Purpose:

To identify clinically relevant mechanisms of resistance to ER-directed therapies in ER+ breast cancer.

Experimental Design:

We conducted a genome-scale functional screen spanning 10,135 genes to investigate genes whose overexpression confer resistance to selective estrogen receptor degraders. In parallel, we performed whole-exome sequencing in paired pretreatment and postresistance biopsies from 60 patients with ER+ metastatic breast cancer who had developed resistance to ER-targeted therapy. Furthermore, we performed experiments to validate resistance genes/pathways and to identify drug combinations to overcome resistance.

Results:

Pathway analysis of candidate resistance genes demonstrated that the FGFR, ERBB, insulin receptor, and MAPK pathways represented key modalities of resistance. The FGFR pathway was altered via FGFR1, FGFR2, or FGF3 amplifications or FGFR2 mutations in 24 (40%) of the postresistance biopsies. In 12 of the 24 postresistance tumors exhibiting FGFR/FGF alterations, these alterations were acquired or enriched under the selective pressure of ER-directed therapy. In vitro experiments in ER+ breast cancer cells confirmed that FGFR/FGF alterations led to fulvestrant resistance as well as cross-resistance to the CDK4/6 inhibitor palbociclib. RNA sequencing of resistant cell lines demonstrated that FGFR/FGF induced resistance through ER reprogramming and activation of the MAPK pathway. The resistance phenotypes were reversed by FGFR inhibitors, a MEK inhibitor, and/or a SHP2 inhibitor.

Conclusions:

Our results suggest that FGFR pathway is a distinct mechanism of acquired resistance to ER-directed therapy that can be overcome by FGFR and/or MAPK pathway inhibitors.

Translational Relevance

A genome-scale overexpression screen revealed a broad spectrum of resistance mechanisms against SERDs, which can provide a resource for researchers studying resistance to estrogen receptor (ER)–directed therapies as well as the biology of ER dependencies in ER+ breast cancer. We demonstrate that activating FGFR/FGF alterations are a mechanism of acquired resistance to ER-directed therapies and CDK4/6 inhibitors in ER+ metastatic breast cancer and can be overcome by combination therapy targeting both the ER and the FGFR pathway. The detection of targetable, clonally acquired genetic alterations in metastatic tumor biopsies highlights the value of serial tumor testing to dissect mechanisms of resistance in human breast cancer and its potential application in directing clinical management.

Approximately 70% of breast cancers express the estrogen receptor (ER), and estrogen signaling drives breast cancer cell growth and progression (1). Although endocrine therapies, including tamoxifen, aromatase inhibitors (AI), and the selective ER degrader (SERD) fulvestrant have improved survival for patients with ER+ breast cancer, within the metastatic setting resistance to endocrine therapies is nearly universal (2).

Although various resistance mechanisms have been proposed for tamoxifen and AI resistance, including loss or modification in ER expression (ESR1-activating mutations and ESR1 fusions; refs. 3–7), and regulation of alternative signal transduction pathways (PI3K/AKT/mTOR and EGFR/ERBB2/MAPK; refs. 8–10), mechanisms of resistance to SERDs remain understudied. Mechanisms of endocrine resistance identified in patients include acquired mutations in the ER itself (4–7), acquired activating mutations in ERBB2 (HER2; refs. 11, 12), loss of function of NF1 (13), and other alterations in MAPK pathway genes (14). Additional clinically relevant mechanisms remain to be identified.

Gain-of-function screens have played a pivotal role in identification of resistance mechanisms to targeted therapies in various cancer types (15–17). In breast cancer, several functional screen studies identified IGF1R, KRAS, and ESR1 as mechanisms of resistance to tamoxifen and/or estrogen deprivation (18–20). However, genome-scale functional screens for SERD resistance have not been reported.

We conducted a genome-scale gain-of-function screen in ER+ breast cancer cells spanning 17,255 overexpressed lentiviral open reading frames (ORF) to investigate genes whose overexpression was sufficient to confer resistance to the SERDs fulvestrant and GDC-0810 (21). In parallel, we sought to identify endocrine resistance mechanisms of clinical significance through genomic profiling of paired pretreatment and posttreatment tumor samples from 60 patients with ER+ metastatic breast cancer (MBC) who developed resistance to endocrine therapy.

Cell culture

293T, T47D, and MCF7 cells were obtained directly from ATCC as frozen vials and were cultured as described in the Supplementary Methods. MCF7 and T47D were validated by Western blotting for ER and HER2 (based on known genotype). We used cell lines of passage number ranging from 4 to 15 in the described experiments. All cell lines tested negative for Mycoplasma contamination by ATCC Universal Mycoplasma Detection Kit. The last test was performed in November 2019.

Genome-scale gain-of-function screen

The pooled lentiviral ORF library hORFeome (22) consists of 17,255 barcoded human ORFs, corresponding to 10,135 distinct human genes with at least 99% nucleotide and protein match. These ORFs were cloned into pLX317 vector and pooled together for transfection into 293T cells to make pooled lentivirus (with 2nd generation packaging plasmids). In 6-well plates, pooled lentivirus was infected in cells to achieve approximately 50% infection rate and ensure approximately 1,000 infected cells per ORF for 17,255 ORFs. Media was supplemented with 4 μg/mL polybrene (Thermo Fisher Scientific # TR1003G) to boost transfection efficiency. After infection, cells were pooled and selected with 1.5 μg/mL puromycin for 5 days. Upon completion of selection, cells were plated for three different drug conditions: DMSO, 100 nmol/L fulvestrant, 1 μmol/L GDC-0810. There were three replicates for each condition screened. A subset of cells was saved for sequencing as early time point (ETP) samples to confirm ORF representation. Infected cells were passaged upon confluency and maintained in DMSO or drugs for 21 days to allow sufficient time for cells carrying resistance to be enriched from the population. At the end of the time course, cells were harvested for isolating genomic DNA as late time point samples (LTP). All genomic DNA samples were amplified with PCR primers flanking the ORF region and sequenced. The ORF representation at the final harvesting (LTP) is compared to the representation of ORFs in cells collected before drug addition (ETP). Cells carrying ORFs that are driving resistance will grow and gradually enrich the population and therefore, will be over-represented in the sequencing data for the final passage compared to the early time point. An ORF with significant enrichment (a Z-score >3) is defined as a resistance candidate gene. A secondary validation screen was performed as described in the Supplementary Methods.

Patients and tumor samples

Prior to any study procedures, all patients provided written informed consent for research biopsies and whole-exome sequencing (WES) of tumor and normal DNA, as approved by the Dana-Farber/Harvard Cancer Center Institutional Review Board (DF/HCC Protocol 05-246). Metastatic core biopsies were obtained from patients and samples were immediately snap frozen in OCT and stored in −80°C. Archival FFPE blocks of primary tumor samples were also obtained. A blood sample was obtained during the course of treatment, and whole blood was stored at −80°C until DNA extraction from peripheral blood mononuclear cells (for germline DNA) was performed. In a few instances, cell free DNA was obtained from plasma for circulating tumor DNA analysis, as described previously (23). The studies were conducted in accordance with U.S. Common Rule for ethical guidelines.

WES analysis

DNA was extracted from primary tumors, metastatic tumors, plasma, and peripheral blood mononuclear cells (for germline DNA) from all patients and WES was performed, as detailed in the Supplementary Methods. Sequencing data were analyzed using tools to identify somatic point mutations and small insertions/deletions (indels), and copy number changes using established algorithms (see Supplementary Methods).

To better measure segment-specific copy number, we subtracted the genome ploidy for each sample to compute copy number above ploidy (CNAP). CNAP of at least 3 are considered as amplifications (AMP); CNAP below 3 are considered low amplification and ignored in our analysis. CNAP of at least 6 are considered high amplifications (HighAMP), and CNAP of at least 9 and fewer than 100 genes (24) is considered very high focal amplification (FocalAMP).

The evolutionary classification of amplifications accounts for the magnitude of the observed copy-number difference between the pretreatment and the posttreatment samples. We used the same method as previously described to make the evolutionary classifications (25). If the difference between the CNAP of the posttreatment and the CNAP of the pretreatment is smaller than 50%, the amplification is defined as “Shared”. If the CNAP of the post-treatment is larger than the CNAP by more than 50% and the lower pretreatment CNAP is not at “FocalAMP” level, the evolutionary classification is “Acquired”. If CNAP of the posttreatment is smaller by at least 50%, comparing with the pretreatment sample and the lower posttreatment CNAP is not at “FocalAMP” level, the evolutionary classification is “Loss”. Otherwise, the evolutionary classification of amplifications is defined as “Indeterminate”.

