Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Oct 4.
Published in final edited form as: Int J Gynecol Pathol. 2017 May;36(3):240–252. doi: 10.1097/PGP.0000000000000325

Granulosa Cell Tumors: Novel Predictors of Recurrence in Early-stage Patients

Sharif Sakr 1, Eman Abdulfatah 1, Sumi Thomas 1, Zaid Al-wahab 1, Rafic Beydoun 1, Robert Morris 1, Rouba Ali-Fehmi 1, Sudeshna Bandyopadhyay 1
PMCID: PMC6171102  NIHMSID: NIHMS986170  PMID: 28727617

Summary:

Granulosa cell tumors (GCTs) comprise 2% to 5% of ovarian neoplasms, with unpredictable patterns of recurrence. The HER family, GATA4, and SMAD3 genes are reportedly involved in GCT proliferation and apoptosis and may serve as new predictors of recurrence. The aim of the study was to evaluate novel predictors of recurrence in GCT from a large single institution cohort. Patients diagnosed with GCTs (n = 125) between 1975 and 2014 were identified. Clinicopathologic parameters were obtained and immunohistochemical evaluation was performed of calretinin, inhibin, HER2, CD56, SMAD3, and GATA4. Statistical analyses were conducted using Fisher exact test and Kaplan-Meier survival curves and Cox regression analysis. The median follow-up period was 120 months (range, 1–465 mo). Recurrence was noted in 12/125 (9.6%) patients. Kaplan-Meier analysis showed a shorter mean disease-free interval in whites versus blacks (P = 0.001), stage III-IV versus stage I-II (P = 0.0001), patients treated with surgery+chemotherapy versus surgery (P = 0.0001), mitotic rate ≥4 (P = 0.005), severe nuclear pleomorphism (P = 0.013), high HER2 expression (P = 0.001), high CD56 expression (P = 0.001), and high SMAD3 expression (P = 0.001). On Cox regression analysis, SMAD3 and type of treatment received were the only 2 independent prognostic factors for disease-free interval (P = 0.03 and P = 0.007, respectively). On subanalysis for early-stage (stage I) GCTs, the need for adjuvant chemotherapy and high expression of SMAD3 continued to be independent predictors of recurrence (HR = 10.2, P = 0.01 and HR = 8.9, P = 0.001, respectively).

Keywords: Granulosa cell tumors, Prediction of recurrence, Immunohistochemistry


Ovarian granulosa cell tumors (GCTs) represent approximately 2% to 5% of all ovarian cancers. They are the most common type of ovarian sex-cord stromal tumors (1). GCTs are divided histologically into juvenile and adult forms. Adult granulosa cell tumor (AGCTs) account for 95% of GCTs with a median age at diagnosis of 50 to 55 years (2). GCTs have malignant potential but fortunately, the majority of patients present with stage I disease, for which the long-term disease-free survival is approximately 84% to 95% (3,4). Many of these patients are cured by surgery but insidious recurrence is still reported in up to 20% to 30%, even in early-stage disease (5,6). Because complete tumor resection is a significant prognostic factor (5,7) and because of the indolent nature of GCTs, routine staging has been traditionally recommended with the exception when there is a need for fertility-sparing surgery.

Although prognostic indicators have long been studied, predicting the likelihood of recurrence with certainty remains elusive; hence, postoperative management is usually based on stage of disease, completeness of tumor resection/presence of residual tumor, and whether fertility-sparing surgery is performed.

Chemotherapy has been used for extensive residual disease and inoperable metastases/recurrences (810). Choice of ideal candidates as well as standard postoperative adjuvant therapy is yet to be determined (11), hence the need for more accurate clinical and pathologic predictors of the clinical behavior of GCT.

Prognostic factors investigated can be divided into clinical as well as pathologic factors. Clinical prognostic factors include age (1,12,13), tumor size (1,12,14), stage of disease (1216), extent of surgery (3,15), and residual disease/tumor rupture (5,7). Pathologic factors include tumor histology (4,17), mitotic index (MI) (7,17,18), nuclear atypia (19), and expression of inhibin, epidermal growth factor (EGFR), and Ki67 (20,21).

With the recent diagnosis of the missense point mutation c402>G (codon C134W) in the gene coding for Forkhead Box L2 (FOXL2) transcription factor in 90% to 97% of GCTs (22), FOXL2 pathways were investigated with the aim of identifying new prognostic factors. Absence of and increased FOXL2 expression were described as poor prognostic factors (23,24). Aberrant granulosa cell proliferation and apoptosis have been recently implicated in the pathogenesis of GCTs secondary to interactions between C134-mutated FOXL2, SMAD3, and GATA4 (25). SMAD3, a transcription factor in the tumor growth factor-β (TGF-β) signaling pathway, activates NF-kB which increases cell viability in human GCT cells through a positive feedback loop (26). In murine cell models, SMAD3 interacts with the GATA4, suggesting the involvement of GATA4 in TGF-β signaling pathway as well (27).GATA4 is a zinc finger transcription factor that plays an important role in granulosa cell function and development (27,28), and has recently been associated with advanced stage GCT and increased recurrence risk (29) because of its antiapoptotic effect (30).

Increased expression of human epidermal growth factor receptor 2 (HER2), a transmembrane tyrosine kinase receptor, is prognostic of aggressive disease in breast, gastric, and epithelial ovarian cancer (31,32) with inconsistency in its expression in GCTs; positive in some studies (33,34) and negative in others (35,36).

CD56 is a membrane-bound cell surface sialoglycoprotein and a member of the immunoglobulin supergene family, which induces cell-to-cell interactions during embryonic development, cell migration, and organogenesis (37).

CD56 is expressed by most neuroectodermal-derived cell lines and different tumors and its expression is associated with poor prognosis in a subset of squamous cell carcinoma and renal cell carcinoma (38). CD56 expression in primordial follicles and theca cells was described to have a role in folliculogenesis (39). CD56 expression in GCTs was suggested to be secondary to acquisition of CD56 immunoreactivity during neoplastic transformation or because GCTs originate from theca cells rather than granulosa cells (40). Expression of CD56 was described in primary and recurrent GCT but not as a predictor of recurrence (41).

We report a review of patients diagnosed with GCTs from a single large institution with the aim of identifying new predictors of recurrence, which could aid in patient counseling and decision making postoperatively and providing more targeted therapy.

