Abstract
Background
High-grade serous ovarian cancer (HGSOC) can be treated with platinum-based neoadjuvant chemotherapy (NACT) and delayed primary surgery (DPS). Histopathological response to NACT can be assessed using Böhm’s chemotherapy response score (CRS). We investigated whether germline BRCA1/2 (gBRCA1/2) genotype associated with omental CRS phenotype.
Methods
A retrospective study of patients with newly diagnosed FIGO stage IIIC/IV HGSOC prescribed NACT and tested for gBRCA1/2 pathogenic variants (PVs) between September 2017 and December 2022 at The Christie Hospital. The Cox proportional hazards model evaluated the association between survival and key clinical factors. The chi-square test assessed the association between CRS3 (no/minimal residual tumour) and gBRCA1/2 status.
Results
Of 586 eligible patients, 393 underwent DPS and had a CRS reported. Independent prognostic factors by multivariable analysis were gBRCA1/2 status (PV versus wild type [WT]), CRS (3 versus 1 + 2), surgical outcome (complete versus optimal/suboptimal) and first-line poly (ADP-ribose) polymerase-1/2 inhibitor maintenance therapy (yes versus no) (all P < 0.05). There was a non-significant trend for tumours with a gBRCA2 PV having CRS3 versus WT (odds ratio [OR] = 2.13, 95% confidence intervals [CI] 0.95–4.91; P = 0.0647). By contrast, tumours with a gBRCA1 PV were significantly less likely to have CRS3 than WT (OR = 0.35, 95%CI 0.14–0.91; P = 0.0291).
Conclusions
Germline BRCA1/2 genotype was not clearly associated with superior omental CRS. Further research is required to understand how HGSOC biology defines CRS.
Subject terms: Prognostic markers, Oncology
Background
Ovarian cancer is the second commonest cause of gynaecological cancer-related death worldwide, with more than 200,000 deaths attributed to the disease each year [1]. The most common histological subtype is high-grade serous ovarian cancer (HGSOC), which accounts for approximately 70% of all malignant tumours of the ovary [2]. The predilection of HGSOC to metastasise early to the peritoneal surfaces means most women are diagnosed with advanced-stage disease and cure is unlikely. Advanced HGSOC can be treated with neoadjuvant chemotherapy (NACT) followed by delayed primary surgery (DPS) [3–6]. A NACT plus DPS approach provides an opportunity to assess a tumour’s histopathological response to systemic anticancer therapy. However, there is no universally accepted scoring system to define the pathological response of HGSOC to NACT [7–14]. Moreover, the biology that defines histopathological responses to NACT in HGSOC is unknown. Consequently, all women treated with NACT plus DPS are offered the same platinum-containing chemotherapy in the immediate postoperative setting, potentially missing an opportunity to personalise therapy [15].
The genomic hallmarks of HGSOC are near-ubiquitous somatic TP53 mutations, germline and somatic BRCA1/2 mutations, homologous recombination deficiency (HRD) and genomic instability [16–20]. Germline BRCA1/2 mutations occur in 10–20% of HGSOCs and are now routinely tested for in all newly diagnosed women [21]. Epithelial ovarian cancers harbouring a germline BRCA1/2 mutation are highly sensitive to platinum-containing chemotherapy [22–25]. The putative mechanism for this platinum sensitivity is somatic loss (genetic or epigenetic) of the remaining BRCA1/2 wild type allele, leading to a biallelic loss-of-function, a deficiency in homologous recombination, and an inability to repair lethal DNA double-strand breaks caused by platinum chemotherapy [26]. We hypothesised that because epithelial ovarian cancers containing a germline BRCA1/2 mutation often have a better radiological response to platinum-containing chemotherapy compared to tumours with germline BRCA1/2 wild type, they would also have a better histopathological response to NACT. To investigate this, we used the Böhm scoring system, which defines histopathological responses of HGSOC to NACT by examining omental tissue resected during DPS [12]. Tumours with a complete/near-complete response to NACT (i.e., no/minimal residual omental tumour) have a chemotherapy response score (CRS) of 3, while tumours with no/minimal response to NACT (i.e., viable residual omental tumour) have a CRS of 1 [12]. We hypothesised that HGSOCs harbouring a germline BRCA1/2 mutation were more likely to have CRS3 than those with germline BRCA1/2 wild type.
Methods
Patient cohort
A retrospective study in which eligible patients included those with newly diagnosed FIGO (International Federation of Gynaecology and Obstetrics) stage IIIC or IV [27] ovarian, fallopian tube or primary peritoneal cancer that were prescribed NACT at The Christie NHS Foundation Trust (Manchester, United Kingdom). All patients had histologically confirmed HGSOC and had been tested for germline BRCA1/2 variants by the North West Genomic Laboratory Hub between 1st September 2017 and 31st December 2022. The decision to proceed to DPS after NACT was made by local multidisciplinary teams. All patients who received at least one dose of NACT were included in the overall population. All patients who underwent DPS were included in the DPS group.
BRCA1/2 testing
All patients provided informed consent for germline BRCA1/2 testing. The next generation sequencing (NGS) and multiplex ligation-dependent probe amplification (MLPA) assays used in the North West Genomic Laboratory Hub have been reported previously [28]. Germline testing was performed on DNA extracted from peripheral blood lymphocytes. The NGS assay tested the coding regions of BRCA1 and BRCA2 plus 15 base pairs either side of each exon-intron boundary. Target coverage was at least 90% at a read depth of 100X. Small sequencing variants (<40 base pairs) were reported with a variant allele fraction ≥5%. Copy number variants including whole gene/exon deletions/duplications were detected using MLPA. Only pathogenic (class 5) and likely pathogenic (class 4) variants were reported (hereafter described as ‘pathogenic variants’ or ‘PVs’) [29].
