Purpose: The mechanisms by which trastuzumab imparts clinical benefit remain incompletely understood. Antibody-dependent cellular cytotoxicity via interactions with Fcγ receptors (FcγR) on leukocytes may contribute to its antitumor effects. Single-nucleotide polymorphisms (SNP) in FCGR3A and FCGR2A genes lead to amino acid substitutions at positions 158 and 131, respectively, and affect binding of antibodies to FcγR such that 158V/V and 131H/H bind with highest affinity. This study aimed to determine whether high-affinity SNPs are associated with disease-free survival (DFS) among patients with HER2-positive nonmetastatic breast cancer.

Experimental Design: Genomic DNA was isolated from 1,286 patients enrolled in a trial of adjuvant trastuzumab-based chemotherapy. Genotyping was conducted using Sanger sequencing and Sequenom mass spectrometry.

Results: Patient samples (N = 1,189) were successfully genotyped for FCGR3A and 1,218 for FCGR2A. Compared with the overall results of the BCIRG006 study, in the subset of patients genotyped in this analysis, a less robust improvement in DFS was observed for the trastuzumab arms than control arm (HR, 0.842; P = 0.1925). When stratified for prognostic features, the HR in favor of trastuzumab was consistent with that of the overall study (HR, 0.74; P = 0.036). No correlation between DFS and FCGR3A/2A genotypes was seen for trastuzumab-treated patients (158V/V vs. V/F vs. F/F, P = 0.98; 131H/H vs. H/R vs. R/R, P = 0.76; 158V/V and/or 131H/H vs. others, P = 0.67).

Conclusion: This analysis evaluating the association between FCGR3A/2A genotypes and trastuzumab efficacy in HER2-positive breast cancer did not show a correlation between FCGR3A-V/F and FCGR2A-H/R SNPs and DFS in patients treated with trastuzumab. Clin Cancer Res; 18(12); 3478–86. ©2012 AACR.

Translational Relevance

One proposed but unproven mechanism of action (MoA) of the monoclonal antibody (mAb) trastuzumab is induction of antibody-dependent cellular cytotoxicity, in which the Fc portion of trastuzumab engages with the Fc-γ receptor (FcγR) on immune effector cells leading to tumor cell destruction. We sought to determine whether differences in FcγR affinity resulting from two single-nucleotide polymorphisms in FcγRIIIa and FcγRIIa impact the clinical outcome of trastuzumab-treated patients. An association between FcγR genotype and outcome was reported with other mAbs for lymphoma and colon cancer. Such an association would provide evidence that the immune system plays a role in the MoA of trastuzumab. These results also impact the future practice of cancer medicine because a positive association would support the development of engineered antibodies with an increased affinity for FcγR and would support the use of genotyping to preselect patients most likely to respond to trastuzumab.

Amplification of the HER2 gene is a key driver in the pathogenesis and biologic aggressiveness of approximately 25% of breast cancer (1). Trastuzumab, a humanized anti-HER2 monoclonal IgG1 antibody, is known to significantly improve clinical outcome for both early and advanced HER2-positive breast cancer (2–4). Although the mechanisms of action of trastuzumab are not completely understood (5), preclinical models suggest that growth factor receptor blockade results in critical changes in growth signaling pathways including downregulation of PI3K/AKT signaling leading to decreased cell proliferation and cycle arrest (6). Other mechanisms suggested from preclinical studies also include inhibition of extracellular domain shedding, decreased angiogenesis, and inhibition of DNA repair (7, 8).

Therapeutic antibodies of the IgG1 subtype can also mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This potential mechanism involves antibody binding to HER2 on the surface of tumor cells, followed by the fragment C (Fc) portion of the antibody engaging Fc-γ receptors (FcγR) expressed on immune effector cells, ultimately resulting in target cell lysis. Preclinical evidence for this mechanism in trastuzumab efficacy was shown in immunodeficient mice bearing human breast cancer xenografts (9). Furthermore, afucosylated trastuzumab with enhanced affinity to FcγR exhibits greater antitumor activity in xenograft models than native trastuzumab (10).

Three classes of FcγR [FcγRI (CD64), FcγRII (CD32), and FcγRIII (CD16a)] and their subclasses have been described. Some FcγR display allelic polymorphisms that confer differing functional properties (11). One such polymorphism in the gene encoding FcγRIIIa is a single-nucleotide substitution at position 559 (ref. 12; A559C, rs396991) that leads to the substitution of phenylalanine (F) by valine (V) at amino acid position 158 in the IgG-binding domain (13, 14). IgG1 and IgG3 bind more tightly to FcγRIIIa 158V/V than to 158F/F, increasing effector cell activity in individuals who are homozygous for FcγRIIIa 158V (13, 14). A polymorphism in the gene encoding FcγRIIa (A519G, rs1801274) places either histidine (H) or arginine (R) at position 131. IgG1 binds more strongly to cells that are homozygous for FcγRIIa 131H (15).

Clinical evidence supporting an association between FCGR3A/2A genotypes and outcomes in patients treated with monoclonal antibody therapy was first reported for rituximab in the treatment of lymphoma (11, 16). Subsequently, studies evaluating the monoclonal antibody, cetuximab, for colon cancer showed an association between FCGR3A/2A genotypes and outcome (17, 18). However, definitive clinical evidence for the role of Fc–FcγR interactions in breast cancer is lacking. Three small trials, each with fewer than 65 patients, evaluated the association between FCGR3A/2A genotypes and outcome after treatment with trastuzumab-based therapy. Two studies reported an association between at least one FcγR polymorphism and clinical outcome (19, 20). The other study revealed no such association (21).

