Prognostic Impact of the Time Interval between Surgery and Postoperative Adjuvant Chemotherapy in Epithelial Carcinoma of the Ovary

JNS Cheng, WWL Yip, B Chan, FCS Wong

ORIGINAL ARTICLE
 
Prognostic Impact of the Time Interval between Surgery and Postoperative Adjuvant Chemotherapy in Epithelial Carcinoma of the Ovary
 
JNS Cheng, WWL Yip, B Chan, FCS Wong
Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong
 
Correspondence: Dr JNS Cheng, Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong. Email: cns325@ha.org.hk
 
Submitted: 20 Jul 2020; Accepted: 16 Nov 2020.
 
Contributors: All authors designed the study, acquired and analysed the data. JNSC and BC drafted the manuscript. All authors critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: The research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Ethics Approval: The study was approved by the New Territories West Cluster Research Ethics Committee (Ref NTWC/REC/20084). The need for patients to provide written consent was waived for this retrospective study.
 
 
 
 
 
Abstract
 
Introduction
 
Multiple studies have evaluated the prognostic impact of the time interval (TI) between initial surgery and adjuvant chemotherapy for epithelial ovarian cancer with different time intervals and inconclusive results. The aim of the present study was to evaluate the prognostic impact of a longer interval of 42 days.
 
Methods
 
In a retrospective single-centre analysis, data were collected for all patients with epithelial ovarian cancer treated between 2007 and 2014. We divided patients by TI: ≤42 days and >42 days. The disease-free survival and overall survival (OS) between the two groups were compared. A Cox regression model was used to evaluate different prognostic factors. A p value <0.05 was considered statistically significant.
 
Results
 
The median follow-up time was 73 months. Among those with postoperative residual disease (n = 30), TI of >42 days was associated with significantly worse OS (hazard ratio = 3.37, 95% confidence interval = 1.23-9.25, p = 0.02). In cases with residual disease after surgery, the Cox proportional model showed the presence of ascites (p = 0.03) and postoperative CA125 level (p = 0.03) were independent prognostic factors for DFS. TI >42 days (p = 0.03) was an independent negative prognostic factor for OS along with grading (p = 0.05) and presence of ascites (p < 0.01).
 
Conclusion
 
Our study showed that patients with residual disease after initial surgery had inferior OS when TI was >42 days. Adjuvant chemotherapy in these patients should be started ≤42 days after surgery.
 
 
Key Words: Carcinoma, ovarian epithelial; Chemotherapy, adjuvant; Prognosis
 
 
中文摘要
 
上皮性卵巢癌的手術和術後輔助化療的時間間隔對預後的影響
 
鄭雁心、葉穎鈴、陳柏霖、黃志成
 
引言
多項研究已評估上皮性卵巢癌的手術與術後輔助化療的時間間隔對預後的影響,但使用的時 間間隔不同且結果尚無定論。本研究旨在評估較長的42天間隔的預後影響。
 
方法
在一項回顧性單中心分析中收集2007年至2014年間治療的所有上皮性卵巢癌患者的數據。我 們將患者分成兩組,包括手術與術後輔助化療的時間間隔(1)42天或以下以及(2)超過42天,並 比較兩組間的無病存活期和總存活期。使用Cox迴歸模型評估不同的預後因素。p值<0.05被認為具統 計學意義。
 
結果
中位隨訪時間為73個月。在具有術後殘留疾病(n = 30)的患者中,兩種治療的時間間隔超 過42天與整體存活率顯著下降相關(危險比 = 3.37,95%置信區間 = 1.23-9.25,p = 0.02)。對於術 後殘留疾病的患者,Cox迴歸模型顯示出現腹水(p = 0.03)和術後CA125水平(p = 0.03)是無病存 活期的獨立預後因素。兩種治療的時間間隔超過42天(p = 0.03)、疾病等級(p = 0.05)和出現腹水 (p < 0.01)是總存活的獨立陰性預後因素。
 
結論
我們的研究表明,當初始手術與輔助化療的時間間隔超過42天時,初次手術後有殘留疾病的 患者的總存活期較差。這些患者的輔助化療應在手術後42天或之前開始。
 
 
 
INTRODUCTION
 
Epithelial ovarian cancer is one of the most lethal gynaecological cancers. According to the data released by the Hong Kong Cancer Registry in 2019, ovarian cancer was the 6th most common cancer and the 7th leading cause of cancer-related deaths among women in 2017.[1] The standard treatment for epithelial ovarian cancer remains surgery with optimal debulking followed by adjuvant chemotherapy.
 
