Abstract
Background
Conventional first‐line combination therapy for ovarian cancer comprises 6 cycles of adjuvant or neoadjuvant carboplatin (AUC5‐6) with paclitaxel (175 mg/m2) every 3 weeks (PC‐3W). Weekly scheduling of paclitaxel may maximize its antiangiogenic effect and reduce adverse effects. We compared the efficacy and safety of PC‐3W with a modified protocol of weekly paclitaxel 80 mg/m2 and weekly carboplatin AUC2 administered on days 1, 8, and 15 in a 28‐day cycle (i.e., with 1 week off‐treatment [PC‐W]).
Materials and Methods
Medical records of consecutive patients treated between 2000 and 2018 were reviewed; 707 patients were analyzed for demographic and clinical characteristics, effectiveness and toxicity.
Results
PC‐3W was administered to 402 patients (median age, 60.5 years) and PC‐W to 305 patients (median age, 62.5 years). Most patients (91.4%) were diagnosed at stage III–IV. Notwithstanding a higher proportion of residual disease and older patients in the PC‐W group, median progression‐free survival was 21.4 months and 13.2 months for PC‐W and PC‐3W, respectively; median overall survival was 75.2 and 54.0 months for PC‐W and PC‐3W, respectively. Cox proportional hazards model indicated improved survival for patients treated with PC‐W (hazard ratio, 0.54). Similar results were observed for older patients diagnosed at ≥75 years. PC‐W demonstrated a better safety profile, with lower incidence of neuropathy, neutropenia, and alopecia.
Conclusion
PC‐W is as active and better tolerated than the standard PC‐3W regimen. PC‐W may serve as an alternative option for elderly or frail patients.
Implications for Practice
Weekly scheduling of paclitaxel 80 mg/m2 and carboplatin AUC2, administered on days 1, 8, and 15 in a 28‐day cycle (PC‐W) for first‐line therapy for advanced ovarian cancer, is as active and better tolerated than the standard regimen of carboplatin and paclitaxel (175 mg/m2) every 3 weeks (PC‐3W). It is possible that the weekly holiday on day 21 in the PC‐W regimen may ensure better completion rates (which may result in treatment delays for toxicity in PC‐3W). The results of this retrospective analysis highlight the weekly regimen as a valid treatment option, especially for elderly patients and those with significant comorbidities.
Keywords: Carcinoma, Ovarian epithelial, Weekly, Carboplatin, Paclitaxel
Short abstract
Conventional first‐line combination therapy for ovarian cancer is 6 cycles of adjuvant or neoadjuvant carboplatin with paclitaxel every three weeks. Weekly scheduling of paclitaxel may maximize its antiangiogenic effect and reduce adverse effects. This study compared the efficacy and safety of a weekly versus the three‐weekly protocol in a large cohort of patients with advanced epithelial ovarian cancer.
Introduction
Epithelial ovarian cancer (EOC) is the leading cause of gynecological cancer mortality [1, 2]. The conventional, and most studied, first‐line combination therapy for EOC consists of carboplatin (AUC5‐6) with paclitaxel (175 mg/m2) repeated every 3 weeks for six cycles either as an adjuvant or neoadjuvant administration. In randomized trials comparing this regimen with others, the 3‐weekly regimen was superior or noninferior to other regimens in terms of response rate, progression‐free survival (PFS), and overall survival (OS) [3, 4, 5, 6]. Its adverse events include alopecia, neurotoxicity, myelosuppression, and fatigue [6, 7].
Paclitaxel binds to tubulin and inhibits the disassembly of microtubules, thereby resulting in the inhibition of cell division [8, 9]. Weekly scheduling of paclitaxel has been suggested to increase its efficacy by reducing tumor regrowth between cycles and maximizing the drug's antiangiogenic effect [10, 11]. Weekly administration also reduces acute myalgia and arthralgia, presumably by lowering peak plasma concentrations [12]. Accordingly, several trials have explored paclitaxel dose schedules at shorter intervals [13, 14, 15, 16]. The Japanese Gynecologic Oncology Group (JGOG) reported remarkable superior efficacy in their “dose‐dense protocol” consisting of weekly paclitaxel (80 mg/m2) with carboplatin (AUC6) administered every 3 weeks versus the standard 3‐weekly administration [15]. However, subsequently the Gynecologic Oncology Group (GOG) failed to show such benefit in the GOG262 study that revisited such a comparison but included a percentage of patients with optional bevacizumab randomized to both arms [13]. A third study (MITO 7) reported similar effectiveness but fewer side effects for a weekly protocol consisting of carboplatin AUC2 with paclitaxel 60 mg/m2 compared with the standard 3‐weekly protocol [16]. Therefore, after nearly universal adoption of weekly paclitaxel regimens after the JGOG publications, the trend for induction regimens has moved back to ones that are administered every 3 weeks.
Since our phase II study of 64 patients from 2003 to 2007 evaluating the safety and efficacy of a modified double protocol [17], some of our physicians have continued to use this regimen, which we define as PC‐W. It consists of carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle (PC‐W). In the current study, we retrospectively compared the activity and safety of the PC‐W protocol to the standard 3‐weekly protocol (PC‐3W) in a large cohort of unselected patients with advanced EOC.
Materials and Methods
Setting and Patients
Medical records of all consecutive patients with advanced EOC, tubal carcinoma (TC), or primary peritoneal carcinomatosis (PPC) treated between 2000 and 2018 at the Onco‐Gynecological Unit of Tel Aviv Medical Center (629 cases, 89.0%) and at other medical centers (78 cases, 11.0%) were reviewed retrospectively.