Efficacy outcomes
Chemotherapy response scores were determined by gynaecological Histopathologists by examining omental tissue removed at DPS using the Böhm scoring system [12]. Reporting Histopathologists were based at two tertiary referral centres, The Christie NHS Foundation Trust and Manchester University NHS Foundation Trust (Manchester, United Kingdom). The outcome of cytoreductive surgery was categorised as complete (no macroscopically visible tumour), optimal (1–10 mm of residual disease) or suboptimal (>10 mm of residual disease) [30]. Efficacy outcomes included progression-free survival (PFS), time to first subsequent therapy (TFST) and overall survival (OS). Progression-free survival was defined as the time interval from cycle 1 day 1 of NACT to the date of clinical or radiological progression or death, whichever occurred first. Time to first subsequent therapy was defined as the time interval from cycle 1 day 1 of NACT to the start of the subsequent therapy or death, whichever occurred first. Overall survival was defined as the time interval from cycle 1 day 1 of NACT to the date of death. The final date of data cut-off was 8th January 2024 to allow a minimum of 12 months follow-up from cycle 1 day 1 of NACT in all patients. The GCIG CA 125 criteria was used to define response to NACT but not progression [31]. The GCIG CA 125 response was determined by comparing the pre-treatment serum CA 125 value with the pre-surgical serum CA 125 value. Only patients with a pre-treatment serum CA 125 value twice the upper limit of normal (range 0–30 U/mL) were eligible for GCIG CA 125 response assessment. The CA 125 ELIMination Rate Constant K (KELIM) was also used to define response to NACT [32]. It was calculated using the same model as implemented on the online calculator (https://www.biomarker-kinetics.org/) [33]. Only those patients with at least three CA 125 values measured within the first 100 days (or less) after starting NACT were eligible for KELIM.
Statistical analysis
The chi-square test was used to determine whether there was an association between omental CRS3 (no/minimal residual tumour) and germline BRCA1/2 status. Odds ratios (ORs) and corresponding 95% confidence intervals (95% CI) and P values were calculated. Kaplan–Meier statistics were used to estimate median survival times and corresponding 95% CI. Univariable and multivariable Cox proportional hazards regression was applied to evaluate the association between survival outcomes and key clinical factors in the overall population and DPS group. Hazard ratios (HRs) together with their corresponding 95% CI and P values were calculated. All reported P values were two-sided. Statistical analyses were performed with R version 4.3.1.
The study was approved by the Quality Improvement & Clinical Audit Committee at The Christie NHS Foundation Trust. The study was performed in line with the principles of the Declaration of Helsinki.
Results
Overall population
In total, 586 eligible women were identified (Fig. 1). Of these, 581 patients received at least one dose of NACT and were included in the overall population (Supplementary Table S1). There were 77 (13%) germline BRCA1/2 PVs detected (40 BRCA1, 37 BRCA2) in 77 women (Supplementary Data). The median PFS, TFST and OS for the overall population were 14.3 (95% CI 13.5–15.3; 474 events), 16.1 (95%CI 15.4–17.1; 462 events) and 35.7 months (95% CI 33.4–40.1; 337 events), respectively (Supplementary Fig. S1). The median follow-up for patients alive at the date of data cut-off was 30.1 months (interquartile range 19.8–34.9).
Fig. 1. CONSORT diagram.
Key: C1 cycle 1, CT computed tomography, CRS chemotherapy response score, DPS delayed primary surgery, NACT neoadjuvant chemotherapy.
Univariable and multivariable analyses showed that ECOG performance status (0–1 versus 2–4), germline BRCA1/2 status (PV versus wild type), cytoreductive surgery (yes versus no) and use of first-line poly (ADP-ribose) polymerase-1/2 inhibitor (PARPi) maintenance therapy (yes versus no) were prognostic factors in the overall population (Supplementary Tables S2 and S3). The median PFS was significantly longer in patients who received first-line maintenance therapy versus those who underwent surveillance (Supplementary Table S4). Taken together, these observations validated our overall population dataset and supported the next stage of evaluating omental CRS as a prognostic factor in women undergoing DPS.
Delayed primary surgery group
Of the 581 patients who received at least one dose of NACT, 402 (69%) underwent DPS and 393 (98%) had an omental CRS reported (Supplementary Table S5). The 9 patients without a documented CRS had no evidence of neoplasia or chemotherapy-related regression changes in the omentum (n = 6) or no CRS reported (n = 3). Of the 179 patients who did not undergo DPS, the commonest reason for not undergoing debulking surgery was the extent of residual disease seen radiologically following NACT (n = 99 [55%]) (Fig. 1). Of these 99 patients, 57 (58%) had FIGO stage IV disease.
Of the 393 patients in the DPS group with a CRS reported, 336 (85%) received 3–4 cycles of NACT and 246 (63%) had complete debulking (Table 1). The proportion of patients who received 3–4 cycles of NACT in the CRS3 versus CRS1 + 2 group did not differ significantly (114/131 [87%] versus 222/262 [85%], respectively; P = 0.60) (Table 1). Fifty-nine women received pre-operative bevacizumab (Table 1). There was no significant difference between the proportion of patients who received bevacizumab prior to DPS in the CRS3 versus CRS1 + 2 group (19/131 [15%] versus 40/262 [15%], respectively; P = 0.84) (Table 1). However, patients with CRS3 were more likely to achieve complete debulking than those with CRS1 + 2 (108/131 [82%] versus 138/262 [53%], respectively; P < 0.0001) (Table 1). Moreover, patients with CRS3 were more likely to have a GCIG CA 125 response (120/121 [99%] versus 229/246 [93%], respectively; P = 0.0085) or KELIM CA 125 favourable score (78/109 [72%] versus 112/218 [51%], respectively; P = 0.0005) than those with CRS1 + 2 (Table 2 and Supplementary Table S6). The median PFS, TFST and OS were significantly longer in the CRS3 group compared to the CRS1 + 2 group (Table 2, Fig. 2 and Supplementary Table S7).
Table 1.