Preclinical models had shown surgical removal of any one of several tumours might accelerate the growth of the residual tumours.[2] [3] Studies of cancers including primary breast cancer, colorectal cancer, gastric cancer and pancreatic cancer have reported significantly worse outcomes with a delay in the initiation of systemic therapy after surgery.[4] [5] [6] [7] [8]
 
Focusing on epithelial ovarian cancer, there is always a struggle between earlier initiation of systemic therapy after surgery and allowing more time for postoperative recovery. Most patients with residual disease should benefit from earlier chemotherapy after debulking surgery. However, debulking surgery is a major operation that carries significant morbidity. Patients usually require significant time for wound healing and nutritional recovery. Surgical series reported a range of inpatient stays of 4 to 14 days after primary surgical staging for patients with ovarian cancer.[9] [10] Data from a study focusing on primary surgery for ovarian cancer versus neoadjuvant chemotherapy also reported a median time to initiation of chemotherapy after primary surgery in advanced ovarian cancer of 32 days, with a range of 5 to 82 days.[11]
 
Few studies have used a cut-off of 42 days. The study by Paulsen et al[12] mainly focused on locally advanced disease and it showed a significant negative impact on overall survival (OS) if adjuvant chemotherapy was delayed ≥6 weeks. Another study by Wright et al[13] focused on patients aged >65 years with locally advanced disease also found a significant negative impact on OS if adjuvant chemotherapy was delayed ≥6 weeks. Focusing on early-stage disease, only one study investigated the prognostic impact of administration of chemotherapy <2, 2 to 4, or >4 weeks after surgery and found no significant impact on disease-free survival (DFS) or OS.[14] Thus, it was uncertain if a delay of >42 days after surgery would have any prognostic impact on ovarian cancer.
 
The aim of this study was to evaluate the prognostic impact of time interval (TI) in epithelial patients with ovarian cancer in a single centre with a standardised protocol on use of adjuvant chemotherapy and a detailed record of residual disease after surgery.
 
METHODS
 
Study Design
 
This retrospective study was conducted on the data of all patients who underwent primary surgical staging with debulking followed by at least one dose of adjuvant chemotherapy at the Department of Clinical Oncology, Tuen Mun Hospital between 1 January 2007 and 31 December 2014.
 
Inclusion Criteria
 
Cases of epithelial ovarian, fallopian tube, or primary peritoneal carcinoma aged >18 years, undergoing surgery with the intention of maximum debulking and followed by at least one dose of adjuvant chemotherapy, were included in this study. Cases receiving neoadjuvant chemotherapy, with incomplete information on chemotherapy administration or starting chemotherapy >90 days after surgery were excluded.
 
Case Selection
 
The Clinical Data Analysis and Reporting System of the Hospital Authority was used to identify all cases of epithelial ovarian, fallopian tube or primary peritoneal cancer first seen at the Department of Clinical Oncology between 1 January 2007 and 31 December 2014 in Tuen Mun Hospital. Only those cases fulfilling the inclusion criteria were included.
 
EVALUATION OF OUTCOME MEASURES
 
All patients were regularly followed up by an oncologist after the completion of adjuvant chemotherapy. The follow-up procedure in our department was every 3 to 4 months for the first to second year, every 4 to 6 months for the third to fourth year, every 9 to 12 months for the fifth year, and yearly afterwards. Clinical assessment with history taking and physical examination would be done on every follow-up. Serum CA125 test was advised to be included at every visit. Imaging studies were performed when there was clinical suspicion of disease recurrence.
 
Patients’ demographic data, including date of birth and date of death were collected from the patient medical records.
 
The tumour stage and histological diagnosis of each patient were determined according to the International Federation of Gynaecology and Obstetrics criteria and the histologic typing system of the World Health Organization.[15] [16] Tumours were graded as either well differentiated (Grade 1), moderately differentiated (Grade 2), or poorly differentiated (Grade 3). Surgical reports, inpatient discharge summaries, histology reports, chemotherapy charts, and consultation notes of individual cases were collected and reviewed.
 
The consultation notes of every follow-up were reviewed. The progress of each case from the date of surgery, including any recurrences and their dates, and the date of death or latest follow-up date were recorded.
 
