Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Original article Annals of Oncology 13: 251–257, 2002 DOI: 10.1093/annonc/mdf038 Chemotherapy versus hormonal treatment in platinum- and paclitaxel-refractory ovarian cancer: a randomised trial of the German Arbeitsgemeinschaft Gynaekologische Onkologie (AGO) Study Group Ovarian Cancer A. du Bois1,2*, W. Meier3,4, H. J. Lück5, G. Emons6,7, V. Moebus 8,9, W. Schroeder10,11, S. Costa12, T. Bauknecht13,14, S. Olbricht15, C. Jackisch16, B. Richter17 & U. Wagner18 1 Department of Gynecology & Gynecologic Oncology, Dr-Horst-Schmidt-Kliniken Wiesbaden; 2Department of Gynecology & Obstetrics, St Vincentius Krankenhaus Karlsruhe; 3Department of Gynecology & Obstetrics, ev. Krankenhaus Düsseldorf; 4Universitaetsfrauenklinik München-Grosshadern; 5 Department of Gynecologic Oncology, Universitaetsfrauenklinik Hannover; 6Universitaetsfrauenklinik Göttingen; 7Universitaetsfrauenklinik Marburg; 8 Department of Gynecology & Obstetrics, Städtische Kliniken Frankfurt-Höchst; 9 Universitaetsfrauenklinik Ulm; 10Department of Gynecologic Oncology, Zentralkrankenhaus Bremen; 11Universitaetsfrauenklinik Aachen; 12 Universitaetsfrauenklinik Frankfurt; 13Universitaetsfrauenklinik Bonn; 14 Universitaetsfrauenklinik Freiburg; 15Universitaetsfrauenklinik Magdeburg; 16 Universitaetsfrauenklinik Münster; 17Universitaetsfrauenklinik Dresden; 18 Universitaetsfrauenklinik Tübingen, Germany Received 30 April 2001; revised 8 August 2001; accepted 24 August 2001 Background: The majority of patients with ovarian cancer are not cured by first-line treatment. Until now, no study could demonstrate any substantial benefit when exposing ovarian cancer patients to second-line chemotherapy. However, most treatment regimens induce toxicity, thus negatively influencing the quality of rather limited life spans. Here we evaluate whether a second-line chemotherapy can offer any benefit compared with a less toxic hormonal treatment. Patients and methods: Patients with ovarian cancer progressing during platinum-paclitaxel containing first-line therapy or experiencing relapse within 6 months were eligible. Patients were stratified for response to primary treatment (progression versus no change/response), and measurable versus nonmeasurable disease. Treatment consisted of either treosulfan 7 g/m2 infused over 30 min or leuprorelin 3.75 mg injected subcutaneously or intramuscularly. Both regimens were repeated every 4 weeks. Results: This study began in late 1996, and after 2.5 years accrual an interim analysis was performed when several investigators reported their concern about a suspected lack of efficacy. Following this analysis the recruitment was stopped early and the 78 patients already enrolled were followed up. The majority of patients received treatment until progressive disease was diagnosed or death occurred. Treatment delay was observed rarely and dose reduction was performed only in the treosulfan arm in 5% of 150 courses. Overall, both treatment arms were well tolerated. No objective responses were observed. The median survival time was 36 and 30 weeks in the treosulfan and leuprorelin arms, respectively. Overall survival did not differ between patients with relapse 3–6 months after first-line chemotherapy compared with patients with progressive disease within 3 months. Conclusions: The selected patient population represents a subgroup with extremely poor prognosis. Accordingly, results were not impressive. Both treatment arms showed favourable toxicity data, but failed to show remarkable activity, thus adding only limited evidence to the issue of whether patients with refractory ovarian cancer might benefit from second-line chemotherapy. Even stratified analysis did not identify any subgroup of patients in whom the administration of second-line chemotherapy could demonstrate a clinically relevant survival benefit. Key words: chemotherapy, hormonal treatment, ovarian cancer, relapse, second-line therapy *Correspondence to: Department of Gynecology & Gynecologic Oncology, Dr Horst Schmidt Kliniken, Ludwig-Erhard-Strasse 100, D-65199 Wiesbaden, Germany. Tel: +49-611-43-3203; Fax: +49-611-43-3205; E-mail: [email protected] © 2002 European Society for Medical Oncology 252 Introduction Despite the considerable progress that has been achieved in the treatment of advanced ovarian cancer during the last decades, the majority of patients are still not cured by first-line treatment. Therefore, development of effective second-line