MATERIALS AND METHODS

After obtaining Institutional Review Board approval, patients diagnosed with GCTs (n = 125) between 1975 and 2014 were identified. Clinical parameters (age, race, body mass index, treatment, time to recurrence, and vital status) and pathologic variables (histologic type, tumor size, and stage) were obtained from electronic medical records and pathology records. Tumor stage was assigned according to the International Federation of Gynecology and Obstetrics (FIGO) criteria (42). Morphologic features including mitotic rate, nuclear pleomorphism (NP), and histologic patterns were evaluated from review of whole-tissue sections of each tumor. Mitotic rate was assessed by enumerating the number of mitotic figures in 10 consecutive HPFs (× 400, field diameter = 0.55 mm); <4/10 HPF is low and ≥4/10 HPF is high. Immunohistochemical (IHC) evaluation was performed on tissue microarrays for 88 primary GCT cases. Tissue microarrays were constructed from formalin-fixed, paraffin embedded blocks using a duplicate of 1-mm-diameter core obtained from a representative area of each tumor (43). The antibodies used were SMAD3 (Catalog no. 51–1500, 1:100 dilution; Invitrogen, Camarillo, CA), VEGF (sc-152, 1:100 dilution; Santa Cruz Biotechnology, Santa Cruz, CA), Her2 (sc-33684, 1:50–1:500 dilution; Santa Cruz Biotechnology), Inhibin (mcA951ST, 1:25 dilution; Serotec, Raleigh, NC), Calretinin (PA5–16681, dilution 1:100; Life Technologies, Chicago, IL), CD56 (123C307–5603, 1:100 dilution; Invitrogen, Carlsbad, CA), and GATA4 (LS-B7989, 1:100 dilution; Lifespan Biosciences Inc.). Immunostaining was performed using heat-induced epitope retrieval. Slides were blocked with 3% H2O2 and then incubated with the antibody for 1 hour at 37°C. Detection of antibody binding was obtained using Optiview (Ventana) after a polymer-based amplification step (Ultraview; Ventana). Slides were then counterstained with hematoxylin and coverslipped. Positive controls were used; for calretinin, mesothelioma tissue section; for inhibin, adrenal gland tissue section; for CD56, brain tissue section; for GATA4, prostate tissue section; for HER2, HER2-positive breast tumor tissue section; and for SMAD3, breast tissue was used. With regards to negative controls, ovarian tissue section without any pathology was used. All diagnoses and IHC reads were confirmed by 2 gynecologic oncology pathologists and in a few cases, where discrepancy was noted, a multihead microscope was used to discuss the areas of discrepancy to reach a consensus on the final read.

H Score

Each marker was scored for % and intensity and H score was established by combining both. Expression of GATA4 was categorized as negative for 0%, low for <80% with +1 nuclear staining, intermediate for <80% with +2/+3 or for >80% with +1, and high for ≥ 80% with +2/+3 staining (25). Expression of inhibin was categorized as negative for 0%, low for ≤50% with +1 cytoplasmic staining, intermediate for ≤50% with +2/+3 or >50% with +1, and high for >50% with +2/+3 staining (44); for SMAD3, negative for 0%, low for any % with +1 cytoplasmic staining, intermediate for <70% with +2/+3, and high for ≥70% with +2/+3 staining (25); for VEGF, negative for <20% with +1 cytoplasmic staining, low for 20% to <60% with +2, intermediate for ≤60% with +1/+3 or >60% with +1/+2, and high for >60% with +3 staining (45). Scoring of membranous antigen for Her2 was negative for 0%, low for <80% with +1 staining, intermediate for <80% with +2/+3 or ≥80% with +1, and high for ≥80% with +2 or +3 staining (19). Scoring for membranous and cytoplasmic antigen for CD56 was negative for 0%, low for <50% with +1 or +2 staining, intermediate for <50% with +3 or ≥50% with +1/+2, and high for ≥50% with +3 staining (46). Expression of calretinin was scored as low for <50% with +1 nuclear and cytoplasmic staining, intermediate for ≤50% with +2/+3 or >50% with +1, and high for >50% with +2/+3 staining (44). Figure 1 illustrates the different IHC stains used. Statistical analyses were conducted using Fisher exact test (2-tailed) and Kaplan-Meier (KM) survival curves. Significance was set at P<0.05.

FIG. 1.

FIG. 1.

IHC stains of primary GCT showing high mitoitc index and high expression of different the pathologic markers [Calretinin (A), CD56 (B), GATA4 (C), HER2 (D), Inhibin (E), SMAD3 (F)] investigated.

RESULTS

The patient clinical characteristics (125 patients with the diagnosis of primary GCT) with respect to recurrence were analyzed using x2 and Fisher exact test to perform univariate analyses and are summarized in Table 1. The median follow-up period was 120 months (range, 1–465 mo). Recurrence was noted in 12/125 (9.6%) patients and was significantly higher in whites and patients who had surgery and chemo-therapy (P = 0.001 and 0.0001, respectively). Early-stage GCT (stages I–II) were significantly less likely to have a recurrence as compared with stages III-IV (P = 0.001). Patients with recurrent GCT had a shorter overall survival (83 vs. 138 mo) and disease-free interval (DFI) (60 vs.135 mo), without a statistical difference noted.

TABLE 1.

Correlation of clinical characteristics of primary GCT to the recurrence status

Parameter Variable n (%) P
Nonrecurrent (n = 113)* Recurrent (n = 12)
Age at diagnosis ≤50 64 (59.3) 4 (33.3) 0.47
> 50 44 (40.7) 8 (66.7)
Race White 12 (16.7) 8 (66.7) 0.001
Black 46 (63.9) 3 (25.0)
Other 14 (19.4) 1 (8.3)
BMI (median) 31.5 27.7 0.41
Stage Stage I-II 111 (98.2) 8 (66.7) 0.001
Stage III-IV 2(1.8) 4 (33.3)
Lymph node status Negative 28 (93.3) 5 (100.0) 1.0
Positive 2 (6.7) 0 (0.0)
Lymphadenectomy Yes 30 (27.3) 4 (66.7) 0.66
No 80 (72.7) 2 (33.3)
Treatment Surgery only 63 (92.6) 2 (16.7) 0.0001
Surgery+chemo 3 (4.4) 10 (83.3)
Chemotherapy 2 (2.9) 0 (0.0)
Type of surgery USO 27 (40.9) 4 (33.3) 0.54
BSO 7 (10.6) 1 (8.3)
TAH/USO 26 (39.4) 7 (58.3)
TAH/BSO 6 (9.1) 0 (0.0)
Median survival (mo) 138 83 0.94
DFI (mo) 135 60 0.12
Vital status Alive 77 (68.1) 6 (50.0) 0.21
Dead 36 (31.9) 6 (50.0)
*

Data is missing in some cases.

