You are on page 1of 7

ELIGARD: LEUPROLIDE ACETATE IN A NOVEL

SUSTAINED-RELEASE DELIVERY SYSTEM


OLIVER SARTOR

ABSTRACT
Androgen deprivation as a treatment for prostate cancer has evolved since the pioneering studies of Huggins
and Hodges 60 years ago using surgical castration or estrogen treatments. The most common hormonal
treatments today use injections of luteinizing hormone–releasing hormone (LHRH) agonists. The US Food
and Drug Administration (FDA) has approved 5 LHRH agonist formulations for treatment of prostate cancer
in the United States. Of these approved products, 3 involve different delivery systems for the LHRH
superagonist leuprolide acetate. Sustained-release formulations of 2 distinct LHRH agonists, goserelin
acetate and triptorelin pamoate, are also commercially available. This review focuses on new data on a novel
formulation of leuprolide acetate (Eligard; Atrix Laboratories Inc., Fort Collins, CO) that incorporates a
unique mixture of selected polymers and solvents to achieve sustained drug delivery after subcutaneous
injection. The FDA has approved 1-month and 3-month formulations of Eligard, and 4-month and 6-month
products are in development. In clinical trials, Eligard achieves reliable and sustained suppression of serum
testosterone to castration levels (ⱕ50 ng/dL). Of the patients treated with the 1-month and 3-month
formulations, 98% (115 of 117) and 94% (104 of 111), respectively, reached testosterone levels of ⱕ20
ng/dL. Breakthroughs, defined as testosterone levels ⬎50 ng/dL after achievement of castration levels,
occurred rarely at 0% (0 of 117) for the 1-month and at 0.9% (1 of 111) for the 3-month formulations in
patients receiving Eligard. The degree of disease control and the adverse-events profile are commensurate
with the effectiveness of the testosterone suppression; the incidence of severe hot flashes is actually lower
than anticipated. Additional studies with these novel formulations are warranted. UROLOGY 61 (Suppl 2A):
25–31, 2003. © 2003, Elsevier Science Inc.

R ecognition of the sensitivity of prostate cancer


to androgenic stimuli dates back ⬎6 decades
to the landmark studies published by Huggins and
thalamic-pituitary axis. In 1971, Andrew Schally et
al.2 isolated luteinizing hormone–releasing hor-
mone (LHRH) from porcine hypothalamic extracts
Hodges1 in 1941. These studies established andro- and demonstrated that this decapeptide was both
gen deprivation, accomplished via orchiectomy or necessary and sufficient for release of luteinizing
estrogen treatment, as the primary treatment for hormone (LH) from the pituitary. In 1977, Dr.
advanced prostate cancer. For his work on hor- Schally shared the Nobel Prize for discoveries on
monal treatment of prostate cancer, Dr. Huggins peptide production in the brain (reviewed in
was awarded the Nobel Prize for Physiology or Schally et al.2).
Medicine in 1966. Research into the regulatory These basic physiologic findings provided the in-
pathways controlling testosterone production sub- tellectual foundation for the next advance in hor-
sequently centered on the critical role of the hypo- monal therapy for prostate cancer. Shortly after the
isolation of LHRH, experimental substitution of
From the Stanley S. Scott Cancer Center, Louisiana State Univer-
sity School of Medicine, New Orleans, Louisiana, USA
amino acids in the basic LHRH structure resulted
This supplement was funded by Sanofi-Synthelabo. Dr. Sartor in analogs with potent agonist activity. Although
is a member of the scientific advisory board for Atrix Laborato- the initial studies of LHRH agonists were aimed at
ries, and is a paid consultant to Sanofi-Synthelabo. developing treatments for infertility, these com-
Reprint requests: Oliver Sartor, MD, Stanley S. Scott Cancer
Center, Suite 620, 2025 Gravier Street, Louisiana State Univer-
pounds, when administered in either a continuous
sity Health Sciences Center, New Orleans, Louisiana 70112. E- or daily manner, resulted in a paradoxical de-
mail: osarto@Isumc.edu crease, rather than increase, in both LH and testos-

