Abstract
Importance of the field
Ovarian cancer remains a deadly malignancy because most patients develop recurrent disease that is resistant to chemotherapy, including platinum. Because response rates for current treatment regimens are relatively similar and unfortunately low, no standard chemotherapy for platinum-resistant ovarian cancer exists.
Areas covered in this review
A systematic literature review of clinical studies published between January 2005 and March 2010 was conducted using search engines, PubMed and MEDLINE with the entry keywords, ovarian cancer and platinum resistance. This search revealed 40 clinical trials (1793 patients).
What the reader will gain
Gemcitabine was the most common drug used in clinical trials reporting higher response rates, ≥+1 SD of overall response rate (5 out of 8). Gemcitabine-based combination therapy showed an average response rate of 27.2% (95%CI 22.4–32.0). Combination of gemcitabine and pegylated liposomal doxorubicin (PLD) was the most common regimen (n=3) and was associated with possible additive effects in platinum-resistant ovarian cancer patients: response rate, gemcitabine alone 6.1%, PLD alone 19.8%, and gemcitabine with PLD 28.7% (95%CI 20.4–37.0), respectively.
Take home message
Analysis of recent clinical trials showed that gemcitabine-based combination chemotherapy was associated with the highest anti-tumor effects in platinum-resistant ovarian cancer patients during the study period.
Keywords: clinical trial, gemcitabine, ovarian cancer, platinum resistance, recurrence, review
1. Introduction
In 2009, an estimated 21,550 women in the United States were diagnosed with ovarian cancer and approximately 14,600 died from this disease.1 The majority of ovarian cancer patients respond to initial therapy with tumor cytoreductive surgery and platinum-based chemotherapy.2 However, approximately 70% of advanced stage patients will develop recurrent cancer and eventually succumb to recurrent disease typically characterized by multiple drug resistance.3–4 Nevertheless, platinum-based chemotherapy remains the mainstay for treatment of recurrent disease. Platinum resistance, defined as tumor progression during or within six months after completion of prior platinum therapy, is common in recurrent disease.5–6 Due to the poor survival of women with platinum-resistant ovarian cancer, understanding the mechanisms contributing to platinum resistance as well as improved therapeutic approaches are urgently needed. Historically, a number of chemotherapeutic agents have been used for patients with platinum-resistant ovarian cancer. However, due to relatively similar response rates with salvage therapy, no standardized treatment exists for these patients. The current study presents a systematic review of contemporary clinical studies in platinum-resistant ovarian cancer to evaluate the effectiveness of various therapies tested.
2. Methods
A systematic review of literature was conducted using public search engines, PubMed and MEDLINE, with entry keywords: ovarian cancer and platinum resistance. The review was limited to the English literature published between January 1, 2005 and March 1, 2010. This time period was chosen to focus on contemporary clinical studies for platinum-resistant ovarian cancer. Of the 319 studies identified during the 5-year period, 60 (18.9%) were original clinical articles that were further evaluated. Only the patients with platinum-resistant epithelial ovarian cancer were evaluated for these analyses. Patients with platinum-sensitive cancer as well as trials presented only in an abstract form were excluded from this study. The following parameters were identified for each article: year of publication, study design (clinical trial or non-trials such as, case series and retrospective reviews), type of chemotherapy regimen, class of drug, number of patients enrolled, number of patients that responded to therapy, and response rate (defined as sum of rates for complete response and partial response by RECIST criteria).7 Stable disease as defined by RECIST criteria was not included in response rate. Survival outcomes for progression-free survival (PFS) and overall survival (OS) from the start of current chemotherapy for the recruited clinical studies were also abstracted. Studies reporting survival outcomes only for the responding patients were not included. Cross-comparison analyses among clinical trials that used the same chemotherapy regimen unifying the variables (subject number enrolled and patients responding to therapy) were performed in our study. Statistical comparisons of response rates to chemotherapy regimens were compared between the trials and between the non-trial studies. Correlation of the response rate to chemotherapy and survival outcome was determined using the curve of best fit. Continuous variables were tested for normality using the Kolmogorov-Smirnov (KS) test and expressed either by mean (±SD) or by median (range), as appropriate. Student t-test or Mann-Whitney U test was performed to determine the statistical significance of observed differences between study groups. Categorical variables were evaluated using Fisher’s exact test with odds ratios and 95% confidence intervals (95%CI). All statistical tests were 2-tailed, and a p value <0.05 was considered statistically significant. The statistical significance of the data was determined using Statistical Package for Social Scientists software (SPSS, version 18.0, Chicago, IL). Information for type of chemotherapy for ongoing clinical trials was abstracted from the public website (http://clinicaltrials.gov).
3. Recent clinical studies
Sixty clinical studies were published during the study period. Of those, 40 (66.7%) were clinical trials (phase II, 37 studies) and the remaining 20 (33.3%) were non-trial studies. A total of 35 drugs over 17 classes were evaluated, which resulted in 40 regimens (Table 1). The most commonly used drugs are shown in Table 2. Among recent studies, gemcitabine was studied in 10 (25%) out of 40 clinical trials and 6 (30%) out of 20 non-trial studies, respectively, making it the most common drug studied. Topotecan-based therapies showed the lowest response rates among the most commonly used drugs (19.9% in clinical trials and 17.8% in non-trials, respectively). Tables 3 and 4 list all the studies evaluated for the analyses of clinical trials and non-trial studies, respectively. Figure 1 demonstrates the response rate based on type of regimen (monotherapy versus combination therapy). There were 1793 patients enrolled in clinical trials, and an average response rate of 16.0% (±12.0) was noted in the 40 trials. The response rate was normally distributed (K-S test, p=0.75). There were 505 patients evaluated in 20 non-trial clinical studies with an average response rate of 26.3% (±16.2) and a normal distribution (K-S test, p=0.97). Average response rate in clinical trials was significantly lower than that found in non-trial clinical studies (p=0.02). The response rate to therapy was significantly lower in platinum-resistant compared to platinum-sensitive ovarian cancer patients. For example, the response rate to topotecan-based therapy in platinum-resistant patients was only 18% whereas the response rate was 44% in platinum-sensitive recurrent ovarian cancer patients.8 Use of gemcitabine (5 out 8) or pegylated liposomal doxorubicin (4 out of 8) was commonly seen in clinical trials that showed higher response rates (≥+1 SD [≥28%] above overall response rate, Table 3 and Figure 1). Conversely, monotherapy (7 out of 8) was commonly seen in the clinical trials that showed substantially lower response rates (≤−1 SD [≤4%] below overall response rate). Non-trial studies that showed ≥+1 SD of response rate (≥42.5%) were common in regimens using paclitaxel (2 out of 4, Table 4). The mean value of median PFS among the evaluated studies was 4.1 (±1.7) months in clinical trials and 4.4 (±2.0) months in non-trial studies, respectively. Mean value of median OS among the evaluated studies from the start of the current treatment was 12.3 (±3.3) months in clinical trials and 13.4 (±5.1) months in non-trial studies, respectively. The proportion of studies with mono- or combination therapy for platinum-resistant ovarian cancer was similar (30 studies, respectively). The efficacy of combination therapy was statistically higher than monotherapy for platinum-resistant ovarian cancer patients (clinical trials, 21.7 vs. 10.1%, p=0.009; non-trial studies, 33.7 vs. 18.9%, p=0.038).
Table 1.
Drug agents used in recent clinical studies for platinum-resistant ovarian cancer.
| Class | |||||
|---|---|---|---|---|---|
| Alkylating agents | canfosfamide | cyclophosphamide | ifosamide | irofulven | |
| Anthracyclines | doxorubicin | epirubicin | PLD | sabarubicin | |
| Cryptophycin analog | LY355703 | ||||
| Cytokine | interferon-α | ||||
| Epothilone B analog | ixabepilone | ||||
| Folate antimetabolites. | pemetrexed | ||||
| 5-Fu prodrug | capecitabine | ||||
| Hormonal related | goserelin | letrozole | 2-methoxyestradiaol | tamoxifen | |
| Immunosuppressive | cyclosporine | ||||
| Marine-derived agent | trabectedin | ||||
| Monoclonal antibody | bevacizumab | matuzumab | |||
| Nucleoside analog | gemcitabine | ||||
| Platinum agents | carboplatin | cisplatin | oxaliplatin | ||
| Small molecular inhibitors | gefitinib | imatinib | |||
| Taxanes | docetaxel | paclitaxel | |||
| Topoisomerase inhibitors | 9-aminocamptothecin | etoposide | irinotecan | CKD-602 | topotecan |
| Vinca alkaloids | navelbine |
Shown in alphabetical order.
Abbreviation: PLD, pegylated liposomal doxorubicin
Table 2.
Common drugs used in recent clinical studies for platinum-resistant ovarian cancer.
| Number (%) | Subjects | RR (%) | |
|---|---|---|---|
| Clinical trials (n=40) | |||
| Gemcitabine | 10 (25%) | 430 | 22.3 |
| Pegylated liposomal doxorubicin | 8 (20%) | 339 | 22.4 |
| Topotecan | 4 (10%) | 96 | 19.9 |
| Cisplatin | 3 (7.5%) | 114 | 21.9 |
| Docetaxel | 3 (7.5%) | 61 | 19.7 |
| Non-trial studies (n=20) | |||
| Gemcitabine | 6 (30%) | 141 | 22.7 |
| Paclitaxel | 4 (20%) | 89 | 39.3 |
| Topotecan | 3 (15%) | 113 | 17.8 |
| Carboplatin | 3 (15%) | 62 | 27.4 |
| Pegylated liposomal doxorubicin | 2 (10%) | 53 | 28.3 |
| Bevacizumab | 2 (10%) | 32 | 37.5 |
Analysis of 60 clinical studies published between January 2005 and March 2010.