treatment strategies remains a clinically relevant issue. Today standard first-line regimens in many countries contain both paclitaxel and a platinum analogue (e.g. cisplatin [1, 2] or carboplatin [3–5]). There are only limited data available reporting results gained from second-line therapy following failure of this new first-line regimen. Until now, no guidelines for the selection of second-line treatment regimens have reached universal acceptance [6]. Furthermore, the definitions of recurrent or relapsed disease according to the status of platinum resistance [7] were solely based on data from patients who did not receive the actual standard first-line regimens containing paclitaxel, and therefore have to be re-evaluated. The treatment-free interval, which offers a chance of gaining a benefit from re-treatment with paclitaxel and/or platinum, remains to be defined. However, patients progressing during or relapsing shortly after platinum-paclitaxel probably have a poor prognosis and can be regarded as refractory to both of the drugs they were exposed to. Until now, no study has demonstrated clearly any substantial benefit for these patients when treating them with second-line chemotherapy. However, most treatment regimens induce toxicity, thus negatively influencing the quality of rather limited life spans in this strictly palliative setting. Therefore, the AGO Study Group set about evaluating whether a second-line chemotherapy could offer any benefit compared with a less toxic hormonal treatment. The decision to use an alkylating agent for second-line chemotherapy was based on the assumption that these agents, which had been part of first-line treatment of ovarian cancer for decades, could offer some benefit as second-line agent after removal from first-line regimens. Treosulfan (Ovastat®, medac, Germany) was chosen as alkylating agent because it has been registered and used frequently in older first-line regimens in Germany, due to a more favourable non-haematological toxicity profile compared with cyclophosphamide [8, 9]. The published data for treosulfan as second-line treatment after platinum failure had been partially contradictory. Two studies using intravenous treosulfan reported response rates of up to 20% in 25 and 72 patients, respectively [10, 11]. The latter trial included 43 patients with platinum refractory ovarian cancer and showed a 21% response rate. Orally administered treosulfan resulted in response rates of 3, 14 and 19% in 30, 22 and 16 platinum pre-treated patients, respectively [12–14]. The only study reporting results of oral treosulfan in platinum refractory patients observed only one response in 30 patients. Therefore, we decided to use intravenous treosulfan as standard chemotherapy arm in this trial. Leuprorelin (leuproreline acetate; Enantone®, Takeda, Germany), a gonadotropin-releasing hormone (GnRH) analogue, was selected as hormonal treatment in the experimental arm of this study. It could be administered in a similar time schedule as the chemotherapy regimen (monthly injections) and had shown some activity in previously reported studies in platinum pre-treated ovarian cancer. In these trials, leuprorelin had been used either as single agent [15–17] or in combination with megestrole acetate or tamoxifen [18, 19]. Overall, nine responses have been reported in 46 platinum pre-treated patients [cumulative odds ratio (OR) 19.6%; 95% confidence interval (CI) 9% to 34%]. A retrospective review reported higher efficacy for leuprorelin compared with goserelin, thus providing further support for selecting leuprorelin in favour of other GnRH analogues [17]. However, platinum resistance had been reported inconsistently in all these studies, thus leaving some questions unanswered regarding efficiency in this particular group of patients. Toxicity profiles of leuprorelin had been uniformely reported as being mild, making this option potentially useful in this strictly palliative setting. Tamoxifen, another hormonal treatment with an 11% overall response rate reported in a meta-analysis in recurrent ovarian cancer [20], was not selected for this study, because the study group felt that the different mode of application could hamper comparability. Patients and methods This study was performed according to the Good Clinical Practice guidelines and was registered and certified by the German Cancer Society. It gained acceptance from ethics committees in each state of Germany and patients were only enrolled after giving