117
Views
18
CrossRef citations to date
0
Altmetric
Review

Leuprorelin depot injection: patient considerations in the management of prostatic cancer

&
Pages 513-526 | Published online: 11 Apr 2008

Abstract

Hormone therapy is well established for treating patients with prostate cancer and remains the mainstay of the treatment of metastatic and locally advanced disease, this article reviews the rationale for its use, its different forms, and complications and controversies still surrounding some of its modalities. Availability of long-acting synthetic luteinizing hormone-releasing hormone (LHRH) agonists revolutionized the hormonal treatment of prostate cancer, and helped to avoid the emotional and psychological effects related to surgical castration. The depot formula has gained wide acceptance from both patients and physicians. This review emphasizes the newer, long-acting formula, leuprorelin (leuprolide acetate), especially the 6-month formula, its advantage over over shorter-acting depot products, and its potential to become a standard of care for patients eligible for androgen deprivation therapy.

Introduction

In the post prostate-specific antigen (PSA) era, screening for prostate cancer has not only led to a stage migration, but also to a higher incidence of the disease. Such a trend has decreased the incidence of metastatic disease at diagnosis and paralleled the decrease in the mortality rate from prostate cancer.

The incidence of metastatic disease was only 1.6% of patients enrolled in the Cancer of the Prostate Strategic Urologic Research Endeavor (CaPSURE), diagnosed between 1998 and 2003 (CitationRyan et al 2006). On the other hand the population failing the initial treatment is increasing, and 40% of the patients treated with curative intent eventually end up with PSA recurrence (CitationWard and Moul 2005), raising the question of whether there is any evidence that aggressive intervention in these patients may result in cure.

Because many patients with T3 disease or local lymph node metastases progress to distant metastases, some authors recommended that the concept of advanced prostate cancer should also include stages C and D1 (CitationMoul 2004) (T3, T4, and any T N1).

Today, in addition to its well-established role in treating patient with metastatic disease, androgen deprivation therapy (ADT) is sometimes used to treat patients with biochemical failure even without evidence of local or systemic recurrence. This therapy is also used as an adjunct to radiation for high risk localized disease.

The treatment of locally advanced and metastatic prostate cancer is only palliative, and most patients who respond initially to ADT ultimately evolve towards hormone refractory disease within 2 years, and at this stage, unfortunately, alternative modalities such as chemotherapy become of limited value, with a survival of only 18–24 months.

This inevitable evolution led to new strategies being explored for delaying progression to androgen independence, which may involve targeting of anti-apoptotic factors, use of chemotherapy at the time of androgen ablation, or blockage or down-regulation of androgen receptor (AR) activity (CitationPetrylak 2005). Among those novel options is a broad range of promising strategies such as targeting signal transduction pathways, cell cycle regulation and differentiation, and angiogenesis (CitationCavarretta et al 2005).

Prostate cancer: a hormone sensitive malignancy

The prostate – testis relationship has been known since the 18th century when John Hunter (CitationAndroutsos 1998) demonstrated in 1786 that castration in young male animals prevented further growth of the prostate, whereas in the adult it caused atrophy. Hunter also observed that aging eunuchs never suffered from symptoms of a hypertrophied prostate (CitationHome 1811).

The second half of the 19th century marked a growing interest in orchiectomy for the treatment of prostatic hyperplasia, particularly urinary retention. Symptomatic improvement in over half of patients with an enlarged prostate treated with castration was reported by William White in 1895 and 1904 (CitationWhite 1895, Citation1904).

The development of prostate cancer requires the presence of a prostate gland and a source of androgens. It has been shown that androgens produced elsewhere can be activated in the prostatic tissue, as prostate cancer never occurs in those with testicular feminization syndrome and in patients with 5-alfa reductase deficiency (CitationNewling 1996).

Before 1940s there was no effective treatment for advanced prostate cancer. In 1939 Charles Huggins, aware of androgens sensitivity of the prostate gland, proposed orchiectomy to control prostate cancer. He demonstrated that castration decreased the height of prostatic epithelial cells in normal prostatic tissue, and that testosterone administration stimulated the secretory activity of dogs’ prostatic cells (CitationHuggins and Clark 1940); furthermore, he proved that castration produced clinical pain relief and a stabilization or regression of metastatic osseous lesions (CitationHuggins et al 1941) and reduction in acid phosphatase level (CitationHuggins and Hodges 1941). Huggins soon realized, however, that the same results could be achieved by the less drastic procedure of the administration of female sex hormones to neutralize the effect of androgens produced by the testicles. Consequently, in 1941 he began to inject his patients with the hormones stilbestrol and hexestrol.

Other forms of castration have been developed, such as the technique of radio-orchiectomy (CitationKeyes and Ferguson 1936), consisting of irradiation of the testis in patients with advanced prostate cancer.

Ninety to 95% of the androgens are produced by the testes and only 5%–10% by the adrenals. These small amounts of androgens continue to support tumor growth.

The involvement of the adrenal gland in hormonal control of prostate malignancies was supported by reports of an increase in the urinary concentration of 17-keto-steroids in patients after castration (CitationSatterthwaite et al 1941; CitationSchröder 2002). The recognition of the role of adrenal androgens in prostate cancer led to the development of the therapeutic strategy known as combined androgen blockade.

The early form of this approach combined orchiectomy and bilateral adrenalectomy. The first procedure in a patient with prostate cancer was carried out by Huggins and William Wallace Scott in 1945 (CitationHuggins and Scott 1945), who demonstrated a clinical benefit of such procedure in men with disease recurrence after orchiectomy.

The initial success of hormonal therapy led on the one hand to less interest in radical surgical and radiation treatment, even for localized disease. On the other hand, it led to a tendency to remove sound organs with vital functions such as adrenals (adrenalectomy) and the pituitary gland (surgical ablation [CitationSilverberg and Britt 1979]) and external irradiation (CitationMurphy and Schwippert 1951) of the pituitary gland, as well as the implantation of radioactive material (CitationFergusson 1957), in hope of completely suppressing androgens and therefore curing or slowing disease progression.