Bold values are statistically significant.

BMI indicates body mass index; DFI, disease-free interval; GCT, granulosa cell tumors.

Histomorphologic Features and Tumor Characteristics Associated With Recurrent GCT

AGCT comprised the majority of patients (113/125, 90%) and 12 (10%) patients were juvenile type (JGCT) (Table 2). Tumors with a mitotic rate ≥4/10 HPF and severe NP were found to have a significantly higher likelihood of recurrence (P = 0.021 and 0.016, respectively). JGCT were more likely to recur (2/12, 17%) as compared with AGCT (10/113, 9%). Tumor subtype, size, and capsule rupture did not show a significant effect on recurrence.

TABLE 2.

Correlation of histomorphologic features and tumor characteristics of primary GCT with recurrence

Parameter Variable n (%) P
Nonrecurrent (n = 113)* Recurrent (n = 12)
Tumor type Adult 103 (91.2) 10 (83.3) 0.32
Juvenile 10 (8.8) 2 (16.7)
Tumor subtype Solid 39 (61.9) 5 (50.0) 0.55
Cystic 9 (14.3) 1 (10.0)
Solid cystic 15 (23.8) 4 (40.0)
Tumor size <10 cm 43 (47.8) 6 (60.0) 0.52
≥10 cm 47 (52.2) 4 (40.0)
Capsule rupture Yes 13 (28.3) 2 (50.0) 0.57
No 33 (71.7) 2 (50.0)
Histopathologic type Diffuse 45 (61.6) 7 (70.0) 0.98
Nondiffuse types 28 (38.4) 3 (30.0)
Mitotic rate <4/10 HPF 56 (70.0) 4 (33.3) 0.021
≥4/10 HPF 24 (30.0) 8 (66.7)
Nuclear pleomorphism Mild 42 (63.6) 2 (20.0) 0.016
Moderate 19 (28.8) 5 (50.0)
Severe 5 (7.6) 3 (30.0)
*

Data is missing in some cases.

Bold values are statistically significant.

GCT indicates granulosa cell tumors.

Univariate analysis for IHC assessment of protein expression is illustrated in Table 3. High levels of Inhibin, HER2, CD56, and SMAD3 expression were significantly associated with increased recurrence (P = 0.02, 0.014, 0.001, and 0.0001, respectively). On multivariate logistic regression analysis of clinicopathologic variables, treatment type continued to have a significant impact on recurrence, with the addition of chemotherapy to surgery predicting increased likelihood of recurrence by 59-fold (P = 0.001; 95% CI, 5.39–646.34). Mitotic count ≥4/10 HPF and high levels of expression of CD56 and SMAD3 were also found to be independent predictors of recurrence (OR = 13.3, P = 0.038;OR = 9.8, P = 0.04; and OR = 14.2, P = 0.001, respectively) (Table 4). HER2 expression was omitted from the multivariate analysis because of not having nonrecurrent patients with HER2 expression.

TABLE 3.

Correlation of IHC markers with recurrent and nonrecurrent GCT

Marker H Score n (%) P
Nonrecurrent (n = 71)* Recurrent (n = 12)
Calretinin Negative 15 (21.1) 1 (10.0) 0.71
Low: ≤50% 25 (35.2) 4 (40.0)
High: >50% 31 (43.7) 5 (50.0)
Inhibin Negative 41 (59.4) 3 (30.0) 0.02
Low: ≤50% 24 (34.8) 4 (40.0)
High: >50% 4 (5.8) 3 (30.0)
Her2 Negative 71 (100.0) 8 (80.0) 0.014
High≥80% 0 (0.0) 2 (20.0)
CD56 Negative 28 (38.4) 0 (0.0) 0.001
Low: ≤80% 17 (23.3) 0 (0.0)
High: >80% 28 (30.4) 10 (100.0)
VEGF Negative 2 (2.8) 0 (0.0) 0.74
Low: ≤80% 2 (2.8) 0 (0.0)
High: >80% 67 (94.4) 10 (100.0)
SMAD3 Negative 51 (72.9) 0 (0.0) 0.0001
Low: ≤80% 11 (15.7) 2 (20.0)
High: >80% 8 (11.4) 8 (80.0)
GATA4 Negative 53 (74.6) 6 (60.0) 0.15
Low: ≤80% 15 (21.1) 2 (20.0)
High: >80% 3 (4.2) 2 (20.0)
*

Data is missing in some cases.

Bold values are statistically significant.

GCT indicates granulosa cell tumors; IHC, immunohistochemistry.

TABLE 4.

Multivariate analysis using forced stepwise logistic regression and cox regression analyses

Variable OR 95% CI P HR 95% CI P
Treatment 59.04 5.39–646.34 0.001 7.02 1.69–29.17 0.007
SMAD3 14.23 2.87–70.65 0.001 5.02 1.16–21.64 0.03
Mitotic rate 13.33 1.15–154.41 0.038 0.86 0.95–7.78 0.89
CD56 9.88 1.06–91.44 0.04 5.54 0.26–118.66 0.27

Bold values are statistically significant.

CI indicates confidence interval; HR, hazard ratio; OR, odds ratio.

Using KM analysis, shorter mean DFI was noted in whites (142.7 vs. 212.9 and 199.52 mo in blacks and others, respectively, P = 0.001), stage III-IV versus stage I-II (77.2 vs. 432.4 mo, P = 0.0001), and in patients treated with surgery+chemotherapy versus surgery (73.5 vs. 227.2 mo) (P = 0.0001). It is noted that the chemotherapy-only group had longer DFI, but these were 2 patients only and the results were censored.