Abbreviations: Number, number of clinical studies; Subjects, sum of enrolled patients in clinical studies; and RR, average response rate in any regimens using the listed agent.
Table 3.
Clinical trials conducted for platinum-resistant ovarian cancer.
| Year | Chemotherapy regimen | Subjects | RR (%) |
|---|---|---|---|
| 2006 | gemcitabine + epirubicin13 | 30 | 43.3 |
| 2008 | pegylated liposomal doxorubicin (biweekly)24 | 50 | 40 |
| 2008 | gemcitabine + pegylated liposomal doxorubicin14 | 48 | 37.5 |
| 2009 | gemcitabine + oxaliplatin17 | 50 | 37 |
| 2007 | gemcitabine + cisplatin (biweekly)19 | 35 | 31.5 |
| 2007 | irinotecan + doxorubicin67 | 13 | 30.8 |
| 2006 | gemcitabine + pegylated liposomal doxorubicin15 | 30 | 33.3 |
| 2006 | topotecan + pegylated liposomal doxorubicin26 | 27 | 28 |
| 2009 | topotecan + docetaxel (weekly)68 | 29¶ | 25 |
| 2006 | pegylated liposomal doxorubicin25 | 29 | 23.1 |
| 2007 | paclitaxel + ifosfamide + cisplatin30 | 22 | 22.7 |
| 2005 | gemcitabine + pegylated liposomal doxorubicin16 | 37 | 22 |
| 2009 | pemetrexed55 | 51 | 21 |
| 2005 | docetaxel + irinotecan69 | 30 | 20 |
| 2006 | paclitaxel (weekly)41 | 48 | 20.9 |
| 2008 | CKD-602 (camptotecan analog)58 | 5 | 20 |
| 2008 | gemcitabine + topotecan21 | 23 | 17 |
| 2006 | gemcitabine + cisplatin20 | 57 | 16 |
| 2007 | bevacizumab50 | 44 | 15.9 |
| 2005 | canfosfamide 70 | 34 | 15 |
| 2010 | ixabepilone53 | 49 | 14.3 |
| 2005 | 9-aminocamptothecin54 | 56 | 14 |
| 2006 | LY355703 (cryptophycin analog)57 | 24 | 12.5 |
| 2005 | tamoxifen + goserelin59 | 26* | 11.5 |
| 2007 | carboplatin + cyclosporine + interferon-α31 | 30** | 10 |
| 2009 | pemetrexed56 | 91 | 9.9 |
| 2007 | gemcitabine + oxaliplatin18 | 21 | 9.5 |
| 2008 | topotecan + pegylated liposomal doxorubicin27 | 22 | 9.1 |
| 2007 | pegylated liposomal doxorubicin†,12 | 96 | 8.3 |
| 2006 | capecitabine71 | 36 | 7.3 |
| 2007 | trabectedin72 | 79 | 6.3 |
| 2007 | gemcitabine†,12 | 99 | 6.1 |
| 2005 | sabarubicin73 | 19 | 5.3 |
| 2009 | canfosfamide74 | 232 | 4.3 |
| 2009 | capecitabine75 | 32 | 3.1 |
| 2008 | letrozole76 | 31 | 3 |
| 2006 | irofulven77 | 58 | 1.7 |
| 2006 | imatinib mesylate78 | 12 | 0 |
| 2007 | tamoxifen + gefitinib60 | 56 | 0 |
| 2007 | docetaxel (weekly)44 | 7 | 0 |
| 2007 | matuzumab79 | 30 | 0 |
Analysis of 40 clinical trials published between January 2005 and March 2010.
Different arms in the same study.
19 out of 29 were ovarian or peritoneal cancer.
17 out of 26 were platinum-resistant.
19 out of 30 were platinum-resistant.
Abbreviations: Subjects, number of patients enrolled in the trial; and RR, response rate (complete response rate + partial response rate or objective response rate if response rate is not available).
Table 4.
Non-trial clinical studies for platinum-resistant ovarian cancer.
| Year | Chemotherapy regimen | Subjects | RR (%) |
|---|---|---|---|
| 2009 | paclitaxel + carboplatin (extended weekly)32 | 20 | 60 |
| 2006 | paclitaxel (weekly)42 | 32 | 53 |
| 2007 | irinotecan + etoposide80 | 27 | 44.4 |
| 2008 | bevacizumab + cyclophosphamide51 | 9 | 44 |
| 2007 | paclitaxel + carboplatin (dose-dense)33 | 8 | 37.5 |
| 2006 | bevacizumab + cytotoxic agent*,52 | 23 | 34.8 |
| 2009 | gemcitabine + pemetrexed81 | 10 | 30 |
| 2006 | irinotecan (weekly)82 | 28 | 29 |
| 2009 | gemcitabine + pegylated liposomal doxorubicin83 | 35 | 28.6 |
| 2008 | gemcitabine + pegylated liposomal doxorubicin (biweekly)84 | 18 | 28 |
| 2009 | gemcitabine + cisplatin + ifosamide36 | 16 | 25 |
| 2008 | gemcitabine (fixed dose)85 | 41 | 22 |
| 2008 | docetaxel + imatinib86 | 23 | 21.7 |
| 2008 | topotecan (weekly)87 | 69 | 20.3 |
| 2008 | topotecan + navelbine8 | 22 | 18 |
| 2007 | tamoxifen88 | 29 | 10 |
| 2008 | topotecan (weekly)89 | 22 | 9.1 |
| 2005 | carboplatin35 | 34 | 5.9 |
| 2006 | gemcitabine90 | 21 | 4.7 |
| 2009 | 2-methoxyestradiol91 | 18 | 0 |
Analysis of 20 non-trial clinical studies published between January 2005 and March 2010.
combined cytotoxic agents include in the study included: cyclophosphamide (65%), 5-fluorouracil (26%), docetaxel (4%), and gemcitabine + liposomal doxorubicin (4%).
Abbreviations: Subjects, number of patients enrolled in the study; and RR, response rate (complete response rate + partial response rate or objective response rate if response rate is not available).
Figure 1.

4. Approaches for Treating Platinum Resistance
4.1. Gemcitabine
Gemcitabine was the most commonly used drug for the treatment of platinum-resistant ovarian cancer patients during the study period. This finding is interesting because gemcitabine has been commonly used in platinum-sensitive recurrent ovarian cancer, but less commonly in platinum-resistant disease. In these studies with gemcitabine-based therapy, 571 subjects were enrolled in 16 gemcitabine-based studies (Table 2). Gemcitabine is a deoxycytidine analog, disrupting DNA polymerization after incorporation during DNA synthesis into cancer cells.9 Gemcitabine also shows cytotoxic activity by inhibiting ribonucleotide reductase thereby disrupting DNA repair mechanisms.9 This approach is useful for platinum-resistant cancers because enhanced DNA repair is commonly observed in recurrent tumors previously exposed to platinum-based treatment.10 Furthermore, the combination of gemcitabine with cisplatin has been shown to have synergistic activity with regard to platinum-DNA adduct formation in pre-clinical studies.11
Gemcitabine monotherapy resulted in among the lowest response rates in platinum-resistant ovarian cancer, as demonstrated in a phase III randomized controlled trial (6.1%).12 However, once combined with other cytotoxic agents, response rates were significantly higher: combination therapy with any cytotoxic agents versus monotherapy, 27.2 vs. 6.1%, odds ratio 5.79, 95%CI 2.45–13.7, p<0.0001 (Figure 2). Gemcitabine-based therapy combined with epirubicin (48.1%, p<0.0001),13 with pegylated liposomal doxorubicin (28.7%, p<0.0001),14–16 with oxaliplatin (28.2%, p=0.0001),17–18 or with cisplatin (21.7%, p=0.003)19–20 demonstrated significantly higher response rates than treatment with gemcitabine alone. Combination therapy with gemcitabine and pegylated liposomal doxorubicin also showed a higher response rate than monotherapy with pegylated liposomal doxorubicin alone although it did not reach statistical significance (28.7 vs 19.8%, p=0.087). These studies suggest a possible additive or synergistic effect in the combined use of gemcitabine and pegylated liposomal doxorubicin. There were no trials that investigated monotherapy with cisplatin, oxaliplatin, or topotecan alone for platinum-resistant ovarian cancer patients during the study period. Combination of gemcitabine with topotecan did not reach statistical significance compared to gemcitabine monotherapy (17.4%, p=0.093).21
Figure 2.