written informed consent. Study procedures followed the AGO Standard Operating Procedures including central randomisation, regular on-site monitoring and double data entry. Patients with epithelial ovarian cancer and progressing during platinumpaclitaxel containing first-line therapy or experiencing relapse within the first 6 months following one platinum-paclitaxel containing regimen were eligible. Elevated CA 125 as the only marker of progressive disease was not accepted as a criterion for inclusion or for defining progression in this study. Further inclusion criteria were an Eastern Collaborative Oncology Group (ECOG) performance status of <3, a life expectancy of >12 weeks, being at least the age of consent, and adequate haematologic function [white blood cell (WBC) count 3000/mm 3, platelet count 100 000/mm3), liver function (bilirubin 1.25 × upper limit) and renal function (serum creatinine 1.25 × upper limit, glomerular filtration rate 40 ml/min estimated according to Jeliffe [21]). Exclusion criteria were: more than one prior chemotherapy regimen, one prior chemotherapy regimen not including platinum and paclitaxel, borderline tumours, Brenner tumours, known allergy to one of the treatment arms, disabling concomittant disease, bowl obstruction and second malignancies, except surgically controlled basal skin cancer or carcinoma in situ of the uterine cervix. Patients were stratified for response to primary treatment (progression during first-line chemotherapy versus no change/response), and bi-dimensionally (measurable versus non-measurable disease). Treatment consisted of either treosulfan 7 g/m2 infused over 30 min on day 1 or leuprorelin 3.75 mg injected s.c. or i.m. Both regimens were repeated every 4 weeks until progression or unacceptable toxicity occurred, or until the patient decided to end treatment. Reductions of treosulfan dose to 5 g/m2 was allowed for myelosuppression, and the administration of subsequent treatment courses was not allowed within 6 weeks. Patients were followed up until death at 253 3-monthly intervals. Toxicity was recorded for each treatment course and, in addition, patients with measurable disease were staged after every three Results courses using the methods that were appropriate for tumour measurement This study (AGO protocol OVAR-2.1) began in late 1996, and recruitment was slower than expected. After 2.5 years accrual an interim analysis was performed when several investigators reported their concern about a suspected lack of efficacy. A discussion among the study coordination group revealed that no significant response had been observed. The principal investigator decided not to continue further patient recruitment until there was clarification. Therefore, this initially unplanned interim analysis was performed. Following this analysis the recruitment was stopped early and patients already enrolled were followed up until this final analysis. Up to that time, 78 patients had been enrolled, of whom five were found to be ineligible. Three of these patients had been randomised to the chemotherapy arm. Reasons for ineligibility were withdrawal of consent (one patient), ECOG performance status of 3 (one), no proven progression after first-line therapy (one), prior treatment with platinum but without paclitaxel (one), relapse later than 6 months after completion of prior at baseline. The same evaluation methods were performed every 3 months during follow-up. The minimum requirements for evaluation in patients without measurable tumours included physical examination, bi-manual gynaecological examination and vaginal ultrasound. Responses were classified according to the WHO standard criteria [22]. The primary endpoint was survival; secondary end-points were progression-free survival, response in patients with measurable disease, and toxicity rates (NCI Common Toxicity Criteria). Patient number calculations were based on the assumption that treosulfan results in 60% survival after 6 months. A difference of 20% survival after 6 months (40% 6 month survival) should be detected with an 80% power and an error of 5%; thus, 58 patients needed to be enrolled per arm. Statistical analysis and data management were performed with SAS, FoxPro, East, Report/Dialog/Testimate/Topograph/N /Nsurv (IDV software). Primary end-point comparison was carried out utilising the Kaplan–Meier life-table method and the log-rank and Wilcoxon tests. The impact of treatment-free interval on results was analysed by bi-factorial