhibitory concentration (MIC): lowest concentration of a drug that prevents growth of a particular bacterium.

The minimal bactericidal concentration (MBC) is the lowest concentration that kills the bacterium

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

Discriminate between selective toxicity and therapeutic index

A

selective toxicity: growth of the infecting organism is selectively inhibited or killed without damage to the cells of the host

therapeutic ratio: the ratio of the maximum non-toxic dose over the minimum effective dose of a drug.

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

What is the selective toxicity and therapeutic index of penicillin

What drug has a selective toxicity and therapeutic index that is the opposite of this

A

high degree of selective toxicity, which corresponds with a large therapeutic index

Polyene antibiotic (eg amphotericin B) have both a low
degree of selective toxicity and a low therapeutic index
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Are the examples of penicillin and polyene antibiotics representative of most antibiotics’ selective toxicity and therapeutic index relationship?

Give an example

A

no
there is often little relationship between selective
toxicity and therapeutic index.

aminoglycosides

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

Describe the selective toxicity and therapeutic index of aminoglycosides

A

very selective with respect to killing bacterial versus host cells, but for some members of this class, unrelated effects on the patient’s nervous system, kidneys, or inner ear (hearing, balance) result in a much lower margin for therapeutic error that would be predicted on the basis of their selective action on cell viability

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

Where does the selectivity of anthracyclines come from

A

differential accumulation:
some tumour cells have enhanced rates of glycolysis, and as a result, reduced pH. Drugs (e.g., anthracyclines) that are trapped in the cell through protonation (thus becoming cationic) are accumulated more in tumour cells than in normal cells.

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

Give an example of differential activation in cancer chemotherapy

A

drugs that are activated by reduction (e.g. alkylating agent Mitomycin C), have enhanced toxicity in hypoxic tumour cell in solid tumours

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

How can cancer drug selectivity be based on differential importance

A

Alkylating and cross-linking agents, antimicrotubule drugs and antifolates are effective because cancer cells have a high demand in DNA replication and cell
division.

Antibodies and small molecule inhibitors target signaling pathways that are permanently switched on in cancer cells.

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

What are the 4 major targets for antibiotic drugs

A

(i) cell wall biosynthesis (sometimes membrane is targeted),
(ii) protein biosynthesis,
(iii) DNA replication, repair and expression
(iv) folate coenzyme
biosynthesis

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

One guiding concept for selectively killing bacteria while sparing the mammalian
host would be for the antibiotic to act against a target present in bacteria but not
found in animals or humans. Which antibiotic types is this true for

A

bacterial cell wall biosynthesis targetters

Enzymes in protein biosynthesis, DNA replication, repair and expression, and folic acid synthesis clearly have mammalian counterparts, but there are enough structural differences between the prokaryotic and eukaryotic synthesis machineries that selective inhibition is achievable.

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

What does the bacterial cell wall do in essence

Why can the cell membrane not do this

A

encases a volume hypertonic to the environment of the organism

Although it is critical to the maintenance of osmotic gradients between the cytosol and the extracellular environment, it is not strong enough to keep the hypertonic sac from rupturing by osmotic shock

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

What do gram positive and negative bacteria both have in common in their cell walls

How does this feature differ between them

A

a peptidoglycan (PG) layer

thicker and mutlilayered in gram +ve
gram +ve also has polymers of teichoic acids associated with it.

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

Describe the crosslinking of the PG layer of the cell wall in bacteria

What is the purpose

Which types of bacteria does this happen in

A

PG, with orthogonal glycan and peptide strands, undergoes enzymatic cross-linking of the glycan strands, by transglycosylase action, and of the peptide strands, by transpeptidase action.

introduce covalent connectivity to the meshwork, impart mechanical strength, and provide the major structural barrier in the bacterial cell wall to osmotic pressure forces

both Gram + and -ve

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

What is the first of the 3 stages of PG synthesis for the bacterial cell wall

A

UDP-NAG + L-Ala + D-Glu + L-Lys (or DAP) + PEP —> UDP N-acetyl
muramyl-tripeptide

D-Ala-D-Ala dipeptide is joined to produce UDP N-acetyl muramyl pentapeptide.

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

How can the formation of UDP-NAM-pentapeptide be inhibited by antibiotics

A

D-cyclosporine (analogue of D-Ala):
inhibits L-alanine racemase, D-Ala-D-Ala synthetase and ligase

Fosfomycin:
inhibits pyruvyl transferase

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

What do the following enzymes do
L-Ala racemase

Pyruvyl transferase

A

L-Ala racemase:
converts L-alanine into D-alanine

Pyruvyl transferase:
that catalyses the transfer of the PEP group to UDP NAG in the production of UDP NAM-tripeptide

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

What is the 2nd of the 3 stages in bacterial PG biosynthesis

A

UDP-NAM-pentapeptide is bound to bactoprenol via a pyrophosphate bridge and then a UDP-NAG is added to it, forming Lipid II

this can then be transferred from the cytosol to the outer leaflet of the phospholipid bilayer

on the outside the the basic repeating subunits can be put together into a polymer by the action of PG transglucosylase and are cross linked by peptidoglycan transpeptidase

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

Where does the first stage of PG synthesis take place

A

inside the cell, at the inner leaflet of the phospholipid bilayer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is bactoprenol phosphate

A

a lipid anchor also referred to as C55 undecaprenyl phosphate

involved in PG synthesis

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

What does the final stage of PG synthesis involve

A

reorientation of bactoprenol pyrophosphate to the inner membrane surface, and its dephosphorylation to
bactoprenol phosphate to be recycled

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Which antibiotic interferes with the final stage of PG synthesis

how

A

bacitracin

forms a very tight complex with Mg2+ and bactoprenol pyrophosphate, this cyclic peptide antibiotic inhibits the regeneration of the lipid carrier, and hence, the biosynthesis of PG.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Which antibiotics inhibit transpeptidation of PG strand cross linking

A

Penicillins, cephalosporins and other beta-lactam antibiotics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

How does the pentapeptide in PG synthesis differ between different bacteria

A

contains diaminopimelic acid (DAP) in Escherichia coli, and L-lysine in Staphylococcus species.

In the transpeptidation
reaction DAP can be used directly, whereas L-lysine is first linked to a pentapeptide interbridge
composed of glycine, alanine and/or serine residues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What type of enzyme are transpeptidases

A

serine hydrolases

active site has serine as a nucleophile

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is the mechanism for the transpeptidase reaction (3)

A
  1. serine attacks D-Ala-D-Ala amide bond -> acyl-O-Ser formed and one is D-Ala released
  2. The acyl-O-Ser enzyme intermediate has the glycan-tetrapeptidyl moiety as the transiently linked acyl group. Hence, the transpeptidase enzyme forms an intermediate in which it is covalently linked to PG
  3. acyl transfer to amino moiety of DAP (or L-Lysine) in a neighbouring pentapeptide, resulting in cross-linking of the two peptide strands with the regeneration of the enzyme for another catalytic cycle.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

How do transpeptidases interact with beta lactam antibiotics

A

Ser adds into the strained four-ring lactam carbonyl and generates an acyl enzyme intermediate in which the b-lactam ring has opened.

The covalent penicilloyl enzyme, thus formed, is very slow to hydrolyse, as H2O is excluded from the active site of transpeptidase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What happens without PG biosynthesis in bacteria treated with beta-lactam antibiotics

A

continued activity of cell wall autolytic enzymes (autolysins), which normally enable re-shaping of PG during cell growth and division, induce lytic death of the cell
Hence, only proliferating cells (in
which autolysins are active) are sensitive to β-lactam antibiotics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

How does vancomycin block cell wall biosynthesis

A

binds to pentapeptidyl tails in the PG repeating unit terminating in D-Ala4-D-Ala5

This substrate sequestration shuts down transpeptidation by making the D-Ala-D-Ala terminus
unavailable to the transpeptidase enzyme

also reduces the accessibility of the PG repeating unit to the transglycosylase enzyme

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Which drugs contain both a beta lactam ring and a thiazolidine ring

A

penicillins and cephalosporins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

How do carbapenems differ from penicillin in structure

A

different stereochemical configuration of constituents on the beta lactam ring

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the ratio of protein to RNA in a ribosome

A

1: 2
protein: RNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the subunits of the bacterial ribosome

A

A bacterial 70S ribosome consists of two subunits, 30S and 50S

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What is the first stage of protein synthesis

A

formation of the initiation complex

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is the first N terminal amino acid for all bacterial proteins

A

formylmethionine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

How does the bacterial ribosome attach to the bacterial RNA (2)

How is the initiation complex completed

A

mRNA becomes attached to the 30S subunit (requires initiation factor 3).

The formylmethionine-charged tRNA then combines with the mRNA-30S ribosomal
complex - requires two additional initiation factors (1 and 2) and GTP

the 50S ribosomal subunit becomes bound to the mRNA-30S-tRNA amino acid complex, the bound GTP is hydrolysed, and the initiation factors are released.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What can the ribosome do once the initiation complex is complete

A

translate the reading frame associated with the initiation codon, with which the formylmethionine-charged tRNA was associated.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Describe protein synthesis in the bacterial ribosome once the initiation complex has been completed

A

The first two tRNAs orient themselves appropriately towards the mRNA, with their attached amino acids adjacent to each other on the surface of the 50S portion of the ribosomes. The two amino acids then become linked by a peptide bond by the peptidyltransferase activity associated with the 50S subunit. The carboxyl group of formylmethionine is linked to the amino group of the second amino acid, and the dipeptide is attached to the second tRNA, which is occupying the A (aminoacyl) site. The tRNA for formylmethionine is now moved from the P (peptidyl) site to the E (exit) site and is released; the tRNA with the attached dipeptide moves from the A to the P site, and the 30S subunit moves one codon along the mRNA. The process of elongation continues with the addition of single amino acid units until a termination sequence signals that the protein is complete

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Name 2 drugs that target the 30S bacterial ribosomal subunit

A

tetracycline

streptomycin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Name 2 drugs that target the 50S ribosomal subunit

A

chloramphenicol

erythromycin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Where does streptomycin bind to the ribosome

which other drug is associated with this site

A

streptomycin bind to the 30S subunit near the A
site for aminoacyl-tRNA binding

Paromomycin also binds near the A site and these drugs’ binding is cooperative

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What happens to bacterial translation in the presence of streptomycin

A

disruption in decoding and translational accuracy, resulting in a decrease in the fidelity of translation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Where does tetracycline bind

what are the chemical reactions involved

A

on the 30S subunit
contains rRNA, not protein, in a groove of 20 Å wide and 7 Å deep

oxygens of internucleotide phosphodiester links in 16S rRNA form electrostatic interactions, directed through a Mg2+ ion to the bottom edge of tetracycline.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

How does tetracycline interfere with translation

A

rotation of aminoacyl-tRNA into the A site would be blocked by tetracycline. The aminoacyl-tRNA would then be prematurely released, terminating that cycle without peptide bond formation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Where does erythromycin bind

A

to E site of the 50S subunit

and interacts with 23S rRNA, allowing about a 6- to 8-oligopeptidyl-tRNA build-up before elongation is blocked and prematurely terminated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are macrolide antibiotics

Name 3

A

bind to E site on 50S subunit
Erythromycin
clarithromycin and roxithromycin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Which antibiotics do macrolide antibiotics interact with

why is this surprising

A

competitive with lincosamide antibiotics (such as lincomycin) that are direct peptidyltransferase inhibitors.

