The surgical, histopathological characteristics, and survival outcome of ovarian clear cell carcinoma: a retrospective case series sharing the experience of a tertiary cancer centre
Original Article

The surgical, histopathological characteristics, and survival outcome of ovarian clear cell carcinoma: a retrospective case series sharing the experience of a tertiary cancer centre

Sally El Tawab1,2 ORCID logo, Sabina Nistor1, Rene Roux3, Sanjiv Manek4, Kezia Gaitskell4, Ahmed Ashour Ahmed1,5,6, Sean Kehoe1,6, Hooman Soleymani majd1,6 ORCID logo

1Oxford Gynaecological Cancer Centre, Churchill Hospital, Oxford University Hospitals Foundation Trust, Headington, UK; 2Elshatby Maternity University Hospital, Gynaecology Oncology Center, Faculty of Medicine, Alexandria University, Alexandria, Egypt; 3Department of Medical Oncology, Churchill Hospital, Oxford University Hospitals Foundation Trust, Headington, UK; 4Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals Foundation Trust, Headington, UK; 5Ovarian Cancer Cell Laboratory, Medical Research Council MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; 6Nuffield Department of Women’s & Reproductive Health, Medical Science Division, Oxford University, Oxford, UK

Contributions: (I) Conception and design: S El Tawab, H Soleymani majd; (II) Administrative support: H Soleymani majd; (III) Provision of study materials or patient: H Soleymani majd; (IV) Collection and assembly of data: S El Tawab, S Manek, K Gaitskell; (V) Data analysis and interpretation: S El Tawab, S Nistor, H Soleymani majd; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Hooman Soleymani majd, MD, MRCOG. Oxford Gynaecological Cancer Centre, Churchill Hospital, Oxford University Hospitals Foundation Trust, Old Road, Headington OX3 7LE, UK; Nuffield Department of Women’s & Reproductive Health, Medical Science Division, Oxford University, Headley Way Road, Headington OX3 9DU, UK. Email: hooman.soleymanimajd@msd.ox.ac.uk.

Background: Ovarian clear cell carcinoma (OCCC) is a rare and distinct subtype of epithelial ovarian cancer (EOC). It is unique in several biological aspects. This study analyzes the clinicopathological features and survival outcome of patients with OCCC, aiming to identify factors affecting recurrence, progression-free survival (PFS) and overall survival (OS).

Methods: A retrospective study included 49 women with OCCC between January 2009 and December 2021 at Oxford Cancer Center. All demographic and pathological characteristics, pre-operative biomarkers, surgical procedure, complications, hospital stay, chemotherapy regimen, and disease status on follow-up, were collected from electronic medical records.

Results: No residual disease (R0) was achieved in 39 out of 49 women who underwent cytoreductive surgery. The follow-up time had a mean of 8.75 years. The 3-year OS was 73.4%, and the 3-year PFS was 81.3% [95% confidence interval (CI): 84.63–118.93]. Women with stage 1 disease had the best outcome. There was a marked difference (P<0.001) in OS in the presence of residual disease. No residual disease conferred a 3-year OS of 88.6% (95% CI: 108.6–141.8), compared to only 12.5% in the presence of residual disease (95% CI: 4.48–32.11). In multivariant analysis, the International Federation of Gynecology and Obstetrics (FIGO) stage was the only independent prognostic indicator of OS with (P<0.05), including carbohydrate antigen (CA) 125, hemoglobin, albumin, associated endometriosis, ascites, residual disease and FIGO staging.

Conclusions: Surgery to achieve no residual disease is necessary to improve the prognosis in advanced OCCC. The true challenge is to predict which patients with early-stage disease at higher risk of recurrence and would most benefit from adjuvant treatments.

Keywords: Ovarian clear cell carcinoma (OCCC); residual disease; recurrence; survival


Submitted Jan 12, 2024. Accepted for publication Jul 23, 2024. Published online Sep 11, 2024.

doi: 10.21037/tcr-24-83


Highlight box

Key findings

• The 3-year overall survival (OS) was 73.4%, and the 3-year progression-free survival was 81.3%. Women with stage 1 disease had the best outcome. In comparing OS in respect to absence or presence of residual disease, the 3-year OS was 88.6% [95% confidence interval (CI): 108.6–141.8], compared to 12.5% (95% CI: 4.48–32.11) respectively (P<0.001). In multivariant analysis, the International Federation of Gynecology and Obstetrics (FIGO) stage was the only independent prognostic indicator of OS with (P<0.05), including carbohydrate antigen 125, hemoglobin, albumin, associated endometriosis, ascites, residual disease and FIGO staging.

What is known and what is new?

• The standard therapeutic treatment for ovarian clear cell carcinoma (OCCC) according to the National Comprehensive Cancer Network guidelines (version 1.2020) is optimal cytoreduction combined with systemic chemotherapy. Patients with OCCC tend to be less sensitive to conventional platinum-based chemotherapy, where only 11–27% of patients with OCCC respond to platinum-based chemotherapy, resulting in poorer outcomes.

• This paper confers a long-term follow-up of over 8 years and reinforces the importance of radical cytoreductive surgery with the intention to achieve no residual disease R0 in rare subtypes of epithelial ovarian cancers which are notoriously not chemo-sensitive.

What is the implication, and what should change now?

• Surgery to achieve no residual disease is necessary to improve the prognosis in advanced OCCC. The true challenge is to predict which patients with early-stage disease are at higher risk of recurrence and would most benefit from adjuvant treatments.


Introduction

Ovarian clear cell carcinoma (OCCC) is a rare and distinct subtype of epithelial ovarian cancer (EOC). It is unique in several biological aspects, starting with its significant prevalence variation according to geographic distribution, where it is rare accounting for only 5–10% of all EOC in the United States and Western countries. It has a higher prevalence in Asian countries, especially Japan, where it reaches 25–30%, but the cause of this variation is unclear (1). While OCCC is type 1 ovarian cancer, it does not fit typically for this subtype as it is considered a high grade at presentation, regardless of its stage (2). It behaves aggressively, especially in advanced stages, unlike other type 1 tumors. OCCC is associated with endometriosis, suggesting that retrograde menstruation is the origin of these tumors (3). The ovary’s atypical endometriosis and atypical adenofibroma have been considered precancerous lesions (4,5). Another distinctive feature is the association between thromboembolism and a poor prognosis in OCCC (6,7), but the biological mechanism of hypercoagulation in OCCC remains unclear (8). Carbohydrate antigen (CA) 125, the standard tumor marker for high-grade serous carcinoma, is elevated only in about 57% of OCCC (9).

