Solid papillary carcinoma with reverse polarity (SPCRP) is a rare breast cancer subtype with an obscure etiology. In this study, we sought to describe its unique histopathologic features and to identify the genetic alterations that underpin SPCRP using massively parallel whole-exome and targeted sequencing. The morphologic and immunohistochemical features of SPCRP support the invasive nature of this subtype. Ten of 13 (77%) SPCRPs harbored hotspot mutations at R172 of the isocitrate dehydrogenase IDH2, of which 8 of 10 displayed concurrent pathogenic mutations affecting PIK3CA or PIK3R1. One of the IDH2 wild-type SPCRPs harbored a TET2 Q548* truncating mutation coupled with a PIK3CA H1047R hotspot mutation. Functional studies demonstrated that IDH2 and PIK3CA hotspot mutations are likely drivers of SPCRP, resulting in its reversed nuclear polarization phenotype. Our results offer a molecular definition of SPCRP as a distinct breast cancer subtype. Concurrent IDH2 and PIK3CA mutations may help diagnose SPCRP and possibly direct effective treatment. Cancer Res; 76(24); 7118–29. ©2016 AACR.

Breast cancer is a heterogeneous disease at the clinical, morphologic, and genetic level (1–3). The current World Health Organization (WHO) classification recognizes 21 histologic types (1), but few have been found to harbor specific genetic alterations, such as CDH1 alterations in invasive lobular carcinoma (4) and recurrent ETV6–NTRK3 and MYB–NFIB fusion genes in secretory and adenoid cystic carcinomas, respectively (5–7). Additional rare breast cancers with distinctive morphologic features also exist, but have not yet been included in the WHO classification. One such malignancy has been described as “breast tumor resembling the tall cell variant of papillary thyroid carcinoma (PTC)” (8, 9), which has a unique histologic appearance consisting of solid, circumscribed nodules of epithelial cells, many of which have fibrovascular cores, resulting in a solid papillary growth pattern. Thirteen such tumors have been reported to date (8, 9), of which approximately half lack estrogen (ER) and progesterone receptor (PR) expression, and all lack HER2 overexpression (8, 9). These tumors are generally associated with a favorable prognosis (8, 9); however, two of 13 patients have been reported to develop metastatic disease, one to an intra-mammary lymph node and another to the bone (8, 9).

Although these tumors have some histologic similarities to PTC, they consistently lack expression of thyroid-specific markers, including TTF-1 and thyroglobulin (8, 9). RET rearrangements and BRAF exon 15 mutations commonly found in PTC have also not been detected in these tumors (9). Given the morphologic overlap with other papillary lesions of the breast and lack of immunohistochemical and genetic evidence of an association with PTC, it has recently been suggested that these neoplasms should be considered morphologic variants of papillary breast carcinoma (9, 10).

In this study, we sought to characterize the morphologic and genetic landscape of this rare and morphologically unusual breast tumor and determine whether it represents a distinct subtype of breast cancer underpinned by disease-specific genetic alterations. To this end, we performed an extensive immunohistochemical characterization and whole exome (WES), targeted, and Sanger sequencing of 13 previously unreported tumors. We found that 10 of 13 (77%) tumors harbored R172 IDH2 mutations, of which eight had a concurrent pathogenic mutation affecting PI3K pathway canonical genes (i.e., six PIK3CA hotspot mutations and two PIK3R1 likely pathogenic mutations). In addition, a PIK3CA-mutant but IDH2-wild-type tumor was found to harbor a TET2 Q548* truncating mutation. Functional studies using nonmalignant breast epithelial cells demonstrated that IDH2 and PIK3CA mutations constitute likely drivers of this tumor and contribute to its characteristic phenotype. Because of their unique histologic and genetic properties, we redefine these tumors here as a discrete subtype of breast carcinoma: solid papillary carcinoma with reverse polarity (SPCRP).

Patient selection

Between 2005 and 2014, 13 SPCRPs were identified by two of the authors (S.J. Schnitt and E. Brogi) on the Breast Pathology Consultation (11 cases) or Breast Pathology (1 case) Services at Beth Israel Deaconess Medical Center (Boston, MA), and the Breast Pathology Consultation Service (1 case) at Memorial Sloan Kettering Cancer Center (MSKCC; New York, NY). Hematoxylin-and-eosin–stained sections were reviewed to delineate the morphologic features of these lesions. Available clinical data were reviewed for demographics, presentation, clinical and family history, treatment, and outcome. The study was approved by the Institutional Review Boards from the respective authors' institutions, and all samples were anonymized prior to analysis.

Immunohistochemistry and Western blotting

Primary antibodies for IHC were as follows: calponin, smooth muscle myosin-heavy chain (SMMHC), p63, AE1/AE3, cytokeratin (CK) 7, CK5/6, CK34βE12, ER, PR, androgen receptor (AR), HER2, mammaglobin, H3K27me3, gross cystic disease fluid protein-15 (GCDFP-15), TTF-1, thyroglobulin, E-cadherin, and MUC1 (Supplementary Table S1). For H3K27me3, we semiquantitatively assessed its nuclear expression using the H-score system as previously described (11); 500 nuclei were counted per case.

Standard Western blotting was performed as previously described (12) using primary antibodies against IDH2 and H3K9me3 (Abcam); phospho-Rb (S807/811), Rb (4H1), histone H3 (D1H2), and α-tubulin (DM1A; Cell Signaling Technology); H3K27me3 (EMD Millipore), and E-cadherin (BD Biosciences). Secondary conjugated IRDye 680RD/800CW antibodies (LI-COR Biosciences) were used. Detection and quantification were performed using the Image Studio Software from LI-COR (LI-COR Biosciences; refs. 13, 14).

Whole exome and targeted massively parallel sequencing

Eight micron–thick representative formalin-fixed paraffin-embedded (FFPE) tumor and normal tissue sections from two SPCRPs (cases 10 and 13), and tumor sections from two SPCRPs (cases 5 and 11) were stained with nuclear fast red and microdissected using a sterile needle under a stereomicroscope (Olympus SZ61), to ensure >80% tumor cell content and that normal tissue was devoid of neoplastic cells as previously described (5, 14). DNA was extracted from microdissected tumor and normal samples using the DNeasy Blood and Tissue Kit (Qiagen) following the manufacturer's guidelines. Tumor and matched germline DNA from SPCRP10 and SPCRP13 were subjected to WES on a Illumina HiSeq2000 at the Integrated Genomics Operation (IGO) at MSKCC (14), and tumor DNA from SPCRP5 and SPCRP11 to Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) sequencing assay (15) targeting all exons and selected regulatory regions and introns of 410 key cancer genes on an Illumina HiSeq2500 (16). Massively parallel sequencing analysis was performed as previously described (Supplementary Methods; refs. 16, 17). WES and MSK-IMPACT data have been submitted to SRA under the accessions SRP066429 and SRP066430, respectively.

SNaPshot genotyping

Representative FFPE sections of each case were reviewed to select the most appropriate tumor-enriched area for analysis. Tumor tissue was manually microdissected from serial 5-μm–thick unstained sections. Total nucleic acid was extracted from FFPE tumor tissue using a modified FormaPure system (Agencourt Bioscience Corporation) as previously described (18). Genotyping analysis of specific mutations in 22 clinically relevant cancer genes (Supplementary Table S2) was performed on six SPCRPs (cases 2, 4, 7–9, and 12) for which insufficient material for massively parallel sequencing was available using a custom Applied Biosystem (ABI) Prism SNaPshot Multiplex platform, as previously reported (18).

Sanger sequencing

For three cases (SPCRP1, 3, and 6) with very limited tissue availability, DNA was subjected to PCR amplification using primer sets encompassing the IDH2 R140 and R172 hotspot residues and the PIK3CA E542/545 and H1047 hotspot residues (Supplementary Table S3), followed by standard Sanger sequencing as previously described (14). All analyses were performed in duplicate. Sequences of the forward and reverse strands were analyzed using MacVector software (MacVector, Inc; ref. 14).