The aim of this study was to further clarify whether FCGR3A and FCGR2A genotypes are correlated with clinical outcome in trastuzumab-treated patients. Such an association would substantiate a role for FcγR-bearing immune effector cells in the antitumor activity of trastuzumab.

FcγR polymorphism genotyping

DNA was purified from serum and whole blood samples using a QIAamp DNA Blood mini kit (Qiagen) and used for nested PCR amplification of regions containing the FCGR3A 158 V/F and FCGR2A 131 H/R SNPs using primers listed in Supplementary Table S1. PCR was carried out using Phusion Hot Start High-Fidelity DNA Polymerase (New England Biolabs) and manufacturer-recommended protocols. The PCR products were purified using a Qiagen PCR clean kit (Qiagen) and then sequenced on an ABI3730XL (Applied Biosystems) using BigDye Terminator v3.1 chemistry. PCR products were also analyzed on a MassARRAY analyzer (Sequenom) using Sequenom's iPLEX Gold assay. For FCGR3A, rs396991 primers were used to identify the A559C (22) polymorphism. For FCGR2A, rs1801274 primers were used to identify the A519G (22) polymorphism. Each sample underwent a total of 4 independent rounds of analyses (2 Sanger and 2 Sequenom). The genotype was included for further analysis if there were 4 concordant results for a given sample. For samples where there were 3 concordant results and a fourth data point had failed for technical reasons, the genotype was called and included further in data analysis.

Patient population

Adjuvant breast cancer cohort (BCIRG-006).

Genomic DNA from serum and whole blood samples was obtained from patients treated in the Breast Cancer International Research Group (BCIRG)006 study (23). This adjuvant study compared 2 trastuzumab-containing arms with a non-trastuzumab–containing control arm for treatment of HER2-positive, early breast cancer. In total, 3,222 patients were randomly assigned to 1 of 3 treatment arms: (i) AC-T: 4 cycles of every 3 weekly doxorubicin (A, 60 mg/m2 i.v.) plus cyclophosphamide (C, 600 mg/m2 i.v.) followed by 4 cycles of every 3 weekly docetaxel (T, 75 mg/m2 i.v.), (ii) AC-TH: AC-T plus trastuzumab (H, 8 mg/kg i.v. loading dose with first dose of docetaxel followed by 6 mg/kg every 3 weeks for 1 year), or (iii) TCH: 6 cycles of every 3 weekly docetaxel, carboplatin [C, area under curve (AUC) 6], trastuzumab (as above, for 1 year). Of these 3,222 patients, 1,286 signed an optional consent upon enrollment to have blood/serum samples sent to our central laboratory for exploratory analyses. A total of 1,189 patient samples (37%) were successfully genotyped for FCGR3A and 1,218 samples (38%) genotyped for FCGR2A. Genotyping failed in 97 samples (7.5%) for FCGR3A and in 68 samples (5.3%) for FCGR2A. Approximately, 860 samples sequenced were from whole blood, and the success rate was more than 99% for both polymorphisms from these specimens. The remainder of patients (>400) only had serum provided. The concentration of DNA is lower in serum than in whole blood, thus making it technically more challenging to extract an adequate amount of DNA for reliable sequencing from serum. The vast majority of sequencing failures were from serum samples. That said, the fail rate in serum for FCGR3A was higher than that for FCGR2A, so there may be a contributing factor that depends on the primers. Because of high homology with FCGR3B, there are unfortunately very limited options for designing primers specific for FCGR3A. The proportion of patients who were genotyped for FCGR3A/2A was well-balanced between the treatment arms (Fig. 1).

Figure 1.

REMARK (24) diagram for adjuvant cohort.

Figure 1.

REMARK (24) diagram for adjuvant cohort.

Close modal

Advanced disease breast cancer cohort.

Blood samples from 177 participants in the PolyomX and Canadian Breast Cancer Foundation (CBCF—Edmonton, AB, Canada) tumor banks were collected from 2001 to 2007. All participants had HER2-positive breast cancer and had received at least one course of trastuzumab. A total of 53 participants had unresectable, local/regional recurrence (N = 12) or distant metastases (N = 41) and had successful determination of at least one FcγR allele. The FCGR3A 158V/F genotype was successfully determined in 52 participants (29%) and FCGR2A 131H/R in 53 participants (30%).

Both the early and advanced disease cohort studies were conducted according to Institutional Review Board/Ethics Committee–approved protocols. Informed consent was obtained from all participating patients. REMARK guidelines (24) were followed in the reporting of these results.

Statistical methods and association testing

For the adjuvant cohort, disease-free survival (DFS) was calculated from the date of randomization to the date of disease recurrence as declared by the treating physician or death from any cause. This retrospective data analysis was based on the third planned analysis of the BCIRG006 study (23). For the advanced disease cohort, progression-free survival (PFS) was calculated from start of first exposure to trastuzumab (in the metastatic or locally recurrent setting) to the time of disease progression or death from any cause. DFS and PFS curves were estimated using the method of Kaplan–Meier. The effect of trastuzumab and the prognostic impact of genotype were assessed using the log-rank test. The predictive impact of genotype on the effect of trastuzumab was assessed through interaction tests in Cox regression models.

