Abstract
Purpose: Although ovarian clear cell carcinomas (OCCC) are commonly resistant to platinum-based chemotherapy, good clinical outcomes are observed in a subset of patients. The explanation for this is unknown but may be due to misclassification of high-grade serous ovarian cancer (HGSOC) as OCCC or mixed histology.
Experimental Design: To discover potential biomarkers of survival benefit following platinum-based chemotherapy, we ascertained a cohort of 68 Japanese and Australian patients in whom progression-free survival (PFS) and overall survival (OS) could be assessed. We performed IHC reclassification of tumors, and targeted sequencing and immunohistochemistry of known driver genes. Exome sequencing was performed in 10 patients who had either unusually long survival (N = 5) or had a very short time to progression (N = 5).
Results: The majority of mixed OCCC (N = 6, 85.7%) and a small proportion of pure OCCC (N = 3, 4.9%) were reclassified as likely HGSOC. However, the PFS and OS of patients with misclassified samples were similar to that of patients with pathologically validated OCCC. Absent HNF1B expression was significantly correlated with longer PFS and OS (P = 0.0194 and 0.0395, respectively). Mutations in ARID1A, PIK3CA, PPP2R1A, and TP53 were frequent, but did not explain length of PFS and OS. An exploratory exome analysis of patients with favorable and unfavorable outcomes did not identify novel outcome-associated driver mutations.
Conclusions: Survival benefit following chemotherapy in OCCC was not associated with pathological misclassification of tumor histotype. HNF1B loss may help identify the subset of patients with OCCC with a more favorable outcome.
Translational Relevance
Ovarian clear cell carcinomas (OCCC) are relatively resistant to platinum-based chemotherapy with lower response rates and shorter progression-free survival compared with high-grade serous cancers. Identification of pathologic and molecular markers in the small proportion of patients who have good outcomes following chemotherapy may help select patients likely to benefit from standard treatment and identify patients with a low probability of response to proceed directly to clinical trials with novel agents. It is speculated that pathological misclassification of OCCC, particularly those with mixed serous and clear cell morphologies, or those with a TP53 mutation account for occasional responses to platinum-based chemotherapy. Our findings, from a cohort of Australian and Japanese patients, show that survival benefit following chemotherapy is not limited to misclassified OCCC, or to immmunohistochemically validated OCCC with TP53 mutations, or other driver mutations. Low-level expression of HNF1B and Ki67 appear to be associated with particularly favorable outcome.
Introduction
Epithelial ovarian cancer (EOC) predominantly consists of five histologic subtypes: high-grade serous, low-grade serous, endometrioid, clear cell, and mucinous carcinomas, and these differ in clinicopathologic characteristics including chemo-sensitivity and molecular features as well as patient outcomes (1, 2). Ovarian clear cell carcinoma (OCCC) is uncommon, and diagnosed in only approximately 12% of women with EOC in Western countries. For reasons that are unclear the incidence of OCCC is higher in Japan, where it accounts for 25% of EOC (3). OCCC is associated with a poorer prognosis, especially in advanced stage disease, compared with other EOC subtypes and is typically resistant to platinum-based therapy (2, 4, 5). Response rates of OCCC to conventional platinum-based chemotherapy are variable and range from 11% to 56% in the first-line setting, and contrast sharply with the responses rates of >80% in the more common high-grade serous ovarian carcinomas (HGSOC; refs. 2, 4, 5).
HGSOC is molecularly characterized by almost ubiquitous TP53 mutations (6), frequent alterations in homologous recombination DNA repair pathway genes, including BRCA1 and BRCA2 (5, 7, 8), and extensive copy number change. OCCC are molecularly distinct from HGSOC, with much lower rates of TP53 mutations and homologous recombination repair deficiency; which appear to impart platinum sensitivity to HGSOC. Conversely, point-mutated driver events in ARID1A, PIK3CA, and deregulated PI3K/AKT/mTOR, RAS/RAF/REK, and receptor tyrosine kinase (RTK) signaling pathways are more common in OCCC (9–12). ARID1A variants have been reported as poor prognostic and/or chemo-resistant markers in OCCC in some previous studies (13–15). The association of TP53 mutation with chemo-sensitivity in OCCC is unclear.
Diagnostic histopathologic classification of EOC has advanced significantly in the last decade, propelled by a deeper understanding of the molecular features of different histotypes and algorithms using specific IHC markers (16). As these developments are recent, pathologic review of cohort series of EOC banked in past years usually results in the re-evaluation of a proportion of initial diagnostic reports. In particular, recent clinicopathologic and IHC studies have described a subset of OCCC that are likely to be misclassified HGSOC, rather than OCCC (11, 17–19). The misclassified tumors often contained both serous and clear cell components, referred to as mixed histology, but historically were regarded as OCCC when clear cell was the dominant morphology. Whether misclassified OCCC accounts for the small proportion of patients who respond to platinum-based chemotherapy is unknown.