Methylation profiling

Methylation profiling was performed using the Infinium MethylationEPIC Kit (Illumina). DNA samples from five IDH2-mutant SPCRPs (cases 3–5, 7, and 9), one TET2-mutant SPCRP (case 11), and two invasive ductal carcinomas of no special type (IDCs, one ER-positive/HER2-negative and one ER-negative/HER2-negative) were bisulfite-converted using the EZ-96 DNA Methylation Kit (Zymo Research), restored using the Illumina Infinium HD FFPE DNA Restore Kit, and whole-genome amplified (Supplementary Methods). The BeadChips were scanned, and the raw data files containing the fluorescence intensity data for each probe were generated. Data analysis was performed using RnBeads (Supplementary Methods; ref. 19).

DNA transfections and analysis of transgene expression

The human IDH2 (NM_002168) cDNA ORF clone pCMV6-IDH2::Myc-DDK was purchased from Origene (RC201152), and the R172S mutation introduced using the Q5 Site-Directed Mutagenesis Kit (New England Biolabs) following the manufacturer's recommendations. The presence of the correct mutation was confirmed by Sanger sequencing (for primers, see Supplementary Table S3). IDH2 wild-type and mutant (R172S) open reading frames were cloned into the pCMV6-TagRFP or pCMV6-TagGFP vectors to generate pCMV6-IDH2::TagRFP/GFP and pCMV6-IDH2 (R172S)::TagRFP/GFP plasmids, respectively, as previously described (13, 14). Transfections of plasmids were performed using Lipofectamine 3000 reagent according to the manufacturer's guidelines (13). IDH2 and IDH2 R172S expression was verified by immunofluorescence-based mitochondrial colocalization using MitoTracker staining (Thermo Fisher Scientific), quantitative RT-PCR, and whole-cell lysate protein detection.

Quantitative RT-PCR

TaqMan quantitative RT-PCR (qRT-PCR; Life Technologies) was performed for IDH2 (Hs00158033_m1), Twist1 (Hs01675818_s1), SNAI1 (Hs00195591_m1), SNAI2 (Hs00161904_m1), FN1 (Hs01549976_m1), and using GAPDH (Hs99999905_m1) for expression assay normalization, as previously described (5, 20).

2-Hydroxyglutarate assay of tissue samples and cell lines

Ten serial 10 μm–thick representative tumor sections were obtained from SPCRP12 and five IDCs, the latter identified in the pathology database at Massachusetts General Hospital and diagnosis confirmed. Tumors were manually microdissected. 2-Hydroxyglutarate (2HG) was analyzed in lysates from these samples using gas chromatography/mass spectrometry, as previously described (21).

Parental MCF10A (MCF10AP) and MCF10A PIK3CA H1047R (MCF10AH1047R) knock-in cells were obtained from Horizon in December 2014. The identities of the cell lines were confirmed by short tandem repeat profiling using the GenePrint 10 System (Promega) in the IGO at MSKCC three months after receipt. In addition, cells were tested for mycoplasma infection using a PCR-based test (ATCC). Confluent cells (MCF10AP and MCF10AH1047R, expressing vector control, IDH2 wild-type, or IDH2 R172S plasmids) were collected from 6-well plates, and 2HG detection was performed in conditioned media and in cell lysates, as previously reported (22; Supplementary Methods). Fluorometric detection was carried out in triplicate with excitation at 540 nm and emission of 610 nm (Victor X4 Multimode Plate Reader; Supplementary Methods).

Cell proliferation and cell migration assays

MCF10AP and MCF10H1047R cells were transfected with pCMV6-TagRFP empty vector, pCMV6-TagRFP-IDH2WT or pCMV6-TagRFP-IDH2R172S using Lipofectamine 3000 (Invitrogen) as per manufacturer's instructions. After 48 hours, cells were trypsinized, collected and RFP-, RFP-IDH2WT-, and RFP-IDH2R172S–expressing cells selected by flow cytometry sorting. For proliferation, the sorted cells were seeded in 96-well plates (1,000 cells/well) in triplicates. Cell growth was assessed every 24 hours by CellTiter-Blue assay (Promega) and fluorometric detection performed with 560 nm excitation and 590 nm emission using a VICTOR X4 Multilabel Plate Reader (PerkinElmer). For migration, cells were starved for 16 hours in 2% horse serum DMEM/F12 medium without EGF. Cells were seeded (0.5 × 105) in the top compartment of a 24-well Transwell insert (8-μm pore membranes; Corning) and allowed to adhere and migrate at 37°C toward 10% horse serum and EGF-containing DMEM/F12 medium for 16 hours. Cells adherent to the top side of the Transwell membrane were removed, and cells on the inferior side of were fixed in 4% paraformaldehyde (PFA), permeabilized (0.5% Triton X-100 in PBS), and stained with crystal violet (0.3%). Transwell membranes were imaged (EVOS XL Imaging System, Life Technologies) and analyzed by ImageJ.

3D cell culture

3D cell culture assays were carried out as previously described (13, 14, 23, 24). Phase-contrast images of acinar structures were acquired after six days (EVOS XL Imaging System). Size (mm3) of 3D acinar structures was determined using Fiji (ImageJ software) to define Feret diameter (D) and minimum Feret diameter (d) and applying the formula (D × d2)/2 = acini volume. Immunofluorescent staining of 3D acini (12 days) was performed as previously described (24). The structures were fixed in 4% PFA and permeabilized (0.5% Triton X-100 in PBS), stained using antibodies against GM130 (D6B1) and E-cadherin (Cell Signaling Technology), and Alexa Fluor–conjugated secondary antibodies and ProLong Gold antifade reagent with DAPI (Life Technologies), and imaged by confocal analyses [Leica SP5 DM, including an ultraviolet (UV) diode (405 nm), argon laser (458, 476, 488, and 514 nm), 543-nm HeNe laser, and 633-nm HeNe laser]. Polarity assessment was performed with the observer blinded to the genotype of the cell line being analyzed, with apical polarity defined as nuclei at the basal pole (near the periphery of the acini) and Golgi apparatus facing upward (toward the lumen), and reverse polarity as nuclei at the apical pole (toward to the lumen) and Golgi apparatus facing away from the lumen (near the periphery of the acini). A minimum of 100 abluminally located cells were counted per experimental condition in each cell line.

Oxidative stress cell viability assay

A cell viability assay for the assessment of oxidative stress was performed basically as previously described (25). Vector-, IDH2- or IDH2 R172S–expressing MCF10AP and MCF10AH1047R were grown overnight on a 96-well plate at a density of 1 × 104 cells/well, treated with 1 mmol/L hydrogen peroxide (H2O2) in serum-free medium for 48 hours at 37°C, and cell viability was assessed using CellTiter-Blue as described above and reported as compared with absorbance measured in untreated control cells. Fluorometric detection was carried out in triplicate in two independent experiments.

Clinicopathologic characteristics of SPCRP

SPCRP has a very distinctive histologic appearance that allowed us to identify a series of 13 such tumors from our consultation and institutional files. They consisted of solid, circumscribed nodules of columnar epithelial cells, some exhibiting a geographic, jigsaw-like growth pattern (Fig. 1A–C). Many nodules contained fibrovascular cores with foamy histiocytes and a double-layered epithelium without a clear lumen, resulting in a solid papillary appearance (Fig. 1B). The cells in many nodules appeared back-to-back and, in particular, the nuclei were present at the apical rather than basal pole of the cells, creating the impression of reverse polarization (Fig. 1C).

Figure 1.