OCCC characteristically contains clear or hobnail cells with eccentric, rounded, bulbous nuclei, multiple complex papillae, densely hyaline basement membrane material, and hyaline bodies. Compared with other types of EOC, the frequency of mitoses is lower [usually <5/10 high power field (HPF)] (10). No single immunohistochemistry marker has been reported to be beneficial in distinguishing between high-grade serous and clear-cell ovarian carcinoma. A four-marker immunohistochemical panel [WT1/p53/napsin A/progesterone receptor (PR)] can distinguish EOC subtypes with high accuracy, and additional immunohistochemical markers can be used if needed. WT1 is the most critical marker diffusely expressed in almost all high-grade and low-grade serous ovarian cancers and is virtually absent in almost all OCCCs and mucinous ovarian cancers (11-14). Fadare et al. recommended a panel of immuno-histochemical markers, including Napsin-A, hepatocyte nuclear factor-1 beta (HNF-1β), and alpha-methylacyl-CoA racemase (AMACR), to obtain the highest sensitivity and specificity when OCCC is a diagnostic consideration (15).

Different molecular pathways and genetic alterations have been identified in OCCC, including the most common mutations of AT-rich interaction domain 1A (ARID1A) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3) catalytic subunit alpha (PIK3CA). OCCC shows high frequency of PIK3CA mutation (40%), leading to higher activity of the phosphoinositide 3 kinase (PI3K)/Akt/mammalian (or mechanistic) target of rapamycin (mTOR) pathway (16). The standard therapeutic treatment for OCCC according to the National Comprehensive Cancer Network (NCCN) guidelines (version 1.2020) is optimal cytoreduction combined with systemic chemotherapy. Patients with OCCC tend to be less sensitive to conventional chemotherapy, as only 11–27% of patients with OCCC respond to platinum-based chemotherapy, resulting in poorer outcomes (17). OCCC chemoresistance might be attributed to the increased antioxidant capacity of cancer cells that are able to neutralize the effects of platinum-derived chemotherapeutics (18,19).

This study aimed to have a closer insight into the clinico-pathological features and outcome of patients with OCCC, by identifying factors affecting recurrence, progression-free survival (PFS), and OS, through a retrospective review of all OCCC cases diagnosed and treated at Oxford Cancer Centre over 13 years. We present this article in accordance with the STROBE reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-83/rc).


Methods

Setting and ethical consideration

A retrospective review of all medical records of women diagnosed with primary OCCC between January 2009 and December 2021—with ≥12 months follow-up—at the Churchill Gynecology Oncology Cancer Center, Oxford University Hospitals Foundation Trust was performed. The center is a tertiary hospital receiving referrals from other cancer units in the Thames Valley Cancer Network. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Institutional Review Board following Oxford University Hospitals Foundation Trust clinical governance processes and service evaluation number was obtained (registration No. 7049). Informed consent was obtained from all individual participants.

Data collection

All demographic and pathological characteristics, pre-operative biomarkers, surgical procedure, complications, hospital stay, chemotherapy regimen, and disease status on follow-up, were collected from electronic medical records. Patient records and information were anonymized before analysis; thus, consent was not required. The International Federation of Gynecology and Obstetrics (FIGO 2014) staging system was used for all tumor staging. For cases before 2014, the stage of disease was classified retrospectively based on surgical and pathological assessment. PFS was defined as the time from initial surgical staging or cytoreductive surgery to the date of disease progression or recurrence, and overall survival (OS) was defined as the time from surgical staging or cytoreductive surgery to the date of death, or to the last follow-up date, if still alive. Recurrence was documented by histologic evidence of disease in tumour biopsy/or the appearance of new lesions on imaging examination. Exclusion criteria included patients whose data was available only for pathology review, women presented in our center only at the time of recurrence (primary treatment given elsewhere), and women with synchronous malignancy or data lost at follow-up.

Statistical analysis

Statistical analysis was performed using SPSS v.28.0 software (IBM Corp., Armonk, NY, USA). Summary statistics were used to describe the data. Continuous variables were expressed as mean, standard deviation, and range; categorical variables were expressed as percentages. Comparisons between groups were analyzed using the Student’s t-test or Wilcoxon-Mann-Whitney test according to the data distribution for continuous variables or the χ2 or Fisher’s exact test for categorical variables. Univariate and multivariable Cox regression analyses were performed to identify predictors of PFS and OS. A P value of <0.05 was considered statistically significant, and all P values reported were two-sided.


Results

Two hundred and twenty-seven women were diagnosed with non-high-grade serous epithelial ovarian tumor, and 64 were diagnosed with OCCC between January 2009 and December 2021 at our tertiary Cancer Center. Of 64 women, nine cases were referred for pathology review only, three cases presented at the time of disease recurrence, two had synchronous malignancies, and one was lost from follow-up (due to traveling), leaving 49 cases for this study analysis.

Demographic data and laboratory findings

The mean age at diagnosis was 63 years (range, 37–83 years, with 77.6% post-menopausal women, and mean body mass index (BMI) of 25.86 kg/m2 (range, 16.4–43 kg/m2). More than half of cases presented in early-stage disease, 27 women (55.1%) had stage I, 9 women (18.4%) had stage II, 9 women (18.4%) had stage III, and four women (8.2%) had stage IV; 57.14% were associated with endometriosis, as seen on their histopathology records, and 32.65% had ascites at the time of presentation (Table 1). The mean CA 125 was 402 IU/mL, while 20% of women presented with regular CA 125 levels. The mean hemoglobin (HB) level was 122 g/L, while 26.5% were anemic (HB <115 g/L) at the presentation time.