All patients treated with a carboplatin‐paclitaxel regimen as a first‐line treatment (either PC‐W or PC‐3W) were included in the analysis. The first 64 patients on weekly schedule were those that were also included in the prospective, single‐arm, phase II study previously published [17].
The collected data included demographic and clinical characteristics, as well as survival status and toxicities.
Treatment Schedule
Patients underwent either primary surgical debulking or interval debulking after three to four neoadjuvant chemotherapy cycles. The 3‐weekly regimen consisted of the conventional doublet paclitaxel 175 mg/m2 and carboplatin AUC6, on day 1 of a 21‐day cycle repeated for 6 cycles (PC‐3W). The weekly regimen consisted of paclitaxel 80 mg/m2 and carboplatin AUC2 administered on days 1, 8, and 15 of a 28‐day cycle, repeated for six cycles (PC‐W). It is important to note that some patients with R2 and R1 residual disease received more than six cycles.
The choice to administer the PC‐W regimen was often based on the patient's performance status (i.e., older women or women with advanced diseases and especially severe ascites). In addition, the regimen was offered to women who preferred having severe grade 1 alopecia compared with grade 2 alopecia (i.e., complete hair loss). An additional requirement was the ability to come to the medical center every week to receive treatment. The physician explained to the patients that in phase II studies the PC‐W regimen showed similar effectiveness to the PC‐3W regimen, as well as lower toxicities (specifically alopecia and neuropathy), but that this was not confirmed in larger phase III studies.
Assessment of Outcomes and Safety
Serum cancer antigen (CA)‐125 levels were evaluated every three cycles during chemotherapy treatment. Complete blood count was performed before each treatment. A confirmed increase in serum CA‐125 levels to more than twice the upper limit of normal values (35 U/dL) was considered as disease progression for patients with no corresponding imaging evidence of disease.
OS was calculated from the initiation of treatment with carboplatin and paclitaxel to either death or to the last known follow‐up. PFS was calculated from the last treatment with carboplatin and paclitaxel to either progression or death or to the last known follow‐up. Platinum sensitivity was defined as recurrence of disease more than 6 months after the last platinum‐based treatment.
At the end of each treatment cycle, toxicity was evaluated according to the National Cancer Institute Common Terminology Criteria for Adverse Events version 2. At each follow‐up visit (for the duration of patient follow‐up) we also asked the patient if she had peripheral sensory neuropathy.
Statistical Analysis
For each treatment group, continuous variables were summarized as median and range, and categorical variables were summarized as number and percentage. Survival functions were demonstrated using the Kaplan‐Meier method, and the effect of each treatment group was assessed using the Log‐Rank test. Univariate comparisons between groups were performed using either the independent Student's t test or the Mann‐Whitney U test for continuous variables and Fisher's exact test or chi‐squared test for categorical variables.
The Cox proportional hazards model was used to compare the risk of death and the risk of a composite endpoint of death or recurrence between the two treatment regimens, adjusting for age, stage of disease, histology (serous papillary, adenocarcinoma, other), BRCA 1 and BRCA 2 status (carriers vs. noncarriers), and debulking status. The model for OS was also adjusted for platinum sensitivity (i.e., platinum‐free interval > 6 months). Time to recurrence and/or death was defined as the time from diagnosis until the relevant endpoint. Results are presented as the hazard ratio (HR) and the 95% confidence interval (CI). The level of significance used for all of the above analyses was two‐tailed, and a p value of <.05 was considered significant. Statistical analyses were performed using R software, version 3.6 (R Development Core Team, Vienna, Austria).
Results
Between 2000 and 2018, 707 patients with EOC, TC, or PPC were treated with first‐line PC‐3W (402, 56.9%) or PC‐W (305, 43.1%). Baseline characteristics are shown in Table 1. The median age of diagnosis of the patients treated with PC‐W was higher than the age of those treated by PC‐3W (62.5 vs. 60.0 years, p < .001). The majority of patients in both groups were diagnosed at stage III–IV. A higher proportion of patients treated by PC‐W had residual disease (R1 or R2) than those treated by PC‐3W (41.3% vs. 31.2%, p = .024). Median CA‐125 values at diagnosis were similar in both groups although widely distributed (849 U/mL in the PC‐3W group and 796 U/mL in the PC‐W group, p = .452). BRCA carrier status was tested in 475 patients (67.2%). Of these 475 patients, 317 (66.7%) were noncarriers, 120 (25.3%) were BRCA1 carriers, and 38 (8.0%) were BRCA2 carriers. Patients who were not tested for BRCA status (244/707, 34.5%) were considered BRCA negative in the analyses. A higher rate of patients in the PC‐W group were BRCA1 carriers compared with their rate in the PC‐3W group (22.3% vs. 12.8%, p = .005). The rate of patients who received neoadjuvant chemotherapy was higher in the PC‐W group compared with the PC‐3W with a trend for statistical significance (53.5% vs. 46.9%, p = .083). A similar percentage of patients in the PC‐W group received more than six treatment courses compared with the PC‐3W group (22.7% vs. 20.3%).
Table 1.