Also, shorter mean DFI was noted in patients with mitotic rate ≥4 (162.3 vs. 432.9 mo, P = 0.005), severe NP (104.2 vs. 181.2 vs. 440.5 mo, P = 0.013), high HER2 expression (89.0 vs. 401.6 mo, P = 0.001), high CD56 expression (156.8 vs. 453.9 mo, P = 0.001), and high SMAD3 expression (220.6 vs. 441.5 mo, P = 0.001) (Fig. 2). On Cox regression analysis, SMAD3 and the need for adjuvant chemo-therapy were the only 2 independent prognostic factors for DFI (P = 0.03 and P = 0.007, respectively) (Table 4).

FIG. 2.

FIG. 2.

A–H, Kaplan-Meier plots of disease-free interval to first recurrence according to race, stage, treatment, mitotic rate, nuclear polymorphism, and expression of HER2, CD56, SMAD3.

To evaluate the utility of the prognostic factors in early-stage (stage 1) GCTs, a subanalysis was performed (Table 5 and 6 and Fig. 3). Treatment (need for adjuvant chemotherapy), and high expression of CD56 and SMAD3 were found to be significant predictors of recurrence on univariate analysis (Table 5), but only treatment was found to be an independent predictor of recurrence on multivariate analysis (P = 0.01) (Table 7).

TABLE 5.

Univariate subanalysis (early stage: Stage I)

Parameter Variable n (%) P
Nonrecurrent (n = 79)* Recurrent (n = 6)
Treatment Surgery only 62 (95.3) 1 (16.7) 0.0001
Surgery+chemo 3 (4.6) 5 (83.3)
Mitotic rate <4/HPF 55 (69.6) 3 (50.0) 0.377
≥4/HPF 24 (30.4) 3 (50.0)
CD56 Neg+low: ≤80% 44 (61.1) 0 (0.0) 0.012
High: >80% 28 (38.9) 5 (100.0)
SMAD3 Neg+low: ≤80% 62 (89.9) 1 (20.0) 0.001
High: >80% 7 (10.1) 4 (80.0)
CD56 and SMAD3 Low-low 22 (78.6) 1 (20.0) 0.007
Low-high 2 (7.1) 0 (0.0)
High-high 4 (14.3) 4 (80.0)
*

Data is missing in some cases.

Bold values are statistically significant.

TABLE 6.

Multivariate analysis using logistic regression (forced stepwise) method and cox regression analyses (forced stepwise) regarding disease-free survival (subanalysis: early stage: stage I)

Variable Odds Ratio 95% CI P Hazard Ratio 95% CI P
Treatment 17.25 1.73–172.02 0.01 10.26 1.69–62.40 0.01
High CD56+high SMAD3 14.87 0.93–237.42 0.05 7.7 0.8–37.9 0.78

Bold values are statistically significant.

CI indicates confidence interval.

FIG. 3.

FIG. 3.

A–E, Kaplan-Meier plots of disease-free interval to first recurrence according. treatment, mitotic rate, and expression of CD56 and SMAD3: subanalysis (stage I).

TABLE 7.

Clinical characteristics of the 12 cases of GCT recurrence

Case No. Age Race Surgery Chemotherapy Stage LN Status DOR (mo) Vital Status Recurrence Site
1 25 CA USO (Lt), staging BEP II 0/5 24 Alive Ascitis, rt ovarian pelvic-abdominal mass. Large necrotic left inguinal LN or possibly a direct hernia
2 68 CA TAHBSO, omentectomy Depolupron IIA 90 Alive Pelvic side wall, ischeorectal fossa
3 60 CA USO, staging BEP IIIA 0/10, 0/1 31 Alive Pelvis, diaphragm/liver
4 58 CA TAHBSO None IIB 0/15, 0/5 85 Dead Pelvic, pararectal
5 50 O BSO BEP IC 0/2, 0/8 38 Alive Pelvic, lung, liver
6 63 AA TAHBSO None IA 30 Dead Abdomen, pelvis, peritoneum
7 22 AA USO BEP IIIC 26 Dead Ovarian
8 36 AA TAHBSO BEP, 2nd cytored, cytox/Avast IIIA 0 49 Alive Pelvis, omentum
9 63 CA TAHBSO Vinbl/BP NA 60 Dead Ascitis, intermediate iliac LN, pelvis, abdominal, lung, liver
10 57 CA TAHBSO Carbo/taxol IIIC 64 Alive Diaphr, peritoneum, pelvis, abdominal wall, omentum
11 51 CA TAHBSO CT-BEP-Cytox/Av-2nd cytored-Gem-Doxil-phase I IC 42 Alive Liver, bowel
12 80 CA TAHBSO Yes (NA) IIIC 240 Dead Pelvis, peritoneum, omentum

Bold values are statistically significant.

Avast. indicates avastin; BEP, bleomycin, etoposide, platinum; BP, bleomycin, platinum; Carbo, carboplatin; CT, carboplatin, taxol; Cytored, cytoreduction; cytox, cytoxan; DOR, date of recurrence; Gem, gemcitabine; NA, not available; O, other; TAHBSO, total abdominal hysterectomy with bilateral salpingo-oophorectomy; Vinbl, vinblastine.

In KM analysis, a significantly shorter DFI was noted with the surgery+chemotherapy treatment group and with high expression of CD56 and SMAD3 (P = 0.0001, P = 0.008, and P = 0.0001, respectively) (Fig. 2). We further investigated the value of high simultaneous expression of CD56 and SMAD3 in stage I GCT. Although significant on univariate and KM analysis (P = 0.007 and P = 0.04), simultaneous high expression of CD56 and SMAD3 showed a tendency toward significance on multivariate analysis (P = 0.05). Given that treatment is not an inherent characteristic in patients, repeat Cox regression analysis in stage I (excluding treatment) showed that high expression of SMAD3 was significantly associated with increased risk of recurrence (HR = 8.9; 95% CI, 2.34–34.5; P = 0.001). Tables 7 and 8 summarize the clinicopathologic characteristics of the 12 patients who developed a recurrence.

TABLE 8.

