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.

Absorption, Fate, and Excretion. Pharmacokinetics and systemic

actions of teriparatide on mineral metabolism are the same as for

PTH. Teriparatide is administered by once-daily subcutaneous injection

of 20 μg into the thigh or abdomen. With this regimen, serum

PTH concentrations peak at 30 minutes after the injection and

decline to undetectable concentrations within 3 hours, whereas the

serum calcium concentration peaks at 4-6 hours after administration.

Based on aggregate data from different dosing regimens, teriparatide

bioavailability averages 95%. Teriparatide clearance averages

62 L/hour in women and 94 L/hour in men, which exceeds normal

liver plasma flow, consistent with both hepatic and extrahepatic PTH

removal. The serum t 1/2

of teriparatide is ~1 hour when administered

subcutaneously versus 5 minutes when administered intravenously.

The longer t 1/2

following subcutaneous administration reflects the

time required for absorption from the injection site. The elimination

of PTH(1-34) and full-length PTH proceeds by nonspecific enzymatic

mechanisms in the liver, followed by renal excretion.

Clinical Effects. In postmenopausal women with osteoporosis, teriparatide

increases BMD and reduces the risk of vertebral and nonvertebral

fractures. Several laboratories have examined the effects of

intermittent PTH on BMD in patients with osteoporosis. In these studies,

teriparatide increased axial bone mineral, although initial reports

of effects on cortical bone were disappointing. Co-administration of

hPTH(1-34) with estrogen or synthetic androgen led to impressive

gains in vertebral bone mass or trabecular bone. However, in some

early studies there was only maintenance or even loss of cortical

bone. Vitamin D insufficiency in patients at baseline or pharmacokinetic

differences involving bioavailability or circulating half-life may

have contributed to observed differences on cortical bone. The most

comprehensive studies to date established the value of daily

hPTH(1–34) administration on total BMD, with significant elevations

of BMD in lumbar spine and femoral neck and with significant reductions

of vertebral and nonvertebral fracture risk in osteoporotic

women (Neer et al., 2001) and men (Finkelstein et al., 2003).

Candidates for teriparatide treatment include women who

have a history of osteoporotic fracture, who have multiple risk factors

for fracture, or who failed or are intolerant of previous osteoporosis

therapy. Men with primary or hypogonadal osteoporosis are

also candidates for treatment with teriparatide. However, the effect

on fracture incidence in this group has not been established.

Adverse Effects. In rats, teriparatide increased the incidence of

osteosarcoma (Vahle et al., 2004). Because of these findings, a black

box warning appears on the FORTEO package insert. The clinical relevance

of this finding is unclear, especially because patients with

primary hyperparathyroidism have marked elevation of serum PTH

without a greater incidence of osteosarcoma. Nonetheless, teriparatide

should not be used in patients who are at increased baseline

risk for osteosarcoma (including those with Paget’s disease of

bone, unexplained elevations of alkaline phosphatase, open epiphyses,

or prior radiation therapy involving the skeleton). The safety and

efficacy of FORTEO have not been evaluated for extended use beyond

2 years of treatment and therapy should be limited to no more than

2 years. A tumor registry-based analysis of the occurrence of

osteosarcoma in teriparatide-treated patients is ongoing; cases of

osteosarcoma associated with the drug should be reported to the FDA.

Increases in bone density acquired during therapy with teriparatide

can maintained or extended with subsequent use of bisphosphonates

(Black et al., 2005).

Full-length PTH(1-84) has been associated with osteosarcomas

in rats (Jolette et al., 2006) although not in humans. The significance

and interpretation of the toxicological findings has been questioned

(Tashjian and Goltzman, 2008). However, adverse effects include exacerbation

of nephrolithiasis and elevation of serum uric acid levels.

CALCIUM SENSOR MIMETICS:

CINACALCET

Calcimimetics are drugs that mimic the stimulatory effect

of calcium on the calcium-sensing receptor (CaSR) to

inhibit PTH secretion by the parathyroid glands. By

enhancing the sensitivity of the CaSR to extracellular

Ca 2+ , calcimimetics lower the concentration of Ca 2+ at

which PTH secretion is suppressed. Inorganic di- and

trivalent cations, along with polycations such as spermine,

aminoglycosides (e.g., streptomycin, gentamicin,

and neomycin) and polybasic amino acids (e.g., polylysine)

are full agonists and are referred to a type I calcimimetics.

Phenylalkylamine derivatives that are

allosteric CaSR modulators that require the presence of

Ca 2+ or other full agonists to enhance the sensitivity of

activation without altering the maximal response are designated

type II calcimimetics. Cinacalcet (SENSIPAR)

(Figure 44–12) is approved for the treatment of secondary

hyperparathyroidism owing to chronic renal disease

and for patients with hypercalcemia associated with

parathyroid carcinoma. Cinacalcet lowers serum PTH

levels in patients with normal or reduced renal function

(Padhi and Harris, 2009).

In clinical trials, cinacalcet at 20- to 100-mg doses lowered

PTH levels in a concentration-dependent manner by 15–50% and

the serum calcium × phosphate product by 7% compared with

placebo (Franceschini et al., 2003). Cinacalcet also effectively

reduced PTH levels in patients with primary hyperparathyroidism

and provided sustained normalization of serum calcium without

altering bone mineral density (Shoback et al., 2003). Long-term control

of PTH levels was achieved during a 3-year study of cinacalcet,

suggesting that resistance does not develop. There currently is insufficient

information to know whether reducing serum PTH levels with

H 3 C

NH

CF 3

Figure 44–12. Structure of cinacalcet. Cinacalcet exists as optical

isomers. The R-enantiomer, which is more active, is shown.

1297

CHAPTER 44

AGENTS AFFECTING MINERAL ION HOMEOSTASIS AND BONE TURNOVER

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

Saved successfully!

Ooh no, something went wrong!