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.

508 doses of 20-25 mg morphine given subcutaneously. The use of

higher doses of propoxyphene is accompanied by untoward effects

including toxic psychoses. Very large doses produce some respiratory

depression in morphine-tolerant addicts, suggesting that crosstolerance

between propoxyphene and morphine is incomplete.

Abrupt discontinuation of chronically administered propoxyphene

hydrochloride (up to 800 mg/day given for almost 2 months) results

in mild abstinence phenomena, and large oral doses (300-600 mg)

produce subjective effects that are considered pleasurable by postaddicts.

The drug is quite irritating when administered either intravenously

or subcutaneously, so abuse by these routes results in

severe damage to veins and soft tissues.

SECTION II

NEUROPHARMACOLOGY

Therapeutic Uses. In 2009, the European Medicines Agency completed

a review of the safety and effectiveness of dextropropoxyphene-containing

medicines and concluded that the benefits

do not outweigh the risks and recommended a gradual withdrawal of

marketing authorization throughout the EU (European Medicines

Agency, 2009). The conclusion reached by the agency included an

assessment that dextropropoxyphene-containing medicines are weak

painkillers with a limited effectiveness in the treatment of pain and a

narrow therapeutic index. Furthermore, the agency judged acetaminophen

(paracetamol) containing combinations with dextropropoxyphene

to be no more effective than acetaminophen alone. In

the U.S., propoxyphene has been approved for mild to moderate pain

since 1957. Recently the drug has been labeled with additional warnings

related to the risk of rapidly fatal effects in the event of an overdose

(FDA, 2009). Thus, in the U.S., propoxyphene should not be

prescribed to patients who are suicidal or have a history of suicidal

ideation. In addition, higher than expected serum levels of

propoxyphene should be anticipated from the concommitant administration

of strong CYP3A4 inhibitors such as ritonavir, ketoconazole,

itraconazole, clarithromycin, nelfinavir, nefazadone, amiodarone,

amprenavir, aprepitant, diltiazem, erythromycin, fluconazole, fosamprenavir,

grapefruit juice, and verapamil. Propoxphene alternatives

should be considered for patients receiving a strong CYP3A4

inhibitor and others at risk for overdose, particularly those with preexisting

heart disease.

Other Opioid Agonists

Tramadol. Tramadol (ULTRAM) is a synthetic codeine analog that is

a weak MOR agonist. Part of its analgesic effect is produced by inhibition

of uptake of norepinephrine and serotonin. In the treatment

of mild to moderate pain, tramadol is as effective as morphine or

meperidine. However, for the treatment of severe or chronic pain,

tramadol is less effective. Tramadol is as effective as meperidine in

the treatment of labor pain and may cause less neonatal respiratory

depression.

Pharmacokinetics. Tramadol is 68% bioavailable after a single oral

dose and 100% available when administered intramuscularly. Its

affinity for the μ opioid receptor is only 1/6000 that of morphine.

However, the primary O-demethylated metabolite of tramadol is

two to four times as potent as the parent drug and may account for

part of the analgesic effect. Tramadol is supplied as a racemic

mixture, which is more effective than either enantiomer alone. The

(+)-enantiomer binds to the receptor and inhibits serotonin uptake.

The (–)-enantiomer inhibits norepinephrine uptake and stimulates

α 2

adrenergic receptors (Lewis and Han, 1997). Tramadol undergoes

extensive hepatic metabolism by a number of pathways, including

CYP2D6 and CYP3A4, as well as by conjugation with subsequent

renal excretion. The rate of formation of the active metabolite is

dependent on CYP2D6 and therefore is subject to both metabolic

induction and inhibition. The elimination t 1/2

is 6 hours for

tramadol and 7.5 hours for its active metabolite. Analgesia begins

within an hour of oral dosing and peaks within 2-3 hours. The duration

of analgesia is ~6 hours. The maximum recommended daily

dose is 400 mg.

Side Effects and Adverse Effects. Common side effects of tramadol

include nausea, vomiting, dizziness, dry mouth, sedation, and

headache. Respiratory depression appears to be less than with

equianalgesic doses of morphine, and the degree of constipation

is less than that seen after equivalent doses of codeine (Duthie,

1998). Tramadol can cause seizures and possibly exacerbate

seizures in patients with predisposing factors. While tramadolinduced

analgesia is not entirely reversible by naloxone, tramadolinduced

respiratory depression is reversed by naloxone. However,

the use of naloxone increases the risk of seizure in patients

exposed to tramadol. Misuse, diversion, physical dependence,

abuse, addiction, and withdrawal have been reported in conjunction

with the use of tramadol. Tramadol has been shown to reinitiate

physical dependence in some patients who have previously

been dependent on other opioids, thus it should be avoided in

patients with a history of addiction. Precipitation of withdrawal

necessitates that tramadol be tapered prior to discontinuation.

Tramadol should not be used in patients taking MAO inhibitors

(Lewis and Han, 1997), SSRIs, or other drugs that lower the

seizure threshold (described earlier).

Tapentadol. Tapentadol (NUCYNTA) is structurally and mechanistically

similar to tramadol. It displays a mild opioid activity and possesses

monoamine reuptake inhibitor activity. It is considered similar

to tramadol in activity, efficacy, and side-effect profile. Like tramadol,

it should not be used concurrently with agents that enhance

monamine activity or lower the seizure threshold, such as MAO

inhibitors and SSRIs. The major pathway of tapentadol metabolism

is conjugation with glucuronic acid; ~70% of the dose is excreted in

urine in the conjugated form.

OPIOID AGONISTS/ANTAGONISTS

AND PARTIAL AGONISTS

The drugs described in this section differ from clinically

used μ opioid receptor agonists. Drugs such as nalbuphine

and butorphanol are competitive MOR antagonists but

exert their analgesic actions by acting as agonists at KOR

receptors. Pentazocine qualitatively resembles these

drugs, but it may be a weaker MOR receptor antagonist

or partial agonist while retaining its KOR agonist activity.

Buprenorphine, on the other hand, is a partial agonist

at MOR. The stimulus for the development of mixed

agonist–antagonist drugs was a desire for analgesics with

less respiratory depression and addictive potential.

However, the clinical use of these compounds is limited

by undesirable side-effects and limited analgesic effects.

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

Saved successfully!

Ooh no, something went wrong!