Demographic and clinical parameters of the study population
Variable | All patients, n = 707 | Patients ≥75 yr, n = 84 | ||||
---|---|---|---|---|---|---|
PC‐W, n = 305 | PC‐3W, n = 402 | p value | PC‐W, n = 46 | PC‐3W, n = 38 | p value | |
Age at diagnosis, yr | 62.5 (27.0–86.3) | 60.0 (21.5–86.9) | <.001 | 78.4 (75.1–86.3) | 77.9 (75.2–86.9) | .645 |
Stage, n (%) | .250 | .425 | ||||
Ic–IIc | 20 (6.6) | 40 (10.1) | 2 (4.4) | 0 (0) | ||
III | 234 (83.0) | 301 (84.1) | 36 (81.8) | 31 (81.6) | ||
IV | 48 (17.0) | 57 (15.9) | 8 (18.2) | 7 (18.4) | ||
Residual disease, n (%) | .024 | .280 | ||||
R0 | 175 (58.7) | 269 (68.8) | 20 (45.5) | 23 (62.2) | ||
R1 | 89 (29.9) | 89 (22.8) | 15 (34.1) | 10 (27.0) | ||
R2 | 34 (11.4) | 33 (8.4) | 9 (20.5) | 4 (10.8) | ||
Histology, n (%) | .013 | .057 | ||||
Serous papillary | 216 (70.8) | 249 (62.4) | 32 (69.6) | 17 (44.7) | ||
Adenocarcinoma (endometrioid) | 52 (17.1) | 105 (26.3) | 12 (26.1) | 16 (42.1) | ||
Poorly differentiated, muscinous, clear cell | 37 (12.1) | 45 (11.3) | 2 (4.4) | 5 (13.2) | ||
BRCA status, a n (%) | ||||||
BRCA1 | 68 (22.3) | 52 (12.9) | .005 c | 2 (4.4) | 3 (7.89) | .077 c |
BRCA2 | 16 (5.3) | 22 (5.5) | 2 (4.4) | 2 (5.26) | ||
Negative or unknown | 221 (72.5) | 328 (81.6) | 42 (91.3) | 33 (86.8) | ||
CA‐125, prior to treatment initiation, U/mL | 796 (5–17,877) | 849 (6–20,000) | .452 | 999 (12–16,700) | 803 (9–7,742) | .378 |
Neoadjuvant chemotherapy, n (%) | 161 (53.5) | 188 (46.9) | .083 c | 34 (73.9) | 19 (50.0) | .024 c |
Treatment courses | 6 (2–11) | 6 (1–14) | .145 | 6 (4–10) | 6 (4–8) | .214 |
>6 courses, n (%) | 67 (22.7) | 80 (20.3) | .435 | 12 (26.7) | 6 (15.8) | .231 |
Bevacizumab maintenance treatment, n (%) | 73 (23.9) | 68 (16.9) | .021 c | 4 (8.7) | 2 (5.3) | .543 c |
PARP inhibitors b maintenance treatment, n (%) | 20 (6.6) | 24 (6.0) | .749 | 20 (6.6) | 24 (6.0) | .749 |
Categorical variables are shown as n (%) and continuous variables are shown as median (range). The p values are based on chi‐squared tests for categorical variables and t test for continuous variables.
BRCA status was tested in 475 patients. Patients who were not tested were considered negative for BRCA.
Olaparib, rucaparib, or niraparib.
The difference between treatment regimens may reflect the chronology of entries, which was lower from 2000 to 2007.
Abbreviations: CA‐125, cancer antigen 125; PARP, Poly (ADP‐ribose) polymerase; PC‐W, neoadjuvant carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle; PC‐3W, neoadjuvant carboplatin (AUC6) with paclitaxel (175 mg/m2) every 3 weeks.
The disease recurred in 197 patients (64.6%) in the PC‐W group and in 298 patients (74.1%) in the PC‐3W group (unadjusted HR, .66; 95% CI, 0.55–0.80; p < .001, log‐rank test). After adjustment for age, stage, debulking, BRCA, and histological type, the HR was 0.53 (95% CI, 0.44–0.64; p < .001) in favor of the PC‐W group. Kaplan‐Meier survival analysis showed that median PFS was 21.4 months (95% CI, 18.8–24.2) for the PC‐W regimen and 13.2 months (95% CI, 11.6–16.6) for the PC‐3W regimen (p < .001, Fig. 1). A higher percentage of patients in the PC‐W compared with the PC‐3W group had platinum‐sensitive disease (88.9% vs. 76.3%, p < .001).
Figure 1.
Kaplan‐Meier curves of progression‐free survival (PFS) according to the time interval from initiation of treatment. Weekly: neoadjuvant carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle, median PFS is 21.37 months (95% confidence interval [CI], 18.8–24.2; range, 0‐175.4). Three weekly: neoadjuvant carboplatin (AUC6) with paclitaxel (175 mg/m2) every 3 weeks, median PFS is 13.2 months (95% CI, 11.6–16.6; range, 0–188.9). Log‐rank p value <.001, n = 688.
During the follow‐up period, 151 patients (49.5%) died in the PC‐W group and 222 (55.2%) died in the PC‐3W group (unadjusted HR, 0.70; 95% CI, 0.57–0.86; p < .001, log‐rank test). After adjustment for age, stage, debulking, BRCA, and histological type, the HR was 0.54 (95% CI, 0.43–0.67; p < .001) in favor of the PC‐W group. Kaplan‐Meier survival analysis showed that median OS was 75.2 months (95% CI, 68.4–86.4) for the PC‐W regimen compared with 54.0 months (95% CI, 48.4–65.7) for the for the PC‐3W regimen (p = .001, Fig. 2).
Figure 2.