univariate analysis. Table 1. Patients characteristics Treosulfan Leuprorelin All 39 39 78 3 2 5 No. eligible patients 36 37 73 Age, years: mean (range) 58.0 (36–75) 59.5 (27–75) 58.8 (27–75) ECOG 0 14 11 25 ECOG 1 16 19 35 ECOG 2 5 6 11 No. randomised patients No. ineligible patients Performance status Unknown 1 1 2 19 19 38 25 24 49 Cisplatin–paclitaxel 9 6 15 Epirubicin–carboplatin– paclitaxel 2 7 9 0–13 weeks 22 13 35 13–26 weeks 14 24 38 Measurable disease First-line regimen Carboplatin–paclitaxel Treatment-free interval Stage at initial diagnosis FIGO II 1 2 3 FIGO III 24 22 46 FIGO IV 10 13 23 <1 cm tumour residuals 13 10 23 1–2 cm tumour residuals 5 5 10 >2 cm tumour residuals 18 20 38 Status after initial surgery 254 therapy (one). Therefore, 73 patients were included in this analysis, of whom 36 received treosulfan and 37 received leuprorelin. One further patient was lost after receiving one treatment course and was classified as not evaluable for efficacy. The treatment groups were well balanced regarding patient characteristics and treatment history (Table 1). In contrast, treatment-free intervals differed between both study arms in favour of the leuprorelin arm. The latter included more patients with treatment-free intervals exceeding 13 weeks (63% versus 37% comparing leuprorelin with treosulfan). The median observation period was 22.5 months for all patients. The early termination of recruitment resulted in a statistical power of 80% to detect a 20% survival difference (50% versus 69.9%) after 6 months with two-sided testing and an α error of 0.05. Treatment and tolerability leuprorelin arm. Hot flushes were reported by one patient in each arm. Three further patients in the treosulfan arm reported grade 3 pain (two patients) and neurotoxicity (one). The latter was due to remaining toxicity from prior platinum-paclitaxel. Alopecia was common but was due to prior treatment. Regrowth of patients’ hair took longer in the treosulfan arm than in the leuprorelin arm. About one-third of patients still had alopecia after treatment with treosulfan compared with 11% in the leuprorelin arm. Fatigue was reported more frequently in the chemotherapy arm (eight of 36 patients versus one of 37 patients, treosulfan versus leuprorelin; P = 0.014, Fisher’s exact test). Overall, both treatment arms were relatively well tolerated resulting in only one treatment cessation due to toxicity. Efficacy The majority of patients received treatment until progressive disease was diagnosed or death occurred. The mean and median treatment periods, respectively, were 18 and 16 weeks in the treosulfan arm, and 13 and 10 weeks in the leuprorelin arm. Treatment delay was observed rarely and median intervals per course were 30.8 and 28.6 days in the treosulfan and leuprorelin arms, respectively. Dose reduction was performed only in the treosulfan arm in eight of 150 courses (5%) because of myelosuppression. Overall, 150 chemotherapy courses and 122 hormonal treatment courses were evaluable for toxicity. Haematological toxicities higher than grade 2 were observed in only a few patients. Thrombocytopenia grade 3/4 occurred in four and one courses in the treosulfan and leuprorelin arms, respectively. Neutropenia grade 3/4 was only observed in one course in each arm and no infections or neutropenic fever was reported. Anaemia greater than grade 2 was observed after seven courses in the treosulfan arm and after two courses in the leuprorelin arm. Non-haematological toxicities grade 3 or 4 were reported in only few patients in both arms. Treosulfan induced nausea and emesis in 9% of patients compared with 6% of patients in the No objective responses were observed in either of the treatment arms. Disease stabilisation lasting at least 4 weeks (no change) was reported in nine and four patients in the chemotherapy and hormonal treatment arm, respectively. All but one patient showed progressive disease within a median observation period of 22 months. Median progression-free survival was 17 weeks for treosulfan and 10 weeks for leuprorelin (P = 0.035, Wilcoxon test). The difference between both treatment arms remained significant in favour of treosulfan after adjusting for treatment-free interval before study entry (P = 0.028). However, after 6 months only 23% and 14% of patients in the treosulfan and leuprorelin arms had not progressed; corresponding figures for the 12 month observation period were 9% and 5%, respectively (Figure 1). At the time of this analysis, seven