Histologic features and immunophenotype of SPCRP. A, Circumscribed, solid papillary tumor cell nests infiltrating around and between normal ducts. B, Solid papillary nests with fibrovascular cores containing aggregates of foamy histiocytes. C, Columnar cells with abundant eosinophilic cytoplasm, mild nuclear atypia, and striking reverse nuclear polarity. D, Neoplastic epithelium demonstrating diffuse and strong expression of high molecular weight cytokeratin CK5/6. E, Absence of myoepithelial cells around tumor nests (left) on p63 immunostain with positive internal control (right). F, Absence of ER staining with positive internal control (top left). G, Absence of apical and basal E-cadherin staining with preservation of lateral membrane expression. H, Strong expression of MUC1 in apical membranes. Scale bars, 150 μm.

Figure 1.

Histologic features and immunophenotype of SPCRP. A, Circumscribed, solid papillary tumor cell nests infiltrating around and between normal ducts. B, Solid papillary nests with fibrovascular cores containing aggregates of foamy histiocytes. C, Columnar cells with abundant eosinophilic cytoplasm, mild nuclear atypia, and striking reverse nuclear polarity. D, Neoplastic epithelium demonstrating diffuse and strong expression of high molecular weight cytokeratin CK5/6. E, Absence of myoepithelial cells around tumor nests (left) on p63 immunostain with positive internal control (right). F, Absence of ER staining with positive internal control (top left). G, Absence of apical and basal E-cadherin staining with preservation of lateral membrane expression. H, Strong expression of MUC1 in apical membranes. Scale bars, 150 μm.

Close modal

In all cases, all tumor nodules lacked a surrounding myoepithelial cell layer as assessed by immunohistochemical analysis for p63 (Fig. 1E), calponin, and SMMHC, supporting the invasive nature of these lesions. Tumor cells showed strong cytoplasmic expression of low molecular weight CK7, but also strongly expressed high-molecular weight/basal CK5/6 (Fig. 1D) and CK34βE12. GCDFP-15 and mammaglobin expression were detected in 62% and 58% of cases, respectively, supporting the breast origin of the tumors, whereas thyroglobulin and TTF-1 were negative in all tumors, excluding metastases of a thyroid carcinoma. In 62% of cases, tumor cells were entirely ER-negative (Fig. 1F); ER expression was seen in 1% to 10% of cells in the remaining tumors. PR expression was seen in 15% of cases, and all cases studied were negative for HER2 protein overexpression. AR was focally positive in only one case. In two cases studied, the Ki67 proliferation rate was low (<5%). E-cadherin staining revealed strong lateral membrane expression with absent apical or basal expression (Fig. 1G), suggesting that the epithelium is polarized. MUC1 staining, which highlights the apical membranes of columnar epithelial cells, was identified at the end of the cell closest to the nucleus, indicating that the nuclei were in an abnormal location (apical rather than basal) and creating the impression of reversed cell polarity (Fig. 1H). Clinical data are summarized in Table 1. 

Table 1.

Clinicopathologic characteristics of SPCRP analyzed in this study

12345678910111213
Clinical findings 
 Age, y 68 62 63 79 64 51 64 58 66 65 70 65 65 
 Laterality 
 Size, cm 0.9 0.8 1.2 1.8 0.8 1.4 0.6 0.9 1.5 1.3 1.2 0.9 
 LN metastasis/sampled 0/2 NP 0/2 0/3 0/1 0/3 0/2 0/2 0/4 
 Outcome/mo NED/77 NED/31 NED/30 NED/29 NED/20 NED/37 NED/12 
 Treatment S/XRT S/XRT S/C S/XRT/C 
 Cancer history − − − − − 
Immunohistochemical features 
 CK5/6 Rare + − Focal + Focal + 
 CK7 − NP 
 CK 34βE12 NP − NP 
 CK AE1/AE3 NP NP NP NP 
 p63 − − − − − − − − − − − − − 
 SMMHC − − − − − − − − − − − NP − 
 Calponin − − − − − − − − − NP − − NP 
 ER − − <10% + − − <10% + <10% + − − <10% + <10% + − − 
 PR − − <10% + − − <10% + − − − − − − − 
 HER2 NP − NP NP − − − − − − − − − 
 AR NP − − − − − − − − NP − − Focal + 
 GCDFP-15 Focal + − Focal + Focal + Focal + Focal + − − Focal + − Focal + − Focal + 
 Mammaglobin Focal + Focal + Focal + Focal + − − − − − Focal + NP 
 TTF-1 − − NP − − − − − − − − − − 
 Thyroglobulin NP − NP − − NP − − − − − − − 
12345678910111213
Clinical findings 
 Age, y 68 62 63 79 64 51 64 58 66 65 70 65 65 
 Laterality 
 Size, cm 0.9 0.8 1.2 1.8 0.8 1.4 0.6 0.9 1.5 1.3 1.2 0.9 
 LN metastasis/sampled 0/2 NP 0/2 0/3 0/1 0/3 0/2 0/2 0/4 
 Outcome/mo NED/77 NED/31 NED/30 NED/29 NED/20 NED/37 NED/12 
 Treatment S/XRT S/XRT S/C S/XRT/C 
 Cancer history − − − − − 
Immunohistochemical features 
 CK5/6 Rare + − Focal + Focal + 
 CK7 − NP 
 CK 34βE12 NP − NP 
 CK AE1/AE3 NP NP NP NP 
 p63 − − − − − − − − − − − − − 
 SMMHC − − − − − − − − − − − NP − 
 Calponin − − − − − − − − − NP − − NP 
 ER − − <10% + − − <10% + <10% + − − <10% + <10% + − − 
 PR − − <10% + − − <10% + − − − − − − − 
 HER2 NP − NP NP − − − − − − − − − 
 AR NP − − − − − − − − NP − − Focal + 
 GCDFP-15 Focal + − Focal + Focal + Focal + Focal + − − Focal + − Focal + − Focal + 
 Mammaglobin Focal + Focal + Focal + Focal + − − − − − Focal + NP 
 TTF-1 − − NP − − − − − − − − − − 
 Thyroglobulin NP − NP − − NP − − − − − − − 

Abbreviations: AR, androgen receptor; C, chemotherapy (carboplatin and taxol); CK, cytokeratin; ER, estrogen receptor; GCDFP-15, gross cystic disease fluid protein-15; L, left; LN, lymph node; mo, months; NED, no evidence of disease; NP, not performed; PR, progesterone receptor; R, right; S, surgery; SMMHC, smooth muscle myosin heavy chain; TTF-1, thyroid transcription factor-1; U, unknown; XRT, radiation; y, year.

SPCRP harbors IDH2 hotspot mutations and mutations affecting PI3K pathway canonical genes