To our knowledge, pathologic, molecular, and clinical correlates have not been evaluated in a cohort of patients with OCCC, where survival benefit following chemotherapy could be assessed. Given the substantial clinical utility of prospective identification of the subset of patients with OCCC who are more likely to benefit from platinum-based chemotherapy, we ascertained a cohort of patients with OCCC with appropriate biospecimens and clinical history for detailed pathologic and molecular analyses. IHC re-classification and targeted sequencing were performed to identify molecular characteristics in all tumors. Clinicopathologic information, including morphology and sensitivity to chemotherapy, assessed by progression-free survival (PFS), were intersected with the molecular characteristics of tumors. Additional whole-exome sequencing was performed in patients with OCCC with an exceptionally long or short survival.
Materials and Methods
Patients
The study population consisted of women diagnosed with pure and mixed OCCC from the Australian Ovarian Cancer Study (AOCS), Melbourne, Australia, and The Jikei University School of Medicine (JIKEI), Tokyo, Japan. AOCS patients were ascertained from Australian hospitals between 2002 and 2009. Patients from JIKEI received primary surgery and adjuvant chemotherapy at hospitals affiliated with the Department of Obstetrics and Gynecology in the JIKEI between 2000 and 2014. OCCC cases with insufficient clinical information to evaluate PFS following chemotherapy and those without tumor tissue available were excluded (Fig. 1). Clinicopathologic information including age, pathologic diagnosis, primary surgery, surgical staging, size of residual tumor (RT) at primary surgery, regimen(s) and period(s) of 1st (and 2nd) line(s) of chemotherapy, disease progression(s), and outcome were collected by review of clinicopathologic records. Surgical staging was assessed in accordance with International Federation of Gynecology and Obstetrics (FIGO) classification. This study was conducted in accordance with the Declaration of Helsinki ethical guidelines and the principles of Good Clinical Practice, and was approved by Human Research Ethics Committees at all participating centers. Written informed consent was obtained from all participants.
Outline of cohort selection. Cases included in the study were from the AOCS and the JIKEI with pure or mixed OCCC. Patients were selected only if assessable for the impact of chemotherapy on survival, that is, those with residual tumor or those without residual tumor who progressed (evaluable). Patients with no residual tumor who had not progressed (unevaluable) and those without biospecimens available were excluded.
Clinical definitions
Disease progression was determined by attending physicians based on imaging, Gynecological Cancer InterGroup (GCIG) CA125 criteria (20), and/or clinical evaluation. PFS was defined as time from primary surgery to first disease progression on or after first-line chemotherapy. Treatment-free interval (TFI) was defined as the time interval between the last date of previous chemotherapy and the date of disease progression or recurrence, and was only used to select patients with exceptional outcomes following second-line chemotherapy in the outlier analysis. Overall survival (OS) was defined as the time interval between histologic diagnosis at primary surgery to death or last contact. Patients were not treated on clinical trials and serial imaging data was not available, therefore we were unable to reliably assess tumor response using RECIST guidelines in most patients, and PFS and TFI were used as surrogates for response.
Pathology review
The initial pathologic classification was extracted from diagnostic reports. All eligible cases subsequently underwent pathology review by expert gynecologic pathologists using a complete set of hematoxylin and eosin (H&E)-stained diagnostic slides or 1 to 10 representative diagnostic block(s) of tumor(s). Pure OCCC was defined as typical clear or hobnail cells present in papillary, solid, or tubulocystic patterns. Mixed OCCC was defined as the presence of another histologic type with OCCC, usually serous, with the minor component consisting of at least 10% of the tumor (21).
Selection of clinical outliers
We defined exceptional clinical outliers as patients with advanced stage disease (stage III or IV) whose PFS, TFI following second-line chemotherapy or OS were two SDs beyond the median of the advanced stage disease cohort (Supplementary Figs. S13 and S14; Supplementary Table S6A and S6B). Conversely, for those with unusually poor outcomes, we selected patients with stage I OCCC with a PFS of less than 12 months and/or an OS of less than 2 years.
Statistical analyses
Distributions of patient survival was estimated with Kaplan–Meier curve analysis, and the statistical significance of apparent differences in survival between groups was tested using the log-rank test, with P < 0.05 considered significant. Fisher exact and Mann–Whitney tests were used to determine P values in indicated analyses. This study is insufficiently powered for a multivariate analysis.