CitationSchally and associates (1971) purified the decapeptide gonadotropin-releasing hormone, referred to as LHRH. The availability of long-acting synthetic luteinizing hormone-releasing hormone (LHRH) agonists in the 1980s revolutionized the hormonal treatment of prostate cancer, enabling many men to avoid the emotional and psychological effects of surgical castration (CitationMcLeod 2003).

Anti-androgens block testosterone action in target tissues by interfering with testosterone or dihydrotesterone (DHT) binding to the steroid-binding domain of the androgen receptor. Steroidal anti-androgens, such as cyproterone acetate, also have some ability to block luteinizing hormone (LH) secretion, and are therefore associated with many of the sexual effects seen with LHRH agonists (CitationSee 2003).

By contrast, the non-steroidal anti-androgens have been associated with a far more beneficial sexual side effect profile. Non-steroidal anti-androgens, such as flutamide, were first used in the mid 1970s (CitationSogani et al 1975) and steroidal anti-androgens such as cyproterone acetate were approved for use in the 1980s (CitationJacobi 1983).

The gonadotropin-releasing hormone (GnRH) antagonists represent the newest class of agents introduced for the hormonal treatment of prostate cancer. The advantages of GnRH antagonists are that they do not cause the initial surge in testosterone associated with LHRH agonists and they rapidly achieve castrate androgen levels.

In clinical trials, monotherapy with abarelix was shown to achieve medical castration more rapidly than leuprorelin (leuprolide acetate) alone or in combination with bicalutamide and without an early surge in testosterone (CitationTrachtenberg et al 2002).

Physiology: hypothalamus – pituitary – testis axis

LHRH, also known as GnRH, is secreted in a pulsatile fashion by the hypothalamus. The increase in LHRH stimulates the anterior pituitary to secrete LH and follicle-stimulating hormone into the systemic circulation (CitationDenmeade and Isaacs 2004). LH stimulates the Leydig cells of the testes to secrete testosterone.

Testosterone is converted into a more potent compound, DHT. Conversion occurs in the cytoplasm of prostatic cells by the enzyme 5-alfa reductase. In addition to testosterone, adrenal androgens may be converted to DHT (CitationSandow et al 1988). In the cytosol, DHT binds to the androgen receptor and crosses into the nucleus. The activated androgen receptor binds to specific DNA sequences that influence gene expression, stimulate the synthesis of specific proteins such as PSA, and trigger cell proliferation.

In the normal prostate, androgen is the major growth factor through two mechanisms: on the one hand, it stimulates cellular proliferation of prostatic epithelial cells; on the other, it blocks the potentially high apoptotic rate of epithelial cells. The net balance determines the overall number of prostate cells (CitationIsaacs 1984, Citation1994).

Testosterone-lowering strategies ()

The first method of permanent castration was bilateral orchiectomy, and the first reversible method was diethylstilbestrol (DES) (CitationByar and Corle 1988).

Table 1 Methods to interrupt the production of and/or block the action of testosterone

Therapeutic approaches to interrupt the production and/or block the action of testosterone include:

  1. Suppression of testosterone via surgical removal of the testes or use of estrogens;

  2. LHRH agonists to down-regulate the pituitary and prevent release of LH or by using LHRH antagonists to directly suppress LH release;

  3. Inhibition of 5-alfa reductase conversion of testosterone to DHT in the prostate;

  4. Blocking the binding of DHT to the androgen receptors in the prostate.

These strategies can be used individually or in various combinations.

In 1983, ketoconazole in high dose was identified as able to produce castrate levels of androgens within 4–8 h of the first dose (CitationTrachtenberg and Point 1983), and represents an alternative treatment where urgent androgen ablation is required (CitationLowe and Bamberger 1990).

Biological effects of androgen deprivation

Anterior pituitary – testis axis

With longer-term administration, a resetting of the anterior pituitary receptor occurs, with subsequent reduction in LH along with follicle-stimulating hormone (FSH) release, resulting in achievement of castrate levels of testosterone (CitationConn et al 1984). Chronic exposure to LHRH ultimately suppressed testosterone by desensitizing pituitary cells through down-regulation of the LHRH receptors (CitationVan Loenen et al 2002).

Castration, the time-honored frontline treatment for metastatic prostate cancer, was previously defined by induction of a serum testosterone level of <50 ng/mL (CitationCoppage and Cooner 1965; CitationYoung and Kent 1968). Recent literature redefines this upper limit as <20 ng/mL (CitationOefelein et al 2000).

Prostate cell level

Effects of androgen deprivation include apoptosis of both cancer cells and benign prostatic epithelium (CitationDenmead et al 1996; CitationButtyan et al 2000). One of the earliest events, occurring prior to the initiation of epithelial apoptosis, is degeneration of the prostatic vasculature. This has been observed in animal models as well as in the human prostate (CitationFranck-Lissbrant et al 1998; CitationShabisgh et al 1999). A recent study has shown that in patients on complete androgen blockade therapy (CAB), there were morphological changes within the prostatic tissues as early as 7 days after initiating ADT, similar to the response to castration. These changes induce significant involution within prostatic tissues over 7–28 days, but allow the persistence of some viable tumor cells capable of proliferation (CitationMercader et al 2007). This event is mediated at least in part by decreased expression of vascular endothelial growth factor (VEGF), an androgen-regulated gene, in the prostatic epithelium and stroma (CitationBurchardt et al 2000). This phenomenon of apoptosis does not affect basal cells which remain intact. A high percentage of androgen-independent cancers appear to maintain expression of the AR gene, but many have genetic alterations that change receptor activity. Altered expression of AR co-activators may occur in recurrent cancer, and AR mutations may occur in as many as 40%–50% of such cancers. Proteins involved in cell survival and resistance to apoptosis are over-expressed in androgen-independent prostate cancer cells or tumors (CitationPeehl 2001).

Indication of ADT in prostate cancer

Today, in addition to its well-established role in treating patients with metastatic disease, ADT is sometimes used to treat patients with biochemical failure even without evidence of local or systemic recurrence. This therapy is also used as an adjunct in patients undergoing radiation for high-risk localized disease.