Two SPCRPs (cases 10 and 13), for which sufficient tumor and normal DNA samples were available, and two SPCRPs (cases 5 and 11) with sufficient tumor DNA were subjected to WES and the MSK-IMPACT sequencing assay (15), respectively. In cases subjected to WES (cases 10 and 13), the median sequencing depth of coverage was 127× (range, 111–212×), and 45 and 24 nonsynonymous somatic mutations were identified (Supplementary Tables S4 and S5). Cases 5 and 11, subjected to MSK-IMPACT sequencing at a coverage if 734× and 573×, respectively, were found to harbor eight and 26 nonsynonymous somatic mutations, respectively (Supplementary Tables S4 and S5). Sequencing analysis revealed that three of these cases harbored somatic single nucleotide variants (SNV) affecting the R172 residue in the substrate-binding pocket of the isocitrate dehydrogenase 2 (NADP+), mitochondrial (IDH2) gene (Fig. 2A; Supplementary Table S5), the same IDH2 codon targeted by recurrent hotspot mutations in glioma, acute myeloid leukemia (AML), cholangiocarcinoma, and chondrosarcoma (26). In SPCRP11, which lacked an IDH2 somatic mutation, a TET2 Q548* truncating mutation was detected. We also identified mutations affecting canonical genes of the PI3K/AKT/mTOR pathway in all four SPCRPs, with two cases harboring PIK3R1 frameshift mutations and two cases harboring PIK3CA somatic mutations (Fig. 2A; Supplementary Table S5). No other recurrent somatic mutations were found. To validate these findings, we subjected DNA derived from an additional six SPCRPs (cases 2, 4, 7–9, and 12) to SNaPshot profiling assessing mutations in 22 cancer genes (Supplementary Table S2) and from three SPCRPs (cases 1, 3, and 6) to IDH2 and PIK3CA hotspot Sanger sequencing (Supplementary Fig. S1; Supplementary Table S3), which confirmed the presence of IDH2 R172 hotspot mutations in seven cases (70%), and PIK3CA H1047R and E542K hotspot mutations in six cases and one case (78%), respectively. No other hotspot mutations were identified in the SPCRPs subjected to SNaPshot profiling. In total, 10 of 13 (77%) SPCRPs analyzed here harbored an IDH2 R172 mutation, of which five had a concurrent PIK3CA H1047R hotspot mutation, one a concurrent PIK3CA C420R mutation, and two a concurrent PIK3R1 frameshift mutation (Fig. 2A). Given that one of the IDH2 wild-type SPCRPs harbored a truncating mutation in TET2, which encodes for an α-ketoglutarate (αKG)-dependent enzyme that catalyzes cytosine 5-hydroxymethylation resulting in demethylation of DNA (27), we sought to define whether the IDH2 wild-type cases SPCRP1 and SPCRP6 would also harbor mutations affecting this gene; however, both were found to be TET2 wild-type. No histologic differences were observed among the IDH2-mutant, TET2-mutant, and IDH2/TET2 wild-type SPCRPs.

Figure 2.

IDH2 and PI3K pathway mutations, 2HG levels, and methylation profiles of SPCRP. A, Nonsynonymous somatic IDH2, TET2, PIK3CA, and PIK3R1 mutations identified in the 13 SPCRPs studied here by massively parallel sequencing (WES or MSK-IMPACT), SNaPshot, or Sanger sequencing. B, Mutation plot shows the domain structure of IDH2 and the nonsynonymous IDH2 mutations identified in SPCRP and in common forms of breast cancer published by TCGA available from cBioPortal (28). C, 2HG analysis in IDH2-mutant SPCRP (case 12) and IDH wild-type invasive ductal carcinoma of no special type (IDC). D,IDH2/TET2-mutant SPCRPs display global DNA hypermethylation assessed by Infinium MethylationEPIC BeadChip as compared with IDH2/TET2 wild-type IDCs (left). Unsupervised hierarchical clustering using all methylation values/genes of six IDH2/TET2-mutant SPCRPs and two IDH2/TET2 wild-type IDCs using complete linkage and Euclidean distance (right). The 1,000 genes with the highest variance across samples are displayed. Methylation status is color coded according to the legend. E, H3K27me3 immunohistochemical analysis of four IDH2-mutant SPCRPs (top) and four IDH2 wild-type IDCs (bottom). Nuclear immunoreactivity for H3K27me3 was quantified using the H-score. *, P < 0.05; Mann–Whitney U test.

Figure 2.

IDH2 and PI3K pathway mutations, 2HG levels, and methylation profiles of SPCRP. A, Nonsynonymous somatic IDH2, TET2, PIK3CA, and PIK3R1 mutations identified in the 13 SPCRPs studied here by massively parallel sequencing (WES or MSK-IMPACT), SNaPshot, or Sanger sequencing. B, Mutation plot shows the domain structure of IDH2 and the nonsynonymous IDH2 mutations identified in SPCRP and in common forms of breast cancer published by TCGA available from cBioPortal (28). C, 2HG analysis in IDH2-mutant SPCRP (case 12) and IDH wild-type invasive ductal carcinoma of no special type (IDC). D,IDH2/TET2-mutant SPCRPs display global DNA hypermethylation assessed by Infinium MethylationEPIC BeadChip as compared with IDH2/TET2 wild-type IDCs (left). Unsupervised hierarchical clustering using all methylation values/genes of six IDH2/TET2-mutant SPCRPs and two IDH2/TET2 wild-type IDCs using complete linkage and Euclidean distance (right). The 1,000 genes with the highest variance across samples are displayed. Methylation status is color coded according to the legend. E, H3K27me3 immunohistochemical analysis of four IDH2-mutant SPCRPs (top) and four IDH2 wild-type IDCs (bottom). Nuclear immunoreactivity for H3K27me3 was quantified using the H-score. *, P < 0.05; Mann–Whitney U test.

Close modal

To date, IDH2 mutations have not been described in breast cancer, and a reanalysis of breast cancers reported by The Cancer Genome Atlas (TCGA) revealed that only one of the 971 invasive ductal and invasive lobular carcinomas harbored an IDH2 mutation, however affecting a different codon (E345K, Fig. 2B; www.cBioPortal.org, accessed on November 30, 2015; Fig. 2B; refs. 4, 28). Taken together, our results indicate that SPCRPs are uniquely characterized by highly recurrent IDH2 R172 hotspot mutations often in combination with mutations affecting PI3K pathway canonical genes, in particular in the form of PIK3CA H1047R hotspot mutations, and suggest that at least in a subset of SPCRPs lacking IDH2 mutations, somatic genetic alteration affecting TET2 may be present. These observations suggest that SPCRPs may constitute an example of a convergent phenotype stemming from alterations of genes leading to similar epigenetic defects and potentially function genetically in the same pathway (26, 29), as IDH1/IDH2 mutations inhibit TET2 function and are mutually exclusive with TET2 mutations in hematologic malignancies (30).

IDH2 and PIK3CA mutations constitute likely drivers of SPCRP

IDH2 hotspot mutations are enzymatic gain-of-function alterations that lead to an increased conversion of αKG to 2HG. Increased levels of 2HG result in hypermethylation of epigenetic targets and a subsequent block in cellular differentiation (31, 32). We first sought to define whether IDH2 mutations result in accumulation of 2HG oncometabolite in SPCRP as reported in glioblastoma (33) and AML (34) harboring this mutation. We measured intratumoral 2HG by gas chromatography/mass spectrometry in one IDH2-mutant SPCRP (case 12), where adequately preserved tissue was available, and five IDH-wild-type IDCs with available material. The SPCRP harboring the IDH2 R172T mutation demonstrated an elevated 2HG level of 49.36 pmol/μg protein (Fig. 2C). In four IDH2-mutant SPCRPs, tissue samples were inadequate and rendered uninterpretable levels of 2HG (data not shown). All five IDH2-wild-type IDCs harbored undetectable levels of 2HG (Fig. 2C). We next sought to define levels of methylation in IDH2/TET2-mutant SPCRPs, for which DNA samples were available. Genome-wide DNA methylation analysis using the Illumina Infinium MethylationEPIC BeadChip revealed that IDH2/TET2-mutant SPCRPs showed a genome-wide hypermethylation profile as compared with IDH2 wild-type IDCs, consistent with the hypermethylation profile reported in IDH1/IDH2-mutant cancers (Fig. 2D and Supplementary Fig. S2; refs. 30, 35). In fact, hierarchical clustering revealed that IDH2/TET2–mutant SPCRPs clustered together based on their methylation profile, and separate from the two IDH2/TET2 wild-type IDCs analyzed (Fig. 2D). Finally, the protein expression levels of H3K27me3 were assessed by IHC in four IDH2-mutant SPCRPs for which adequate histologic sections were available, and in four IDCs (two ER-positive/HER2-negative and two ER-negative/HER2-negative). Compared with IDH2 wild-type IDCs, IDH2-mutant SPCRPs displayed significantly higher levels of trimethylation of H3K27 (H3K27me3, P = 0.029, Mann–Whitney U test; Fig. 2E), consistent with the results reported in IDH-mutant gliomas (31).