Table 1

Patients and tumor characteristics

Variables Values (N=49)
Age (years)
   Minimum–maximum 37–83
   Mean ± standard deviation 62.6±11.04
   <40, n [%] 1 [2]
   40–60, n [%] 18 [36.7]
   >60, n [%] 30 [61.2]
Menopausal status, n [%]
   Pre-menopausal 11 [22.4]
   Post-menopausal 38 [77.6]
Body mass index (kg/m2)
   Minimum–maximum 16.4–43
   Mean 25.86
   Underweight <18.5, n [%] 1 [2.6]
   Normal 18.5–24.9, n [%] 18 [47.4]
   Overweight 25–29.9, n [%] 10 [26.3]
   Obese ≥30, n [%] 9 [23.7]
   NA 11
Laterality, n [%]
   Right 23 [53.5]
   Left 19 [44.2]
   Bilateral 1 [2.3]
   NA 6
Endometriosis-associated, n [%]
   Yes 28 [57.14]
   No 21 [42.86]
Ascites, n [%]
   No 33 [67.35]
   Yes 16 [32.65]
FIGO 2014 staging, n [%]
   Stage I 27 [55.1]
    IA 9 [18.4]
    IB 0 [0.0]
    IC 18 [36.7]
      IC1 4 [8.2]
      IC2 4 [8.2]
      IC3 5 [10.2]
      IC NA 5 [10.2]
   Stage II 9 [18.4]
    IIA 5 [10.2]
    IIB 4 [8.2]
   Stage III 9 [18.4]
    IIIA 3 [6.1]
    IIIB 2 [4]
    IIIC 4 [8.2]
   Stage IV 4 [8.2]
    IV A 2 [4]
    IV B 2 [4]

NA data: not available in medical records. FIGO, International Federation of Gynecology and Obstetrics.

Management data

In our retrospective analysis, five women (10%) were enrolled in clinical trials, and neoadjuvant chemotherapy (NACT) was given to four women (8.2%). Diagnostic laparoscopy was performed in 11 women (22.4%), and all patients had cytoreductive surgery. The summarized details of surgical procedures with intra-and postoperative complications are shown in Table 2, with no macroscopic residual disease (R0) being achieved in 39 women (79.59%) of cases. Post-operative adjuvant therapy was offered in 39 women (79.6%); see summary in Table 3.

Table 2

Surgical procedures and complications

Variables Number of patients Percentage
Neoadjuvant chemotherapy
   Yes 4 8.2
   No 45 91.8
Pre-operative laparoscopy
   Not done 36 73.5
   Done 11 22.4
    Done in separate setting 6 12.2
    Done in the same setting 5 10.2
   Done as total laparoscopic hysterectomy 2 4.08
Surgical procedure 47 (NA =2)
   Hysterectomy 40+7 hysterectomy 85.1
   Bilateral salpingo-ophrectomy 46 97.9
   Unilateral salpingo-ophrectomy 1 2.1
   Pelvic lymph node sampling 8 (positive in 1) 17 (2.1)
   Pelvic lymph node dissection 6 (positive) 12.8
   Para aortic lymph node sampling 8 (positive in 1) 17 (2.1)
   Infracolic omentectomy 47 (positive in 5) 100 (positive 10.6)
   Appendectomy 10 (all negative) 21.3
   Falciform ligament resection 3 (all negative) 6.4
   Excision of gastric nodule 1 (negative) 2.1
   Excision of liver nodule 1 (positive) 2.1
   Bladder peritoneum resection 7 (positive in 2) 14.9 (positive 4.3)
   Pelvic peritonectomy 8 17
   Paracolic peritonectomy 6 12.8
   Small bowel resection & end to end anastomosis 2 4.3
   Large bowel resection & end to end anastomosis 1 2.1
   Colostomy 1 2.1
R0 no residual achieved 39 79.59
R0 not achieved 10 20.41
Intra-operative complications 9 19.1
Post operative complications 40 (NA =9)
   None 24 60
   Ileus (all managed conservatively) 6 15
   Severe wound infection 2 5
   Wound dehiscence and return to theatre 1 2.5
   Pelvic collection (readmission) 1 2.5
   Peritonitis + pulmonary embolism 1 2.5
   Pulmonary embolism 1 2.5
   Deep vein thrombosis in left subclavian vein 1 2.5
   Ureteric leaking 1 2.5
   Lymphocyst 1 2.5
   Complete heart block and percutaneous pacing 1 2.5
   Need for intensive care unit admission 4 10

NA data: not available in medical records.

Table 3

Adjuvant therapy

Variables Values
Adjuvant therapy, n 49
   Adjuvant offered, n [%] 39 [79.6]
    Adjuvant chemotherapy, n [%] 37 [75.5]
    Palliative, n [%] 2 [4]
   Adjuvant chemotherapy offered but declined or hold, n/N [%] 4/39 [10.3]
Adjuvant chemotherapy types
   Carboplatin ± paclitaxel, n
    Completed 6 cycles 22
    <6 cycles 7
   Data not clear on medical records, n 10

Follow-up, disease progression, survival, PFS, and recurrence

A follow-up time for non-progress survivors ranged between 12–144 months, with a mean of 105.5 months [95% confidence interval (CI): 87.25–123.9], with a median of 53 months based on the reverse Kaplan-Meier method. Among our cohort group, residual disease (R0 was not achieved) in 10 women (20.4%), who progressed rapidly over the following months or even weeks, and recurrence occurred in 9 women (18.4%), with a total 14 deaths (28.5%) during follow-up period among the whole cohort, with OS 71.4%. The 3-year OS was 73.4%, and the 3-year PFS was 81.3%, with a mean of 101.7 months (95% CI: 84.63–118.93) (Figure 1).

Figure 1 Kaplan Meier survival analysis—OS (A) and PFS (B). OS, overall survival; PFS, progression-free survival.