Among the five types of combination therapy, gemcitabine with epirubicin showed a significantly higher response rate compared to gemcitabine with cisplatin (48.1 vs. 21.7%, odds ratio 3.36, 95% CI 1.36–8.25, p=0.013) and compared to gemcitabine with topotecan (48.1 vs. 17.4%, odds ratio 4.41, 95%CI 1.18–16.4, p=0.036). The remaining combination therapies did not show statistically significant differences between gemcitabine with pegylated liposomal doxorubicin, oxaliplatin, cisplatin, and topotecan. Epirubicin is a new generation anthracycline; therefore, no previous studies investigating its efficacy in combination with gemcitabine in ovarian cancer patients had been reported. Although anti-tumor activity with gemcitabine and epirubicin was the highest among the 40 clinical trials in platinum-resistant ovarian cancer (response rate 48.1%, 95%CI 29.3–67.0), the study size was relatively small, making it susceptible to type II error (n=30).13 Further trials are needed to better characterize the efficacy of this combination treatment among patients with platinum-resistant ovarian cancer. Combination gemcitabine with pegylated liposomal doxorubicin was the most common regimen (n=3, 7.5%) in the 40 clinical trials during the study period.14–16 This regimen was also associated with the third highest response rate among the evaluated chemotherapy regimens in the recent clinical trials (28.7%, 95%CI 20.4–37.0). The combination of these two agents was chosen because of differing mechanisms of action, possible synergistic effects, and minimally overlapping toxicity profiles.14 Although topotecan was commonly used in recent clinical trials, only one study evaluated the efficacy of gemcitabine combined with topotecan.21 The response rate of this trial was below the average of other clinical trials utilizing combination therapies (17.4 vs. 21.7%, Figure 1), and no statistical difference was seen in the response rate between gemcitabine alone and gemcitabine with topotecan (Figure 2). Prior to this study period, only one other study evaluating the combination of gemcitabine and topotecan in platinum-resistant ovarian cancer had been published,22 and a similar response rate was reported (12.5%). Therefore, in these limited clinical studies, adding gemcitabine with topotecan does not appear to confer any additional therapeutic benefit in platinum-resistant ovarian cancer.
4.2. Pegylated liposomal doxorubicin
Pegylated liposomal doxorubicin is polyethylene glycol coated liposomal doxorubicin.23 The strength of pegylated liposomal doxorubicin is that monotherapy alone produces sufficient anti-tumor effects in platinum-resistant ovarian cancer. In our analyses of clinical trials for platinum-resistant ovarian cancer, average response rate from 3 studies in which monotherapy was given was among the highest of all tested monotherapy agents (response rate 19.4%, Figure 1).12, 24–25 Of note, the average response rate of monotherapy in the setting of platinum-resistant cancer was only 10.1% (95%CI 8.4–11.8). The average response rate of pegylated liposomal doxorubicin monotherapy was similar to the average response rate from 13 clinical trials with combination therapy given to platinum-resistant ovarian cancer patients (21.7%, 95%CI 18.3–25.0). This information is useful for patients treated with pegylated liposomal doxorubicin because adverse effects during chemotherapy are often associated with multi-drug regimens and can be a limiting factor for discontinuation of chemotherapy.
Despite the multiple trials with pegylated liposomal doxorubicin, the second most commonly tested drug during the study period, there were only two types of combination therapy that were used with it (2 studies with topotecan26–27, 3 studies with gemcitabine14–16). The rationale to combine topotecan and pegylated liposomal doxorubicin originated from differences in dose-limiting toxicity profiles.26 However, the addition of topotecan to pegylated liposomal doxorubicin does not appear to confer additional therapeutic benefit (monotherapy versus topotecan combination, 19.4 vs. 19.1%, p=1.0).26–27 Conversely, the combination of pegylated liposomal doxorubicin with gemcitabine may result in additional benefit. Although it did not reach statistical significance, this is most likely due to the small sample size and the fact that the average response rate among trials with gemcitabine tended to be higher than trials with pegylated liposomal doxorubicin alone (response rate, 28.7 vs. 19.4%, odds ratio 1.67, 95%CI 0.96–2.9, p=0.087). These observations provide a rationale for developing additional combination regimens with pegylated liposomal doxorubicin.
The sequence of drug administration in the two trials of pegylated liposomal doxorubicin and topotecan was: pegylated liposomal doxorubicin followed by topotecan (response rate 28%)26; and simultaneous initiation of both drugs in each cycle (response rate 9.1%, p=0.14 compared to the other regimen).27 Although these two regimens did not demonstrate a statistically significant difference, the response rate of the regimen with simultaneous delivery was lower than the sequentially dosed regimen. Prior to the study period, there was a phase II clinical trial that showed a higher response rate with sequential administration of topotecan followed by etoposide (response rate 38%) in platinum-resistant ovarian cancer.28 In that study, pretreatment with topotecan was postulated to increase inhibition of topoisomerase II-alpha, which would allow sensitization to etoposide.28 Taken together, the sequence of administration of topotecan appears to be a potentially important factor affecting response to therapy. Due to the limited number of studies on this topic, a definitive answer remains to be determined. Further investigations are needed to determine the optimal sequence of topotecan-based chemotherapy.
4.3. Platinum agents
Platinum-based chemotherapeutics elicit their cytotoxic effects by forming intra-strand cross-links of DNA and platinum that induces cell apoptosis.29 Platinum remains the mainstay of treatment of recurrent disease in ovarian cancer patients. Current active members of available platinum agents include cisplatin, carboplatin, and oxaliplatin. In our analyses of published studies, 10 (16.7%) of the 60 clinical studies used platinum agents for patients with platinum resistance.17–20, 30–35 Among clinical trials, 6 (10%) studies used platinum-based therapies such as cisplatin (n=3),19–20, 30 carboplatin (n=1),31 and oxaliplatin (n=2).17–18 None of these studies used solely platinum-based monotherapy. Rather, all were combined with other cytotoxic agents such as gemcitabine (n=4), ifosfamide (n=1), paclitaxel (n=1), cyclosporine (n=1), and interferon (n=1) (Table 3). Among the non-trial studies, 4 (20%) studies evaluated cisplatin (n=1)36 or carboplatin (n=3).32–33, 35 One of 4 studies tested carboplatin monotherapy,35 and the remaining three tested combination therapy with paclitaxel (Table 4).
The issue of re-treatment with platinum agents in the setting of recurrent platinum-resistant ovarian cancer has been extensively debated. See and colleagues evaluated the effectiveness of re-treatment with carboplatin monotherapy in 34 patients diagnosed with platinum-resistant recurrent ovarian cancer.35 This approach was based on the premise that a prolonged platinum-free interval of more than 1 year may restore sensitivity to platinum drugs in patients with previously diagnosed platinum-resistant ovarian cancer. However, this study reported only 2 (5.9%) patients with a partial response, which was one of the lowest response rates in our analyses (more than 1 SD below the average response rate). Although some effect with platinum-monotherapy re-treatment may be evident in patients with platinum-resistant ovarian cancer, the broad use of this therapeutic strategy must be carefully evaluated prior to clinical implementation. On the other hand, when platinum agents were combined with other cytotoxic agents, these regimens were among the more effective treatments in platinum-resistant disease (response rate, 6 clinical trials 21.3%, 95%CI 16.0–26.7; and 3 non-trial studies 43.2%, 95% CI 28.5–57.8). A newer generation of platinum, oxaliplatin, was developed with the goal of overcoming platinum resistance. Indeed, oxaliplatin as a single agent or in combination with other agents in ovarian cancer demonstrated minimal cross-resistance with cisplatin or carboplatin,37 while simultaneously showing some therapeutic activity in recurrent ovarian cancer.38 In our analyses of clinical trials, oxaliplatin combined with gemcitabine showed similar response rates to cisplatin combined with gemcitabine (28.7 vs. 21.7%, p=0.37); however, many on-going pre-clinical studies are actively investigating new synthetic platinum compounds that may have future clinical applications.39–40
4.4. Taxanes
Paclitaxel and docetaxel are both taxanes that inhibit microtubule dis-aggregation. Both agents are commonly used in first line chemotherapy after primary cytoreductive surgery in ovarian cancer. Paclitaxel monotherapy showed the highest response rate among the tested monotherapy drugs in clinical trials given to platinum-resistant recurrent ovarian cancer patients (20.9%).41 Though not clinical trials, paclitaxel-based regimens employing a weekly dosing schedule also resulted in the two highest response rates (60% and 53%, respectively, Table 4).32, 42 Dose-dense combination therapy with carboplatin and paclitaxel to platinum-resistant ovarian cancer patients also showed a high response rate although it showed a wide 95% CI due to the limited subject number tested (n=8, response rate 37.5%, 95% CI 4–71%).33 While the original basis for the once weekly dose-dense schedule for paclitaxel administration was to maintain intense and prolonged exposure of the drug to the target cells,42 emerging data contends that the therapeutic effects may also reflect semi-metronomic dosing resulting in anti-angiogenic effects rather than purely a dose-density effect. In this setting, combination of paclitaxel with bevacizumab may be an attractive approarch for patients with platinum-resistant disease. A retrospective analysis of 55 patients with recurrent ovarian cancer treated with weekly paclitaxel and biweekly bevacizumab showed showed a 60% (95% CI 47.1–72.9) response rate including a 25% complete response rate.43 Future clinical development of this regimen is expected in platinum-resistant patients. Although docetaxel did not demonstrate a favorable therapeutic response in our analyses during the study period, this was most likely due to the limited patient sample size (n=7).44 Prior to the study period, 4 published phase II clinical trials evaluating docetaxel monotherapy in platinum-resistant ovarian cancer reported a wide range of response rates ranging from 6.9% to 35%.45–48 The overall response rate with docetaxel in platinum-resistant ovarian cancer patients among these five trials was 17.6% (95%CI 10.9–24.3) in 125 patients, which was comparable to paclitaxel monotherapy.44–48
4.5. Bevacizumab
Bevacizumab is a human monoclonal antibody against the critical pro-angiogenic factor, vascular endothelial growth factor (VEGF) that contributes to angiogenesis, tumor growth, and metastasis.49 In an analysis of recent clinical trials, bevacizumab monotherapy showed one of the highest response rates in platinum-resistant disease (bevacizumab versus all other monotherapy agents evaluated in our study, 15.9 vs. 10.1%, Figure 1).50 Although there were no published clinical trials evaluating combination therapy with bevacizumab during the study period, a retrospective analysis of 2 studies studying bevacizumab-based combination therapy (with cyclophosphamide, 5-fluorouracil, docetaxel, or gemcitabine + liposomal doxorubicin) reported promising response rates in platinum-resistant disease (37.5%, 95%CI 20.7–54.3, Table 4).51–52 Currently, multiple phase II clinical trials testing bevacizumab-based combination therapy either with docetaxel, topotecan, temsirolimus, pemetrexed, sorafenib, or erlotinib for platinum-resistant disease are ongoing.