We next tested the functional impact of the IDH2 R172S mutation on the growth and phenotype of nonmalignant breast epithelial cells and investigated potential epistatic interactions between IDH2 R172S and the most frequent PIK3CA (H1047R) mutations concurrently detected in SPCRPs. We used the MCF10A model system, including parental MCF10A cells and MCF10A cells harboring a stable knock-in of the PIK3CA H1047R mutation. As expected, forced expression of wild-type IDH2 (IDH2WT) and R172S mutant IDH2 (IDH2R172S) in parental MCF10A cells (MCF10AP) and in PIK3CA H1047R–mutant MCF10A cells (MCF10AH1047R) resulted in increased IDH2 mRNA expression (Supplementary Fig. S3A) and IDH2 protein mitochondrial localization (Supplementary Fig. S3B). When expressed at similar protein levels, IDH2R172S resulted in significantly higher neomorphic enzymatic activity than IDH2WT based on the analysis of the production and secretion of 2HG, regardless of the presence of the H1047R PIK3CA mutation (Fig. 3A).

Figure 3.

Downstream effects of IDH2 expression in nonmalignant breast epithelial cells. A, Detection of 2HG as a readout of IDH2WT and IDH2R172S enzymatic activity in total protein lysates (left) and in conditioned media (right) of MCF10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S. The same cell lysates from MCF10AP and MCF10AH1047R cells were analyzed for IDH2 protein expression by Western blotting. Total tubulin was used as loading control; ns: not significant; ***, P < 0.001; ****, P < 0.0001; error bars, standard deviation of mean. B, Whole-cell lysates of MCF10AP and MCF10AH1047R cells were analyzed for IDH2, total and phosphorylated RB, E-cadherin, and tubulin protein expression by Western blotting (left), and quantified using near-infrared detection (LI-COR; Odyssey; right); error bars, SD of mean.

Figure 3.

Downstream effects of IDH2 expression in nonmalignant breast epithelial cells. A, Detection of 2HG as a readout of IDH2WT and IDH2R172S enzymatic activity in total protein lysates (left) and in conditioned media (right) of MCF10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S. The same cell lysates from MCF10AP and MCF10AH1047R cells were analyzed for IDH2 protein expression by Western blotting. Total tubulin was used as loading control; ns: not significant; ***, P < 0.001; ****, P < 0.0001; error bars, standard deviation of mean. B, Whole-cell lysates of MCF10AP and MCF10AH1047R cells were analyzed for IDH2, total and phosphorylated RB, E-cadherin, and tubulin protein expression by Western blotting (left), and quantified using near-infrared detection (LI-COR; Odyssey; right); error bars, SD of mean.

Close modal

IDH1 and IDH2 mutations have been shown to constitute drivers of glioma (36, 37), AML (34, 38), and spindle cell hemangioma (39, 40) among others, resulting in a differentiation block and tumorigenesis (31, 32). Although in our transient transfection system, no statistically significant differences in the expression levels of H3K27 and H3K9 trimethylation were observed upon forced expression of IDH2WT or IDH2R172S as compared with cells expressing empty vector (Supplementary Fig. S4), expression of IDH2R172S resulted in changes in expression of cell-cycle and epithelial–mesenchymal transition (EMT) markers in the nonmalignant breast epithelial cells, MCF10A. Forced expression of IDH2R172S in MCF10AH1047R cells led to significantly increased phosphorylation of RB1 protein compared with empty vector, as detected by immunoblot (Fig. 3B). Consistent with these observations, we observed a significant increase in cell growth upon forced expression of IDH2WT and IDH2R172S in MCF10AH1047R cells (Fig. 4A), as well as a significant increase in migration of MCF10AWT and MCF10AH1047R cells expressing R172S-mutant IDH2 (Fig. 4B). Furthermore, we observed that forced expression of IDH2WT and IDH2R172S led to a reduction of E-cadherin protein expression in MCF10AH1047R cells (Fig. 3B) and resulted in significantly increased levels of EMT markers in MCF10A cells harboring wild-type or H1047R-mutant PIK3CA (Fig. 4C).

Figure 4.

Effects of IDH2 on proliferation, migration, mesenchymal marker expression, and oxidative stress in nonmalignant breast epithelial cells. A, Effect of empty vector control (blue), IDH2WT (green), or IDH2R172S (red) expression on survival and growth of MCF10AP (top) and MCF-10AH1047R (bottom), assessed by MTT assay. **, P < 0.01; error bars, SD of mean. B, Effect of empty vector control, IDH2WT, or IDH2R172S expression on Transwell migration of MCF10AP (top) and MCF-10AH1047R (bottom). *, P < 0.05; error bars, SD of mean. C, Relative transcript expression for mesenchymal markers in MCF10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S as determined by qRT-PCR. Two-fold increase above empty vector control for each cell line was used as a cut-off point. D, Effect of H2O2 treatment of MCF-10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S on cell viability fraction, assessed by MTT assay as compared with untreated control cells. ns, not significant; **, P < 0.01; ***, P < 0.001; error bars, SD of mean.

Figure 4.

Effects of IDH2 on proliferation, migration, mesenchymal marker expression, and oxidative stress in nonmalignant breast epithelial cells. A, Effect of empty vector control (blue), IDH2WT (green), or IDH2R172S (red) expression on survival and growth of MCF10AP (top) and MCF-10AH1047R (bottom), assessed by MTT assay. **, P < 0.01; error bars, SD of mean. B, Effect of empty vector control, IDH2WT, or IDH2R172S expression on Transwell migration of MCF10AP (top) and MCF-10AH1047R (bottom). *, P < 0.05; error bars, SD of mean. C, Relative transcript expression for mesenchymal markers in MCF10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S as determined by qRT-PCR. Two-fold increase above empty vector control for each cell line was used as a cut-off point. D, Effect of H2O2 treatment of MCF-10AP and MCF10AH1047R cells expressing empty vector control, IDH2WT, or IDH2R172S on cell viability fraction, assessed by MTT assay as compared with untreated control cells. ns, not significant; **, P < 0.01; ***, P < 0.001; error bars, SD of mean.

Close modal

Given that cancer cells can avoid cell death by inhibiting the initial deleterious effects of oxidative stress (25, 41), we sought to define the impact of IDH2R172S on the viability of nonmalignant breast epithelial cells. In MCF10AP cells treated with exogenous H2O2, forced expression of wild-type, but not mutant IDH2, displayed a higher viability than empty vector expression (Fig. 4D). In H2O2-treated MCF10AH1047R cells, however, forced expression of both wild-type and mutant IDH2 resulted in increased viability compared with the empty vector control (Fig. 4D). These results suggest that IDH2R172S may protect against reactive oxygen species (ROS) in nonmalignant breast epithelial cells, consistent with observations described previously for IDH1 and IDH2 in other nonneoplastic tissue types (25, 41).

We next sought to define the impact of IDH2R172S on the growth and glandular architecture of MCF10A cells grown in a 3D model system, which has been previously used to assess the oncogenic properties of somatic mutations (13, 23, 42). Forced expression of IDH2R172S in MCF10AP and MCF10AH1047R cells resulted in acinar structures that were significantly larger than those observed in MCF10A cells transfected with empty vector or IDH2WT (Fig. 5A), a phenotype previously reported to be elicited by the expression of bona fide oncoproteins in this model system (13, 23, 42). In fact, IDH2R172S expression in MCF10AH1047R cells led to the formation of anastomosing solid cell nests/cell nodules (Fig. 5A and Supplementary Fig. S5). Consistent with the reverse polarization observed in the primary tumors, IDH2R172S expression in MCF10A cells grown in the 3D system also resulted in an increase in cells displaying a reversed nuclear polarity, as defined by expression of adhesion molecule E-cadherin (CDH1) and apical Golgi marker GM130. This reversed nuclear polarity phenotype was particularly evident in MCF10AH1047R cells expressing IDH2R172S (Fig. 5B). Taken together, our data provide evidence that IDH2R172S and PIK3CAH1047R hotspot mutations constitute likely drivers of SPCRP, and that together these somatic genetic alterations are likely sufficient, but not necessarily required, to cause its unusual reverse polarization phenotype.

