22.05.2022 Views

DƯỢC LÍ Goodman & Gilman's The Pharmacological Basis of Therapeutics 12th, 2010

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

1688 platinum complexes do not form carbonium ion intermediates

like other alkylating agents or formally alkylate

DNA, they covalently bind to nucleophilic sites on

DNA and share many pharmacological attributes with

alkylators.

SECTION VIII

CHEMOTHERAPY OF NEOPLASTIC DISEASES

Chemistry. Cisplatin and carboplatin are divalent, inorganic, watersoluble,

platinum-containing complexes. Oxaliplatin, a tetravalent

complex, does not display cross-resistance to the divalent compounds

in some experimental tumors. The coordination of di- or

tetravalent platinum with various organic adducts reduces its renal

toxicity and stabilizes the metal ion, as compared to the inorganic

divalent platinum ion.

Mechanism of Action. Cisplatin, carboplatin, and oxaliplatin

enter cells by an active Cu 2+ transporter, CTR1,

and in doing so rapidly degrade the transporter (Kruh,

2003). The compounds are actively extruded from cells

by ATP7A and ATP7B copper transporters and by multidrug

resistance protein 1 (MRP 1); variable expression

of these transporters may contribute to clinical

resistance (Dolan and Fitch, 2007). Inside the cell, the

chloride, cyclohexane, or oxalate ligands of the three

analogs are displaced by water molecules, yielding a

positively charged and highly reactive molecule. In the

primary cytotoxic reaction, the aquated species of the

drug then reacts with nucleophilic sites on DNA and

proteins.

Aquation of cisplatin is favored at the low concentrations of

chloride inside the cell and in the urine. High concentrations of chloride

stabilize the drug, explaining the effectiveness of chloride diuresis

in preventing nephrotoxicity (see “Clinical Toxicities”). The

activated platinum complexes can react with electron-rich molecules,

such as sulfhydryls, and with various sites on DNA, forming both

intrastrand and interstrand cross-links. The N-7 of guanine is a particularly

reactive site, leading to platinum cross-links between adjacent

guanines (GG intrastrand cross-links) on the same DNA strand;

guanine–adenine cross-links also form and may contribute to cytotoxicity.

Interstrand cross-links form less frequently. DNA-platinum

adducts inhibit replication and transcription, lead to single- and

double-stranded breaks and miscoding, and if recognized by p53 and

other checkpoint proteins, cause induction of apoptosis. Although

no quantitative relationship between platinum-DNA adduct formation

and efficacy has been documented, the ability of patients to form

and sustain platinum adducts appears to be an important predictor of

clinical response (Reed et al., 1988). The analogs differ in the conformation

of their adducts and the effects of adduct on DNA structure

and function. Oxaliplatin and carboplatin are slower to form

adducts. The oxaliplatin adducts are bulkier and less readily repaired,

create a different pattern of distortion of the DNA helix, and differ

from cisplatin adducts in the pattern of hydrogen bonding to adjacent

segments of DNA (Sharma et al., 2007).

Unlike the other platinum analogs, oxaliplatin exhibits a cytotoxicity

that does not depend on an active MMR system, which may

explain its greater activity in colorectal cancer. It also seems less

dependent on the presence of high mobility group (HMG) proteins

that are required by the other platinum derivatives. Testicular cancers

have a high concentration of HMG proteins and are quite sensitive

to cisplatin. Basal-type breast cancers, such as those with BRCA1 and

BRCA2 mutations, lack Her 2 amplification and hormone-receptor

expression and appear to be uniquely susceptible to cisplatin

through their upregulation of apoptotic pathways governed by p63

and p73 (Deyoung and Ellisen, 2007). The specificity of cisplatin

with regard to phase of the cell cycle differs among cell types,

although the effects of cross-linking are most pronounced during

the S phase.

The platinum analogs are mutagenic, teratogenic, and carcinogenic.

Cisplatin- or carboplatin-based chemotherapy for ovarian

cancer is associated with a 4-fold increased risk of developing

secondary leukemia.

Resistance to Platinum Analogs. Resistance to the platinum analogs

likely is multifactorial, and the compounds differ in their degree

of cross-resistance. Carboplatin shares cross-resistance with cisplatin

in most experimental tumors, while oxaliplatin does not. A

number of factors influence sensitivity to platinum analogs in

experimental cells, including intracellular drug accumulation, as

determined by the uptake and efflux transporters; intracellular levels

of glutathione and other sulfhydryls such as metallothionein

that bind to and inactivate the drug (Meijer et al., 1990); and rates

of repair of DNA adducts. Repair of platinum-DNA adducts

requires participation of the NER pathway. Inhibition or loss of

NER increases sensitivity to cisplatin in ovarian cancer patients,

while overexpression of NER components is associated with poor

response to cisplatin or oxaliplatin-based therapy in lung, colon,

and gastric cancer (Paré et al., 2008). Higher levels of expression

of the NER component, ERCC1, in tumor cells and peripheral

white blood cells are associated with a lower response rate in

patients with solid tumors (Dolan and Fitch, 2007).

Resistance to cisplatin, but not oxaliplatin, appears to be

partly mediated through loss of function in the MMR proteins

(hMLH1, hMLH2, or hMSH6), which recognize platinum-DNA

adducts and initiate apoptosis.

In the absence of effective repair of DNA-platinum adducts,

sensitive cells cannot replicate or transcribe affected portions of the

DNA strand. However, it is clear that some DNA polymerases can

bypass adducts, especially those created by cisplatin. Oxaliplatin

adducts are less easily bypassed. It remains unproven whether these

polymerases contribute to resistance. Cisplatin resistance related to

loss of active uptake has been demonstrated in yeast; overexpression

of copper efflux transporters, ATP7A and ATP7B, correlates with poor

survival after cisplatin-based therapy for ovarian cancer (Kruh, 2003).

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!