Further stratification of the cohort group concerning endometriosis association was done; for univariant analysis, OCCC was associated with endometriosis, in 27 women (55.1%), and not associated in 22 women (44.8%). The 3-year OS was 68.1% and 79.9% (95% CI: 87.25–123.9) (in endometriosis-associated and non-endometriosis associated respectively), but with no statistically significant difference using log-rank (Mantel-Cox) analysis. Again, 3-year PFS was 84.4% and 77.9% (95% CI: 84.6–119.93) (in endometriosis-associated and non-endometriosis-associated, respectively), but no statistical significance was found in the analysis.

Stratification of our study group based on the FIGO stage, and estimating the impact of the stage on OS, showed a statistically significant difference (P<0.001), with stage I having the best prognosis and stage IV the worst prognosis (Figure 2). Univariant analysis using log-rank (Mantel-Cox test), for the impact of residual disease on OS, showed a statistically significant difference (P<0.001), the 3-year OS in no-residual disease group 88.6% (95% CI: 108.6–141.8), compared to 12.5% (95% CI: 4.48–32.11) in the residual disease group (Figure 3). In multivariant analysis, CA 125 (<200 IU/mL), HB (<115 g/L), albumin (<40 g/L), endometriosis-association, ascites, residual disease and FIGO staging were included, but only the FIGO stage was an independent prognostic indicator of OS with (P<0.05). None of the other factors had statistical significance (Table 4).

Figure 2 Kaplan Meier survival analysis—stratified by stage. OS, overall survival.
Figure 3 Kaplan Meier survival analysis—stratified by residual disease. OS, overall survival.

Table 4

Multivariant analysis using Cox regression

Predictive factor Independent covariate
HR (95% CI) P value
CA 125 (>200 IU/mL) 0.3 (0.04–2.341) 0.25
Hemoglobin (<115 g/L) 3.2 (0.804–13.53) 0.09
Albumin (<40 g/L) 1.8 (0.276–12.27) 0.52
Ascites 1.471 (0.250–8.66) 0.66
Endometriosis-association 1.980 (0.367–10.691) 0.42
FIGO stage I 1.213 (0.102–14.462) 0.87
FIGO stage II 18.747 (1.029–341.687) 0.048*
FIGO stage III 234.817 (2.765–19940) 0.01*
Residual disease 0.385 (0.020–7.251) 0.52

*, statistically significant. HR, hazard ratio; CI, confidence interval; CA, carbohydrate antigen; FIGO, International Federation of Gynecology and Obstetrics.


Discussion

Our study described OCCC medical data—with a total of 49 women—from a single cancer center in the UK from January 2009 to December 2021. Surprisingly, in our study, the mean age at presentation was 62.6±11.04 years (range, 37–83 years), with >61% presenting above the age of 60 years. This is different from previous retrospective studies that showed a younger mean age at presentation, as a recent publication (20) involving 86 patients stated the median age at diagnosis was 49.21±9.91 years (range, 25–70 years) with only 12% presenting above the age of 60 years. Another recently published study (21), stated the age of onset had two peaks, namely 36 and 77 years, but in our cohort, only one woman presented below the age of 40 years. These studies were conducted in China, with different disease prevalence and possible genetic basis that may explain the findings. However, the Memorial Sloan Kettering (MSK) experience (22) from the USA, including 176 OCCC patients, had a mean age of 53 years at diagnosis.

Our study diagnosed 36 women (73.5%) at stage I or II. This aligns with prior studies (22,23), in which most women with OCCC present with early-stage disease. Part of this may be due to its tendency to present as a large unilateral pelvic mass, compared to the early peritoneal dissemination, which is common in HGSC.

The histopathology protocol followed in our institution for OCCC diagnosis is based generally on the hematoxylin and eosin (H&E) stain. There are a few well-recognized patterns—solid, papillary, and tubule-cystic. There can be a mixture of these in the same malignancy. The carcinoma cells can be transparent or pink (eosinophilic) and there are characteristic features such as pink globules and hob-nail cells/nuclei seen. If there is a need to differentiate from serous carcinomas or endometrioid carcinomas with secretory changes, then Napsin A is the most widely used immunomarker. Estrogen receptor (ER) and PR should be negative, as should WT1, unless there is a component of serous carcinoma admixed. We tend to use vimentin to look for endometrioid epithelium. AMACR can also be used for clear cell tumours but Napsin A is the best.

During our 13-year retrospective period, five patients have been enrolled in 5 different trials: 1st patient was enrolled in Javelin Ovarian 100, NCT02718417: a phase 3 open-label study on avelumab in previously untreated patients with EOC. The study was terminated based on the results of a planned interim analysis that showed futility of efficacy (24). Another patient was enrolled in the OSI trial, identifier: NCT00889382, a multi-center, randomized, open-label, phase 1/2 study of continuous weekly paclitaxel and escalating doses of intermittent or continuous OSI-906 in patients with recurrent/relapsed ovarian and other solid tumors (25). A 3rd patient was enrolled in Chorus trial identifier: NCT00075712, Timing of Surgery and Chemotherapy in Treating Patients with Newly Diagnosed Advanced Ovarian Epithelial, Fallopian Tube, or Primary Peritoneal Cavity Cancer (26). According to her medical notes, the 4th patient was enrolled in the Ovpsch trial, but no further details were found either on medical records or in the trust intranet registered trials. A 5th patient was enrolled in the Peacock trial (27); phase II, multicenter, single-arm trial in patients with advanced clear cell gynaecology cancer (CCGC) who had ≥1 prior line of chemotherapy with progression at study entry.

Since OCCC is a high-risk group, adjuvant chemotherapy is recommended even if the stage is Ia, but the guidelines vary slightly between different gynecologic oncology societies. In our study group, where the British Gynecology Cancer Society guideline is followed, adjuvant chemotherapy was not offered in 10 women (20.4%), as the guideline states that low stages Ia /Ib OCCC are excluded from offering adjuvant platinum-based chemotherapy. According to the GOG 157 trial in which OCCC represented one third of cases (n=130), the benefit of using 6 vs. 3 cycles adjuvant paclitaxel and carboplatin chemotherapy in early-stage ovarian cancer in reducing recurrence risk was not evident in OCCC group (RFS HR =0.9, 95% CI: 0.43, 1.91) (28,29). ACTION study reported similar PFS for patients with early-stage clear-cell carcinoma with or without adjuvant chemotherapy (30) and two retrospective analyses (31,32) reported no benefit from adjuvant chemotherapy after completing surgical staging for stage IA–B clear-cell carcinoma. On the contrary, other studies suggest a high level of biological heterogeneity among clear-cell tumors that can sometimes justify an aggressive appearance even among patients with stage I disease (33). The true challenge is to predict which patients with early-stage disease are at higher risk of recurrence and would most benefit from additional treatments.