Demographic data for delayed primary surgery group.
| CRS1 + 2 | CRS3 | |
|---|---|---|
| 262 patients | 131 patients | |
| Age at diagnosis/years | ||
| Median (range) | 63 (37–83) | 67 (36–86) |
| ECOG performance status | ||
| 0–1 | 218 (83%) | 109 (83%) |
| 2–4 | 44 (17%) | 22 (17%) |
| FIGO stage | ||
| IIIC | 180 (69%) | 96 (73%) |
| IVA | 30 (11%) | 9 (7%) |
| IVB | 52 (20%) | 26 (20%) |
| Germline BRCA1/2 status | ||
| Pathogenic variant | 40 (15%) | 18 (14%) |
| Wild type | 222 (85%) | 113 (86%) |
| Neoadjuvant chemotherapy | ||
| Carboplatin-Paclitaxel | 256 | 126 |
| 3-weekly | 202 (77%) | 96 (73%) |
| Weekly | 54 (21%) | 30 (23%) |
| Carboplatin-Caelyx | 4 (2%) | 0 |
| Carboplatin | 2 (<1%) | 5 (4%) |
| Cycles of neoadjuvant chemotherapy | ||
| <3 | 1 (<1%) | 0 |
| 3-4 | 222 (85%) | 114 (87%) |
| >4 | 39 (15%) | 17 (13%) |
| Median (range) | 4 (2–6) | 4 (3–6) |
| Pre-operative bevacizumab | ||
| Yes | 40 (15%) | 19 (15%) |
| Median (range) | 3 (1–6) | 3 (2–4) |
| Hyperthermic Intraperitoneal Chemotherapy | ||
| Yes | 15 (6%) | 6 (2%) |
| Surgical outcome | ||
| Complete | 138 (53%) | 108 (82%) |
| Optimal | 102 (39%) | 20 (15%) |
| Suboptimal | 22 (8%) | 3 (2%) |
| Total cycles of first-line chemotherapy | ||
| <6 | 17 (6%) | 5 (4%) |
| ≥6 | 245 (94%) | 126 (96%) |
| Median (range) | 6 (2–8) | 6 (3–8) |
| First-line maintenance therapy | ||
| None | 73 (28%) | 45 (34%) |
| Bevacizumab | 90 (34%) | 40 (31%) |
| Olaparib | 15 (6%) | 13 (10%) |
| Niraparib | 52 (20%) | 23 (18%) |
| Bevacizumab and olaparib | 32 (12%) | 10 (8%) |
DPS delayed primary surgery, ECOG Eastern Cooperative Oncology Group, FIGO International Federation of Gynaecology and Obstetrics.
Table 2.
Efficacy outcomes in delayed primary surgery group.
| CRS1 + 2 | CRS3 | Hazard ratio (95% CI) | P value | |
|---|---|---|---|---|
| 262 patients | 131 patients | |||
| GCIG CA 125 response – number (%) | ||||
| Eligible | 247 (94%) | 121 (92%) | – | – |
| CA 125 Response | 229 (93%) | 120 (99%) | – | 0.0085 |
| No CA 125 response | 18 (7%) | 1 (<1%) | – | |
| CA 125 KELIM – number (%) | ||||
| Eligible | 218 (83%) | 109 (83%) | – | – |
| Favourable score (≥1.0) | 112 (51%) | 78 (72%) | – | 0.0005 |
| Unfavourable score (<1.0) | 106 (49%) | 31 (28%) | – | |
| Progression-free survival | ||||
| Events – number (%) | 218 (83%) | 80 (61%) | - | – |
| Median (95% CI) – months | 14.8 (13.9–15.9) | 24.4 (21.9–31.9) | 2.45 (1.9–3.2) | <0.0001 |
| Time to first subsequent therapy | ||||
| Events – number (%) | 210 (80%) | 80 (61%) | – | – |
| Median (95% CI) – months | 16.4 (15.7–17.8) | 26.81 (23.2–34.4) | 2.42 (1.9–3.2) | <0.0001 |
| Overall survival | ||||
| Events – number (%) | 148 (56%) | 47 (36%) | – | – |
| Median (95% CI) – months | 38.0 (33.1–42.7) | 58.8 (50.1–NR) | 2.28 (1.6–3.2) | <0.0001 |
95% CI 95% confidence interval, CRS chemotherapy response score, DPS delayed primary surgery, GCIG Gynecologic Cancer Intergroup, KELIM ELIMination Rate Constant K.
Fig. 2. Kaplan–Meier curves for progression-free survival (PFS) and overall survival (OS) in CRS groups.
a, c show Kaplan–Meier curves for PFS (a) and OS (c) for CRS1 versus CRS2 versus CRS3; b, d show Kaplan–Meier curves for PFS (b) and OS (d) for CRS1+2 versus CRS3.
Univariable and multivariable analyses showed that germline BRCA1/2 status (PV versus wild type), surgical outcome (complete versus optimal/suboptimal debulking), CRS (3 versus 1 + 2) and use of first-line PARPi maintenance therapy (yes versus no) were prognostic factors in the DPS group (Table 3 and Supplementary Table S8). These observations validated our DPS group dataset and supported the next stage of evaluating the association between omental CRS3 and germline BRCA1/2 status.
Table 3.
Multivariable analysis of delayed primary surgery group.
| Progression-free survival | Time to first subsequent therapy | Overall survival | |||||||
|---|---|---|---|---|---|---|---|---|---|
| HR | 95% CI | P value | HR | 95% CI | P value | HR | 95% CI | P value | |
| Age at diagnosis | 1.00 | 0.98–1.01 | 0.6925 | 1.00 | 0.98–1.01 | 0.6069 | 1.00 | 0.99–1.02 | 0.7802 |
| ECOG performance status | |||||||||
| 0–1 (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| 2–4 | 1.25 | 0.92–1.69 | 0.1603 | 1.22 | 0.89–1.67 | 0.2083 | 1.40 | 0.95–2.05 | 0.0858 |
| FIGO stage | |||||||||
| IIIC (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| IV | 1.27 | 0.99–1.63 | 0.0572 | 1.27 | 0.99–1.64 | 0.0614 | 1.22 | 0.89–1.66 | 0.2161 |
| Germline BRCA1/2 status | |||||||||
| Pathogenic variant (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| Wild type | 1.44 | 1.01–2.07 | 0.0447 | 1.47 | 1.02–2.13 | 0.0398 | 1.70 | 1.08–2.68 | 0.0223 |
| Number of cycles of NACT | |||||||||
| 3–4 (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| >4 | 0.84 | 0.60–1.18 | 0.3109 | 0.92 | 0.65–1.30 | 0.6351 | 0.91 | 0.60–1.38 | 0.6499 |
| Surgical outcome | |||||||||
| Complete (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| Optimal/suboptimal | 1.74 | 1.35–2.23 | <0.0001 | 1.79 | 1.39–2.31 | <0.0001 | 1.60 | 1.18–2.17 | 0.0026 |
| Chemotherapy response score | |||||||||
| 3 (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| 1 + 2 | 2.34 | 1.77–3.09 | <0.0001 | 2.27 | 1.71–3.01 | <0.0001 | 2.08 | 1.45–2.97 | 0.0001 |
| First–line PARPi therapy | |||||||||
| Yes (Ref) | 1.00 | – | – | 1.00 | – | – | 1.00 | – | – |
| No | 2.35 | 1.79–3.09 | <0.0001 | 2.37 | 1.79–3.14 | <0.0001 | 1.64 | 1.07–2.51 | 0.0231 |
95% CI 95% confidence interval, DPS delayed primary surgery, ECOG Eastern Cooperative Oncology Group, HR hazard ratio, NACT neoadjuvant chemotherapy, PARPi poly (ADP-ribose) polymerase-1/2 inhibitor.