macrolide antibiotics do not directly block the peptide bond-forming step at the peptidyltransferase centre of the 50S subunit

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Which antibiotics are affected by mutations in the 23S RNA of the bacterial ribosome

A

alter the binding of macrolides, lincosamides and streptogramin B family members,

suggesting the partial physical overlap of binding sites of these antibiotics.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What does clindamycin interact with

A

both the A site and the P site of the peptidyltransferase centre, competing with the binding of loaded tRNA molecules at these positions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Where does chloramphenicol bind

A

to 50S subunit

blocks aminoacyl-tRNA interaction with the A site of the peptidyl transferase centre

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What does fusidic acid do

A

binds to and inhibits the elongation factor G, thereby blocking translocation in bacteria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is synercid

A

a synergistic nonribosomal peptide combination (quinupristin plus dalfopristin) that blocks polypeptide translocation at the 50S subunit of the ribosome by binding to 23S rRNA sites partially overlapping with those targeted by macrolides.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Why does stopping transcription of proteins not kill bacteria

A

bacteria are unable to influence the composition or nutrient availability in the extracellular environment. The temporary inability to synthesize cellular proteins is therefore not unusual, and bacteria have evolved stress response mechanisms to survive periods of starvation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Are aminoglycosides bactericidal

A

yes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Name 2 aminoglycoside antibiotics

A

streptomycin (or the newer, less toxic gentamycin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

How do streptomycin and gentamycin affect translation

A

decreases the fidelity of mRNA translation, growing bacteria frequently insert the “wrong” amino acid with a consequent alteration in the activity of proteins. When this happens in membrane proteins, the permeability of the membrane will be affected, resulting in the leakage of small ions followed by larger molecules and eventually by whole proteins from the bacterial cell prior to aminoglycoside-induced death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Is erythromycin bacteriostatic or bactericidal

A

both:

depends on the bacterial species, the drug concentration and the bacterial density.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Which class of enzymes have been used for many years and led to the discovery of Topo II enzymes (specifically DNA gyrase)

A

the coumarins, represented by streptomycete metabolites such as novobiocin and coumermycin A1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Name a fluoroquinolone

A

ciprofloxacin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Why is ciprofloxacin so widely used

A

because of its
activity against both gram-negative and gram-positive bacteria in urinary tract
infections, osteomyelitis, community-acquired pneumonia, and gastroenteritis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

How do newer quinolones differ from older fluoroquinolones

A

newest generation of quinolones, such as gatifloxacin, have increased potency against gram-positive pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Why are Topo enzymes essential for cell viability

A

changes in supercoiling of DNA are required during replication and gene expression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What are etoposide and camptothecin used for

A

mammalian Topo II inhibitors

used in cancer to kill rapidly dividing tumour cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

what is topo IV

A

Like DNA gyrase, topo IV is a bacterial type II DNA topoisomerase, but unlike the DNA gyrase it cannot supercoil DNA.

Instead, topo IV carries out the ATP-dependent relaxation of DNA and is a more potent decatenase than DNA gyrase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

How do quinolones affect DNA enzymes

A

affect the double-strand
cleavage/double-strand religation equilibrium in DNA gyrase and topo IV catalytic cycles, such that the cleaved complex accumulates

may speed up the double-cleavage step of bound DNA or selectively slow the double-religation step

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is the killing action of quinolones

A

As the quinolone-covalent DNA gyrase-doubly cut DNA intermediate accumulates,
it blocks the progression of the replication forks,

DNA repair machinery is recruited which, upon failure, turns on signalling pathways that lead to rapid, directed cell death (apoptosis).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What does rifampin do

A

binds in a very
tight, but non-covalent manner to an allosteric site on the b subunit of the DNAdependent RNA polymerase at a ratio of one mole of drug per mole of enzyme.

directly blocks the elongating RNA chain at the di- or tri-nucleotide stage by binding in the DNA/RNA tunnel associated with the b subunit (analogous to the binding of macrolide antibiotics to the protein exit tunnel in the 50S ribosomal subunit). As a result of the binding, rifampin inhibits the initiation of RNA synthesis, but synthesis in progress at the time of drug exposure is not affected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the 2 categories of antibiotic that interact with DNA

A

those that interact noncovalently with DNA and those that form covalent bonds with DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Name some drugs which intercalate DNA

A

Many rigid planar polycyclic antibiotics (e.g., daunorubicin and actinomycin D) and synthetic compounds (e.g. ethidium)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Why is the planar character of daunorubicin important

A

allows intercalation between the adjacent stacked base-pairs of the double DNA helix

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

How do intercalating drugs disrupt replication and DNA transcription

A

insertion event is based on a preliminary local unwinding of the double helix to produce spaces between the stacked pairs into which the planar polycyclic molecules can move

this partial unwinding affects the molecular dimensions of the major and minor grooves in the DNA, and therefore the interaction of template DNA with DNA polymerases, RNA polymerases and transcription factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What are Bleomycins

Which organisms do they particularly affect

A

a family of metal-chelating glycopeptide antibiotics

especially toxic to Gram-positive bacteria and mammalian cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What does mitomycin C do in bacteria

A
alkylates DNA (at
guanine bases at GC positions in complementary DNA strands)

Thus, mitomycin induces the cross-linking of two Gs, one in each strand of the double helix, and prevents strand separation during DNA replication and transcription.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What is the chemical interactions underpinning bleomycins’ action

A

based on the interaction between O2 and the bound iron,

which generates superoxide and hydroxyl radicals causing single- and DSB in DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What is Co-trimoxazole

A

a combination drug of sulfamethoxazole and trimethoprim

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What is the class of synthetic chemicals in the longest use as effective antibiotics

A

sulfa drugs, first tested in the 1930s as bacteria-killing molecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What does sulfamethoxazole block

A

dihydropteroate synthase (DHPS)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What does trimethoprim inhibit

A

dihydrofolate reductase (DHFR)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What are DHPS and DHFR important for

A

synthesis of tetrahydrofolate, a methyl carrier coenzyme required for the biosynthesis of dTMP, and hence, DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Why is folate metabolism different in eukaryotes and bacteria

how is this reflected in their enzymes

A

Bacteria have to make the folate skeleton de novo, while eukaryotes can scavenge
folate from dietary sources and transport it into cells

DHPS is totally absent from mammals while bacterial versus mammalian DHFR have enough structural differences that selective inhibition can be achieved

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Why is it useful for a DHPS inhibitor and a DHFR inhibitor to be used in combination

A

while sulfonamides shut off de novo synthesis of folate, folate pool levels would take several bacterial generations to decline, giving a slow killing mechanism.
Addition of trimethoprim traps the folate coenzyme molecules in the useless dihydrofolate form after each cycle of dTMP synthesis, leading to a rapid depletion of the tetrahydrofolate form of the coenzyme

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Name 3 bacterial nonribosomal peptide antibiotics

What do they all act on

A

valinomycin,
gramicidin A,
polymixin

act on the cytoplasmic membrane.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

How is valinomycin made

How is it toxic

A

produced by Streptomyces species,

exerts toxicity by its
ionophoretic capacity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

Describe the MOA of valinomycin

A

contains three repeating units of (L-lactate)-(Lvaline)-(D-hydroxyisovalerate)-(D-valine), which form a circular structure

A dehydrated K+ ion is coordinated precisely and specifically to carbonyl groups in the hydrophilic interior of valinomycin.

Because valinomycin is electroneutral, it carries the single (+) charge of the bound K+ ion.

As valinomycin diffuses across the membrane, it functions as a K+ uniporter.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What is the MOA of monensin

A

The ionophores nigericin and monensin lose a proton as they bind
K+ or Na+, respectively, and function as K+-H+ or Na+-H+ antiporters
(similar to valinomycin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe gramicidin A

What is its MOA

A

a hydrophobic linear polypeptide antibiotic consisting of 15 aa and a carboxyterminal ethanolamine

Upon dimerisation in the membrane, this molecule forms a transmembrane ion channel that permits passive diffusion of monovalent cations with diameters of up to 5 Å

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What is the net charge of polymixin

What is its structure

A

+5

a cyclic amphipatic protein

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

How is polymixin proposed to work

A

associate with the negatively charged phosphate head group region on the outer surface of the membrane, and then, to aggregate into micelle-like complexes which bind lipids and affect the permeability of the cytoplasmic membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

It is possible that all lipopeptide antibiotics have some component of
membrane penetration and membrane disruption to their bioactivity. Give an example

A

the cyclic lipopeptidolactone daptomycin, in complex with Ca2+ ions, may exert part of its antibacterial activity through such membrane-seeking, surface-active behaviour.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What makes antibiotics that interact with the bacteria cell membrane toxic

What is a problem with a lot of these drugs

A

dissipation of transmembrane ion gradients, which disturb ion homeostasis and the energy metabolism, and induce leakage of macromolecules from the bacteria affected

lack of selectivity,
(presence of a phospholipid bilayer is not restricted to bacteria.)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Which antifungal agents target cell membranes

How are they selective

A

polyenes amphotericin B and nystatin

exploit differences in the sterol composition of the fungal plasma membrane (ergosterol) and mammalian plasma membrane (mainly cholesterol)

bind preferentially to
ergosterol, facilitating the formation of pores for ions and macromolecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Name some antibiotics which indirectly affect the barrier function of membranes

A

Antifungal synthetic triazoles (eg fluconazole) and imidazoles (eg miconazole) inhibit enzymes involved in ergosterol biosynthesis.

Depletion of ergosterol alters fluidity of the membrane, thereby affecting permeability and the activity of membrane-associated enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Where are parasitic diseases most prevalent

A

under conditions of crowding, poverty, and poor sanitation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What is the most important distinction of pharmacological significance to the
treatment of parasitic infections

How do drugs act between these groups

A

whether the infecting organism is a unicellular protozoan, or a multicellular helminth

Most antiparasitic agents have a relatively broad spectrum of activity within one of these groups, but virtually no crossover activity in the other group

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Why are the molecular mechanisms of antiparasitic drugs often not known

A

difficulties in culturing parasites under controlled experimental conditions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What are the 2 broad categories for the MOA of anti-parasitic drugs

A

acting on (i) cellular integrity and (ii) biosynthesis of essential cofactors and macromolecules.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What do trivalent organic arsenicals react with

Why are they toxic

A

hiol groups (especially dithiols) in cofactors (e.g., lipoic acid; this
inhibits lipoic acid-dependent enzymes) and enzymes (e.g. pyruvate kinase and
phospofructokinase),

hence, ultimately inhibit ATP synthesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is melarsen oxide

A

an arsenical generated from prodrug melarsoprol are preferentially toxic for Trypanosoma species.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What makes melarsen oxide selective against Trypanosoma species

A

probably due to selective uptake in these organisms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Which parasites does suramin attack

How does it work

A

Trypanosoma

inhibits glycerol-3-phosphate oxidase
and NAD+-dependent glycerol-3-phosphate dehydrogenase

interfere with the reoxidation of NADH and inhibit ATP synthesis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Which antifungals act against Leishmania (2)

Why

A

amphotericin B and
miconazole

membrane of certain Leishmania species contains ergosterol, generated
in an ergosterol biosynthesis pathway

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What do trophozoites of Plasmodium do when in RBCs

A

digest hemoglobin to obtain amino acids for biosynthesis (haem is released as a byproduct)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Where does digestion of haemoglobin occur in trophozoites

A

inside the food vacuole of the parasite

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is the toxic waste problem that trophozoites face

A

If the released heme were allowed to accumulate within the food vacuole, the heam level could easily reach 200 – 500 mM!

oxidation of haem iron results in the production of ROS
(superoxide anion, H2O2, and hydroxyl radicals), which would harm the parasite.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

How do trophozoites prevent haem accumulation

A

by polymerizing the haem into nontoxic

crystals of hemozoin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How does chloroquine work

which drug works in a similar way

A

inhibits haem polymerisation reaction in trophozoite food vacuoles, causing haem to build up

mefloquine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

How does artemisinin extert antimalarial activity

A

y through the generation of highly reactive

organic free radicals.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How do causal prophylactic drugs block malaria development

A

block the link between the exo-erythrocytic stage and the erythrocytic stage, and thus prevent the development of malarial attacks

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What is true causal prophylaxis in malaria

How do we do this

A

the prevention of infection by the killing
of the sporozoites on entry into the host

not feasible with the drugs at present in use, although it may be achieved in the future with vaccines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is the antiparasitic drug sulfadoxin

A

p-aminobenzoate analogue

competitively inhibit the action of dihydropteroate synthase (DHPS)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is sulfadoxin usually given in combination with when treating a parasitic infection

What is this combination called

Which parasites can it treat

A

trimethoprim analogue pyrimethamine (blocks DHFR)

Fansidar

Plasmodium falciparum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Why is the potential for finding drugs that target parasitic protein synthesis limited

However 2 antibiotics show activity against certain protozoa. What are these drugs?