Tissue microarray construction and IHC
Tissue microarrays (TMA) were constructed from representative formalin-fixed paraffin-embedded (FFPE) tumor tissue blocks selected by expert gynecological pathologists using duplicate 0.6 mm (AOCS) or 2.0 mm (JIKEI) cores. In three mixed cases, J-44, J-45, and J-46, tumor cores were separately obtained from both OCCC and HGSOC regions to compare molecular characteristics. IHC analyses of WT1, HNF1B, Napsin A, p53, estrogen receptor (ER), ARID1A, PAX8, FOLR1, IGF2BP3, Ki67 and four mismatch repair (MMR) proteins MLH1, MSH2, MSH6, and PMS2 were performed using TMA sections as described in Supplementary Materials and Methods and Supplementary Table S1.
Nucleic acid isolation
H&E stained sections of FFPE tumor blocks were marked by a gynecological pathologist and tissue containing >80% tumor cells was obtained from five to ten 4 or 10 μm adjacent sections. For samples with an estimated tumor cellularity of <80%, needle dissection was performed on up to 25, 4 or 10 μm sections. Normal DNA was obtained from tumor-free tissue removed at diagnosis. Genomic DNA was obtained using the DNeasy Blood and Tissue Kit (Qiagen), and quantified using the Qubit dsDNA BR assay (Thermo Fisher Scientific). Tumor DNA was separately obtained from both OCCC and HGSOC regions for three mixed cases, J-44, J-45, and J-46.
DNA sequence analysis
For all cases with tumor samples available (n = 68), we assessed tumor DNA for pathogenic and likely pathogenic mutations in 103 genes associated with OCCC and HGSOC (Supplementary Table S2), using a custom capture panel (SureDesign; Agilent). For clinical outliers (n = 10), whole-exome sequencing was performed on tumor DNA and matched normal DNA using the SureSelect Human All Exon V6 Kit following the SureSelectXT recommended protocol (Agilent Technologies). Further details are provided in the Supplementary Materials and Methods.
Results
Selection of patients with OCCC with evaluable disease
Ovarian cancer, including OCCC, is typically treated with debulking surgery followed by adjuvant chemotherapy. To evaluate survival outcomes following platinum-based chemotherapy in patients with OCCC, we first excluded those with tumors who may have been cured by surgery alone and selected patients with residual disease (stages II–IV) following primary debulking surgery and who subsequently received adjuvant chemotherapy. We also included patients who were diagnosed with disease recurrence on/after adjuvant chemotherapy despite having no macroscopic residual disease after primary surgery, irrespective of their initial stage (Fig. 1; Supplementary Fig. S3). Disease relapse in these patients suggested that these cancers were inherently chemoresistant.
Given the stringent selection criteria and that OCCC is an uncommon histotype, we chose to ascertain patients from both a large national cohort study of Australian ovarian cancer patients (AOCS) and a high volume Japanese university clinical network (JIKEI), where the incidence of OCCC is higher than in Caucasian populations. One hundred and thirty-three patients (32%) from an initial cohort of 413 met our selection criteria. We further filtered based on the availability of material for pathologic, IHC, and molecular analyses, resulting in a final cohort of 68 patients. Despite differences in ethnicity and location, there were no significant clinical differences between the AOCS and JIKEI cohorts, although there was a trend towards younger patients among those from Japan (Table 1). Seven of 68 patients were mixed OCCC cases. Six of seven cases had clear cell and serous components, and one case, A-2486, consisted of three histologic regions: clear cell, serous, and endometrioid (Supplementary Table S3). Clinicopathologic characteristics are summarized in Supplementary Fig. S3. Although most known prognostic factors including age of diagnosis, stage, and extent of residual tumor were not significantly associated with survival in this cohort, only stage was correlated with PFS but not OS (Supplementary Fig. S4).
Patient characteristics collected from each ovarian cancer cohort
IHC reclassification of mixed OCCC as likely HGSOC
Morphological classification of OCCC recognizes tumors with uniform clear cell histologic features (pure) and those with components of clear cell and other histotypes (mixed), usually serous or endometrioid (18, 19). Our cohort of 68 patients was identified following initial diagnostic and subsequent expert morphologic review of diagnostic samples. Recently, probabilistic algorithms applied to panels of IHC markers have been used to more effectively subtype ovarian cancer in large patient cohorts (16, 22). We therefore applied this validated algorithm, using staining of WT1, HNF1B, Napsin A, p53, and ER to confirm or refute histotype. A majority of tumor samples showed a pattern of staining consistent with OCCC and all but one of these were of pure morphology (Fig. 2A; Supplementary Fig. S5). Nine samples (13.2%) had a staining pattern consistent with HGSOC, including aberrant p53 staining, positivity for WT1 and an absence of OCCC markers HNF1B and Napsin A. While most were of mixed histology, one third were classed as pure. Therefore, although mixed histology is highly suggestive of a misclassified HGSOC tumor, pure morphology was not an entirely reliable indicator of true OCCC status.