4.6. Other new agents
Several new agents have been evaluated for platinum-resistant disease (Table 1). Response rates of monotherapy with ixabepilone (14.3%), 9-aminocamptothecin (14%), pemetrexed (13.4%), and LY355703 (12.5%) were comparable or higher across 19 evaluated regimens (overall response rate 10.1%, Figure 1). Ixabepilone is a microtubule-stabilizing epothilone B analog with anti-tumor activity in taxane-resistant breast cancer.53 DeGeest and colleagues extended these observations and reported the anti-tumor efficacy of ixabepilone in platinum-resistant ovarian cancer to be higher than average compared to other monotherapies tested.53 No published studies reporting the efficacy of combination therapy of ixabepilone were identified. 9-aminocamptothecin, a camptothecin analog, inhibits the activity of topoisomerase I.54 Despite its better than average anti-tumor activity among evaluated monotherapy agents in our study, the authors concluded that the prolonged infusion schedule (continuous infusion for 120 hours) may be a barrier to adopting this treatment in the future.54 Pemetrexed is an anti-folate agent that inhibits all folate-dependent metabolic enzymes involved in thymidine and purine nucleotide pathways.55–56 One of the studies that used pemetrexed monotherapy reported encouraging anti-tumor activity with a response rate among the highest of evaluated monotherapy regimens (21%, Table 3).55 Clinical trials for recurrent platinum-resistant and -sensitive ovarian cancer testing pemetrexed in combination with bevacizumab and with carboplatin are currently ongoing. LY355703 is a cytotoxic cryptophycin analog that destabilizes microtubules during mitosis.57 This mechanism of action is opposite to the one of taxanes. In pre-clinical experiments, LY355703 elicited anti-tumor effects in multidrug resistant cells, including taxane-resistant cells.57 CKD-602 is a newly developed camptothecin analog that inhibits topoisomerase I. CKD-602 showed superiority over parental drugs by overcoming the difficulty of toxicity and poor water solubility.58 Although the overall response rate of CKD-602 monotherapy was encouraging, the relatively small number of subjects (n=5) mandates further exploration of this agent before treatment recommendations can be developed. Tamoxifen-based therapy demonstrates very low response rates in platinum-resistant ovarian cancer in our analyses (average response rate, 3.6%).59–60
5. Role of evaluating progression-free survival in chemotherapy response
Chemotherapy response is generally determined by RECIST criteria, the mainstay of the standard evaluation of solid tumors, including ovarian cancer. These evaluations are based on the change in the size of tumors, and do not include survival functions such as PFS. Because PFS directly reflects the durability of the effect of a given chemotherapy in trials or studies, we evaluated potential relationships between PFS and chemotherapy response rates. As expected, PFS significantly correlated with chemotherapy response rates in platinum-resistant ovarian cancer studies (clinical trials, p<0.001; non-trial studies, p=0.01, Figure 3). However, a select sub-group of studies did not fit the results of this correlation and merit special attention. First, we divided results of clinical trials into 4 categories based on the cutoff values of PFS and response rate as determined by the mean values, 4.1 months and 16%, respectively. These cutoffs divide the trials into the following categories: Category A, both response rate and PFS were above the average; Category B, although response rate is above the average, PFS is below the average; Category C, although response rate is below the average, PFS is above the average; and Category D, both parameters were below the average (Figure 3). We established that Category A trials included desirable regimens while Category D regimens were not desirable. Category B regimens merit special attention because these studies actually, yet unexpectedly, showed significantly shorter PFS than studies with Category A regimens despite reportedly higher response rates (mean PFS in clinical trials, Category B vs. A, 3.1 vs. 5.1 months, p<0.001, Figure 3). Category C therapies, on the other hand, hold some promise of anti-tumor effects because these regimens resulted in longer PFS than average despite somewhat lower response rates. There was no statistical difference in PFS in Category C studies compared to Category A trials (mean PFS Category C vs. A, 5.8 vs. 5.1 months, p=0.24). Categories B or C, regarded as “gray zone regimens,” comprised approximately one third of the regimens with response rates higher or lower than the average of clinical trials, respectively (Category B, 30.8% in Category A+B; and Category C 27.3% in Category C+D). Table 5 shows the actual chemotherapy regimens based on response rates and PFS. Gemcitabine-based combination therapy remained a favorable regimen in more than half of trials in Category A (5 out of 9).
Figure 3.

Table 5.
Combining response rate with PFS in evaluating chemotherapy.
| Year | Chemotherapy regimen | RR (%) | PFS | OS |
|---|---|---|---|---|
| Category A: both RR and PFS above average (n=9) | ||||
| 2006 | gemcitabine + epirubicin13 | 43.3 | 8 | |
| 2008 | pegylated liposomal doxorubicin (biweekly)24 | 40 | 4.1 | 16.5 |
| 2008 | gemcitabine + pegylated liposomal doxorubicin14 | 37.5 | 6.9 | 18.2 |
| 2009 | gemcitabine + oxaliplatin17 | 37 | 4.6 | 11.4 |
| 2007 | gemcitabine + cisplatin (biweekly)19 | 31.5 | 4.9 | 13.2 |
| 2006 | topotecan + pegylated liposomal doxorubicin26 | 28 | 7.5 | 10.3 |
| 2006 | pegylated liposomal doxorubicin25 | 23.1 | 5.4 | 13.8 |
| 2005 | docetaxel + irinotecan69 | 20 | 5 | 11 |
| 2006 | gemcitabine + cisplatin20 | 16 | 5.4 | 14.9 |
| Category B: RR above average but PFS below average (n=4) | ||||
| 2006 | gemcitabine + pegylated liposomal doxorubicin15 | 33.3 | 3.8 | 15.8 |
| 2005 | gemcitabine + pegylated liposomal doxorubicin16 | 22 | 2.7 | 8.4 |
| 2009 | pemetrexed55 | 21 | 2.9 | 11.4 |
| 2008 | gemcitabine + topotecan21 | 17 | 3 | 12.6 |
| Category C: RR below average but PFS above average (n=6) | ||||
| 2007 | bevacizumab50 | 15.9 | 4.4 | 10.7 |
| 2005 | canfosfamide 70 | 15 | 7.2 | 14.1 |
| 2010 | ixabepilone53 | 14.3 | 4.4 | 14.8 |
| 2006 | LY355703 (cryptophycin analog)57 | 12.5 | 5.1 | |
| 2007 | gemcitabine + oxaliplatin18 | 9.5 | 5 | 9.2 |
| 2005 | sabarubicin73 | 5.3 | 4.2 | 2.1 |
| Category D: both RR and PFS below average (n=10) | ||||
| 2005 | 9-aminocamptothecin54 | 14 | 2.9 | 10 |
| 2009 | pemetrexed56 | 9.9 | 2.8 | 11.1 |
| 2008 | topotecan + pegylated liposomal doxorubicin27 | 9.1 | 2 | 10.5 |
| 2007 | pegylated liposomal doxorubicin†,12 | 8.3 | 3.1 | 13.5 |
| 2007 | gemcitabine†,12 | 6.1 | 3.6 | 12.7 |
| 2009 | canfosfamide74 | 4.3 | 2.3 | 13.5 |
| 2009 | capecitabine75 | 3.1 | 2.3 | |
| 2007 | tamoxifen + gefitinib60 | 0 | 1.9 | 8.4 |
| 2007 | docetaxel (weekly)44 | 0 | 3.1 | 12.3 |
| 2007 | matuzumab79 | 0 | 1.8 | 13.3 |
Total 29 clinical trials that both response rate and PFS were available were classified into 4 groups based on the cutoff values of overall response rate (16%) and PFS (4.1 months) among clinical trials.
Abbreviations: RR, response rate; PFS, median progression-free survival (months); and OS, median overall survival (months).
Platinum resistance, as defined by the Gynecologic Oncology Group (GOG), covers a relatively broad range of time frame for tumor recurrence. It is possible that the biology of tumors growing during postoperative chemotherapy administration (i.e., platinum refractory) may differ from tumors that recur at around the six month time period following completion of postoperative chemotherapy. It is also possible that tumors in Category B were more aggressive than Category A while tumors in Category C were less aggressive than Category D. In this setting, it may be useful to refine the definition of platinum resistance to cover a more narrow range of time than the current 6 months time frame. Some groups such as Gynecologic Cancer Intergroup (GCIG) are considering the 3 month time point for defining platinum-resistance instead of 6 months. In the current study, we proposed grouping PFS and response rate into 4 categories, which we believe may provide a more biologically pertinent classification. However, the classification of regimens into four categories based on chemotherapy response and PFS may potentially be arbitrary (Table 5). Due to the limited number of patients, it cannot be excluded that certain regimens may have been misclassified above or below average just by chance. Our classification describes the common observation that some patients may obtain a significant, but short response, while others may demonstrate modest but durable remissions. The general impression is that this phenomenon could be due to different biological behavior of the tumor rather than truly different activity of the treatment. The latter hypothesis could safely be tested only in the context of large prospective randomized trials.
