Nucleic Acid Based Cancer Therapies

?
  • Created by: LBCW0502
  • Created on: 27-01-20 11:38
State features of DNA as a drug target
Drugs target either major groove or minor groove. DNA possess a number of nucleophilic sites (reactive towards electrophiles). Intercalation (drugs can slide between base pairs). Non-covalent interactions. Drugs have different selectivities
1 of 132
What are the types of DNA adducts?
DNA cleavage, intercalation, intra-strand cross-link, interstrand cross-link, topoisomerase adduct, mono-adduct
2 of 132
Give examples of methylating agents
Dacarbazine, procarbazine, temozolomide
3 of 132
Give examples of alkylating agents
Carbinolamines, cyclopropanes
4 of 132
Give examples of cross-linking agents
Nitrogen mustards, aziridines, mitomycin C, epoxides, methanesulphonates, nitrosoureas, platinum complexes, sequence selective DNA cross-linking agents
5 of 132
Give examples of intercalating agents
Antracyclines, antracenes, phenoxazines
6 of 132
What are the types of topoisomerase inhibitors?
Topoisomerase I Inhibitors. Topoisomerase II Inhibitors
7 of 132
Give examples of DNA cleaving agents
Bleomycins, enediynes
8 of 132
Give examples of novel nucleic acid based approaches
Quadruplex binding and telomere inhibition. Transcription factor binding inhibition. DNA repair inhibition (synthetic lethality). Epigenetics. RNA targeting (e.g. RNAi and anti-sense). CRISPR- Cas9 gene editing, other approaches
9 of 132
What is the basis for their selective toxicity towards tumour cells? (1)
Difference in growth rate of populations of cancer cells compared to normal cells (explains toxicity). Reduced capability of cancer cells to repair DNA lesions compared to normal cells (apoptosis)
10 of 132
What is the basis for their selective toxicity towards tumour cells? (2)
Some DNA interactive agents selectively target specific DNA regions (G-C) which may be prevalent in some tumour cells (Burkitt's lymphoma). May block action of key transcription factors up-regulated in tumour cells compared to healthy cells
11 of 132
Describe the importance of DNA repair pathways in mechanism of action of DNA-interactive anticancer agents
If a cell cannot repair DNA damage caused by DNA-Interactive Agents, then p53 will direct cell into “Programmed Cell Death” (also known as “Apoptosis”). DNA Repair Pathways may be damaged in cancer cells - genomes evolve/become increasingly mutated
12 of 132
Which toxicities are associated with DNA-interactive anti-cancer agents?
Short term S/E of DNA interactive drugs - alopecia, GI toxicity (mucositis, diarrhoea), nausea and vomiting, reversible bone marrow suppression (most dangerous). Lung and skin toxicities (e.g. bleomycin)
13 of 132
What are the two problems associated with prolonged usage? (1)
Gametogenesis is affected and sperm storage is recommended in male patients prior to treatment. Ovarian tissue can also be stored for women
14 of 132
What are the two problems associated with prolonged usage? (2)
Prolonged use in combination with radiotherapy can increase incidence of acute non-lymphocytic leukaemia/sarcomas occurring later in life du to DNA damage caused by treatments
15 of 132
Give examples of points of nucleophilicity in DNA
Major and minor grooves. Within nucleophilic bases. All potential reaction points are nucleophilic. Electrophilic BNF drugs interact with nucleophilic DNA. GC base pairing. AT base pairing (various drugs sensitive to particular base pairs)
16 of 132
Which class of drugs was the first DNA-interactive agents?
Nitrogen mustards
17 of 132
Describe features of methylating agents (1)
E.g. Dacarbazine, procarbazine and temozolomide. Work by methylating guanine bases within major groove of DNA at guanine N7 and O6 positions
18 of 132
Describe features of methylating agents (2)
This is distinct from ET-743 and the PBDs which alkylate the N2 of guanine in the minor groove thus forming bulky adducts which block DNA processing.
19 of 132
Describe features of dacarbazine (1)
Wide spectrum of activity ranging from malignant lymphomas to melanomas and sarcomas. Initially thought to inhibit purine biosynthesis but actually demonstrated that enzymic N-demethylation occurs in vivo