SNPStats software (http://bioinfo.iconcologia.net/SNPstats; ref. 25) was used for determining allele frequencies and Hardy–Weinberg equilibrium (HWE) and the Haploview program (http://www.broadinstitute.org; ref. 26) for pairwise linkage disequilibrium (LD; measured as D′) patterns between markers. A sample size of N = 1,133 was used for which we have complete genotype data to determine LD between FCGR2A and FCGR3A gene polymorphisms. The Fisher exact test was used to assess deviations from HWE, with P < 0.05 suggesting significant deviation from HWE.

Patient characteristics

Adjuvant breast cancer cohort.

The prognostic clinical and pathologic features of patients according to treatment arm are shown in Table 1. At the third planned analysis of BCIRG006 (N = 3,222), DFS was significantly improved for patients who received trastuzumab-based therapy compared with control arm therapy [AC-TH vs. AC-T: HR, 0.64, 95% confidence interval (CI), 0.53–0.78; P < 0.001; TCH vs. AC-T: HR, 0.75; 95% CI, 0.63–0.90; P = 0.002; Supplementary Fig. S1], indicating that trastuzumab-based therapy significantly extends DFS compared with chemotherapy alone (23). The clinical and tumor characteristics of the patients genotyped in our study compared with the patients who were not genotyped are shown in Supplementary Table S2. In the subset of patients genotyped in our study (N = 1,286), a less robust improvement in DFS was observed for patients treated with trastuzumab compared with control arm therapy (combined trastuzumab-arms vs. AC-T: HR, 0.842; P = 0.1925; Supplementary Fig. S2). Stratified analysis showed that this may be due to genotyped patients in the trastuzumab arms numerically having worse prognostic features than patients in the AC-T arm (Table 1). When stratified for age, node status, hormone receptor status, size and surgery type, the HR in favor of trastuzumab was consistent with that of the overall patient population and statistically significant (HR, 0.74; P = 0.036; Supplementary Fig. S3). Baseline patient characteristics by genotype for this adjuvant cohort are shown in Table 2.

Table 1.

Patient characteristics by treatment arm (adjuvant cohort)

AC-TH (%), N = 406TCH (%), N = 402AC-TH and TCH (combined; %), N = 808AC-T (%), N = 381
Number of positive lymph nodes     
 0 112 (28) 133 (33) 245 (30) 120 (31) 
 1 157 (39) 163 (41) 320 (40) 153 (40) 
 >4 137 (34) 106 (26) 243 (30) 108 (28) 
Hormone receptor     
 ER/PR-negative 222 (55) 224 (56) 446 (55) 194 (51) 
 ER and/or PR-positive 184 (45) 178 (44) 362 (45) 187 (49) 
Tumor size, cm     
 >2 248 (61) 240 (60) 488 (60) 210 (55) 
 ≤2 158 (39) 162 (40) 320 (40) 171 (45) 
Menopause status     
 Premenopause 177 (44) 168 (42) 345 (43) 170 (45) 
 Postmenopause 229 (56) 234 (58) 463 (57) 211 (55) 
Age, y     
 Median 49 48 48 49 
 Range 24–74 23–73 23—74 26–74 
Tumor grade     
 G1 3 (0.7) 6 (1.5) 9 (1.1) 6 (1.6) 
 G2 102 (25) 113 (28) 215 (27) 83 (22) 
 G3 293 (72) 274 (68) 567 (70) 282 (74) 
 G4 1 (0.2) 2 (0.5) 3 (0.4) 3 (0.8) 
 GX 7 (1.7) 7 (1.7) 14 (1.7) 7 (1.8) 
AC-TH (%), N = 406TCH (%), N = 402AC-TH and TCH (combined; %), N = 808AC-T (%), N = 381
Number of positive lymph nodes     
 0 112 (28) 133 (33) 245 (30) 120 (31) 
 1 157 (39) 163 (41) 320 (40) 153 (40) 
 >4 137 (34) 106 (26) 243 (30) 108 (28) 
Hormone receptor     
 ER/PR-negative 222 (55) 224 (56) 446 (55) 194 (51) 
 ER and/or PR-positive 184 (45) 178 (44) 362 (45) 187 (49) 
Tumor size, cm     
 >2 248 (61) 240 (60) 488 (60) 210 (55) 
 ≤2 158 (39) 162 (40) 320 (40) 171 (45) 
Menopause status     
 Premenopause 177 (44) 168 (42) 345 (43) 170 (45) 
 Postmenopause 229 (56) 234 (58) 463 (57) 211 (55) 
Age, y     
 Median 49 48 48 49 
 Range 24–74 23–73 23—74 26–74 
Tumor grade     
 G1 3 (0.7) 6 (1.5) 9 (1.1) 6 (1.6) 
 G2 102 (25) 113 (28) 215 (27) 83 (22) 
 G3 293 (72) 274 (68) 567 (70) 282 (74) 
 G4 1 (0.2) 2 (0.5) 3 (0.4) 3 (0.8) 
 GX 7 (1.7) 7 (1.7) 14 (1.7) 7 (1.8) 

Abbreviations: A, adriamycin; C, cytoxan; GX, grade unknown; H, Herceptin; T, Taxotere.

Table 2.