Outcome Measurements
 
The primary endpoint of this study was OS, which was defined as the TI from surgery to a patient’s death. The secondary endpoint was DFS, which was defined as the TI from surgery to the date when the patient was diagnosed with recurrence or the date of death, whichever occurred first. The cut-off date for follow-up was 17 July 2020. Imaging or histology was required to diagnose recurrence; a rise in CA125 alone was considered insufficient for diagnosis.
 
Sample Size and Rationale
 
Sample size calculation was estimated using the open source software Power and Sample Size Program version 3.0 for survival analysis with log rank tests. On review of previous evidence, reported effect size ranged from 1.02 to 3.44, assuming two tails and aiming to detect an effect size of 1.7. Assuming achievement of a significance level of 0.05, median survival for the patients with TI >42 days of approximately 50 months, and power = 0.7, the required sample size would be 134. If the power were to be increased to 0.8, the sample size required would be 170. However, as this study is a single-centre retrospective study, including more cases would involve gathering the data of cases who were first seen by oncology before 2007; this was not possible as records before that year had been destroyed.
 
Statistics
 
The TIs from surgery to chemotherapy of all included cases were calculated. Patients were divided into two groups according to the TI of ≤42 days or >42 days. The 42-day cut-off point was chosen because most patients who received adjuvant chemotherapy >42 days after initial surgery were excluded from most clinical chemotherapy trial protocols.[11] [17] [18] Therefore, those patients were among the least studied population.
 
SPSS (Window version 26.0; IBM Corp, Armonk [NY], United States) was used for the statistical analyses. Descriptive statistics included frequency and percentage for categorical variables. Clinical data were compared by Chi squared test or Fisher’s exact test for categorical variables.
 
To visualise the crude DFS and OS for the two groups with different TIs, Kaplan–Meier curves were constructed. A log rank estimate was used to compare the number of recurrences and deaths between the two TI groups. The same analysis was also performed after dividing patients into those with or without residual disease. Cox regression analysis was further performed to quantify the effect.
 
The Cox proportional hazards analysis was used to evaluate the effect of different prognostic factors. A p value of <0.05 was considered statistically significant and all p values reported were two sided. Prognostic factors included in the analysis were patient’s age at operation, performance status, stage, histology, tumour grading, size of residual, presence or absence of ascites, number of chemotherapy cycles given, postoperative CA125 value, and the TI between surgery and adjuvant chemotherapy. The reason for choosing the above factors was based on reviewing the significant prognostic factors found in previous studies (Table 1 [12] [13] [14],[19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36]).
 
Table 1. Ovarian cancer studies on the effect of interval from surgery to initiation of adjuvant therapy.
 
Ethical Considerations
 
This study was approved by New Territories West Cluster Research Ethics Committee (EC Ref. No.: NTWC/REC/2084). The need for informed consent was waived.
 
RESULTS
 
Characteristics of the Study Population
 
A total of 133 cases were included in the study. The baseline characteristics of the study subjects are summarised in Table 2. The median follow-up duration was 72.6 (1.8-155.9) months. The median DFS and OS were not reached. The most common disease stage in the current population was stage IC (33.8%) and stage IIIC (24.8%). The median TI from surgery to adjuvant chemotherapy was 34 days (interquartile range, 27-42 days) [Figure 1]. In total, 98 patients had TI of ≤42 days and 35 patients had TI of >42 days.
 
Table 2. Characteristics of patients with regard to different intervals from surgery to start of chemotherapy.
 
Figure 1. Distribution of time interval between initial surgery and chemotherapy initiation.
 
In all, 121 (91%) patients achieved optimal debulking while only 12 (9%) patients had suboptimal debulking. For those with optimal debulking, 18 had residual disease of ≤1 cm. For those with suboptimal debulking, residual disease ranged from 1.5 to 10 cm.
 
Patient characteristics across the two groups were similar. Reasons for delaying initiation of chemotherapy to >42 days after surgery are summarised in Table 3. In all, 29% of the delays was due to chemotherapy clinic waiting time while 26% were due to oncology new case waiting time. There was also a delay in 17% of cases due to the need to start hepatitis B prophylaxis before chemotherapy, as hepatitis B carrier status is prevalent in our area.
 
Table 3. Reasons for delay of >42 days after surgery for chemotherapy initiation (n = 35).
 