Controversies around hormonal therapy

Several controversies still surround the modalities of this therapy ():

Table 2 Controversies around hormonal therapy for prostate cancer

  • Monotherapy or CAB?

  • Early versus delayed?

  • Continuous or intermittent?

  • Adjuvant or neoadjuvant?

  • Secondary manipulation after failure of initial hormonal therapy?

  • Hormone after PSA recurrence?

  • Hormone for localized prostate cancer.

  • Role of anti-androgens alone?

  • Role of anti-5AR + antiandrogens?

LHRH agonists flare issue

During the initial 1–2 weeks the LHRH receptor is overstimulated, initially resulting in testosterone surge and potential for clinical flare. There is a delay in testosterone reduction and a surge in testosterone and dihydro-testerone as well as elevation of PSA. Symptomatic flare may result. The impact of the increase in FSH is only now undergoing evaluation, but this too may have an untoward effect on prostatic carcinoma. The flare phenomenon may be life threatening if an LHRH agonist is administered to men with high-volume metastatic disease. The clinical consequence of the flare is prevented by pretreatment with an anti-androgen, which inhibits the stimulatory effect of the testosterone surge at the level of the androgen receptor (CitationThompson 2001).

Patients with symptomatic advanced prostate cancer such as those with significant obstructive voiding symptoms merit a flare-free induction of hormonal therapy, as do patients with significant back pain and early neurologic sequelae: both urinary retention and paraplegia are side effects to be avoided.

Optimal blockade is achieved by pretreatment with an appropriate agent (eg, anti-androgen), or for the patient in whom maximum blockade is desirable immediately, consideration could be given to the use of ketoconazole, which dramatically reduces testosterone in only a matter of hours.

Failure to achieve castrate level

A small but potentially important subgroup of men on depot LHRH agonist therapy fail to achieve or maintain a castrate level of testosterone (CitationOefeien and Comum 2000), a finding that supports the need for monitoring testicular response during LHRH agonist therapy (CitationMorote et al 2006).

Cost of treatment

For an individual patient the cost of LHRH agonist treatment surpassed the cost of surgery at less than 4.2–5.3 months, and for combined androgen blockade (LHRH agonists and non-steroidal anti-androgens) at less than 2.7–3.4 months (CitationMariani et al 2001). Except for patients with short anticipated survival, current medical androgen suppressive treatment options are more costly than bilateral orchiectomy.

For men who accept it, bilateral orchiectomy is likely to be the most cost-effective androgen suppression strategy. Combined androgen blockade is the least economically attractive option, yielding small health benefits at high relative costs (CitationBayoumi et al 2000).

Complications of hormonal therapy

The common side effects of hormonal manipulation are hot flushes. Changes in the hormonal balance induced by hormonal manipulation in men with prostate cancer can increase the likelihood of gynecomastia, which usually resolves spontaneously after cessation of hormonal manipulation during the first year (CitationKumar et al 2005).

Men with prostate cancer who are initiating ADT have a 5- to 10-fold increased loss of bone density at multiple skeletal sites compared with either healthy controls or men with prostate cancer who are not on ADT. Bone loss is maximal in the first year after initiation of ADT, suggesting initiation of early preventive therapy (CitationGreenspan et al 2005; CitationShahinian et al 2005). Hormone therapy does not appear to cause clinically significant changes in depression among men with locally advanced prostate cancer. However, fatigue increased significantly over the study period (CitationKoupparis et al 2004).

The current data suggest that men with prostate cancer who are receiving long-term ADT are at risk for developing insulin resistance, hyperglycemia (CitationBasaria et al 2006), and the occurrence of metabolic syndrome (CitationBraga-Basari et al 2006), thus leading to an increased risk of cardiovascular complications.

Compared with normal men, total and free testosterone levels during treatment were 1.8-fold and 2.3-fold higher in obese men. These differences may contribute to the association between obesity and increased prostate cancer mortality (CitationSmith 2007), loss of sexual desire, erectile dysfunction, and anemia.

Pituitary adenomas (CitationMassoud et al 2006) together with subcutaneous granulomas (CitationWhitaker et al 2002) mimicking metastatic metastatic nodules have also been described with ADT.

Leuprorelin

Chemical composition and different formulas

Natural LHRH was first isolated and identified in 1971 (CitationSchally et al 1971). Leuprorelin (leuprolide acetate) was first synthesized in 1974 by Takeda Chemical Industries, Japan (CitationFujino et al 1974).

Leuprorelin is a synthetic non-peptide analogue of naturally occurring porcine LHRH. It has a longer half-life than natural LHRH due to its enhanced binding affinity and increased resistance to peptidase degradation. At least two alterations in chemical structure serve to enhance the biologic activity of GnRH. These alterations include substitution of a D-amino acid for the glycine molecule at position 6 and deletion of the glycine molecule at position 10, and usually replacement by an N-ethylamide group (CitationMonahan et al 1973). Leuprorelin is 80 times more potent than natural LHRH (CitationChrisp and Sorkin 1991).

Leuprorelin is orally inactive and originally was generally given subcutaneously or intramuscularly; leuprorelin 1 mg was given by daily injection. However, a depot formulation was soon developed to enable convenient subcutaneous or intramuscular injection at 1-month intervals. The clinical benefit of treating prostate cancer with LHRH analogues was reported from 1982 (CitationTolis et al 1982).

Leuprorelin was initially administered subcutaneously to men with metastatic prostate cancer as a daily 1-mg injection, starting in 1985.

The monthly depot was first launched in Europe in France in April 1989, and now is well established as the leading LHRH analogue. Since the publication of earlier reviews (eg, CitationChrisp and Sorkin 1991), new formulations have also been developed to maximize flexibility and convenience of administration for both doctor and patient.

Microsphere technology enables leuprorelin to be given as a depot formulation. Leuprorelin microspheres range in mean diameter between 10 and 20 mm for the 1-month depot and between 10 and 30 mm for the 3-month depot. Clinically, this means that leuprorelin can be given as a liquid injection through a fine-gauge needle using conventional injection techniques.