RNA-Seq characterization of genomically perturbed cells under various drug conditions

We performed RNA sequencing (RNA-Seq) on T47D cells perturbed to overexpress FGFR pathway activation including FGFR1, FGFR2 (WT, K660N, M538I, and N550K), and FGF3, as well as GFP and parental as a control, as described above. Cells were plated in 96-well plates, and then treated with DMSO, fulvestrant (100 nmol/L), palbociclib (1 μmol/L), FIIN-3 (100 nmol/L), and trametinib (500 nmol/L) as single agent and in combinations for 24 hours. FGFR1/2 cell lines were treated with or without FGF2 (10 ng/mL) in various conditions. RNA was extracted from cells and sequencing libraries prepared as described in Supplementary Methods. For each specific construct and treatment combination, we performed at least 6 replicates, for a total of 672 RNA-Seq profiles.

Generation of plasmids and engineered cells

T47D or MCF7 cells were infected with lentivirus to derive stable cell lines overexpressing wild type (WT) or mutant ORFs. All WT ORFs were obtained from the Broad Institute. Mutant ORFs (FGFR2 M538I, N550K and K660N) were made using QuickChange II Site-Directed Mutagenesis Kit (Agilent Technologies #200523). Most stable cells lines express ORFs in pLX317 vector and were selected with puromycin (Life Technologies #A1113803). Stable cell lines expressing CCND1 and PIM1 in pLX304 vector were selected with blasticidin (Life Technologies #A1113903).

Kill curves and CellTiter-Glo viability assay

Cells were plated in 96-well tissue culture ViewPlate (PerkinElmer # 6005181) on day 1 and treated with drug on day 2. Media with or without drugs was refreshed on day 5. On day 8, cells were equilibrated to room temperature, media was removed, and cells were lysed in a mixture of 50 μL media and 50 μL CellTiter-Glo 2.0 reagent (Promega # G9243) per well. Plates were then incubated on an orbital shaker for 2 minutes. Following another 10 minutes of incubation at room temperature to stabilize signal, luminescence was recorded to measure cell viability on Infinite M200 Pro microplate reader (Tecan).

Western blotting

Western blotting was performed as described in Supplementary Methods.

Data availability

Tumor and germline WES data generated and analyzed for this study have been deposited in the access-controlled public repository dbGAP with accession code phs001285 (https://www.ncbi.nlm.nih.gov/gap). RNA-Seq data are available through GEO under accession GSE153509. Additional data generated in this study including tumor exome analysis and RNA-Seq data are available within the paper and in the supplementary information files.

Statistical analysis

Statistical analyses related to drug response curve were performed with two-tailed Student t test in GraphPad Prism. Fisher exact test was used to calculate OR and q-value for volcano plots in the RNA-Seq analysis. Cohen D test with Hedges correction and Welch t test were used to estimate the effect size and significance for signature strength of gene sets.

A genome-scale gain-of-function screen for resistance to selective estrogen receptor degraders

To identify the spectrum of genes whose overexpression confers resistance to SERDs in vitro, we expressed a pooled lentiviral library of 17,255 human ORFs, corresponding to 10,135 distinct human genes, in ER+ T47D breast cancer cells in the presence of fulvestrant or GDC-0810 (22). Genes that confer drug resistance will be enriched under drug selection for 21 days, indicated by a positive log fold change (LFC) for ORF representation before and after DMSO/drug selection.

Using a Z-score >3 as a criterion to identify resistance candidates, we identified 64 genes (93 ORFs) that conferred resistance to fulvestrant and 57 genes (83 ORFs) that conferred resistance to GDC-0810 (Fig. 1A; Supplementary Table S1). Thirty-seven genes (55 ORFs) conferred resistance to both drugs, a degree of overlap which was anticipated given the mechanistic similarities between fulvestrant and GDC-0810. The LFC and corresponding Z-score for each ORF in fulvestrant and GDC-0810 treatment arms were highly correlated, with a correlation coefficient of 0.77 (Fig. 1A).

Figure 1.

A genome-scale gain-of-function screen identified resistance genes to fulvestrant and GDC-0810. 17,255 human ORFs, corresponding to 10,135 distinct genes, were expressed in ER+ T47D breast cancer cells in the presence of fulvestrant or GDC-0810. Fulvestrant (100 nmol/L), 1 μmol/L GDC-0810, or vehicle control (DMSO) was added following infection and selection. ORF representation was assessed by sequencing after 21 days of drug exposure. Genes that confer drug resistance will be enriched under drug selection, indicated by a positive LFC for ORF representation before and after DMSO/drug selection. A, The average Z-score for LFC of each ORF was plotted for both the fulvestrant (x-axis) and GDC-0810 (y-axis) arms. The average Z-score was calculated from three replicates in each condition. The ORFs with a Z-score > 3 in both drug arms are highlighted and labeled with gene ID. The shape of each data point represents the total number of ORFs for that gene in the library. B, Heatmap of top ORF hits with a Z-score > 3 in fulvestrant or GDC-0810 arm. The Z-score in the DMSO arm is also presented. ORF hits are grouped by their molecular function according to Uniprot annotation. Information on the complete list of ORFs can be found in Supplementary Table S1. C, Individual ORFs were overexpressed in T47D cells and validated to confer resistance to fulvestrant by drug response curves. KRAS G12D ORF was used for overexpression in T47D cells while other selected ORFs are wild type. Cell viability was measured by CellTiter-Glo and all the data points were normalized to growth under DMSO condition. Results shown are ±SD and representative of three independent experiments. D, GSEA was performed for the gene list ranked by LFC in the fulvestrant arm. For genes with multiple ORFs, the ORF with highest LFC was selected. 1000 permutations were performed for the analysis. NES, normalized enrichment score. The full list of nominated pathways is shown in Supplementary Table S2.

Figure 1.

A genome-scale gain-of-function screen identified resistance genes to fulvestrant and GDC-0810. 17,255 human ORFs, corresponding to 10,135 distinct genes, were expressed in ER+ T47D breast cancer cells in the presence of fulvestrant or GDC-0810. Fulvestrant (100 nmol/L), 1 μmol/L GDC-0810, or vehicle control (DMSO) was added following infection and selection. ORF representation was assessed by sequencing after 21 days of drug exposure. Genes that confer drug resistance will be enriched under drug selection, indicated by a positive LFC for ORF representation before and after DMSO/drug selection. A, The average Z-score for LFC of each ORF was plotted for both the fulvestrant (x-axis) and GDC-0810 (y-axis) arms. The average Z-score was calculated from three replicates in each condition. The ORFs with a Z-score > 3 in both drug arms are highlighted and labeled with gene ID. The shape of each data point represents the total number of ORFs for that gene in the library. B, Heatmap of top ORF hits with a Z-score > 3 in fulvestrant or GDC-0810 arm. The Z-score in the DMSO arm is also presented. ORF hits are grouped by their molecular function according to Uniprot annotation. Information on the complete list of ORFs can be found in Supplementary Table S1. C, Individual ORFs were overexpressed in T47D cells and validated to confer resistance to fulvestrant by drug response curves. KRAS G12D ORF was used for overexpression in T47D cells while other selected ORFs are wild type. Cell viability was measured by CellTiter-Glo and all the data points were normalized to growth under DMSO condition. Results shown are ±SD and representative of three independent experiments. D, GSEA was performed for the gene list ranked by LFC in the fulvestrant arm. For genes with multiple ORFs, the ORF with highest LFC was selected. 1000 permutations were performed for the analysis. NES, normalized enrichment score. The full list of nominated pathways is shown in Supplementary Table S2.