20 of 132
Describe features of dacarbazine (2)
Forms 5-aminoimidazole-4-carboxamide and a transient methyldiazonium ion. Through radiolabeling experiments it has been shown that the latter methylates DNA at guanine N7 positions. Has irritant properties. S/E - myelosuppression, nausea/vomiting
21 of 132
Describe features of dacarbazine (3)
Used for Hodgkin's disease
22 of 132
Describe features of procarbazine (1)
First synthesized as a mono-amine oxidase inhibitor. Later discovered that it had significant activity in lymphomas/carcinoma of bronchus. Also a prodrug form of CH3+ ion
23 of 132
Describe features of procarbazine (2)
MOA - metabolic N-oxidation to an azoprocarbazine species, followed by subsequent rearrangement to produce either methyl diazonium or methyl radicals which act as DNA methylating agents towards guanine residues.
24 of 132
Describe features of procarbazine (3)
Used for Hodgkin's disease in combination treatment. S/E - nausea, myelosuppression, hypersensitivity rash.
25 of 132
Describe features of procarbazine (4)
The mild monoamine-oxidase inhibition effect of procarbazine does not require any dietary restriction, however alcohol ingestion may cause a disulfiram-like reaction
26 of 132
Describe features of temozolomide (1)
Treatment for malignant glioma and melanoma. Dose-limiting toxicity (bone marrow suppression). Good PKs. Penetrates CNS (brain tumours)
27 of 132
Describe features of temozolomide (2)
MOA - chemical cleavage of tetrazinone ring to liberate 5-aminoimidazole-4-carboxamide and the highly reactive methyldiazonium methylating species
28 of 132
Describe features of temozolomide (3)
After methylating DNA (or otherwise decomposing by reacting with water to give methanol) the latter forms N2, and it is the generation of the small stable molecules 5-aminoimidazole-4-carboxamide, CO2 and N2 that provides the driving force for MOA
29 of 132
Describe features of temozolomide (4)
Also prodrug for CH3+ ion form. A fragment of duplex DNA is shown with a methylated guanine base
30 of 132
Describe features of the biological mechanism of action (1)
Antitumour activity of temozolomide correlates with the accumulation of agent in tumours where it methylates the guanine-O6 and N7 positions in the major groove of DNA in tumour cells
31 of 132
Describe features of the biological mechanism of action (2)
Selectivity at cellular level may be attributable to slightly different pH environments of normal vs malignant tissues in brain, coupled with differential capacities to repair methylated lesions by O6-alkyl-DNA alkyltranferase (ATase)
32 of 132
Describe features of the biological mechanism of action (3)
At a biochemical level, it has now been established that temozolomide’s activity depends on the operation of the mismatch repair enzymes (MMRs) which detect the wobble base-pair formed during replication of drug-modified DNA
33 of 132
Describe features of the biological mechanism of action (4)
Enzymes replicating DNA get confused (G-T base pairing resembles A-T) – wobble base pair (MMR enzyme remove T and try to incorporate suitable base, results in adding T back on again, endless cycle), P53 protein recognises issue, triggers apoptosis
34 of 132
Describe the development of resistance (1)
To protect against such events, repair proteins have evolved, O6- and N7-alkylguanine-DNA alkyltranferases (ATases), that cleave O6- and N7-modifications stoichiometrically to restore native guanine bases
35 of 132
Describe the development of resistance (2)
Base modified by temozolimide leads to mechanism of resistance - response to tumour to drug depends on MMR levels and ATase expression. Screen patients (maximum treatment benefit with ATase-ve and MMR +ve)
36 of 132
Describe features of carbinolamines (1)
Ecteinascidin-743. Alkylates the N2 of guanine in the minor groove thus forming a bulky adduct that interferes with DNA processing. Used against melanoma, breast, ovarian, colon, renal, lung, prostate carcinomas
37 of 132
Describe features of carbinolamines (2)