Pathologic characteristics of the 12 cases of GCT recurrence

Case No. Type Solid/Cyst Size Rupture ME MR NP CD56 SMAD3
1 JGCT C 16 No Diffuse 3 Moderate High High
2 AGCT S 3.5 Diffuse 9 Severe High Low
3 AGCT S/C 20 Yes Diffuse 5 Severe High High
4 AGCT S 16 Micronodular 5 Moderate High High
5 AGCT NA 7 NA NA 2 NA NA NA
6 AGCT S 0.4 Trabecular 4 Severe High High
7 JGCT S/C 34 No Diffuse 12 Mild High Low
8 AGCT 2.5 Yes NA 2 NA NA NA
9 AGCT S/C Diffuse 1 Mild High Low
10 AGCT S/C Diffuse 9 Moderate Intermediate High
11 AGCT S 8 Diffuse 9 Moderate High High
12 AGCT S 1.5 Trabecular 4 Mild High High

Bold values are statistically significant.

AGCT indicates adult granulosa cell tumor; C, cystic; JGCT, juvenile granulosa cell tumor; ME, microscopic exam; MR, mitotic rate; NP, nuclear pleomorphism, S, solid; S/C, solid cystic.

DISCUSSION

GCTs are low-grade malignant neoplasms, in which intraoperative and postoperative management decisions are challenging often times. This stems from the rarity of GCTs and the need for prolonged follow-up periods, which limit the availability of high-quality evidence. Recurrences after therapy have typically occurred within 5 to 10 years (47,48), and up to as long as 37 years (49), which makes the establishment of reliable prognostic indicators or predictors of recurrence a challenge. This may account for the discrepancy noted in the overall 10-year versus the 25-year survival in AGCTs in general and in stage I; 60% to 90% versus 40% to 60% and 85% to 95% versus 69% to 90%, respectively (47,48,50,51). As the majority (60%–90%) of GCTs present with stage I disease (52), the development of reliable predictors of recurrence may help in identifying early-stage GCTs with a higher likelihood for recurrence that may have gone without adjuvant therapy or inadequately followed after surgery. We sought to investigate some of the previously studied clinicopathologic prognostic factors and to explore novel markers of recurrence.

Of the clinical factors investigated, patient’s age and tumor size/rupture remain to be of controversial prognostic value. Although some studies support worse prognosis with older age (1,12), tumor size >10 to >15 cm (53,54), and tumor rupture (16), others as in our study do not (16,18,51,55).

In contrast, stage of disease has been consistently shown to be the most important independent prognostic factor with long-term follow-up studies showing the 10-year survival to be 84% to 95%, 50% to 65%, and 17% to 33% for stage I, II, and III-IV, respectively (3,13).

Although in our study, a significant difference was only noted on univariate and KM analyses for stage but not on multivariate analysis, this may have been explained by the relatively small sample size and the majority of patients being in early stage. In addition, the significant association of chemotherapy administration (reflective of disease stage) and recurrence may support the anticipated effect of stage on prognosis/recurrence.

However, in a subanalysis of stage I, patients receiving adjuvant chemotherapy (stage IC, residual disease) were significantly more likely to have a recurrence and to have a shorter DFI (OR = 17.25; 95% CI, 1.73–172.02; P = 0.01 and 104.3 vs. 58.9 mo, P = 0.0001).

In some studies, postoperative chemotherapy showed a clinical benefit in advanced stage and recurrent disease (5659), but others failed to show any improvement in progression-free survival (PFS) and overall survival with the addition of chemo-therapy (6,53,6062). In agreement with our study, Meisel et al (63) reported that patients who did not receive adjuvant chemotherapy had a significantly longer median PFS and a significantly higher 3-year PFS rate than those that did.

The effcacy of pathologic factors (MI, NP) as predictors of recurrence remains controversial partly because of the subjectivity (interobserver variation), use of different cutoffs, and not controlling for stage. Some studies reported a worse prognosis with increased NP (12,47) and MI (different cutoffs >3 to >10) (12,14,16,17,64), whereas others reported no prognostic significance (1,34,51,55,65). In agreement with the findings of Fox and colleagues and Lassus and colleagues, the subanalysis performed in our study showed no predictive value for MI in early-stage (stage I) GCTs. However, the mitotic rate was found to be an independent predictor of recurrence on logistic regression analysis of the whole data with a greater likelihood for recurrence with a mitotic rate ≥4. This may be explained by possible skewness of the data by the higher stage GCTs, and thus highlights the importance of performing subanalysis on early-stage GCTs.

Although high serum levels of and high expression of VEGF were noted in GCT (45), 1 study failed to show a prognostic significance in predicting recurrence/PFS as shown in our study (66). Also, even though GATA4 was recently found to be an independent predictor of recurrence (19), our data failed to show any prognostic utility for GATA4 as well as HER2 (33,35). However, expression of HER2 may be of importance in predicting resistance to treatment with bevacizumab.

CD56 has been recently found to be expressed in primary and recurrent GCTs (39), with a high degree of sensitivity for sex-cord stromal tumors (46). In our study, high expression of CD56 independently predicted a higher likelihood of recurrence and a significantly longer DFI.

The expression of SMAD3 in GCTs has not been studied before. High expression of SMAD3 independently predicted a higher likelihood of recurrence and a significantly longer DFI. In addition, on a subanalysis for early-stage (stage I) GCTs, high expression of CD56 or SMAD3 showed a significant prediction of recurrence on univariate analysis only; however, simultaneous high expression of CD56 and SMAD3 predicted a higher likelihood of recurrence and significantly shorter DFI on multivariate analysis. More importantly, the true utility of SMAD3 in predicting recurrence in early-stage GCTs was illustrated upon exclusion of treatment (which is not an inherent characteristic of patients) from the analysis. High expression of SMAD3 was the only independent predictor of recurrence. This may prove to be a useful tool in counseling patients postoperatively about recurrence risk, closer surveillance, and need for chemotherapy and may prove to be a future therapeutic target.

Ideally, a predictive model for recurrence of early-stage (stage I) GCTs could be developed based on combining clinical and pathologic factors. An example of such model was suggested by Nosov et al. (20), where it was suggested that patients younger than 48 years whose tumor has a higher expression of inhibin and Ki67 are at a higher risk for recurrence and hence would benefit from adjuvant chemotherapy. Another model predicting recurrence-free survival in a retrospective cohort of 127 patients from multiple institutions created a nomogram based on clinical stage, body mass index, tumor diameter, and MI (67).