© 2003, ELSEVIER SCIENCE INC. 0090-4295/03/$30.00


ALL RIGHTS RESERVED PII S0090-4295(02)02396-8 25
terone levels. The decrease was a consequence of CURRENTLY APPROVED LUTEINIZING
downregulated LHRH receptors at the pituitary HORMONE–RELEASING HORMONE
level.2 In 1980, LHRH agonists were first used to AGONISTS
treat patients with prostate cancer.3 The US Food The FDA has approved 5 LHRH agonist formu-
and Drug Administration (FDA) approval of LHRH lations for the treatment of prostate cancer: 3 de-
analogs for treatment of prostate cancer was livery systems that administer leuprolide acetate in
granted in 1985, after pivotal randomized studies a sustained manner, a goserelin acetate implant
demonstrated that the time to cancer progression (Zoladex; AstraZeneca Pharmaceuticals, Maccles-
was equivalent in patients treated with daily 3-mg field, London, UK), and a sustained-release form of
doses of diethylstilbestrol (DES) and daily 1-mg triptorelin pamoate (Trelstar; Pharmacia, Peapack,
subcutaneous LHRH injections.4 Toxicity varied NJ). Of the 3 sustained-release delivery systems for
between the 2 treatment groups, favoring the leuprolide acetate, 2 are injectable forms: Lupron
LHRH-treated subjects: fluid retention, nausea, gy- Depot (TAP Pharmaceuticals, Lake Forest, IL) and
necomastia, and breast tenderness were more com- Eligard (Atrix Laboratories, Inc., Fort Collins,
monly reported in the patients treated with DES. CO), and the third is a subcutaneously implanted
Hot flashes were more commonly reported in pa- minipump approved for 1-year dosing (Viadur;
tients treated with the LHRH analog. Given these Alza Corporation, Mountain View, CA). Each of
findings, and the strong preference patients have these agents has been shown to maintain mean tes-
expressed for injections as opposed to orchiec- tosterone levels ⬍50 ng/dL. Limited data are avail-
tomy, LHRH agonists have virtually replaced both able about which products meet the more stringent
estrogens and orchiectomy as the initial hormonal testosterone suppression guidelines established by
treatment of choice for patients with prostate can- the NCCN.
cer in the United States. The remainder of this article describes a compre-
hensive series of data from clinical trials of the
Eligard formulation of leuprolide acetate for sub-
cutaneous injection. The leuprolide acetate is ad-
TESTOSTERONE SUPPRESSION AND ministered via an implanted depot delivery system,
CANCER RESPONSIVENESS which releases the drug in a controlled manner
The studies of Huggins and Hodges1 clear- over defined intervals—1, 3, 4, or 6 months. The
ly established that androgen deprivation re- FDA has approved the 1-month and 3-month for-
mulations for the palliative treatment of advanced
sulted in improved outcomes for patients with
prostate cancer. The 4-month formulation has
advanced prostate cancer. Although the broad
completed clinical trials and is awaiting FDA ap-
relation between androgen suppression and re- proval; clinical trials of the 6-month formulation
sponsiveness of tumors in patients with prostate are currently under way.
cancer has now been well documented, the exact
relation between testosterone levels (after thera-
peutic intervention) and length of survival or ELIGARD DELIVERY SYSTEM
time to cancer progression has yet to be defined.
In their most recently issued guidelines, the The implant system used in the Eligard product
National Comprehensive Cancer Network (NCCN) consists of leuprolide acetate admixed with a
has recommended that orchiectomy or addition of combination of biodegradable polymers in a liquid
an oral antiandrogen be considered if a patient’s carrier. The polymeric delivery system (Atrix
Laboratories, Inc.) is composed of a biodegrad-
serum testosterone levels remain ⬎20 ng/dL dur-
able poly (DL-lactide-co-glycolide) polymer for-
ing monotherapy with an LHRH agonist.5 This
mulation dissolved in a biocompatible solvent,
threshold is not based on published clinical evi- N-methyl-2-pyrrolidone.8 Leuprolide acetate is
dence. The FDA has established serum testoster- physically mixed with the polymer/solvent combi-
one levels of ⱕ50 ng/dL as being consistent with nation immediately before injection. The drug/
levels obtained after surgical castration. This level polymer combination is then injected via a 20-
of testosterone has been used as the standard for gauge needle into the subcutaneous tissue, where
approval of new hormonal therapies in the United it forms a solid drug-delivery implant, slowly re-
States since the advent of LHRH agonists. Current leasing the leuprolide acetate at a controlled
studies demonstrate that only about 5% of patients rate as the polymer is degraded by normal biologic
being treated with LHRH agonist therapy fail to processes. Variations in the molecular weight of
achieve testosterone suppression ⬍50 ng/dL.6 The the polymer and solvent concentrations allow
consequences of this relative lack of suppression sustained release of the drug for the specified du-
are unknown.7 ration.