Close modal

To confirm these results, we conducted a secondary screen using a smaller pooled library consisting of 570 ORFs in T47D and MCF7 cells to validate candidates nominated by the primary screen. Both cell lines are commonly used and well-characterized ER+ breast cancer cell and both are sensitive to SERDs, although they have different levels of ER signaling and harbor some genomic differences (https://portals.broadinstitute.org/ccle; refs. 26, 27). Top resistance genes found in the primary screen were again enriched in the secondary screen for T47D cells, including FGF genes, FOXR1, AKT genes, PIM genes and several GPCR genes (Supplementary Fig. S1A). Many top ranked resistance genes (CSF1R, FGF3, FGF6, FOXR1, and PIM2) were shared between T47D and MCF7 cells (Supplementary Fig. S1B). However, distinct resistance genes were also observed in each cell line, suggesting some resistance mechanisms may be cell context-dependent.

Functional categories of candidate resistance genes include serine/threonine kinases (PIK3CA, AKT1/2/3, PIM1/2/3), receptor tyrosine kinases (EGFR, ERBB2, PDGFRB), growth factors (FGF3/6/10/22), cell-cycle regulatory proteins (CCND1, CCND2, CCND3, CDK6), and G-protein–coupled receptors (GPCR; Fig. 1B). As further validation, we overexpressed 13 ORFs belonging to these categories individually in T47D cells and they conferred resistance to fulvestrant and GDC-0810 to various degrees (Fig. 1C; Supplementary Fig. S2A and S2B).

Gene set enrichment analysis (GSEA) of the candidate resistance genes demonstrated enrichment in 4 functional pathways: FGFR signaling, ERBB signaling, insulin receptor signaling, and the MAPK pathway (Fig. 1D; Supplementary Fig. S3; Supplementary Table S2). Consistent with this, we and others recently demonstrated that ERBB2-activating mutations (11) and alterations in MAPK pathway genes can cause endocrine resistance in patients with ER+ MBC (13, 14, 25). We sought to further examine the role of FGFR and FGF genes in resistance to SERDs in MBC.

Identification of acquired FGFR and FGF alterations in metastatic biopsies from patients with resistant ER+ MBC

To examine the potential role of FGFR and FGF alterations in the development of endocrine resistance clinically, we analyzed WES data from paired pretreatment and posttreatment metastatic tumor biopsies or cell free DNA from 60 patients with ER+ MBC who had received at least one endocrine therapy (tamoxifen, AI, SERDs) for more than 120 days between the two biopsies (28).

Amongst the 60 posttreatment samples, we found FGFR1 amplifications in 15% (9/60), FGFR2 amplifications in 5% (3/60), FGFR2-activating mutations in 3.3% (2/60), and FGF3 amplifications in 28.3% (17/60), for a total of 40% (24/60) of the cohort with at least one alteration in one of these three genes (Fig. 2A). Overall, the prevalence of FGFR1, FGFR2, and FGF3 alterations in the resistant metastatic setting seen here is increased compared to what was observed in previously published cohorts of primary ER+ breast cancer, such as The Cancer Genome Atlas (TCGA; ref. 29), and comparable to other datasets of metastatic ER+ breast cancer (Supplementary Table S3). The incidence of FGFR2 alterations (6.7%), in particular, is markedly increased compared with primary treatment-naive breast cancer, in which the incidence is less than 2% in TCGA (Supplementary Table S3).

Figure 2.

Identification of acquired FGFR and FGF alterations in metastatic biopsies from patients with resistant ER+ MBC. A, Evolutionary status of ESR1, FGFR1, FGFR2, ERBB2, and FGF3 alterations is presented by comparing the pretreatment and posttreatment mutational status for each patient (red = acquired, blue = lost, black = shared, gray = indeterminate). These 24 pairs of samples included 23 tumor biopsies and one cell-free DNA sample at the pretreatment timepoint, and 22 tumor biopsies and two cell-free DNA samples at the posttreatment timepoint. The evolutionary inference of copy number changes was based on measuring differences in copy number amplitudes between pretreatment and posttreatment samples, while accounting for differences in cancer cell fraction (“purity”) in the sample and correcting for differences in ploidy. The resultant purity-corrected values provide an estimate of “copy number above ploidy” (CNAP; see Materials and Methods). The evolutionary inference and clonal dynamics of mutations was based on changes in the estimated fraction of tumor cells harboring each genomic alteration (CCF) as previously shown for acquired HER2 mutations (11). Activating SNVs are denoted with a purple asterisk. Clinical and pathology tracks depict the site of biopsy for both matched samples, and the duration between the pretreatment and posttreatment biopsies. The lines of endocrine therapies received between the early and late sample for a duration of at least 120 days are depicted with three tracks including SERD (fulvestrant), SERM (tamoxifen), and any AIs. B, Copy number alterations for FGFR1, FGFR2, and FGF3 in pre- and posttreatment tumor samples are shown with CNAP depicted to illustrate the magnitude of the acquired amplification in each case. To better measure segment-specific copy number, we subtracted the genome ploidy for each sample to compute CNAP. The purity and ploidy for tumor samples are shown in Supplementary Table S4. C, Clonal evolution analysis showing the overall clonal structure and acquisition for FGFR2 mutations observed in two patients- FGFR2 M538I (chr10:123258070C>T, GRCh37, also denoted as M537I, depending on the isoform) FGFR2 N550K (chr10:123258034A>T, GRCh37, also denoted as N549K, depending on the isoform). In the pretreatment biopsies, FGFR2 M538I (ID 0300348) and FGFR2 N550K (ID 0300350) were with CCF of 2% (single read) and 0% (unobserved), respectively, while being observed as clonal mutations in the posttreatment sample with a CCF of 1. The phylogenetic relationships among clones are reconstructed for each patient starting from the normal cell (white circle) connected to the ancestral cancer cells (gray trunk). The phylogenetic divergence to the pretreatment clones (and subclones) is depicted with blue edges, and phylogenetic divergence to the metastatic clones (and subclones) is in red. Selected mutations in cancer genes are marked on the corresponding branches of the cancer phylogeny.

Figure 2.

Identification of acquired FGFR and FGF alterations in metastatic biopsies from patients with resistant ER+ MBC. A, Evolutionary status of ESR1, FGFR1, FGFR2, ERBB2, and FGF3 alterations is presented by comparing the pretreatment and posttreatment mutational status for each patient (red = acquired, blue = lost, black = shared, gray = indeterminate). These 24 pairs of samples included 23 tumor biopsies and one cell-free DNA sample at the pretreatment timepoint, and 22 tumor biopsies and two cell-free DNA samples at the posttreatment timepoint. The evolutionary inference of copy number changes was based on measuring differences in copy number amplitudes between pretreatment and posttreatment samples, while accounting for differences in cancer cell fraction (“purity”) in the sample and correcting for differences in ploidy. The resultant purity-corrected values provide an estimate of “copy number above ploidy” (CNAP; see Materials and Methods). The evolutionary inference and clonal dynamics of mutations was based on changes in the estimated fraction of tumor cells harboring each genomic alteration (CCF) as previously shown for acquired HER2 mutations (11). Activating SNVs are denoted with a purple asterisk. Clinical and pathology tracks depict the site of biopsy for both matched samples, and the duration between the pretreatment and posttreatment biopsies. The lines of endocrine therapies received between the early and late sample for a duration of at least 120 days are depicted with three tracks including SERD (fulvestrant), SERM (tamoxifen), and any AIs. B, Copy number alterations for FGFR1, FGFR2, and FGF3 in pre- and posttreatment tumor samples are shown with CNAP depicted to illustrate the magnitude of the acquired amplification in each case. To better measure segment-specific copy number, we subtracted the genome ploidy for each sample to compute CNAP. The purity and ploidy for tumor samples are shown in Supplementary Table S4. C, Clonal evolution analysis showing the overall clonal structure and acquisition for FGFR2 mutations observed in two patients- FGFR2 M538I (chr10:123258070C>T, GRCh37, also denoted as M537I, depending on the isoform) FGFR2 N550K (chr10:123258034A>T, GRCh37, also denoted as N549K, depending on the isoform). In the pretreatment biopsies, FGFR2 M538I (ID 0300348) and FGFR2 N550K (ID 0300350) were with CCF of 2% (single read) and 0% (unobserved), respectively, while being observed as clonal mutations in the posttreatment sample with a CCF of 1. The phylogenetic relationships among clones are reconstructed for each patient starting from the normal cell (white circle) connected to the ancestral cancer cells (gray trunk). The phylogenetic divergence to the pretreatment clones (and subclones) is depicted with blue edges, and phylogenetic divergence to the metastatic clones (and subclones) is in red. Selected mutations in cancer genes are marked on the corresponding branches of the cancer phylogeny.