6. Conclusion
We conducted a systematic review of 60 recent clinical studies that included a total of 2298 patients with platinum-resistant ovarian cancer. All together, the chemotherapy response rate in clinical trials for patients receiving any type of chemotherapy for platinum-resistant recurrent ovarian cancer was 16.0% (±12.0) in clinical trials, and 26.3% (±16.2) in non-trial clinical studies, respectively. As expected, combination therapy showed higher anti-tumor activity than monotherapy (clinical trial, 21.7 vs. 10.1%; and non-trial studies, 33.7 vs. 18.9%, respectively). Gemcitabine-based therapy was the most common regimen and reported the highest response rates to platinum-resistant ovarian cancer. Gemcitabine with pegylated liposomal doxorubicin and with platinum agents were well-studied regimens that demonstrated sufficient anti-tumor activity with possible synergistic effects demonstrated in relatively large study populations.
7. Expert Opinion
In 2006, the US Food and Drug Administration (FDA) approved the use of gemcitabine only for platinum-sensitive recurrent ovarian cancer patients. As of 2009, the FDA approved the use of gemcitabine combined with another cytotoxic agent for patients with advanced ovarian cancer who do not show response to other chemotherapy agents. Because gemcitabine-based combination therapy was among the most effective regimens for platinum-resistant ovarian cancer in our analyses (5 out of 8 clinical trials with ≥+1 SD improved response rates were gemcitabine-based therapy, Table 3), a gemcitabine-based combination therapy appears to be a favorable approach to overcome platinum-resistance in ovarian cancer patients. In our analyses combining PFS with response rate, gemcitabine-based combination therapy was the most common regimen in the subgroup showing substantial response rates and PFS (Category A in Table 5 and Figure 3). While modest improvements in response rates with combination chemotherapy may be attractive, the decision to use such regimens must carefully balance the potential for increased toxicity in a setting where cure is unattainable. Indeed, gemcitabine-based therapy was associated with considerable incidence of grade 3 or 4 neutropenia, and 62.5% of clinical trials with gemcitabine-based therapy reported a 50% or higher incidence of grade 3 or 4 neutropenia (range 26% to 80%).12–21 Furthermore, more toxicity was associated with gemcitabine-based therapy compared to non gemcitabine-based therapy. For example, in a phase III randomized trial, gemcitabine therapy was associated with increased risk of severe neutropenia compared to pegylated liposomal doxorubicin (grade 3 or 4, 38.4 vs. 18.8%, p=0.003).12 This toxicity profile associated with gemcitabine-based therapy could explain the reason for its lack of integration as the standard therapy for recurrent ovarian cancer. Further trials are expected to elucidate the efficacy of gemcitabine-based combination therapy.
The development of liposomal doxorubicin has resulted in improvement of drug delivery with a concomitant decrease in toxicity compared to free doxorubicin alone.23 A liposomal carrier is designed to have a longer circulating half-life and thus enhance the tumoral uptake of drug. Increased efflux of platinum is one of the known mechanisms associated with developing platinum-resistance in ovarian cancer.61 Therefore, developing mechanisms to either increase the uptake of cytotoxic agents into tumor cells or improve retention of cytotoxic agents within tumor cells may be an attractive approach to overcome platinum resistance. Pre-clinical studies of a newly synthesized pegylated liposomal cisplatin showed promising results with regard to increased intracellular cisplatin concentrations.62 Additional clinical development with pegylated liposomal cisplatin is warranted in platinum-resistant ovarian cancer. One delivery mechanism of liposomal drugs includes the endocytotic pathway. Inducing cellular endocytosis with macromolecular drug complexes was associated with increased drug uptake into platinum-resistant cell lines.39 Developing targeted drug delivery systems with developing new liposomal carriers is an important strategy to overcome platinum resistance and requires further investigation in this area.
Targeting DNA repair mechanisms is an attractive therapeutic approach in platinum-resistant ovarian cancer. Most of the platinum-based combination therapies in our analyses are fundamentally expected to have the additional benefit of targeting one of the major mechanisms of platinum resistance (i.e., enhanced repair mechanism of damaged DNA).10 The platinum and gemcitabine regimen was the most common combination among platinum-based trials, and this pairing was designed to inhibit ribonucleotide reductase to then block the repair of damaged DNA by gemcitabine.9 The response rates of platinum with gemcitabine were overall higher than the average response rates across all trials using combination therapy (24.5%, 95%CI 17.9–31.1). Other approaches targeting DNA repair mechanisms in platinum-resistant tumors in recent published trials have added cyclosporine to platinum.31 Cyclosporine alters the expression of oncogenes as well as other genes associated with the DNA repair mechanism, thereby reversing platinum resistance.31 Important molecular targets in the DNA repair mechanism in ovarian cancer include poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP),63 excision repair cross-complementation group 1 (ERCC1),64 and breast cancer gene (BRCA).65 Multiple pre-clinical studies have demonstrated that alterations of these enzymes are associated with platinum resistance in ovarian cancer, and phase II clinical trials with PARP inhibitors are underway in ovarian cancer patients. Future clinical trials are expected to further evaluate the efficacy of targeting DNA repair mechanisms. Of additional interest may be to further evaluate the combination of a platinum agent with an inhibitor of the DNA damage repair mechanism. Bearing in mind that altered DNA repair mechanisms are not the only pathways of platinum resistance, other mechanisms contributing to platinum resistance worthy of exploration include abnormal platinum transport.61 Increased expression of ATP7B, a metal transporter, is associated with platinum-resistant ovarian cancer. Inhibiting ATP7B with small interference RNA (siRNA) reversed cisplatin resistance in platinum-resistant cells.61 RNA interference therapy with siRNA offers a unique therapeutic option because (i) many important targets are not “drugable” by other methods, (ii) the functional protein structure is not known for many targets, (iii) many tyrosine kinases also have known and unknown kinase-independent functions, and (iv) multiple important and functionally relevant phosphorylation sites often exist within a target. Although RNA interference therapy is currently in pre-clinical stages, the potential clinical utility of RNA interference therapy deserves special consideration.
Some potential limitations of our study include the following: (i) The number of lines of chemotherapy administered to the patient is an important aspect that should be considered in reference to response rates; (ii) although our studies evaluated for the analysis were predominantly similar, that is, platinum-resistant ovarian cancer, indirect comparisons across different studies makes it difficult to draw definitive conclusions regarding the efficacy of chemotherapy. Nevertheless, our study offers valuable information for design of future trials; (iii) the primary end-point of therapy in the setting of recurrent ovarian cancer is palliation; therefore, the quality of life should also be considered.66
8. Article Highlights.
Ovarian cancer remains the leading cause of death among gynecologic malignancies. Platinum resistance is commonly seen in recurrent ovarian cancer patients. There is currently no standard chemotherapy for recurrence.
Overall chemotherapy response rate in clinical trials for patients who received any type of chemotherapy for platinum-resistant recurrent ovarian cancer was 16.0% with mean progression-free survival of 4.1 months from the treatment. Combination therapy showed higher response rates than monotherapy (21.7 vs. 10.1%).
Gemcitabine was the most common drug used in clinical trials. Gemcitabine-based combination therapy showed an average response rate of 27.2%, and was the most common therapy among the group of regimens with above average response rate and progression-free survival.
Footnotes
Declaration of interest
K Matsuo is supported by the GCF/OCRF Ann Schreiber Ovarian Cancer Research grant, the Betty Anne Asche Murray Distinguished Professorship and an award from the Meyer and Ida Gordon Foundation #2. Portions of this work were supported by the NIH (P50 CA083639, P50 CA098258, CA128797, RC2GM092599), the Ovarian Cancer Research Fund, Inc. (Program Project Development Grant), the DOD (OC073399, OC093146, BC085265) to AK Sood and K Matsuo. YG Lin is supported by the Woman’s Cancer Programme, University of Southern California. L Roman has nothing to disclose.
References
- 1.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59:225–49. doi: 10.3322/caac.20006. [DOI] [PubMed] [Google Scholar]
- 2.Omura G, Blessing JA, Ehrlich CE, et al. A randomized trial of cyclophosphamide and doxorubicin with or without cisplatin in advanced ovarian carcinoma. A Gynecologic Oncology Group Study. Cancer. 1986;57:1725–30. doi: 10.1002/1097-0142(19860501)57:9<1725::aid-cncr2820570903>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
- 3.Matsuo K, Bond VK, Eno ML, Im DD, Rosenshein NB. Low drug resistance to both platinum and taxane chemotherapy on an in vitro drug resistance assay predicts improved survival in patients with advanced epithelial ovarian, fallopian and peritoneal cancer. Int J Cancer. 2009;125:2721–7. doi: 10.1002/ijc.24654. [DOI] [PubMed] [Google Scholar]
- 4.Matsuo K, Eno ML, Im DD, Rosenshein NB, Sood AK. Clinical relevance of extent of extreme drug resistance in epithelial ovarian carcinoma. Gynecol Oncol. 116:61–5. doi: 10.1016/j.ygyno.2009.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Markman M, Rothman R, Hakes T, et al. Second-line platinum therapy in patients with ovarian cancer previously treated with cisplatin. J Clin Oncol. 1991;9:389–93. doi: 10.1200/JCO.1991.9.3.389. [DOI] [PubMed] [Google Scholar]
- 6.Gore ME, Fryatt I, Wiltshaw E, Dawson T. Treatment of relapsed carcinoma of the ovary with cisplatin or carboplatin following initial treatment with these compounds. Gynecol Oncol. 1990;36:207–11. doi: 10.1016/0090-8258(90)90174-j. [DOI] [PubMed] [Google Scholar]
- 7.Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205–16. doi: 10.1093/jnci/92.3.205. [DOI] [PubMed] [Google Scholar]
- 8.Herzog TJ, Powell MA, Rader JS, et al. Phase II evaluation of topotecan and navelbine in patients with recurrent ovarian, fallopian tube or primary peritoneal cancer. Gynecol Oncol. 2008;111:467–73. doi: 10.1016/j.ygyno.2008.08.005. [DOI] [PubMed] [Google Scholar]
- 9.Peters GJ, Van Moorsel CJ, Lakerveld B, et al. Effects of gemcitabine on cis-platinum-DNA adduct formation and repair in a panel of gemcitabine and cisplatin-sensitive or -resistant human ovarian cancer cell lines. Int J Oncol. 2006;28:237–44. [PubMed] [Google Scholar]
- 10.Wynne P, Newton C, Ledermann JA, Olaitan A, Mould TA, Hartley JA. Enhanced repair of DNA interstrand crosslinking in ovarian cancer cells from patients following treatment with platinum-based chemotherapy. Br J Cancer. 2007;97:927–33. doi: 10.1038/sj.bjc.6603973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.van Moorsel CJ, Pinedo HM, Veerman G, et al. Mechanisms of synergism between cisplatin and gemcitabine in ovarian and non-small-cell lung cancer cell lines. Br J Cancer. 1999;80:981–90. doi: 10.1038/sj.bjc.6690452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mutch DG, Orlando M, Goss T, et al. Randomized phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer. J Clin Oncol. 2007;25:2811–8. doi: 10.1200/JCO.2006.09.6735. [DOI] [PubMed] [Google Scholar]
- 13.Galligioni E, Arcuri C, Sorio R, Griso C. Gemcitabine and anthracyclines in platinum-resistant ovarian cancer. Ann Oncol. 2006;17 (Suppl 5):v195–8. doi: 10.1093/annonc/mdj980. [DOI] [PubMed] [Google Scholar]
- 14.Pectasides D, Xiros N, Papaxoinis G, et al. Gemcitabine and pegylated liposomal doxorubicin alternating with cisplatin plus cyclophosphamide in platinum refractory/resistant, paclitaxel-pretreated, ovarian carcinoma. Gynecol Oncol. 2008;108:47–52. doi: 10.1016/j.ygyno.2007.08.061. [DOI] [PubMed] [Google Scholar]
- 15.Petru E, Angleitner-Boubenizek L, Reinthaller A, et al. Combined PEG liposomal doxorubicin and gemcitabine are active and have acceptable toxicity in patients with platinum-refractory and -resistant ovarian cancer after previous platinum-taxane therapy: a phase II Austrian AGO study. Gynecol Oncol. 2006;102:226–9. doi: 10.1016/j.ygyno.2005.12.017. [DOI] [PubMed] [Google Scholar]
- 16.Skarlos DV, Kalofonos HP, Fountzilas G, et al. Gemcitabine plus pegylated liposomal doxorubicin in patients with advanced epithelial ovarian cancer resistant/refractory to platinum and/or taxanes. A HeCOG phase II study. Anticancer Res. 2005;25:3103–8. [PubMed] [Google Scholar]
- 17.Ray-Coquard I, Weber B, Cretin J, et al. Gemcitabine-oxaliplatin combination for ovarian cancer resistant to taxane-platinum treatment: a phase II study from the GINECO group. Br J Cancer. 2009;100:601–7. doi: 10.1038/sj.bjc.6604878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Harnett P, Buck M, Beale P, et al. Phase II study of gemcitabine and oxaliplatin in patients with recurrent ovarian cancer: an Australian and New Zealand Gynaecological Oncology Group study. Int J Gynecol Cancer. 2007;17:359–66. doi: 10.1111/j.1525-1438.2007.00763.x. [DOI] [PubMed] [Google Scholar]
- 19.Bozas G, Bamias A, Koutsoukou V, et al. Biweekly gemcitabine and cisplatin in platinum-resistant/refractory, paclitaxel-pretreated, ovarian and peritoneal carcinoma. Gynecol Oncol. 2007;104:580–5. doi: 10.1016/j.ygyno.2006.09.006. [DOI] [PubMed] [Google Scholar]
- 20.Brewer CA, Blessing JA, Nagourney RA, Morgan M, Hanjani P. Cisplatin plus gemcitabine in platinum-refractory ovarian or primary peritoneal cancer: a phase II study of the Gynecologic Oncology Group. Gynecol Oncol. 2006;103:446–50. doi: 10.1016/j.ygyno.2006.03.018. [DOI] [PubMed] [Google Scholar]
- 21.Goff BA, Holmberg LA, Veljovich D, Kurland BF. Treatment of recurrent or persistent platinum-refractory ovarian, fallopian tube or primary peritoneal cancer with gemcitabine and topotecan: a phase II trial of the Puget Sound Oncology Consortium. Gynecol Oncol. 2008;110:146–51. doi: 10.1016/j.ygyno.2008.04.037. [DOI] [PubMed] [Google Scholar]
- 22.Greggi S, Salerno MG, D’Agostino G, et al. Topotecan and gemcitabine in platinum/paclitaxel-resistant ovarian cancer. Oncology. 2001;60:19–23. doi: 10.1159/000055291. [DOI] [PubMed] [Google Scholar]
- 23.Gabizon A, Martin F. Polyethylene glycol-coated (pegylated) liposomal doxorubicin. Rationale for use in solid tumours. Drugs. 1997;54 (Suppl 4):15–21. doi: 10.2165/00003495-199700544-00005. [DOI] [PubMed] [Google Scholar]
- 24.Strauss HG, Hemsen A, Karbe I, Lautenschlager C, Persing M, Thomssen C. Phase II trial of biweekly pegylated liposomal doxorubicin in recurrent platinum-refractory ovarian and peritoneal cancer. Anticancer Drugs. 2008;19:541–5. doi: 10.1097/CAD.0b013e3282fcbbf7. [DOI] [PubMed] [Google Scholar]
- 25.Chou HH, Wang KL, Chen CA, et al. Pegylated liposomal doxorubicin (Lipo-Dox) for platinum-resistant or refractory epithelial ovarian carcinoma: a Taiwanese gynecologic oncology group study with long-term follow-up. Gynecol Oncol. 2006;101:423–8. doi: 10.1016/j.ygyno.2005.10.027. [DOI] [PubMed] [Google Scholar]
- 26.Verhaar-Langereis M, Karakus A, van Eijkeren M, Voest E, Witteveen E. Phase II study of the combination of pegylated liposomal doxorubicin and topotecan in platinum-resistant ovarian cancer. Int J Gynecol Cancer. 2006;16:65–70. doi: 10.1111/j.1525-1438.2006.00298.x. [DOI] [PubMed] [Google Scholar]
- 27.Rose PG, Smrekar M, Haba P, Fusco N, Rodriguez M. A phase I study of oral topotecan and pegylated liposomal doxorubicin (doxil) in platinum-resistant ovarian and peritoneal cancer. Am J Clin Oncol. 2008;31:476–80. doi: 10.1097/COC.0b013e31816a6221. [DOI] [PubMed] [Google Scholar]
- 28.Sood AK, Lush R, Geisler JP, et al. Sequential intraperitoneal topotecan and oral etoposide chemotherapy in recurrent platinum-resistant ovarian carcinoma: results of a phase II trial. Clin Cancer Res. 2004;10:6080–5. doi: 10.1158/1078-0432.CCR-04-0574. [DOI] [PubMed] [Google Scholar]
- 29.Zwelling LA, Kohn KW. Mechanism of action of cis-dichlorodiammineplatinum(II) Cancer Treat Rep. 1979;63:1439–44. [PubMed] [Google Scholar]
- 30.Polyzos A, Kosmas C, Tsavaris N, et al. Paclitaxel-ifosfamide-cisplatin as salvage chemotherapy in ovarian cancer patients pretreated with platinum compounds and paclitaxel. Anticancer Res. 2007;27:1645–51. [PubMed] [Google Scholar]
- 31.Morgan RJ, Jr, Synold TW, Gandara D, et al. Phase II trial of carboplatin and infusional cyclosporine with alpha-interferon in recurrent ovarian cancer: a California Cancer Consortium Trial. Int J Gynecol Cancer. 2007;17:373–8. doi: 10.1111/j.1525-1438.2007.00787.x. [DOI] [PubMed] [Google Scholar]
- 32.Sharma R, Graham J, Mitchell H, Brooks A, Blagden S, Gabra H. Extended weekly dose-dense paclitaxel/carboplatin is feasible and active in heavily pre-treated platinum-resistant recurrent ovarian cancer. Br J Cancer. 2009;100:707–12. doi: 10.1038/sj.bjc.6604914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cadron I, Leunen K, Amant F, Van Gorp T, Neven P, Vergote I. The “Leuven” dose-dense paclitaxel/carboplatin regimen in patients with recurrent ovarian cancer. Gynecol Oncol. 2007;106:354–61. doi: 10.1016/j.ygyno.2007.04.003. [DOI] [PubMed] [Google Scholar]
- 34.Polyzos A, Gogas H, Markopoulos C, et al. Salvage chemotherapy with oxaliplatin and capecitabine for breast cancer patients pretreated with anthracyclines and taxanes. Anticancer Res. 2009;29:2851–6. [PubMed] [Google Scholar]
- 35.See HT, Freedman RS, Kudelka AP, et al. Retrospective review: re-treatment of patients with ovarian cancer with carboplatin after platinum resistance. Int J Gynecol Cancer. 2005;15:209–16. doi: 10.1111/j.1525-1438.2005.15205.x. [DOI] [PubMed] [Google Scholar]
- 36.Polyzos A, Tsavaris N, Gogas H, et al. Cisplatin-Ifosfamide-gemcitabine as salvage chemotherapy in ovarian cancer patients pretreated with platinum compounds and Paclitaxel. Anticancer Res. 2009;29:2681–6. [PubMed] [Google Scholar]
- 37.Raymond E, Chaney SG, Taamma A, Cvitkovic E. Oxaliplatin: a review of preclinical and clinical studies. Ann Oncol. 1998;9:1053–71. doi: 10.1023/a:1008213732429. [DOI] [PubMed] [Google Scholar]
- 38.Fu S, Kavanagh JJ, Hu W, Bast RC., Jr Clinical application of oxaliplatin in epithelial ovarian cancer. Int J Gynecol Cancer. 2006;16:1717–32. doi: 10.1111/j.1525-1438.2006.00654.x. [DOI] [PubMed] [Google Scholar]
- 39.Garmann D, Warnecke A, Kalayda GV, Kratz F, Jaehde U. Cellular accumulation and cytotoxicity of macromolecular platinum complexes in cisplatin-resistant tumor cells. J Control Release. 2008;131:100–6. doi: 10.1016/j.jconrel.2008.07.017. [DOI] [PubMed] [Google Scholar]
- 40.Marques-Gallego P, den Dulk H, Brouwer J, et al. Cytotoxic activity and cellular processing in human ovarian carcinoma cell lines of a new platinum(II) compound containing a fluorescent substituted propylene diamine ligand. Biochem Pharmacol. 2009;78:365–73. doi: 10.1016/j.bcp.2009.04.027. [DOI] [PubMed] [Google Scholar]
- 41.Markman M, Blessing J, Rubin SC, Connor J, Hanjani P, Waggoner S. Phase II trial of weekly paclitaxel (80 mg/m2) in platinum and paclitaxel-resistant ovarian and primary peritoneal cancers: a Gynecologic Oncology Group study. Gynecol Oncol. 2006;101:436–40. doi: 10.1016/j.ygyno.2005.10.036. [DOI] [PubMed] [Google Scholar]
- 42.Le T, Hopkins L, Baines KA, Rambout L, Al Hayki M, Kee Fung MF. Prospective evaluations of continuous weekly paclitaxel regimen in recurrent platinum-resistant epithelial ovarian cancer. Gynecol Oncol. 2006;102:49–53. doi: 10.1016/j.ygyno.2005.11.025. [DOI] [PubMed] [Google Scholar]
- 43.Hurt JD, Richardson DL, Seamon LG, et al. Sustained progression-free survival with weekly paclitaxel and bevacizumab in recurrent ovarian cancer. Gynecol Oncol. 2009;115:396–400. doi: 10.1016/j.ygyno.2009.08.032. [DOI] [PubMed] [Google Scholar]
- 44.Tinker AV, Gebski V, Fitzharris B, et al. Phase II trial of weekly docetaxel for patients with relapsed ovarian cancer who have previously received paclitaxel--ANZGOG 02-01. Gynecol Oncol. 2007;104:647–53. doi: 10.1016/j.ygyno.2006.10.006. [DOI] [PubMed] [Google Scholar]
- 45.Francis P, Schneider J, Hann L, et al. Phase II trial of docetaxel in patients with platinum-refractory advanced ovarian cancer. J Clin Oncol. 1994;12:2301–8. doi: 10.1200/JCO.1994.12.11.2301. [DOI] [PubMed] [Google Scholar]
- 46.Katsumata N, Tsunematsu R, Tanaka K, et al. A phase II trial of docetaxel in platinum pre-treated patients with advanced epithelial ovarian cancer: a Japanese cooperative study. Ann Oncol. 2000;11:1531–6. doi: 10.1023/a:1008337103708. [DOI] [PubMed] [Google Scholar]
- 47.Markman M, Zanotti K, Webster K, Peterson G, Kulp B, Belinson J. Phase 2 trial of single agent docetaxel in platinum and paclitaxel-refractory ovarian cancer, fallopian tube cancer, and primary carcinoma of the peritoneum. Gynecol Oncol. 2003;91:573–6. doi: 10.1016/j.ygyno.2003.08.001. [DOI] [PubMed] [Google Scholar]
- 48.Berkenblit A, Seiden MV, Matulonis UA, et al. A phase II trial of weekly docetaxel in patients with platinum-resistant epithelial ovarian, primary peritoneal serous cancer, or fallopian tube cancer. Gynecol Oncol. 2004;95:624–31. doi: 10.1016/j.ygyno.2004.08.028. [DOI] [PubMed] [Google Scholar]
- 49.Burger RA. Experience with bevacizumab in the management of epithelial ovarian cancer. J Clin Oncol. 2007;25:2902–8. doi: 10.1200/JCO.2007.12.1509. [DOI] [PubMed] [Google Scholar]
- 50.Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol. 2007;25:5180–6. doi: 10.1200/JCO.2007.12.0782. [DOI] [PubMed] [Google Scholar]
- 51.Jurado JM, Sanchez A, Pajares B, Perez E, Alonso L, Alba E. Combined oral cyclophosphamide and bevacizumab in heavily pre-treated ovarian cancer. Clin Transl Oncol. 2008;10:583–6. doi: 10.1007/s12094-008-0254-7. [DOI] [PubMed] [Google Scholar]
- 52.Wright JD, Hagemann A, Rader JS, et al. Bevacizumab combination therapy in recurrent, platinum-refractory, epithelial ovarian carcinoma: A retrospective analysis. Cancer. 2006;107:83–9. doi: 10.1002/cncr.21969. [DOI] [PubMed] [Google Scholar]
- 53.De Geest K, Blessing JA, Morris RT, et al. Phase II clinical trial of ixabepilone in patients with recurrent or persistent platinum- and taxane-resistant ovarian or primary peritoneal cancer: a gynecologic oncology group study. J Clin Oncol. 2010;28:149–53. doi: 10.1200/JCO.2009.24.1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Miller DS, Blessing JA, Waggoner S, et al. Phase II evaluation of 9-aminocamptothecin (9-AC, NSC #603071) in platinum-resistant ovarian and primary peritoneal carcinoma: a Gynecologic Oncology Group Study. Gynecol Oncol. 2005;96:67–71. doi: 10.1016/j.ygyno.2004.09.015. [DOI] [PubMed] [Google Scholar]
- 55.Miller DS, Blessing JA, Krasner CN, et al. Phase II evaluation of pemetrexed in the treatment of recurrent or persistent platinum-resistant ovarian or primary peritoneal carcinoma: a study of the Gynecologic Oncology Group. J Clin Oncol. 2009;27:2686–91. doi: 10.1200/JCO.2008.19.2963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Vergote I, Calvert H, Kania M, Kaiser C, Zimmermann AH, Sehouli J. A randomised, double-blind, phase II study of two doses of pemetrexed in the treatment of platinum-resistant, epithelial ovarian or primary peritoneal cancer. Eur J Cancer. 2009;45:1415–23. doi: 10.1016/j.ejca.2008.12.013. [DOI] [PubMed] [Google Scholar]
- 57.D’Agostino G, del Campo J, Mellado B, et al. A multicenter phase II study of the cryptophycin analog LY355703 in patients with platinum-resistant ovarian cancer. Int J Gynecol Cancer. 2006;16:71–6. doi: 10.1111/j.1525-1438.2006.00276.x. [DOI] [PubMed] [Google Scholar]
- 58.Lee HP, Seo SS, Ryu SY, et al. Phase II evaluation of CKD-602, a camptothecin analog, administered on a 5-day schedule to patients with platinum-sensitive or -resistant ovarian cancer. Gynecol Oncol. 2008;109:359–63. doi: 10.1016/j.ygyno.2007.11.023. [DOI] [PubMed] [Google Scholar]
- 59.Hasan J, Ton N, Mullamitha S, et al. Phase II trial of tamoxifen and goserelin in recurrent epithelial ovarian cancer. Br J Cancer. 2005;93:647–51. doi: 10.1038/sj.bjc.6602752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wagner U, du Bois A, Pfisterer J, et al. Gefitinib in combination with tamoxifen in patients with ovarian cancer refractory or resistant to platinum-taxane based therapy--a phase II trial of the AGO Ovarian Cancer Study Group (AGO-OVAR 2.6) Gynecol Oncol. 2007;105:132–7. doi: 10.1016/j.ygyno.2006.10.053. [DOI] [PubMed] [Google Scholar]
- 61.Mangala LS, Zuzel V, Schmandt R, et al. Therapeutic Targeting of ATP7B in Ovarian Carcinoma. Clin Cancer Res. 2009;15:3770–80. doi: 10.1158/1078-0432.CCR-08-2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Krieger ML, Eckstein N, Schneider V, et al. Overcoming cisplatin resistance of ovarian cancer cells by targeted liposomes in vitro. Int J Pharm. 389:10–7. doi: 10.1016/j.ijpharm.2009.12.061. [DOI] [PubMed] [Google Scholar]