Germline BRCA1/2 status and chemotherapy response score
In the 393 patients in the DPS group with an omental CRS reported, there was no significant difference between the proportion of tumours with CRS3 in the germline BRCA1/2 PV group versus the germline BRCA1/2 wild type group (OR 0.88, 95% CI 0.49–1.63; P = 0.6875) (Table 4). When the subgroups of women with a germline BRCA1 and BRCA2 PV were analysed separately, there was a trend towards a significantly greater proportion of tumours with CRS3 in the germline BRCA2 PV subgroup compared to the germline BRCA1/2 wild type group (OR 2.13, 95% CI 0.95–4.91; P = 0.0647) (Table 4 and Fig. 3). In contrast, there was a significantly smaller proportion of tumours with CRS3 in the germline BRCA1 PV subgroup compared to the germline BRCA1/2 wild type group (OR 0.35, 95% CI 0.14–0.91; P = 0.0291) (Table 4 and Fig. 3).
Table 4.
Association between chemotherapy response score and germline BRCA1/2 (gBRCA1/2) status.
| Group | CRS1 + 2 | CRS3 | Odds ratio (95% CI) | P value |
|---|---|---|---|---|
| Wild type | 222 (66%) | 113 (34%) | – | – |
| gBRCA1 | 28 (85%) | 5 (15%) | 0.35 (0.14–0.91) | 0.0291 |
| gBRCA2 | 12 (48%) | 13 (52%) | 2.13 (0.95–4.91) | 0.0647 |
| gBRCA1/2 | 40 (69%) | 18 (31%) | 0.88 (0.49–1.63) | 0.6875 |
95% CI 95% confidence interval, CRS chemotherapy response score.
Fig. 3.
Chemotherapy response score reported for patients with a germline BRCA1/2 pathogenic variant.
Discussion
A key question in HGSOC research is what is the biology that drives histopathological responses to NACT. Improving our understanding of the biology that underpins responses to NACT may help optimise the use of postoperative therapies. At present, the standard approach for women with newly diagnosed, advanced-stage HGSOC, who are treated with NACT and DPS, is to offer the same platinum-based chemotherapy postoperatively that was used preoperatively, regardless of how well the tumour had responded histopathologically [15]. This treatment strategy misses a potential opportunity to develop personalised postoperative therapies through improved understanding of the biology that underpins a tumour’s histopathological response to NACT.
Epithelial ovarian cancers that harbour a germline BRCA1/2 PV respond better radiologically to platinum-based chemotherapy compared to those with germline BRCA1/2 wild type [23]. Moreover, women with a germline BRCA1/2 PV have longer survival outcomes than those with germline BRCA1/2 wild type, largely due to enhanced primary platinum sensitivity and retained platinum sensitivity in the relapsed setting [22, 24, 25]. In their original study, Böhm and colleagues reported that omental CRS3 had a negative predictive value of 94.6% for primary platinum resistance, implying that HGSOCs with CRS3 were highly unlikely to develop primary platinum-resistant disease [12]. Taken together, these observations suggest that one factor underpinning histopathological responses to NACT in HGSOC could be germline BRCA1/2 status. Thus, we hypothesised that tumours with CRS3 were more likely to have a germline BRCA1/2 PV compared to tumours with germline BRCA1/2 wild type.
To evaluate an association between omental CRS phenotype and germline BRCA1/2 genotype, we firstly confirmed that key clinical factors in our overall population and DPS group were prognostically reproducible. Here, we showed that undergoing cytoreductive surgery and achieving complete debulking were prognostic in our overall population and DPS group. Moreover, we showed that having CRS3 or a germline BRCA1/2 PV were independent prognostic factors in our DPS group, regardless of the use of first-line PARPi maintenance therapy. These observations provided us with confidence that data from our cohort aligned with data reported in prospective ovarian cancer cohorts [30, 34–39]. Following this validation exercise, we were able to show a trend towards omental CRS3 in tumours harbouring a germline BRCA2 PV compared to tumours with germline BRCA1/2 wild type. In contrast, and surprisingly, we found that tumours with CRS3 were less likely to have a germline BRCA1 PV than those with germline BRCA1/2 wild type.
It is unclear why we found no clear association between germline BRCA1/2 genotype and CRS phenotype. It is notable that in their original study, Böhm and colleagues did not use BRCA1/2 status to develop the CRS, because germline BRCA1/2 status was not known in most women included in their discovery or validation cohorts [12]. Thus, it is plausible that the histopathological changes seen in the omentum that define CRS, including the fibro-inflammatory response and extent of residual disease, are not biologically related to the presence of a germline BRCA1/2 PV. A multicentre, individual patient data meta-analysis performed by The HGSC CRS Collaborative Network did report a positive associated between tumours with CRS3 and a germline BRCA1/2 PV (P = 0.027) [40]. However, in this meta-analysis only 306 patients (35%) out of 807 included had known germline BRCA1/2 status. Moreover, 80/306 (26%) had a germline BRCA1/2 PV and 33/80 (41%) had CRS3. The prevalence of germline BRCA1/2 PVs in this meta-analysis was higher than expected in an unselected population of women with ovarian cancer [41]. Thus, by unavoidably including a disproportionally high number of germline BRCA1/2 heterozygotes, the meta-analysis may have overestimated the association between CRS3 and germline BRCA1/2 status [40]. Nonetheless, the lack of a clear association between CRS3 and germline BRCA1/2 status in our study, and other smaller observational studies [42–50], implies that CRS biology cannot be explained using the current dogma linking BRCA1/2 status and platinum sensitivity in HGSOC. To deconvolute the complexity of CRS biology, detailed analysis of tumour-intrinsic factors and the tumour immune microenvironment is required, using spatial multiomic platforms and next-generation high-dimensional tissue imaging. Indeed, detailed understanding of the biology of the residual omental tumour and/or the tumour immune microenvironment in patients with CRS1 + 2 may lead to optimisation of post-operative treatments through personalised medicine.