Figure 5.

Impact of IDH2 mutations on glandular architecture and polarity of nonmalignant breast epithelial cells. A, Impact of forced expression of IDH2WT or IDH2R172S on glandular architecture of MCF10AP (top) and MCF10AH1047R (bottom) cells grown in 3D basement membrane cultures (scale bar, 0.1 mm). The volume of the MCF10AP and MCF10AH1047R acinar structures expressing IDH2WT and IDH2R172S was quantified; *, P < 0.05; ***, P < 0.001; ****, P < 0.0001; error bars, SD of mean. B, Immunofluorescent analysis of MCF10AP and MCF10AH1047R acinar structures expressing IDH2WT or IDH2R172S with antibodies against E-cadherin (red), cis-Golgi marker GM130 (green), and nucleus (DAPI, blue; scale bars, 25 μm; left). Arrows, cells with reverse polarity. MCF10AP and MCF10AH1047R acinar structures showing either apical or reversed polarity were quantified. MCF10AH1047R IDH2R172S acini showed significantly more frequently inverted polarity than MCF10AH1047R vector control and MCF10AH1047R IDH2WT acini (*, P < 0.05 each; t test).

Figure 5.

Impact of IDH2 mutations on glandular architecture and polarity of nonmalignant breast epithelial cells. A, Impact of forced expression of IDH2WT or IDH2R172S on glandular architecture of MCF10AP (top) and MCF10AH1047R (bottom) cells grown in 3D basement membrane cultures (scale bar, 0.1 mm). The volume of the MCF10AP and MCF10AH1047R acinar structures expressing IDH2WT and IDH2R172S was quantified; *, P < 0.05; ***, P < 0.001; ****, P < 0.0001; error bars, SD of mean. B, Immunofluorescent analysis of MCF10AP and MCF10AH1047R acinar structures expressing IDH2WT or IDH2R172S with antibodies against E-cadherin (red), cis-Golgi marker GM130 (green), and nucleus (DAPI, blue; scale bars, 25 μm; left). Arrows, cells with reverse polarity. MCF10AP and MCF10AH1047R acinar structures showing either apical or reversed polarity were quantified. MCF10AH1047R IDH2R172S acini showed significantly more frequently inverted polarity than MCF10AH1047R vector control and MCF10AH1047R IDH2WT acini (*, P < 0.05 each; t test).

Close modal

Here, we characterize the morphologic, immunohistochemical, and genomic profile of SPCRP, a rare and histologically distinct subtype of invasive breast carcinoma. Through whole exome and targeted massively parallel sequencing analysis, we have identified IDH2 hotspot and TET2 truncating mutations in 77% and 8% of these cases, respectively. In fact, SPCRPs may constitute an example of a convergent phenotype as IDH2 and TET2 mutations lead to similar epigenetic defects (26, 29). This is the first report of IDH2 hotspot mutations detected in breast cancer, expanding the spectrum of solid tumors and hematologic malignancies in which IDH mutations may drive tumorigenesis (43). The high frequency of IDH2 mutations found in SPCRP highlights the association between genotype and the unique tumor morphology (29). In addition, mutations affecting canonical genes of the PI3K pathway were found in 85% of SPCRPs, including 54% harboring PIK3CA H1047R mutations, a common genetic alteration in breast cancer that may be enriched in this subset of tumors (44).

A striking feature of SPCRP is the unique epithelial morphology of a double layer of columnar cells with apical nuclei. The absence of expression of E-cadherin, a protein known to play an important role in cell polarity, in apical and/or basal membranes combined with strong MUC1 staining of apical membranes indicates that the cells are indeed polarized. The presence of nuclei near the apical membrane indicates reverse nuclear polarization, and loss of apical polarity was also observed in our cell line models, in particular in cells expressing mutant forms of IDH2 and PIK3CA, suggesting that this phenotype may result from epistatic interactions between mutations affecting these genes. Although our findings provide a novel example of a genotypic–phenotypic correlation in breast cancer, further analyses are required to define the genetic basis of the reverse polarity in SPCRPs lacking IDH2, TET2, and/or PI3K pathway gene mutations.

The mechanism by which IDH1 or IDH2 mutations cause human cancer remains to be fully elucidated. It has been found, however, that these alterations lead to a gain-of-function enzymatic activity that allows NADPH-dependent reduction of αKG to 2HG within tumor cells (33, 34), which inhibits αKG-dependent dioxygenases and alters genome-wide histone and DNA methylation, cell differentiation and survival, and extracellular matrix maturation (26, 31, 45). High 2HG concentrations disrupt TET2 catalytic function and prevent hydroxylation of 5-methylcytosine, resulting in attenuated TET2-dependent demethylation of DNA (30, 45). Akin to IDH-mutant glioma, AML and common forms of breast carcinoma harboring IDH1 mutations (33, 34, 46), we found high concentrations of intratumoral 2HG in the IDH2-mutant SPCRP successfully tested; the use of archival material makes this analysis challenging as formalin fixation and paraffin embedding may lead to oncometabolite loss (21). In addition, we observed global DNA hypermethylation and H3K27 trimethylation in IDH2/TET2–mutant SPCRPs as compared with IDH2 wild-type IDCs, consistent with those reported in IDH1/IDH2–mutant cancers (26, 30, 31, 35). IDH1 and IDH2 have been described previously to protect cells from ROS (25, 41). We observed that IDH2R172S provided ROS protection in MCF10AH1047R but not in MCF10AP cells. Further studies are warranted to assess the potential mechanistic interaction between mutant IDH2, mutant PI3K and ROS, in particular because intracellular ROS levels have been reported to affect the PI3K pathway (for a review, see ref. 47).

IDH2 mutations in SPCRP may serve as a novel target for therapeutic intervention in breast cancer. Acquired enzymatic activity resulting in 2HG accumulation is specific to tumor cells, making mutant-specific small-molecule inhibitors an attractive alternative to chemotherapy regimens with systemic toxic effects. IDH2 inhibitors have now entered phase I clinical trials for the treatment of AML with favorable safety profiles and durable clinical activity (48). Clinical benefit in the solid tumor patient population has yet to be demonstrated.

This study has important limitations. First, owing to the rarity of SPCRP, we studied 13 bona fide cases of this entity. Despite the small sample size, this is the largest collection of SPCRPs reported to date. Second, most samples were individual contributions from different institutions; hence, we were unable to perform a survival analysis to ascertain the impact of IDH2 mutations on the outcome of SPCRP. Importantly, given that none of the patients included in this study developed a distant relapse during the follow-up period available and that less than 25% of SPCRPs lack IDH2 hotspot mutations, it is unlikely that these mutations would be of prognostic significance. Third, we were unable to identify the driver genetic alterations in two SPCRPs lacking IDH2 hotspot and TET2 mutations by Sanger sequencing; however, due to the rarity of IDH2/TET2–wild-type SPCRPs and the limited material available from the cases analyzed, we were unable to sequence the entire coding region of TET2 in the remaining cases. Further studies of IDH2 wild-type SPCRPs are warranted.