Kaplan‐Meier curves of overall survival (OS) according to the time interval from initiation of treatment. Weekly: neoadjuvant carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle median, OS is 75.2 months (95% confidence interval [CI], 68.4–86.4; range, 2.7–211.5). Three weekly: neoadjuvant carboplatin (AUC6) with paclitaxel (175 mg/m2) every 3 weeks, median OS is 54.0 months (95% CI, 48.4–65.7; range, 0.0–249.0). Log‐rank p value <.001, n = 707.
We also analyzed the population of patients who were ≥75 years at diagnosis (n = 84). Baseline demographic characteristics were similar in both treatment groups (Table 1). In this group of older patients, the disease recurred in 32 patients (70%) in the PC‐W group and in 31 patients (82.0%) in the PC‐3W group (unadjusted HR, 0.63; 95% CI, 0.38–1.04; p = .068, log‐rank test). After adjustment for age, stage, debulking, BRCA, and histological type, the HR was 0.41 (95% CI, 0.22–0.76; p = .005) in favor of the PC‐W group. Kaplan‐Meier analysis showed a median PFS of 14.8 months (95% CI, 9.3–18.1) for the PC‐W regimen and 6.1 months (95% CI, 4.5–9.3) for the PC‐3W regimen (p = .07, Fig. 3).
Figure 3.
Kaplan‐Meier curves of progression‐free survival (PFS) according to the time interval from initiation of treatment in patients diagnosed at ≥75 years. Weekly: neoadjuvant carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle, median PFS is 14.8 months (95% confidence interval [CI], 9.3–18.1; range, 0–99.2). Three weekly: neoadjuvant carboplatin (AUC6) with paclitaxel (175 mg/m2) every 3 weeks, median PFS is 6.1 months (95% CI, 4.5–9.3; range, 0–188.9). Log‐rank p value = .068, n = 83.
During the follow‐up period, 27 patients ≥75 years (58.7%) died in the PC‐W group and 30 (79.0%) ‐ in the PC‐3W group (unadjusted HR, 0.54; 95% CI, 0.31–0.93; p = .0236, log‐rank test). After adjustment for age, stage, debulking, BRCA, and histological type, the HR was 0.28 (95% CI, 0.14–0.58; p < .001) in favor of the PC‐W group. Median OS by Kaplan‐Meier survival analysis was 50.8 months (95% CI, 34.0–68.1) for the PC‐W regimen compared with 25.0 months (95% CI, 17.2–32.3) for the PC‐3W regimen (p = .024, Figure 4).
Figure 4.
Kaplan‐Meier curves of overall survival (OS) according to the time interval from initiation of treatment in patients diagnosed at ≥75 years. Weekly: neoadjuvant carboplatin (AUC2) and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle, median OS is 50.8 months (95% confidence interval [CI], 34.0–68.1; range, 7.3–109.0). Three weekly: neoadjuvant carboplatin (AUC6) with paclitaxel (175 mg/m2) every 3 weeks, median OS is 25.0 months (95% CI, 17.2–32.3; range, 5.0–194.9). Log‐rank p value = .024, n = 84.
Toxicities
A significantly lower proportion of patients treated by the PC‐W regimen had grade ≥ 2 alopecia compared with those treated with the PC‐3W regimen (39.3%% vs. 88.8%, p < .001, Table 2). The subjective reports on grade 3–4 weakness were lower in the PC‐W regimen compared with the PC‐3W (3.6% vs. 6.2%, p = .15). The rate of grade ≥ 2 neutropenia was significantly lower in the PC‐W group compared with the PC‐3W group (44.9% vs. 53.0%, p = .007), but a similar percentage of patients received GCSF support in both groups (33.7% and 35.8% in the PC‐W and the PC‐3W groups, respectively, p = .570). The rate of anemia grade 3–4 was higher in the PC‐W group compared with the PC‐3W group (12.5% vs 4.5%, p = .001). The rate of thrombocytopenia grade 3–4 was higher for PC‐W than PC‐3W (10.2% vs. 4.5%) but not statistically significantly different. The rate of grade 3–4 neuropathy was significantly lower in the PC‐W group compared with the PC‐3W group (1.6% vs. 5.7%, p = .042). The rate of long‐term peripheral sensory neuropathy after >2 years of follow‐up was also significantly lower in the PC‐W group. None of the patients in the PC‐W group reported grade ≥ 3 nausea compared with 1.2% of patients in the PC‐3W group. In contrast, hypersensitivity reaction was diagnosed in 15 patients (9.7%) in the PC‐W group and in 22 patients (9.4%) in the PC‐3W (p = .937).
Table 2.