What is evident from our study is that a large number of tumours with CRS3 have germline BRCA1/2 wild type. These tumours may have a BRCA1/2 wild type/HRD-positive genotype, although this remains unknown as routine genomic instability score (GIS) testing was not available during most of the study period [51]. Indeed, tumour HRD/GIS testing was only performed on 215/586 (37%) patients in the overall population (151/402 [38%] patients in the DPS group) because it only became available as a standard test in the North West of England in April 2021. Thus, further research is required to determine if omental CRS3 associates with tumour HRD/GIS status [52, 53]. Nevertheless, our study does validate CRS3 as an independent prognostic factor for primary platinum efficacy. It is well established that HRD-positive tumours are highly sensitive to PARPi, and that PARPi efficacy is linked to primary platinum response [34–39]. All women with newly diagnosed, advanced-stage HGSOC now undergo routine tumour HRD testing to access first-line olaparib-bevacizumab maintenance therapy [37]. Tumours with better histopathological responses to NACT are more likely to fail HRD testing due to the paucity of residual viable tumour cells available for testing in the DPS specimen [52]. In this scenario, when tumour HRD testing fails, omental CRS3 may offer a surrogate biomarker of PARPi efficacy. Thus, we advocate prospectively investigating omental CRS3 as a surrogate biomarker of PARPi efficacy; the main advantage of CRS being that it is cost-free [12].
Our retrospective study has some limitations based on the study design, which may have led to unavoidable selection bias. Firstly, germline BRCA1/2 testing occurred over a five-year period, in which standard eligibility criteria for testing changed from relapsed, platinum-sensitive HGSOC to any patient diagnosed with HGSOC. As a result, not all women with newly diagnosed HGSOC that were treated at The Christie Hospital will have undergone germline BRCA1/2 testing between 2017 and 2022. Indeed, patients who did not undergo germline BRCA1/2 testing were not included in our study. Secondly, only those patients that underwent DPS could be included in our analysis of omental CRS phenotype versus germline BRCA1/2 genotype, meaning the conclusions drawn about HGSOC biology cannot be applied to patients who did not receive NACT or undergo DPS. Thirdly, the decision to proceed with DPS was made by two multidisciplinary teams working in isolation, thus there may have been subtle differences between patients chosen for upfront primary surgery versus DPS. Fourthly, histology reporting occurred across two gynaecology pathology laboratories, so there may have been interobserver variation in CRS amongst reporting histopathologists, although this was not assessed in the study. One way to mitigate against interobserver variability would be to include a centralised review panel of expert gynaecology pathologists and/or use a digital pathology tool. Finally, we have not included patients with somatic BRCA1/2 PVs in the analysis, largely because local testing showed that many of these mutations do not have variant allele frequencies consistent with biallelic loss-of-function [54]. Overall, however we were reassured by the fact that the Böhm scoring system was prognostic in our study, consistent with data reported by other observational studies [43, 45, 46, 48, 49, 55–64]. Moreover, median survival data in our overall population aligned with prospective trial data for patients with epithelial ovarian cancer treated with NACT [65].
To our knowledge, we report the largest observational study assessing the association between omental CRS and germline BRCA1/2 status in newly diagnosed, advanced-stage HGSOC. Unexpectedly, we found no evidence to show a clear association between established pathology (omental CRS) and genetic (germline BRCA1/2 status) biomarkers of platinum sensitivity in HGSOC. We also add to the growing evidence that the modified two-tier Böhm scoring system (CRS1 + 2 versus CRS3) is an independent prognostic biomarker in FIGO stage IIIC/IV HGSOC. Finally, we demonstrate that patients with CRS1 + 2 are less likely to have a GCIG CA 125 or KELIM CA 125 response to platinum-based NACT or achieve complete debulking surgery compared to those with CRS3.
Supplementary information
Acknowledgements
The data was presented at the European Society for Medical Oncology 2023 Congress (Annals of Oncology 2023;34 (Suppl 2): S527-S528).
Author contributions
Conceptualization: RDM; Methodology: RDM, XW; Formal Analysis: RDM, XW, AC; Data Curation: RDM, XW, BMB, LS, AC; Supervision: BY, DGRE, GCJ, SST; Writing - Original Draft: RDM, XW, AC, GCJ, SST; Writing - Review & Editing: RDM, XW, BMB, LS, AC, DN, JH, CM, ZS, SD, JS, BW-R, HS, GJB, ARC, RJE, BY, DGRE, GCJ, SST.
Funding
This work was funded by a generous gift to Ovarian Cancer Research Team at The Christie NHS Foundation Trust, Manchester, by the family of Mrs Margaret Evans from North Ferriby, East Yorkshire. Robert Morgan is supported by a National Institute for Health Research (NIHR) Clinical Lectureship (CL-2022-06-002). Bethany Barnes and Stephen Taylor are funded by a Cancer Research UK Programme Grant awarded to Stephen Taylor (C1422/A19842 and C1422/A31334); and the Medical Research Council (MR/X008088/1); with co-funding from the NIHR Manchester Biomedical Research Centre (NIHR203308); and the Cancer Research UK Manchester Centre (C147/A25254). The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care.
Data availability
The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials.
Competing interests
The authors declare no competing interests.
Ethics approval and consent to participate
The study was approved by the Quality Improvement & Clinical Audit Committee at The Christie NHS Foundation Trust. The Genetic Variants in Gynaecological Cancer database has been approved by The Christie NHS Foundation Trust. The study was performed in line with the principles of the Declaration of Helsinki. Informed consent was obtained from all subjects involved in the study.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
The online version contains supplementary material available at 10.1038/s41416-024-02874-6.
References
- 1.Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229–63. [DOI] [PubMed] [Google Scholar]
- 2.WHO Classification of Tumours of Female Reproductive Organs, 5th ed. Lyon, France: IARC Publications; 2020.
- 3.Vergote I, Trope 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. [DOI] [PubMed] [Google Scholar]
- 4.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. [DOI] [PubMed] [Google Scholar]
- 5.Fagotti A, Ferrandina G, Vizzielli G, Fanfani F, Gallotta V, Chiantera V, et al. Phase III randomised clinical trial comparing primary surgery versus neoadjuvant chemotherapy in advanced epithelial ovarian cancer with high tumour load (SCORPION trial): Final analysis of peri-operative outcome. Eur J Cancer. 2016;59:22–33. [DOI] [PubMed] [Google Scholar]
- 6.Onda T, Satoh T, Saito T, Kasamatsu T, Nakanishi T, Nakamura K, et al. Comparison of treatment invasiveness between upfront debulking surgery versus interval debulking surgery following neoadjuvant chemotherapy for stage III/IV ovarian, tubal, and peritoneal cancers in a phase III randomised trial: Japan Clinical Oncology Group Study JCOG0602. Eur J Cancer. 2016;64:22–31. [DOI] [PubMed] [Google Scholar]
- 7.Sassen S, Schmalfeldt B, Avril N, Kuhn W, Busch R, Hofler H, et al. Histopathologic assessment of tumor regression after neoadjuvant chemotherapy in advanced-stage ovarian cancer. Hum Pathol. 2007;38:926–34. [DOI] [PubMed] [Google Scholar]
- 8.Ferron JG, Uzan C, Rey A, Gouy S, Pautier P, Lhomme C, et al. Histological response is not a prognostic factor after neoadjuvant chemotherapy in advanced-stage ovarian cancer with no residual disease. Eur J Obstet Gynecol Reprod Biol. 2009;147:101–5. [DOI] [PubMed] [Google Scholar]
- 9.Samrao D, Wang D, Ough F, Lin YG, Liu S, Menesses T, et al. Histologic parameters predictive of disease outcome in women with advanced stage ovarian carcinoma treated with neoadjuvant chemotherapy. Transl Oncol. 2012;5:469–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Muraji M, Sudo T, Iwasaki S, Ueno S, Wakahashi S, Yamaguchi S, et al. Histopathology predicts clinical outcome in advanced epithelial ovarian cancer patients treated with neoadjuvant chemotherapy and debulking surgery. Gynecol Oncol. 2013;131:531–4. [DOI] [PubMed] [Google Scholar]
- 11.Petrillo M, Zannoni GF, Tortorella L, Pedone Anchora L, Salutari V, Ercoli A, et al. Prognostic role and predictors of complete pathologic response to neoadjuvant chemotherapy in primary unresectable ovarian cancer. Am J Obstet Gynecol. 2014;211:632. [DOI] [PubMed] [Google Scholar]
- 12.Bohm S, Faruqi A, Said I, Lockley M, Brockbank E, Jeyarajah A, et al. Chemotherapy Response Score: Development and Validation of a System to Quantify Histopathologic Response to Neoadjuvant Chemotherapy in Tubo-Ovarian High-Grade Serous Carcinoma. J Clin Oncol. 2015;33:2457–63. [DOI] [PubMed] [Google Scholar]
- 13.Ebata T, Yunokawa M, Yoshida H, Bun S, Shimoi T, Shimomura A, et al. The Prognostic Impact of the Pathological Response to Neoadjuvant Dose-Dense Therapy for Ovarian Carcinoma. Int J Gynecol Cancer. 2017;27:1850–5. [DOI] [PubMed] [Google Scholar]
- 14.Liang MI, Prendergast EN, Staples JN, Holschneider CH, Cohen JG, Cass I. Prognostic role of pathologic response and cytoreductive status at interval debulking surgery after neoadjuvant chemotherapy for advanced epithelial ovarian cancer. J Surg Oncol. 2019;120:779–85. [DOI] [PubMed] [Google Scholar]
- 15.Gonzalez-Martin A, Harter P, Leary A, Lorusso D, Miller RE, Pothuri B, et al. Newly diagnosed and relapsed epithelial ovarian cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2023;34:833–48. [DOI] [PubMed] [Google Scholar]
- 16.Ahmed AA, Etemadmoghadam D, Temple J, Lynch AG, Riad M, Sharma R, et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol. 2010;221:49–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15. [DOI] [PMC free article] [PubMed]
- 18.Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N, Sander C. Emerging landscape of oncogenic signatures across human cancers. Nat Genet. 2013;45:1127–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Huang KL, Mashl RJ, Wu Y, Ritter DI, Wang J, Oh C, et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell. 2018;173:355–370.e314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hauke J, Hahnen E, Schneider S, Reuss A, Richters L, Kommoss S, et al. Deleterious somatic variants in 473 consecutive individuals with ovarian cancer: results of the observational AGO-TR1 study (NCT02222883). J Med Genet. 2019;56:574–80. [DOI] [PubMed] [Google Scholar]
- 21.Morgan, RD, Burghel, GJ, Flaum, N, Schlecht, H, Clamp, AR, Hasan, J, et al. Extended panel testing in ovarian cancer reveals BRIP1 as the third most important predisposition gene. Genet Med. 2024;26:101230. [DOI] [PubMed]
- 22.Tan DS, Rothermundt C, Thomas K, Bancroft E, Eeles R, Shanley S, et al. BRCAness” syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530–6. [DOI] [PubMed] [Google Scholar]
- 23.Vencken P, Kriege M, Hoogwerf D, Beugelink S, van der Burg MEL, Hooning MJ, et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol. 2011;22:1346–52. [DOI] [PubMed] [Google Scholar]
- 24.Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol. 2012;30:2654–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Norquist BM, Brady MF, Harrell MI, Walsh T, Lee MK, Gulsuner S, et al. Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study. Clin Cancer Res. 2018;24:777–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Maxwell KN, Wubbenhorst B, Wenz BM, De Sloover D, Pluta J, Emery L, et al. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers. Nat Commun. 2017;8:319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Prat J, Oncology, F. C. O. G. Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet. 2014;124:1–5. [DOI] [PubMed] [Google Scholar]
- 28.Morgan RD, Burghel GJ, Flaum N, Bulman M, Clamp AR, Hasan J, et al. Prevalence of germline pathogenic BRCA1/2 variants in sequential epithelial ovarian cancer cases. J Med Genet. 2019;56:301–7. [DOI] [PubMed] [Google Scholar]
- 29.Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.du Bois A, Reuss A, Pujade-Lauraine E, Harter P, Ray-Coquard I, Pfisterer J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: a combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d’Investigateurs Nationaux Pour les Etudes des Cancers de l’Ovaire (GINECO). Cancer. 2009;115:1234–44. [DOI] [PubMed] [Google Scholar]
- 31.Rustin GJ, Vergote I, Eisenhauer E, Pujade-Lauraine E, Quinn M, Thigpen T, et al. Definitions for response and progression in ovarian cancer clinical trials incorporating RECIST 1.1 and CA 125 agreed by the Gynecological Cancer Intergroup (GCIG). Int J Gynecol Cancer. 2011;21:419–23. [DOI] [PubMed] [Google Scholar]
- 32.Colomban O, Tod M, Leary A, Ray-Coquard I, Lortholary A, Hardy-Bessard AC, et al. Early Modeled Longitudinal CA-125 Kinetics and Survival of Ovarian Cancer Patients: A GINECO AGO MRC CTU Study. Clin Cancer Res. 2019;25:5342–50. [DOI] [PubMed] [Google Scholar]
- 33.You B, Freyer G, Gonzalez-Martin A, Lheureux S, McNeish I, Penson RT, et al. The role of the tumor primary chemosensitivity relative to the success of the medical-surgical management in patients with advanced ovarian carcinomas. Cancer Treat Rev. 2021;100:102294. [DOI] [PubMed] [Google Scholar]
- 34.Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med. 2018;379:2495–505. [DOI] [PubMed] [Google Scholar]
- 35.Gonzalez-Martin A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med. 2019;381:2391–402. [DOI] [PubMed] [Google Scholar]
- 36.Coleman RL, Fleming GF, Brady MF, Swisher EM, Steffensen KD, Friedlander M, et al. Veliparib with First-Line Chemotherapy and as Maintenance Therapy in Ovarian Cancer. N Engl J Med. 2019;381:2403–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ray-Coquard I, Pautier P, Pignata S, Perol D, Gonzalez-Martin A, Berger R, et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med. 2019;381:2416–28. [DOI] [PubMed] [Google Scholar]
- 38.Monk BJ, Parkinson C, Lim MC, O’Malley DM, Oaknin A, Wilson MK, et al. A Randomized, Phase III Trial to Evaluate Rucaparib Monotherapy as Maintenance Treatment in Patients With Newly Diagnosed Ovarian Cancer (ATHENA-MONO/GOG-3020/ENGOT-ov45). J Clin Oncol. 2022;40:3952–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wu, X, Liu, J, Wang, J, Wang, L, Lin, Z, Wang, X, et al. Senaparib as first-line maintenance therapy in advanced ovarian cancer: a randomized phase 3 trial. Nat Med. 2024;30:1612–21. [DOI] [PubMed]
- 40.Cohen PA, Powell A, Bohm S, Gilks CB, Stewart CJR, Meniawy TM, et al. Pathological chemotherapy response score is prognostic in tubo-ovarian high-grade serous carcinoma: A systematic review and meta-analysis of individual patient data. Gynecol Oncol. 2019;154:441–8. [DOI] [PubMed] [Google Scholar]
- 41.Kurian AW, Ward KC, Howlader N, Deapen D, Hamilton AS, Mariotto A, et al. Genetic Testing and Results in a Population-Based Cohort of Breast Cancer Patients and Ovarian Cancer Patients. J Clin Oncol. 2019;37:1305–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Barrington DA, Felix AS, Owda R, Suarez AA, Cohen DW, Senter L, et al. Pathologic chemotherapy response score in epithelial ovarian cancer: Surgical, genetic, and survival considerations. Surg Oncol. 2020;34:40–45. [DOI] [PubMed] [Google Scholar]
- 43.Lawson BC, Euscher ED, Bassett RL, Liu J, Ramalingam P, Zhong Y, et al. A 3-Tier Chemotherapy Response Score for Ovarian/Fallopian Tube/Peritoneal High-grade Serous Carcinoma: Is it Clinically Relevant? Am J Surg Pathol. 2020;44:206–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lee YJ, Kim HS, Rim JH, Lee JY, Nam EJ, Kim SW, et al. Germline BRCA, chemotherapy response scores, and survival in the neoadjuvant treatment of ovarian cancer. BMC Cancer. 2020;20:185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhong Y, Liu J, Li X, Westin SN, Malpica A, Lawson BC, et al. A Modified 2 Tier Chemotherapy Response Score (CRS) and Other Histopathologic Features for Predicting Outcomes of Patients with Advanced Extrauterine High-Grade Serous Carcinoma after Neoadjuvant Chemotherapy. Cancers. 2021;13:704. [DOI] [PMC free article] [PubMed]
- 46.De Jong D, Otify M, Chen I, Jackson D, Jayasinghe K, Nugent D, et al. Survival and Chemosensitivity in Advanced High Grade Serous Epithelial Ovarian Cancer Patients with and without a BRCA Germline Mutation: More Evidence for Shifting the Paradigm towards Complete Surgical Cytoreduction. Medicina. 2022;58:1611. [DOI] [PMC free article] [PubMed]
- 47.Ergasti R, Marchetti C, Tudisco R, Iervolino A, Naldini A, Oliva R, et al. BRCA status and platinum sensitivity in advanced ovarian cancer according to Chemotherapy Response Score. Int J Gynecol Cancer. 2022;32:639–45. [DOI] [PubMed] [Google Scholar]
- 48.Marsh LA, Kim TH, Zhang M, Kubalanza K, Treece CL, Chase D, et al. Pathologic response to neoadjuvant chemotherapy in ovarian cancer and its association with outcome: A surrogate marker of survival. Gynecol Oncol. 2023;177:173–9. [DOI] [PubMed] [Google Scholar]
- 49.Rodolakis I, Liontos M, Pergialiotis V, Haidopoulos D, Kaparelou M, Efthimios Vlachos D, et al. Chemotherapy response score as a predictor of survival in ovarian cancer patients. Eur J Obstet Gynecol Reprod Biol. 2024;296:233–8. [DOI] [PubMed] [Google Scholar]
- 50.Lee, YJ, Shin, YK, Kim, NR, Kim, SI, Lee, YY, Park, JY et al. Chemotherapy response score no longer predicts survival outcomes in high-grade serous ovarian cancer patients with BRCA mutation and/or maintenance therapy. J Gynecol Oncol. 2024. 10.3802/jgo.2024.35.e73. [DOI] [PMC free article] [PubMed]
- 51.Morgan RD, Clamp AR, Barnes BM, Timms K, Schlecht H, Yarram-Smith L, et al. Homologous recombination deficiency in newly diagnosed FIGO stage III/IV high-grade epithelial ovarian cancer: a multi-national observational study. Int J Gynecol Cancer. 2023;33:1253–9. [DOI] [PubMed] [Google Scholar]
- 52.Zalaznick H, Clegg B, Cogan E, Perry M, Trost J, Mancini-DiNardo D, et al. Rates of homologous recombination deficiency across different subtypes of ovarian cancer and in pre- and post-neoadjuvant chemotherapy tumor samples. Gynecol Oncol. 2022;166:S86–S87. [Google Scholar]
- 53.Marchetti, C, Ergasti, R, Capomacchia, FM, Giannarelli, D, Sassu, CM, Zannoni, GF, et al. The role of Chemotherapy Response Score (CRS) in the decision making process for patients with advanced high grade serous ovarian cancer undergoing neoadjuvant chemotherapy. J Clin Oncol. 2023;41. https://ascopubs.org/doi/10.1200/JCO.2023.41.16_suppl.5591.
- 54.Morgan RD, Burghel GJ, Flaum N, Bulman M, Smith P, Clamp AR, et al. Predicting the likelihood of a BRCA1/2 pathogenic variant being somatic by testing only tumour DNA in non-mucinous high-grade epithelial ovarian cancer. J Clin Pathol. 2023;76:684–9. [DOI] [PubMed] [Google Scholar]
- 55.Coghlan E, Meniawy TM, Munro A, Bulsara M, Stewart CJ, Tan A, et al. Prognostic Role of Histological Tumor Regression in Patients Receiving Neoadjuvant Chemotherapy for High-Grade Serous Tubo-ovarian Carcinoma. Int J Gynecol Cancer. 2017;27:708–13. [DOI] [PubMed] [Google Scholar]
- 56.Lee JY, Chung YS, Na K, Kim HM, Park CK, Nam EJ, et al. External validation of chemotherapy response score system for histopathological assessment of tumor regression after neoadjuvant chemotherapy in tubo-ovarian high-grade serous carcinoma. J Gynecol Oncol. 2017;28:e73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Said I, Bohm S, Beasley J, Ellery P, Faruqi AZ, Ganesan R, et al. The Chemotherapy Response Score (CRS): Interobserver Reproducibility in a Simple and Prognostically Relevant System for Reporting the Histologic Response to Neoadjuvant Chemotherapy in Tuboovarian High-grade Serous Carcinoma. Int J Gynecol Pathol. 2017;36:172–9. [DOI] [PubMed] [Google Scholar]
- 58.Michaan N, Chong WY, Han NY, Lim MC, Park SY. Prognostic Value of Pathologic Chemotherapy Response Score in Patients With Ovarian Cancer After Neoadjuvant Chemotherapy. Int J Gynecol Cancer. 2018;28:1676–82. [DOI] [PubMed] [Google Scholar]
- 59.Rajkumar S, Polson A, Nath R, Lane G, Sayasneh A, Jakes A, et al. Prognostic implications of histological tumor regression (Bohm’s score) in patients receiving neoadjuvant chemotherapy for high grade serous tubal & ovarian carcinoma. Gynecol Oncol. 2018;151:264–8. [DOI] [PubMed] [Google Scholar]
- 60.Singh P, Kaushal V, Rai B, Rajwanshi A, Gupta N, Dey P, et al. The chemotherapy response score is a useful histological predictor of prognosis in high-grade serous carcinoma. Histopathology. 2018;72:619–25. [DOI] [PubMed] [Google Scholar]
- 61.Ditzel HM, Strickland KC, Meserve EE, Stover E, Konstantinopoulos PA, Matulonis UA, et al. Assessment of a Chemotherapy Response Score (CRS) System for Tubo-Ovarian High-Grade Serous Carcinoma (HGSC). Int J Gynecol Pathol. 2019;38:230–40. [DOI] [PubMed] [Google Scholar]
- 62.Santoro A, Angelico G, Piermattei A, Inzani F, Valente M, Arciuolo D, et al. Pathological Chemotherapy Response Score in Patients Affected by High Grade Serous Ovarian Carcinoma: The Prognostic Role of Omental and Ovarian Residual Disease. Front Oncol. 2019;9:778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liang WF, Li H, Wu JY, Liu CH, Wu MF, Li J. Identification of Ovarian Cancer Patients Most Likely to Achieve Chemotherapy Response Score 3 Following Neoadjuvant Chemotherapy: Development of a Predictive Nomogram. Front Oncol. 2020;10:560888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Heayn M, Skvarca LB, Zhu L, Edwards RP, Olawaiye AB, Modugno F, et al. Impact of Ki-67 Labeling Index on Prognostic Significance of the Chemotherapy Response Score in Women With Tubo-ovarian Cancer Treated With Neoadjuvant Chemotherapy. Int J Gynecol Pathol. 2021;40:278–85. [DOI] [PubMed] [Google Scholar]
- 65.Morgan RD, McNeish IA, Cook AD, James EC, Lord R, Dark G, et al. Objective responses to first-line neoadjuvant carboplatin-paclitaxel regimens for ovarian, fallopian tube, or primary peritoneal carcinoma (ICON8): post-hoc exploratory analysis of a randomised, phase 3 trial. Lancet Oncol. 2021;22:277–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials.