Sustained-release parenteral depot formulations, in which the hydrophilic leuprorelin is entrapped in biodegradable highly lipophilic synthetic polymer microspheres, have been developed to avoid daily injections (CitationSharifiet al 1997; CitationPeriti et al 2002). In-situ forming drug delivery systems are prepared by dissolving a drug and a biodegradable polymer (poly (D, L-lactide-co-glycolide; PLGA) in a biocompatible organic solvent. In clinical studies of prostatic cancer, use of the depot formulation has effectively reduced the dose required to as low as one-eighth of that needed for administration by daily injection (CitationTogushi 1992).

Three- and 4-month formulations of leuprorelin were approved by the FDA for this indication in 2002 and 2003. Within a few years of its introduction, the 3-month depot accounted for 45% of the LHRH agonist market, and the 4-month formulation accounted for 40% of the market in its first year.

The development of depot forms of the LHRH agonists provided a well accepted approach to effective castration. Current formulations of 3- or 4-month (or longer) preparations are widely utilized; the most recent addition is the 6-month formula. Other benefits of the LHRH agonists include no cardiovascular toxicity and the fact that the “castration” is reversible.

Subcutaneous implants have been developed; the implant is designed to be removed after 12 months, and then another implant can be inserted for continued therapy.

Implants require a surgical incision and have greater injection site reactions; however, patients avoid multiple injections. The leuprorelin implant (Viadur®) effectively suppressed testosterone concentrations to less than the castrate threshold and maintained that suppression throughout the study period (CitationFowler et al 2000a). Some authors claimed testosterone suppression to the castrate range of 100% (CitationFowler et al 2000b; CitationMark 2003).

Histrelin acetate 50 mg subdermal implant (Vantas®) also seems to achieve similar results in men with advanced prostatic cancer. The hydrogel implant provided consistent delivery of histrelin over 1 year. Testosterone suppression was maintained throughout the 52 weeks after implantation in more than 99% of patients. No testosterone or LH surge was observed with re-implantation. PSA, a secondary end point for effectiveness, was also suppressed significantly from baseline (CitationSchlegel 2006).

Pharmacokinetics

After injection of the 1-month depot formulation of leuprorelin 3.75 mg, peak serum levels are achieved within 1 hour, followed by a rapid fall over the next 24 hours. A dose-dependent plateau is maintained over at least 5 weeks, representing a constant rate of release of leuprorelin from the copolymer (CitationMazzei et al 1990).

As with the 1-month injection, the 3-month injection (11.25 mg), results in an initial rise in serum levels of leuprorelin, followed by continuous linear release. A serum level of about 200–287 pg/mL is maintained over at least 3 months (CitationWechsel et al 1996) (equivalent to that achieved with a 1-month dose of 3.75 mg after repeated injections).

Mechanism of action

Leuprorelin acts as a potent inhibitor of gonadotropin secretion when given continuously in therapeutic doses. Animal and human studies indicate that after an initial stimulation as with other LHRH analogues (3–4 days), chronic administration of leuprorelin suppresses testicular and ovarian steroidogenesis. In humans, administration of leuprorelin initially increases circulating levels of LH and FSH, leading to a transient increase in levels of the gonadal steroids. However, continuous administration of leuprorelin decreases levels of LH and FSH, and subsequently reduces testosterone. These decreases occur within 2–4 weeks after initiation of treatment.

To obtain optimal therapeutic effect against androgen-dependent tumor cells, serum testosterone levels must be reduced to castrate levels (=50 ng/dL) (CitationFergusson 1957). This level can be achieved by a 1-month depot injection of 3.75 mg leuprorelin (CitationWhitaker et al 2002) or by at 3-month depot injection of 11.25 mg (CitationKhan and O’Brien 1998), or other new depot formulations.

GnRH agonist administration was shown by Redding and Schally to reduce the growth of prostate tumors in rats (CitationRedding and Schally 1983). Early clinical studies in metastatic or advanced prostate cancer established the efficacy of daily s.c. injection of leuprorelin (1–20 mg) in suppressing testosterone levels, delaying tumor progression, and alleviating symptoms of locally advanced and metastatic prostate cancer (CitationTrachtenberg 1983; CitationYamanak et al 1984). This effect is reversible upon discontinuation of drug therapy and this therapeutic approach has now replaced surgical castration.

Comparative studies on other androgen suppression modalities (): comparison of different depot formulas

The US Food and Drug Administration (FDA) has emphasized the importance of testosterone in prostate cancer therapy by utilizing decrease in testosterone associated with ADT as a surrogate endpoint for prostate cancer treatment (CitationBrawerr 2001). It is intriguing that the agency uses testosterone and not PSA level in this regard. Therapies directed at treating prostate cancer on a hormonal basis need only to demonstrate achievement of castrate levels of testosterone for approval by the FDA.

Table 3 Leuprorelin: comparative studies to other androgen suppression modalities

Another requirement of GnRH agonists and other endocrine therapies for prostate cancer is the ability to maintain castration levels of testosterone while on long-term therapy. Different formulas have been proven to be meet those criteria (). Selecting the best product may be based on: patient comfort, side effects or injection-site reactions, ease of use such as patient preparation, time, and staffing required for administration.

Table 4 Leuprorelin formulas and testosterone suppression

Six-month formulation of leuprorelin depot (Eligard® 45-mg)

Six-month leuprorelin acetate (Eligard® 45-mg; Atrix Laboratories) is a new formulation for the palliative treatment of advanced prostate cancer.

In recent study conducted by CitationCrawford et al (2006), the 6-month depot formulation of leuprorelin 45-mg (Eligard®) was evaluated clinically in a 12-month, open-label, multicenter study in patients with prostate cancer. The mean time required to reach castrate T levels (<50 ng/dL) was 21 days, 99% had 12-month testosterone (T) levels <50 ng/dL, and 88% patients had T levels <20 ng/dL. Mean PSA levels decreased from 39.8 ± 21.5 ng/mL at baseline to 1.2 ± 0.3 ng/mL at 12 months.