Appreciable differences are noted among different studies as well as our study with regards to the degree of expression of different pathologic prognostic factors, which are potential therapeutic targets (such as VEGF, HER2). Also potential novel pathologic prognostic factors/therapeutic targets (SMAD3 and other TGF-β pathway targets) are being investigated. In an era of precision medicine, individualizing patient care postoperatively and making a choice of adjuvant chemotherapy based on which of these IHC pathologic factors is highly expressed, patients (determined to be at higher risk for recurrence) can be treated with bevacizumab (45,68), trastuzumab/imatinib (69), peptide aptemers, and ligand traps/antisense oligonucleotides (70) if they have GCTs that have a high expression of VEGF, HER2, SMAD3, and TGF-β receptor, respectively.

Our study is not without limitations; this is a retrospective review with inherent bias but because of the rare nature of GCTs and tendency for late recurrence, this makes accruing patients in a prospective randomized study very diffcult, even from multiple institutions. Also, the percentage of recurrence was relatively low (12/125, 9.6%), compared with higher reported rates of recurrence by other authors.

However, this review is one of the larger series published on GCTs from a single institution and spans over 39 years. Because many studies investigating prognostic factors in AGCTs do not include a pathologic review of the cases, one of the major strengths of our study is the stringent pathology review by the same gynecologic oncology pathologist to confirm the diagnosis. Also performing a sub-analysis to predict recurrence and DFI in early-stage patients was rarely done in studies evaluating GCTs because of small sample size.

To the best of our knowledge, this is the first report of the utility of CD56 and SMAD3 IHC expression in primary GCTs as independent predictors of recurrence. This will need to be validated on a larger scale, possibly through multisite collaboration, to confirm the utility of these prognostic factors.

Footnotes

The authors declare no conflict of interest.