patients in the treosulfan arm and eight patients in the leuprorelin arm are still alive with disease [hazard ratio (HR) 0.98; 95% CI 0.58–1.67]. The differences observed between the treatment arms did not reach statistical significance (P = 0.87, Wilcoxon test; Figure 2). Again, adjusting for a treatment-free interval before study entry did not alter results. The median survival time was 36 and Figure 1. Progression-free survival (median 17 and 10 weeks for treosulfan Figure 2. Survival (median 36 and 30 weeks for treosulfan and leupro- and leuprorelin, respectively; P <0.05, log rank test; Kaplan–Meier curves). relin, respectively; P = 0.87, log rank test; Kaplan–Meier curves). 255 30 weeks in the treosulfan and leuprorelin arm, respectively. About one-third of patients in each arm were alive after 12 months. The impact of time to treatment failure after first-line chemotherapy on second-line therapy efficacy was analysed bicategorially. The cut-off was set at 13 weeks, thus comparing patients with progression under first-line therapy or early relapse within 3 months with patients who relapsed 3–6 months after completion of first-line chemotherapy. Overall, the difference between the groups with respect to progression-free survival did not reach statistical significance. Median progression-free survival was 11 and 12 weeks, respectively, for the two groups (P = 0.46, log rank test; HR 0.83; 95% CI 0.51–1.35). Furthermore, overall survival did not differ significantly between patients with relapse 3–6 months after first-line chemotherapy compared with patients with progressive disease within 3 months (P = 0.34, log rank test; HR 0.77; 95% CI 0.46–1.31). However, median survival was slightly superior in the group with a longer progression-free interval after first-line therapy (42 versus 25 weeks). The latter difference did not reach statistical significance and the Kaplan–Meier curves almost fell on top of each other shortly after 1 year of observation (data not shown). The presence of bi-dimensionally measurable disease had a negative impact on treatment results. Patients with measurable disease showed a median progression-free survival of 11 weeks compared with 19 weeks in patients with non-measurable disease (P = 0.0006, log rank test). Again, overall survival was superior in the group of patients with non-measurable disease, but this difference did not reach statistical significance (median 47 versus 24 weeks; P = 0.18, log rank test). Only 29% of patients with measurable disease compared with 46% of patients with non-measurable disease were alive after 12 months (HR 1.93; 95% CI 0.73–5.16). Subsequent treatment In the treosulfan arm, 15 patients received third-line treatments, of whom three were changed over to leuprorelin. The remaining eight patients received: radiotherapy (one), tamoxifen (one) or chemotherapeutic drugs [topotecan (six), etoposide (one), liposomal doxorubicin (one), carboplatin (one), carboplatin-paclitaxel (one)]. Furthermore, 14 patients received fourth-line treatment, including tamoxifen (two), MPA (one), etoposide (two), topotecan (two), and one patient each idarubicin, gemcitabin or mitoxantrone i.p. Finally, three patients received fifth-line cyclophosphamide (one), etoposide (one) or radiotherapy (one). In the leuproreline arm, almost all patients received third-line therapy. Sixteen patients were crossed over to treosulfan, four received intraperitoneal mitoxantrone, two had liposomal doxorubicin and one patient each received etoposide, topotecan, carboplatin, paclitaxelmitoxantrone or carboplatin-paclitaxel. Two patients received hormonal third-line treatment (one each received tamoxifen and MPA). Fourth-line treatment was offered to seven patients, including radiotherapy (one), topotecan (two), and one patient each liposomal doxorubicin-etoposide, etoposide or etoposide–5-fluorouracil (5-FU). Fifth-line treatment was offered to three patients, including paclitaxel, gemcitabin and 5-FU–platinum. The considerable use of third-line therapies after progression of disease might have hampered survival analysis, which in fact showed no significant difference between the treatment arms (although progression-free survival differed). Discussion This study represents a prospectively randomised trial in a very homogenous population. Only patients who were refractory to the standard first-line treatment of advanced ovarian cancer (i.e. platinum plus paclitaxel) were recruited. This selection represents a patient group