A

their protein synthesis is essentially similar to mammals

tetracyclines and lincomycins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Why is it difficult to develop antiviral drugs that act only on infected cells

A

Viral replication depends on many of the same cellular processes that operate in normal uninfected mammalian cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the genome of influenza like

what contains the genome

A

eight segments of negative-sense single-stranded RNA

nucleocapsid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

How is the influenza genome affected by pH

A

The nucleocapsid encasing the genome contains an ion channel

protein, M2, that triggers uncoating of the genome when the virus is exposed to low pH.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are the roles of influenza’s neuraminidase

A

helps prevent viral aggregation,

facilitates release from the host cells, and may have a role as a virulence factor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

Name 2 major drugs used against influenza A

What are their MOAs

A

amantadine and rimantadine

At an early step in viral replication, they
block the function of the M2 channel protein. At a later stage, they interfere with hemagglutinin processing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

How does amantadine work

A

At an early step in viral replication, block the function of the M2 channel protein.

At a later stage, interfere with hemagglutinin processing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is sialidase

A

another name for neuraminidase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What is zanamivir used to treat

What is its MOA

What is another drug that works the same way

A

Influenza A and possibly B

Inhibits neuraminidase, enhancing viral aggregration and inhibiting release from host cells
also reduces viral movement in the upper respiratory tract

oseltamivir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

Compare oseltamivir and zanamivir

A

both inhibit neuraminidase in Influenza A and (possibly) B

Zanamivir is an active drug whereas oseltamivir is an ethyl ester pro-drug, which is cleaved by esterases in the plasma and in cells of the gut upon the adsorption of oseltamivir.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What do all available anti-herpes agents target

A

the virally encoded DNA polymerases

that replicate the double-stranded DNA genome of these viruses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

How do viral DNA pol work

A

in the same manner as cellular DNA polymerases, (i.e., join the 5’-OH group of the base being added to the 3’-OH group of a 2’-deoxyribose sugar in the polymerized strand of DNA)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is the origin of the ‘acyclo’ in acyclovir

A

purine analogues used against herpes viruses all lack the cyclic sugar of 2’- deoxyguanosine and acyclovir was the first drug of this ring-lacking class

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What happens when acyclovir is taken up by cells (3)

A

monophosphorylated by a herpesvirus-encoded thymidine kinase. Cellular enzymes then convert it to the triphosphate form.

Aciclovir triphosphate competitively inhibits viral DNA polymerase.

In addition, it can be added by DNA polymerase to the 3’-OH of a strand of DNA, but it has no corresponding 3’-OH to which additional nucleotides may be added. This terminates the DNA strand and permanently inactivates the enzyme

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

Why is acyclovir selective against viral cells

A

(1) only virally infected cells have the thymidine kinase required to monophosphorylate the drug,
(2) the drug preferentially binds to the virally encoded DNA polymerase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

Does acyclovir affect healthy cells

A

yes but is 30 times more potent against the viral DNA polymerase that the host
enzyme

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What is ganciclovir

What happens to it in infected cells

A

competitive inhibitors of DNA polymerase, but they have 3’-OH moieties and will permit chain extension

monophosphorylated by a phosphotransferase encoded by cytomegalovirus (CMV)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What is the drug of choice against CMV

A

Ganciclovir is more effective that aciclovir against CMV-infected cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Name a pyrimidine analogue used as an antiviral

A

Cidofovir

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

How does cidofovir work

A

is a nucleoside phosphonate analogue of cytosine, which is converted to a diphosphoryl derivative that selectively inhibits the DNA polymerase of CMV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What is the antiviral drug foscarnet

How does it work

A

an organic analogue of pyrophosphate

selectively binds to viral DNA pol of CMV and herpes simplex virus (HSV) and others and prevents the cleavage of pyrophosphate from nucleoside triphosphate during DNA
polymerization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What is contained in a HIV virion (for MoDA purposes)

A

two copies of a single-stranded RNA genome, a reverse transcriptase, and an aspartic protease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What happens to the HIV RNA early in the viral replication cycle

A

reverse
transcriptase converts the RNA into double-stranded DNA, which is then integrated
(via integrase) into the host cell DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What is the fidelity of the HIV reverse transcriptase like

What does this lead to

A

poor

frequent transcription errors, and a high degree of sequence variation among the viral genome copies that are produced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

How are HIV reverse transcriptase inhibitors divided

A

into Non-nucleoside reverse transcriptase inhibitors (NNRTI) or Nucleoside reverse transcriptase inhibitors (NRTI)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

Name a NNRTI

what is the MOA

A

nevirapine

binds to the target enzyme near the catalytic site and denature it.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

Name a NRTI

A

zidovudine

azidothymidine, AZT - a thymidine analogue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

How do NRTI drugs work

A

bind to the target enzyme by mimicking naturally occurring nucleosides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

How do NRTIs and NNRTIs differ in action

A

NRTIs (but not NNRTIs) must be phosphorylated by host cell enzymes before becoming active

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

How do NRTIs terminate synthesis

What part of the viral life cycle are they important for

A

When incorporated into DNA, these compounds lack 3’-OH groups.

Reverse transcription is an early event in the replication cycle, so these agents have no effect on a cell in later stages of viral infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What is often used in conjunction with AZT

Why is this given with pyrimidine analogues

A

Hydroxyurea

inhibits ribonucleotide reductase.
This decreases the intracellular pool of pyrimidine nucleotides, and thereby potentiates the effect of pyrimidine analogues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

How many HIV protease inhibitors are there currently

Name 2

What are they approved for

A

6

saquinavir and ritonavir

clinical use against HIV-1 and HIV-2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What is the purpose of the Asp protease in HIV

A

the mRNA transcribed from the provirus is translated into two biochemically inert polypeptides, termed gag proteins. HIV protease then converts these polypeptides into functional proteins by cleavage at the appropriate positions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Why is the HIV protease a good target

A

this protease does not occur in the host

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

When does HIV protease usually perform its function

What would happen without it

A

as the virus is budding from the cell membrane or shortly
thereafter.

Without these cleavages, the newly produced virus is not infectious.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

Name something of note that is present in the HIV protease active site

A

HIV protease contains two aspartyl residues in its active site

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What is viral interference

A

biochemical changes that inhibit viral propagation, brought about by inteferons

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What are the 3 classes of interferon

Which have antiviral activity

A

IFN-a, IFN-b, and IFN-g

a and b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What do antiviral Interferons bind to and what does this cause

A

specific ganglioside receptors on host cell membranes and promote in host cell ribosomes the production of enzymes that inhibit the translation of viral mRNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

Name 2 inteferons and state their use in treating viral infections

A

IFN-a-2a: used in treatment of hepatitis B infections and AIDS related Kaposi sarcomas.

IFN-a-2b: used for hepatitis C.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

Name 3 uncommon cancers that have whose treatment has rapidly advanced through development of cytotoxic drugs

A

childhood cancers,
lymphomas
teratomas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

When was nitrogen mustard introduced into clinical practice

What was quickly established

A

1946

The effectiveness of this class of compounds in producing regression in lymphomas

also gastrointestinal and haematological toxicity they produced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What was aminopterin shown to do in 1947

What was this quickly followed by

A

produces remission in acute leukaemia

(it is an analogue of folate)

production of methotrexate in ‘49

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

How do most anticancer drugs work generally

A

reducing rate of cell growth and division in the tumour

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

Give 3 ways to block cancerous cell proliferation

A
  1. By preventing effective DNA replication through direct binding to nucleobases or impairing the DNA synthesis machinery,
  2. By damaging the mechanisms of cell division such as formation of the mitotic spindle,
  3. By blocking the pathways involved in cell growth that are activated by signals such as growth factors or hormones.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

Name 2 nitrogen mustards

A

melphalan

cyclophosphamide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

Which anticancer agents bind directly to DNA

name a drug of each type

A

nitrogen mustards (melphalan),

nitrosoureas (lomustine),

aziridines (mitomycin C),

Pt compounds
(cisplatin).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What is the important chemical feature of nitrogen mustard

A

its 2 chloroethyl side chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

How does nitrogen mustard crosslink DNA

A

One of the
chloroethyl side chains undergoes a cyclization and forms an immonium-ion
intermediate with the release of a chloride
This strained three-membered ring is highly reactive and can attack the 7-nitrogen group of guanine.
This repeats with the other chloroethyl side chain with another G in DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

What are the possible results of the reaction of nitrogen mustard with DNA

A

may result in cross-linking between DNA strands
or linking between bases within the same strand of DNA - inhibiting replication and gene expression

If the second side chain reacts with
H2O instead of a guanine, a monoalkylated guanine is produced, which bonds incorrectly with T, causing a GC>AT transition

In addition, the DNA containing the drug adduct is recognized by DNA repair systems, and strand scission may occur as a result of endonuclease attack when the cell attempts to repair the alkylated DNA.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

Where does the general toxicity of nitrogen mustards come from

A

Alkylating agents have less favoured reactions with other nucleophilic
groups in DNA, RNA and protein

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What is melphalan

A

L-phenylalanine mustard

a synthetic nitrogen mustard

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What is the benefit of using melphalan

A

as phenylalanine is a precursor of melanin, it is accumulated in melanomas and thereby produces a relatively selective effect.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What is the most commonly used anticancer alkylating agent

Why is this

A

cyclophosphamide

has a a broad application in cancer chemotherapy (e.g. lymphoid tumours, and carcinomas of the breast, lung, ovary and endometrium).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

Briefly state2 facts about the PK of cyclophosphamide

A

well absorbed orally and needs to be metabolised in the liver by the cytochrome P450 system to become activated to a phosphoramide mustard.