IHC profiling the study cohort. A, Cases were reclassified as confirmed OCCC, Likely HGSC, or Unclassified using five IHC markers (red) and standardized scoring. Additional IHC stains are shown in black. Antibodies to MMR proteins included those to MLH1, MSH2, MSH6, and PMS2. B, Lack of association of pathologic and IHC reclassification with patient outcome. C, Significant correlation of progression free and OS with the level of expression in HNF1B.
For another five cases without Napsin A and HNF1B staining the possibility of endometrioid carcinomas could not entirely be excluded and those cases were regarded as uncertain/unclassified. The re-classification of all three groups (clear cell, likely HGSOC, uncertain) based on the algorithmic markers was consistent with observed immunoreactivity to ARID1A, which is typically lost in OCCC rather than HGSOC (Fig. 2A; Table 2; refs. 9, 10).
Correlations between immunochemical reclassification and clinicopathological and molecular characteristics
The proportion of likely HGSOC within the cohort was similar to reported rates of response to chemotherapy in OCCC (2, 5), suggesting that misclassified samples could account for occasional responses seen in some patients. We first considered possible confounders of age, stage, and extent of residual disease but found no statistically significant difference between the immunohistochemically-defined subtypes (Table 2). We then performed Kaplan–Meier survival analyses, but surprisingly found no difference in PFS or OS between patients with tumors of pure or mixed morphologies, or tumor classifications assigned immunohistochemically (Fig. 2B). Importantly, survival benefit was not confined to those likely HGSOC that were originally misclassified as OCCC.
HNF1B loss was associated with longer PFS and OS in OCCC
A number of IHC markers, including those used in this study, have been associated with clinical outcome in OCCC. However, most prior studies have not focused on those with an assessable tumor burden to evaluate clinical outcomes following chemotherapy. We therefore assessed known prognostic factors (age of diagnosis, stage, extent of residual tumor) and expression patterns of a panel of IHC markers (HNF1B, ARID1A, Ki67, FOLR1, IGF2BP3) for associations with clinical characteristics and survival in the 54 confirmed patients with OCCC (Supplementary Table S4; Supplementary Figs. S6 and S7). All known prognostic factors were not significantly associated with survival in 54 confirmed OCCC cases (Supplementary Fig. S6). Loss of ARID1A expression was frequently observed in cases with advanced disease and residual tumor compared with early stage and completely resected cases (P = 0.035 and 0.027, respectively; Supplementary Table S4). Interestingly, Ki67 scores in JIKEI cases were significantly higher than those in AOCS cases (P = 0.020), but we are uncertain whether this is related to differences in ethnicity between patient populations or potential differences in tissue processing (Supplementary Table S4). Interestingly, loss of HNF1B expression was significantly associated with longer PFS (P = 0.0194) and OS (P = 0.0395; Fig. 2C). To our knowledge, there has been no previous study showing an association between loss of HNF1B expression and favorable outcomes in OCCC.
We also investigated the correlation of IHC markers with outcomes in some clinical subgroups. In 24 OCCCs with residual tumor at primary surgery, only loss of HNF1B showed significant correlation with longer PFS (P = 0.0152) and a trend towards longer OS (P = 0.0948; Supplementary Fig. S8). In 30 OCCCs with no residual tumor at primary surgery, loss of ARID1A and higher Ki67 expression were associated with shorter PFS (P = 0.0378 and 0.0257, respectively), but not significantly with OS (P = 0.0727 and 0.1351, respectively; Supplementary Fig. S9).
Patients with OCCC with stage I tumors generally have a favorable prognosis, however a relatively small proportion of stage I patients experience recurrence on or after first-line chemotherapy. Therefore, we investigated the relationship of PFS with expression of IHC markers in 13 patients with stage I disease. A high level of Ki67 and loss of ARID1A were associated with shorter PFS (P = 0.0029 and 0.0716, respectively) and OS (P = 0.0216 and 0.0428, respectively; Supplementary Fig. S10).
Concordant IHC and mutational profiles in OCCC samples
OCCC is characterized by frequent mutations in ARID1A and PIK3CA, and a low frequency of TP53 mutations (7, 9, 10). Given that some previous studies may have erroneously included non-OCCC samples in their analysis, we used our immunopathologically-characterized cohort to better define the frequency of mutations in genes commonly altered in OCCC. We performed targeted sequencing using a custom capture panel of 103 genes that have previously been reported as mutated in OCCC or HGSOC (Supplementary Table S2).
