22.05.2022 Views

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

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

1744

O

HO

cell

membrane

IGF1R

Other TK receptors

PI3K

stress

energy depletion

amino acid status

Ras

SECTION VIII

O

HO

N

O

O O

O O

O

O

OH

O

Rapamycins

FKBP12

Akt

mTORC1

mTORC2

MAPK

CHEMOTHERAPY OF NEOPLASTIC DISEASES

RAPAMYCIN (SIROLIMUS)

Mechanisms of Action and Resistance. The rapamycins inhibit an

enzyme complex, mTORC1, which occupies a downstream position

in the PI3 kinase pathway (Figure 62–4). mTOR forms the mTORC1

complex with a member of the FK506-binding protein family,

FKBP12. Among other actions, mTORC1 phosphorylates S6 kinase

and also relieves the inhibitory effect of 4EBP on initiation factor elf-

4E, therby promoting protein synthesis and metabolism. The antitumor

actions of the rapamycins result from their binding to FKBP12

and inhibition of mTORC1. Rapamycin and its congeners have

immunosuppressant effects, inhibit cell-cycle progression and angiogenesis,

and promote apoptosis.

Resistance to mTOR inhibitors is incompletely understood

but may arise through the action of a second mTOR complex,

mTORC2, which is unaffected by rapamycins and which regulates

AKT kinase. Experimental work suggests that inhibition of

mTORC1 leads to mTORC2 activation of AKT kinase and the MAP

kinase pathway, and these actions may be responsible for incomplete

responses or resistance of rapamycins (Carracedo et al., 2008). Dual

mTORC1 and mTORC2 inhibitors are in clinical development.

S6K1

translation

autophagy

4EBP

translation

metabolism

proliferation

survival

proliferation

metabolism

Figure 62–4. Insulin-like growth factor 1 receptor (IGF-1R) and

other tyrosine kinase (TK) growth factor receptors signal through

multiple pathways. A key pathway is regulated by phosphatidylinositol-3

kinase (PI3K) and its downstream partner, the mammalian

target of rapamycin (mTOR). Rapamycins complex with

FKBPP12 to inhibit the mTORC1 complex. mTORC2 remains

unaffected and responds by upregulating Akt, driving signals

through the inhibited mTORC1. The various downstream outputs

of the two complexes are shown. Phosphorylation of 4EBP

by mTOR inhibits the capacity of 4EBP to inhibit eif-4E and

slow metabolism. 4EBP, eukaryotic initiation factor 4e (eif-4E)

binding protein; S6K1, S6 kinase 1; FKBP12, the immunophilin

target (binding protein) for tacrolimus (FK506).

Absorption, Fate, and Excretion. The FDA has approved both temsirolimus

and everolimus for treatment of renal cancer. Temsirolimus

prolongs survival and delays disease progression in patients with

advanced and poor- or intermediate-risk renal cancer, as compared

to standard interferon-α treatment. Everolimus, as compared to

placebo, prolongs survival in patients who had failed initial treatment

with anti-angiogenic drugs (Motzer et al., 2008). mTOR

inhibitors also have antitumor activity against mantle cell lymphomas

(Ansell et al., 2008) and are under active investigation in

combination with hormonal therapies, EGFR inhibitors, and cytotoxic

drugs.

For renal-cell cancer, temsirolimus is given in weekly doses

of 25 mg, intravenously, while everolimus is administered orally

in doses of 10 mg daily. Both drugs should be administered in the

fasting state at least 1 hour before a meal. Both parent molecules

are metabolized by CYP3A4. Temsirolimus has a plasma t 1/2

of

30 hours; its primary metabolite, sirolimus, has a longer t 1/2

of

53 hours. Because sirolimus has equivalent activity as an inhibitor

of mTORC1, and has a greater AUC, sirolimus is likely the more

important contributor to antitumor action in patients (Hutson

et al., 2008). Everolimus has a plasma t 1/2

of 30 hours, and on a

weekly schedule at doses of 20 mg, it maintains inhibition of

mTORC1 for 7 days in white blood cells (O’Donnell et al., 2008).

At their usual clinical doses, both agents provide peak drug levels

of ~1 μM.

Both drugs are susceptible to interactions with other agents

that affect CYP3A4 activity. Ketoconazole, a CYP3A4 inhibitor,

causes a 2- to 3-fold increase in the AUCs of both temsirolimus and

sirolimus, while inducers of metabolism such as rifampin or phenytoin

can decrease the C Pmax

of temsirolimus by 36% and decrease

the AUC of its metabolite by >50%. The dose of temsirolimus

should be doubled in the presence of inducers and reduced by half

in the presence of ketoconazole (Hutson et al., 2008). Hepatic dysfunction

delays drug clearance; for everolimus, the dose should be

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

Saved successfully!

Ooh no, something went wrong!