MOA involves covalent binding to the N2 position of guanine in the minor groove of DNA causing the double helix to bend towards the major groove. Subunits A/B (framework of covalent interaction with minor groove of DNA double helix)
38 of 132
Describe features of carbinolamines (3)
Subunit C (protrudes DNA duplex, interacts with adjacent nuclear proteins, accounts for cytotoxicity)
39 of 132
What is the biological mechanism of action for ET-743? (1)
Traps nucleases in a malfunctioning nuclease-(ET-743)-DNA adduct complex, thereby inducing irreparable single-strand breaks in the DNA
40 of 132
What is the biological mechanism of action for ET-743? (2)
Induces strong perturbation of the cell cycle with delay of cell progression from G1 to G2 phase, an inhibition of DNA synthesis and cell cycle arrest in G2 phase resulting in p53- independent apoptosis.
41 of 132
What is the biological mechanism of action for ET-743? (3)
Selectively inhibits transcriptional activation of the multidrug-resistance (MDR1) gene in human sarcoma cells. S/E not observed
42 of 132
Describe features of cyclopropanes (1)
Cyclopropane moieties are the homocarbon analogues of aziridines and epoxides, and are also electrophilic and capable of alkylating DNA. Release of strain within 3-membered ring (driving force for alkylation). Duocarmycin interacts with cycloproprane
43 of 132
Describe features of cyclopropanes (2)
CPIs and CBIs. Cyclopropane-Containing DNA A-Mono-Alkylating ADC Payloads. Cyclopropylbenzindole. Trastuzumab duocarmazine
44 of 132
Describe features of cross-linking agents (1)
Contain two alkylating moieties separated by various distances by linkers.
45 of 132
Describe features of cross-linking agents (2)
By alkylating two nucleophilic functional groups either on the same or opposite strands of DNA, the adducts formed represent either “intrastrand” or “interstrand” cross-links, respectively
46 of 132
Describe features of nitrogen mustards
Molecules “staple” the two strands of DNA together via covalent interactions in the major groove, and presumably block replication. It can also be demonstrated that enzymes such as RNA polymerase are blocked by mustard-DNA adducts. Telomeric effect
47 of 132
What are the possibilities for basis of selectivity?
Kinetic explanation (killing faster growing cells). Some cancer cells less proficient at repairing mustard adducts compared to healthy cells. GC selectivity. Some transcription factor binding sites also GC rich
48 of 132
What are the possible mechanisms for development of resistance? (1)
MDR-related resistance (up-regulation of p-glycoproteins). Increase in concentration of glutathione in tumour cells (nucleophilic, forms adducts with mustards, no longer electrophilic/unable to react with DNA)
49 of 132
What are the possible mechanisms for development of resistance? (2)
Some cancer cells resistant to nitrogen mustards by up-regulating repair processes, mustard adducts excised and damaged DNA re-synthesised
50 of 132
Describe features of aliphatic nitrogen mustards - chlormethine (1)
The high chemical reactivity of the aliphatic nitrogen mustards towards DNA and also their vulnerability to attack by a wide range of other nucleophiles is thought to account for the observed toxicities.
51 of 132
Describe features of aliphatic nitrogen mustards - chlormethine (2)
Forms a cyclic aziridinium ion. Cation formed, reacts readily with nucleophile. Attack of the N7-atoms of guanine residues in the major groove of DNA (both strands, lock strands together, SN2 process)
52 of 132
Describe features of aromatic nitrogen mustards (1)
Central nitrogen atom of an aromatic mustard is not sufficiently basic to form a cyclic aziridinium ion since the nitrogen electron pair is delocalized by interaction with the π electrons of the aromatic ring
53 of 132
Describe features of aromatic nitrogen mustards (2)
SN1 reaction, normal carbocation formed (Cl ion ejection, provides rate determining step). Analogues deactivated to reach DNA target sites before being degraded by nucleophiles. Oral treatment
54 of 132
Describe features of chlorambucil
COOH group (improve water solubility of aromatic mustard analogue, electron withdrawing effect)
55 of 132