Our study’s mean follow-up time was 105.5 months (range, 7–144 months), with 3-year OS 73.4%. The study group showed significant variation in OS for women among stages (P<0.001), 3-year OS rate was 92.1% for stage I, which is consistent with previous studies (21,22,34), 85.7% in stage II, dropping to 25% in stage III, with the worst prognosis for stage IV with 1-year OS 25%.

In our study group, endometriosis was found in 28 women (57.14%), confirmed in the histopathology report; the percentage is consistent with other studies that reported 18% to 43% of women with OCCC have a history of endometriosis (35,36), and a different study has demonstrated that this benign disease is significantly related to the pathogenesis of OCCC, with three-fold increased risk of OCCC [odds ratio (OR) =3.05] when endometriosis being reported (37). In our study, OS and PFS showed no statistically significant difference when Kaplan-Meier analysis stratified by endometriosis. It is unclear in the literature the role of endometriosis on ovarian cancer prognosis. Published studies have generally shown that patients with endometriosis-associated ovarian cancer had higher survival rates. Still, in most series, this finding has been linked to an earlier stage at diagnosis among ovarian cancer cases with endometriosis rather than the association with endometriosis as a prognostic factor or their published data had no statistically significant difference (38,39). Our results agreed with a recently published study that concluded that the presence of endometriosis did not affect either the OS (87.99 vs. 75.30, P value =0.25) or the PFS (111.13 vs. 117.42, P value =0.48) (40).

In our study, in a univariant analysis, a statistically significant difference (P<0.001) was found when survival analysis was stratified according to the presence of residual disease (suboptimal cytoreduction), showing 3-year OS in no-residual disease group 88.6% (95% CI: 108.6–141.8), compared to 12.5% (95% CI: 4.48–32.11) in residual disease group, keeping suboptimal cytoreduction a significant prognostic predictor for OCCC. The residual disease was linked to advanced stages, and the small number of advanced cases in our cohort (13) did not allow further analysis. In published data, other studies examined the association between residual disease and prognosis in clear cell carcinoma (41,42). In a retrospective cohort study, that examined the association between residual disease and prognosis by histological type of ovarian cancer using data from the National Cancer Database of America, OS differed significantly according to residual disease status not only in patients with high-grade serous carcinoma but also in those with clear cell carcinoma and, while not statistically significantly different, the survival benefit associated with complete cytoreduction was more significant in clear cell carcinoma (HR =0.39, 95% CI: 0.22 to 0.69) than in high-grade serous carcinoma (HR =0.58, 95% CI: 0.49 to 0.68) (42). Different studies have elicited the significance of complete cytoreduction and its impact on survival in ovarian cancer patients (43-62).

In our study, in multivariant analysis, CA 125, HB, albumin, endometriosis-association, ascites, residual disease and FIGO staging were included; only FIGO stage was an independent prognostic indicator of OS with (P<0.05), and none of the remaining factors had statistical significance. A similar analysis in a study published by Park et al. (63), found in the univariate survival analysis, significant prognostic factors for both DFS and OS were coexisting endometriosis, elevated preoperative serum CA 125 levels, advanced FIGO stage (stage I vs. stages II–IV), ovarian surface involvement, positive peritoneal cytology, and suboptimal debulking. In the multivariate analysis, coexisting endometriosis and advanced FIGO stage were significant factors for both DFS and OS.

As a recent review of the literature, we added a table of previously published studies over the last 5 years, involving OCCC in retrospective analysis, and the studies have been summarized as shown in (1,20,21,63-66) (Table 5).

Table 5

Summarizes similar retrospective studies involving OCCC in the last years

Authors Year of publication Study design Study period No. of patients Endometriosis association Early stage
I & II
Residual disease Recurrence PFS OS
Gallego (65) 2022 Retrospective 1992–2013 56 58.9% 40.5% 12.5% ~60% (3 years)
Tranoulis (64) 2022 Retrospective 2010–2019 94 51% ~50% stage I 25% with endo; 10.9% with no endo 39 months (in both groups ) 55 months (no endo); 71 months (with endo)
Kim (1) 2016 Retrospective 1999–2018 2,962 74% 74.9% (5 years)
Zhu (20) 2021 Retrospective 2010–2022 86 18.6% 68.6% 10.46% 26.53% 3 years, 78.95% (early); 22.2% (advanced) 3 years, 89.47% (early); 44.44% (advanced)
Zhou (21) 2021 Retrospective 1998–2018 91 54% 72.5% 6.2% 17.2% 89.9% (5 years) 88% (5 years)
Park (63) 2018 Retrospective 1991–2012 155 50.3% 76.7% 14.19% 32.9% 5 years, 83% (endo); 51% (no endo) 5 years, 84% (endo); 54% (no endo)
Scarfone (66) 2014 Retrospective 1990–2012 73 36.9% 56.1% 5 years, 60% (no endo); 73% (endo)

OCCC, ovarian clear cell carcinoma; PFS, progression-free survival; OS, overall survival; endo, endometriosis.

Limitations

The main limitations of this study are the small number of patients in our cohort and the retrospective study design, with potential selection and information bias.


Conclusions

The true challenge is to predict which patients with early-stage disease are at higher risk of recurrence and would most benefit from additional treatments. Surgery to achieve no residual tumor is necessary to improve the prognosis in advanced-stage clear cell carcinoma. Due to the rarity of clear cell carcinoma, international collaboration will be essential to power large-scale clinical trials required to answer the many remaining questions regarding the optimal treatment of this disease.


Acknowledgments

The abstract has been presented as a poster in British Gynaecological Cancer Society June 2023 annual scientific meeting.