Disease-Free Survival
 
There was no significant effect on DFS when comparing patients with TI of ≤42 days versus >42 days (hazard ratio [HR] = 0.72, 95% confidence [CI] = 0.39-1.32, p = 0.29) [Figure 2]. The 5-year DFS rate for patients with TI of ≤42 days was 51.0% and that for patients with TI of >42 days was 62.2%.
 
Figure 2. Disease-free survival for chemotherapy initiated ≤42 days or >42 days after surgery.
 
Overall Survival
 
There was no significant effect on OS when comparing TIs of ≤42 days versus >42 days in all cases (HR = 0.82, 95% CI = 0.42-1.60, p = 0.56) [Figure 3]. The 5-year OS rates for patients with TI of ≤42 days was 64.7% and that for patients with TI of >42 days was 71.4%.
 
Figure 3. Overall survival for chemotherapy initiated ≤42 days or >42 days after surgery.
 
Subgroup Analysis with Presence or Absence of Residual Disease
 
For patients with residual disease (n = 30), their OS was statistically significantly worse for patients with TI of >42 days (HR = 3.37, 95% CI = 1.23-9.25, p = 0.02) [Figure 4]. The median OS for patients with TI of ≤42 days was 58 months (95% CI = 32.5-83.5) and that for patients with TI >42 days was 13.5 months (95% CI = 12.9-14.1). The 5-year OS rate for patients with TI of ≤42 days was 65.2% and that for patients with TI of >42 days was 14.3%.
 
Figure 4. Overall survival in patients with residual disease for chemotherapy initiated ≤42 days or >42 days after surgery.
 
There was no significant difference in DFS when comparing patients with TI of ≤42 days versus >42 days among patients with residual disease (HR = 1.77, 95% CI = 0.71-4.48, p = 0.22).
 
For patients with no residual disease after surgery, there was no DFS or OS difference when comparing patients with TI of ≤42 days and >42 days (HR = 2.59, 95% CI = 0.26-1.35, p = 0.21 and HR = 0.50, 95% CI = 0.19-1.30, p = 0.16, respectively).
 
Prognostic Factors
 
In cases with residual disease after surgery, the Cox proportional model showed the presence of ascites (p = 0.03) and postoperative CA125 level (p = 0.03) were independent prognostic factors for DFS. TI >42 days (p = 0.03) was an independent negative prognostic factor for OS (Table 4) along with grading (p = 0.05) and presence of ascites (p < 0.01).
 
Table 4. Cox proportional hazards model for disease-free survival and overall survival among those who had residual disease after surgery.
 
DISCUSSION
 
The current study did not find an effect of TI on DFS or OS in patients with epithelial ovarian cancer without residual disease. However, for those with residual disease, delaying chemotherapy to >42 days after surgery was significantly associated with shorter OS.
 
It is well known that adjuvant chemotherapy is associated with improved survival in epithelial ovarian cancer.[37] However, patients do recur after surgery, especially those with residual disease. In a study by Polterauer et al,[38] 3-year OS rates were 72.4%, 65.8%, and 45.2% for patients with no residual disease, minimal residual disease, and gross residual disease (>1 cm), respectively.
 
Our study showed there was a statistically significant shortening of OS in patients with residual disease and a TI of >42 days. We had 82 (61.7%) cases with stage I/II disease and 51 (38.3%) cases with stage III/IV disease. Our findings concur with a study by Seagle et al[27] involving cases in stage I to IV that revealed a negative prognostic effect of delaying chemotherapy ≥36 days after surgery. That study consisted mainly of stage III or IV patients (55.7% and 16.1%, respectively). Tewari et al[25] reported in stage IV disease patients with microscopic residual disease that a >25 days interval from surgery to adjuvant chemotherapy was associated with a worse OS. Lee et al[29] suggested that patients with residual disease size ranging from 1 to 9 mm after surgery were associated with significantly worsened OS when there was delay in initiating chemotherapy of ≥10 days after surgery. Although their study was limited to patients with serous ovarian cancer, Hofstetter et al[24] suggested that there would be a worsened OS with HRs of 2.24 for patients with gross residual disease after primary surgery and chemotherapy initiated after a TI >28 days. Our study also had similar findings.
 
However, one of the limitations in interpreting our data is that our study sample size is small, with only 30 cases with residual disease after operation and only seven initiating chemotherapy >42 days after surgery. This finding can serve as hypothesis generation only and further studies should be conducted to confirm this hypothesis (Tables 1 [12] [13] [14],[19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] and 5 [39] [40]).
 