Close modal

To determine whether this enrichment of FGFR/FGF alterations in the metastatic setting was due to acquisition/selection under the selective pressure of endocrine therapy, we performed an evolutionary analysis to evaluate clonal structure and dynamics, including changes in mutations and copy number for the 24 patients harboring FGFR/FGF alterations. For this analysis, we define “acquired” alterations as alterations with higher representation in the posttreatment sample as compared with the pretreatment sample (see Materials and Methods). Although we use the term “acquired”, we recognize that when the mutation is not detected in the pretreatment sample, we cannot distinguish between preexisting alteration that was selected for and clonally enriched versus de novo alterations that developed during the treatment.

In 12 of the 24 patients with FGFR or FGF alterations (50%), the alterations were acquired in the posttreatment sample as compared with the pretreatment sample (Fig. 2A, marked in red). Five of nine FGFR1 amplifications were acquired (55.6%), while all four FGFR2 alterations were acquired (100%), including one patient (Pt 0300350) with acquisition of both an FGFR2 mutation and amplification. FGF3 amplifications were acquired in 4 of 17 tumors (23.5%), including one case in which an FGFR1 amplification was coacquired. The concurrent acquisition may suggest that the evolutionary selection of both the ligand and receptor provided additional fitness in this tumor. Among the other 12 patients, the alterations in 8 patients were shared in both pretreatment and posttreatment samples (Fig. 2A, marked in black), and evolutionary status of alterations in the remaining 4 patients was inconclusive (Fig. 2A, marked in gray). The increase in copy number (corrected for tumor purity and ploidy, Supplementary Table S4) from pretreatment to posttreatment for FGFR1, FGFR2, and the FGF3 amplicon in all 12 patients is depicted in Fig. 2B.

Two of the acquired alterations found in these 12 patients were single-nucleotide variants (SNV) in the FGFR2 gene: M538I and N550K. N550K is the most common FGFR2 mutation in breast cancer while M538I was previously identified in lung cancer but has not yet been characterized in breast cancer (30). Figure 2C illustrates the change in the estimated fraction of tumor cells harboring each genomic alteration (CCF) from the pretreatment biopsy to the resistant biopsy. In both patients, the FGFR2 mutations were either not detected in the primary tumor (N550K in Pt 0300350) or detected in a small fraction (CCF of 2%) of the pretreatment tumor (M538I in Pt 0300348). In both patients, the activating FGFR2 mutations in the posttreatment biopsies were clonally acquired (CCF of 100%; Fig. 2B).

Notably, the acquired alterations in FGFR1, FGFR2, and FGF3 were largely mutually exclusive with acquired ESR1 mutations and HER2 mutations (Fig. 2A). ESR1 mutations are the most common mechanism described for acquired endocrine resistance (31). Although the overall rate of acquired ESR1 mutation in this cohort is 22% (13/60), among the 12 cases of acquired FGFR and FGF alterations, only one patient also has an acquired ESR1 mutation (Fig. 2A). Similarly, only one of these 12 patients had acquired a HER2 mutation ERBB2 I628M (which was an alteration of unknown significance), suggesting that these are also mutually exclusive mechanisms of resistance.

Although we highlight FGF3 as the key gene in the amplicon (given the results of the gain-of-function screen), FGF3 resides in genomic proximity to FGF4, FGF19, and CCND1 and these four genes are often coamplified. Here, in 3 of the 4 cases with acquired FGF3 amplification, FGF3 copies were gained without coacquisition of CCND1 amplification, suggesting that this acquisition can occur as an independent genomic event (Supplementary Fig. S4). Similarly, 2 of the 4 cases with acquired FGF3 did not have coacquisition of FGF19 amplification. In all 4 of the cases with FGF3 amplification, FGF4 was also co-amplified. The other concurrent genetic alterations for the 12 patients with acquired FGFR/FGF alterations are shown in Supplementary Fig. S5 and Supplementary Tables S5 and S6.

Figure 3 depicts clinical vignettes for 6 of the patients with acquired FGFR1, FGFR2, and/or FGF3 alterations in their posttreatment biopsies. All patients were treated with ER-directed therapy before acquiring FGF or FGFR alterations, including tamoxifen (3 patients), AIs (6 patients), and fulvestrant (3 patients). Vignettes for the other 6 patients with acquired FGFR1, FGFR2, and/or FGF3 alterations in their posttreatment biopsies are shown in Supplementary Fig. S6A. Detailed clinicopathologic features and therapy details for all 12 patients are found in Supplementary Table S7.

Figure 3.

Clinical vignettes of patients who acquired FGFR/FGF alterations following endocrine therapy. The clinical vignettes for selected patients with acquired alterations in FGFR1, FGFR2, and/or FGF3 illustrate detailed information on age and stage of disease at diagnosis, therapies patients received, duration of response to each therapy, and time of biopsies collected during the clinical course. For each biopsy, available information on biopsy type, tissue site, receptor status, and selected genomic alterations detected by whole exome sequencing is shown. In each case, the asterisk indicates the time that metastatic disease was diagnosed. The complete clinicopathologic information for each patient is provided in Supplementary Table S7. IDC, invasive ductal carcinoma; IDLC, invasive ductal-lobular carcinoma; yo: years old; Bx: biopsy; PR: progesterone receptor; wt: wild type.

Figure 3.

Clinical vignettes of patients who acquired FGFR/FGF alterations following endocrine therapy. The clinical vignettes for selected patients with acquired alterations in FGFR1, FGFR2, and/or FGF3 illustrate detailed information on age and stage of disease at diagnosis, therapies patients received, duration of response to each therapy, and time of biopsies collected during the clinical course. For each biopsy, available information on biopsy type, tissue site, receptor status, and selected genomic alterations detected by whole exome sequencing is shown. In each case, the asterisk indicates the time that metastatic disease was diagnosed. The complete clinicopathologic information for each patient is provided in Supplementary Table S7. IDC, invasive ductal carcinoma; IDLC, invasive ductal-lobular carcinoma; yo: years old; Bx: biopsy; PR: progesterone receptor; wt: wild type.

Close modal

In addition to these 12 patients in our cohort, we identified several additional patients with acquired activating mutations in FGFR1(N546K) and FGFR2 (N550K, K660N) following the development of resistance to endocrine therapy (Supplementary Fig. S6B; Supplementary Table S8).

In summary, we observed acquired alterations in FGFR1, FGFR2, or FGF3 in 20% (12/60) of patients with endocrine resistant ER+ MBC, comparable with the known frequency of acquired mutations in ESR1, highlighting the important role of the FGFR pathway in acquired resistance to ER-directed therapies.

Active FGFR signaling leads to resistance to SERDs through activation of the MAP kinase pathway

To further investigate how FGFR/FGF genes may confer resistance to ER-directed therapy, we treated T47D cells with FGF3, FGF6, FGF10, or FGF22 ligand. Each of these ligands resulted in resistance to fulvestrant (Fig. 4A). This effect was reversed by PD173074, a pan-FGFR inhibitor (Fig. 4A). The addition of these FGF ligands enhanced phosphorylation of ERK and AKT, which was reversed by PD173074 (Supplementary Fig. S7A). FGF3, FGF6, FGF10, and FGF22 also reduced fulvestrant sensitivity in MCF7 cells (Supplementary Fig. S7B and S7C).

Figure 4.

Active FGFR signaling leads to resistance to SERDs and CDK4/6 inhibitors through activation of MAPK pathway. A, FGF ligands lead to resistance to fulvestrant, which was blocked by FGFR inhibitor PD173074. Recombinant FGF ligands were added into media every three days at the concentration of 100 ng/mL with or without 1 μmol/L PD173014. T47D cells were treated with heparin (1 μg/mL) that facilitates the binding between FGF ligand and receptor, and sensitivity to 100 nmol/L fulvestrant over 6 days was normalized to DMSO control. *** P < 0.001. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments. B, FGFR1 or FGFR2 overexpression leads to resistance to fulvestrant, which was blocked by PD173074. GFP, FGFR1, or FGFR2 was overexpressed in T47D cells to establish stable T47D_GFP, T47D_FGFR1, and T47D_FGFR2 cells. The fulvestrant sensitivity of various cell lines were determined in the presence or absence of 10 ng/mL FGF2 and 1 μmol/L PD173074 over 6 days of drug treatment. Results shown are ±SD and representative of three independent experiments. C, FGFR1 and FGFR2 induced phosphorylation of ERK and AKT in the presence of FGF2, which was blocked by PD173074. Results shown are representative of two independent experiments. Cells were treated with indicated conditions for one hour before protein harvest. FGF2 (10 ng/mL) and 1 μmol/L PD173074 were used. D, Trametinib abrogated the resistance to fulvestrant (top) or the combination of fulvestrant and palbociclib (bottom) conferred by FGFR1 or FGFR2. Cells were treated with different conditions as indicated: 10 ng/mL FGF2; 100 nmol/L fulvestrant (Fulv); 1 μmol/L palbociclib (Palbo); 500 nmol/L trametinib. CellTiter-Glo assay was performed to measure cell viability after 6 days for all dose response curves, data are ±SD. Results shown are representative of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001, n.s. not significant. Student t test was performed for pairwise comparisons. E, Trametinib blocked ERK phosphorylation and reduced CCND1 and p-Rb levels. Cells were treated as indicated daily for two days before protein harvest and western blot: 10 ng/mL FGF2; 100 nmol/L fulvestrant; 1 μmol/L palbociclib; 500 nmol/L trametinib. Results shown are representative of two independent experiments.

Figure 4.

Active FGFR signaling leads to resistance to SERDs and CDK4/6 inhibitors through activation of MAPK pathway. A, FGF ligands lead to resistance to fulvestrant, which was blocked by FGFR inhibitor PD173074. Recombinant FGF ligands were added into media every three days at the concentration of 100 ng/mL with or without 1 μmol/L PD173014. T47D cells were treated with heparin (1 μg/mL) that facilitates the binding between FGF ligand and receptor, and sensitivity to 100 nmol/L fulvestrant over 6 days was normalized to DMSO control. *** P < 0.001. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments. B, FGFR1 or FGFR2 overexpression leads to resistance to fulvestrant, which was blocked by PD173074. GFP, FGFR1, or FGFR2 was overexpressed in T47D cells to establish stable T47D_GFP, T47D_FGFR1, and T47D_FGFR2 cells. The fulvestrant sensitivity of various cell lines were determined in the presence or absence of 10 ng/mL FGF2 and 1 μmol/L PD173074 over 6 days of drug treatment. Results shown are ±SD and representative of three independent experiments. C, FGFR1 and FGFR2 induced phosphorylation of ERK and AKT in the presence of FGF2, which was blocked by PD173074. Results shown are representative of two independent experiments. Cells were treated with indicated conditions for one hour before protein harvest. FGF2 (10 ng/mL) and 1 μmol/L PD173074 were used. D, Trametinib abrogated the resistance to fulvestrant (top) or the combination of fulvestrant and palbociclib (bottom) conferred by FGFR1 or FGFR2. Cells were treated with different conditions as indicated: 10 ng/mL FGF2; 100 nmol/L fulvestrant (Fulv); 1 μmol/L palbociclib (Palbo); 500 nmol/L trametinib. CellTiter-Glo assay was performed to measure cell viability after 6 days for all dose response curves, data are ±SD. Results shown are representative of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001, n.s. not significant. Student t test was performed for pairwise comparisons. E, Trametinib blocked ERK phosphorylation and reduced CCND1 and p-Rb levels. Cells were treated as indicated daily for two days before protein harvest and western blot: 10 ng/mL FGF2; 100 nmol/L fulvestrant; 1 μmol/L palbociclib; 500 nmol/L trametinib. Results shown are representative of two independent experiments.

Close modal

We next overexpressed FGFR1, FGFR2, or GFP in T47D cells through lentiviral transduction and examined the impact on susceptibility to SERDs. Overexpression of FGFR1 or FGFR2 alone did not affect sensitivity to fulvestrant. However, with the addition of FGF2 ligand, both FGFR1 and FGFR2 rendered cells highly resistant to fulvestrant (Fig. 4B). A similar resistance phenotype was also observed when T47D cells expressing FGFR1 or FGFR2 were treated with GDC-0810 or tamoxifen (Supplementary Fig. S8A–S8C). In comparison, FGF2 ligand alone reduced sensitivity to SERDs in control cells expressing GFP to a much lesser extent than in the FGFR1 or FGFR2-expressing cells, suggesting the potent resistance phenotype requires both FGF ligand and receptor. This requirement for the presence of both FGF ligand and receptor for maximal resistance phenotype may also explain why only FGFs but not FGFR1 or FGFR2 scored in the resistance screen (Fig. 1A and B). The resistance phenotype resulting from FGFR1 and FGFR2 overexpression was completely reversed by the addition of PD173074 (Fig. 4B). Similar results were obtained in MCF7 cells (Supplementary Fig. S8E and S8F).

FGFR1 and FGFR2 overexpression (in the presence of FGF2 ligand) induced more potent phosphorylation of AKT and ERK than the GFP control, which was reversed by PD173074 (Fig. 4C; Supplementary Fig. S8D and S8G). These results are consistent with previous findings that FGFR1 activation led to MAPK activation and fulvestrant resistance (32). Collectively, these findings suggest that FGFR1 and FGFR2 cause SERD resistance through the activation of MAPK and/or PI3K/AKT pathways.

We examined the sensitivity of cells overexpressing FGFR1 or FGFR2 to several inhibitors of downstream effectors: the MEK inhibitor trametinib, the AKT inhibitor AZD5363, and the mTOR inhibitor everolimus. FGFR1 or FGFR2 overexpression in the presence of FGF2 led to hypersensitivity to trametinib (Supplementary Fig. S9A), but reduced sensitivity to AKT and mTOR inhibitors (Supplementary Fig. S9A).

We attempted to reverse FGFR-induced resistance to fulvestrant by inhibiting the MAPK pathway. Treatment of FGFR1-overexpressing cells with trametinib partially resensitized cells to fulvestrant, and treatment of FGFR2 overexpressing cells with trametinib fully resensitized the cells to fulvestrant (Fig. 4D). In contrast, trametinib did not reverse an increase in viability induced by FGF2 in the GFP control cells. This is likely because FGF2 alone may not sufficiently increase MAPK signaling to induce a dependency, which is consistent with much lower sensitivity to trametinib in GFP cells as compared to FGFR1 and FGFR2 cells in the presence of FGF2 (Supplementary Fig. S9A and S9B). Colony formation assays produced similar results (Supplementary Fig. S9C). Treatment with the mTOR inhibitor everolimus also partially reversed resistance conferred by FGFR1 or FGFR2 overexpression (Supplementary Fig. S9D). Together, these results suggest that the MAPK pathway is the primary downstream effector of FGFR activation resulting in endocrine resistance.

FGFR activation confers cross-resistance to CDK4/6 inhibitors

Because the combination of endocrine therapy and CDK4/6 inhibitors is now a standard-of-care treatment for patients with ER+ MBC, we also examined the effect of FGFR signaling on sensitivity to the combination of fulvestrant and the CDK4/6 inhibitor palbociclib. In T47D cells, FGFR1 and FGFR2 overexpression in the presence of FGF2 also conferred resistance to combination treatment of fulvestrant and palbociclib. Resistance to fulvestrant and palbociclib was abrogated by PD173074 and also partially reversed by trametinib (Fig. 4D; Supplementary Fig. S9E), further providing the support for the role of MAPK pathway activation in FGFR-mediated drug resistance. The reversal of resistance by trametinib was accompanied by reduced ERK phosphorylation (Fig. 4E). Similar results were achieved in MCF7 cells, although everolimus was more effective than trametinib in reversing the resistance phenotype by FGFR1 or FGFR2 overexpression in this cell line (Supplementary Fig. S10). In the presence of fulvestrant and palbociclib, FGFR1 or FGFR2 overexpression was accompanied by increased p-Rb and CCND1 levels, both of which were partially reversed by trametinib (Fig. 4E). This is consistent with prior results suggesting that CCND1 was involved in FGF2-mediated drug resistance (33).

Clinical evidence also supports the finding that FGFR alterations can cause resistance to CDK4/6 inhibitors. Following the acquisition of FGFR2 N550K (along with FGFR2 amplification), Pt 0300350 did not respond to the combination of letrozole and palbociclib (Fig. 3), suggesting that FGFR2 alterations may lead to intrinsic resistance to the combination of endocrine therapy and CDK4/6 inhibitors. Another patient with an FGFR2 N550K mutation (FM Patient 2) also did not respond to the combination of fulvestrant and palbociclib (Supplementary Fig. S6B). Collectively, this suggests targeting the FGFR pathway may also be a viable strategy to overcome FGFR/FGF–mediated resistance to SERDs and CDK4/6 inhibitors.

FGFR2 mutations found in patients are activating with differential sensitivity to FGFR inhibitors

We identified 3 acquired mutations in the kinase domain of FGFR2 in patients who developed resistance to endocrine therapy. Two of these, FGFR2 N550K and K660N, are known activating FGFR2 mutations that have been previously identified in breast cancer (30, 34). The third mutation, FGFR2 M538I, has not been previously reported in breast cancer, but was identified through in vitro screening as a reversible pan FGFR inhibitor resistance mutation and confirmed to increase kinase activity in vitro (Fig. 5A; ref. 35).

Figure 5.

FGFR2 M538I, N5550K, and K660N were activating mutations and can be targeted by irreversible kinase inhibitors FIIN-2 and FIIN-3. A, Crystal structure of activated FGFR2 protein with mutations shown. FGFR2 is in complex with ATP analogue (in yellow) and substrate peptide (PDB ID: 2PVF). FGFR2 N550K is part of the molecular brake at the kinase hinge region, which allows the receptor to adopt an active conformation more easily (35). FGFR2 K660N is located in a conserved region in the tyrosine kinase domain and has been confirmed to increase kinase activity (34, 35). B, Stable cell lines overexpressing FGFR2 wild type (WT), M538I, N550K, and K660N were treated with 10 ng/mL FGF2 and/or 1 μmol/L PD173014 for one hour before protein harvest. Results shown are representative of two independent experiments. C, Stable cell lines constitutively overexpressing GFP or FGFR2 constructs (as described previously) were examined for sensitivity to fulvestrant or combination of fulvestrant and palbociclib with or without the treatment of FGF2 and/or PD173074. *, P < 0. 05; **, P < 0.01; ***, P < 0.001, calculated as compared with GFP group in all conditions. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments. D, T47D cells overexpressing FGFR2 N550K cells were treated as indicated for 3 days and retreated for 3 hours before protein harvest and Western blot analysis. Results shown are representative of two independent experiments. E, All stable cells lines expressing GFP or FGFR2 constructs were treated with fulvestrant under the following conditions: control, 10 ng/mL FGF2, 10 ng/mL FGF2 with 1 μmol/L PD173074, 10 ng/mL FGF2 with 1 μmol/L AZD4547, 10 ng/mL FGF2 with 1 μmol/L FIIN-2, or 10 ng/mL FGF2 with 100 nmol/L FIIN-3. Drug response curves were determined by CellTiter-Glo. Results shown are ±SD and representative of three independent experiments.

Figure 5.

FGFR2 M538I, N5550K, and K660N were activating mutations and can be targeted by irreversible kinase inhibitors FIIN-2 and FIIN-3. A, Crystal structure of activated FGFR2 protein with mutations shown. FGFR2 is in complex with ATP analogue (in yellow) and substrate peptide (PDB ID: 2PVF). FGFR2 N550K is part of the molecular brake at the kinase hinge region, which allows the receptor to adopt an active conformation more easily (35). FGFR2 K660N is located in a conserved region in the tyrosine kinase domain and has been confirmed to increase kinase activity (34, 35). B, Stable cell lines overexpressing FGFR2 wild type (WT), M538I, N550K, and K660N were treated with 10 ng/mL FGF2 and/or 1 μmol/L PD173014 for one hour before protein harvest. Results shown are representative of two independent experiments. C, Stable cell lines constitutively overexpressing GFP or FGFR2 constructs (as described previously) were examined for sensitivity to fulvestrant or combination of fulvestrant and palbociclib with or without the treatment of FGF2 and/or PD173074. *, P < 0. 05; **, P < 0.01; ***, P < 0.001, calculated as compared with GFP group in all conditions. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments. D, T47D cells overexpressing FGFR2 N550K cells were treated as indicated for 3 days and retreated for 3 hours before protein harvest and Western blot analysis. Results shown are representative of two independent experiments. E, All stable cells lines expressing GFP or FGFR2 constructs were treated with fulvestrant under the following conditions: control, 10 ng/mL FGF2, 10 ng/mL FGF2 with 1 μmol/L PD173074, 10 ng/mL FGF2 with 1 μmol/L AZD4547, 10 ng/mL FGF2 with 1 μmol/L FIIN-2, or 10 ng/mL FGF2 with 100 nmol/L FIIN-3. Drug response curves were determined by CellTiter-Glo. Results shown are ±SD and representative of three independent experiments.

Close modal

We expressed all three FGFR2 kinase domain mutants in T47D cells through lentiviral transduction, as well as wildtype (WT) FGFR2 and GFP as negative controls. All three mutants elicited higher kinase activity than WT FGFR2 constitutively, demonstrated by levels of p-FRS2 (a direct substrate for FGFR2), p-ERK, and p-AKT (Fig. 5B). The addition of FGF2 ligand further enhanced downstream signaling for all FGFR2 mutants, and the enhanced signaling was blocked by PD173074 for FGFR2 M538I and K660N, but not for N550K (Fig. 5B). FGFR2 mutants were also expressed under a tetracycline responsive promoter in T47D cells grown in low doses of doxycycline to determine the functionality at lower expression levels. All three FGFR2 mutations acquired in patients with breast cancer are functionally active; FGFR2 N550K is constitutively active while FGFR2 M538I and K660N may be more ligand-dependent at low levels of expression (Supplementary Fig. S11A).

All 3 FGFR2 mutants led to modest resistance to fulvestrant or the combination of fulvestrant and palbociclib (Fig. 5C), which was enhanced in the presence of FGF2 ligand. PD173074 reversed the resistance phenotype for cells overexpressing FGFR2 M538I and FGFR2 K660N as well as WT FGFR2 to fulvestrant, but not cells overexpressing FGFR2 N550K (Fig. 5C). Similar results were obtained in MCF7 cells (Supplementary Fig. S12).

Activating FGFR2 mutations can be targeted with irreversible FGFR inhibitors

Because of the differential responses of these FGFR2 mutants to PD173074, we tested the ability of additional FGFR inhibitors to resensitize cells expressing these mutants to fulvestrant. FIIN-2 and FIIN-3 are two irreversible covalent pan-FGFR inhibitors that target a cysteine conserved in FGFR1-4 and have exquisite selectivity for some FGFR2 mutations including M538I and K659N (36). In addition, AZD4547 is a selective FGFR inhibitor that was previously shown to inhibit FGFR2 N550K (37).

Both FIIN-2 and FIIN-3 were more effective in inhibiting the downstream signaling (p-FRS2, p-ERK, and p-AKT) induced by FGFR2 N550K as compared with PD173074 and AZD4547 (Fig. 5D), with FIIN-3 being more potent than FIIN-2 (Supplementary Fig. S13). T47D cells stably overexpressing FGFR2 mutant were exquisitely sensitive to FIIN-2 and FIIN-3 as compared with cells expressing GFP or FGFR2 WT (Supplementary Fig. S11B). Although resistance to WT FGFR1/2 and FGFR2 M538I and FGFR2 K660N can be reversed by multiple FGFR inhibitors, for some mutants like FGFR2 N550K, only the irreversible pan-FGFR inhibitors FIIN-2 and FIIN-3 successfully resensitized cells to fulvestrant (Fig. 5E), highlighting the fact that specific resistance mutations might require different strategies to overcome or preempt endocrine resistance.

Transcriptional changes induced by FGFR/FGF include ER reprograming and MAPK activation

To examine transcriptional changes associated with FGFR pathway activation, we performed RNA-Seq on T47D cells overexpressing FGFR1, FGFR2 (wild type), FGFR2-activating mutants (M538I, N550K, K660N), or FGF3, as well as on GFP and parental lines as controls (Supplementary Table S9). We combined these profiles with transcriptional profiles we previously generated (11) from of T47D cells overexpressing wild-type and kinase-dead HER2, HER2-activating mutants (S653C, L755S, V777L, and L869R), and ESR1 Y537S, for a total of 15 genetic perturbations examined.

Linear discriminant analysis of these profiles indicated that samples from cells overexpressing FGFR1, wild-type FGFR2, FGFR2-activating mutants, FGF3, wild-type HER2, or HER2-activating mutants are separated from controls (parental cells, GFP expressing, or kinase-dead HER2 D845A) along the first LD component (LD1), as well as from a separate group of cells overexpressing mutant ESR1 along the second LD component (LD2), indicating a common receptor tyrosine kinase (RTK)/growth factor–driven cell state that is distinct from the mutant ER state (Fig. 6A). Along LD1, the cells overexpressing FGF3 have distinct scores from those overexpressing FGFR (P = 2.12 × 10−10, Welch t test), suggesting that in our model system, FGF ligand overexpression is different from FGFR overexpression (Fig. 6A). The overall signature strength of RTKs and growth factor signaling genes (defined in Supplementary Methods and shown in Supplementary Table S10) were highly correlated with scores on LD1 (Spearman ρ = 0.926), suggesting that LD1 broadly represents RTK and growth factor pathway activation.

Figure 6.

Transcriptional cell-state analysis of FGF/R activating perturbations reveals MAPK activation and ER-reprograming, and FGFR-induced resistance phenotype was reversed by MEK and SHP2 inhibition. A, Linear discriminant analysis (LDA) projection of FGFR/FGF–activated cells and controls. Two-dimensional visualization of the transcriptional footprints driven by FGFR1, FGFR2, FGFR2 activating mutants, FGF3, and GFP (all shown as circles), as well as previously published (11) transcriptional footprints of HER2-activating mutants, wild-type HER2, kinase-dead HER2 D845A, and ESR1 Y537S (all shown as triangles), all treated with DMSO. “Parental” refers to cells without ORF overexpression. B, Volcano plot representation of differentially expressed genes (DEG) comparing the transcriptional footprints from cells expressing FGFR1, FGFR2, FGFR2 activating mutants, and FGF3, collectively versus controls, all under DMSO condition. The FGF/R-ACT transcriptional state was derived from significant DEGs. C, Selective volcano plots and GSEA plots indicate several pathways, which are activated in the FGF/R-ACT state, including RTK/growth factor receptors signaling genes (624 genes), RAS/MAPK genes (701 genes), ER signaling driven by growth factor genes (95 genes), and MTOR pathway genes (953 genes). See Supplementary Table S11 for gene sets, Supplementary Table S12 for a comprehensive list of gene sets and pathway associations. NES, normalized enrichment score; n.s., not significant. D, RAS/MAPK signature was compared across various drug conditions for resistant FGFR/FGF cell lines. The signature strength for each sample is depicted in the y-axis with a Z-score scaled across all 647 samples. Each box plot represents the distribution of the signature strength among replicates of each experimental condition as indicated. Drug conditions include: “DMSO” (no drug), “Fulv” (fulvestrant), “Palbo” (palbociclib), “Fulv+Palbo” (fulvestrant and palbociclib), “FIIN-3”, “Fulv+FIIN-3” (fulvestrant and FIIN-3), “Palbo+FIIN-3” (palbociclib and FIIN-3), “Fulv+Palbo+FIIN-3” (fulvestrant, palbociclib, and FIIN-3), “Fulv+Tram” (fulvestrant and trametinib), “Fulv+Palbo+Tram” (fulvestrant, palbociclib, and trametinib). Concentration of drugs used: fulvestrant (100 nmol/L), palbociclib (1 μmol/L), FIIN-3 (100 nmol/L), trametinib (500 nmol/L). Construct perturbations include overexpression of GFP (control), FGF3, FGFR1, and FGFR2 (WT, K660N, M538I, and N550K). For cell lines overexpressing FGFR1/2, 10 ng/mL FGF2 was supplemented in the media. See Supplementary methods for details and Supplementary Table S11 for gene sets definitions. E and F, SHP099 as a single agent and in combination with trametinib rescued resistance to fulvestrant (E) and the combination of fulvestrant and palbociclib (F) conferred by FGFR1 or FGFR2 wild type and mutant constructs. Concentration of drugs used: 10 ng/mL FGF2; 100 nmol/L fulvestrant; 1 μmol/L palbociclib; 500 nmol/L trametinib; 10 μmol/L SHP099. *, P < 0.05; **, P < 0.01; ***, P < 0.001. n.s. not significant. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments.

Figure 6.

Transcriptional cell-state analysis of FGF/R activating perturbations reveals MAPK activation and ER-reprograming, and FGFR-induced resistance phenotype was reversed by MEK and SHP2 inhibition. A, Linear discriminant analysis (LDA) projection of FGFR/FGF–activated cells and controls. Two-dimensional visualization of the transcriptional footprints driven by FGFR1, FGFR2, FGFR2 activating mutants, FGF3, and GFP (all shown as circles), as well as previously published (11) transcriptional footprints of HER2-activating mutants, wild-type HER2, kinase-dead HER2 D845A, and ESR1 Y537S (all shown as triangles), all treated with DMSO. “Parental” refers to cells without ORF overexpression. B, Volcano plot representation of differentially expressed genes (DEG) comparing the transcriptional footprints from cells expressing FGFR1, FGFR2, FGFR2 activating mutants, and FGF3, collectively versus controls, all under DMSO condition. The FGF/R-ACT transcriptional state was derived from significant DEGs. C, Selective volcano plots and GSEA plots indicate several pathways, which are activated in the FGF/R-ACT state, including RTK/growth factor receptors signaling genes (624 genes), RAS/MAPK genes (701 genes), ER signaling driven by growth factor genes (95 genes), and MTOR pathway genes (953 genes). See Supplementary Table S11 for gene sets, Supplementary Table S12 for a comprehensive list of gene sets and pathway associations. NES, normalized enrichment score; n.s., not significant. D, RAS/MAPK signature was compared across various drug conditions for resistant FGFR/FGF cell lines. The signature strength for each sample is depicted in the y-axis with a Z-score scaled across all 647 samples. Each box plot represents the distribution of the signature strength among replicates of each experimental condition as indicated. Drug conditions include: “DMSO” (no drug), “Fulv” (fulvestrant), “Palbo” (palbociclib), “Fulv+Palbo” (fulvestrant and palbociclib), “FIIN-3”, “Fulv+FIIN-3” (fulvestrant and FIIN-3), “Palbo+FIIN-3” (palbociclib and FIIN-3), “Fulv+Palbo+FIIN-3” (fulvestrant, palbociclib, and FIIN-3), “Fulv+Tram” (fulvestrant and trametinib), “Fulv+Palbo+Tram” (fulvestrant, palbociclib, and trametinib). Concentration of drugs used: fulvestrant (100 nmol/L), palbociclib (1 μmol/L), FIIN-3 (100 nmol/L), trametinib (500 nmol/L). Construct perturbations include overexpression of GFP (control), FGF3, FGFR1, and FGFR2 (WT, K660N, M538I, and N550K). For cell lines overexpressing FGFR1/2, 10 ng/mL FGF2 was supplemented in the media. See Supplementary methods for details and Supplementary Table S11 for gene sets definitions. E and F, SHP099 as a single agent and in combination with trametinib rescued resistance to fulvestrant (E) and the combination of fulvestrant and palbociclib (F) conferred by FGFR1 or FGFR2 wild type and mutant constructs. Concentration of drugs used: 10 ng/mL FGF2; 100 nmol/L fulvestrant; 1 μmol/L palbociclib; 500 nmol/L trametinib; 10 μmol/L SHP099. *, P < 0.05; **, P < 0.01; ***, P < 0.001. n.s. not significant. Student t test was performed for pairwise comparisons. Results shown are ±SD and representative of three independent experiments.