Patients characteristics by genotype (adjuvant cohort)

V/V (%), N = 169V/F (%), N = 471F/F (%), N = 549H/H (%), N = 323H/R (%), N = 597R/R (%), N = 298
Number of positive lymph nodes       
 0 62 (37) 131 (28) 172 (31) 93 (29) 187 (31) 94 (32) 
 1–3 55 (32) 207 (44) 211 (38) 129 (40) 242 (41) 113 (38) 
 >4 52 (31) 133 (28) 166 (30) 101 (31) 168 (28) 91 (31) 
Hormone receptor       
 ER/PR-negative 92 (54) 261 (55) 287 (52) 183 (57) 328 (55) 149 (50) 
 ER and/or PR-positive 77 (46) 210 (45) 262 (48) 140 (43) 269 (45) 149 (50) 
Tumor size, cm       
 >2 93 (55) 279 (59) 326 (59) 195 (60) 341 (57) 176 (59) 
 ≤2 76 (45) 192 (41) 223 (41) 128 (40) 256 (43) 122 (41) 
Menopause status       
 Premenopause 77 (46) 192 (41) 246 (45) 143 (44) 250 (42) 133 (45) 
 Postmenopause 92 (54) 279 (59) 303 (55) 180 (56) 347 (58) 165 (55) 
Age, y       
 Median 49.3 48.5 48.6 49.0 48.0 49.0 
 Range 27.0–74.0 23.0–74.0 24.0–74.0 28.0–74.0 22.0–74.0 24.0–73.0 
HER2 ratio       
n 154 435 496 290 557 263 
 Mean 8.3 8.2 8.0 8.3 8.1 8.1 
 Range 2.2–19.3 2.2–19.5 2.3–18.1 2.2–23.5 2.3–19.5 2.2–19.2 
Treatment arm       
 AC-T 56 (33) 149 (32) 176 (32) 110 (34) 182 (30) 95 (32) 
 AC-TH 53 (31) 162 (34) 191 (35) 114 (35) 210 (35) 91 (31) 
 TCH 60 (36) 160 (34) 182 (33) 99 (31) 205 (34) 112 (38) 
V/V (%), N = 169V/F (%), N = 471F/F (%), N = 549H/H (%), N = 323H/R (%), N = 597R/R (%), N = 298
Number of positive lymph nodes       
 0 62 (37) 131 (28) 172 (31) 93 (29) 187 (31) 94 (32) 
 1–3 55 (32) 207 (44) 211 (38) 129 (40) 242 (41) 113 (38) 
 >4 52 (31) 133 (28) 166 (30) 101 (31) 168 (28) 91 (31) 
Hormone receptor       
 ER/PR-negative 92 (54) 261 (55) 287 (52) 183 (57) 328 (55) 149 (50) 
 ER and/or PR-positive 77 (46) 210 (45) 262 (48) 140 (43) 269 (45) 149 (50) 
Tumor size, cm       
 >2 93 (55) 279 (59) 326 (59) 195 (60) 341 (57) 176 (59) 
 ≤2 76 (45) 192 (41) 223 (41) 128 (40) 256 (43) 122 (41) 
Menopause status       
 Premenopause 77 (46) 192 (41) 246 (45) 143 (44) 250 (42) 133 (45) 
 Postmenopause 92 (54) 279 (59) 303 (55) 180 (56) 347 (58) 165 (55) 
Age, y       
 Median 49.3 48.5 48.6 49.0 48.0 49.0 
 Range 27.0–74.0 23.0–74.0 24.0–74.0 28.0–74.0 22.0–74.0 24.0–73.0 
HER2 ratio       
n 154 435 496 290 557 263 
 Mean 8.3 8.2 8.0 8.3 8.1 8.1 
 Range 2.2–19.3 2.2–19.5 2.3–18.1 2.2–23.5 2.3–19.5 2.2–19.2 
Treatment arm       
 AC-T 56 (33) 149 (32) 176 (32) 110 (34) 182 (30) 95 (32) 
 AC-TH 53 (31) 162 (34) 191 (35) 114 (35) 210 (35) 91 (31) 
 TCH 60 (36) 160 (34) 182 (33) 99 (31) 205 (34) 112 (38) 

Advanced disease breast cancer cohort.

Patient characteristics by genotype in this 53-patient cohort are shown in Supplementary Table S3. HER2 overexpression/amplification was verified in 50 and was unknown in 3 participants. Tumors were positive for one or both hormone receptors in 70% (n = 37) of patients, negative for both in 23% (n = 12), and unknown in 8% (n = 4). A total of 42% of patients were postmenopausal. Tumor grade was grade I in 2%, grade II in 30%, grade III in 53%, and unknown in 15% of patients. Visceral metastases were present in 66% of participants. Of the 53 patients, 43 had not received prior chemotherapy and 10 had received 1 to 4 previous chemotherapy regimens. In terms of specific trastuzumab-based regimens received by patients, 18 (34%) received trastuzumab alone, 28 (53%) received single-agent chemotherapy plus trastuzumab, and 7 (13%) received doublet (taxane-platinum) chemotherapy plus trastuzumab.

Genotype and allele frequencies

Adjuvant breast cancer cohort.

The frequency of FCGR3A/2A genotypes did not differ significantly among treatment arms (Table 2). We observed a minor allele frequency of 0.34 and 0.48 for FCGR3A and FCGR2A, respectively. The frequencies of FCGR3A genotypes deviated from HWE, whereas the genotype distributions for FCGR2A were in conformity with the HWE assumptions (Supplementary Table S4). The influence of genotyping errors on the observed deviations from HWE for FCGR3A were ruled out or minimized as the genotyping data from 2 independent technology platforms (see Patients and Methods) were concordant. We do not have genotype data from apparently healthy control subjects to assess conformity with HWE assumptions in a case–control setting to suggest putative association of this locus with breast cancer risk or the associated phenotypes, thus limiting the interpretability of our findings. We nonetheless included this allele for further analysis to permit comparisons with the previously reported, smaller studies (19, 20). The LD (D′ = 0.32) we observed between FCGR2A and FCG3RA was entirely concordant with those previously reported in the literature (27).