The 6-month formula of leuprorelin is the new addition which has a number of advantages over shorter-acting depot products, while reducing the need to schedule actual treatment time to only twice yearly.

Patients who may benefit from include those who:

  • Are stable and well controlled on palliative therapy

  • Travel for extended periods of time

  • Have difficulty getting to the physician’s office (eg, due to limited mobility or long travel distance)

  • Receive an LHRH agonist as neoadjuvant therapy.

Moreover, the change in Medicare reimbursement (CitationPainter 2005) for injections to “budget neutral” removes any incentive for favoring a formulation that is given more often over one that is given every 6 months. Of the urologists polled, 77% said that they were likely to use a 6-month depot rather than the shorter treatment regimens currently available. This novel delivery system allows for a low-volume injection, with very small changes in volume with increased dosage.

The Atrigel® Delivery System is composed of a biodegradable polymer (DL-lactide-coglycolide) dissolved in a biocompatible liquid solvent (N-methyl-2-pyrrolidone, or NMP). Because of its larger volume, leuprorelin depot is typically injected into the buttocks, necessitating the use of a treatment room. Patients receiving Lupron® Depot may experience pain at the injection site lasting up to 2 days, large abscesses, and bruising. However, there was no statistically significant difference in pain experienced on injection of goserelin and leuprorelin when patients were unaware of needle size (CitationMontgomery et al 2005).

Conclusion

After seven decades, ADT has stood the test of time and gained its place as the mainstay of treatment for advanced and metastatic prostate cancer. It has relieved the symptoms and prolonged the life of many patients. Its discovery and refinement have brought two Nobel prizes to the medical community, first with the seminal work of Charles Higgins on the influence of the endocrine system on the development of a human malignancy in 1966, and then in 1977 with Andrew Schally’s work on the hypothalamic hormones.

ADT went through several steps and refinement processes to reach its current status. In the 1940s the only option available was the irreversible castration through orchiectomy or the administration of female hormones, today the options are myriad.

The availability of long-acting synthetic LHRH agonists in the 1980s revolutionized the hormonal treatment of prostate cancer, enabling many men to avoid the emotional and psychological effects of surgical castration. The depot formulas have gained wide acceptance from both patients and physicians.

The 6-month formula of leuprorelin represents a new addition to the armentarium that has a number of advantages over shorter-acting depot products. It suppresses the testosterone to the castrate level in the majority of patients completing treatment without any breakthrough responses.

Compared with a 3-month depot, the 6-month depot is associated with two rather than four opportunities for patients to lose therapeutic effect if they delay their injection beyond the formulation’s normal duration, thus improving the patient’s compliance to therapy and quality of life.

The limited number of injections reduces the anxiety due to physician’s office visit and fear of pain; injection takes minimal time which is the most desirable option from the nursing standpoint. The drug is well tolerated while maintaining a side effect profile comparable to other products in its class.

These advantages coupled with the recent change to Medicare legislation will make long-duration formulation an attractive option for physicians, and will certainly tip the balance in favor of this option in treating prostate cancer patients fulfilling the criteria for receiving such products.