REFERENCES

  • 1.Fox H, Agrawal K, Langley FA. A clinicopathologic study of 92 cases of granulosa cell tumor of the ovary with special reference to the factors influencing prognosis. Cancer 1975;35:231–41. [DOI] [PubMed] [Google Scholar]
  • 2.Stuart GC, Dawson LM. Update on granulosa cell tumours of the ovary. Curr Opin Obstet Gynecol 2003;15:33–7. [DOI] [PubMed] [Google Scholar]
  • 3.Pankratz E, Boyes DA, White GW, et al. Granulosa cell tumors. A clinical review of 61 cases. Obstet Gynecol 1978;52:718–23. [PubMed] [Google Scholar]
  • 4.Schwartz PE, Smith JP. Treatment of ovarian stromal tumors. Am J Obstet Gynecol 1976;125:402–11. [DOI] [PubMed] [Google Scholar]
  • 5.Lee YK, Park NH, Kim JW, et al. Characteristics of recurrence in adult-type granulosa cell tumor. Int J Gynecol Cancer 2008;18:642–7. [DOI] [PubMed] [Google Scholar]
  • 6.Sun HD, Lin H, Jao MS, et al. A long-term follow-up study of 176 cases with adult-type ovarian granulosa cell tumors. Gynecol Oncol 2012;124:244–9. [DOI] [PubMed] [Google Scholar]
  • 7.Sehouli J, Drescher FS, Mustea A, et al. Granulosa cell tumor of the ovary: 10 years follow-up data of 65 patients. Anticancer Res 2004;24(2C):1223–9. [PubMed] [Google Scholar]
  • 8.Colombo N, Sessa C, Landoni F, et al. Cisplatin, vinblastine, and bleomycin combination chemotherapy in metastatic granulosa cell tumor of the ovary. Obstet Gynecol 1986;67:265–8. [DOI] [PubMed] [Google Scholar]
  • 9.Gershenson DM, Morris M, Burke TW, et al. Treatment of poor-prognosis sex cord-stromal tumors of the ovary with the combination of bleomycin, etoposide, and cisplatin. Obstet Gynecol 1996;87:527–31. [DOI] [PubMed] [Google Scholar]
  • 10.Homesley HD, Bundy BN, Hurteau JA, et al. Bleomycin, etoposide, and cisplatin combination therapy of ovarian granulosa cell tumors and other stromal malignancies: a Gynecologic Oncology Group study. Gynecol Oncol 1999;72:131–7. [DOI] [PubMed] [Google Scholar]
  • 11.Schumer ST, Cannistra SA. Granulosa cell tumor of the ovary. J Clin Oncol 2003;21:1180–9. [DOI] [PubMed] [Google Scholar]
  • 12.Stenwig JT, Hazekamp JT, Beecham JB. Granulosa cell tumors of the ovary. A clinicopathological study of 118 cases with long-term follow-up. Gynecol Oncol 1979;7:136–52. [DOI] [PubMed] [Google Scholar]
  • 13.Zhang M, Cheung MK, Shin JY, et al. Prognostic factors responsible for survival in sex cord stromal tumors of the ovary—an analysis of 376 women. Gynecol Oncol 2007;104: 396–400. [DOI] [PubMed] [Google Scholar]
  • 14.Miller BE, Barron BA, Wan JY, et al. Prognostic factors in adult granulosa cell tumor of the ovary. Cancer 1997;79: 1951–5. [DOI] [PubMed] [Google Scholar]
  • 15.Evans AT III, Gaffey TA, Malkasian GD Jr, et al. Clinicopathologic review of 118 granulosa and 82 theca cell tumors. Obstet Gynecol 1980;55:231–8. [PubMed] [Google Scholar]
  • 16.Bjorkholm E, Silfversward C. Prognostic factors in granulosa-cell tumors. Gynecol Oncol 1981;11:261–74. [DOI] [PubMed] [Google Scholar]
  • 17.Fujimoto T, Sakuragi N, Okuyama K, et al. Histopathological prognostic factors of adult granulosa cell tumors of the ovary. Acta Obstet Gynecol Scand 2001;80:1069–74. [DOI] [PubMed] [Google Scholar]
  • 18.Malmstrom H, Hogberg T, Risberg B, et al. Granulosa cell tumors of the ovary: prognostic factors and outcome. Gynecol Oncol 1994;52:50–5. [DOI] [PubMed] [Google Scholar]
  • 19.Farkkila A, Andersson N, Butzow R, et al. HER2 and GATA4 are new prognostic factors for early-stage ovarian granulosa cell tumor-a long-term follow-up study. Cancer Med 2014;3: 526–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nosov V, Silva I, Tavassoli F, et al. Predictors of recurrence of ovarian granulosa cell tumors. Int J Gynecol Cancer 2009;19: 628–33. [DOI] [PubMed] [Google Scholar]
  • 21.Juric G, Zarkovic N, Nola M, et al. The value of cell proliferation and angiogenesis in the prognostic assessment of ovarian granulosa cell tumors. Tumori 2001;87:47–53. [DOI] [PubMed] [Google Scholar]
  • 22.Shah SP, Kobel M, Senz J, et al. Mutation of FOXL2 in granulosa-cell tumors of the ovary. N Engl J Med 2009;360: 2719–29. [DOI] [PubMed] [Google Scholar]
  • 23.Kalfa N, Philibert P, Patte C, et al. Extinction of FOXL2 expression in aggressive ovarian granulosa cell tumors in children. Fertil Steril 2007;87:896–901. [DOI] [PubMed] [Google Scholar]
  • 24.D’Angelo E, Mozos A, Nakayama D, et al. Prognostic significance of FOXL2 mutation and mRNA expression in adult and juvenile granulosa cell tumors of the ovary. Mod Pathol 2011;24:1360–7. [DOI] [PubMed] [Google Scholar]
  • 25.Anttonen M, Pihlajoki M, Andersson N, et al. FOXL2, GATA4, and SMAD3 co-operatively modulate gene expression, cell viability and apoptosis in ovarian granulosa cell tumor cells. PLoS One 2014;9:e85545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bilandzic M, Chu S, Wang Y, et al. Betaglycan alters NFkappaB-TGFbeta2 cross talk to reduce survival of human granulosa tumor cells. Mol Endocrinol 2013;27: 466–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Anttonen M, Parviainen H, Kyronlahti A, et al. GATA-4 is a granulosa cell factor employed in inhibin-alpha activation by the TGF-beta pathway. J Mol Endocrinol 2006;36:557–68. [DOI] [PubMed] [Google Scholar]
  • 28.Kyronlahti A, Ramo M, Tamminen M, et al. GATA-4 regulates Bcl-2 expression in ovarian granulosa cell tumors. Endocrinology 2008;149:5635–42. [DOI] [PubMed] [Google Scholar]
  • 29.Anttonen M, Unkila-Kallio L, Leminen A, et al. High GATA-4 expression associates with aggressive behavior, whereas low anti-Mullerian hormone expression associates with growth potential of ovarian granulosa cell tumors. J Clin Endocrinol Metab 2005;90:6529–35. [DOI] [PubMed] [Google Scholar]
  • 30.Kyronlahti A, Kauppinen M, Lind E, et al. GATA4 protects granulosa cell tumors from TRAIL-induced apoptosis. Endocr Relat Cancer 2010;17:709–17. [DOI] [PubMed] [Google Scholar]
  • 31.Lassus H, Leminen A, Vayrynen A, et al. ERBB2 amplification is superior to protein expression status in predicting patient outcome in serous ovarian carcinoma. Gynecol Oncol 2004;92: 31–9. [DOI] [PubMed] [Google Scholar]
  • 32.Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987;235:177–82. [DOI] [PubMed] [Google Scholar]
  • 33.Furger C, Fiddes RJ, Quinn DI, et al. Granulosa cell tumors express erbB4 and are sensitive to the cytotoxic action of heregulin-beta2/PE40. Cancer Res 1998;58:1773–8. [PubMed] [Google Scholar]
  • 34.King LA, Okagaki T, Gallup DG, et al. Mitotic count, nuclear atypia, and immunohistochemical determination of Ki-67, cmyc, p21-ras, c-erbB2, and p53 expression in granulosa cell tumors of the ovary: mitotic count and Ki-67 are indicators of poor prognosis. Gynecol Oncol 1996;61:227–32. [DOI] [PubMed] [Google Scholar]
  • 35.Menczer J, Schreiber L, Czernobilsky B, et al. Is Her-2/neu expressed in nonepithelial ovarian malignancies? Am J Obstet Gynecol 2007;196:79.e1–e4. [DOI] [PubMed] [Google Scholar]
  • 36.Higgins PA, Brady A, Dobbs SP, et al. Epidermal growth factor receptor (EGFR), HER2 and insulin-like growth factor-1 receptor (IGF-1R) status in ovarian adult granulosa cell tumours. Histopathology 2014;64:633–8. [DOI] [PubMed] [Google Scholar]