FcγR polymorphisms and outcome

Adjuvant breast cancer cohort.

Baseline patient and tumor characteristics did not differ significantly between the FCGR3A V/V, V/F, and F/F polymorphism groups, nor between FCGR2A H/H, H/R, and R/R groups (Table 2). In the population of patients genotyped who were in the non-trastuzumab–containing control arm (AC-T), there was no statistically significant difference in DFS based on FCGR3A/2A genotypes (FCGR3A V/V vs. V/F vs. F/F, log-rank test: P = 0.33; FCGR3A H/H vs. H/R vs. R/R, log-rank test: P = 0.81). Among those who received trastuzumab (TCH and AC-TH arms combined), there was no statistically significant difference in DFS by FCGR3A genotype (P = 0.98; Fig. 2A) or FCGR2A genotype (P = 0.76; Fig. 2B). When cases having the “favorable” FCGR3A V/V and/or FCGR2A H/H genotypes were compared with others, there was also no statistically significant difference in DFS (P = 0.67; Fig. 2C). When the trastuzumab-containing treatment arms were analyzed separately, again there was no difference in DFS by the FCGR3A (TCH: P = 0.96, AC-TH: P = 0.94), FCGR2A (TCH: P = 0.98, AC-TH: P = 0.47), or by combined FCGR3A V/V and/or FCGR2A H/H genotypes (TCH V/V and/or H/H vs. TCH others vs. AC-TH V/V and/or H/H vs. AC-TH others, log-rank: P = 0.97; Supplementary Fig. S4). To evaluate whether genotype is prognostic independently of trastuzumab, the non-trastuzumab–containing AC-T arm was analyzed separately. There was no difference in DFS by the FCGR3A (P = 0.33), FCGR2A (P = 0.81), or by combined FCGR3A V/V and/or FCGR2A H/H genotypes (P = 0.39; Supplementary Fig. S5). There was also no difference in overall survival when comparing FCGR3A/2A genotypes (Supplementary Fig. S6). Finally, we compared the trastuzumab-containing treatment arms with AC-T in all of the FCGR3A and FCGR2A genotypes. Despite an apparent trend toward a larger effect of trastuzumab in the FCGR3A V/V and/or FCGR2A H/H genotypes (Fig. 3), the difference did not reach significance in any genotype, and the tests for interaction between trastuzumab and genotype were all nonsignificant.

Figure 2.

Kaplan–Meier estimates of DFS in trastuzumab-treated patients within the adjuvant cohort according to FcγR polymorphisms. A, FCGR3A 158 V/F genotype, P = 0.98 (V/V vs. V/F vs. F/F). B, FCGR2A 131 H/R genotype, P = 0.76 (H/H vs. H/R vs. R/R). C, FcγRIIIa and FcγRIIa polymorphisms. Others represent patients with neither FCGR3A 158V/V nor FCGR2A 131H/H genotype, P = 0.67 (H/H and/or V/V vs. others).

Figure 2.

Kaplan–Meier estimates of DFS in trastuzumab-treated patients within the adjuvant cohort according to FcγR polymorphisms. A, FCGR3A 158 V/F genotype, P = 0.98 (V/V vs. V/F vs. F/F). B, FCGR2A 131 H/R genotype, P = 0.76 (H/H vs. H/R vs. R/R). C, FcγRIIIa and FcγRIIa polymorphisms. Others represent patients with neither FCGR3A 158V/V nor FCGR2A 131H/H genotype, P = 0.67 (H/H and/or V/V vs. others).

Close modal
Figure 3.

Kaplan–Meier estimates of DFS in trastuzumab-treated (AC-TH/TCH combined) versus no trastuzumab–treated (AC-T) patients, by FcγR genotype. A, V/V (P = 0.13). B, V/F (P = 0.422). C, F/F (P = 0.81). D, H/H (P = 0.35). E, H/R (P = 0.985). F, R/R (P = 0.58).

Figure 3.

Kaplan–Meier estimates of DFS in trastuzumab-treated (AC-TH/TCH combined) versus no trastuzumab–treated (AC-T) patients, by FcγR genotype. A, V/V (P = 0.13). B, V/F (P = 0.422). C, F/F (P = 0.81). D, H/H (P = 0.35). E, H/R (P = 0.985). F, R/R (P = 0.58).

Close modal

Advanced disease breast cancer cohort.

In the 53-patient advanced disease cohort, baseline prognostic features including age, tumor grade, and disease-free interval did not differ significantly between the 3 FCGR3A genotypes and the 3 FCGR2A genotypes (Supplementary Table S3). Menopausal status, hormone receptor status, and presence of visceral metastases differed significantly between genotypes for one or both SNPs. Menopausal status was statistically different between genotypes for both SNPs (χ2, P = 0.0448 for FCGR3A and P = 0.0287 for FCGR2A). Hormone receptor status was statistically different between genotypes with FCGR3A [V/V more frequently estrogen receptor (ER)- and/or progesterone receptor (PR)-positive than other genotypes, χ2, P = 0.0488]. Presence of visceral metastases differed significantly between genotypes for FCGR2A (visceral metastases less frequent in H/R genotype than others, χ2, P = 0.013). For convenience, we have reported all P values unadjusted for multiplicity, which is standard practice in retrospective analyses, when the number of comparisons is not prespecified. In actual fact, as many comparisons were conducted, P values much less extreme than P < 0.05 are to be considered statistically significant. As a rough guidance, the Bonferroni correction can be used in the interpretation: if M comparisons are conducted, the level of significance that applies to each comparison is equal to 0.05 divided by M. Nonetheless, no statistically significant difference in PFS was detected by FCGR3A genotype (FCGR3A V/V vs. V/F vs. F/F, log-rank test, P = 0.88; Supplementary Fig. S7A) and by FCGR2A genotypes (FCGR2A H/H vs. H/R vs. R/R, log-rank test, P = 0.52; Supplementary Fig. S7B).