180
Q

Name a nitrosourea

Which features give it anticancer applications

A

lomustine

has a chloroethyl chain and is both a alkylating and carbamoylating agent

181
Q

What does it mean to say that a drug is both an alkylating and carbamoylating agent

Which anticancer drugs have these properties

A

they react with a
variety of groups to attach an alkyl (R-CH2-) or a carbamoyl (R-N-CO-) moiety

nitrosoureas

182
Q

Can nitrosoureas produce DNA crosslinks

A

yes
all of the nitrosoureas (even those with only one chloroethyl side chain) can produce interstrand cross-links in duplex DNA in which N7 and O6 positions in guanine are preferred sites of attack.

183
Q

What is mitomycin C used for

A

both antibiotic and anticancer (inhibits DNA synthesis via cross linking and alkylation)

184
Q

Give a fact about the kinetics of mitomycin C

A

has no effect on purified DNA in vitro unless a cell extract is added - activated by chemical or enzymatic reduction of the quinone group

185
Q

What kind of anticancer drug is mitomycin C

A

aziridines

186
Q

What is the MOA of mitomycin C

A

After reduction of the quinone moiety, a tertiary methoxy group is spontaneously eliminated.

The aziridine ring is then broken, creating a semiquinone radical that reacts with nucleophilic groups in DNA (most commonly guanine).

Then, intramolecular displacement of the carbamate group yields the cross-linked DNA-drug adduct.

187
Q

Which drug is only active in cis form

A

Cis-diamine dichloroplatinum (cisplatin)

188
Q

What are the principal sites of reaction of cisplatin at physiological pH

A

N7 atoms of guanine and adenine

189
Q

True or false

cisplatin only works in vivo

A

false
regardless of whether it is purified DNA, intact cells, or tumour-bearing patients that are exposed to cisplatin, the principal coordinate is an intrastrand cross-link formed by binding of the drug to two neighbouring guanines (pGpG).

190
Q

What was found in a trial of the DNA adducts were analysed in white cells of cancer patients treated with cisplatin?

A

65% of adducts represented intrastrand cross-links on pGpG, 22% on pApG (but not pGpA) sequences, and 13% a mixture of other adducts

191
Q

How doese the binding of cisplatin inhibit DNA synthesis/ gene expression

A

intrastrand cross-linking induces major bending of the DNA duplex towards the major groove, together with the physical block provided by the platinum adduct on the template strand

192
Q

Name 2 types of drugs that non covalently bind to DNA in cancer treatment

A

anthracyclines

actinomycins

193
Q

Name 2 anthracyclines

A

doxorubicin, daunorubicin

194
Q

Name ant actinomycin

A

actinomycin D

195
Q

What is the in the structure of anthracyclines and actinomycins that allows them to noncovalently bond to DNA

A

usually have planar aromatic ring structures that allow them to intercalate between stacked nucleobases at the centre of duplex DNA

196
Q

What does intercalation of anthracyclines and actinomycins cause in DNA

What does it affect (3)

A

local unwinding on the helix with changes in the geometry of the minor and major groove

binding of DNA and RNA pol, and transcription factors.

197
Q

What is the basis of the severe side effects of anthracyclines

A

generation of free radicals due to the presence of
hydroxyquinone moiety

while these free radicals attack DNA and can induce DNA cleavage, radicals can also cause lipid peroxidation, e.g. in cardiac tissue

198
Q

How was the free radical problem with anthracyclines overcome

A

Anthracycline analogues with a modified structure, (eg mitoxantrone) lack this property of quinone-type free radical generation.

199
Q

What is mitoxantrone

A

anthracycline analogue which lacks the property of quinone free radical generation

200
Q

How can antimetabolites interfere with production of DNA and RNA (2)

A

(i) inhibition of normal precursor production, and

(ii) substitution of purines and pyrimidines in nucleic acid synthesis.

201
Q

Name a folic acid antagonist in clinical use

A

methotrexate

202
Q

What is the antifolate action of methotrexate

A

inhibition of dihyrofolate reductase (DHFR)

competes with folic acid for active transport into mammalian cells

203
Q

Name a) an antibiotic, b) antiprotozoal and c) an anticancer drug that inhibits DHFR

A

a) trimethoprim
b) pyrimethamine
c) methotrexate

204
Q

Antifolate medication can lead to folate depletion in healthy cells. How can mammalian cells be saved from this

A

by also giving Leucovorin (N5

-formyl-tetrahydrofolate)

205
Q

How does Leucovorin work

A

can be readily converted to other reduced folic acid derivatives (e.g. tetrahydrofolate), and thus has vitamin activity that is equivalent to folic acid.

However, since it does not require the action of dihydrofolate reductase for its conversion, its function as a vitamin is unaffected by inhibition of this enzyme by drugs eg methotrexate.

Leucovorin, therefore, allows for some pyrimidine synthesis to occur in the presence of dihydrofolate
reductase inhibition,

206
Q

Why does Leucovorin not simply inhibit methatexate

A

Due to the difference in tetrahydrofolate requirement between normal cells and rapidly proliferating tumour cell (low versus high, respectively), leucovorin is able to reduce the toxicity of
methotrexate in normal cells only

207
Q

What are the most importnat members of the group of anticancer drugs that directly block thymidylate synthetase

A

pyrimidine antagonists (dUMP analogues)

208
Q

name a pyrimidine antagonist

How does it kill cancer cells

A

5-fluorouracil

Although it is also incorporated into rRNA and mRNA, with disruption of further transcription, intracellular distribution and translation, inhibition of thymidylate synthetase appears to be its primary cytotoxic mechanism

209
Q

Name 2 purine antagonists

what are they analogues of

What do they cause

A

6-mercaptopurine and 6-thioguanine are analogues of
hypoxanthine and guanine, respectively

cause nucleotide synthesis inhibition as well as being incorporated into nucleic acids (after their activation to triphosphate nucleotides).

210
Q

Name 3 inhibitors of mammalian Topo II

A

etoposide

daunorubicin and doxorubicin

211
Q

Which drugs inhibit mammalian Topo I

A

Camptothecins (derived from the Chinese tree Camptotheca) and topotecan

212
Q

Which drugs were extracted from the periwinkle Catharanthus roseus and the bark of the Pacific yew Taxus brevifolia

What did they cause in animals

How did they do this

A

Vinca alkaloids (e.g. vinblastine and vincristine) and taxol (paclitaxel) respectively

granulocytopenia

poison the mitotic spindle by acting on microtubule formation

213
Q

What is the major component of microtubules

A

tubulin dimer arranged head to tail in linear protofilaments

214
Q

What is the tubulin dimer

A

a protein complex containing two non-identical (alpha and beta) subunits

215
Q

describe the structure of a microtubule

A

13 protofilaments together form a hollow structure with a minus end which is anchored to an organising centre and a plus end where growth or shrinkage of the microtubule takes place

216
Q

Are microtubules stable

A

they are in cilia and neuronal axons but in chromosome segregation are very labile

217
Q

How does the microtubule population change with changing conditions

A

Individual microtubules can oscillate between polymerization and depolymerisation, and the net status of the microtubules population is very sensitive to factors affecting the equilibrium between free tubulin dimers and assembled polymers

218
Q

What may affect the equilibrium of microtubule formation and breakdown

A

availability of numerous factors including tubulin, GTP, Mg2+, and non-tubulin protein

219
Q

How do vinca alkaloids work

A

bind to free tubulin dimers. Although these agents do not all share the same binding site, their interactions with the tubulin dimer prevent microtubule assembly, and hence, result in the disappearance of microtubules

220
Q

How does taxol work

A

it is a taxane drug that disrupts the equilibrium between free tubulin dimers and microtubules by shifting it in the direction of assembly rather than disassembly.

As a result, taxol treatment causes both the stabilization of ordinary cytoplasmic microtubules and the formation of abnormal bundles of microtubules.

221
Q

Name a cancer that responds to body hormone levels

A

Many breast

cancers grow more rapidly in the presence of female steroid hormones

222
Q

How can you predict whether a breast cancer will respond to hormone therapy

A

by the amount of estrogen and progesterone receptors in the tumour tissue.

223
Q

What is tamoxifen

A

an anti-oestrogen that competitively binds to the estrogen receptor,
but with a lower affinity than estrogen

224
Q

What happens when tamoxifen binds to the oestrogen receptor

A

complex is translocated to the nucleus, transcription of estrogen-responsive genes involved in the development and growth of breast cancers is attenuated.

225
Q

How many breast cancers can respond to tamoxifen

A

Approximately 70% of breast cancers are positive for estrogen and progesterone receptors and can respond to tamoxifen

226
Q

What is an issue with tamoxifen

A

can exert oestrogen agonist effects in healthy tissue eg bone and uterus

227
Q

Name a tamoxifen analogue that lacks the oestrogen agonist properties

A

toremifene

228
Q

What is aromatase

A

converts androgen precursors into estradiol

229
Q

Name an aromatase inhibitor

What is it used for

A

anastrozole

treatment of estrogen-dependent breast cancer either instead of, or after
treatment with tamoxifen.

also used in post-menopausal women with breast cancer to prevent the formation of estrogens (from androgen precursors) at
peripheral sites such as muscle and fat tissue

230
Q

What does GnRH do

A

binds to receptors in the pituitary gland and stimulates the production of luteinising
hormone (LH) and follicle-stimulating hormone (FSH)

231
Q

What happens to LH secretion with continuous GnRH production

A

immediate increase in LH and FSH followed complete inhibition of their release

232
Q

What does goseraline do

A

cause a biochemical castration (no synthesis of testosterone) by binding very strongly to the GnHR receptor.

233
Q

What do anti-androgens do

name one

A

antagonise the interaction of endogenous androgens at their nuclear receptors and are also used against prostate cancer.

flutamide

234
Q

Name a drug that utilises the fact that hormones can directly inhibit growth

A

prednisone,

has a lymphocytolytic effect and are useful against leukemias.

235
Q

What is Rituximab

A

a chimeric monoclonal antibody against CD20

236
Q

What does Rituximab target

A

CD20, a protein present on

the surface of many transformed B-cells (lymphocytes) in Hodgkin’s disease and non-Hodgkin’s lymphoma

237
Q

What has Rituximab been shown to do

A

lyses CD20 cells through antibody-dependent cell-mediated cytotoxicity, activation of the complement cascade, and introduction of apoptosis.

238
Q

What was the first monoclonal Ab approved against cancer

A

rituximab

239
Q

What is Trastuzumab

A

a humanised monoclonal antibody against the

human epidermal growth factor receptor 2 (HER2).

240
Q

How is HER2 related to cancer

A

over-expressed in 25 to 30% of human breast cancers and is associated with a poorer prognosis

241
Q

How is trastuzumab administered

A

Trastuzumab is administered intravenously, once weekly, either alone or in
combination with classical anticancer agents for metastatic breast cancer that
overexpress HER2.

242
Q

Name 4 common side effects of trastuzumab

A

chills, asthenia, fever, and nausea.

243
Q

What is a risk with trastuzumab

A

Rarely, trastuzumab can cause cardiac dysfunction.

However, the risk of cardiac dysfunction increases significantly if trastuzumab is given together with other cardiotoxic agents such as anthracyclines.

244
Q

What is bevacizumab

A

a humanised monoclonal antibody that binds to and inhibits the human vascular endothelial growth factor (VEGF)

245
Q

What is VEGF

Why is it important for tumours?

A

a soluble protein that plays an important role in inducing blood vessel formation,

thereby allowing
tumours to grow beyond a few millimetres in size.