References

  • AkazaHTrends in primary androgen depletion therapy for patients with localized and locally advanced prostate cancer: Japanese perspectiveCancer Sci200697243716630114
  • AkazaHEfficacy of primary hormone therapy for localized or locally advanced prostate cancer:results of a 10-year follow-upBJU Int200698573916925756
  • AndroutsosGJohn Hunter (1728–1793):fondateur de la chirurgie scientifique et précurseur de l’urologieProg Urol199881087969894276
  • BarqawiAAkdumanBAbouelfadelZThe use of flutamide as a single antiandrogen treatment for hormone-refractory prostate cancerBJU Int200392695814616448
  • BarqawiABMoulJWZiadaACombination of low-dose flutamide and finasteride for PSA-only recurrent prostate cancer after primary therapyUrology200362872614624911
  • BasariaSMullerDCCarducciMAHyperglycemia and insulin resistance in men with prostate carcinoma who receive androgen-deprivation therapyCancer2006106581816388523
  • BayoumiAMBrownADGarberAMCost-effectiveness of androgen suppression therapies in advanced prostate cancerJ Natl Cancer Inst2000921731911058616
  • BeerTMExperimental use of GnRH antagonists as second-line hormonal therapyRev Urol20046S33816985935
  • BergesRBelloUEffect of a new leuprorelin formulation on testosterone levels in patients with advanced prostate cancerCurr Med Res Opin2006226495516684425
  • Boccon-GibodLFournierGBottetPFlutamide versus orchidectomy in the treatment of metastatic prostate carcinomaEur Urol19973239159412794
  • Braga-BasariaMDobsASMullerDCThe metabolic syndrome in men with prostate cancer undergoing long term androgen deprivation therapyJ Clin Oncol20062439798316921050
  • BrawerMKThe evolution of hormonal therapy for prostatic carcinomaRev Urol20013S11116986002
  • BurchardtMBurchardtTChenMWVascular endothelial growth factor-A expression in the rat ventral prostate gland and the early effects of castrationProstate2000431849410797493
  • ButtyanRGhafarMAShabsighAThe effects of androgen deprivation on the prostate gland:cell death mediated by vascular regressionCurr Opin Urology20001041520
  • ByarDPCorleDKHormone therapy for prostate cancer: results of the Veterans Administration Cooperative Urological Research Group studiesNCI Monogr19881651703050535
  • CassilethBRPatients’ choice of treatment in stage D prostate cancerUrology19893357592523612
  • CavarrettaITRCuligZKlockerHNovel experimental therapeutic approaches for prostate cancerEUA Update Series2005322739
  • ChodakGWA critical review of maximal androgen blockade for advanced prostate cancerRev Urol200461823
  • ChodakGWMaximum androgen blockade: a clinical updateRev Urol200571317
  • ChrispPSorkinEMLeuprorelin:a review of its pharmacology and therapeutic use in prostatic disordersDrugs Aging199114875091794035
  • ChuFMJaysonMDineenMKA clinical study of 22.5 mg. La-2550: A new subcutaneous depot delivery system for leuprolide acetate for the treatment of prostate cancerJ Urol2002168119920312187267
  • ConnPMRogerSCSeaySGBiphasic regulation of gonadotropin-releasing hormone receptor by receptor micro-aggregation and intracellular Ca2 levelsMed Pharmacol1984255155
  • CoppageWSJrCoonerAETestosterone in human plasmaN Engl J Med196527390275832873
  • CrawfordEDEisenbergerMAMcLeonDGA controlled trial of leuprolide with and without flutamide in prostatic carcinomaN Engl J Med1989321419242503724
  • CrawfordEDSartorOChuFA 12-month clinical study of LA-2585 (45.0 mg):a new 6-month subcutaneous delivery system for leuprolide acetate for the treatment of prostate cancerJ Urol2006175533616406989
  • DebruyneFMJGonadotropin-releasing hormone antagonist in the management of prostate cancerRev Urol20046S253216985933
  • DenisLJKeuppensFSmithPHMaximal androgen blockade: final analysis of EORTC phase III trial 30853Eur Urol199833144519519355
  • DenmeadeSRIsaacsJTDevelopment of prostate cancer treatment: the good newsProstate2004582112414743459
  • DenmeadeSRLinXSIsaacsJTRole of programmed (apoptotic) cell death during the progression and therapy for prostate cancerProstate199628251658602401
  • FergussonJDImplantation of radioactive material into the pituitary for the control of prostatic cancerBr J Urol1957292152213471943
  • FowlerJEFlanaganMGleasonDMEvaluation of an implant that delivers leuprolide for 1 year for the palliative treatment of prostate cancerUrology2000a556394210792069
  • FowlerJEGottelsmanJEReidCFSafety and efficacy of an implantable leuprolide delivery system in patients with advanced prostate cancerJ Urol2000b164730410953135
  • Franck-LissbrantIHaggstromSDamberJETestosterone stimulates angiogenesis and vascular re-growth in the ventral prostate in castrated adult ratsEndocrinology199813945169449610
  • FreedlandSJPartinAWEarly hormonal therapy for prostate cancer: the good, the bad and the uglyRev Urol20057187816985828
  • FujinoMFukudaTShinagawaSSynthetic analogs of luteinizing hormone releasing hormone (LH-RH) substituted in position 6 and 10Biochem Biophys Res Commun197460406134608231
  • GleaveMEGoldenbergSLChinJLRandomized comparative study of 3 versus 8-month neoadjuvant hormonal therapy before radical prostatectomy: biochemical and pathological effectsJ Urol2001166500611458055
  • GreenspanSICoatesPSereikaSMBone loss after initiation of androgen deprivation therapy in patients with prostate cancerJ Clin Endoc Metab20059064107
  • HallEJRadiobiology for radiobiologist20005PhiladelphiaLippincot Wilkins
  • HomeEPractical observations on the treatment of the diseases of the prostate gland1811LondonNicol
  • HugginsCClarkPJQuantitative studies of prostatic secretion. II. The effect of castration and of estrogenic injections on the normal and on the hyperplasic prostate glands of dogsJ Exp Med19407274761
  • HugginsCHodgesCUStudies on prostate cancer:the effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostateCancer Res19411293
  • HugginsCScottWWBilateral adrenalectomy in prostatic cancer:clinical features and urinary excretion of 17-ketosteroids and estrogenAnn Surg194512210314117858696
  • HugginsCStevensREJrHodgesCVStudies in prostatic cancerArch Surg19414320923
  • IsaacsJTAntagonistic effect of androgen on prostatic cell deathProstate19845545576483690
  • IsaacsJTRole of androgens in prostate cancerVitam Horm1994494335027810075
  • IversenPOrchidectomy and oestrogen therapy revisitedEur Urol1998347119854189
  • IversenPMelezinekISchmidtANonsteroidal antiandrogens:a therapeutic option for patients with advanced prostate cancer who wish to retain sexual interest and functionBJU Int200187475611121992
  • JacobiGHIntramuscular cyproterone acetate for advanced prostatic carcinoma:results of the first multicentre trialIn Androgens and anti-androgens1983NetherlandsSchering16170