In addition to perturbation of HER2 signaling, trastuzumab-mediated FcγR engagement by immune effector cells could represent a potential mechanism of action for the antibody in HER2-positive breast cancer. The present study was conducted to determine whether differences in FcγR affinity resulting from SNPs in FCGR3A and FCGR2A had any impact on the outcome of patients treated with trastuzumab-based therapy.

To date, there has been no prospective evaluation of FCGR3A/2A genotypes as determinants of trastuzumab outcome. Three previous retrospective studies investigating the correlation of FCGR3A/2A genotypes with clinical outcome to trastuzumab-based therapy yielded discordant results (19–21). Each of these studies was limited by a small sample size. Moreover, in contrast to the large adjuvant cohort in the current study, the majority of patients (91%) evaluated in the 3 published studies had metastatic breast cancer. The first report was a retrospective analysis of a subset of patients enrolled in the pivotal trial of trastuzumab. No difference in the distribution of the FCGR3A 158V/F genotype was detected among 63 patients who achieved an objective response (OR) and those who had progressive disease (21). Conversely, a subsequent study by Musolino and colleagues reported improved response rates and PFS for those patients with FCGR3A V/V and, to a lesser extent, FCGR2A H/H genotypes among 54 patients with HER2-positive metastatic breast cancer who received trastuzumab and taxane (19). Tamura and colleagues evaluated whether FCGR3A/2A genotypes are associated with pathologic complete response (pCR) or OR in patients treated with chemotherapy plus trastuzumab in the neoadjuvant setting (n = 15) and whether the genotypes are associated with PFS in patients with metastatic breast cancer (n = 35) who received single-agent trastuzumab (20). They also showed a correlation with clinical outcome. Specifically, they found that FCGR2A H/H genotype was significantly associated with pCR (P = 0.015) and OR (P = 0.043) in the neoadjuvant setting. They also found a correlation with PFS (P = 0.034) in the metastatic setting; however, FCGR3A genotype was not significantly associated with clinical outcome in that study (20).

The current study involves the largest retrospective analysis to date evaluating an association between FCGR3A/2A genotypes and clinical outcome in trastuzumab-treated HER2-positive breast cancer in the adjuvant setting. No statistically significant correlation between FCGR3A and FCGR2A genotypes and DFS was detected in a cohort of 1,286 patients treated with trastuzumab-based therapy in early breast cancer. Moreover, to expand this study to advanced disease, the retrospective analysis of a cohort of 53 women treated with trastuzumab-based therapy for metastatic breast cancer was conducted and also revealed no significant correlation between FCGR3A and FCGR2A genotypes and PFS. While these data do not completely rule out the possibility that trastuzumab acts in part via ADCC, it does suggest that any differences in Fc–FcγR affinity attributed to the SNPs tested does not result in detectable differences in clinical outcome. We acknowledge that these data are limited by the fact that only 38% of the patients enrolled in the BCIRG006 study were genotyped. Thus, it is not possible to generalize conclusions originating from the genotyped subset to the entire BCIRG006.

The trastuzumab benefit in this study appeared less robust in the adjuvant cohort compared with the benefit seen in the overall BCIRG006 study population, most likely due to the fact that random sampling of study patients for genotyping could not be conducted. This was because only those patients who provided informed consent and had separate blood/serum samples sent into the centralized laboratory for biomarker testing were evaluated. As a result, the sample in which FCGR3A/2A genotyping was conducted was not representative of the entire patient population. In fact, in this sample, the lower benefit of trastuzumab may have been due to the imbalance in poorer than average prognostic features of trastuzumab-treated patients consenting to provide samples in this substudy. However, stratified analyses adjusting for known prognostic factors support a statistically significant trastuzumab benefit of the same magnitude in the current cohort as in the overall population. That said, we cannot completely exclude a contribution of FCGR polymorphisms to the trastuzumab-treated patient outcomes.

Our results differ from those of Musolino and colleagues (19), and Tamura and colleagues (20), and these discrepancies may reflect differences in intrinsic population factors or chemotherapy regimens. For instance, while the study used by Musolino and colleagues administered only paclitaxel or docetaxel in combination with trastuzumab, patients in our adjuvant cohort received doxorubicin, cyclophosphamide, and docetaxel, or carboplatin and docetaxel in combination with trastuzumab. In patients with lymphoma treated with rituximab, the strong influence of FCGR3A/2A genotypes on outcomes appears to be diminished with concurrent chemotherapy (28). On the other hand, the majority (87%) of patients with breast cancer in our advanced disease breast cancer cohort received either trastuzumab monotherapy or single-agent chemotherapy plus trastuzumab. Despite this, PFS still did not correlate with FCGR3A/2A genotypes.

The 2 receptor polymorphisms considered in this study did not show a significant association with treatment outcomes. The minor allele frequencies and LD between these markers are consistent with previous findings (19, 22).