26 UROLOGY 61 (Supplement 2A), February 2003


FIGURE 1. Testosterone suppression in dogs (N ⫽ 6) with Eligard (Atrix Laboratories, Inc., Fort Collins, CO) and
Lupron Depot (TAP Pharmaceuticals, Lake Forest, IL) 3-month products. T ⫽ testosterone. (Data on file, Atrix
Laboratories, Inc.9).

FIGURE 2. Testosterone levels in dogs with Eligard (45 mg) 6-month depot. LA ⫽ leuprolide acetate; NMP ⫽
N-methyl-2-pyrrolidone; PLG ⫽ poly (DL-lactide-co-glycolide). (Data on file, Atrix Laboratories, Inc.9)

ELIGARD PRECLINICAL STUDIES (Figure 2). These data clearly demonstrate utility
of the leuprolide acetate delivery system to reduce
Preclinical studies conducted with various for-
testosterone levels in a preclinical model.
mulations of Eligard in dogs established proof of
principle. With the 3-month formulation, testos-
ELIGARD CLINICAL TRIALS
terone levels increased within the first week after
administration but then decreased to well below THE 1-MONTH FORMULATION
canine castration levels within 14 days after admin- The 1-month depot formulation of Eligard (7.5
istration (Figure 1).9 The 6-month formulations of mg leuprolide acetate) has been evaluated clini-
Eligard have been shown to effectively suppress cally in an open-label, multicenter study of 120
testosterone levels in dogs over the 6-month period patients with prostate cancer.10 Of these patients,

UROLOGY 61 (Supplement 2A), February 2003 27


FIGURE 3. Testosterone levels in humans after subcutaneous injection of Eligard (Atrix Laboratories, Inc., Fort
Collins, CO) 7.5 mg 1-month depot. (Adapted with permission from Clin Ther.10)

TABLE I. Testosterone values with Eligard (7.5 mg) 1-month


depot11
Testosterone Value
Mean value
Baseline (N ⫽ 120) 361.2 ng/dL
Month 6 (N ⫽ 117) 6.1 ng/dL (3.0*–27.0)
Median time to suppression (range)
ⱕ50 ng/dL 21 d (10–42 d)
ⱕ20 ng/dL 28 d (14–49 d)
Proportion with suppression at month 6
ⱕ50 ng/dL 117 patients (100%)
ⱕ20 ng/dL 115 patients (98%)
* Lower limits of detection.

89 had Jewett stage C disease and 31 had stage D ng/dL on day 28. Median time to suppression to
disease. (TNM staging was not done.) A total of ⱕ50 ng/dL was 21 days; median time to suppres-
117 patients completed 6 months of treatment, sion to ⱕ20 ng/dL was 28 days. At the conclusion
consisting of 6 subcutaneous injections (1 subcu- of the study (month 6), the mean testosterone con-
taneous injection every 28 days in the upper abdo- centration was 6.1 ng/dL (Table I). By day 28
men). The 3 patients who withdrew cited nonmed- (week 4), 112 of 119 patients (94.1%) had
ical reasons for not completing the study. Over a achieved castration levels of testosterone, defined
6-month period, the following were evaluated: (1) as serum testosterone levels ⱕ50 ng/dL. By day 42,
safety and efficacy, measured in terms of testoster- 100% of the patients had achieved testosterone lev-
one levels; (2) levels of prostate-specific antigen els ⱕ50 ng/dL. Once serum testosterone levels
(PSA); and clinical adverse events. Secondary effi- were ⱕ50 ng/dL, no patient demonstrated a break-
cacy endpoints included performance status, bone through response (concentration ⬎50 ng/dL) at
pain, urinary symptoms, and urinary pain. any time in the study. All 117 patients providing
As would be expected at the initiation of ther- evaluable data had testosterone concentrations
apy with an LHRH agonist, the mean testosterone ⱕ50 ng/dL at month 6. In addition, at 6 months,
concentration increased from 361.2 ng/dL at 98% (115 of 117) of the patients treated with Eli-
baseline to 574.6 ng/dL at day 3 after the initial gard had testosterone levels at or below the more
subcutaneous injection (Figure 3).11 Mean test- stringent NCCN threshold of ⱕ20 ng/dL. The
osterone concentration then decreased to below baseline mean serum PSA level for the 117
baseline by day 10, reaching a mean level of 21.8 patients enrolled in the study was 31.0 ng/mL