Despite these limitations, here we have identified somatic IDH2 hotspot mutations or TET2 mutations in conjunction with mutations affecting PI3K pathway genes in SPCRP, validating this rare breast cancer as a unique clinicopathologic entity underpinned by a distinctive constellation of somatic mutations. Detection of IDH2 mutations may serve as an ancillary marker for the diagnosis of SPCRP. Broad-base genetic profiling in breast cancer patients who develop progressive disease with this tumor type may efficiently identify those eligible for clinical trials for IDH inhibition.

S. Pusch is a patent holder of 2 HG assay patent. A.J. Iafrate has ownership interest (including patents) in ArcherDX and is a consultant/advisory board member for Roche. No potential conflicts of interest were disclosed by the other authors.

The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Conception and design: S. Chiang, B. Weigelt, H.-C. Wen, A.J. Iafrate, J.S. Reis-Filho, S.J. Schnitt

Development of methodology: H.-C. Wen, A. Raghavendra, S. Piscuoglio, A.A. Jungbluth, A.v. Deimling, A.J. Iafrate, S.J. Schnitt

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Chiang, H.-C. Wen, F. Pareja, L.G. Martelotto, T. Basili, A. Li, F.C. Geyer, S. Piscuoglio, A.A. Jungbluth, J. Balss, G.M. Baker, K.S. Cole, J.M. Batten, J.D. Marotti, J. Serrano, K.P. Siziopikou, S. Lu, X. Liu, T. Hammour, E. Brogi, M. Snuderl, A.J. Iafrate, J.S. Reis-Filho, S.J. Schnitt

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Chiang, B. Weigelt, H.-C. Wen, A. Raghavendra, L.G. Martelotto, K.A. Burke, S. Piscuoglio, C.K.Y. Ng, J. Balss, R.S. Lim, A.J. Iafrate, J.S. Reis-Filho, S.J. Schnitt

Writing, review, and/or revision of the manuscript: S. Chiang, B. Weigelt, H.-C. Wen, A. Raghavendra, L.G. Martelotto, K.A. Burke, S. Piscuoglio, A.v. Deimling, J.D. Marotti, K.P. Siziopikou, E. Brogi, M. Snuderl, A.J. Iafrate, J.S. Reis-Filho, S.J. Schnitt

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Pusch, A.v. Deimling, B.L. McCalip, J. Serrano, A.J. Iafrate, S.J. Schnitt

Study supervision: A.J. Iafrate, S.J. Schnitt

Other (provided one of the breast cases involved in this study): H.-C. Soh

S. Piscuoglio was funded in part by a Susan G. Komen Postdoctoral Fellowship Grant (PDF14298348). J.S. Reis-Filho is funded in part by BCRF, and M. Snuderl is supported by the Friedberg Charitable Foundation. S. Chiang, B. Weigelt, H.-C. Wen, F. Pareja, A. Raghavendra, L.G. Martelotto, K.A. Burke, T. Basili, A. Li, F.C. Geyer, S. Piscuoglio, C.K.Y. Ng, A.A. Jungbluth, R.S. Lim, E. Brogi, and J.S. Reis-Filho were funded in part by a NIH/NCI Cancer Center Support Grant (P30 CA008748).