Funding: None.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-83/rc

Data Sharing Statement: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-83/dss

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-83/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-83/coif). H.S.m. serves as an unpaid editorial board member of Translational Cancer Research from September 2023 to August 2025. A.A.A. reports patents planned, issued or pending: through Oxford University Innovation (one patent and four pending). He also reports participation on a Data Safety Monitoring Board or Advisory Board: Well-being of Women and The Eve Appeal; and he is a founder, director and consultant of Singula Bio Ltd. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Institutional Review Board following Oxford University Hospitals Foundation Trust clinical governance processes and service evaluation number was obtained (registration No. 7049). Informed consent was obtained from all individual participants.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Kim SI, Lim MC, Lim J, et al. Incidence of epithelial ovarian cancer according to histologic subtypes in Korea, 1999 to 2012. J Gynecol Oncol 2016;27:e5. [Crossref] [PubMed]
  2. Zannoni GF, Morassi F, Prisco MG, et al. Clinicopathologic and immunohistochemical features of ovarian clear cell carcinomas in comparison with type I and type II tumors. Int J Gynecol Pathol 2012;31:507-16. [Crossref] [PubMed]
  3. Terada KY, Ahn HJ, Kessel B. Differences in risk for type 1 and type 2 ovarian cancer in a large cancer screening trial. J Gynecol Oncol 2016;27:e25. [Crossref] [PubMed]
  4. Kobayashi H, Sumimoto K, Moniwa N, et al. Risk of developing ovarian cancer among women with ovarian endometrioma: a cohort study in Shizuoka, Japan. Int J Gynecol Cancer 2007;17:37-43. [Crossref] [PubMed]
  5. Yamamoto S, Tsuda H, Takano M, et al. Clear-cell adenofibroma can be a clonal precursor for clear-cell adenocarcinoma of the ovary: a possible alternative ovarian clear-cell carcinogenic pathway. J Pathol 2008;216:103-10. [Crossref] [PubMed]
  6. Matsuura Y, Robertson G, Marsden DE, et al. Thromboembolic complications in patients with clear cell carcinoma of the ovary. Gynecol Oncol 2007;104:406-10. [Crossref] [PubMed]
  7. Diaz ES, Walts AE, Karlan BY, et al. Venous thromboembolism during primary treatment of ovarian clear cell carcinoma is associated with decreased survival. Gynecol Oncol 2013;131:541-5. [Crossref] [PubMed]
  8. Tamura R, Yoshihara K, Matsuo K, et al. Proposing a molecular classification associated with hypercoagulation in ovarian clear cell carcinoma. Gynecol Oncol 2021;163:327-33. [Crossref] [PubMed]
  9. Liu H, Xu Y, Ji J, et al. Prognosis of ovarian clear cell cancer compared with other epithelial cancer types: A population-based analysis. Oncol Lett 2020;19:1947-57. [Crossref] [PubMed]
  10. Prat J. Ovarian carcinomas: five distinct diseases with different origins, genetic alterations, and clinicopathological features. Virchows Arch 2012;460:237-49. [Crossref] [PubMed]
  11. Köbel M, Kang EY. The Evolution of Ovarian Carcinoma Subclassification. Cancers (Basel) 2022;14:416. [Crossref] [PubMed]
  12. Nistor S, El-Tawab S, Wong F, et al. The clinicopathological characteristics and survival outcomes of primary expansile vs. infiltrative mucinous ovarian adenocarcinoma: a retrospective study sharing the experience of a tertiary centre. Transl Cancer Res 2023;12:2682-92. [Crossref] [PubMed]
  13. Filippini C, Smyth S, Soleymani Majd H, et al. Primary peritoneal mucinous cystadenocarcinoma mimicking possible recurrent ovarian mucinous cystadenoma: coincidental pathology or a spectrum of disease? BMJ Case Rep 2021;14:e242478. [Crossref] [PubMed]
  14. Hunter A, Addley S, Soleymani Majd H. Aggressive anaplastic ovarian carcinoma in a young nulliparous patient. BMJ Case Rep 2021;14:e241461. [Crossref] [PubMed]
  15. Fadare O, Zhao C, Khabele D, et al. Comparative analysis of Napsin A, alpha-methylacyl-coenzyme A racemase (AMACR, P504S), and hepatocyte nuclear factor 1 beta as diagnostic markers of ovarian clear cell carcinoma: an immunohistochemical study of 279 ovarian tumours. Pathology 2015;47:105-11. [Crossref] [PubMed]
  16. Enomoto T, Aoki D, Hattori K, et al. The first Japanese nationwide multicenter study of BRCA mutation testing in ovarian cancer: CHARacterizing the cross-sectionaL approach to Ovarian cancer geneTic TEsting of BRCA (CHARLOTTE). Int J Gynecol Cancer 2019;29:1043-9. [Crossref] [PubMed]
  17. Kobayashi H, Sugimoto H, Onishi S, et al. Novel biomarker candidates for the diagnosis of ovarian clear cell carcinoma. Oncol Lett 2015;10:612-8. [Crossref] [PubMed]
  18. Tossetta G, Fantone S, Goteri G, et al. The Role of NQO1 in Ovarian Cancer. Int J Mol Sci 2023;24:7839. [Crossref] [PubMed]
  19. Fantone S, Piani F, Olivieri F, et al. Role of SLC7A11/xCT in Ovarian Cancer. Int J Mol Sci 2024;25:587. [Crossref] [PubMed]
  20. Zhu C, Zhu J, Qian L, et al. Clinical characteristics and prognosis of ovarian clear cell carcinoma: a 10-year retrospective study. BMC Cancer 2021;21:322. [Crossref] [PubMed]
  21. Zhou L, Yao L, Dai L, et al. Ovarian endometrioid carcinoma and clear cell carcinoma: A 21-year retrospective study. J Ovarian Res 2021;14:63. [Crossref] [PubMed]