246
Q

What is the monoclonal antibody against VEGF

How is it administered

For which cancer in particular
Which other cancers has it been shown to be effective against

A

bevacizumab

in combination with 5-fluoro-uracil

metastatic colorectal cancer
NSCLC and breast cancer

247
Q

Give some side effects of taking bevacizumab (4)

A

Due to its anti-angiogenic action, bevacizumab
can interfere with wound healing and increase the risk of bleeding or gastrointestinal
perforation

hyper tension
proteinuria

248
Q

What kind of antibody is Cetuximab and what does it bind

A

a human/mouse chimeric monoclonal antibody that binds to the extracellular domain of EGFR

249
Q

What does EGFR consist of

A

a transmembrane glycoprotein composed of an extracellular ligand-binding domain, a short transmembrane domain, and an intracellular domain that has tyrosine kinase activity

250
Q

What does binding of EGF to EGFR lead to

A

induces conformational changes within the receptor and increases the activity of associated tyrosine kinases.

This results in autophosphorylation and increased biological activity, which might include cell proliferation and/or cells differentiation.

251
Q

Why is the EGFR important for cancer understanding

A

Abnormal EGFR expression, either through a mutation or over-expression, has been demonstrated in many malignancies.

252
Q

How can inhibition of EGFR in cancer cells be achieved

A

by preventing ligand binding with an anti-EGFR antibody or by inhibiting EGFR-associated tyrosine kinases.

253
Q

What does Cetuximab do (3)

A

binds to the extracellular part of EGFR and inhibits it

reverses the resistance of colorectal cancer to the topoisomerase inhibitor, irinotecan

used in combination with irinotecan in the treatment of EGFR overexpressing metastatic colorectal cancer in patients who are refractory to irinotecan.

254
Q

What are the monoclonal antibodies you need to know for cancer treatment

A

cetuximab
bevacizumab
Trastuzumab (sold as Herceptin)
Rituximab

255
Q

What are the 4 key small molecule inhibitors that are

A

erlotinib
imatinib mesylate
toceranib
mastinib

256
Q

What is erlotinib and gefitinib

What have they been approved to treat

What are some side effects

A

orally active, potent, and selective quinazoline
derivatives that inhibit EGFR tyrosine kinases

advanced NSCLC

rashes and diarrhea

257
Q

Which experiments have shown the effects of erlotinib

A

Growth inhibition and tumour regression have been seen in human xenograft models in lung, prostate, breast and colorectal cancers

258
Q

What forms the Philadelphia Chr

A

reciprocal exchange of genetic material between the long arms of chromosomes 9 and 22. As a result of this translocation, a fusion gene, bcr-abl, is formed

259
Q

Describe the activity of BCR-ABL

A

tyrosine kinase activity and is

constitutively active. It is thought to be the main cause of CML

260
Q

Which drug can treat CML

A

Imatinib mesylate

an agent that specifically inhibits the BCR-ACL protein kinase activity, and it is active in chronic and blast phases of CML

261
Q

What are the 2 uses of imatinib mesylate

A

inhibits BCR-ABL in CML

inhibits c-kit which is overexpressed in patients with gastrointestinal stromal tumours (GISTs)

262
Q

What kind of actrivty does c-kit have

A

tyrosine kinase activity

which is commonly over-expressed in patients with GISTs

263
Q

Name 2 tyrosine kinase inhibitors that are used in veterinary medicine

what do they do

Are they relevant to real medicine

A

toceranib
mastinib

used in the treatment of non-resectable grade II – III mast cell tumours (mastocytomas) in connective tissue in dogs

yes - both drugs used for c-kit mutations

264
Q

What are Grade II and III cancer cells

A

Grade II cells are intermediately differentiated with a potential for locally invasive metastasis.

Grade III cells are poorly differentiated or undifferentiated with a high potential for metastasis.

265
Q

What are some other small molecular ligands that are being generated against new drug targets in
mammals

A

check-point protein kinases (referred to as mammalian target of rapamycin, mTOR), telomerase, and epigenetic drug targets, such as histone deacetylases (HDACs)

266
Q

What was the causative agent of plague

Which antibiotics is it susceptible to

A

Yersinia pestis

uniformly susceptible to the antibiotics streptomycin, chloramphenicol, and tetracycline, but has been reported to have high level resistance to multiple antibiotics

267
Q

Why is TB now a problem again

A

a result of the emergence of Mycobacterium tuberculosis strains resistant to multiple antiTB drugs.

268
Q

Define drug resistance

A

a condition in which there is insensitivity or decreased sensitivity to drugs that ordinarily cause inhibition of cell growth or cell death

269
Q

What does intrinsic resistance refer to

A

a microbes inherent insensitivity to a drug

eg if the organism lacks the receptor for a drug

270
Q

Which antifungal drugs do bacteria have an inherent resistance to

Why

A

antifungal polyenes

polyenes require presence of ergosterol in the membrane, which bacteria do not have

271
Q

Describe the intrinsic resistance of fungi to rifampin

How can we make the fungus more susceptible

A

Although fungal DNA-dependent RNA polymerase is inhibited by rifampin, this drug is not particularly effective against fungi because the drug does not readily pass through the fungal cell envelope to its site of action.

simultaneous exposure to polyene antibiotics, which facilitates drug entry

272
Q

Name a bacteria that has a significant intrinsic resistance to antimicrobial agents compared to other bacteria

Why is this

A

TB

because of the high content of mycolic acids in a complex lipid layer outside of their peptidoglycan, which is impermeable to many drugs

273
Q

What is a first line agent against TB

why

How well does this drug work on other bacteria

A

Isoniazid

inhibits synthesis of mycolic acids for the lipid layer

Other bacteria, which do not use mycolic acids in their cell envelope, have a high intrinsic resistance against this drug.

274
Q

What is acquired antibiotic resistance to

A

when populations of microorganisms that are initially sensitive to a drug undergo a change so that they become less sensitive or insensitive.

275
Q

Does acquired resistance always mean the patient can no longer be treated by that drug

A

no
resistance can be slight but often organisms become resistant to any clinically achievable concentration of
drug.

276
Q

How does the abundance of resistant organisms in a population change as the treatment continues

What is this called

A

increases relatively as non resistant microbes are killed off

selection - continued use of the drug exerts selection pressure on the population

277
Q

What are the 3 ways microbes can become insensitive to antibiotics

A

(i) Enzymatic inactivation of drugs,
(ii) replacement, amplification or modification of drug
targets,
(iii) decreased drug uptake or increased efflux of drugs

278
Q

Which kind of antibiotic is resistance via enzymatic inactivation of drugs a real problem for?

What does this reflect

A

natural product antibiotic classes but has not yet been observed as a major route of resistance development for the classes of synthetic antibacterials

time of exposure of bacteria to natural products, putatively hundreds of millions of years, versus the 70 years of less for the man-made antibiotics

279
Q

What do b-lactamases do

A

destroy the chemical warhead of b-lactam antibiotics by hydrolysing their strained b-lactam
ring, which is the chemically reactive acylating group for modifying the active-site serine residue in transpeptidases in PG cross-linking

280
Q

How effective is resistance through beta-lactamase production?
Use an example

A

very effective as one b-lactam-resistant E. coli cell can excrete up to 105 b-lactamase molecules, each with an ability to hydrolyse ~1000 b-lactams per second.

281
Q

True or false

beta lactamases have developed because of clinical antibiotic use

A

false
b-Lactamase activity was detected a few years before clinical use of penicillins, indicating its presence in soil bacteria that combat the natural product penicillins

282
Q

How many beta-lactmases exist now

how are they classified

A

> 190

into A, B, C, and D
A,C and D are Ser enzymes similar to PG transpeptidases

283
Q

What kind of intermediate do A, C, and D beta lactamases form

What does this suggest

A

same type of penicilloyl-O-Ser enzyme covalent intermediate as PG transpeptidases

evolution from these transpeptidases.

284
Q

Why is resistance to aminoglycoside antibiotics different to penicillins

A

aminoglycoside antibiotics do not have a reactive chemical warhead comparable to the b-lactam

285
Q

aminoglycoside antibiotics do not have a reactive chemical warhead comparable to the b-lactam. What do aminoglycosides do instead

A

read specific regions in the 16S rRNA in the 30S ribosome subunit by a hydrogen-bonding network through the various hydroxyl and amino substituents on the cyclitol rings to provide a high-affinity docking site

286
Q

What is the enzymatic destruction strategy for aminoglycoside resistant bacteria

A

to covalently modify those specificity-conferring OH and NH2 groups in the aminoglycosides and thereby interfere with recognition by the 16 S rRNA

287
Q

How does beta lactamases A, C and D confer resistance to penicillin

A

usually penicillin irreversibly binds to transpeptidase (half life of 90 mins)

b-lactamase binds to penicillin and can incorporate water into the active site which rapidly hydrolyses the ring in miliseconds (similar to the speed of action of transpeptidase without penicillin present)

288
Q

Why do class B lactamases fail to be inhibited by A, C and D lactamase inhibitors

A

There is no such covalent penicilloyl enzyme intermediate in the catalytic cycle of the zinc-dependent, class B lactamases. Instead, the B class enzymes use zinc to activate a water molecule and catalyse its direct addition to the b-lactam ring

289
Q

What led to the development of extended spectrum cephalosporins

A

he growing number of b-lactamases in gram-negative E. coli and Klebsiella pneumoniae, as well as the emergence of these enzymes in other pathogens (for example, Haemophilus influenza and Neisseria gonorrhoeae)

290
Q

Why were extended spectrum cephalosporins not effective for long

A

selective pressure quickly fostered the emergence of extended spectrum b-lactamases
(ESBLs), which could hydrolyse many of the oxyimino-cephalosporins

291
Q

How did ESBLs arise

Name an example of an ESBL

A
by plasmid transfer from pre-existing chromosomal ESBL genes from Kluyvera spp., which typically is a non-pathogenic commensal organism. 
Other ESBLs harbour point-mutations that led to single amino acid changes in existing class A b-lactamases.

CTX-M

292
Q

How many ESBLs have been identified

A

> 200

293
Q

What are the 3 kinds of enzymatic modification of OH and NH2 on aminoglycosides

what have these enzymes likely evolved from

A

(i) N-acetylation of NH2 groups by acetyl-CoA,
(ii) O-phosphoryl transfer of the gamma-phosphate group of ATP to a OH moiety on the aminoglycoside,
(iii) O-adenylyl transfer of the a-phosphate group of ATP, resulting in the transfer of the AMP moiety to a OH moiety on the aminoglycoside

adenylyltransferases, phosphotransferases and N-acetyltransferases that had been utilized for normal biosynthetic processes in the bacterial cells.

294
Q

What are the inactivating enzyme equivalents for bacteria other than aminoglycosides

A

Analogous to the aminoglycoside-inactivating acetyltransferases are families of inactivating acetyltransferases for streptogramin, chloramphenicol, and others.