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.
Lakhani
SR
,
Ellis
IO
,
Schnitt
SJ
,
Tan
PH
,
van de Vijver
MJ
,
editors
.
WHO Classification of Tumours of the Breast
.
Lyon, France
:
IARC
; 
2012
.
2.
Ng
CK
,
Schultheis
AM
,
Bidard
FC
,
Weigelt
B
,
Reis-Filho
JS
. 
Breast cancer genomics from microarrays to massively parallel sequencing: paradigms and new insights
.
J Natl Cancer Inst
2015
;
107
:
djv015
.
3.
Weigelt
B
,
Reis-Filho
JS
. 
Histological and molecular types of breast cancer: is there a unifying taxonomy?
Nat Rev Clin Oncol
2009
;
6
:
718
30
.
4.
Ciriello
G
,
Gatza
ML
,
Beck
AH
,
Wilkerson
MD
,
Rhie
SK
,
Pastore
A
, et al
Comprehensive molecular portraits of invasive lobular breast cancer
.
Cell
2015
;
163
:
506
19
.
5.
Martelotto
LG
,
De Filippo
MR
,
Ng
CK
,
Natrajan
R
,
Fuhrmann
L
,
Cyrta
J
, et al
Genomic landscape of adenoid cystic carcinoma of the breast
.
J Pathol
2015
;
237
:
179
89
.
6.
Persson
M
,
Andren
Y
,
Mark
J
,
Horlings
HM
,
Persson
F
,
Stenman
G
. 
Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck
.
Proc Natl Acad Sci U S A
2009
;
106
:
18740
4
.
7.
Tognon
C
,
Knezevich
SR
,
Huntsman
D
,
Roskelley
CD
,
Melnyk
N
,
Mathers
JA
, et al
Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma
.
Cancer Cell
2002
;
2
:
367
76
.
8.
Colella
R
,
Guerriero
A
,
Giansanti
M
,
Sidoni
A
,
Bellezza
G
. 
An additional case of breast tumor resembling the tall cell variant of papillary thyroid carcinoma
.
Int J Surg Pathol
2015
;
23
:
217
20
.
9.
Masood
S
,
Davis
C
,
Kubik
MJ
. 
Changing the term "breast tumor resembling the tall cell variant of papillary thyroid carcinoma" to "tall cell variant of papillary breast carcinoma"
.
Adv Anat Pathol
2012
;
19
:
108
10
.
10.
Hameed
O
,
Perry
A
,
Banerjee
R
,
Zhu
X
,
Pfeifer
JD
. 
Papillary carcinoma of the breast lacks evidence of RET rearrangements despite morphological similarities to papillary thyroid carcinoma
.
Mod Pathol
2009
;
22
:
1236
42
.
11.
Budwit-Novotny
DA
,
McCarty
KS
,
Cox
EB
,
Soper
JT
,
Mutch
DG
,
Creasman
WT
, et al
Immunohistochemical analyses of estrogen receptor in endometrial adenocarcinoma using a monoclonal antibody
.
Cancer Res
1986
;
46
:
5419
25
.
12.
Weigelt
B
,
Warne
PH
,
Downward
J
. 
PIK3CA mutation, but not PTEN loss of function, determines the sensitivity of breast cancer cells to mTOR inhibitory drugs
.
Oncogene
2011
;
30
:
3222
33
.
13.
Ng
CK
,
Martelotto
LG
,
Gauthier
A
,
Wen
HC
,
Piscuoglio
S
,
Lim
RS
, et al
Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification
.
Genome Biol
2015
;
16
:
107
.
14.
Weinreb
I
,
Piscuoglio
S
,
Martelotto
LG
,
Waggott
D
,
Ng
CK
,
Perez-Ordonez
B
, et al
Hotspot activating PRKD1 somatic mutations in polymorphous low-grade adenocarcinomas of the salivary glands
.
Nat Genet
2014
;
46
:
1166
9
.
15.
Cheng
DT
,
Mitchell
TN
,
Zehir
A
,
Shah
RH
,
Benayed
R
,
Syed
A
, et al
Memorial Sloan Kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology
.
J Mol Diagn
2015
;
17
:
251
64
.
16.
Piscuoglio
S
,
Ng
CK
,
Murray
M
,
Burke
KA
,
Edelweiss
M
,
Geyer
FC
, et al
Massively parallel sequencing of phyllodes tumours of the breast reveals actionable mutations, and TERT promoter hotspot mutations and TERT gene amplification as likely drivers of progression
.
J Pathol
2016
;
238
:
508
18
.
17.
Schultheis
AM
,
Ng
CK
,
De Filippo
MR
,
Piscuoglio
S
,
Macedo
GS
,
Gatius
S
, et al
Massively parallel sequencing-based clonality analysis of synchronous endometrioid endometrial and ovarian carcinomas
.
J Natl Cancer Inst
2016
;
108
:
djv427
.
18.
Dias-Santagata
D
,
Akhavanfard
S
,
David
SS
,
Vernovsky
K
,
Kuhlmann
G
,
Boisvert
SL
, et al
Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine
.
EMBO Mol Med
2010
;
2
:
146
58
.
19.
Assenov
Y
,
Muller
F
,
Lutsik
P
,
Walter
J
,
Lengauer
T
,
Bock
C
. 
Comprehensive analysis of DNA methylation data with RnBeads
.
Nat Methods
2014
;
11
:
1138
40
.
20.
Piscuoglio
S
,
Ng
CK
,
Martelotto
LG
,
Eberle
CA
,
Cowell
CF
,
Natrajan
R
, et al
Integrative genomic and transcriptomic characterization of papillary carcinomas of the breast
.
Mol Oncol
2014
;
8
:
1588
602
.
21.
Sahm
F
,
Capper
D
,
Pusch
S
,
Balss
J
,
Koch
A
,
Langhans
CD
, et al
Detection of 2-hydroxyglutarate in formalin-fixed paraffin-embedded glioma specimens by gas chromatography/mass spectrometry
.
Brain Pathol
2012
;
22
:
26
31
.
22.
Balss
J
,
Pusch
S
,
Beck
AC
,
Herold-Mende
C
,
Kramer
A
,
Thiede
C
, et al
Enzymatic assay for quantitative analysis of (D)-2-hydroxyglutarate
.
Acta Neuropathol
2012
;
124
:
883
91
.
23.
Debnath
J
,
Brugge
JS
. 
Modelling glandular epithelial cancers in three-dimensional cultures
.
Nat Rev Cancer
2005
;
5
:
675
88
.
24.
Debnath
J
,
Muthuswamy
SK
,
Brugge
JS
. 
Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures
.
Methods
2003
;
30
:
256
68
.
25.
Someya
S
,
Yu
W
,
Hallows
WC
,
Xu
J
,
Vann
JM
,
Leeuwenburgh
C
, et al
Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction
.
Cell
2010
;
143
:
802
12
.
26.
Yang
H
,
Ye
D
,
Guan
KL
,
Xiong
Y
. 
IDH1 and IDH2 mutations in tumorigenesis: mechanistic insights and clinical perspectives
.
Clin Cancer Res
2012
;
18
:
5562
71
.
27.
Ko
M
,
An
J
,
Pastor
WA
,
Koralov
SB
,
Rajewsky
K
,
Rao
A
. 
TET proteins and 5-methylcytosine oxidation in hematological cancers
.
Immunol Rev
2015
;
263
:
6
21
.
28.
Gao
J
,
Aksoy
BA
,
Dogrusoz
U
,
Dresdner
G
,
Gross
B
,
Sumer
SO
, et al
Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal
.
Sci Signal
2013
;
6
:
pl1
.
29.
Ashworth
A
,
Lord
CJ
,
Reis-Filho
JS
. 
Genetic interactions in cancer progression and treatment
.
Cell
2011
;
145
:
30
8
.
30.
Figueroa
ME
,
Abdel-Wahab
O
,
Lu
C
,
Ward
PS
,
Patel
J
,
Shih
A
, et al
Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation
.
Cancer Cell
2010
;
18
:
553
67
.
31.
Lu
C
,
Ward
PS
,
Kapoor
GS
,
Rohle
D
,
Turcan
S
,
Abdel-Wahab
O
, et al
IDH mutation impairs histone demethylation and results in a block to cell differentiation
.
Nature
2012
;
483
:
474
8
.
32.
Turcan
S
,
Rohle
D
,
Goenka
A
,
Walsh
LA
,
Fang
F
,
Yilmaz
E
, et al
IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype
.
Nature
2012
;
483
:
479
83
.
33.
Dang
L
,
White
DW
,
Gross
S
,
Bennett
BD
,
Bittinger
MA
,
Driggers
EM
, et al
Cancer-associated IDH1 mutations produce 2-hydroxyglutarate
.
Nature
2009
;
462
:
739
44
.
34.
Ward
PS
,
Patel
J
,
Wise
DR
,
Abdel-Wahab
O
,
Bennett
BD
,
Coller
HA
, et al
The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate
.
Cancer Cell
2010
;
17
:
225
34
.
35.
Kernytsky
A
,
Wang
F
,
Hansen
E
,
Schalm
S
,
Straley
K
,
Gliser
C
, et al
IDH2 mutation-induced histone and DNA hypermethylation is progressively reversed by small-molecule inhibition
.
Blood
2015
;
125
:
296
303
.
36.
Parsons
DW
,
Jones
S
,
Zhang
X
,
Lin
JC
,
Leary
RJ
,
Angenendt
P
, et al
An integrated genomic analysis of human glioblastoma multiforme
.
Science
2008
;
321
:
1807
12
.
37.
Yan
H
,
Parsons
DW
,
Jin
G
,
McLendon
R
,
Rasheed
BA
,
Yuan
W
, et al
IDH1 and IDH2 mutations in gliomas
.
N Engl J Med
2009
;
360
:
765
73
.
38.
Mardis
ER
,
Ding
L
,
Dooling
DJ
,
Larson
DE
,
McLellan
MD
,
Chen
K
, et al
Recurring mutations found by sequencing an acute myeloid leukemia genome
.
N Engl J Med
2009
;
361
:
1058
66
.
39.
Amary
MF
,
Bacsi
K
,
Maggiani
F
,
Damato
S
,
Halai
D
,
Berisha
F
, et al
IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours
.
J Pathol
2011
;
224
:
334
43
.
40.
Pansuriya
TC
,
van Eijk
R
,
d'Adamo
P
,
van Ruler
MA
,
Kuijjer
ML
,
Oosting
J
, et al
Somatic mosaic IDH1 and IDH2 mutations are associated with enchondroma and spindle cell hemangioma in Ollier disease and Maffucci syndrome
.
Nat Genet
2011
;
43
:
1256
61
.
41.
Itsumi
M
,
Inoue
S
,
Elia
AJ
,
Murakami
K
,
Sasaki
M
,
Lind
EF
, et al
Idh1 protects murine hepatocytes from endotoxin-induced oxidative stress by regulating the intracellular NADP(+)/NADPH ratio
.
Cell Death Differ
2015
;
22
:
1837
45
.
42.
Bose
R
,
Kavuri
SM
,
Searleman
AC
,
Shen
W
,
Shen
D
,
Koboldt
DC
, et al
Activating HER2 mutations in HER2 gene amplification negative breast cancer
.
Cancer Discov
2013
;
3
:
224
37
.
43.
Dang
L
,
Yen
K
,
Attar
EC
. 
IDH mutations in cancer and progress toward development of targeted therapeutics
.
Ann Oncol
2016
;
27
:
599
608
.
44.
Cancer Genome Atlas Network
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
45.
Xu
W
,
Yang
H
,
Liu
Y
,
Yang
Y
,
Wang
P
,
Kim
SH
, et al
Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases
.
Cancer Cell
2011
;
19
:
17
30
.
46.
Fathi
AT
,
Sadrzadeh
H
,
Comander
AH
,
Higgins
MJ
,
Bardia
A
,
Perry
A
, et al
Isocitrate dehydrogenase 1 (IDH1) mutation in breast adenocarcinoma is associated with elevated levels of serum and urine 2-hydroxyglutarate
.
Oncologist
2014
;
19
:
602
7
.
47.
Sullivan
LB
,
Chandel
NS
. 
Mitochondrial reactive oxygen species and cancer
.
Cancer Metab
2014
;
2
:
17
.
48.
Stein
EM
. 
Molecular pathways: IDH2 mutations-co-opting cellular metabolism for malignant transformation
.
Clin Cancer Res
2016
;
22
:
16
9
.