  22. Shu CA, Zhou Q, Jotwani AR, et al. Ovarian clear cell carcinoma, outcomes by stage: the MSK experience. Gynecol Oncol 2015;139:236-41. [Crossref] [PubMed]
  23. Köbel M, Kalloger SE, Huntsman DG, et al. Differences in tumor type in low-stage versus high-stage ovarian carcinomas. Int J Gynecol Pathol 2010;29:203-11. [Crossref] [PubMed]
  24. Avelumab in Previously Untreated Patients With Epithelial Ovarian Cancer (JAVELIN OVARIAN 100). NCT02718417. Available online: https://clinicaltrials.gov/study/NCT02718417
  25. A Study Evaluating Intermittent and Continuous OSI-906 and Weekly Paclitaxel in Patients With Recurrent Epithelial Ovarian Cancer (and Other Solid Tumors). NCT00889382. Available online: https://clinicaltrials.gov/study/NCT00889382
  26. Kehoe S, Hook J, Nankivell M, 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] [PubMed]
  27. Miller RE, Clamp A, Gourley C, et al. 2022-RA-1276-ESGO Pembrolizumab monotherapy for advanced clear cell gynaecological cancer: phase II PEACOCC trial. International Journal of Gynecologic Cancer 2022;32:A316.
  28. Bell J, Brady MF, Young RC, et al. Randomized phase III trial of three versus six cycles of adjuvant carboplatin and paclitaxel in early stage epithelial ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2006;102:432-9. [Crossref] [PubMed]
  29. Chan JK, Tian C, Fleming GF, et al. The potential benefit of 6 vs. 3 cycles of chemotherapy in subsets of women with early-stage high-risk epithelial ovarian cancer: an exploratory analysis of a Gynecologic Oncology Group study. Gynecol Oncol 2010;116:301-6. [Crossref] [PubMed]
  30. Trimbos JB, Vergote I, Bolis G, et al. Impact of adjuvant chemotherapy and surgical staging in early-stage ovarian carcinoma: European Organisation for Research and Treatment of Cancer-Adjuvant ChemoTherapy in Ovarian Neoplasm trial. J Natl Cancer Inst 2003;95:113-25. [Crossref] [PubMed]
  31. Mizuno M, Kajiyama H, Shibata K, et al. Adjuvant chemotherapy for stage i ovarian clear cell carcinoma: is it necessary for stage IA? Int J Gynecol Cancer 2012;22:1143-9. [Crossref] [PubMed]
  32. Takada T, Iwase H, Iitsuka C, et al. Adjuvant chemotherapy for stage I clear cell carcinoma of the ovary: an analysis of fully staged patients. Int J Gynecol Cancer 2012;22:573-8. [Crossref] [PubMed]
  33. Friedlander ML, Russell K, Millis S, et al. Molecular Profiling of Clear Cell Ovarian Cancers: Identifying Potential Treatment Targets for Clinical Trials. Int J Gynecol Cancer 2016;26:648-54. [Crossref] [PubMed]
  34. Chan JK, Teoh D, Hu JM, et al. Do clear cell ovarian carcinomas have poorer prognosis compared to other epithelial cell types? A study of 1411 clear cell ovarian cancers. Gynecol Oncol 2008;109:370-6. [Crossref] [PubMed]
  35. Ye S, Yang J, You Y, et al. Comparative study of ovarian clear cell carcinoma with and without endometriosis in People's Republic of China. Fertil Steril 2014;102:1656-62. [Crossref] [PubMed]
  36. Paik ES, Kim TJ, Choi CH, et al. Clinical outcomes of patients with clear cell and endometrioid ovarian cancer arising from endometriosis. J Gynecol Oncol 2018;29:e18. [Crossref] [PubMed]
  37. King CM, Barbara C, Prentice A, et al. Models of endometriosis and their utility in studying progression to ovarian clear cell carcinoma. J Pathol 2016;238:185-96. [Crossref] [PubMed]
  38. Orezzoli JP, Russell AH, Oliva E, et al. Prognostic implication of endometriosis in clear cell carcinoma of the ovary. Gynecol Oncol 2008;110:336-44. [Crossref] [PubMed]
  39. Noli S, Cipriani S, Scarfone G, et al. Long term survival of ovarian endometriosis associated clear cell and endometrioid ovarian cancers. Int J Gynecol Cancer 2013;23:244-8. [Crossref] [PubMed]
  40. Kim HS, Kim MA, Lee M, et al. Effect of Endometriosis on the Prognosis of Ovarian Clear Cell Carcinoma: A Two-Center Cohort Study and Meta-analysis. Ann Surg Oncol 2015;22:2738-45. [Crossref] [PubMed]
  41. Iida Y, Okamoto A, Hollis RL, et al. Clear cell carcinoma of the ovary: a clinical and molecular perspective. Int J Gynecol Cancer 2021;31:605-16. [Crossref] [PubMed]
  42. Melamed A, Manning-Geist B, Bregar AJ, et al. Associations between residual disease and survival in epithelial ovarian cancer by histologic type. Gynecol Oncol 2017;147:250-6. [Crossref] [PubMed]
  43. Tozzi R, Hardern K, Gubbala K, et al. En-bloc resection of the pelvis (EnBRP) in patients with stage IIIC-IV ovarian cancer: A 10 steps standardised technique. Surgical and survival outcomes of primary vs. interval surgery. Gynecol Oncol 2017;144:564-70. [Crossref] [PubMed]
  44. Tozzi R, Giannice R, Cianci S, et al. Neo-adjuvant chemotherapy does not increase the rate of complete resection and does not significantly reduce the morbidity of Visceral-Peritoneal Debulking (VPD) in patients with stage IIIC-IV ovarian cancer. Gynecol Oncol 2015;138:252-8. [Crossref] [PubMed]
  45. Pinelli C, Morotti M, Casarin J, et al. Interval Debulking Surgery for Advanced Ovarian Cancer in Elderly Patients (≥70 y): Does the Age Matter? J Invest Surg 2021;34:1023-30. [Crossref] [PubMed]
  46. Tozzi R, Ferrari F, Nieuwstad J, et al. Tozzi classification of diaphragmatic surgery in patients with stage IIIC-IV ovarian cancer based on surgical findings and complexity. J Gynecol Oncol 2020;31:e14. [Crossref] [PubMed]
  47. Addley S, Morotti M, Pinelli C, et al. Transdiaphragmatic resection of enlarged cardiophrenic lymph node during interval debulking surgery for advanced ovarian cancer. Gynecol Oncol Rep 2021;37:100807. [Crossref] [PubMed]
  48. Tate S, Nishikimi K, Matsuoka A, et al. Bevacizumab in First-Line Chemotherapy Improves Progression-Free Survival for Advanced Ovarian Clear Cell Carcinoma. Cancers (Basel) 2021;13:3177. [Crossref] [PubMed]
  49. Tozzi R, Casarin J, Baysal A, et al. Morbidity of multiple bowel resection compared to single bowel resection after debulking surgery for ovarian cancer. Eur J Obstet Gynecol Reprod Biol 2019;240:215-9. [Crossref] [PubMed]
  50. Elena Buruiana F, Ismail L, Ferrari F, et al. The Role of Ultra-Radical Surgery in the Management of Advanced Ovarian Cancer: State or Art. IntechOpen. 2021. doi: 10.5772/intechopen.97638.10.5772/intechopen.97638
  51. Tozzi R, Traill Z, Campanile RG, et al. Diagnostic flow-chart to identify bowel involvement in patients with stage IIIC-IV ovarian cancer: Can laparoscopy improve the accuracy of CT scan? Gynecol Oncol 2019;155:207-12. [Crossref] [PubMed]
  52. Tozzi R, Casarin J, Garruto-Campanile R, et al. Morbidity and reversal rate of ileostomy after bowel resection during Visceral-Peritoneal Debulking (VPD) in patients with stage IIIC-IV ovarian cancer. Gynecol Oncol 2018;148:74-8. [Crossref] [PubMed]
  53. Soleymani Majd H, Ferrari F, Manek S, et al. Diaphragmatic peritonectomy vs. full thickness resection with pleurectomy during Visceral-Peritoneal Debulking (VPD) in 100 consecutive patients with stage IIIC-IV ovarian cancer: A surgical-histological analysis. Gynecol Oncol 2016;140:430-5. [Crossref] [PubMed]
  54. Tozzi R, Casarin J, Baysal A, et al. Bowel resection rate but not bowel related morbidity is decreased after interval debulking surgery compared to primary surgery in patents with stage IIIC-IV ovarian cancer. J Gynecol Oncol 2019;30:e25. [Crossref] [PubMed]
  55. Bogani G, Ditto A, Martinelli F, et al. Surgical Techniques for Diaphragmatic Resection During Cytoreduction in Advanced or Recurrent Ovarian Carcinoma: A Systematic Review and Meta-analysis. Int J Gynecol Cancer 2016;26:371-80. [Crossref] [PubMed]
  56. Tozzi R, Soleymani Majd H, Campanile RG, et al. Feasibility of laparoscopic diaphragmatic peritonectomy during Visceral-Peritoneal Debulking (VPD) in patients with stage IIIC-IV ovarian cancer. J Gynecol Oncol 2020;31:e71. [Crossref] [PubMed]
  57. Gadducci A, Tana R, Landoni F, et al. Analysis of failures and clinical outcome of advanced epithelial ovarian cancer in patients with microscopic residual disease at second-look reassessment following primary cytoreductive surgery and first-line platinum-based chemotherapy. Eur J Gynaecol Oncol 2013;34:213-7. [PubMed]
  58. Soleymani Majd H, Ismail L, Hardern K, et al. Comparison of survival outcome of patients with primary peritoneal and fallopian tube carcinoma treated with neoadjuvant chemotherapy versus primary debulking surgery. J Obstet Gynaecol 2017;37:89-92. [Crossref] [PubMed]
  59. Smyth SL, Majd HS. The application of pringle manoeuvre, type three liver mobilisation, full thickness diaphragmatic resection with primary closure technique and peritonectomy in the management of advanced ovarian malignancy. Obstet Gynecol Sci 2023;66:459-61. [Crossref] [PubMed]
  60. Pinelli C, Morotti M, Casarin J, et al. The Feasibility of Cardiophrenic Lymphnode Assessment and Removal in Patients Requiring Diaphragmatic Resection During Interval Debulking Surgery for Ovarian Cancer. J Invest Surg 2021;34:756-62. [Crossref] [PubMed]
  61. Addley S, Yao DS, Soleymani Majd H. Primary diaphragmatic closure following diaphragmatic resection and cardiophrenic lymph node dissection during interval debulking surgery for advanced ovarian malignancy. Gynecol Oncol Rep 2021;36:100744. [Crossref] [PubMed]
  62. EL-Tawab S. Soleymani Majd H. Evolutions in the management of advanced ovarian cancer. Gynecol Pelvic Med 2023;6:1. [Crossref]
  63. Park JY, Kim DY, Suh DS, et al. Significance of Ovarian Endometriosis on the Prognosis of Ovarian Clear Cell Carcinoma. Int J Gynecol Cancer 2018;28:11-8. [Crossref] [PubMed]
  64. Tranoulis A, Buruiana FH, Gupta B, et al. Friend or foe? The prognostic role of endometriosis in women with clear cell ovarian carcinoma. A UK population-based cohort study. Arch Gynecol Obstet 2022;305:1279-89. [Crossref] [PubMed]
  65. Gallego A, Mendiola M, Hernando B, et al. Prognostic markers of inflammation in endometrioid and clear cell ovarian cancer. Int J Gynecol Cancer 2022;32:1009-16. [Crossref] [PubMed]
  66. Scarfone G, Bergamini A, Noli S, et al. Characteristics of clear cell ovarian cancer arising from endometriosis: a two center cohort study. Gynecol Oncol 2014;133:480-4. [Crossref] [PubMed]
Cite this article as: El Tawab S, Nistor S, Roux R, Manek S, Gaitskell K, Ahmed AA, Kehoe S, Soleymani majd H. The surgical, histopathological characteristics, and survival outcome of ovarian clear cell carcinoma: a retrospective case series sharing the experience of a tertiary cancer centre. Transl Cancer Res 2024;13(9):5037-5049. doi: 10.21037/tcr-24-83

Download Citation