28 UROLOGY 61 (Supplement 2A), February 2003


FIGURE 4. Testosterone levels in humans after subcutaneous injection of Eligard (Atrix Laboratories, Inc., Fort
Collins, CO) 22.5 mg 3-month depot. (Adapted with permission from J Urol.11)

TABLE II. Testosterone values with Eligard (22.5 mg) 3-month


depot11
Testosterone Value
Mean value ⫾ SD (range)
Baseline (N ⫽ 117) 367 ng/dL ⫾ 13.2 ng/dL (140–839 ng/dL)
Month 6 (N ⫽ 111) 10.1 ⫾ 0.7 ng/dL (3.0*–50.0 ng/dL)
Median time to suppression (range)
ⱕ50 ng/dL 21 d (14–35 d)
ⱕ20 ng/dL 28 d (14–84 d)
Proportion with suppression at month 6
ⱕ50 ng/dL 115 patients (98%)
ⱕ20 ng/dL 104 patients (94%)
* Lower limit of detection.

(range, 0.1 to 639.0 ng/mL). At month 6, the mean duration, or recurrence of adverse events with sub-
PSA value was 1.9 ng/nL (range, 0.1 to 93.9 ng/ sequent injections. No treatment-related serious
mL), representing a 94% mean reduction from adverse events occurred, and no deaths were re-
baseline. As expected with an effective hormonal ported during the study. No patients discontinued
therapy, serum PSA levels decreased in all patients the study because of a treatment-related adverse
whose baseline values were elevated above the event.
normal limit.
A total of 716 injections of Eligard 7.5 mg were THE 3-MONTH FORMULATION
administered in the 120 patients enrolled in the A multicenter, open-label study investigated a
study. Overall, adverse events were consistent with 3-month subcutaneous depot formulation of leu-
androgen deprivation. The most common adverse prolide acetate in patients with prostate cancer
events reported by patients were hot flashes. Side (Jewett stage A, B, C, and D). Of the 117 patients
effects related to sexual function were not volun- enrolled, 111 patients completed 6 months of
tarily reported by the patients in the trial. Hot treatment consisting of 2 subcutaneous injections
flashes were reported in 68 of the 120 patients of 22.5 mg, with 1 injection administered to the
(56.7%). Of the reported hot flashes, 83% were upper abdomen every 3 months. Safety and efficacy,
mild, 16% moderate, and 1% severe. Injection site as assessed by testosterone and PSA levels, were
adverse events were reported either as mild in 60 evaluated. After an initial increase in testosterone
patients (50%) or moderate in 14 patients (11.7%). levels, mean testosterone concentrations de-
There were no trends toward increased severity, creased to ⱕ50 ng/dL by day 21 and to ⱕ20 ng/dL

UROLOGY 61 (Supplement 2A), February 2003 29


TABLE III. Adverse events with Eligard (22.5 mg) 3-month depot
(6-month study, N ⴝ 117)11
Incidence of Possibly or
Probably
Treatment-Related Systemic
Adverse Events
Reported by >2% of Patients
Adverse Event Patients (N) Percent
Hot flashes/flushes 66 56.4
Nausea 4 3.4
Fatigue 7 6.0
Arthralgia 4 3.4
Urinary frequency 3 2.6
Pruritus (not otherwise specified) 3 2.6

TABLE IV. Summary of Eligard products10,11


Final Mean Testosterone Volume of
Patients Testosterone Breakthrough Injection
Product (Completed) (ng/mL) (Patients) [mL]
1-Month Depot 117 6.1 0/117 0.25
3-Month Depot 111 10.1 1/111 0.375

by day 28 (Figure 4; Table II). The mean testoster- and 3-month formulations, respectively (Table
one values remained ⬍20 ng/dL for the rest of the IV).10,11 Each formulation has been shown to be
evaluation period (168 days). There was 1 patient highly effective in reducing mean testosterone lev-
who had a transient increase in testosterone to 112 els to below castration levels established by the
ng/dL on day 49 after achieving castration levels, FDA (ⱕ50 ng/dL) as well as below the values rec-
but he regained suppression (27.0 ng/dL) on day ommended by the NCCN for LHRH agonist mono-
98, 14 days after the second injection. No studies therapy (ⱕ20 ng/dL). Breakthrough increases in
were performed to establish a mechanism for the testosterone levels (concentrations ⱖ50 ng/dL)
breakthrough. Baseline PSA values decreased from were infrequent and transient. Among the patients
a mean baseline value of 86.4 ng/mL to a mean being treated with the 1-month formulation, none
value at 6 months of 1.7 ng/mL, representing a experienced breakthrough testosterone increases;
mean reduction of ⬎98%. At the end of the evalu- with the other formulations, transient break-
ation period, 103 of 111 patients (93%) had through occurred at 1 time point in 1 patient
achieved a PSA level ⱕ4 ng/mL.11 treated with the 3-month formulation and in 3 pa-
As with the 1-month formulation, hot flashes tients treated with the 4-month formulation. As
were the most frequent adverse events in patients expected, PSA levels normalized in most patients
treated with the Eligard 22.5-mg, 3-month depot treated with the Eligard products. Although time
formulation, occurring in 56.4% of patients (Table to normalization of PSA values is tumor dependent
III). The incidence of severe hot flashes/sweats was and varies from patient to patient, 25% to 33% of
low. Although hot flashes/sweats were not pro- patients treated in the 3 trials began the trial with
spectively evaluated during the trial, of the 84 hot baseline values of ⬍4 ng/mL, and by the end of
flashes/sweats reported by 66 of the 117 patients, each trial, 92% to 95% of patients achieved normal-
87% were considered mild, 13% moderate, and 0% ization. No data are available for time to progres-
severe. Other reported adverse events occurred in sion or time to death, because these trials were not
⬍5% of patients, with the exception of fatigue, designed to assess these endpoints. In all 3 clinical
which occurred in 6%. Injection site reactions trials, the frequency of severe hot flashes with Eli-
overall were mild and brief in duration. gard treatment was low. Prospective trials designed
to carefully monitor hot flashes are needed to es-
tablish the comparative incidence of hot flashes
DISCUSSION
with Eligard and other available medical therapies
There have been 2 clinical trials that have now used to induce androgen ablation. Additional stud-
been published for Eligard evaluating the 1-month ies are needed to clarify the relation between the

30 UROLOGY 61 (Supplement 2A), February 2003


extent of testosterone suppression, time to cancer 6. Oefelein MG, and Cornum R: Failure to achieve cas-
progression, and cancer-specific survival. trate levels of testosterone during luteinizing hormone releas-
ing hormone agonist therapy: the case for monitoring serum
testosterone and a treatment decision algorithm. J Urol 164:
REFERENCES 726 –729, 2000.
1. Huggins C, and Hodges CV: Studies on prostate cancer. 7. Oefelein MG, Feng A, Scolieri MJ, et al: Reassessment
I. The effect of castration, of estrogen and of androgen injec- of the definition of castrate levels of testosterone: impli-
tion on serum phosphatases in metastatic carcinoma of the cations for clinical decision making. Urology 56: 1021–1024,
prostate. Cancer Res 1: 293–297, 1941. 2000.
2. Schally AV, Comaru-Schally AM, Plonowski A, et al:
8. Yewey GL, Duysen EG, Cox SM, et al: Delivery of pro-
Peptide analogs in the therapy of prostate cancer. Prostate 45:
teins from a controlled release injectable implant. Pharm Bio-
158 –166, 2000.
technol 10: 93–117, 1997.
3. Tolis G, Ackman D, Stellos A, et al: Tumor growth
inhibition in patients with prostatic carcinoma treated with 9. Data on file, Atrix Laboratories, Fort Collins, Colorado,
luteinizing hormone–releasing hormone agonists. Proc Natl 2002.
Acad Sci U S A 79: 1658 –1662, 1982. 10. Perez R, Chu FM, Gleason D, et al: A clinical study of
4. Leuprolide Study Group: Leuprolide versus diethylstil- LA-2500 7.5 mg: a new subcutaneous depot delivery system
bestrol for metastatic prostate cancer. N Engl J Med 311: for leuprolide acetate and the treatment of advanced prostate
1281–1286, 1984. cancer. Clin Ther 24: 1902–1914, 2002.
5. National Comprehensive Cancer Network. Prostate 11. Chu FM, Jayson M, Dineen MK, et al: A clinical study of
cancer. Treatment guidelines for patients. Version II. Last 22.5 mg. La-2550: a new subcutaneous depot delivery system
update: 2001. Available at: http://www.nccn.org/patient_gls/ for leuprolide acetate for the treatment of prostate cancer.
_english/_prostate_cancer/index.htm. Accessed July 2, 2002. J Urol 168: 1199 –1203, 2002.

UROLOGY 61 (Supplement 2A), February 2003 31

You might also like