We identified 145 likely pathogenic variants in 28 genes, present in 63 of 68 cases (92.6%; Fig. 3A; Supplementary Table S5). Mutations in ARID1A, PIK3CA, PPP2R1A, RAS pathway members (KRAS and BRAF), and TP53 occurred most frequently. To further validate findings, we examined the consistency between IHC and mutational results using informative antibodies for ARID1A and p53. Abnormal IHC staining patterns and presence of mutations in TP53 and ARID1A were strongly associated (P < 0.0001 in both TP53 and ARID1A; Supplementary Fig. S11). Sensitivity and specificity were 0.82 and 1.00 for TP53, and 0.88 and 0.91 for ARID1A, respectively.
Mutational profiling of OCCC. A, Likely pathogenic alterations detected by custom capture sequencing of tumor DNA from the study cohort. Colored boxes indicate types of mutations detected. Cross-hatching indicates existence of other variants. Vertical bars indicate number of mutated genes, and horizontal bars show percentages of cases with mutation. B, Association of PFS with mutational status of TP53, ARID1A, and PIK3CA. C, Frequently altered pathways in 54 CCCs. PI3K pathway alterations include PIK3CA, PTEN, PIK3R1, and PIK3R2. RTK mutations include mutations in ERBB2, ERBB3, ERBB4, EGFR, MET, FGFR2, PDGFRB, and FLT4. RAS pathway mutations have been grouped as KRAS and BRAF. SMI/SNF pathway alterations include ARID1A, ARID1B, and ARID5B.
ARID1A, PIK3CA, PPP2R1A, and BRAF mutations were restricted to tumors that had been classified immunohistochemically as OCCC (Table 2). Mutations in KRAS were observed in OCCC tumors, but also in one likely HGSOC and three unclassified cases. Mutations in TP53 were observed in all likely HGSOC cases (100%), consistent with almost invariant TP53 mutation in this histotype (6, 7). By contrast, TP53 mutation was detected in only five of 54 OCCC cases (9.3%). We tested whether the small proportion of TP53 mutation-positive OCCC were associated with differential PFS. Kaplan–Meier analysis showed a nonsignificant trend to longer PFS (Fig. 3B). ARID1A mutations tended to be associated with shorter PFS, although this did not reach statistical significance, possibly due to limited sample size. ARID1A mutations have been reported to be associated with poorer prognosis and chemo-resistance in some studies, whereas others have failed to confirm this (13–15). PPP2R1A mutations were only seen in tumors from patients who were optimally debulked (Supplementary Table S4). No other frequent mutations, including PIK3CA, KRAS, and PPP2R1A, were associated with differences in PFS (Fig. 3B; Supplementary Fig. S12A).
Components of the SWI/SNF complex, including ARID1A, ARID1B, and ARID5B, were the most commonly altered signaling pathway, with 63% of tumors tested having at least one mutation in a pathway component (Fig. 3C). Mutations or amplification in PI3K family members, RTK and RAS pathways were common, detected in 51.9%, 22.2%, and 16.7% of tumors, respectively. RTK and RAS mutations were largely mutually exclusive. Overall, we did not observe differences between OCCC patients with and without clearly discernable pathway aberration, for either PFS or OS (Supplementary Fig. S12B).
We next evaluated the association between frequent mutations and outcomes in clinical subgroups. In 24 cases with residual tumor at primary surgery, no frequent mutations showed significant differences in PFS, but ARID1A mutation tended to be associated longer survival contrary to previous reports (Supplementary Fig. S13). By contrast, in 30 cases with optimal debulking, ARID1A mutations were associated with shorter PFS, supporting the observation that loss of ARID1A expression is associated with shorter PFS (Supplementary Fig. S14). In the subset of patients with stage I disease, we did not identify any associations between mutation status and PFS (Supplementary Fig. S15).
To further support the conclusion that mixed tumors were mostly misclassified HGSOC rather than two distinct coexisting tumors, we evaluated the mutational profile of three cases in which we performed targeted sequencing on separate regions of clear cell or serous histology. In all patients, TP53 mutations were identical in the matching regions showing that they had shared origins rather than being collision tumors (Supplementary Fig. S16).
HGSOC and OCCC display distinct DNA copy number profiles (23–25). To examine the correlation between copy number alterations and IHC subtype, we compared DNA gains and losses in 54 OCCCs and 9 likely HGSOC by making use of read depth from the targeted sequencing data. Although a relatively low-resolution method given the number of genes assayed, unsupervised hierarchical clustering cleanly separated the groups as defined by IHC analysis (Supplementary Fig. S17A). OCCC samples segregated into three groups but these were not associated with differential clinical outcome (Supplementary Fig. S17B).
Molecular aberrations in clinical outliers
Recently, patients with unusually favorable or unfavorable responses to conventional and targeted therapies have provided unique insights into the molecular determinants of clinical outcomes (26, 27). Having carefully characterized the patient cohort, we then performed a more detailed molecular analysis on 10 patients who had either exceptionally favorable (Supplementary Fig. S18) or unexpectedly poor clinical outcomes (see Materials and Methods).