Table 5. Meta-analyses of the effect of interval from surgery to initiation of adjuvant therapy.
 
One of the greatest limitations in the current study was sampling bias with confounding by indication. As with many other retrospective studies, clinicians could decide at their discretion on the timing of initiation of chemotherapy. In our study, interestingly, we noted a trend towards worse survival in those who initiated chemotherapy earlier, although it was statistically insignificant. It might be postulated that clinicians opted to start chemotherapy early in patients who were deemed to be at high risk of recurrence.
 
Another limitation of our study would be the follow-up procedure. In our patients, the interval of surveillance computed tomography (CT) or checking of tumour marker CA125 was decided by the treating physician. The lack of standardisation might have an impact on the DFS. OS would be a more robust endpoint that is less sensitive to the impact of diagnosing recurrence earlier with more frequent imaging or blood tests. Indeed, it had been shown that earlier initiation of palliative chemotherapy based on elevated CA125 alone did not improve OS.[41] [42]
 
The reasons for delaying initiation of chemotherapy in this study were mainly the prolonged waiting time for oncology new case appointments or chemotherapy clinic appointments. Further arrangements of fast-track service for this group of patients to improve their potential OS should be considered. The prevalence of hepatitis B carriage in the Asian population also warrants earlier detection of hepatitis B status to allow earlier initiation of hepatitis B prophylaxis medications to avoid delays in chemotherapy administration.
 
CONCLUSION
 
Our study showed that patients with residual disease after initial surgery may have inferior OS when the adjuvant chemotherapy is initiated >42 days after surgery. Further studies should be conducted to see if this finding can be reproduced. Adjuvant chemotherapy in these patients should be started ≤42 days after surgery.
 
REFERENCES
 
1. Hospital Authority, Hong Kong SAR Government. Ovarian cancer in 2017. Available from: https://www3.ha.org.hk/cancereg/pdf/factsheet/2017/ovary_2017.pdf. Accessed 5 Apr 2020.
 
2. Gunduz N, Fisher B, Saffer EA. Effect of surgical removal on the growth and kinetics of residual tumor. Cancer Res. 1979;39:3861-5.
 
3. Fisher B, Gunduz N, Coyle J, Rudock C, Saffer E. Presence of a growth-stimulating factor in serum following primary tumor removal in mice. Cancer Res. 1989;49:1996-2001.
 
4. Lohrisch C, Paltiel C, Gelmon K, Speers C, Taylor S, Barnett J, et al. Impact on survival of time from definitive surgery to initiation of adjuvant chemotherapy for early-stage breast cancer. J Clin Oncol. 2006;24:4888-94. Crossref
 
5. Kupstas AR, Hoskin TL, Day CN, Habermann EB, Boughey JC. Effect of surgery type on time to adjuvant chemotherapy and impact of delay on breast cancer survival: a National Cancer Database analysis. Ann Surg Oncol. 2019;26:3240-9. Crossref
 
6. Cai L, Tong Y, Zhu X, Shen K, Zhu J, Chen X. Prolonged time to adjuvant chemotherapy initiation was associated with worse disease outcome in triple negative breast cancer patients. Sci Rep. 2020;10:7029. Crossref
 
7. Petrelli F, Zaniboni A, Ghidini A, Ghidini M, Turati L, Pizzo C, et al. Timing of adjuvant chemotherapy and survival in colorectal, gastric, and pancreatic cancer. A systematic review and meta-analysis. Cancers (Basel). 2019;11:550. Crossref
 
8. Noh GT, Han J, Cho MS, Hur H, Lee KY, Kim NK, et al. The impact of early adjuvant chemotherapy in rectal cancer. PLoS One. 2020;15:e0228060. Crossref
 
9. Ghezzi F, Cromi A, Uccella S, Bergamini V, Tomera S, Franchi M, et al. Laparoscopy versus laparotomy for the surgical management of apparent early stage ovarian cancer. Gynecol Oncol. 2007;105:409-13. Crossref
 
10. Nezhat FR, DeNoble SM, Liu CS, Cho JE, Brown DN, Chuang L, et al. The safety and efficacy of laparoscopic surgical staging and debulking of apparent advanced stage ovarian, fallopian tube, and primary peritoneal cancers. JSLS. 2010;14:155-68. Crossref
 