  • JanoffDMPetersonCMongoue-TchokoteSValue of androgen ablation in conjunction with radiation for locally advanced Pca: clinical outcomes of androgen deprivation as the sole therapy for localized and locally advanced prostate cancerBJU Int200596503716104900
  • KeyesELFergusonRSKeyesELFergusonRSTreatment of malignant tumors of the prostateUrology19366New YorkAppleton-Century4256
  • KhanMSO’BrienAAn evaluation of pharmacokinetics and pharmacodynamics of leuprorelin acetate 3M-depot in patients with advanced and metastatic carcinoma of the prostateUrol Int19986033409519419
  • KirkDImmediate vs. delayed hormone treatment for prostate cancer: how safe is androgen deprivation?Brit J Uro Int200086220 Abstract MP6.1.07
  • KirkDComment on: Can androgen deprivation be considered a primary treatment for prostate cancer?Nat Clin Pract Urol20063127
  • KoupparisARamsdenAPersadRCognitive effects of hormonal treatment for prostate cancerBJU Int2004939151615142135
  • KumarRJBarqawiAlCrawfordEDAdverse events associated with hormonal therapy for prostate cancerRev Urol200573743
  • LabrieFCandasBGomezJLCan combined androgen blockade provide long-term control or possible cure of localized prostate cancer?Urology200260115912100935
  • LeibovitzRLTuckerSJTreatment of localized prostate cancer with intermittent triple androgen blockade:preliminary results in 110 consecutive patientsOncologist20016177811306729
  • LoweFCBambergerMHIndications for use of ketoconazole in management of metastatic prostate cancerUrology1990365412247927
  • [LPG] The Leuprolide Study GroupLeuprolide versus diethylstilbestrol for metastatic prostate cancerN Engl J Med1984311128166436700
  • [LSG] The Leuprolide Study GroupLeuprolide versus diethylstilbesterol for metastatic prostate cancerN Engl J Med1984311128166436700
  • LyonsJAKupelianPAMohanDSImportance of high radiation doses (72 Gy or greater) in the treatment of stage T1-T3 adenocarcinoma of the prostateUrology200055859010654900
  • MaloneSPerryGEapenLMature results of the Ottawa Phase II study of intermittent androgen-suppression therapy in prostate cancer: clinical predictors of outcome Prostate-specific Antigen-only RecurrenceInt J Radiat Oncol Biol Phys20076869970617379438
  • MaloneSPerryGSegalRLong-term side-effects of intermittent androgen suppression therapy in prostate cancer:results of a phase II studyBJU Int2005965142016104902
  • MarianiAJGloverMAritaSMedical versus surgical androgen suppression therapy for prostate cancer:a 10-year longitudinal cost studyJ Urol2001165104711125375
  • MarkLSLuteinizing hormone-releasing hormone agonists in the treatment of men with prostate cancer:timing, alternatives, and the 1-year implantUrology2003623642
  • MassoudWPaparelPLopezJGDiscovery of a pituitary adenoma following a gonadotropin-releasing hormone agonist in a patient with prostate cancerInt J Urol200613303416643633
  • MazzeiTMiniERizzoMHuman pharmacokinetic and pharmacodynamic profiles of leuprorelin acetate depot in prostatic cancer patientsJ Int Med Res19901842562108885
  • McLeodDZinnerNTomeraKAberalix Study Group.A phase 3, multicenter, open-label, randomized study of abarelix versus leuprolide acetate in men with prostate cancerUrology2001587566111711355
  • McLeodDGTolerability of nonsteroidal antiandrogens in the treatment of advanced prostate cancerOncologist19972182710388026
  • McLeodDGHormonal therapy:historical perspective to future directionUrology2003613712667881
  • MercaderMSenguptaSBodnerBKEarly effects of pharmacological androgen deprivation in human prostate cancerBJU Int200799606717227493
  • MessingEMMonolaJSarodsyMImmediate hormonal therapy compared with observation following radical prostatectomy and pelvic lymphadenectomy in men with node-positive prostate cancerN Engl J Med19993411781810588962
  • MigliariRMuscasGMurruMAntiandrogens:a summary review of pharmacodynamic properties and tolerability in prostate cancer therapyArch Ital Urol Androl19997129330210673793
  • MillerKSteinerULingnauRandomized prospective study of intermittent versus continuous androgen suppression in advanced prostate cancerASCO20075015 Abstract
  • MonahanMWAmossMSAndersonHASynthetic analogs of the hypothalamic luteinizing hormone releasing factor with increased agonist or antagonist propertiesBiochemistry1973124616204589943
  • MontgomeryBSBorwellJPHigginsDMDoes needle size matters? Patient experience with luteinizing hormone releasing hormone analogueProstate Cancer Prostatic Dis2005866815700050
  • MoroteJEsquenaSAbascalJMFailure to maintain a suppressed level of serum testosterone during long-acting depot luteinizing hormone-releasing hormone agonist therapy in patients with advanced prostate cancerUrol Int200677135816888418
  • MoulJWWuHSunLEarly versus delayed hormonal therapy for prostate specific antigen only recurrence of prostate cancer after radical prostatectomyJ Urol20041711141714767288
  • MoulJWThe evolving definition of advanced prostate cancerRev Urol20046S101616985915
  • [MRC] The Medical Research Council Prostate Cancer Working Party Investigators GroupImmediate versus deferred treatment for advanced prostatic cancer:initial results of the Medical Research Council TrialBr J Urol199779235469052476
  • MurphyWTSchwippertHPituitary irradiation in prostatic cancerRadiology1951563768014834413
  • NewlingDWWAnti-androgens in the treatment of prostate cancerBr J Urol199677776848705208
  • OefeieinMGComumRFailure to achieve castrate levels of testosterone during luteinizing hormone releasing hormone agonist therapy:the case for monitoring serum testosterone and a treatment decision algorithmJ Urol2000164726910953134
  • OefeleinMGFengAScolieriMJReassessment of the definition of castrate levels of testosterone:implications for clinical decision makingUrology2000561021411113751
  • OrnsteinDKBeiserJAAndrioleGLAnaemia in men receiving combined finasteride and flutamide therapy for advanced prostate cancerBJU Int19998343610233450
  • PainterMRReimbursement issues with hormonal therapies for prostate cancerRev Urol20057S44716985884
  • ParmarHLightmanSLAllenLRandomised controlled study of orchidectomy vs. long-acting D-Trp-6-LHRH microcapsules in advanced prostatic carcinomaLancet19852120152866289
  • Pavone-MacalusoMFlutamide monotherapy versus combined androgen blockade in advanced prostate cancer. Interim report of an Italian multicentre, randomised studySIU 23rd Congress1994A354
  • PeehlDMBasic science of hormonal therapy for prostate cancerRev Urol200131522
  • Perez-MarenoRChuFMGleasonDA six-month, open-label study assessing a new formulation of leuprolide 7.5 mg for suppression of testosterone in patients with prostate cancerCli Ther200224190214