295
Q

Give a key feature of the methicillin structure

What is it used to treat

How did it dev

A

a bulky 2,6-dimethoxybenzoyl substituent on the 6-aminopenicillin scaffold

Gram positive bacteria that had become resistant to penicillin via inducible b-lactamase hydrolysis of the antibiotic

296
Q

What was the benefit of the bulky side chain of methicillin

A

enhanced the lifetime of the covalent penicillyol-O-lactamase acyl enzyme intermediate against hydrolysis, thereby effectively deactivating the b-lactamase enzyme

297
Q

What is MRSA

How common is it

A

methicillin-resistant
Staphylococcus aureus

In hospital environment in the USA, MRSA can now reach an incidence of 20 to 40%; in Japan an incidence of up to 60% has been reported

298
Q

Where is MRSA particularly prevalent

A

burn centres and long term facilities

299
Q

What is the problem with MRSA and beta lactam administration in hospitals

A

MRSA is resistant to essentially all b-lactam molecules, including penicillins, cephalosporins, carbapenems, and monobactams. Therefore, b-lactam administration selects for MRSA in the clinical setting

300
Q

True or false

MRSA is elaborating an improved version of beta lactamase

A

False
in >90% of cases MRSA has acquired mecA gene, which encodes a new b-lactam-insensitive, bifunctional transglycosylase/transpeptidase (termed penicillin-binding protein 2A).
Auxiliary genes eg fem are also important

301
Q

Describe the auxiliary genes that confer beta lactam resistance in MRSA when combined with mecA

A

fem (factor essential for expression of methicillin resistance) genes

adds a pentaglycyl cross bridge to PG before cross linking which is a better substrate for mecA transpeptidase than the original PG strand

302
Q

What causes Commuity acquired pneumonia, meningitis, otitis media and sinusitis?

Why is this bacteria relevant to drug resistance

A

Streptococcus pneumonia

mechanism of b-lactam resistance based on changes in the composition of PG transpeptidase and other penicillin-binding proteins has been observed

303
Q

Why did the use of vancomycin increase in the 1980/90s

What did this lead to

A

to treat infections caused by gram positive MRSA

selected for drug-resistant enterococci, less potent pathogens than staphylococci but opportunistic in the space vacated by other bacteria and in patients with compromised immune systems

304
Q

Which species accounts for >90% of clinical isolates that are resistant to vancomycin?

Which patients are particularly vulnerable to this bacteria (2)

A

Enterococcus faecalis

patients with indwelling catheters, including dialysis patients and those undergoing cancer chemotherapy who have chemotherapy-induced white cell depletion in the middle of treatment cycles

305
Q

What is the leading cause of endocarditis

A

enterococci

306
Q

What is VanA

A

The first major clinical phenotype of vancomycin-resistant enterococcus

this was followed by VanB

307
Q

How does VanA differ from VanB

A

essentially the same molecular mechanism but differs in the continuing sensitivity to the glycopeptide teicoplanin (a vancomycin analogue).

308
Q

Which genes and gene products are necessary for both VanA and VanB

A

5 tandemly arranged genes

the products are:
3 enzymes, VanH, VanA, and VanX, involved in reprogramming of the PG termini
from N-acyl-D-Ala-D-Ala to N-acyl-D-Ala-D-lactate,

and

2 proteins, VanS and VanR forming a two-component (sensor and response regulator) signal transduction
pathway for inducible reprogramming to vancomycin resistance

309
Q

What accounts for the 1000-fold decrease in vancomycin binding in resistant bacteria

A

switch from D-Ala to D-lactate as the terminal residue in the pentapeptide of the uncross-linked PG

310
Q

Which class of antibiotic is erythromycin involved in

What is this class used to treat

A

macrolide

widely used for respiratory tract infections, but erythromycin resistance has become problematic.

311
Q

Which bacteria are particularly important in erythromycin resistance

A

Pneumococcal

MRSA

312
Q

What is a major route of resistance to macrolides

A

methylation of a specific adenine
(A2058) in the 23S rRNA in the 50S ribosomal subunit by RNA Nmethyltransferases, which is close to the macrolide-binding site

this reduces affinity of the rRNA for lincosamides and streptogramin B, without affecting rRNA function

313
Q

Reduced drug influx is an effective drug resistance mechanism for which drugs

A

hydrophilic drugs which hardly diffuse passively across phospholipid bilayers

314
Q

Give an example of bacteria using drug efflux to promote resistance

A

In Gram -ve P. aeruginosa, aminoglycosides are taken into the periplasm via facilitated diffusion through porin channels in the outer membrane and are then taken across PM by oligopeptide transporters

resistance arises from a decreased porin count, modification of the lipopolysaccharide outer leaflet, and
mutations in uptake trasnporters

315
Q

What drug combination is usually given to patients with an S. aureus infection

Why

A

Aminoglycosides are often combined with a b-lactam drug

b-lactam drug affects cell wall synthesis and increases the passive diffusion of the aminoglycoside into the cell.

enhances bactericidal activity, whereas aminoglycoside monotherapy may allow resistant staphylococci to persist during therapy and cause a clinical relapse once the antibiotic is discontinued.

316
Q

Are hydrophobic drugs affected by decreased influx mechanisms

A

can slow down entry, but cannot not prevent it due to the non-protein mediated diffusion of the drug across cellular membranes.

Active efflux by transport proteins in the cytoplasmic membrane is then the only alternative to prevent the entry of drug in the cytosol

317
Q

Name 4 types of drug where active efflux is relevant for resistance

A

b-lactams,
macrolides,
fluoroquinolones,
tetracyclines

318
Q

Are bacterial efflux transporters dedicated to a single drug?

A

some are eg tetracycline transporters but some have a much broader range

319
Q

Why do bacteria have efflux pumps normally

What does this allow

A

used physiologically for the export of specific metabolites and to pump foreign toxic substances

The integrated array of transporters with overlapping drug specificities can lead to a remarkable capacity to efflux drugs either as a chromosomally encoded metabolic capacity, which makes Pseudomonas aeruginosa intrinsically antibiotic insensitive, or by the acquisition of transport genes on plasmids and transposons.

320
Q

How can drug transporters be divided bioenergetic POV

Which is more common

A

primary active systems (couple drug efflux to the hydrolysis of ATP) - more common in eukaryotic organisms

secondary (couple drug efflux with influx of Na+ or H+) - more common in prokaryotes

321
Q

In gram-negative bacteria, what are

secondary-active drug transporters often associated with

A

an accessory protein, which spans the periplasm, and an outer membrane porin to allow drug transport across the cell envelope into the external environment

322
Q

What is multiple drug resistance

A

The simultaneous expression of various antibiotic resistance mechanisms, each specific for a drug or class of drugs

323
Q

What is a regulon

A

a ‘master switch’ which controls the co-expression of various drug resistance mechanisms, encoded by genes localised at different positions on the genome

324
Q

Other than a regulon, how can multiple drug resistance arise

A

from the co-localization of antibiotic resistance genes on the same resistance plasmid (also called R-factor), which can be transferred from one bacterium to another by conjugation or transformation.

325
Q

How can the R factor be transferred between bacteria

A

by conjugation or transformation

326
Q

What causes multidrug resistance

A

expression of a multidrug efflux pump in the PM confers resistance to a wide variety of drugs due to the enormously broad specificity of the pump.

(not multiple drug resistance - be careful)

327
Q

Why is multidrug resistance difficult to deal with clinically

A

many of the first-line classes of antibiotics can be effluxed from the cell.

328
Q

Name an antifungal azole

What do these do

A

fluconazole

inhibit enzymes involved in ergosterol biosynthesis

329
Q

What is azole resistance based upon

A

alterations in the activity and amount of ergosterol biosynthesis enzymes, and on active azole efflux.

330
Q

What is resistance of herpes to purine analogues based upon

A

a change in the substrate specificity of the viral purine-activating enzyme thymidine kinase, disabling the phosphorylation of purine analogues.

331
Q

Breifly what does the resistance to anti-HIV drugs arise from

A

Resistance to HIV reverse transcriptase inhibitors (e.g. zidovudine) or HIV protease inhibitors (e.g.,
saquinavir) is due to mutations in these enzymes that disable the interaction between enzyme and inhibitor.

332
Q

How do parasites develop resistance to chloroquine

A

resistant parasites accumulates chloroquine in their food vacuoles
much less efficiently than chloroquine-sensitive strains, suggesting that drug resistance results mainly from exclusion of the drug from the site of action.

333
Q

What might the lack of drug accumulation in chloroquine resistant parasites indicate

A

increased drug efflux from resistant parasites, and an ATP-dependent P-glycoprotein was implicated as the pump responsible

or

chloroquine efflux from the vacuole might be mediated by a mutated secondary-active transporter, termed CRT (chloroquine resistance transporter).

334
Q

How can cancer cells use enzymes to deactivate drugs inside the cell

A

Drug can be detoxified by drug metabolism based on cytochrome P450 systems (CYP450) and on conjugation by glutathione S-transferase (GST) and other conjugating systems.

335
Q

How can cancer cells modify drug targets to become resistant

A

Resistance can develop due to mutations in drug targets that alter specificity.
eg cells resistant to topo poisons (e.g. etoposides) possess modified topoisomerases.

336
Q

How can altering gene expression/ enhancing gene repair increase cancer cell resistance to drugs

A

Nitrosourea-resistant cells have high levels of alkyltransferases that repair guanine lesions and so prevent DNA
cross-linking.

Cisplatin-resistant cells have higher levels of enzymes involved in DNA repair.

Resistance can also arise from altered gene expression. For instance, cells will not enter the apoptotic pathway if mutations result in the loss of p53 expression.

337
Q

Guve an example of cancer cells using metabolic bypass to confer drug resistance

A

methotrexate resistance can be based on enhanced expression of dihydrofolate reductase (DHFR).

Methotrexate resistance can also result from reduced uptake due to mutations in the folate carrier, which reduces the affinity of this membrane transporter for methotrexate

338
Q

How can cancer cells use drug efflux to confer resistance

use eg

A

Drug resistance (e.g. against vinca alkaloids, anthracyclines, and mitoxantrone) can result from enhanced drug efflux by multidrug transporters such as the multidrug resistance P-glycoprotein MDR1 (also termed ABCB1), multidrug resistance-associated proteins (MRP1 and 2, also termed ABCC1 and 2) and the breast cancer resistance protein (ABCG2).

339
Q

Give 5 ways we can combat drug resistance

A

Identification of new drug targets

Specific inhibitors of drug resistance mechanisms

Development of new classes of antibiotics/ cancer drugs

Combination therapy

Extending antibiotic lifespan

340
Q

How has genomics helped the battle against antibiotic resistance

A

Approaches involving gene disruption
have begun to narrow the list of genes in pathogens that are essential either for virulence or for survival to perhaps a few hundred genes

we can find chemicals that inhibit the products of these essential genes

341
Q

How did the way we tackled b-lactam resistance change from when resistance was first seen to when it became a real menace

A

originally just tinkered with periphery of the ring so maintain effectiveness for a time

later, attention switched to approaches to neutralize the antibiotic-destroying hydrolase, both by screening against lactamase producers and by mechanism-based inhibition of the active-site serine hydrolases

342
Q

What is clavulanate

How is it administered

A

a suicide inhibitor of lactamase

in combination with amoxicillin (as clavulanate was not an effective antibiotic by itself)

this combination is called Co-amoxiclav and it augments the range of amoxicillin

343
Q

What is Unasyn

A

the combination fo sublactam and ampicillin

344
Q

What is sublactam

A

a penicillin analogue with a five-ring sulphur atom oxidized to the sulphone

has a weaker C-S bond disposed the ring of the acyl-lactamase intermediate to open and create a long-lived covalent enzyme intermediate that was inactive.

345
Q

What is Primaxin an example of

A

Another successful combination of a beta-lactamase inactivator and b-lactam antibiotic (imipenem-cilastatin)

346
Q

What is an important feature of 3rd gen erythromycins currently in development

A

less prone to induce resistance due to methylation of the antibiotic

347
Q

What combination is being designed to combat macrolide and tetracycline resistance

A

a separate inhibitor of efflux pumps, once identified as having sufficient potency and safety, could be combined with the macrolide antibiotic or the tetracycline.