- 63.Nguewa PA, Fuertes MA, Cepeda V, et al. Poly(ADP-ribose) polymerase-1 inhibitor 3-aminobenzamide enhances apoptosis induction by platinum complexes in cisplatin-resistant tumor cells. Med Chem. 2006;2:47–53. doi: 10.2174/157340606775197697. [DOI] [PubMed] [Google Scholar]
- 64.Dabholkar M, Bostick-Bruton F, Weber C, Bohr VA, Egwuagu C, Reed E. ERCC1 and ERCC2 expression in malignant tissues from ovarian cancer patients. J Natl Cancer Inst. 1992;84:1512–7. doi: 10.1093/jnci/84.19.1512. [DOI] [PubMed] [Google Scholar]
- 65.Sakai W, Swisher EM, Karlan BY, et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature. 2008;451:1116–20. doi: 10.1038/nature06633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Herzog TJ. Recurrent ovarian cancer: how important is it to treat to disease progression? Clin Cancer Res. 2004;10:7439–49. doi: 10.1158/1078-0432.CCR-04-0683. [DOI] [PubMed] [Google Scholar]
- 67.Nishimura S, Tsuda H, Hashiguchi Y, et al. Phase II study of irinotecan plus doxorubicin for early recurrent or platinum-refractory ovarian cancer: interim analysis. Int J Gynecol Cancer. 2007;17:159–63. doi: 10.1111/j.1525-1438.2006.00728.x. [DOI] [PubMed] [Google Scholar]
- 68.Gupta D, Owers RL, Kim M, et al. A phase II study of weekly topotecan and docetaxel in heavily treated patients with recurrent uterine and ovarian cancers. Gynecol Oncol. 2009;113:327–30. doi: 10.1016/j.ygyno.2009.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Polyzos A, Kosmas C, Toufexi H, et al. Docetaxel in combination with irinotecan (CPT-11) in platinum-resistant paclitaxel-pretreated ovarian cancer. Anticancer Res. 2005;25:3559–64. [PubMed] [Google Scholar]
- 70.Kavanagh JJ, Gershenson DM, Choi H, et al. Multi-institutional phase 2 study of TLK286 (TELCYTA, a glutathione S-transferase P1-1 activated glutathione analog prodrug) in patients with platinum and paclitaxel refractory or resistant ovarian cancer. Int J Gynecol Cancer. 2005;15:593–600. doi: 10.1111/j.1525-1438.2005.00114.x. [DOI] [PubMed] [Google Scholar]
- 71.Wolf JK, Bodurka DC, Verschraegen C, et al. A phase II trial of oral capecitabine in patients with platinum--and taxane--refractory ovarian, fallopian tube, or peritoneal cancer. Gynecol Oncol. 2006;102:468–74. doi: 10.1016/j.ygyno.2005.12.040. [DOI] [PubMed] [Google Scholar]
- 72.Krasner CN, McMeekin DS, Chan S, et al. A Phase II study of trabectedin single agent in patients with recurrent ovarian cancer previously treated with platinum-based regimens. Br J Cancer. 2007;97:1618–24. doi: 10.1038/sj.bjc.6604088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Caponigro F, Willemse P, Sorio R, et al. A phase II study of sabarubicin (MEN-10755) as second line therapy in patients with locally advanced or metastatic platinum/taxane resistant ovarian cancer. Invest New Drugs. 2005;23:85–9. doi: 10.1023/B:DRUG.0000047110.36382.45. [DOI] [PubMed] [Google Scholar]
- 74.Vergote I, Finkler N, del Campo J, et al. Phase 3 randomised study of canfosfamide (Telcyta, TLK286) versus pegylated liposomal doxorubicin or topotecan as third-line therapy in patients with platinum-refractory or -resistant ovarian cancer. Eur J Cancer. 2009;45:2324–32. doi: 10.1016/j.ejca.2009.05.016. [DOI] [PubMed] [Google Scholar]
- 75.Pisano C, Morabito A, Sorio R, et al. A phase II study of capecitabine in the treatment of ovarian cancer resistant or refractory to platinum therapy: a multicentre Italian trial in ovarian cancer (MITO-6) trial. Cancer Chemother Pharmacol. 2009;64:1021–7. doi: 10.1007/s00280-009-0958-0. [DOI] [PubMed] [Google Scholar]
- 76.Ramirez PT, Schmeler KM, Milam MR, et al. Efficacy of letrozole in the treatment of recurrent platinum- and taxane-resistant high-grade cancer of the ovary or peritoneum. Gynecol Oncol. 2008;110:56–9. doi: 10.1016/j.ygyno.2008.03.014. [DOI] [PubMed] [Google Scholar]
- 77.Seiden MV, Gordon AN, Bodurka DC, et al. A phase II study of irofulven in women with recurrent and heavily pretreated ovarian cancer. Gynecol Oncol. 2006;101:55–61. doi: 10.1016/j.ygyno.2005.09.036. [DOI] [PubMed] [Google Scholar]
- 78.Coleman RL, Broaddus RR, Bodurka DC, et al. Phase II trial of imatinib mesylate in patients with recurrent platinum- and taxane-resistant epithelial ovarian and primary peritoneal cancers. Gynecol Oncol. 2006;101:126–31. doi: 10.1016/j.ygyno.2005.09.041. [DOI] [PubMed] [Google Scholar]
- 79.Seiden MV, Burris HA, Matulonis U, et al. A phase II trial of EMD72000 (matuzumab), a humanized anti-EGFR monoclonal antibody, in patients with platinum-resistant ovarian and primary peritoneal malignancies. Gynecol Oncol. 2007;104:727–31. doi: 10.1016/j.ygyno.2006.10.019. [DOI] [PubMed] [Google Scholar]
- 80.Nishio S, Sugiyama T, Shouji T, et al. Pilot study evaluating the efficacy and toxicity of irinotecan plus oral etoposide for platinum- and taxane-resistant epithelial ovarian cancer. Gynecol Oncol. 2007;106:342–7. doi: 10.1016/j.ygyno.2007.03.036. [DOI] [PubMed] [Google Scholar]
- 81.Gasent Blesa JM, Alberola Candel V, Provencio Pulla M, Esteban Gonzalez E, Martin Algarra S. Management of platinum-resistant ovarian cancer with the combination of pemetrexed and gemcitabine. Clin Transl Oncol. 2009;11:35–40. doi: 10.1007/s12094-009-0308-z. [DOI] [PubMed] [Google Scholar]
- 82.Matsumoto K, Katsumata N, Yamanaka Y, et al. The safety and efficacy of the weekly dosing of irinotecan for platinum- and taxanes-resistant epithelial ovarian cancer. Gynecol Oncol. 2006;100:412–6. doi: 10.1016/j.ygyno.2005.10.013. [DOI] [PubMed] [Google Scholar]
- 83.Karaoglu A, Arslan UY, Ozkan M, et al. Efficacy and toxicity of gemcitabine and pegylated liposomal Doxorubicin in recurrent platinum-resistant/refractory epithelial ovarian cancer. Asian Pac J Cancer Prev. 2009;10:63–6. [PubMed] [Google Scholar]
- 84.Tas F, Guney N, Derin D, Aydiner A, Topuz E. A pilot study evaluating the efficacy and toxicity of biweekly gemcitabine and pegylated liposomal doxorubicin in recurrent platinum-resistant epithelial ovarian cancer. Int J Clin Oncol. 2008;13:156–60. doi: 10.1007/s10147-007-0740-4. [DOI] [PubMed] [Google Scholar]
- 85.Ojeda Gonzalez B, Gonzalez Martin A, Bover Barcelo I, et al. A phase II trial of fixed-dosed rate gemcitabine in platinum-resistant ovarian cancer: a GEICO (Grupo Espanol de Investigacion en Cancer de Ovario) Trial. Am J Clin Oncol. 2008;31:481–7. doi: 10.1097/COC.0b013e31816d1c7b. [DOI] [PubMed] [Google Scholar]
- 86.Matei D, Emerson RE, Schilder J, et al. Imatinib mesylate in combination with docetaxel for the treatment of patients with advanced, platinum-resistant ovarian cancer and primary peritoneal carcinomatosis: a Hoosier Oncology Group trial. Cancer. 2008;113:723–32. doi: 10.1002/cncr.23605. [DOI] [PubMed] [Google Scholar]
- 87.Abushahin F, Singh DK, Lurain JR, Grendys EC, Rademaker AW, Schink JC. Weekly topotecan for recurrent platinum resistant ovarian cancer. Gynecol Oncol. 2008;108:53–7. doi: 10.1016/j.ygyno.2007.08.062. [DOI] [PubMed] [Google Scholar]
- 88.Karagol H, Saip P, Uygun K, et al. The efficacy of tamoxifen in patients with advanced epithelial ovarian cancer. Med Oncol. 2007;24:39–43. doi: 10.1007/BF02685901. [DOI] [PubMed] [Google Scholar]
- 89.Le T, Hopkins L, Baines KA, Rambout L, Fung-Kee-Fung M. Prospective evaluation of weekly topotecan in recurrent platinum-resistant epithelial ovarian cancer. Int J Gynecol Cancer. 2008;18:428–31. doi: 10.1111/j.1525-1438.2007.01041.x. [DOI] [PubMed] [Google Scholar]
- 90.Safra T, Ron I, Boaz M, et al. Heavily pretreated ovarian cancer patients treated by single-agent gemcitabine. A retrospective outcome comparison between platinum-sensitive and platinum-resistant patients. Acta Oncol. 2006;45:463–8. doi: 10.1080/02841860500509035. [DOI] [PubMed] [Google Scholar]
- 91.Matei D, Schilder J, Sutton G, et al. Activity of 2 methoxyestradiol (Panzem NCD) in advanced, platinum-resistant ovarian cancer and primary peritoneal carcinomatosis: a Hoosier Oncology Group trial. Gynecol Oncol. 2009;115:90–6. doi: 10.1016/j.ygyno.2009.05.042. [DOI] [PubMed] [Google Scholar]