with an extremely poor prognosis. At the moment, there is only limited evidence that these patients benefit from second-line chemotherapy at all, and more studies in this subgroup are necessary before any recommendations or guidelines can be established. A randomised trial of the National Cancer Institute of Canada has shown an advantage for one arm over another when comparing 3-weekly topotecan days 1–5 to weekly topotecan in 78 patients, of whom 60% had received prior paclitaxel, and 60% were platinum refractory [23]. This advantage was limited to overall response (23% versus 8%). Progression-free survival differed only at a non-significant level (8 versus 13 weeks), and overall survival did not differ at all. Our trial showed a statistically significant advantage of one arm (treosulfan) with respect to progression-free survival, but failed to show any difference in overall survival. In addition, no differences with respect to response rates were observed. In fact, we did not observe any objective response. The latter could indicate a lack of activity of both study drugs, treosulfan and leuprorelin. However, even higher response rates as reported in the literature did not translate to longer progression-free and overall survival. A prospectively randomised trial comparing liposomal doxorubicin with topotecan included 254 platinum refractory patients; in addition, about two-thirds had received paclitaxel as part of prior therapy [24]. No significant differences were observed in the refractory subgroup: response rates were 7% and 12%, median progression-free survival was 9 and 14 weeks, and median survival was 33 and 37 weeks, respectively. Our observations of median progression free survival of 11 and 17 weeks and median survival of 30 and 36 weeks fit well with the reported data in this poor prognostic subgroup, although we did not observe any objective responses. Another randomised trial in 81 platinum refractory patients comparing paclitaxel with paclitaxel–epirubicin reported response rates of 17% and 34% translating to 2-year survival of 10% and 18% [25]. The corresponding 2-year survival in our trial was 19% and 22%, thus indicating the limited value of objective response rates as predictors for survival or 256 progression-free survival in this poor prognostic subgroup of patients with truely refractory ovarian cancer. Nevertheless, achieving an objective response might be beneficial in this palliative setting, especially if bulky tumours induce symptoms such as pain or bowl obstruction. However, objective response rates might not sufficiently reflect this potential benefit. Therefore, different response criteria that better reflect the palliative approach in these patients should be evaluated prospectively (e.g. symptom relief, reduction of pain medication or ability of enteral food intake). The development of better tools for the evaluation of genuine second-line chemotherapies becomes even more necessary when taking into account the fact that ovarian cancer becomes more of a chronic disease: mortality does not change substantially, but median and 5-year survival improves, thus indicating a growing need for efficient second-line and higher treatment. These therapies should allow tumour control and simultaneously should not reduce life quality. This study reports mild toxicity data for both treatment arms, treosulfan and leuprorelin acetate, but, due to the very poor activity levels observed in both arms, adds only limited evidence to the issue of whether patients with refractory ovarian cancer benefit from second-line chemotherapy at all. Even stratified analysis in patients with progressive ovarian cancer versus patients experiencing relapse 3–6 months after firstline therapy, or patients with measurable versus non-measurable diseases, did not demonstrate any subgroup of patients in whom the administration of treosulfan second-line chemotherapy could demonstrate a clinically relevant benefit. Although a very short progression-free interval and the presence of bi-dimensionally measurable disease seemed to turn a bad prognosis into a worse prognosis, none of the differences between the strata showed a consistent and clinically relevant difference in survival. Only progression-free survival was influenced by these factors to some extent. Our data did not indicate that patients with a progression-free interval of >3 months but <6 months after first-line therapy have a better prognosis than those patients progressing within 3 months. Therefore, a progression-free interval of up to 6 months after first-line chemotherapy remained the inclusion criteria in our subsequent trials in this population. However, results were disappointing in all subgroups. A rather small benefit was traded for a higher rate of fatigue in patients receiving chemotherapy. A gain of 6 weeks median progression-free survival in the superior chemotherapy arm in our study and some advantages with respect to response rates in other trials do not convincingly answer the question of whether second-line chemotherapy offers any benefit for patients with refractory ovarian cancer. Further studies are necessary to help to evaluate whether chemotherapy has a role in this subgroup of patients with a very unfavourable prognosis. A randomised comparison between best supportive care and the most active chemotherapy regimen available could theoretically be an appropriate design for such a trial. How- ever, the German AGO investigators did not even broadly accept a randomisation between a hormonal treatment and a chemotherapy arm, as measured by an extremely slow recruitment rate. Furthermore, this study had to be closed prematurely after an interim analysis indicated only very limited activity in both treatment arms. A trial using best supportive care as one treatment arm would probably not be accepted either, although the above-mentioned efficacy data from chemotherapy studies do not provide more optimistic results. Treosulfan showed an acceptable toxicity profile and at least some activity compared with leuprorelin acetate, thus allowing continuation of clinical research with this alkylating agent. Our subsequent trial in the refractory population compares treosulfan with topotecan (AGO protocol OVAR-2.3) and recruitment is much better, indicating that investigators more easily accept trials comparing two chemotherapy regimens. Quality of life evaluation was included in this protocol in an attempt to improve understanding of the nature of potential gains from second-line therapy. Besides treosulfan and topotecan, which are further evaluated by our group, several chemotherapy agents have shown some activity in platinum- and paclitaxel-refractory ovarian cancer, and could serve as comparators in pending further trials: ifosfamide [26], hexamethylmelamine [27], gemcitabin [28] and liposomal doxorubicin [23, 29]. The difficult task of recruiting large homogenous patient populations to secondline trials supports the ongoing discussions and activities in cooperative groups and networks, such as the worldwide Gynecologic Cancer InterGroup (GCIG), which is already planning and performing intergroup trials of second-line treatment of ovarian cancer. Acknowledgements The authors thank Dr Sheila De Liz for helping with the language, and the investigators contributing to this study: K. Friese (Rostock), G. Batzke (Rottweil), H. J. Becker (Gardelegen), H. J. Bettex (Lüneburg), P. Richter (Plauen), O. Behrens (Rendsburg), R. Blumenberg/D. Kramer (Pforzheim), G. Deutsch (Karlsruhe), J. Felz-Süssenbach (Stadthagen), K. Gössel (Leer), H. Gröger (Bad Neuenahr), G. Hasselbauer (Aschersleben), L. Heilmann (Rüsselsheim), W. Herchenhein (Herzberg), U. P. Hoyme (Erfurt), D. Kranzfelder (Würzburg), M. Lange (Riesa), K. V. Maillot (Aalen), W. Meinerz (Paderborn), J. Meyer-Grohbrügge (Sigmaringen), H. Mickan (Esslingen), W. Niedner (Moers), E. Petri (Schwerin), A. Reichel (Burg), K. O. Reichl (Schorndorf), K. Robke (SteinfurtBorghorst), G. Stricker (Wipperfürth), T. Zeiser (HernstedtUlzburg). We also thank the pharmaceutical companies Takeda and medac, Germany for supporting the infrastructure of the AGO Study Group Ovarian Cancer. Furthermore, we thank Takeda for providing the study drug leuprorelin (leuproreline acetate, Enantone®). 257 References 1. Piccart MJ, Bertelsen K, James K et al. Randomized intergroup trial of cisplatin-paclitaxel versus cisplatin–cyclophosphamide in women with advanced epithelial ovarian cancer: three-year results. J Natl Cancer Inst 2000; 92: 699–708. 2. McGuire WP, Hoskins WJ, Brady MF et al. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl J Med 1996; 334: 1–6. 3. du Bois A, Lück HJ, Meier W et al. Cisplatin/paclitaxel vs carboplatin/paclitaxel in ovarian cancer: Update of an AGO trial. Proc Am Soc Clin Oncol 1999; 18: 356a (Abstr 1374). 4. Ozols RF, Bundy BN, Fowler D et al. Randomized phase III study of cisplatin (CIS)/paclitaxel (PAC) versus carboplatin (CARBO)/PAC in optimal stage III epithelial ovarian cancer (OC): a Gynecologic Oncology Group trial (GOG 158). Proc Am Soc Clin Oncol 1999; 18: 356a (Abstr 1373). 