348
Q

What is Oritavancin

What is important about its structure

A

a semisynthetic analogue of vancomycin - used to act against vancomycin resistance enterococci

contains a hydrophobic biphenyl substituent on the vancosamine sugar and is more hydrophobic and may partition the analogue more to the membrane, as well as alter its ratio of inhibition between transpeptidases and transglycosylases

349
Q

What is Synercid

What is it used to treat

A

combination of two non-ribosomal
peptides, quinupristin and dalfopristin, which act synergistically to inhibit protein synthesis in a bactericidal manner

treating infections by vancomycin resistant enterococci.

350
Q

What is the new structural class of synthetic antibiotic molecules with a broad spectrum and acceptable potency

A

oxazolidinones

351
Q

What is the MOA of oxazolidinones

A

o inhibit protein biosynthesis, specifically by interaction with the 23S ribosomal RNA of the 50S ribosomal subunit at or near the peptidyl transferase centre of the ribosome

352
Q

Name an oxazolidinone

A

linezolid

353
Q

What is ramoplanin

A

glycolipodepsipeptide drug against vancomycin-resistant enterococci.

354
Q

What is the MOA of ramoplanin

A

forms a complex with the lipid pentapeptide intermediates in cellwall biosynthesis, and acts in a somewhat analogous way to vancomycin by targeting a substrate rather than an enzyme in the peptidoglycan assembly pathway

355
Q

Name a natural product antibiotic produced by “uncultured”

bacteria discovered in 2015

A

teixobactin

356
Q

Why is resistance to

teixobactin less likely to occur than with other antibiotics

A

binds to lipid-like
peptidoglycan precursors and gene mutations can only affect lipid structure indirectly (through alterations in enzymes in lipid synthesis pathways)

357
Q

What are the different kinds of combination therapy

A

i) Augmentin and Synercid approaches where two components work together to neutralize a single target,
ii) combinations of distinct antibiotic classes that work on different targets concurrently (eg HAART)

358
Q

Why is combination therapy eg HAART so effective

A

fewer cells or viruses will escape from drug action,

and the probability of mutation to clinically significant resistance is reduced

359
Q

What is HAART

A

(highly active antiretroviral therapy

incorporates a mixture of protease inhibitors (e.g., saquinavir) and reverse transcriptase inhibitors (e.g., zidovudine and nevirapine).

360
Q

What is syngergy when it comes to antibiotics

Why is it a therapeutically desired response

A

The characteristic that combinations of antibiotics can have a greater effect than the sum of the two individual drug effects

allows a reduction in the amount of antibiotic required to obtain a therapeutic effect.

361
Q

What is an antagonistic combination of antibiotics

Why does this happen

A

likely to occur when a bactericidal drug (e.g., a penicillin or an aminoglycoside) is combined with a bacteriostatic drug (e.g., a tetracycline)

many bactericidal agents have a killing effect only on cells that are growing or actively synthesizing protein, and that the bacteriostatic drugs prevent growth or protein synthesis and thereby counter the effect of the bacteriostatic agent alone

362
Q

Give an example of a syngerstic combination of antibiotics

A

an aminoglycoside and an inhibitor of cell wall synthesis, which is based on the increased entry of the aminoglycoside into the bacterium

363
Q

Combinations in which two antibiotics inhibit different steps in the same critical metabolic pathway may also result in
synergism. What is an example of this?

A

trimethoprim and sulfamethoxazole (co-trimoxazole), which both inhibit the synthesis of tetrahydrofolate

364
Q

What is an indifferent drug combination

A

drug combination is roughly a summation of the effects of the individual drugs

365
Q

What are some ways to decrease incidence of drug resistance

A

judicious use of antibiotics (which has been recommended by NICE)
rotating use of antibiotics
avoid antibiotic use in farm animals

366
Q

Gvive an example of how we might rotate antibiotic use to preserve efficacy

A

first-line therapy would be to start with one of the 3 main b-lactam categories, such as a b-lactam plus lactamase inhibitor (e.g. the amoxicillin-clavulanate combination).

After 2 months, the unit would cycle to carbapenem antibiotics as the front-line therapy.

At the end of the next 2 months, a third- or fourth-generation (expanded-spectrum and higher) cephalosporin would become the front-line choice.

Then, one would cycle back to the initial combination choice, completing a three-drug traverse in the 6 months

367
Q

Which cancers can be cured by current chemotherapy (5)

A
testicular cancer, 
Hodgkin’s disease,
non-Hodgkin’s lymphoma, 
choriocarcinoma, 
many childhood cancers.
368
Q

How doe we screen naturally occurring products to find new treatments for canacer

A

New compounds are screened for activity against human and animal tumour cell lines in vitro. The most promising agents are tested further to identify the maximally tolerated dose in mice and other species

369
Q

How are new anticancer drugs being developed

A

more drugs are being developed on the basis of know pathways involved in cancer progression and unique activity in preclinical models, rather than through largescale screening of naturally occurring compounds

370
Q

What are the phase I trials in new anticancer drugs

A

performed in cancer patients when

no other treatment is available or when conventional therapy has been unsuccessful

371
Q

What is the starting dose of a phase I anticancer drug

How does dosage change from here

A

based on the patient’s body
surface area, is usually equivalent to one-tenth of the maximally tolerated dose in mice

If this produces no major toxicity, the dose is increased in the next group of patients. In this way, the maximally tolerated dose is determined, which allows Phase II trials

372
Q

What is the major aim of Phase II cancer trials

What happens in Phase III

A

Assessment of anti-tumour efficacy. Sometimes, the efficacy is also tested in combination therapy with existing agents

comparisons are made with the best available current therapy

373
Q

What is D-cycloserine

what is its MOA

A

structural analogue of D-Ala

prevents the synthesis of
pentapeptide by inhibiting L-Ala racemase, D-Ala-D-Ala synthetase and D-Ala-D-Ala/
muramyl tripeptide ligase

374
Q

What is the MOA of fosfomycin and what is it approved for

A

inhibits pyruvyl transferase required for the conversion of NAG into NAM.

Was recently approved in the USA for treatment of urinary tract infections.

375
Q

What is bacitracin and what is its MOA (2)

What is it active against

A

cyclic polypeptide antibiotic;

forms a tight complex with Mg2+ and bactoprenol pyrophosphate;
inhibits the dephosphorylation to bactoprenol phosphate (lipid carrier for NAM-NAG unit).

active against Gram-positive bacteria.

376
Q

What are the following and what is their MOA:
Penicillin, ampicillin, amoxicillin and methicillin

name a combination associated with these drugs

A

ß-lactam antibiotics; inhibit peptidoglycan-crosslinking transpeptidase enzymes by covalent and irreversible binding as a pseudosubstrate. The combination of the ß-lactam amoxicilin and the ß-lactamase inhibitor clavulanate is used as augmentin

377
Q

What is vancomycin and what is its MOA

What is it effective against

A

glycopeptide antibiotic;

binds to D-Ala-D-Ala termini of the pentapeptide in peptidoglycan, thereby preventing its cross-linking to a
neighbouring pentapeptide in another peptidoglycan strand.

Vancomycin is effective against Clostridium difficile, and is used intravenously against
Gram-positive cocci, such as Enterococcus and Staphylococcus

378
Q

What is a drug designed to be similar to vancomycin

A

Oritavancin

a semisynthetic analogue of vancomycin containing a hydrophobic biphenyl substituent on the vancosamine sugar. The drug is more hydrophobic and may partition more to the membrane

379
Q

What is the first line treatment for TB

How does it work

A

isoniazid

used in the prevention and treatment of tuberculosis.

It inhibits the biosynthesis of mycolic acids in the cell envelope of Mycobacterium tuberculosis.

380
Q

What is extended spectrum cephalosporin active against

A

active against Gram-positive bacteria with increased activity against Gram-negative bacteria.

381
Q

What is Teixobactin effective against and what is its MOA

How was it discovered

A

gram-positive bacteria.

It inhibits bacteria by binding to Lipid II in the peptidoglycan biosynthesis pathway

was discovered using a new method of culturing bacteria in soil. This allowed researchers to grow a previously unculturable bacterium, now named Eleftheria terrae, which produces the antibiotic

382
Q

Which antibiotics do we need to know that act on protein synthesis (6)

A
chloramphenicol
tetracycline
erythromycin
fusidic acid
streptomycin and gentamycin
383
Q

What is the MOA of chloramphenicol

when is it used

A

locks aminoacyl tRNA binding to 50S subunit of ribosome. Very effective broad-spectrum antibiotic

use is restricted because of bone
marrow suppression in some cases. Drug is indicated in life-threatening infections eg meningitis.

384
Q

What is tetracycline
what is the MOA
what is it effective against? Is it a first line drug?

is there any toxicity

A

polyketide antibiotic; binds to 16 rRNA in 30S subunit of ribosome and inhibits the movement of aminoacyl-tRNA into the A site.

effective against a wide range of bacteria and are first-line drugs against mycoplasma and cholera.

Drug toxicity is associated with binding of calcium in bones
and teeth.

385
Q

What is erythromycin

what is its action

when is it used

A

14-membered macrolide antibiotic;

binds to 23S rRNA in 50S subunit, and blocks the polypeptide exit tunnel.

Drug has a similar antibacterial spectrum as penicillin and is a suitable second-line drug for patients allergic to penicillin.

386
Q

What is the MOA of fusidic acid

When is it used

A

inhibits elongation factor G, and hence, the movement of the 30S subunit by one codon along the mRNA.

narrow spectrum and is used against staphylococcal infections

387
Q

What are Streptomycin and gentamycin

MOA?

What are they used against

A

aminoglycoside antibiotics

target 30S subunit of ribosome and decrease fidelity of mRNA translation

Used against Gram-negative rods including Pseudomonas and Proteus. Most streptococci (Grampositive) are resistant because gentamycin cannot penetrate the cell. However, penicillin and gentamycin have a synergistic effect against some streptococci.

388
Q

Compare the toxicity of gentamycin and streptomycin

A

Streptomycin exhibits nephrotoxicity and ototoxicity. These effects are reduced for gentamycin

389
Q

What are the 5 antibiotics we learnt that act on DNA and RNA

A
ciprofloxacin 
rifampin 
daunorubicin
bleomycin
mitomycin
390
Q

ciprofloxacin
type?
MOA?
effective against?

A

fluoroquinolone antibiotic;

inhibits type II DNA topoisomerases
(DNA gyrase and topo IV), and hence, inhibits changes in supercoiling required for replication and gene expression.

FDA-approved drug for killing Bacillus anthracis in anthrax infections. It is particularly useful for Pseudomonas infections where oral therapy is preferred, such as respiratory tract infections in patients with cystic fibrosis.

391
Q

What is the MOA of rifampin

used for?

A

binds to b subunit of the DNA-dependent RNA polymerase and
inhibits initiation of RNA synthesis.

Is often used to treat infections by Mycobacterium.

392
Q

Describe daunorubicin (3)

A

planar polycyclic anthracycline antibiotic;

intercalates in dsDNA and causes local unwinding.

Used as anticancer agent

393
Q

Bleomycin
type of drug
MOA

A

metal-chelating glycopeptide antibiotic;

generates superoxide and hydroxyl radicals, causing single- and double-strand breaks in DNA.