11. Kehoe S, Hook J, Nankivell M, Jayson GC, Kitchener H, Lopes T, et al. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): an open-label, randomised, controlled, non-inferiority trial. Lancet. 2015;386:249-57. Crossref
 
12. Paulsen T, Kaern J, Kjaerheim K, Haldorsen T, Tropé C. Influence of interval between primary surgery and chemotherapy on short-term survival of patients with advanced ovarian, tubal or peritoneal cancer. Gynecol Oncol. 2006;102:447-52. Crossref
 
13. Wright JD, Doan T, McBride R, Jacobson J, Hershman D. Variability in chemotherapy delivery for elderly women with advanced stage ovarian cancer and its impact on survival. Br J Cancer. 2008;98:1197-203. Crossref
 
14. Chan JK, Java JJ, Fuh K, Monk BJ, Kapp DS, Herzog T, et al. The association between timing of initiation of adjuvant therapy and the survival of early stage ovarian cancer patients — An analysis of NRG Oncology/Gynecologic Oncology Group trials. Gynecol Oncol. 2016;143:490-5. Crossref
 
15. Shepherd JH. Revised FIGO staging for gynaecological cancer. Br J Obstet Gynaecol. 1989;96:889-92. Crossref
 
16. Scully R, Sobin LH, editors. Histological typing of ovarian tumours. Heidelberg: Springer; 1999. Crossref
 
17. European Platform of Cancer Research. EORTC protocol 55981: a randomized trial of Paclitaxel/Epirubicin/Carboplatin Combination (TEC) versus Paclitaxel/Carboplatin(TC) in the treatment of women with Advanced Ovarian Cancer. 1999. Available from: https://www.eortc.org/research_field/clinical-detail/55981/. Accessed 11 Apr 2020.
 
18. Vergote I, Tropé CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 2010;363:943-53. Crossref
 
19. Omura GA, Bundy BN, Berek JS, Curry S, Delgado G, Mortel R. Randomized trial of cyclophosphamide plus cisplatin with or without doxorubicin in ovarian carcinoma: a Gynecologic Oncology Group Study. J Clin Oncol. 1989;7:457-65. Crossref
 
20. Warwick J, Kehoe S, Earl H, Luesley D, Redman C, Chan KK. Long-term follow-up of patients with advanced ovarian cancer treated in randomised clinical trials. Br J Cancer. 1995;72:1513-7. Crossref
 
21. Sorbe B. Prognostic importance of the time interval from surgery to chemotherapy in treatment of ovarian carcinoma. Int J Gynecol Cancer. 2004;14:788-93. Crossref
 
22. Wright JD, Herzog TJ, Neugut AI, Burke WM, Lu YS, Lewin SN, et al. Effect of radical cytoreductive surgery on omission and delay of chemotherapy for advanced-stage ovarian cancer. Obstet Gynecol. 2012;120:871-81. Crossref
 
23. Mahner S, Eulenburg C, Staehle A, Wegscheider K, Reuss A, Pujade-Lauraine E, et al. Prognostic impact of the time interval between surgery and chemotherapy in advanced ovarian cancer: analysis of prospective randomised phase III trials. Eur J Cancer. 2013;49:142-9. Crossref
 
24. Hofstetter G, Concin N, Braicu I, Chekerov R, Sehouli J, Cadron I, et al. The time interval from surgery to start of chemotherapy significantly impacts prognosis in patients with advanced serous ovarian carcinoma — analysis of patient data in the prospective OVCAD study. Gynecol Oncol. 2013;131:15-20. Crossref
 
25. Tewari KS, Java JJ, Eskander RN, Monk BJ, Burger RA. Early initiation of chemotherapy following complete resection of advanced ovarian cancer associated with improved survival: NRG Oncology/Gynecologic Oncology Group study. Ann Oncol. 2016;27:114-21. Crossref
 
26. Heo EJ, Paik ES, Choi HJ, Kim WY, Lee YY, Choi CH, et al. Impact of interval from definitive surgery to initiation of adjuvant chemotherapy (ISC) on survival in advanced epithelial ovarian cancer. Gynecologic Oncology. 2016 Jun 1;141:116-7. Crossref
 
27. Seagle BL, Butler SK, Strohl AE, Nieves-Neira W, Shahabi S. Chemotherapy delay after primary debulking surgery for ovarian cancer. Gynecol Oncol. 2017;144:260-5. Crossref
 