Comparison of toxicities between the carboplatin with paclitaxel treatment regimens
Toxicity | PC‐W (n = 305), n (%) | PC‐3W (n = 402), n (%) | p value |
---|---|---|---|
Neutropenia | |||
Grade 1–2 | 178 (58.4) | 245 (61.0) | <.001 |
Grade 3–4 | 57 (18.7) | 28 (7.0) | |
Thrombocytopenia | |||
Grade 1–2 | 90 (29.5) | 112 (27.9) | .1889 |
Grade 3–4 | 31 (10.2) | 26 (6.5) | |
Anemia | |||
Grade 1–2 | 198 (64.9) | 244 (60.7) | .001 |
Grade 3–4 | 38 (12.5) | 18 (4.5) | |
Alopecia | |||
Grade 2–4 | 120 (39.3) | 357 (88.8) | <.001 |
Nausea | |||
Grade 1–2 | 50 (16.4) | 69 (17.2) | .061 |
Grade 3–4 | 0 (0) | 5 (1.2) | |
Weakness | |||
Grade 1–2 | 107 (35.1) | 97 (24.1) | .015 |
Grade 3–4 | 11 (3.6) | 25 (6.2) | |
Neuropathy | |||
Grade 1–2 | 122 (40.0) | 208 (51.7) | .042 |
Grade 3–4 | 5 (1.6) | 23 (5.7) | |
Residual neuropathy | |||
None | 234 (76.7) | 249 (61.9) | <.001 |
Grade 1 | 59 (19.3) | 116 (28.9) | |
Grade ≥2 | 12 (3.9) | 37 (9.2) |
Abbreviations: PC‐W, neoadjuvant carboplatin and paclitaxel (80 mg/m2) administered on days 1, 8, and 15 in a 28‐day cycle; PC‐3W, neoadjuvant carboplatin with paclitaxel (175 mg/m2) every 3 weeks.
Concurrent Treatment
Bevacizumab was approved for OC treatment in Israel from 2014 onward for stage IV at diagnosis or for stage III with residual disease after surgery. In our cohort, only 23.9% and 16.9% of patients in the PC‐W and PC‐3W groups, respectively, were treated with bevacizumab. The 3‐year survival rate was higher in women who were treated concurrently with PC and bevacizumab compared with those who were not treated with bevacizumab (83.0% vs. 76.2%). In woman who were treated concurrently with bevacizumab and PC‐W, the 3‐year survival rate was higher compared with women who were treated concurrently with bevacizumab and PC‐3W (86.3% vs. 79.4%).
Discussion
Safe administration of paclitaxel and carboplatin in patients presenting with advanced ovarian cancer may be challenging because of several factors, including dose, schedule, the need for growth factors, and other supportive care measures, which are important issues in individual patient treatment. The thorough analysis of our institutional experience with the standard paclitaxel and carboplatin double and the weekly regimen adopted from our phase II study because of advantages in OS and tolerance suggests improved outcome and better toxicity profile for a modified PC‐W over the standard PC‐3W regimen.
Table 3 presents a summary of studies that evaluated different schedules of carboplatin‐paclitaxel regimens. The first prospective study depicting the dose‐dense method, in which paclitaxel chemotherapy was administrated at a weekly dose of 80 mg/m2 for 6 cycles of 21 days each (with carboplatin AUC6 at day 1), was published in the JGOG 3016 study in 2009 [15], with long‐term follow‐up results published in 2013 [14]. More than 600 women received six cycles of conventional chemotherapy (paclitaxel 180 mg/m2 plus carboplatin for an AUC of 6 mg/mL per minute given on day 1 of a 21‐day cycle) or a dose‐dense paclitaxel (80 mg/m2 given on days 1, 8, and 15) plus carboplatin given on day 21. Median PFS was longer in the dose‐dense treatment group (28.0 months vs. 17.2 months), as was the 5‐year survival (58.6% vs. 51.0%) [14]. Although withdrawal from the trial was higher in the dose‐dense protocol compared with the conventional protocol, because of toxicity (53% vs 37%), the only significant adverse effect greater in this cohort was grade 3 and 4 anemia. Yet, these results were not replicated in further studies. The American GOG‐0262 study, which included 692 patients [13], the European MITO‐7 study, which included 810 patients who received weekly paclitaxel but not in a dose‐dense method [16], and the ICON‐8 study, which included 1,566 patients [18], did not show any significant prolongation, nor shortening, of the OS or the PFS. The difference between these studies was attributed mainly to pharmacogenomic variations among the research populations, as well as to the changes in paclitaxel doses (Table 3) [19, 20, 21]. Notably, in the GOG‐0262 study, among patients who opted not to receive bevacizumab, weekly paclitaxel was associated with a median PFS that was 3.9 months longer than that observed with paclitaxel administered every 3 weeks [13].
Table 3.
Review of studies administering weekly and 3‐weekly regimens of paclitaxel and carboplatin.
Reference | No. of patients | Study type | Regimen | Paclitaxel dose, mg/m2 | Carboplatin dose, AUC | Cycle length, d | No. of cycles | Median OS; 95% CI, mo | Median PFS; 95% CI, mo | Grade 3–4 toxicity (unless specified) | Conclusions |
---|---|---|---|---|---|---|---|---|---|---|---|
Current study | 707 | Retrospective | 3W | 175 | 6 | 21 | 6 | 54.0 (48.4–65.7) | 13.2 (11.6–16.6) | Anemia: 5%; neutropenia: 7%; thrombocytopenia: 7%; neuropathy a : 6% (neuropathy a grade 2–4: 31%) | PC‐W administered in a 28‐day cycle improved PFS and OS and had better tolerability when compared retrospectively with the standard PC‐3W |
W | 80 | 2 | 28 | 6 | 75.2 (68.4–86.4) | 21.4 (18.8–24.2) | Anemia: 13%; neutropenia: 19%; thrombocytopenia: 10%; neuropathy a : 2% (neuropathy a grade 2–4: 21%) | ||||
Chan et al., 2016 [ 13 ]; GOG‐0262 | 692 | Prospective, randomized | 3W | 175 | 6 | 21 | 6 | 39.0 | 14.0 (10.3 without bevazicumab) | Anemia: 16%; neutropenia:83%; neuropathy a (grade 2–4): 18% | Among patients who did not receive bevacizumab, PC‐W was associated with a median PFS that was 3.9 months longer than that observed with PC‐3W |
W | 80 | 6 | 21 | 6 | 40.2 | 14.7 (14.2 without bevazicumab) | Anemia: 36% neutropenia:72%; neuropathy a (grade 2–4): 26% | ||||
Katsumata et al., 2009 [ 15 ]; Katsumata et al., 2013 [ 14 ]; JGOG 3016 | 600 | Prospective, randomized | 3W | 180 | 6 | 21 | 6 | 62.2 (52.1–82·6) | 17.5 (15.7–21.7) | Anemia: 44%; neutropenia: 88%; thrombocytopenia: 38%; neuropathy a : 6% | Dose‐dense treatment offers better survival than conventional treatment |
W | 80 | 6 | 21 | 6 | 100.5 (65.2–not reached) | 28.2 (22.3–33.8) | Anemia: 69%; neutropenia: 92%; thrombocytopenia: 38%; neuropathy a : 7% | ||||
Clamp et al., 2017 [ 18 ]; ICON‐8 | 1,566 | Prospective, randomized | 3W | 175 | 5/6 | 21 | 6 | 24.4 | All grade 3–4 toxicities: 42% | No benefit to any regimen | |
W | 80 | 5/6 | 21 | 6 | 24.9 | All grade 3–4 toxicities: 42% | |||||
W | 80 | 2 | 21 | 6 | 25.3 | All grade 3–4 toxicities: 42% | |||||
Pignata et al., 2014 [ 16 ]; MITO‐7 | 810 | Prospective, randomized | 3W | 175 | 6 | 21 | 6 | 17.3 (15.2–20.2) | Neutropenia: 50%; thrombocytopenia: 7%; neuropathy a grade 2–4): 17% | PC‐W improved quality of life and showed a better toxicity profile, but did not prolong PFS | |
W | 60 | 2 | 21 | 6 | 18.3 (16.8–20.9) | Neutropenia: 42%; thrombocytopenia: 1%; neuropathy a (Grade 2–4): 6% | |||||
Abaid et al., 2013 [28] | 88 | Prospective | W | 80 | 5/6 | 28 | 6 | 31.5 | 22.5 | Neutropenia: 22.7 %; thrombocytopenia: 7.9 %; anemia: 1.1 % | Modified dose‐dense PC‐W preserves the efficacy of traditional dose‐dense chemotherapy, while minimizing hematologic toxicity |
Ebata et al., 2016 [29] | 74 | Retrospective | W | 80 | 6 | 21 | 6 | 55.1 (44.6–not reached) | 19.0 (16.2–23.7) | Anemia: 41.9%; neutropenia: 55.4%; thrombocytopenia: 21.6%; neuropathy a : 8.1% | Dose‐dense carboplatin + paclitaxel was effective and tolerable |
Van der Burg et al., 2014 [ 30 ] | 267 | Prospective, randomized | 3W | 175 | 6 | 21 | 6 | 41.1 (34.4–47.7) | 16.4 (13.5–19.2) | Anemia: 3%; neutropenia: 38.2%; thrombocytopenia: 3%; neuropathy a : none | There was no benefit in terms of OS, PFS, or RR for a weekly regimen nor for extended chemotherapy as first‐line treatment for EOC in European patients |
W | 90 | 4 | 28 | 6 | 44.8 (33.1–56.5) | 18.5 (15.9–21) | Anemia: 13.5%; neutropenia: 61.2; thrombocytopenia: 11.4%; neuropathy a : 1.5% |
Randomized studies are shown in bold.
Peripheral sensory neuropathy.
Abbreviations: CI, confidence Interval; EOC, epithelial ovarian cancer; OS, overall survival; PC‐W, weekly paclitaxel and carboplatin regimen; PC‐3W, three‐weekly paclitaxel and carboplatin regimen; PFS, progression‐free survival; RR, response rate.
Our weekly carboplatin and paclitaxel regimen differs in schedule from others and in particular that of JGOG 3016 [15] and GOG‐262 [13]: we administered carboplatin on days 1, 8, and 15 of a 28‐day cycle at a dose of AUC2, whereas the above “dose‐dense” regimens were given weekly every 21 days. Thus, our protocol allows a “week off” from chemotherapy. In a meta‐analysis [22] that reviewed phase III clinical trials [13, 14, 16], which compared the 3‐weekly and the weekly (dose‐dense) paclitaxel and carboplatin treatment regimens for treatment of ovarian cancer, the authors reported that the dose‐dense regimen was associated with significant improvement of PFS compared with standard schedule, with HR of 0.73 (95% CI 0.61–0.88, p = .001). There was no difference in OS between treatment regimens (HR 0.95, 95% CI, 0.77–1.16, p = .06), as well as in term of severe acute toxicity. These findings corroborate our own experience with the dose‐dense treatment regimen. Some studies also showed an improvement in quality of life and patient satisfaction [16, 17].
As for the toxicity profile, the PC‐3W regimen is known to be toxic, causing alopecia, neurotoxicity, myelosuppression, and fatigue [3, 4, 23]. In the present analysis, the weekly paclitaxel regimen is at least as tolerable as the 3‐weekly one [13, 16]. Although the rates of hematological toxicities were higher in the PC‐W group, this observation might be attributed to the fact that PC‐W–treated patients were followed more closely and were evaluated weekly for toxicity. Therefore, there is a possible underestimation of bone marrow toxicity in the PC‐3W group because additional or midcycle bone marrow toxicity in this group may not have been reported. Of note is the observation that long‐term paclitaxel‐induced peripheral neuropathy was significantly lower in patients treated with the PC‐W regimen. Chemotherapy‐induced peripheral neuropathy is one of the most life‐affecting toxicities of chemotherapy associated with the use of taxanes [24], and it is associated with reduced quality of life in patients with cancer [25, 26]; therefore, reduced long‐term neuropathy in patients treated with PC‐W suggests improved quality of life in these patients.
Although analysis of patient demographics within each one of the treatment regimens (Table 1) suggests that older and more infirm patients received the PC‐W, OS and PFS among patients who were 75 or older at diagnosis also indicate a benefit for treatment with the PC‐W regimen. Better treatment response in PFS and OS of the PC‐W group may be attributed to the 4‐week delay in course completion of the PC‐W regimen (24 weeks instead of 18 weeks for PC‐3W), to the fourth week off‐treatment in each treatment cycle, which allowed the patients to recover, and to the increased completion rate (of 6 cycles) for frail subsets of the population. Thus, our carboplatin weekly regimen with weekly paclitaxel 1, 8, and 15 every 28 days may offer advantages in terms of tolerance in older patients while retaining or improving efficacy, perhaps by allowing more carboplatin to be given.
Our results showed that 3‐year survival rates were somewhat higher for women who were treated concurrently with PC and bevacizumab compared with those who were not, with higher survival rates for the PC‐W regimen at 3 years. The phase‐3 GOG‐0218 study did not find survival differences among patients who received concurrent bevacizumab and PC‐3W compared with PC‐3W alone. A survival benefit was observed in women with BRCA1/2 mutations and in those with non‐BRCA1/2 homologous recombination repair genes [27]. The low percentage of patients in our cohort treated concurrently with carboplatin with paclitaxel and PARP inhibitors prevent us from reaching any conclusion regarding any survival benefit.
Conclusion
Our data suggest that the weekly paclitaxel carboplatin regimen is as active and better tolerated than the standard PC‐3W regimen in a cohort of unselected patients with advanced EOC, TC, and PPC. It is possible that the weekly holiday on day 21 in the PC‐W regimen may ensure better completion rates (that may result in treatment delays for toxicity in PC‐3W). As prospective trials comparing the two regimens are lacking, the results of this analysis highlight the modified dose dense weekly regimen as a valid treatment option, especially for elderly patients and those with significant comorbidities.
Author Contributions
Conception/design: Tamar Safra
Provision of study material or patients: Tamar Safra, Barliz Waissengrin, Talya Levy, Ellie Leidner, Rotem Merose, Diana Matceyevsky, Dan Grisaru, Ido Laskov, Nadav Mishaan, Rotem Shayzaf
Collection and/or assembly of data: Tamar Safra, Barliz Waissengrin, Talya Levy, Ellie Leidner, Rotem Merose, Diana Matceyevsky, Dan Grisaru, Ido Laskov, Nadav Mishaan, Rotem Shayzaf
Data analysis and interpretation: Tamar Safra, Barliz Waissengrin, Ellie Leidner
Manuscript writing: Tamar Safra, Barliz Waissengrin, Ido Wolf
Final approval of manuscript: Tamar Safra, Barliz Waissengrin, Talya Levy, Ellie Leidner, Rotem Merose, Diana Matceyevsky, Dan Grisaru, Ido Laskov, Nadav Mishaan, Rotem Shayzaf, Ido Wolf
Disclosures
The authors indicated no financial relationships.
Disclosures of potential conflicts of interest may be found at the end of this article.
Editor's Note: See the related commentary, “Weekly Carboplatin and Paclitaxel for Ovarian Cancer: The ‘Finer Points’ ” by Franco Muggia, on page 1 of this issue.
No part of this article may be reproduced, stored, or transmitted in any form or for any means without the prior permission in writing from the copyright holder. For information on purchasing reprints contact Commercialreprints@wiley.com. For permission information contact permissions@wiley.com.
References
- 1. Jacobs IJ, Menon U, Ryan A et al. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A randomised controlled trial. Lancet 2016;387:945–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017;67:7–30. [DOI] [PubMed] [Google Scholar]
- 3. du Bois A, Lück HJ, Meier W et al. A randomized clinical trial of cisplatin/paclitaxel versus carboplatin/paclitaxel as first‐line treatment of ovarian cancer. J Natl Cancer Inst 2003;95:1320–1329. [DOI] [PubMed] [Google Scholar]
- 4. Ozols RF, Bundy BN, Greer BE et al. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol 2003;21:3194–3200. [DOI] [PubMed] [Google Scholar]
- 5. Thigpen T, duBois A, McAlpine J et al. First‐line therapy in ovarian cancer trials. Int J Gynecol Cancer 2011;21:756–762. [DOI] [PubMed] [Google Scholar]
- 6. Bookman MA, Brady MF, McGuire WP et al. Evaluation of new platinum‐based treatment regimens in advanced‐stage ovarian cancer: A Phase III Trial of the Gynecologic Cancer Intergroup. J Clin Oncol 2009;27:1419–1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. 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. [DOI] [PubMed] [Google Scholar]
- 8. Jordan MA, Toso RJ, Thrower D et al. Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations. Proc Natl Acad Sci USA 1993;90:9552–9556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Jordan MA, Wendell K, Gardiner S et al. Mitotic block induced in HeLa cells by low concentrations of paclitaxel (Taxol) results in abnormal mitotic exit and apoptotic cell death. Cancer Res 1996;56:816–825. [PubMed] [Google Scholar]
- 10. Norton L. Theoretical concepts and the emerging role of taxanes in adjuvant therapy. The Oncologist 2001;6(suppl):30–35. [DOI] [PubMed] [Google Scholar]
- 11. Torres K, Horwitz SB. Mechanisms of Taxol‐induced cell death are concentration dependent. Cancer Res 1998;58:3620–3626. [PubMed] [Google Scholar]
- 12. Saito K, Kikuchi Y, Fujii K et al. Effect of paclitaxel on vascular endothelial growth factor (VEGF) and interleukin (IL)‐8 in serum of patients with recurrent ovarian cancer: a comparison of weekly vs triweekly regimens. Target Oncol 2006;1:86–89. [Google Scholar]
- 13. Chan JK, Brady MF, Penson RT et al. Weekly vs. every‐3‐week paclitaxel and carboplatin for ovarian cancer. N Engl J Med 2016;374:738–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Katsumata N, Yasuda M, Isonishi S et al. Long‐term results of dose‐dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): A randomised, controlled, open‐label trial. Lancet Oncol 2013;14:1020–1026. [DOI] [PubMed] [Google Scholar]
- 15. Katsumata N, Yasuda M, Takahashi F et al. Dose‐dense paclitaxel once a week in combination with carboplatin every 3 weeks for advanced ovarian cancer: A phase 3, open‐label, randomised controlled trial. Lancet 2009;374:1331–1338. [DOI] [PubMed] [Google Scholar]
- 16. Pignata S, Scambia G, Katsaros D et al. Carboplatin plus paclitaxel once a week versus every 3 weeks in patients with advanced ovarian cancer (MITO‐7): A randomised, multicentre, open‐label, phase 3 trial. Lancet Oncol 2014;15:396–405. [DOI] [PubMed] [Google Scholar]
- 17. Safra T, Menczer J, Bernstein RM et al. Combined weekly carboplatin and paclitaxel as primary treatment of advanced epithelial ovarian carcinoma. Gynecol Oncol 2009;114:215–218. [DOI] [PubMed] [Google Scholar]
- 18. Clamp AR, McNeish I, Dean A et al. ICON8: A GCIG Phase III randomised trial evaluating weekly dose‐ dense chemotherapy integration in first‐line epithelial ovarian/ fallopian tube/ primary peritoneal carcinoma (EOC) treatment: Results of primary progression‐ free survival (PFS) Analysis. Ann Oncol 2017;28(suppl 5):v605–v649. [Google Scholar]
- 19. Bosó V, Herrero MJ, Santaballa A et al. SNPs and taxane toxicity in breast cancer patients. Pharmacogenomics 2014;15:1845–1858. [DOI] [PubMed] [Google Scholar]
- 20. Hertz DL. Germline pharmacogenetics of paclitaxel for cancer treatment. Pharmacogenomics 2013;14:1065–1084. [DOI] [PubMed] [Google Scholar]
- 21. Meyer UA. Pharmacogenetics and adverse drug reactions. Lancet 2000;356:1667–1671. [DOI] [PubMed] [Google Scholar]
- 22. Marchetti C, De Felice F, Musella A et al. Weekly versus three weeks chemotherapy for advanced ovarian cancer: a meta‐analysis. Oncotarget 2016;7:58709–58715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Vasey PA, Jayson GC, Gordon A et al. Phase III randomized trial of docetaxel‐carboplatin versus paclitaxel‐carboplatin as first‐line chemotherapy for ovarian carcinoma. J Natl Cancer Inst 2004;96:1682–1691. [DOI] [PubMed] [Google Scholar]
- 24. Kerckhove N, Collin A, Conde S et al. Long‐term effects, pathophysiological mechanisms, and risk factors of chemotherapy‐induced peripheral neuropathies: A comprehensive literature review. Front Pharmacol 2017;8:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bao T, Basal C, Seluzicki C et al. Long‐term chemotherapy‐induced peripheral neuropathy among breast cancer survivors: Prevalence, risk factors, and fall risk. Breast Cancer Res Treat 2016;159:327–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Jin L, Zhang Y, Yang W. Chemotherapy‐induced peripheral neuropathy among patients with ovarian cancer. Int J Gynaecol Obstet 2020;149:303–308. [DOI] [PubMed] [Google Scholar]
- 27. Tewari KS, Burger RA, Enserro D et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol 2019;37:2317–2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Abaid LN, Micha JP, Rettenmaier MA et al. A phase II study of modified dose‐dense paclitaxel and every 4‐week carboplatin for the treatment of advanced‐stage primary epithelial ovarian, fallopian tube, or peritoneal carcinoma. Cancer Chemother Pharmacol 2013;72:101–107. [DOI] [PubMed] [Google Scholar]
- 29. Ebata T, Yunokawa M, Bun S et al. Dose‐dense paclitaxel plus carboplatin as neoadjuvant chemotherapy for advanced ovarian, fallopian tube, or primary peritoneal carcinomas. Cancer Chemother Pharmacol 2016;78:1283–1288. [DOI] [PubMed] [Google Scholar]
- 30. van der Burg ME, Onstenk W, Boere IA et al. Long‐term results of a randomised phase III trial of weekly versus three‐weekly paclitaxel/platinum induction therapy followed by standard or extended three‐weekly paclitaxel/platinum in European patients with advanced epithelial ovarian cancer. Eur J Cancer 2014;50:2592–2601. [DOI] [PubMed] [Google Scholar]