We performed whole-exome sequencing to investigate further genomic aberrations in the 10 outlier cases. One thousand one hundred ninety-four high confidence variants were identified overall (Supplementary Table S7). There was no difference in the median mutational rates in favorable or unfavorable groups (2.0: range 1.0–3.0 and 2.1: range 1.5–3.9 mutations per Mb, respectively; Fig. 4A). Seventy-nine likely pathogenic variants were identified in 65 genes among the 10 outlier patients. Reflective of the quality of the exome data, all but two of the variants detected by targeted sequencing were also detected by whole-exome sequencing (Fig. 4B; Supplementary Table S8), with a common hotspot mutation in PIK3CA, p.E542K, only found by targeted sequencing due to poor sequence coverage at this locus in the whole-exome data.
Mutational signatures of clinical outliers. A, Somatic mutation burden identified in tumors of clinical outliers by whole-exome sequence analysis. Survival category is shown under the sample names. B, TFI and OS for individual patients and predicted pathogenic mutations. See text for criteria defining given patients as having exceptional survival characteristics. C, Heat map showing differences of the contribution in mutational signatures. Survival category is shown to the left of sample names.
ARID1A, BRAF, and PIK3CA mutations occurred in both the exceptional and poor outcome groups, but no novel pathogenic mutations were common within either group. We noted a germline frameshift deletion mutation in POLE in one exceptional outlier. POLE somatic mutations have been associated with unusually favorable outcomes in other solid cancers, including glioblastoma and endometrial cancers (28, 29), and are characterized by very high mutational load with a distinct pattern of nucleotide change. However, we did not observe a pattern of genomic scarring in this patient consistent with functional inactivation of POLE. In addition to POLE, an in-frame deletion detected in ERBB3 by custom capture sequencing was also confirmed as a germline variant in one short survival patient.
We extended the analysis of DNA base changes to investigate signatures that are reflective of other mutational processes in cancer (30). Signatures 1 (age), 2/13 (APOBEC), 3 (homologous recombination), and 6/15/20/26 (mismatch repair defect) have been reported as common mutational signatures observed in OCCC in recent studies (31, 32). Similar to previous results, broadly similar patterns of mutational signatures associated with age, mismatch repair, and homologous recombination were observed in both the favorable and unfavorable outcome groups (Supplementary Fig. S19). We next compared differences of contribution scores across all samples. Interestingly, signatures 2 and 13 related with APOBEC were closely located by clustering analysis and tended to show high contribution with cases with unfavorable outcome compare to cases with favorable prognosis (Fig. 4C).
We investigated copy number alterations based on the ratio of read coverage between tumor and normal samples (Supplementary Materials and Methods). A similar pattern of copy number change was observed in the clinical outliers to that reported in previous studies (Supplementary Fig. S20A; refs. 23, 25, 33). Although the sample sizes were too small to be statistically meaningful, we determined whether specific regions of copy number change were associated with clinical groups. Wide copy number gain in whole chromosomal arm 8p was observed in two patients with the longest PFS (Supplementary Fig. S20B). Gain of 8p is rare in OCCC, however, amplification at 8p11-12 has been observed in 10% to 15% of human breast cancers (34). In contrast, long deletions in chromosome 9q, rarely observed in acute myeloid leukemia, were seen in three of seven resistant cases for first-line chemotherapy, but not detected in three sensitive cases (35).
Discussion
In a cohort of patients with Australian and Japanese OCCC, we found that superior PFS and OS was not due to pathologic misclassification of HGSOC as OCCC, or due to the presence of TP53 mutations and other driver mutations associated with HGSOC. We did not identify any specific driver mutations that could explain why a subset of patients with OCCC have better clinical outcomes following platinum-based chemotherapy. However, low-level expression of HNF1B and Ki67 appear to be associated with particularly favorable outcomes in patients with OCCC.
The approach to treat all patients with ovarian cancer with platinum-based therapy is based on the exquisite sensitivity of the dominant histotype, HGSOC (5). However, OCCC lack the molecular characteristics that make HGSOC particularly sensitive to platinum, namely frequent impairment of homologous recombination (HR) DNA repair and almost invariant TP53 mutations in HGSOC (7, 8). It is therefore unsurprising that the response rates to platinum-taxane chemotherapy are low in OCCC and typically less than 40% in patients with measureable disease in the first-line setting, and less than 5% in patients with recurrent OCCC (2, 4, 5). Regrettably, the factors that determine response in the patients with OCCC that derive benefit from platinum-based chemotherapy, and how best to identify such patients prospectively, are unknown.