Describe features of melphalan
Also contains COOH. Attachment of an amino acid residue (i.e., phenylalanine) might facilitate selective uptake by tumour cells in which rapid protein synthesis is occurring.
56 of 132
Give examples of oxazaphosphorines
Cyclophosphamide (Endoxana). Ifosfamide (Mitoxana)
57 of 132
Describe features of cyclophosphamide (1)
The design of this mustard prodrug was based on the concept that the P=O group should decrease the availability of the nitrogen lone pair in an analogous manner to the phenyl ring of the aromatic mustards, deactivating molecule to nucleophilic attack
58 of 132
Describe features of cyclophosphamide (2)
P=O group might be cleaved by phosphoramidases which were thought to be over-produced in some tumour cells, thus releasing the nitrogen lone pair and restoring the electrophilicity of the molecule
59 of 132
Describe features of cyclophosphamide (3)
A degree of tumour selectivity might be achieved if the phosporamidase enzymes were confined mainly to tumour cells.
60 of 132
Describe features of cyclophosphamide (4)
It was later demonstrated that activation in vivo is not due to enzyme-catalysed hydrolysis of the P=O group, but rather to oxidation by liver microsomal enzymes
61 of 132
Describe features of using MESNA to reduce toxicity of acrolein produced from cyclophosphamide (1)
The acrolein metabolite is excreted in the urine where it can cause hemorrhagic cystitis.
62 of 132
Describe features of using MESNA to reduce toxicity of acrolein produced from cyclophosphamide (2)
Acrolein is a potent electrophile and can react with nucleophiles on the surface of cells lining the bladder. An increased fluid intake after intravenous injection can help to avoid this problem.
63 of 132
Describe features of using MESNA to reduce toxicity of acrolein produced from cyclophosphamide (3)
MESNA (UromitexanTM) is routinely co-administered to “neutralize” the effect of the acrolein.
64 of 132
Describe features of using MESNA to reduce toxicity of acrolein produced from cyclophosphamide (4)
Acts as a “sacrificial” nucleophile, forming a non-reactive water soluble adduct that is eliminated safely in the urine.
65 of 132
Describe features of ifosfamide
Analogue of cyclophosphamide that is not technically a nitrogen mustard due to the translocation of one chloroethyl moiety to another position within the molecule. But similar spectrum activity to cyclophosphamide. MESNA co-prescribed
66 of 132
Describe features of nitrogen mustards
The combination of a mustard and an oestrogen which appears to provide both an antimitotic effect and (by reducing testosterone levels) a hormonal effect
67 of 132
Describe features of estramustine phosphate
Combination of an oestrogen and chlormethine (mustine) unit joined through a carbamate linkage. Alkylation/cross-link DNA
68 of 132
Describe features of bendamustine
Forms both intra- and inter-strand cross-links between DNA bases
69 of 132
Describe features of aziridines (1)
Ring-opening of the aziridines with nucleophiles is slower compared to the fully-charged aziridinium ions of the mustards. However, depending upon the pKa of the aziridine nitrogen, there is likely to be significant protonation at physiological pH,
70 of 132
Describe features of aziridines (2)
Meaning that, in practice, the aziridinium ion may be the reactive species.
71 of 132
Describe features of mitomycin C (1)
A naturally-occurring antitumour antibiotic rich in chemical functional groups. MOA involves - quinone, aziridine and carbamate moieties. Bioreductive agent. DNA cross linking properties. MOA - bioreductive trigger (chemical mechanism)
72 of 132
Describe features of mitomycin C (2)
Bioreductive conditions that can exist at the centre of tumours (particularly larger ones) are thought to account, in part, for the tumour selectivity of mitomycin (mechanism)
73 of 132
Describe features of mitomycin C (3)
Treats upper GI, breast cancers. Can cause delayed bone-marrow toxicity and so is usually administered at 6-weekly intervals. Prolonged use - permanent bone marrow damage, lung fibrosis, renal damage. Acute skin irritation at injection site
74 of 132
Describe features of epoxides
Alkylate DNA. Treosulfan - prodrug that converts non-enzymatically to L-diepoxybutane via the monoepoxide under physiological conditions. Alkylation occurs at guanine bases
75 of 132
Describe features of methanesulphonates (1)
Forms DNA cross-links, with the methanesulphonyloxy moieties acting as leaving groups after attack by nucleophilic sites on DNA (SN2 alkylation)
76 of 132
Describe features of methanesulphonates (2)
Less S/E, but can cause myelosuppression, irreversible bone marrow aplasia (careful monitoring required). Hyperpigmentation of skin can occur as common S/E
77 of 132
Describe features of nitrosureas (1)
Alkylate DNA and lead to both mono-adducts and interstrand cross-links at a number of different sites. E.g. carmustine and lomustine
78 of 132
Describe features of nitrosureas (2)
MOA - mono-alkylation and cross-linking. Activity to brain/CSF/so-called sanctuary sites due to high lipophilicity. But can cause severe bone marrow toxicity, dose limiting
79 of 132
Describe features of lomustine
It is mainly used to treat Hodgkin’s disease and certain solid tumours. Bone marrow toxicity delayed. Nausea and vomiting common. Permanent bone marrow damage with prolonged use
80 of 132
Describe features of carmustine (1)
It is most commonly used to treat patients with myeloma, lymphoma and brain tumours. Cumulative renal damage and delayed pulmonary fibrosis may occur.
81 of 132
Describe features of carmustine (2)
Gliadel wafer - implanted into socket left from tumour being removed, release drug, dissolve over time, remove cancer stem cells
82 of 132
Describe features of platinum complexes
Cisplatin (testicular/ovarian cancers). MOA - platinum complexes interact with major groove of DNA forming mainly intrastrand links (guanine N7-guanine N7, guanine N7-adenine N7, guanine N7-X-guanine N7). Kinked cisplatin DNA adduct
83 of 132
What is the biological mechanism of action for platinum complexes
Germ cell shave limited ability to repair cisplatin adducts compared to other cells. MOA could involve transcriptor factor inhibition, RNA transcription inhibition, DNA replication inhibition
84 of 132
What are the disadvantages of platinum complexes?
S/E of cytotoxic agents - bone marrow suppression, nausea, vomiting, GI disturbances and hair loss. Peripheral neuropathy and ototoxicity. High cost due to Pt content
85 of 132
Describe the uses and side effects of cisplatin
Used in metastatic germ cell cancers. S/E - nephrotoxicity, ototoxicity, peripheral neuropathy, hypomagnesaemia and myelosuppression. Requires IV hydration treatment for nausea/vomiting
86 of 132
Describe features of carboplatin
Better tolerated than cisplatin (S/E, due to dose determined by renal function rather than BSA)
87 of 132
Describe features of oxaplatin
Licensed for colorectal cancer in combination with fluorouracil and folinic acid. S/E - sensory peripheral neuropathy are dose limiting, GI disturbances, ototoxicity and myelosuppression. Need to monitor renal function
88 of 132
Give examples of new cisplatin analogues in various stages of development
Picoplatin, Nedaplatin, Satraplatin
89 of 132
Describe features of sequence selective cross-linking agents
Aim to develop agents that can span longer sequences of DNA. Agents with lower sequence selectivity leads to larger number of adducts formed, toxic S/E. Longer sequence selectivity, fewer adducts, less S/E
90 of 132
Give an example of a DNA sequence selective minor groove binding agent
SJG-136. (Design concepts of PDB dimers, Rovalpituzumab tesirine, A-A cross linkers)
91 of 132
Describe features of intercalating agents
MOA - insertion between the base pairs of DNA, perpendicular to the axis of the helix. Effect on DNA - changes structure of DNA
92 of 132
What are the types of intercalating agents?
Anthracyclines, anthracene, phenoxazine, radiomimetics
93 of 132
Describe features of anthracyclines
Possible MOAs - inserts between DNA base pairs to interfere with transcription, form complexes with topoisomerase enzymes and DNA to cause strand to break, binds to cell membranes to alter fluidity, free radical production
94 of 132
Describe features of doxorubicin (1)
Used to treat the acute leukemias, lymphomas, and a variety of solid tumours including carcinoma of the breast, lung, thyroid and ovary, as well as soft-tissue carcinomas
95 of 132
Describe features of doxorubicin (2)
Given by injection into a fast running infusion, usually at 21 day intervals, although care must be taken to avoid local extravasation which can cause severe tissue necrosis. S/E - nausea, vomiting, myelosuppression, alopecia, mucositis
96 of 132
Describe features of limiting cardiotoxicity of doxorubicin (1)
Higher cumulative doses are associated with cardiomyopathy, and potentially fatal heart failure can occur.
97 of 132
Describe features of limiting cardiotoxicity of doxorubicin (2)
Liposomal formulations are also available which are thought to reduce the incidence of cardiotoxicity and lower the potential for local necrosis.
98 of 132
Describe features of daunorubicin
Treatment of acute lymphocytic and myelocytic leukemias.
99 of 132
Describe features of aclarubicin
It is used for acute non-lymphocytic leukemia in patients who have relapsed or are resistant or refractory to first line chemotherapy
100 of 132
Describe features of epirubicin
A semi-synthetic analog of doxorubicin differing only in the stereochemistry of the C4-hydroxy group of the sugar moiety.
101 of 132
Describe features of idarubicin
A semi-synthetic analog of daunorubicin with general properties similar to doxorubicin. The only anthracycline that can be given orally as well as intravenously.
102 of 132
Describe features of anthracenes
Have three fused rings e.g. mitoxantrone
103 of 132
Describe features of mitoxantrone (1)
Rich in oxygen- and nitrogen- containing substituents and side chains, and these allow extensive stabilization of the intercalated adduct by hydrogen bonding interactions.
104 of 132
Describe features of mitoxantrone (2)
Binding to GC-rich sequences and, like the anthracyclines, there is evidence that DNA is cleaved although the mechanism is not thought to be linked to the generation of reactive oxygen species.
105 of 132
Describe features of mitoxantrone (3)
Used for metastatic breast cancer, non-Hodgkin’s lymphoma and adult non-lymphocytic leukaemia. Myelosuppression and dose-related cardiotoxicity (S/E)
106 of 132
Describe features of pixantrone
Analogue of mitoxantrone with fewer toxic effects on cardiac tissue. Acts as an intercalating agent and Topoisomerase II inhibitor. It is formulated as pixantrone dimaleate.
107 of 132
Describe features of phenoxazines
Three fused six-membered rings, central ring contains oxygen and nitrogen heteroatoms. The best known member is dactinomycin that contains two cyclic peptide side chains which stabilise the drug-DNA adduct by interacting in the minor groove of DNA.
108 of 132
Describe features of dactinomycin (1)
The mechanism of action appears to be dependent on its concentration, with either blockade of DNA synthesis occurring or inhibition of DNA-directed RNA synthesis thus preventing chain elongation.
109 of 132
Describe features of dactinomycin (2)
Phenoxazone ring intercalates preferentially between GC base pairs where it can interact with the N2-amino groups
110 of 132
Describe features of dactinomycin (3)
The cyclic peptide moieties position themselves in the minor groove and participate in extensive hydrogen bonding and hydrophobic interactions with functional groups in the floor and walls of the groove (stabilise adduct, block RNA polymerase)
111 of 132
Describe features of dactinomycin (4)
Used in combination with vincristine in the treatment of Wilm’s tumour, and combined with methotrexate for gestational choriocarcinoma. The side effects of dactinomycin are similar to those of doxorubicin (cardiac toxicity is less prominent)
112 of 132
In which phase of the cell cycle is cytotoxicity greatest?
In the S phase (when topoisomerase levels are at a maximum)
113 of 132
Describe features of topoisomerases
DNA topoisomerases (enzymes responsible to cleavage, annealing, supercoiling DNA)
114 of 132
Describe features of topoisomerase I enzymes