This is the largest analysis evaluating the association between FCGR3A/2A genotypes and clinical outcome in trastuzumab-treated HER2-positive breast cancer. In contrast to previously published smaller studies, we found no statistically significant correlation between FCGR3A/2A genotypes and DFS in the adjuvant setting or PFS in the metastatic setting for patients with HER2-positive breast cancer treated with trastuzumab. These data do not support the hypothesis that naturally occurring polymorphism-related differences in FcγR affinity result in differential outcome to trastuzumab therapy. However, the data require confirmation in a prospective study and at present time do not completely preclude a potential role for ADCC/FcγR engagement in the treatment of breast cancer with anti-HER2 antibodies The Fc engineering of antibodies with much higher affinity Fc–FcγR interactions than those observed with naturally occurring polymorphisms may provide future opportunities to fully engage this mechanism of action against breast cancer.

S.A. Hurvitz has a commercial research grant and a travel grant from Roche and Genentech. H.M. Stern, J. Stinson, S. Seshagiri, and M.X. Sliwkowski (as Sr. Staff Scientist) have employment in Genentech and Roche and ownership interests. M. Buyse has employment (other than primary affiliation; e.g., consulting) in IDDI as the Chairman and has Ownership Interest (including patents) in IDDI. J. Mackey and J. Crown have honoraria from Speakers' bureau of Roche. J. Crown has received commercial research grant from Roche. C. Falkson has honoraria from speakers' bureau of Genentech. C. Falkson and A. Brufsky are consultants/advisory board members in Genentech. N.J. Robert and D.J. Slamon have honoraria from Speakers' bureau of Genentech and Roche and are consultant/advisory board members for Genentech and Roche. N.J. Robert has commercial research grant from Genentech and Roche. V. Valero has commercial research grant from Genentech and Roche and honoraria from Speakers' bureau of Genentech. T. Pienkowski has a commercial research grant and a travel grant from Roche and is a consultant/advisory board member for Roche. W. Eiermann is a consultant/advisory board member for Roche. No potential conflicts of interest were disclosed by other authors.

Conception and design: S.A. Hurvitz, D.J. Betting, H.M. Stern, M. Buyse, M.X. Sliwkowski, D.J. Slamon, J.M. Timmerman

Development of methodology: S.A. Hurvitz, H.M. Stern, M. Buyse

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D.J. Betting, H.M. Stern, J. Stinson, S. Seshagiri, Y. Zhao, J. Mackey, N.J. Robert, J. Crown, C. Falkson, A. Brufsky, T. Pienkowski, W. Eiermann, M. Martin, D.J. Slamon, J.M. Timmerman

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.A. Hurvitz, H.M. Stern, E. Quinaux, J. Stinson, S. Seshagiri, M. Buyse, A. Driga, S. Damaraju, M.X. Sliwkowski, V. Valero, J. Crown, A. Brufsky, M. Martin, O. Marathe, J.M. Timmerman

Writing, review, and/or revision of the manuscript: S.A. Hurvitz, D.J. Betting, H.M. Stern, M. Buyse, J. Mackey, A. Driga, M.X. Sliwkowski, N.J. Robert, V. Valero, J. Crown, C. Falkson, A. Brufsky, T. Pienkowski, W. Eiermann, M. Martin, O. Marathe, D.J. Slamon, J.M. Timmerman

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.A. Hurvitz, Y. Zhao, A. Driga, V. Valero, V. Bee, D.J. Slamon