5. Neijt JP, Engelholm SA, Tuxen MK et al. Exploratory phase III study of paclitaxel and cisplatin versus paclitaxel and carboplatin in advanced ovarian cancer. J Clin Oncol 2000; 18: 3084–3092. 6. Berek JS, Bertelsen K, du Bois A et al. Advanced epithelial ovarian cancer: 1998 consensus statements. Ann Oncol 1999; 10 (Suppl. 1): 87–92. 7. Markman M, Hoskins W. Responses to salvage chemotherapy in ovarian cancer: a critical need for precise definitions of the treated population. J Clin Oncol 1992; 10: 513–514. 8. Merkle E, Ackermann S, Beck EP, Jäger W, Lang N. High-dose versus low-dose cisplatin chemotherapy plus treosulfan in epithelial ovarian carcinoma FIGO II–IV: results of a prospective randomized trial. Onkologie 2000; 23: 232–238. 9. Breitbach GP, Sass G, Bastert G. PT vs PC (Cis-platinum/treosulfan vs cis-platinum/cyclophosphamide) in advanced ovarian carcinoma: a phase III study. Proc Am Soc Clin Oncol 1993; 12: 261a (Abstr 832). 10. Hoffmann L, Koppermann S, Müllerleile U. Treosulfan in the treatment of ovarian carcinoma. Onkologie 1991; 14 (Suppl 3): 8–9 (Abstr). 11. Gropp M, Meier W, Hepp H. Treosulfan als effiziente second-linetherapie beim ovarialkarzinom. Geburtsh u Frauenheilkd 1997; 57: 356–360. 12. Keldsen N, Madsen EL, Havsteen H et al. Oral treosulfan as secondline treatment in platinum-resistant ovarian cancer: a phase II study. The Danish Ovarian Cancer Study Group. Gynecol Oncol 1998; 69: 100–102. 13. Kühnle H, Hilfrich J. Chemotherapie des fortgeschrittenen ovarialkarzinoms. Onkologie 1985; 8: 374–382. 14. Kramer D, Golz N, Glaser D, Mast H. Treosulfan als second-linetherapie beim fortgeschrittenen ovarialkarzinom. Arch Gynecol Obstet 1991; 250: 195–196. 15. Miller DS, Brady MF, Barrett RJ. A phase II trial of leuprolide acetate in patients with advanced epithelial ovarian carcinoma. A Gynecologic Oncology Group study. Am J Clin Oncol 1992; 15: 125–128. 16. Kavanagh JJ, Roberts W, Townsend P, Hewitt S. Leuprolide acetate in the treatment of refractory or persistent epithelial ovarian cancer. J Clin Oncol 1989; 7: 115–118. 17. Adelson MD, Reece MT. Effects of gonadotropin-releasing hormone analogues on ovarian epithelial tumors. Clin Obstet Gynecol 1993; 36: 690–700. 18. Bruckner HW, Motwani BT. Treatment of advanced refractory ovarian carcinoma with a gonadotropin-releasing hormone analogue. Am J Obstet Gynecol 1989; 161: 1216–1218. 19. Lopez A, Tessadrelli A, Kudelka AP et al. Combination therapy with leuprolide acetate and tamoxifen in refractory ovarian cancer. Int J Gynecol Cancer 1996; 6: 15–19. 20. Williams CJ. Tamoxifen in relapsed ovarian cancer: a systematic review. Int J Gynecol Cancer 1998; 8: 89–94. 21. Jeliffe RW. Creatinine clearance: bedside estimate. Ann Intern Med 1973; 79: 604. 22. Miller AB, Hoogstraten B, Staquet M, Winkler A. Reporting results of cancer treatment. Cancer 1981; 47: 207–214. 23. Hoskins P, Eisenhauer E, Beare S et al. Randomized phase II study of two schedules of topotecan in previously treated patients with ovarian cancer: a National Cancer Institute of Canada Clinical Trials Group study. J Clin Oncol 1998; 16: 2233–2237. 24. Gordon A, Fleagle J, Guthrie D et al. Interim analysis of a phase III randomized trial of doxil/caelyx (D) versus topotecan (T) in the treatment of patients with relapsed ovarian cancer. Proc Am Soc Clin Oncol 2000; 19: 380a (Abstr 1504). 25. Bolis G, Parazzini F, Scarfone G et al. Paclitaxel vs epidoxorubicin plus paclitaxel as second-line therapy for platinum-refractory and -resistant ovarian cancer. Gynecol Oncol 1999; 72: 60–64. 26. Scarfone G, Villa A, Parazzini F et al. A phase I–II trial of high-dose ifosfamide in patients with ovarian cancer refractory or resistant to platinum and/or paclitaxel-containing chemotherapy. Tumori 1999; 85: 217–219. 27. Schink JC, Harris LS, Grosen ER, Balley HH. Altretamine (Hexalen) an effective salvage chemotherapy after paclitaxel (Taxol) in women with recurrent platinum resistant ovarian cancer. Proc Am Soc Clin Oncol 1995; 14: 275 (Abstr 770). 28. Coenen M, Berteloot P, Amant F et al. Gemcitabine in platinum-paclitaxel resistant ovarian carcinoma. Proc Am Soc Clin Oncol 2000; 19: 405a (Abstr 1603). 29. Muggia FM, Safra T, Groshen S et al. Pegylated liposomal doxorubicin: antitumor activity in epithelial ovarian cancer or cancer of peritoneal origin. Oncology 1997; 11 (Suppl 11): 38–44.