OCCC are an uncommon histologic subtype and associated with a significantly worse 5-year disease-free survival compared with other histologic subtypes of ovarian cancer after adjusting for age, stage, and grade. The most important prognostic factor for survival is stage and patients with stage I OCCC have a 5-year survival of 85% (36). In contrast, in an analysis of 1,895 patients with advanced ovarian cancer treated in 6 Gynecologic Oncology Group (GOG) trials, the 62 patients with clear cell cancers (3.3%) had a median OS of 24 months compared with 45, and 56 months for serous, and endometrioid cell types, respectively (36). The relationship between volume of residual disease and prognosis in patients with advanced stage OCCC is uncertain as the majority of patients have no residual disease following surgery, but despite this still have a poor prognosis (37). In this study, we selected a high-risk subset of early-stage patients who had relapsed, and therefore did not show the usual association of early stage with favorable outcome.
In principle, primary surgical debulking and adjuvant chemotherapy can both contribute to the PFS and to OS in OCCC patients. Therefore, to remove as much as possible the confounding effect of surgery, we reviewed over 400 patients with OCCC to identify those with advanced stage disease and/or residual disease remaining after surgery in whom it was possible to evaluate survival benefit following chemotherapy based on PFS and where biospecimens were available. This allowed us to then carry out a detailed pathologic and molecular assessment and correlate the results with patient outcomes.
Our first question was a simple but clinically important one, namely were all patients with OCCC with good outcomes, in fact, misclassified HGSOC? If this was so, it would provide a way forward for identifying patients who were likely to benefit from adjuvant chemotherapy (or not). Additionally, the identification of misclassified HGSOC patients may impact on the subsequent referral and assessment of patients for high-risk germline mutations, given the frequency of BRCA1, BRCA2, and other HR pathway mutations in this histotype. Similar to recent studies (11, 18, 38), we found that in our series around 10% of tumors classified as OCCC at diagnosis were almost certainly HGSOC based on concordant IHC and molecular criteria. Although the presence of mixed histology is a strong indicator of potential misclassification, we found that even a small proportion of tumors with pure clear cell morphology can be misclassified when relying only on histologic features. As newly developed and validated IHC algorithms for scoring become widely adopted, it is expected that the rate of misclassification of ovarian tumors will reduce substantially (16).
Although the misclassified HGSOC were among the patients with better outcomes in the cohort, they did not account for all the patients who showed longer PFS and OS. Enhanced survival could not be simply explained by TP53 mutations, present in only five of 54 validated OCCC, or in mutations in other driver genes such as PIKC3A, KRAS, and ARID1A. Patients with glioblastoma with POLE mutations have shown longer survival in this highly aggressive cancer, and enhanced immunological responses have been observed in individuals with higher mutational burdens (28, 39, 40). Although these genes were included in the targeted sequencing panel, we considered whether patients with exceptional outcomes may harbor unusual germline or somatic mutations not evaluated with the capture-sequence panel. However, as observed with our more restricted analyses, complete exome sequence on a small number of unusually favorable and poor outcome patients failed to identify plausible drivers or mutational signatures associated with atypical outcomes.
HNF1B is a transcription factor correlated with the embryonic development of several organs, and has been reported to be associated with risk of several human cancers, including ovarian cancer (41). Overexpression of HNF1B is a useful diagnostic biomarker with high sensitivity and specificity for OCCC, particularly when used in conjunction with Napsin A (42–44). Surprisingly, we identified a small population of OCCC (14.8%) that were negative for HNF1B immunoreactivity and these had both a longer PFS and OS, including those with residual tumor at primary surgery. It has been suggested that chemo-resistance of OCCC might be due to aberrant retention of the G2 checkpoint through overexpression of HNF1B (45). Low HNF1B is associated with indolent behavior in renal chromophobe adenomas and biliary phenotype in patients with hepatocellular carcinoma (46, 47). OCCCs lacking HNF1B expression may represent a molecularly and clinically distinct population.
Several previous studies have reported that loss of ARID1A is associated with a poorer outcome, but other studies have not replicated this result (9, 13–15, 48). In the subgroup analyses restricted to cases with either no residual tumor at primary surgery or stage I disease, loss of ARID1A expression and/or ARID1A mutations were correlated with shorter PFS, indicating that tumors with ARID1A mutations have a more aggressive phenotype. This survival association was not observed in a subset analysis restricted to cases with residual tumor at primary surgery, perhaps because the majority of cases with residual disease also have loss of ARID1A (79%) compared with those with no residual disease (45%). Further studies are needed to clarify correlations between ARID1A and prognosis, although this is the first analysis in cases with no residual tumor followed by recurrence.
We also found that tumors with a high proportion of cells expressing the proliferative marker Ki67 were associated with shorter PFS, both in cases with no residual tumor and stage I disease. Ki67 staining has not previously been evaluated in a tightly defined clinical cohort of OCCC similar to ours, and the data on these markers in a wider series of patients with OCCC is conflicting (16, 38). Ki67 has been extensively evaluated in other solid cancers, particularly breast cancer where it is associated with poorer outcome (49, 50). This is in contrast to our recent study of exceptional survivors with HGSOC, in which we found high Ki67 was associated with long PFS and long-term survival (26, 27), possibly as tumors with high proliferative activity are more sensitive to chemotherapy. In intrinsically chemo-resistant OCCC, low Ki67 may be associated with longer PFS because such tumors are slow growing.
Although we ruled out some factors that could explain favorable outcomes in OCCC, our inability to clearly associate specific molecular drivers with outcome, particularly in those with more detailed exome analyses, present a conundrum. It is possible that disparate molecular factors converge on outcome, and that much larger numbers of patients are needed to discern a relationship between drivers and response. This is likely to be true for copy number events that involve large genomic regions that are more difficult to associate with outcome than point mutations in individual genes. Alternatively, other analyses may be required, including methylation, expression, or evaluation of non-coding sequences. OCCC molecularly resemble renal clear cell cancers, and both have been associated with favorable responses to immunotherapy. Although the patients in our series predated the use of immune checkpoint inhibitors and agents that target altered genes or pathways, it is possible that the extent of native innate and/or adaptive immune responses may have a dominant effect in determining the length of remission and OS in this cohort.
Disclosure of Potential Conflicts of Interest
M. Friedlander reports receiving speakers bureau honoraria from AstraZeneca and is a consultant/advisory board member for AstraZeneca, MSD, Lilly, and Takeda. D.D. L. Bowtell reports receiving commercial research grants from Roche Genentech and AstraZeneca. No potential conflicts of interest were disclosed by the other authors.
Authors' Contributions
Conception and design: M. Takenaka, S. Isonishi, A. Okamoto, M. Friedlander, D.D.L. Bowtell
Development of methodology: M. Takenaka, M. Köbel, S. Fereday, T. Kiyokawa, S. Isonishi, T. Semple, A. Okamoto, D.D.L. Bowtell
Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Takenaka, M. Köbel, S. Fereday, J. Hendley, A. Kawabata, D. Noguchi, N. Yanaihara, T. Kiyokawa, H. Takano, S. Isonishi, N. Traficante, S.R. Gadipally, T. Semple, D. Vassiliadis, G.M. Arnau
Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Takenaka, M. Köbel, D.W. Garsed, S. Fereday, A. Pandey, D. Etemadmoghadam, A. Kawabata, T. Kiyokawa, K. Christmalee Amarasinghe, J. Li, D.D.L. Bowtell
Writing, review, and/or revision of the manuscript: M. Takenaka, M. Köbel, D.W. Garsed, S. Fereday, D. Etemadmoghadam, J. Hendley, H. Takahashi, T. Kiyokawa, S. Isonishi, T. Semple, A. Okamoto, M. Friedlander, D.D.L. Bowtell
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Takenaka, S. Fereday, A. Pandey, J. Hendley, A. Kawabata, T. Kiyokawa, M. Ikegami, S. Isonishi, T. Semple, A. Okamoto
Study supervision: K. Ochiai, A. Okamoto, D.D.L. Bowtell
Acknowledgments
The AOCS gratefully acknowledges the cooperation of the participating institutions in Australia, and also acknowledges the study nurses, research assistants and all clinical and scientific collaborators including Misato Saito, Kazuya Sakurai, and Haruka Yanagisawa. The complete AOCS Group can be found at www.aocstudy.org. We would like to thank all of the women who participated in the study. This study was financially supported by the National Health and Medical Research Council of Australia (NHMRC) Program Grant APP1092856 “Improving outcomes for women with ovarian cancer” (to M. Friedlander and D.D.L. Bowtell), and the NHMRC Research Fellowship Grant APP1117044 “The molecular biology of human ovarian cancer” (to D.D.L. Bowtell). The AOCS was supported by the U.S. Army Medical Research and Materiel Command under DAMD17-01-1-0729, the Cancer Council Victoria, Queensland Cancer Fund, The Cancer Council New South Wales, The Cancer Council South Australia, The Cancer Council Tasmania, and The Cancer Foundation of Western Australia (Multi-State Applications 191, 211, and 182) and the NHMRC (ID400413 and ID400281). The AOCS gratefully acknowledges additional support from S. Boldeman, the Agar family, Ovarian Cancer Action (UK), Ovarian Cancer Australia, and the Peter MacCallum Foundation.
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Footnotes
Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).
Clin Cancer Res 2019;25:3962–73
- Received November 11, 2018.
- Revision received January 24, 2019.
- Accepted April 2, 2019.
- Published first April 9, 2019.
- ©2019 American Association for Cancer Research.