Study supervision: S.A. Hurvitz, J. Mackey

The funding for this project came from the ASCO Young Investigator Award in 2007 and from Genentech.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Slamon
DJ
,
Clark
GM
,
Wong
SG
,
Levin
WJ
,
Ullrich
A
,
McGuire
WL
. 
Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene
.
Science
1987
;
235
:
177
82
.
2.
Slamon
DJ
,
Leyland-Jones
B
,
Shak
S
,
Fuchs
H
,
Paton
V
,
Bajamonde
A
, et al
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2
.
N Engl J Med
2001
;
344
:
783
92
.
3.
Piccart-Gebhart
MJ
,
Procter
M
,
Leyland-Jones
B
,
Goldhirsch
A
,
Untch
M
,
Smith
I
, et al
Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1659
72
.
4.
Romond
EH
,
Perez
EA
,
Bryant
J
,
Suman
VJ
,
Geyer
CE
 Jr
,
Davidson
NE
, et al
Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer
.
N Engl J Med
2005
;
353
:
1673
84
.
5.
Hudis
CA
. 
Trastuzumab–mechanism of action and use in clinical practice
.
N Engl J Med
2007
;
357
:
39
51
.
6.
Junttila
TT
,
Akita
RW
,
Parsons
K
,
Fields
C
,
Lewis
Phillips GD
,
Friedman
LS
, et al
Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941
.
Cancer Cell
2009
;
15
:
429
40
.
7.
Klos
KS
,
Zhou
X
,
Lee
S
,
Zhang
L
,
Yang
W
,
Nagata
Y
, et al
Combined trastuzumab and paclitaxel treatment better inhibits ErbB-2-mediated angiogenesis in breast carcinoma through a more effective inhibition of Akt than either treatment alone
.
Cancer
2003
;
98
:
1377
85
.
8.
Izumi
Y
,
Xu
L
,
di Tomaso
E
,
Fukumura
D
,
Jain
RK
. 
Tumour biology: herceptin acts as an anti-angiogenic cocktail
.
Nature
2002
;
416
:
279
80
.
9.
Clynes
RA
,
Towers
TL
,
Presta
LG
,
Ravetch
JV
. 
Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets
.
Nat Med
2000
;
6
:
443
6
.
10.
Junttila
TT
,
Parsons
K
,
Olsson
C
,
Lu
Y
,
Xin
Y
,
Theriault
J
, et al
Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer
.
Cancer Res
2010
;
70
:
4481
9
.
11.
Cartron
G
,
Dacheux
L
,
Salles
G
,
Solal-Celigny
P
,
Bardos
P
,
Colombat
P
, et al
Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene
.
Blood
2002
;
99
:
754
8
.
12.
Ravetch
JV
,
Perussia
B
. 
Alternative membrane forms of Fc gamma RIII(CD16) on human natural killer cells and neutrophils. Cell type-specific expression of two genes that differ in single nucleotide substitutions
.
J Exp Med
1989
;
170
:
481
97
.
13.
Koene
HR
,
Kleijer
M
,
Algra
J
,
Roos
D
,
von dem Borne
AE
,
de Haas
M
. 
Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype
.
Blood
1997
;
90
:
1109
14
.
14.
Wu
J
,
Edberg
JC
,
Redecha
PB
,
Bansal
V
,
Guyre
PM
,
Coleman
K
, et al
A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease
.
J Clin Invest
1997
;
100
:
1059
70
.
15.
Warmerdam
PA
,
van de Winkel
JG
,
Vlug
A
,
Westerdaal
NA
,
Capel
PJ
. 
A single amino acid in the second Ig-like domain of the human Fc gamma receptor II is critical for human IgG2 binding
.
J Immunol
1991
;
147
:
1338
43
.
16.
Weng
WK
,
Levy
R
. 
Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma
.
J Clin Oncol
2003
;
21
:
3940
7
.
17.
Zhang
W
,
Gordon
M
,
Schultheis
AM
,
Yang
DY
,
Nagashima
F
,
Azuma
M
, et al
FCGR2A and FCGR3A polymorphisms associated with clinical outcome of epidermal growth factor receptor expressing metastatic colorectal cancer patients treated with single-agent cetuximab
.
J Clin Oncol
2007
;
25
:
3712
8
.
18.
Bibeau
F
,
Lopez-Crapez
E
,
Di Fiore
F
,
Thezenas
S
,
Ychou
M
,
Blanchard
F
, et al
Impact of Fc{gamma}RIIa-Fc{gamma}RIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan
.
J Clin Oncol
2009
;
27
:
1122
9
.
19.
Musolino
A
,
Naldi
N
,
Bortesi
B
,
Pezzuolo
D
,
Capelletti
M
,
Missale
G
, et al
Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer
.
J Clin Oncol
2008
;
26
:
1789
96
.
20.
Tamura
K
,
Shimizu
C
,
Hojo
T
,
Akashi-Tanaka
S
,
Kinoshita
T
,
Yonemori
K
, et al
Fc{gamma}R2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer
.
Ann Oncol
2011
;
22
:
1302
7
.
21.
Foster
CHK
,
Ostland
M
,
Mass
R
,
Chen
R
,
Presta
L
,
Chanock
N
. 
FCGR3A V/F polymorphism does not predict response to Herceptin (H) monotherapy
.
Proc Am Soc Clin Oncol
21
:
57a
, 
2002
(
abstr 227
).
22.
Alizadeh
BZ
,
Broen
J
,
Rueda
B
,
Hesselstrand
R
,
Wuttge
D
,
Simeon
C
, et al
Functional variants of Fc gamma receptor (FCGR2A) and FCGR3A are not associated with susceptibility to systemic sclerosis in a large European Study (EUSTAR)
.
J Rheumatol
2010
;
37
:
1673
9
.
23.
Slamon
D
,
Eiermann
W
,
Robert
N
,
Pienkowski
T
,
Martin
M
,
Press
M
, et al
Adjuvant trastuzumab in HER2-positive breast cancer
.
N Engl J Med
2011
;
365
:
1273
83
.
24.
McShane
LM
,
Altman
DG
,
Sauerbrei
W
,
Taube
SE
,
Gion
M
,
Clark
GM
. 
Reporting recommendations for tumor marker prognostic studies
.
J Clin Oncol
2005
;
23
:
9067
72
.
25.
Sole
X
,
Guino
E
,
Valls
J
,
Iniesta
R
,
Moreno
V
. 
SNPStats: a web tool for the analysis of association studies
.
Bioinformatics
2006
;
22
:
1928
9
.
26.
Barrett
JC
,
Fry
B
,
Maller
J
,
Daly
MJ
. 
Haploview: analysis and visualization of LD and haplotype maps
.
Bioinformatics
2005
;
21
:
263
5
.
27.
Alizadeh
BZ
,
Valdigem
G
,
Coenen
MJ
,
Zhernakova
A
,
Franke
B
,
Monsuur
A
, et al
Association analysis of functional variants of the FcgRIIa and FcgRIIIa genes with type 1 diabetes, celiac disease and rheumatoid arthritis
.
Hum Mol Genet
2007
;
16
:
2552
9
.
28.
Carlotti
E
,
Palumbo
GA
,
Oldani
E
,
Tibullo
D
,
Salmoiraghi
S
,
Rossi
A
, et al
FcgammaRIIIA and FcgammaRIIA polymorphisms do not predict clinical outcome of follicular non-Hodgkin's lymphoma patients treated with sequential CHOP and rituximab
.
Haematologica
2007
;
92
:
1127
30
.

Supplementary data