  • 37.Mayerhofer A, Lahr G, Frohlich U, et al. Expression and alternative splicing of the neural cell adhesion molecule NCAM in human granulosa cells during luteinization. FEBS Lett 1994;346:207–12. [DOI] [PubMed] [Google Scholar]
  • 38.Daniel L, Bouvier C, Chetaille B, et al. Neural cell adhesion molecule expression in renal cell carcinomas: relation to metastatic behavior. Hum Pathol 2003;34:528–32. [DOI] [PubMed] [Google Scholar]
  • 39.Volker HU, Engert S, Cramer A, et al. Expression of CD56 isoforms in primary and relapsed adult granulosa cell tumors of the ovary. Diagn Pathol 2008;3:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McCluggage WG, Young RH. Immunohistochemistry as a diagnostic aid in the evaluation of ovarian tumors. Semin Diagn Pathol 2005;22:3–32. [DOI] [PubMed] [Google Scholar]
  • 41.Nofech-Mozes S, Ismiil N, Dube V, et al. Immunohistochemical characterization of primary and recurrent adult granulosa cell tumors. Int J Gynecol Pathol 2012;31:80–90. [DOI] [PubMed] [Google Scholar]
  • 42.Pecorelli S Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int J Gynaecol Obstet 2009;105:103–4. [DOI] [PubMed] [Google Scholar]
  • 43.Kononen J, Bubendorf L, Kallioniemi A, et al. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 1998;4:844–7. [DOI] [PubMed] [Google Scholar]
  • 44.Movahedi-Lankarani S, Kurman RJ. Calretinin, a more sensitive but less specific marker than alpha-inhibin for ovarian sex cord-stromal neoplasms: an immunohistochemical study of 215 cases. Am J Surg Pathol 2002;26:1477–83. [DOI] [PubMed] [Google Scholar]
  • 45.Farkkila A, Pihlajoki M, Tauriala H, et al. Serum vascular endothelial growth factor A (VEGF) is elevated in patients with ovarian granulosa cell tumor (GCT), and VEGF inhibition by bevacizumab induces apoptosis in GCT in vitro. J Clin Endocrinol Metab 2011;96:E1973–81. [DOI] [PubMed] [Google Scholar]
  • 46.McCluggage WG, McKenna M, McBride HA. CD56 is a sensitive and diagnostically useful immunohistochemical marker of ovarian sex cord-stromal tumors. Int J Gynecol Pathol 2007;26:322–7. [DOI] [PubMed] [Google Scholar]
  • 47.Bjorkholm E Granulosa cell tumors: a comparison of survival in patients and matched controls. Am J Obstet Gynecol 1980;138:329–31. [DOI] [PubMed] [Google Scholar]
  • 48.Sjostedt S, Wahlen T. Prognosis of granulosa cell tumours. Acta Obstet Gynecol Scand Suppl 1961;40(suppl 6):1–26. [PubMed] [Google Scholar]
  • 49.Hines JF, Khalifa MA, Moore JL, et al. Recurrent granulosa cell tumor of the ovary 37 years after initial diagnosis: a case report and review of the literature. Gynecol Oncol 1996;60:484–8. [DOI] [PubMed] [Google Scholar]
  • 50.Pautier P, Lhomme C, Culine S, et al. Adult granulosa-cell tumor of the ovary: a retrospective study of 45 cases. Int J Gynecol Cancer 1997;7:58–65. [DOI] [PubMed] [Google Scholar]
  • 51.Lauszus FF, Petersen AC, Greisen J, et al. Granulosa cell tumor of the ovary: a population-based study of 37 women with stage I disease. Gynecol Oncol 2001;81:456–60. [DOI] [PubMed] [Google Scholar]
  • 52.Savage P, Constenla D, Fisher C, et al. Granulosa cell tumours of the ovary: demographics, survival and the management of advanced disease. Clin Oncol (R Coll Radiol) 1998;10:242–5. [DOI] [PubMed] [Google Scholar]
  • 53.Chan JK, Zhang M, Kaleb V, et al. Prognostic factors responsible for survival in sex cord stromal tumors of the ovary—a multivariate analysis. Gynecol Oncol 2005;96:204–9. [DOI] [PubMed] [Google Scholar]
  • 54.Thrall MM, Paley P, Pizer E, et al. Patterns of spread and recurrence of sex cord-stromal tumors of the ovary. Gynecol Oncol 2011;122:242–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cronje HS, Niemand I, Bam RH, et al. Review of the granulosa-theca cell tumors from the emil Novak ovarian tumor registry. Am J Obstet Gynecol 1999;180(pt 1):323–7. [DOI] [PubMed] [Google Scholar]
  • 56.Uygun K, Aydiner A, Saip P, et al. Clinical parameters and treatment results in recurrent granulosa cell tumor of the ovary. Gynecol Oncol 2003;88:400–3. [DOI] [PubMed] [Google Scholar]
  • 57.Sigismondi C, Gadducci A, Lorusso D, et al. Ovarian Sertoli-Leydig cell tumors. a retrospective MITO study. Gynecol Oncol 2012;125:673–6. [DOI] [PubMed] [Google Scholar]
  • 58.Park JY, Jin KL, Kim DY, et al. Surgical staging and adjuvant chemotherapy in the management of patients with adult granulosa cell tumors of the ovary. Gynecol Oncol 2012;125: 80–6. [DOI] [PubMed] [Google Scholar]
  • 59.Chiara S, Merlini L, Campora E, et al. Cisplatin-based chemotherapy in recurrent or high risk ovarian granulosa-cell tumor patients. Eur J Gynaecol Oncol 1993;14:314–7. [PubMed] [Google Scholar]
  • 60.Lee IH, Choi CH, Hong DG, et al. Clinicopathologic characteristics of granulosa cell tumors of the ovary: a multicenter retrospective study. J Gynecol Oncol 2011;22: 188–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Al-Badawi IA, Brasher PM, Ghatage P, et al. Postoperative chemotherapy in advanced ovarian granulosa cell tumors. Int J Gynecol Cancer 2002;12:119–23. [DOI] [PubMed] [Google Scholar]
  • 62.Gurumurthy M, Bryant A, Shanbhag S. Effectiveness of different treatment modalities for the management of adult-onset granulosa cell tumours of the ovary (primary and recurrent). Cochrane Database Syst Rev 2014;4:CD006912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Meisel JL, Hyman DM, Jotwani A, et al. The role of systemic chemotherapy in the management of granulosa cell tumors. Gynecol Oncol 2015;136:505–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Miller BE, Barron BA, Dockter ME, et al. Parameters of differentiation and proliferation in adult granulosa cell tumors of the ovary. Cancer Detect Prev 2001;25:48–54. [PubMed] [Google Scholar]
  • 65.Costa MJ, Walls J, Ames P, et al. Transformation in recurrent ovarian granulosa cell tumors: Ki67 (MIB-1) and p53 immunohistochemistry demonstrates a possible molecular basis for the poor histopathologic prediction of clinical behavior. Hum Pathol 1996;27:274–81. [DOI] [PubMed] [Google Scholar]
  • 66.Farkkila A, Anttonen M, Pociuviene J, et al. Vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 are highly expressed in ovarian granulosa cell tumors. Eur J Endocrinol 2011;164:115–22. [DOI] [PubMed] [Google Scholar]
  • 67.van Meurs HS, Schuit E, Horlings HM, et al. Development and internal validation of a prognostic model to predict recurrence free survival in patients with adult granulosa cell tumors of the ovary. Gynecol Oncol 2014;134: 498–504. [DOI] [PubMed] [Google Scholar]
  • 68.Tsoi M, Lague MN, Boyer A, et al. Anti-VEGFA therapy reduces tumor growth and extends survival in a murine model of ovarian granulosa cell tumor. Transl Oncol 2013;6: 226–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Stern HM. Improving treatment of HER2-positive cancers: opportunities and challenges. Sci Transl Med 2012;4: 127rv122. [DOI] [PubMed] [Google Scholar]
  • 70.Connolly EC, Freimuth J, Akhurst RJ. Complexities of TGF-beta targeted cancer therapy. Int J Biol Sci 2012;8:964–78. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES