Skip to main content
Erschienen in: Advances in Therapy 2/2017

Open Access 27.12.2016 | Original Research

Efficacy of Testosterone Suppression with Sustained-Release Triptorelin in Advanced Prostate Cancer

verfasst von: Jürgen Breul, Eija Lundström, Daniela Purcea, Werner P. Venetz, Patrick Cabri, Pascale Dutailly, Evan R. Goldfischer

Erschienen in: Advances in Therapy | Ausgabe 2/2017

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Introduction

Androgen deprivation therapy (ADT) is a mainstay of treatment against advanced prostate cancer (PC). As a treatment goal, suppression of plasma testosterone levels to <50 ng/dl has been established over decades. Evidence is growing though that suppression to even lower levels may add further clinical benefit. Therefore, we undertook a pooled retrospective analysis on the efficacy of 1-, 3-, and 6-month sustained-release (SR) formulations of the gonadotropin-releasing hormone (GnRH) agonist triptorelin to suppress serum testosterone concentrations beyond current standards.

Methods

Data of 920 male patients with PC enrolled in 9 prospective studies using testosterone serum concentrations as primary endpoint were pooled. Patients aged 42–96 years had to be eligible for ADT and to be either naïve to hormonal treatment or have undergone appropriate washout prior to enrolment. Patients were treated with triptorelin SR formulations for 2–12 months. Primary endpoints of this analysis were serum testosterone concentrations under treatment and success rates overall and per formulation, based on a testosterone target threshold of 20 ng/dl.

Results

After 1, 3, 6, 9, and 12 months of treatment, 79%, 92%, 93%, 90%, and 91% of patients reached testosterone levels <20 ng/dl, respectively. For the 1-, 3-, and 6-month formulations success rates ranged from 80–92%, from 83–93%, and from 65–97% with median (interquartile range) serum testosterone values of 2.9 (2.9–6.5), 5.0 (2.9–8.7), and 8.7 (5.8–14.1) ng/dl at study end, respectively.

Conclusion

In the large majority of patients, triptorelin SR formulations suppressed serum testosterone concentrations to even <20 ng/dl. Testosterone should be routinely monitored in PC patients on ADT although further studies on the clinical benefit of very low testosterone levels and the target concentrations are still warranted.

Introduction

Based on recent estimates, prostate cancer (PC) ranks first among all new male cancers in both the USA and Europe (21% in 2016 and 22.8% in 2012, respectively) [1, 2]. Testosterone is known to foster PC cell growth; thus, the backbone therapy of advanced PC is androgen deprivation (ADT). Approximately 90% of tumours respond to initial ADT, which may result in a marked symptom reduction and prolonged survival, even if androgen-independent disease eventually develops [35].
ADT was initially achieved surgically via bilateral orchiectomy. Gonadotropin-releasing hormone (GnRH) analogues emerging in the 1980s offered a novel approach to medical castration through suppression of the hypothalamic-pituitary-gonadal axis. One of these was triptorelin, which was shown to be up to 100 times more potent than the native GnRH in vitro and in vivo [68]. Its clinical development started in 1982 [3] and it was registered as the first GnRH agonist SR formulation worldwide in France in 1986. Today, GnRH agonists are used worldwide for ADT [9] with survival rates similar to surgical castration [10]. They reduce serum testosterone to castrate levels via the decrease in pituitary gonadotropin secretion that follows down-regulation of the pituitary GnRH receptors [11]. The ensuing hypoandrogenic environment results in glandular and tumour shrinkage and in an increase of interglandular connective tissue [4, 12, 13].
A testosterone level of 50 ng/dl (1.7 nmol/l) has been established as the standard castration threshold, which has been applied for over 40 years [14]. This threshold is widely accepted by regulatory authorities for the approval of GnRH analogues in the treatment of advanced PC [14, 15] and is also deemed adequate by the National Comprehensive Cancer Network (NCCN) [16]. The European Association of Urology (EAU) indeed also acknowledges the 50 ng/dl cut-off, but suggests 20 ng/dl (0.7 nmol/l) to be more appropriate as “better results are repeatedly observed with lower levels compared to 50 ng/dl” [17].
In fact, there is growing evidence that the extent of testosterone suppression during ADT in hormone-sensitive PC patients predicts a lower risk of and shorter time to androgen-independent progression or castration-resistant PC (CRPC), although so far most studies have been retrospective and sample sizes were rather small [4, 11, 14, 1820]. Advances in the understanding of the principal mechanisms of PC progression and resistance to castration have led to new agents for CRPC such as the testosterone synthesis inhibitor abiraterone and the androgen receptor blocker enzalutamide. The improved survival in patients with metastatic CRPC when adding these agents to first-line ADT with GnRH analogues (or surgical castration) also indicates an important role of low testosterone levels in the treatment of advanced PC [21, 22].
Efficacy results of triptorelin 1-, 3-, and 6-month formulations in patients with advanced PC have been published based on the standard castration limit of serum testosterone of 50 ng/dl. Reports suggesting that serum testosterone levels below this standard threshold might lead to improved clinical outcomes [4, 11, 14, 1820] prompted us to re-examine testosterone data from nine phase II to IV studies, which is so far the largest analysis examining the efficacy of ADT at a cut-off level of 20 ng/dl [3, 9, 23].

Methods

This is a retrospective pooled analysis of nine prospective clinical studies on the efficacy of triptorelin 1-, 3-, and 6-month SR formulations for advanced PC with overall 920 evaluable patients. Four studies were randomised controlled phase II studies, two were randomised controlled phase III studies, and three were non-controlled phase II to IV studies (Table 1). Primary endpoints always included testosterone assessments measured by either validated radioimmunoassay (RIA) or liquid chromatography tandem mass spectrometry (LC-MS/MS) [3, 9, 24, 25], which proved to show comparable results at both high and low testosterone concentrations. At the very low levels expected in men under ADT, RIA and LC–MS/MS are considered the most accurate and reliable methods.
Table 1
Overview of the nine studies combined in this analysis (ITT populations = 920)
Study ID, location; number of patients
Study design
Treatment (triptorelin formulation); duration
Primary endpoints
Assay (sensitivity)
DEB-96-TRI-02
South Africa; n = 30
Phase II, open, 1-arm
Pamoate 3.75 mg (1-month) IM on days 0 and 28; 2 months
Response (inhibition of FSH and LH secretion)
Triptorelin levels on days 0 and 28
Reduction of testosterone levels from day 0 to 56
125I-RIA Kit (LOD 6 ng/dl)b
DEB-99-TRI-04
South Africa; n = 69
Phase II, multicentre, single-blind, randomised, controlled, parallel group
Pamoate 3.75 mg (1-month) IM
Acetate 3.75 mg (1-month) IM, both on days 1 and 29; 2 months
Testosterone pharmacodynamics
125I-RIA Kit (LOD 6 ng/dl)b
DEB-TRI6M-201
Bulgaria; n = 24 (8a)
Phase II, single-blind, randomised, controlled, 3-arm
Pamoate 22.5 mg (6-month) IM; 6 months
Achievement of castrate testosterone levels on day 29
Maintenance of castrate levels from days 57 to 169
Solid-phase RIA (direct/extracted) (LOD 1 ng/dl)
E47-52014-402L
UK; n = 70
Phase II, multicentre, open, randomised
Pamoate 11.25 mg (3-month) IM
Acetate 3 mg (1-month) IM on days 0, 28, 56; 3 months
Testosterone level at month 3 (day 84)
Direct RIA (LOD 5 ng/dl)
E28-52014-701
South Africa, France, UK; n = 131
Part I: phase II, multicentre, open, randomised, 2-arm
Part II: open extension, 1-arm
Acetate 11.25 mg (3-month) IM
Acetate 3 mg (1-month) IM every 28 days; 3 months
Acetate 11.25 mg (3-month) IM on days 91–273; 6 months
% Patients with testosterone levels ≤50 ng/dl at day 84
RIA (LLOQ 5 ng/dl)
DEB-96-TRI-01 1st
South Africa; n = 335
Phase III, multicentre, randomised, controlled, 2-arm
Pamoate 3.75 mg (1-month) IM every 28 days
Pamoate 11.25 mg (3-month) IM every 84 days; 9 months
Achievement of testosterone castration levels at month 1
Maintenance of castration levels from month 2 to 9
125I-RIA Kit (LOD 6 ng/dl)b
DEB-96-TRI-01 2nd
South Africa; n = 277 (137a)
Phase III, multicentre, randomised, controlled, 2-arm
Pamoate 3.75 mg (1-month) IM every 28 days; 9 months
Achievement of testosterone castration levels at day 29
Maintenance of castration levels from month 2 to 9
125I-RIA Kit (LOD 6 ng/dl)b
DEB-TRI6M-301
South Africa; n = 120
Phase III, multicentre, open-label, 1-arm
Pamoate 22.5 mg (6-month) IM on days 1 and 169; 12 months
Achievement of testosterone castration levels at day 29
Maintenance of castration levels from month 2 to 12
LCMS/MS (LLOQ 3 ng/dl)
DEB-01-TRI-04
South Africa; n = 20
Phase IV, open-label, 1-arm
Pamoate 11.25 mg (3-month) IM on days 1, 85, and 169; 9 months
% Castrated patients on days 87 and 171, two days after re-exposures to triptorelin pamoate
DiaSorin RIA (LOD 2.9 ng/dl)
FSH follicle-stimulating hormone, GnRH gonadotropin-releasing hormone, IM intramuscular, LH: luteinising hormone, LLOQ lower limit of quantification, LOD limit of detection
aOnly patients of the triptorelin arm were included in this analysis
bFrom kit instruction manual
Patients were recruited in Europe and South Africa and had to have an indication for ADT, i.e., advanced or metastatic PC or rising prostate-specific antigen (PSA) after failed local therapy, were naïve to hormonal treatment or had undergone a 6-month washout period prior to study treatment, and presented with a normal baseline testosterone level at study entry. None of the patients except those in study E28-52014-701 (flutamide for 10 days) received any ADT. Concomitant treatments affecting the metabolism or secretion of testosterone were prohibited in all studies. Patients were treated with only one of the triptorelin formulations and investigated for 2–12 months (Fig. S1) except in study E28-52014-701 in which some patients were switched from the 1-month to the 3-month formulation (Table 1). All procedures performed in the original studies were in accordance with the ethical standards of institutional and national research committees and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Informed consent was obtained from all patients enrolled in the original studies; for the retrospective pooled analysis no formal consent is required.
Descriptive statistics were used to present demographic data, testosterone concentrations, and success rates. For the calculation of mean testosterone levels over various ranges of months, first the mean value for all available measurements over the indicated range of months was calculated for each patient without imputation for missing values, and then the mean and standard error (SE) for each group were calculated based on these means. For the comparison of baseline characteristics, Student’s t test and chi-square test were used. Data pooling and data management were done in SAS® (version 9.4) and MS Access 2013. Analyses were performed and figures created with R Core Team (2016).

Results

Mean age was similar among formulation groups with ages ranging from 42 to 96 years. Overall, half of the patients were Caucasian and one quarter black (Table 2). The 6-month formulation group had significantly higher testosterone levels as compared to the other two groups at baseline, i.e., means (95% CIs): 358.6 (345.8; 371.4) ng/dl in the 1-month formulation group and 383.1 (363.5; 402.7) ng/dl in the 3-month formulation as compared to 502.6 (467.4; 537.8) ng/dl in the 6-month formulation group (Table 2). The mean BMI was also higher in the 6-month formulation group as compared to the others.
Table 2
Demographic data and baseline characteristics, means (range) or n (%)
Triptorelin formulation
1 month (3.75 mg)
3 month (11.25 mg)
6 month (22.5 mg)
All
Patients enrolled
489
303
128
920
Age (years)
71.1 (42–96)
70.5 (48–93)
71.1 (51–93)
70.9 (42–96)
Weight (kg)
74.2 (40–129)
74.6 (38–132)
83.3 (47–136)
75.8 (38–136)
BMI (kg/m2)
24.8 (13–43)
25.2 (16–44)
27.6 (19–42)
25.4 (13–44)
Testosterone (ng/dl)
358.6 (3–1015)
383.1 (40–1296)
502.6 (54–1171)
386.7 (3–1296)
Racea, n (%)
421 (100)
240 (100)
128 (100)
789 (100)
 Caucasian
231 (54.9)
147 (61.2)
85 (66.4)
463 (58.7)
 Black
128 (30.4)
65 (27.1)
27 (21.1)
220 (27.9)
 Coloured
61 (14.5)
27 (11.3)
16 (12.5)
104 (13.2)
 Other
1 (0.2)
1 (0.4)
0 (0)
2 (0.2)
BMI body mass index
aData on ethnicity were not collected in study E28-52014-701
The pooled data of all studies showed that a high proportion of patients achieved a testosterone level <20 ng/dl at the time points pre-defined in the protocols (i.e., months 1, 3, 6, 9, and 12), regardless of the formulation. Overall success rates based on the castration limit of 20 ng/dl reached 79% (95% CI: 75.9–81.3%) at month 1, 92% (89.7–93.6%) at month 3, 93% (90.4–94.4%) at month 6; 90% (87.2–92.0%) at month 9, and 91% (84.6–95.8%) at month 12. The success rates based on the standard castration limit of 50 ng/dl ranged from 95–99% (Fig. 1).
Overall, mean testosterone levels were maintained <20 ng/dl over study periods for all formulations except for the 6-month formulation at months 1 and 12 (Fig. 2) because of single patients with exceptionally high levels (patient DEB-TRI6M-301-11-11613 with a value 422 ng/dl at month 1 and patient DEB-TRI6M-301-04-04602 with a value of 1213 ng/dl at month 12). Excluding those as outliers would result in mean values of 18.5 and 13.1 ng/dl, respectively.
Success rates per formulation in terms of the proportion of patients with testosterone <20 ng/dl ranged from 80–92% for the 1-month formulation, 83–93% for the 3-month formulation, and 65–97% for the 6-month formulation (Fig. 1b). The large majority of patients (89.7%) maintained stable low testosterone levels without two consecutive increases in serum testosterone to ≥20 ng/dl. Only very few patients (1.1%) experienced such testosterone escapes to ≥50 ng/dl. Pooling data from all formulations resulted in mean testosterone levels just above 10 ng/dl for months 1–6, 1–9, and 1–12. Testosterone still decreased between months 1 and 2 and mean values were <10 ng/dl for months 2–6, 2–9, and 2–12 (Fig. S2). At the end of the studies, median (IQR) serum testosterone values were 2.9 (2.9–6.5) ng/dl for the 1-month formulation, 5.0 (2.9–8.7) ng/dl for the 3-month formulation, and 8.7 (5.8–14.1) ng/dl for the 6-month formulation.

Discussion

Our analysis of pooled testosterone data was based on a total of almost 1000 patients and is, so far, the largest of its kind. The great majority of patients treated with triptorelin were shown to achieve and maintain testosterone levels <20 ng/dl. This is in line with two other retrospective studies reassessing testosterone levels achieved by either orchiectomy [14] or by use of polymer-delivered subcutaneous leuprolide acetate formulations [26]. Suppression of testosterone levels <20 ng/dl through ADT may thus be common and regardless of the formulation, although in our study, the testosterone decrease with the 6-month formulation appeared less rapid. However, this might have been due to a slightly different patient population as reflected by higher baseline BMI and testosterone levels. In fact, obese patients were reported to respond less to GnRH agonist treatment as compared to patients with normal BMI [27]. By contrast, the efficacy of the triptorelin 6-month formulation has recently been confirmed by a Danish study with patients under triptorelin showing significantly lower testosterone levels at months 3 and 6 than after subcapsular orchiectomy [28].
Still, some limitations of our study may need to be considered: Safety data of the nine studies were not pooled and re-analysed as the overall safety profile of triptorelin treatment in PC has recently been confirmed to be well-established and consistent among formulations as well [29]. A more relevant limitation might be inherent to the retrospective design. Although all included clinical trials were quite homogeneous in terms of design, quality, enrolled populations, and analytical methods for the measurement of testosterone, they were of rather short duration and did therefore not allow for an assessment of the time to androgen-independent progression (AIP) or death. Thus, the clinical benefit of achieving testosterone levels <20 ng/dl could actually not be addressed. However, evidence is growing that such a benefit indeed exists, even though most studies so far have been retrospective as well and rather small in size.
In 73 PC patients on ADT followed up for 51 months (range 1–20 years) the lowest serum testosterone cut-off that was able to discriminate regarding AIP-free survival was 32 ng/dl (88 vs. 137 months; P < 0.03). However, only breakthrough testosterone increases >50, but not >20 ng/dl had a significant impact on AIP-free survival [18]. In 225 Japanese PC patients treated with combined androgen blockade and followed up for 45.8 months, multivariate analysis revealed nadir testosterone <20 ng/dl to be the most significant prognostic factor of overall survival [20]. The risk of death was also shown to significantly correlate with 6-month serum testosterone levels in 129 patients with metastatic PC treated with goserelin and followed up for 47.5 months (range 22–72) [11]. In the largest and longest retrospective study so far with 626 PC patients having received ADT for a median of 8 years, nadir testosterone levels <20 ng/dl were shown to be associated with longer times to disease progression [30]. Conversely, patients with a median testosterone level >20 ng/dl had a significantly higher risk of developing CRPC. Maximum testosterone levels ≥50 ng/dl (23% of patients) were associated with a significantly higher rate of progression to CRPC as compared to patients with maximum testosterone <20 ng/dl (27%) and patients with nadir testosterone levels ≥50 ng/dl (1%) had a significantly higher risk of dying from disease as compared to patients with nadir testosterone 20–50 (21%) and <20 ng/dl (78%).
By contrast, there have been only two prospective studies on the additional benefit of suppressing testosterone to lower serum thresholds or minimum levels: In 32 patients followed up for about 2 years, time to CRPC was significantly longer in those with 9-month testosterone <32 ng/dl as compared to those with >32 ng/dl; however no additional predictive value was found for those <20 ng/dl [4]. In the second study, 153 patients with advanced PC were treated with GnRH agonists for 65 months. Testosterone levels <20 ng/dl after 6 months were found to be associated with a significantly lower risk of death and a trend towards a lower risk of disease progression (P = 0.12) as compared to those >20 ng/dl [19].
In conclusion, evidence is growing that sustainable testosterone suppression <20 ng/dl is beneficial in patients with PC. Reassuringly, this is widely achieved with all triptorelin SR formulations. Still, regular assessment of testosterone levels should be routine clinical practice for men on ADT and patients who under GnRH therapy do not succeed in achieving an appropriate testosterone suppression may qualify for an alternative method of ADT. Well-designed and sufficiently powered prospective studies using accurate and reliable testosterone assays are warranted to further evaluate the clinical benefit of a more rigorous testosterone suppression to minimum levels and to establish a new target threshold for testosterone levels.

Acknowledgements

Sponsorship for this study and article processing charges were funded by Debiopharm International, Switzerland, and Ipsen Pharma, France. All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship of this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval to the version to be published. All authors had full access to all of the data in this study and take complete responsibility for the integrity of the data and accuracy of the data analysis. Editorial assistance in the preparation of this manuscript was provided by Camilla Hansson of Debiopharm International and Uwe Totzke of Totzke & Dreher Scientific (TDS). Support for this assistance was funded by Debiopharm International.

Disclosures

E. Lundström is employed by Debiopharm International. D. Purcea is employed by Debiopharm International. W.P. Venetz is employed by Debiopharm International. P. Cabri is employed by Ipsen Pharma. P. Dutailly is employed by Ipsen Pharma. J. Breul and E. R. Goldfischer declare that they have no conflict of interest.

Compliance with Ethics Guidelines

This article is based on previously conducted studies, and does not involve any new studies of human or animal subjects performed by any of the authors.

Data Availability

The data sets generated during and/or analysed during the current study are not publicly available but may be made available by the corresponding author on reasonable request.

Open Access

This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License (http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​), which permits any noncommercial use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (https://​creativecommons.​org/​licenses/​by/​4.​0), which permits use, duplication, adaptation, distribution, and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013;49(6):1374–403.CrossRefPubMed Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013;49(6):1374–403.CrossRefPubMed
2.
3.
Zurück zum Zitat Lundstrom EA, Rencken RK, van Wyk JH, Coetzee LJ, Bahlmann JC, Reif S, et al. Triptorelin 6-month formulation in the management of patients with locally advanced and metastatic prostate cancer: an open-label, non-comparative, multicentre, phase III study. Clin Drug Investig. 2009;29(12):757–65.CrossRefPubMed Lundstrom EA, Rencken RK, van Wyk JH, Coetzee LJ, Bahlmann JC, Reif S, et al. Triptorelin 6-month formulation in the management of patients with locally advanced and metastatic prostate cancer: an open-label, non-comparative, multicentre, phase III study. Clin Drug Investig. 2009;29(12):757–65.CrossRefPubMed
4.
Zurück zum Zitat Dason S, Allard CB, Tong J, Shayegan B. Defining a new testosterone threshold for medical castration: results from a prospective cohort series. Can Urol Assoc J. 2013;7(5–6):E263–7.CrossRefPubMedPubMedCentral Dason S, Allard CB, Tong J, Shayegan B. Defining a new testosterone threshold for medical castration: results from a prospective cohort series. Can Urol Assoc J. 2013;7(5–6):E263–7.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Helgstrand JT, Berg KD, Lippert S, Brasso K, Roder MA. Systematic review: does endocrine therapy prolong survival in patients with prostate cancer? Scand J Urol. 2016;50(3):135–43 (Epub 2016/02/26).CrossRefPubMed Helgstrand JT, Berg KD, Lippert S, Brasso K, Roder MA. Systematic review: does endocrine therapy prolong survival in patients with prostate cancer? Scand J Urol. 2016;50(3):135–43 (Epub 2016/02/26).CrossRefPubMed
6.
Zurück zum Zitat Coy DH, Labrie F, Savary M, Coy EJ, Schally AV. LH-releasing activity of potent LH-RH analogs in vitro. Biochem Biophys Res Commun. 1975;67(2):576–82.CrossRefPubMed Coy DH, Labrie F, Savary M, Coy EJ, Schally AV. LH-releasing activity of potent LH-RH analogs in vitro. Biochem Biophys Res Commun. 1975;67(2):576–82.CrossRefPubMed
7.
Zurück zum Zitat Nestor JJ Jr, Ho TL, Simpson RA, Horner BL, Jones GH, McRae GI, et al. Synthesis and biological activity of some very hydrophobic superagonist analogues of luteinizing hormone-releasing hormone. J Med Chem. 1982;25(7):795–801 (Epub 1982/07/01).CrossRefPubMed Nestor JJ Jr, Ho TL, Simpson RA, Horner BL, Jones GH, McRae GI, et al. Synthesis and biological activity of some very hydrophobic superagonist analogues of luteinizing hormone-releasing hormone. J Med Chem. 1982;25(7):795–801 (Epub 1982/07/01).CrossRefPubMed
8.
Zurück zum Zitat Millar RP, Troskie BE, Flanagan CA. Comparative receptor binding affinity and inositol phosphate production potency of D-Leu6 and D-Trp6 GnRH agonists on COS-1 cells transfected with the human GnRH receptor. XIII International congress of comparative endocrinology; 16–21 November; Yokohoma, Japan: Monduzzi Editore S.p.A. 1997; p. 559–62. Millar RP, Troskie BE, Flanagan CA. Comparative receptor binding affinity and inositol phosphate production potency of D-Leu6 and D-Trp6 GnRH agonists on COS-1 cells transfected with the human GnRH receptor. XIII International congress of comparative endocrinology; 16–21 November; Yokohoma, Japan: Monduzzi Editore S.p.A. 1997; p. 559–62.
9.
Zurück zum Zitat Heyns CF, Simonin MP, Grosgurin P, Schall R, Porchet HC. South African Triptorelin Study G. Comparative efficacy of triptorelin pamoate and leuprolide acetate in men with advanced prostate cancer. BJU Int. 2003;92(3):226–31.CrossRefPubMed Heyns CF, Simonin MP, Grosgurin P, Schall R, Porchet HC. South African Triptorelin Study G. Comparative efficacy of triptorelin pamoate and leuprolide acetate in men with advanced prostate cancer. BJU Int. 2003;92(3):226–31.CrossRefPubMed
10.
Zurück zum Zitat Seidenfeld J, Samson DJ, Hasselblad V, Aronson N, Albertsen PC, Bennett CL, et al. Single-therapy androgen suppression in men with advanced prostate cancer: a systematic review and meta-analysis. Ann Intern Med. 2000;132(7):566–77.CrossRefPubMed Seidenfeld J, Samson DJ, Hasselblad V, Aronson N, Albertsen PC, Bennett CL, et al. Single-therapy androgen suppression in men with advanced prostate cancer: a systematic review and meta-analysis. Ann Intern Med. 2000;132(7):566–77.CrossRefPubMed
11.
Zurück zum Zitat Perachino M, Cavalli V, Bravi F. Testosterone levels in patients with metastatic prostate cancer treated with luteinizing hormone-releasing hormone therapy: prognostic significance? BJU Int. 2010;105(5):648–51.CrossRefPubMed Perachino M, Cavalli V, Bravi F. Testosterone levels in patients with metastatic prostate cancer treated with luteinizing hormone-releasing hormone therapy: prognostic significance? BJU Int. 2010;105(5):648–51.CrossRefPubMed
12.
Zurück zum Zitat Civantos F, Marcial MA, Banks ER, Ho CK, Speights VO, Drew PA, et al. Pathology of androgen deprivation therapy in prostate carcinoma. A comparative study of 173 patients. Cancer. 1995;75(7):1634–41.CrossRefPubMed Civantos F, Marcial MA, Banks ER, Ho CK, Speights VO, Drew PA, et al. Pathology of androgen deprivation therapy in prostate carcinoma. A comparative study of 173 patients. Cancer. 1995;75(7):1634–41.CrossRefPubMed
13.
Zurück zum Zitat Murphy WM, Soloway MS, Barrows GH. Pathologic changes associated with androgen deprivation therapy for prostate cancer. Cancer. 1991;68(4):821–8.CrossRefPubMed Murphy WM, Soloway MS, Barrows GH. Pathologic changes associated with androgen deprivation therapy for prostate cancer. Cancer. 1991;68(4):821–8.CrossRefPubMed
14.
Zurück zum Zitat Oefelein MG, Feng A, Scolieri MJ, Ricchiutti D, Resnick MI. Reassessment of the definition of castrate levels of testosterone: implications for clinical decision making. Urology. 2000;56(6):1021–4.CrossRefPubMed Oefelein MG, Feng A, Scolieri MJ, Ricchiutti D, Resnick MI. Reassessment of the definition of castrate levels of testosterone: implications for clinical decision making. Urology. 2000;56(6):1021–4.CrossRefPubMed
15.
Zurück zum Zitat Tombal B. The importance of testosterone control in prostate cancer. Eur Urol Suppl. 2007;6(15):834–9.CrossRef Tombal B. The importance of testosterone control in prostate cancer. Eur Urol Suppl. 2007;6(15):834–9.CrossRef
16.
Zurück zum Zitat NCCN. Guidelines for treatment of cancer by site: prostate cancer. National comprehensive cancer network (NCCN), 2016 MT/W/0001/pdWS/001. NCCN. Guidelines for treatment of cancer by site: prostate cancer. National comprehensive cancer network (NCCN), 2016 MT/W/0001/pdWS/001.
17.
Zurück zum Zitat EAU. European association of urology (EAU) Guidelines on prostate cancer. 2016 MT/W/0001/pdWS/001. EAU. European association of urology (EAU) Guidelines on prostate cancer. 2016 MT/W/0001/pdWS/001.
18.
Zurück zum Zitat Morote J, Orsola A, Planas J, Trilla E, Raventos CX, Cecchini L, et al. Redefining clinically significant castration levels in patients with prostate cancer receiving continuous androgen deprivation therapy. J Urol. 2007;178(4 Pt 1):1290–5.CrossRefPubMed Morote J, Orsola A, Planas J, Trilla E, Raventos CX, Cecchini L, et al. Redefining clinically significant castration levels in patients with prostate cancer receiving continuous androgen deprivation therapy. J Urol. 2007;178(4 Pt 1):1290–5.CrossRefPubMed
19.
Zurück zum Zitat Bertaglia V, Tucci M, Fiori C, Aroasio E, Poggio M, Buttigliero C, et al. Effects of serum testosterone levels after 6 months of androgen deprivation therapy on the outcome of patients with prostate cancer. Clin Genitourin Cancer. 2013;11(3):325–30.CrossRefPubMed Bertaglia V, Tucci M, Fiori C, Aroasio E, Poggio M, Buttigliero C, et al. Effects of serum testosterone levels after 6 months of androgen deprivation therapy on the outcome of patients with prostate cancer. Clin Genitourin Cancer. 2013;11(3):325–30.CrossRefPubMed
20.
Zurück zum Zitat Kamada S, Sakamoto S, Ando K, Muroi A, Fuse M, Kawamura K, et al. Nadir testosterone after long-term followup predicts prognosis in patients with prostate cancer treated with combined androgen blockade. J Urol. 2015;194(5):1264–70.CrossRefPubMed Kamada S, Sakamoto S, Ando K, Muroi A, Fuse M, Kawamura K, et al. Nadir testosterone after long-term followup predicts prognosis in patients with prostate cancer treated with combined androgen blockade. J Urol. 2015;194(5):1264–70.CrossRefPubMed
21.
Zurück zum Zitat Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–33.CrossRefPubMedPubMedCentral Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–33.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368(2):138–48.CrossRefPubMed Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013;368(2):138–48.CrossRefPubMed
23.
Zurück zum Zitat Teillac P, Heyns CF, Kaisary AV, Bouchot O, Blumberg J. Pharmacodynamic equivalence of a decapeptyl 3-month SR formulation with the 28-day SR formulation in patients with advanced prostate cancer. Horm Res. 2004;62(5):252–8.PubMed Teillac P, Heyns CF, Kaisary AV, Bouchot O, Blumberg J. Pharmacodynamic equivalence of a decapeptyl 3-month SR formulation with the 28-day SR formulation in patients with advanced prostate cancer. Horm Res. 2004;62(5):252–8.PubMed
24.
25.
Zurück zum Zitat Wang C, Catlin DH, Demers LM, Starcevic B, Swerdloff RS. Measurement of total serum testosterone in adult men: comparison of current laboratory methods versus liquid chromatography-tandem mass spectrometry. J Clin Endocrinol Metab. 2004;89(2):534–43 (Epub 2004/02/07).CrossRefPubMed Wang C, Catlin DH, Demers LM, Starcevic B, Swerdloff RS. Measurement of total serum testosterone in adult men: comparison of current laboratory methods versus liquid chromatography-tandem mass spectrometry. J Clin Endocrinol Metab. 2004;89(2):534–43 (Epub 2004/02/07).CrossRefPubMed
26.
Zurück zum Zitat Shore ND, Chu F, Moul J, Saltzstein D, Concepcion R, McLane JA, et al. Polymer-delivered subcutaneous leuprolide acetate formulations achieve and maintain castrate concentrations of testosterone in four open-label studies in patients with advanced prostate cancer. BJU Int. 2016. doi:10.1111/bju.13482 (Epub ahead of print). Shore ND, Chu F, Moul J, Saltzstein D, Concepcion R, McLane JA, et al. Polymer-delivered subcutaneous leuprolide acetate formulations achieve and maintain castrate concentrations of testosterone in four open-label studies in patients with advanced prostate cancer. BJU Int. 2016. doi:10.​1111/​bju.​13482 (Epub ahead of print).
27.
Zurück zum Zitat Smith MR. Obesity and sex steroids during gonadotropin-releasing hormone agonist treatment for prostate cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2007;13(1):241–5.CrossRef Smith MR. Obesity and sex steroids during gonadotropin-releasing hormone agonist treatment for prostate cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2007;13(1):241–5.CrossRef
28.
Zurück zum Zitat Østergren P, Kistorp C, Fode M, Bennedbæk F, Faber J, Sonksen J. Luteinizing hormone releasing hormone agonists lower testosterone levels more than subcapsular orchiectomy: results from a randomized trial. J Urol. 2016;195(4):e324–5.CrossRef Østergren P, Kistorp C, Fode M, Bennedbæk F, Faber J, Sonksen J. Luteinizing hormone releasing hormone agonists lower testosterone levels more than subcapsular orchiectomy: results from a randomized trial. J Urol. 2016;195(4):e324–5.CrossRef
29.
Zurück zum Zitat Merseburger AS, Hupe MC. An update on triptorelin: current thinking on androgen deprivation therapy for prostate cancer. Adv Ther. 2016;33(7):1072–93. doi:10.1007/s12325-016-0351-4. Merseburger AS, Hupe MC. An update on triptorelin: current thinking on androgen deprivation therapy for prostate cancer. Adv Ther. 2016;33(7):1072–93. doi:10.​1007/​s12325-016-0351-4.
30.
Zurück zum Zitat Klotz L, O’Callaghan C, Ding K, Toren P, Dearnaley D, Higano CS, et al. Nadir testosterone within first year of androgen-deprivation therapy (ADT) predicts for time to castration-resistant progression: a secondary analysis of the PR-7 trial of intermittent versus continuous ADT. J Clin Oncol. 2015;33(10):1151–6.CrossRefPubMedPubMedCentral Klotz L, O’Callaghan C, Ding K, Toren P, Dearnaley D, Higano CS, et al. Nadir testosterone within first year of androgen-deprivation therapy (ADT) predicts for time to castration-resistant progression: a secondary analysis of the PR-7 trial of intermittent versus continuous ADT. J Clin Oncol. 2015;33(10):1151–6.CrossRefPubMedPubMedCentral
Metadaten
Titel
Efficacy of Testosterone Suppression with Sustained-Release Triptorelin in Advanced Prostate Cancer
verfasst von
Jürgen Breul
Eija Lundström
Daniela Purcea
Werner P. Venetz
Patrick Cabri
Pascale Dutailly
Evan R. Goldfischer
Publikationsdatum
27.12.2016
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 2/2017
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-016-0466-7

Weitere Artikel der Ausgabe 2/2017

Advances in Therapy 2/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.