28. Timmermans M, van der Aa MA, Lalisang RI, Witteveen PO, Van de Vijver KK, Kruitwagen RF, et al. Interval between debulking surgery and adjuvant chemotherapy is associated with overall survival in patients with advanced ovarian cancer. Gynecol Oncol. 2018;150:446-50. Crossref
 
29. Lee YY, Lee JW, Lu L, Xu W, Kollara A, Brown T, et al. Impact of interval from primary cytoreductive surgery to initiation of adjuvant chemotherapy in advanced epithelial ovarian cancer. Int J Gynecol Obstet. 2018;143:325-32. Crossref
 
30. Flynn PM, Paul J, Cruickshank DJ, Scottish Gynaecological Cancer Trials Group. Does the interval from primary surgery to chemotherapy influence progression-free survival in ovarian cancer? Gynecol Oncol. 2002;86:354-7. Crossref
 
31. Gadducci A, Sartori E, Landoni F, Zola P, Maggino T, Maggioni A, et al. Relationship between time interval from primary surgery to the start of taxane-plus platinum-based chemotherapy and clinical outcome of patients with advanced epithelial ovarian cancer: results of a multicenter retrospective Italian study. J Clin Oncol. 2005;23:751-8. Crossref
 
32. Rosa DD, Clamp A, Mullamitha S, Ton NC, Lau S, Byrd L, et al. The interval from surgery to chemotherapy in the treatment of advanced epithelial ovarian carcinoma. Eur J Surg Oncol. 2006;32:588-91. Crossref
 
33. Aletti GD, Long HJ, Podratz KC, Cliby WA. Is time to chemotherapy a determinant of prognosis in advanced-stage ovarian cancer? Gynecol Oncol. 2007;104:212-6. Crossref
 
34. Lydiksen L, Jensen-Fangel S, Blaakaer J. Is it possible to define an optimal time for chemotherapy after surgery for ovarian cancer? Gynecol Oncol. 2014;133:454-9. Crossref
 
35. Feng Z, Wen H, Bi R, Yang W, Wu X. Prognostic impact of the time interval from primary surgery to intravenous chemotherapy in high grade serous ovarian cancer. Gynecol Oncol. 2016;141:466-70. Crossref
 
36. Anuradha S, Donovan PJ, Webb PM, Brand AH, Goh J, Friedlander M, et al. Variations in adjuvant chemotherapy and survival in women with epithelial ovarian cancer — a population-based study. Acta Oncol. 2016;55:226-33. Crossref
 
37. Kyrgiou M, Salanti G, Pavlidis N, Paraskevaidis E, Ioannidis JP. Survival benefits with diverse chemotherapy regimens for ovarian cancer: meta-analysis of multiple treatments. J Natl Cancer Inst. 2006;98:1655-63. Crossref
 
38. Polterauer S, Vergote I, Concin N, Braicu I, Chekerov R, Mahner S, et al. Prognostic value of residual tumor size in patients with epithelial ovarian cancer FIGO stages IIA–IV: analysis of the OVCAD data. Int J Gynecol Cancer. 2012;22:380-5. Crossref
 
39. Liu Y, Zhang T, Wu Q, Jiao Y, Gong T, Ma X, et al. Relationship between initiation time of adjuvant chemotherapy and survival in ovarian cancer patients: a dose-response meta-analysis of cohort studies. Sci Rep. 2017;7:9461. Crossref
 
40. Usón PL Junior, Bugano DD, França MS, Antunes YP, Taranto P, Kaliks RA, et al. Does time-to-chemotherapy impact the outcomes of resected ovarian cancer? Meta-analysis of randomized and observational data. Int J Gynecol Cancer. 2017;27:274-80. Crossref
 
41. Rustin GJ, van der Burg ME, Griffin CL, Guthrie D, Lamont A, Jayson GC, et al. Early versus delayed treatment of relapsed ovarian cancer (MRC OV05/EORTC 55955): a randomised trial. Lancet. 2010;376:1155-63. Crossref
 
42. Rustin GJ, van der Burg ME. on behalf of MRC and EORTC collaborators. A randomized trial in ovarian cancer (OC) of early treatment of relapse based on CA125 level alone versus delayed treatment based on conventional clinical indicators (MRC OV05/EORTC 55955 trials). J Clin Oncol. 2009;27(Suppl 18):1. Crossref
 
 
 
AttachmentSize
v24n1_Prognostic.pdf600.74 KB