394
Q

describe mitomycin

A

aziridine-containing antibiotic;

alkylates and cross-links DNA, thereby preventing strand separation during DNA replication and transcription.

395
Q

Name 3 antibiotics that act as antimetabolites

A

sulfamethoxazole
trimethoprim
co-trimoxazole

396
Q

How does Sulfamethoxazole work

A

sulfa drug

p-aminobenzoate analogue that competitively inhibits dihydropteroate synthase in the biosynthesis of tetrahydrofolate (methyl carrier required for synthesis of dTMP).

397
Q

what is the MOA of trimethoprim

A

inhibits dihydrofolate reductase in the biosynthesis of tetrahydrofolate.

398
Q

What is Co-trimoxazole

What is it used for

A

combination of sulfamethoxazole and trimethoprim.

has been withdrawn as a routine anti-bacterial agent in this country, as result of its being implicated in cases of Stevens-Johnson syndrome – a severe and sometimes fatal allergic reaction. Co-trimoxazole is used in AIDS patients with fungal Pneumocystis carinii infections.

399
Q

Name 5 drugs that change the bacterial membrane to kill it

A
valinomycin
gramicidin
polymixin
amphotericin B
fluconazole
400
Q

moa of valinomycin

A

cyclic peptide antibiotic that binds K+ and facilitates K+ diffusion across the membrane.

401
Q

structure and moa of gramicidin

A

linear polypeptide antibiotic

forms a homodimeric complex that acts as an ion channel in the membrane.

402
Q

what is polymixin and what is its moa

A

cationic detergent antibiotic containing cyclic peptide and
hydrophobic tail;

binds to membrane and alters its ion permeability.

403
Q

What us amphotericin

what is the moa

used for?

A

polyene antifungal and antiparasitic drug;

forms pores in
the membrane by an ergosterol- dependent mechanism

Is currently the drug of choice for most systemic mycoses: active against Cryptococcus, Candida, and Aspergillus

404
Q

what is fluconazole

what is miconazole

What are their MOA

what are they used for

A

triazole antifungal drug

imidazole antifungal and antiprotozoal drug.

Both inhibit ergosterol biosynthesis.

Fluconazole is often used in the treatment of athlete’s foot and vaginal candidiasis.

405
Q

name 12 anti protozoal drugs

A
melarsen oxide
suramin
amphotericin B and miconazole 
chloroquine and mefloquine 
artemisinin 
sulfadoxin 
pyrimethamine
fansidar
tetracycline and lincomycin
406
Q

describe melarsen oxide

moa

used for

A

arsenic compound;

selectively accumulated in Trypanosoma species, inhibits energy metabolism.

Used for treatment of sleeping sickness and other diseases caused by trypanosomes

407
Q

why was suramin introduced in 1920

what is it used for

what is a benefit of it

moa?

A

as a development of Ehrlich’s Trypan Red,

today still remains the drug of choice for trypanosomiasis.

Does not contain toxic metal, hence, its therapeutic index is much higher than that of melarsen.

Molecular mechanism still unknown; might act on glycolytic enzymes

408
Q

What are Chloroquine and mefloquine and what do they do

A

antimalarial 4-aminoquinoline drug

inhibits haem polymerization in food vacuole of Plasmodium.

409
Q

Describe artemisinin

A

prodrug of the biologically active metabolite dihydroartemisinin,
which is active during the stage when the parasite is located inside RBCs

MOA might be based on the production of oxygen radicals.

410
Q

what is sulfadoxin

A

sulfa drug that inhibits dihydropteroate synthase in Plasmodium.
Analogue of the antibiotic sulfamethoxazole

411
Q

what is the moa of pyrimethamine

A

inhibits dihydrofolate reductase in Plasmodium. Analogue of the antibiotic trimethoprim

412
Q

what is fansidar

what is it used for

A

combination of sulfadoxin and pyrimethamine; analogous to
antibacterial co-trimoxazole

Used in the treatment of patients with P.
falciparum malaria when chloroquine resistance is suspected, and oral treatment is
appropriate.

413
Q

name 10 antivirals

A
amantidine
oseltamivir 
acyclovir
ganciclovir
cidofovir
foscarnet
zidovudine
nevirapine
saquinavir
interferon
414
Q

what is the MOA of amantadine and what is it used for

A

blocks the function of M2 ion channel protein involved in
nucleocapsid uncoating of influenza virus. Is used predominantly in prophylaxis and
treatment of infections by Influenza A virus. Not effective against Influenza B virus

415
Q

What is the MOA of oseltamivir

what is a related drug

A
inhibit neuraminidase (sialidase), a
glycoprotein enzyme that plays an essential role in the release of the virion progeny from infected cells, and that assists in the movement of the virus particles through the upper respiratory tract.

zanamivir

416
Q

what is aciclovir

A

purine analogue;

upon activation by viral thymidine kinase, phosphorylated compound inhibits viral DNA polymerase.

Is present in Zovirax creams against herpes simplex infections of the skin and mucous membranes

417
Q

what is ganciclovir

A

as acyclovir, but selective against cytomegalovirus.

418
Q

what is the moa of cidofovir

A

nucleoside phosphonate analogue of cytosine that inhibits DNA polymerase of cytomegalovirus.

419
Q

What is the moa of foscarnet

A

organic analogue of pyrophosphate that inhibits DNA polymerase
of cytomegalovirus and herpes virus

420
Q

what is nevirapine

A

anti-HIV drug; non-nucleoside reverse transcriptase inhibitor.

421
Q

what is AZT

moa

A

Zidovudine (azidothymidine)

nucleoside reverse transcriptase
inhibitor;

thymidine analogue

422
Q

describe saquinavir

A

inhibitor of HIV protease; this protease is vital for both viral
replication within the cell and release of mature viral particles from an infected cell.

423
Q

which drugs are used in HAART and when is it used

A

nevirapine, zidovudine and saquinavir

during serious manifestations of HIV infections in patients with AIDS

424
Q

what is interferon and how is it used for antiviral therapy

A

immunoregulatory cytokine that is synthesized in response to viral infection.

Interferons are used as antiviral agent against hepatitis B and C, and AIDS related Kaposi sarcomas.

425
Q

melphalan
class?
used for?
derivation?

A

nitrogen mustard alkylating agents,

is used to treat chronic myeloid leukaemia
occasionally used against melanoma.

Melphalan is a phenylalanine derivative of mechlorethamine.

426
Q

cyclophosphamide
class?
PK?
use?

A

nitrogen mustard alkylating agent,

“prodrug”: converted in the liver to active forms that have chemotherapeutic activity.

main use of cyclophosphamide is in the treatment of lymphomas, some forms of leukemia and some solid tumours

427
Q

lomustine
class?
MOA?
use and special ability?

A

nitrosourea class of agents,

acts by crosslinking DNA to other DNA strands or to protein in such a way that dsDNA cannot be replicated.

Lomustine is most commonly used against lymphoma (particularly cutaneous (skin) lymphoma) and melanoma, and tumours in kidney and lung.

Lomustine has the special ability to penetrate the blood/brain barrier and can be used to treat cancers in the brain.

428
Q

describe cis-platin

A

platinum-based drug used to treat various types of cancers, including sarcomas, some carcinomas (e.g. small cell lung cancer, and ovarian cancer), lymphomas and germ cell tumours.

429
Q

describe doxorubicin

A

anthracycline antibiotic that intercalates in DNA.

structurally closely related to daunorubicin, and also intercalates in DNA.

often administered by local injection.

430
Q

mitoxantrone
class
use

A

anthracycline agent

used in the treatment of certain types of cancer, mostly metastatic breast cancer, acute myeloid leukemia, and non-Hodgkin’s lymphoma.

431
Q

Methotrexate
class
moa

A

antimetabolite and antifolate drug.

It acts by inhibiting the metabolism of folic acid.

Methotrexate replaced the more powerful and toxic antifolate
aminopterin.

432
Q

what is leucovorin

A

adjuvant used in cancer chemotherapy involving the drug methotrexate. It is also used in synergistic combination with the chemotherapy agent 5-fluorouracil.

433
Q

5-fluorouracil
class
administration
use

A

belongs to the family of antimetabolites and is a pyrimidine analogue.

typically administered with leucovorin.

its principal use is in colorectal cancer and pancreatic cancer, in which it has been the established form of chemotherapy for decades (platinum-containing drugs are a recent addition).

434
Q

etoposide
moa
use

A

inhibitor of the enzyme topoisomerase II.

used in the treatment of lung cancer, testicular cancer, lymphoma, and non-lymphocytic leukaemia

435
Q

what is topotecan

what is it used to treat

A

topoisomerase I inhibitor.

Is used to treat ovarian cancer and lung cancer.

436
Q

vinblastine
class
use

A

vinca alkaloid and an anti-mitotic drug

used to treat Hodgkin’s lymphoma, non-small cell lung cancer, breast cancer and testicular cancer

437
Q

Taxol

use? (5)

A

belongs to the drug category of the taxanes, and is a mitotic inhibitor

used to treat patients with lung, ovarian, breast cancer, head and neck cancer, and advanced forms of Kaposi’s sarcoma.

438
Q

what is tamoxifen

use

A

orally active oestrogen receptor antagonist

used in the treatment of breast cancer and is currently the world’s largest selling drug for that purpose.

439
Q

what is toremifene

A

tamoxifen analogue without estrogen agonist properties

440
Q

what is anastrozole

A

aromatase inhibitor

441
Q

what is goseraline

A

Decapeptide analogue of gonadotropin-releasing hormone that disrupts endogenous hormonal feedback systems, resulting in the down regulation of testosterone and estrogen production

442
Q

what is flutamide and what is its moa

A

oral anti-androgen drug primarily used to treat prostate cancer.

competes with testosterone and its powerful metabolite, dihydrotestosterone (DHT) for binding to androgen receptors in the prostate gland. By doing so, it prevents their stimulation of growth of prostate cancer cells.

443
Q

what is prednisone

A

synthetic corticosteroid prodrug that is converted by the liver into prednisolone, which is the active agent that inhibits growth of lymphocytes in leukemias.

444
Q

what are the following:

a) rituximab
b) trastuzumab
c) bevacizumab
d) cetuximab
e) erlotinib

A

a) anti-CD20 monoclonal antibody.
b) anti-HER2 monoclonal antibody
c) anti-VEGF monoclonal antibody.
d) anti-EGFR monoclonal antibody
e) small molecule ligand that inhibits EGFR tyrosine kinase.

445
Q

what are all the monoclonal antibodies used against cancer (5)

A

a) rituximab
b) trastuzumab
c) bevacizumab
d) cetuximab
e) erlotinib

446
Q

what is imantinib

what is it used for

what is it marketed as

A

2-phenylaminopyrimidine derivative that functions as a specific inhibitor of a number of tyrosine kinase enzymes.

used in treating chronic myeloid leukemia, and gastrointestinal stromal tumours, and is currently marketed as Gleevec (USA) or Glivec (Europe/Australia).

447
Q

How does beta lactamases A, C and D work

How does B class differ

A
penicillin works by binding to transpeptidase and excluding water from the active site, meaning the complex is stable and has a half life of 90 mins
When an A, C or D class lactamase is present, it brings in water to the transpeptidase-penicillin complex, causing the half life to be reduced to only 4 seconds 
This breaks the lactam ring and releases the transpeptidase to perform its usual task

B class has a Zn ion which breaks the lactam ring before the penicillin-transpeptidase complex is formed. It does NOT form a covalent bond with penicillin