Remove negative supercoils in DNA without leaving damaging nicks, break one DNA strand, attach free phosphate of broken strand to tyrosine residue on enzyme, complexes to reseal two ends of DNA
115 of 132
Describe features of topoisomerase II enzymes
Cleaves both strands of double-stranded DNA followed by resealing of both strands
116 of 132
Describe features of topoisomerase I inhibitors
Induces DNA breaks and cell death. Camptothecin, Hycamtin, Irinotecan. Keep chromosomes wound tight, so cell cannot make proteins, leading to cell death. S/E include diarrhoea, nausea, lowered leukocyte count, damage to bone marrow
117 of 132
Give an example of the use of pharmacogenomic assay to avoid serious ADRs
Test for UGT1A1 - identify patients with increased risk of severe adverse reactions (detect variations in UGT1A1 gene)
118 of 132
Describe features of topoisomerase II inhibitors
Topo IIa and IIb isoforms. Etoposide, Teniposide, Ellipticene, Amsacrine (inhibits isoforms to reseal cleaved DNA duplexes), S/E - alopecia, myelosuppression, nausea, vomiting
119 of 132
Describe features of DNA cleaving agents
Bind to DNA helix, cleave double-stranded helix through radical production e.g. bleomycin, calicheamicin
120 of 132
State features of bleomycins
Bleomycins – doesn’t follow Lipinski’s RO5, three distinct regions (thiazoles – intercalate, between base pairs of DNA), chelation of metal ion (cause free radical formation), stabilisation of adduct. S/E - dose limiting
121 of 132
Describe features of DNA quadruplex binding agents
Potentially selective modulators of gene expression by binding in promoter regions of genes e.g Quarfloxin
122 of 132
Describe features of transcription factor inhibitors
Aim to selectively inhibit gene transcription with small molecules (rather than macromolecules), stop expression of oncogenes. Dervan's hairpain polyamides
123 of 132
Describe features of DNA repair inhibition
PARP inhibitors (Olaparib). Inhibit PARP1 – repair cannot take place (by P53 enzyme), single stranded break becomes a double stranded break, effective for pancreatic cancer
124 of 132
Describe features of epigenetics (1)
A mechanism to regulate gene expression independent of any changes to the DNA sequence. Genomics (epigenetic influence), transcriptomics, proteomics
125 of 132
Describe features of epigenetics (2)
Transcription (de-methylation and acetylation). No transcription (methylation and de-acetylation)
126 of 132
Describe features of RNA targeting
RNAi (cancer therapeutic), controls expression of miRNA, use in mechanism of gene silencing , challenges (IP, stability/delivery, nanoparticle approach)
127 of 132
Describe features of the antisense approach (1)
Nucleus produces mRNA, ribosome, production of protein. Antisense DNA oligonucleotide locks onto RNA to form duplex, blocks transcription. Need to know sequence of gene you want to deregulate. Difficult to give nucleic acid as oral treatment
128 of 132
Describe features of the antisense approach (2)
Fomivirsen and mipomersen (and more development)
129 of 132
Describe features of CRISPR-Cas9 gene editing (1)
CRISPR inhibition (broken/dead Cas9 enzyme will block binding of other proteins e.g. RNA polymerase, prevent expression of gene). CRISPR activation (activated protein attached to dead Cas9 protein to stimulate gene expression of specific gene)
130 of 132
Describe features of CRISPR-Cas9 gene editing (2)
Also use of epigenetics and inducible CRISPR
131 of 132
Give an example of other approaches for treatment
Replace DNA in tumour cells with DNA containing 8 base pairs
132 of 132

Other cards in this set

Card 2

Front

What are the types of DNA adducts?

Back

DNA cleavage, intercalation, intra-strand cross-link, interstrand cross-link, topoisomerase adduct, mono-adduct

Card 3

Front

Give examples of methylating agents

Back

Preview of the front of card 3

Card 4

Front

Give examples of alkylating agents

Back

Preview of the front of card 4

Card 5

Front

Give examples of cross-linking agents

Back

Preview of the front of card 5
View more cards

Comments

No comments have yet been made

Similar Pharmacy resources:

See all Pharmacy resources »See all Nucleic Acid Based Cancer Therapies resources »