  • PeritiPMazzeiTMiniEClinical pharmacokinetics of depot leuprorelinClin Pharmacokinet20024148550412083977
  • PetrylakDPThe Current role of chemotherapy in metastatic hormone-refractory prostate cancerUrology2005653815885271
  • PetrylakDPNew paradigms for advanced prostate cancerRev Urol20079312
  • PilepichMVAndrogen suppression adjuvant to definitive radiotherapy in prostate carcinoma – long-term results of phase III RTOG 85-31Int J Radiat Oncol Biol Phys20056112859015817329
  • PontALong term experience with high dose ketoconazole therapy in patients with D2 prostatic carcinomaJ Urol19871379022437334
  • PoundRPartinAWEisenbergerMANatural history of progression after PSA elevation following radical prostatectomyJAMA19992811591710235151
  • ReddingTWSchallyAVInhibition of mammary tumor growth in rats and mice by administration of agonistic and antagonistic analogs of luteinizing hormone-releasing hormoneProc Natl Acad Sci USA1983801459626219395
  • RyanCJElkinEPSmallEJReduced incidence of bony metastasis at initial prostate cancer diagnosis: data from CaPSUREUrol Oncol20062439640216962488
  • SandfordNLSearleJWKerrJFSuccessive waves of apoptosis in the rat prostate after repeated withdrawal of testosterone stimulationPathology198416406106522105
  • SandowJvon RechenbergWEngelbartKPharmacological studies on androgen suppression in therapy of prostate carcinomaAm J Clin Oncol198811S6103291602
  • SatterthwaiteRWHillJHPackardEEExperimental and clinical evidence on the role of the 17 keto-steroids in prostatic carcinomaJ Urol194146114954
  • SchallyAVArimuraABabaYIsolation and properties of the FSH and LH-releasing hormoneBiochem Biophys Res Commun19714339394930860
  • SchlegelPNHistrelin Study Group. Efficacy and safety of histrelin subdermal implant in patients with advanced prostate cancerJ Urol20061751353816515997
  • ScholzMJennrichRStrumSLong-term outcome for men with androgen independent prostate cancer treated with ketoconazole and hydrocortisoneJ Urol2005173194795215879788
  • SchröderFHWalshPCRetikABVaughnEDJrHormonal therapy of prostate cancerCampbell’s urology20028PhiladelphiaWB Saunders3182208
  • SeeWAAdjuvant hormone therapy after radiation or surgery for localized or locally advanced prostate cancerCurr Treat Options Oncol200343516212941195
  • SeidenfeldJSamsonDJHasselbladVSingle-therapy androgen suppression in men with advanced prostate cancer:a systematic review and meta-analysisAnn Intern Med20001325667710744594
  • ShabisghATanji N’ D’AgatiVEarly effects of castration on the vascular system of the rat ventral prostate glandEndocrinology19991401920192610098532
  • ShahinianVBKuoYFFreemanJLRisk of fracture after androgen deprivation for prostate cancerN Engl J Med20053521546415647578
  • SharifiRKnollLDSmithJLeuprolide acetate (30-mg depot every four months) in the treatment of advanced prostate cancerUrology19985127169495710
  • SharifiRRatanwongCJungATherapeutic effects of leuprorelin microspheres in prostate cancerAdv Drug Deliv Rev1997281213610837568
  • SilverbergGDBrittRHTrans-sphenoidal hypophysectomy in the treatment of metastatic breast and prostate carcinomaWest J Med19791301915425500
  • SmallEJRyanCJThe case for secondary hormonal therapies in the chemotherapy ageJ Urol2006176S667117084172
  • SmithMRObesity and sex steroids during gonadotropin-releasing hormone agonist treatment for prostate cancerClin Cancer Res200713241517200361
  • SoganiPCRayBWhitmoreWFAdvanced prostatic carcinomaUrology1975616461145932
  • StuderUWhelanPAlbrechtWImmediate or deferred androgen deprivation for patients with prostate cancer not suitable for local treatment with curative intent: European Organization for Research and Treatment of Cancer Trial 30 891J Clin Oncol20062418687616622261
  • TabataK8-month neoadjuvant hormonal therapy before radical prostatectomy for high-risk prostate cancerNippon Hinyokika Gakkai Zasshi200697712816898594
  • TanejaSSA multidisciplinary approach to the management of hormone-refractory prostate cancerRev Urol200358591
  • ThompsonIMFlare associated with LHRH-agonist therapyRev Urol20013101416985686
  • TogushiHFormulation study of leuprorelin acetate to improve clinical performanceClin Ther199214121301606592
  • TolisGAckmanDTellosSTumour growth inhibition in patients with prostaic carcinoma treated with luteinising hormone releasing hormone agonistsProc Nat Acad Sci USA1982791658626461861
  • TrachtenbergJGittlemanMSteidleCA phase 3, multicenter, open-label randomized study of abarelix versus leuprolide plus daily antiandrogen in men with prostate cancerJ Urol20021671670411912385
  • TrachtenbergJPointAKetoconazole: a novel and rapid treatment for advanced prostate cancerJ Urol19831301526306286
  • TrachtenbergJThe treatment of metastatic prostatic cancer with a potent luteinizing hormone releasing hormone analogueJ Urol19831291149526406688
  • UchidaTIllingROCathcartPJThe effect of neoadjuvant androgen suppression on prostate cancer-related outcomes after high-intensity focused ultrasound therapyBJU Int200698770216879448
  • UsamiMBicalutamide 80 mg combined with a luteinizing hormone-releasing hormone agonist (LHRH-A) versus LHRH-A monotherapy in advanced prostate cancer: findings from a phase III randomized, double-blind, multicenter trial in Japanese patientsProstate Cancer Prostatic Dis20071019420117199134
  • Van LoenenACHuirneJASchatsRGnRH agonists, antagonists, and assisted conceptionSemin Reprod Med20022034912536358
  • WardJFMoulJWBiochemical recurrence after definitive prostate cancer therapy. Part I:defining and localizing biochemical recurrence of prostate cancerCurr Opin Urol200515181615815195
  • WechselHWZerbibMPaganoFRandomized opabelledled comparative study of the efficacy, safety and tolerability of leuprorelin acetate 1M and 3M depot in patients with advanced prostatic cancerEur Urol199630714 discussion 19–218977984
  • WhitakerISFazelMZJoshiHBLeuprorelin acetate granulomas:recurrent subcutaneous nodules mimicking metastatic deposits at injections sitesBJU Int20029035012133078
  • WhiteJWThe results of double castration in hyper-trophy of the prostateAnn Surg18952213
  • WhiteJWThe present position of surgery of the hypertrophied prostateAnn Surg19044078892
  • YamanakaHMakinoTKumasakaFClinical efficacy of (DLeu6)-des Gly-NH2(10)-LHRH ethylamide against prostatic cancerHinyokika Kiyo19843054566435416
  • YoungHHIIKentJRPlasma testosterone levels in patients with prostatic carcinoma before and after treatmentJ Urol196899788924871357
  • ZietmanALThe case for neoadjuvant androgen suppression before radiation therapyMol Urol200042038 discussion 21511062375