Close modal

Using differential expression analysis to compare activating constructs with GFP and parental controls, we defined transcriptional signatures for all FGFR1/2 constructs (FGFR1/2), FGF3, and all HER2-activating mutants (HER2-MUT). Comparison of the top 200 highly expressed genes in each of the FGFR1/2, FGF3, and HER2-MUT signatures (Supplementary Table S11) highlights the high degree of similarity between these groups, with 32 genes common to the FGFR1/2 and FGF3 signatures (OR = 39.63 P = 4.7 × 10−37, two-sided Fisher exact test), 84 genes common to the FGFR1/2 and HER2-MUT signatures (OR = 219.4, P = 1.025 × 10−140, two-sided Fisher exact test), and 18 upregulated genes common to all 3 groups (Supplementary Fig. S14A).

We next defined an FGF/R-ACT transcriptional state based on 377 genes that are upregulated in FGF3 and FGFR1/2 cell lines compared with controls (Fig. 6B; Supplementary Table S10). GSEA (38) of the FGF/R-ACT state versus 5,150 previously characterized gene sets (Supplementary Table S12) demonstrated that the shared FGF/R-ACT state is enriched for upregulation of RTK/growth factor receptors signaling, RAS/MAPK signaling, and growth factor–induced ER target genes (Fig. 6C), similar to what we have previously shown for HER2-activating mutants (11). This is consistent with a model in which FGFR/FGF activation leads to ER reprogramming via MAPK pathway activation, potentially shifting the transcriptional spectrum from genes activated by transcription factor AF2 to those activated by AF1. Of note, the genes associated with mTOR signaling were variably upregulated and downregulated in FGF/R-ACT state (Fig. 6C), suggesting only moderate mTOR activation as compared with the robust MAPK activation signal observed. Similar transcriptional signatures were seen when the FGF/R-ACT signature was characterized under fulvestrant treatment, demonstrating that the FGF/R-ACT state is present even when ER signaling is inhibited, consistent with the phenotype of fulvestrant resistance (Supplementary Tables S11 and S12).

Next, we evaluated the effect of 10 different drug combinations on the transcriptional signatures in FGFR/FGF expressing cell lines and control cells profiled by RNA-Seq. The activated RAS/MAPK signature, ER signaling driven by growth factors signature, and FGF/R-ACT signature present in cells overexpressing FGFR/FGF constructs persisted under treatment with fulvestrant, palbociclib, or their combination (Fig. 6D; Supplementary Fig. S14B and S14C). Treatment with FIIN-3, alone or in combination with fulvestrant and/or palbociclib largely reversed the activation of all three signatures (Supplementary Table S13). Effective suppression of all three signatures was also achieved by trametinib plus fulvestrant, with or without palbociclib (Fig. 6D; Supplementary Fig. S14B and S14C; Supplementary Table S13).

Targeting MAPK pathway via SHP2 inhibition can overcome FGFR-induced endocrine resistance

Data from the gain-of-function ORF screen, the transcriptomic analysis, and our individual in vitro experiments together suggest that MAPK pathway may represents a common node for drug resistance in ER+ MBC. FGFR2 mutants, in particular, rendered cells more sensitive to trametinib than did GFP or WT FGFR2 (Supplementary Fig. S14D), further supporting the finding that FGFR signaling requires the MAPK pathway in this context. Src homology phosphotyrosyl phosphatase 2 (SHP2) is protein downstream of RTKs that is required for RAS activation. Several recent studies have demonstrated that cotargeting SHP2 prevented adaptive resistance to MEK inhibition in multiple RAS-driven cancer models (39–41). Consistent with this, cell lines expressing FGFR2 mutants were hypersensitive to the SHP2 inhibitor SHP099 (Supplementary Fig. S14E). We next examined the ability of combinations of SHP099, trametinib, fulvestrant, and/or palbociclib to overcome resistance in FGFR-expressing cell lines. Similar to trametinib, SHP099 as a single agent partially rescued resistance to fulvestrant and/or palbociclib conferred by FGFR1, and completely rescued resistance conferred by FGFR2. Cells expressing the FGFR2 constructs (wild type or activating mutations) were particularly sensitive to the treatment regimen of trametinib and SHP099 in addition to fulvestrant and palbociclib (Fig. 6E and F), in comparison with minimal inhibitory effect of MEK and SHP2 inhibition in GFP control cells. These results suggest that targeting MEK and SHP2 may serve as an even more effective strategy to overcome multiple forms of FGFR pathway mediated resistance to endocrine therapy and CDK4/6 inhibitors in MBC, as well as, potentially, other RTK-induced mechanisms of resistance.

Taken together, our results from genome-scale gain-of-function screen and genomic profiling of patient samples suggest that activating FGFR pathway alterations are a distinct mechanism of acquired resistance to multiple forms of ER-directed therapy in MBC that can be overcome by FGFR and/or MAPK pathway inhibitors.

Our findings are consistent with two recently published studies. Formisano and colleagues identified FGFR1 amplification as a mechanism of resistance to CDK4/6 inhibitors (42). In this study, FGFR1 knockdown sensitized FGFR1-amplified CAMA-1 cells to fulvestrant/palbociclib, and the addition of FGFR inhibitor erdafitinib to fulvestrant/palbociclib further induced tumor regression in patient-derived xenografts. In another study, Drago and colleagues demonstrated that FGFR1 amplification confers resistance to ER, PI3K, and CDK4/6 inhibitors while retaining TORC sensitivity (43). Our results extend our understanding of endocrine resistance with multiple novel findings, identifying novel pathways and genes associated with endocrine resistance by functional screen, demonstrating the acquisition of multiple FGFR pathway alterations after the development of resistance to endocrine therapy and/or CDK4/6 inhibitors in matched tumor samples, and highlighting therapeutic agents that can overcome this resistance.

Our genome-scale screen provided a comprehensive view into the resistance mechanisms to SERDs. Similar resistance genes were nominated for fulvestrant and GDC-0810, thereby confirming the two drugs have similar mechanism of action. Of note, two ESR1 ORFs conferred resistance specifically to GDC-0810 but not fulvestrant, possibly due to GDC-0810 having a less potent effect on ER degradation than fulvestrant (21, 44). Among the resistance mechanisms shared by fulvestrant and GDC-0810, many are frequently altered in ER+ MBC, such as CCNDs/CDK6, KRAS/MAPK, EGFR/ERBB2, and PIK3CA/AKTs/PIMs, and agents targeting those alterations are under clinical development to be combined with endocrine therapy (14, 45). We also identified potential resistance mechanisms that are not characterized to the same extent, such as G protein–coupled receptors, Wnt pathway (FZD10, RSPO1, RSPO3) and Src family kinases (YES1, FYN, FGR), providing clues as to the potential crosstalk between these pathways and ER signaling (46–48) and suggesting that patients with breast cancer harboring functional alterations in these pathways may develop resistance to SERDs. We recognize that those screens were performed using cell lines as model system, and the results will need to be further validated in the clinical setting. We have provided the full genome-scale screen data as a resource to the community of researchers interested in resistance to ER-directed therapies as well as the biology of ER dependencies in ER+ breast cancer.

Our ultimate goal is to identify resistance mechanisms that are clinically relevant and can be therapeutically targeted. By comparing paired pretreatment and posttreatment tumors, our evolutionary analyses identified acquired FGFR1 and FGFR2, and FGF3 alterations in 12 of 60 posttreatment samples, further highlighting a potential role for the FGFR pathway in driving drug resistance and disease progression. Most notably, all four alterations in FGFR2 in our cohort were found to be acquired after the development of resistance to endocrine therapy. Our overall findings are consistent with other recent studies which noted some patients with acquired FGFR1 and FGFR2 alterations following treatment of endocrine therapy (42, 43, 49, 50), and provides a mechanistic explanation for these acquisitions.

This analysis was enabled by a novel method we developed to compare the magnitude of amplification in matched pre- and posttreatment samples while considering key confounders to allow for more reliable assignment of copy gain or loss. Because matched tumor samples of the same patient are highly variable in the cancer cell fraction (purity) and often variable in ploidy, we computed the purity-corrected copy number above ploidy and set a relatively stringent threshold of changes in CNAP to define acquired amplification (see Materials and Methods), as cancer clones bearing amplifications with high focality and magnitude in FGFR/FGF genes are more likely to induce dependency on FGFR pathway and result in endocrine resistance.

Our genomic analysis has some limitations and caveats. The observed alterations may not exclusively result from endocrine therapy as some patients received other therapies between the two collected biopsies. Moreover, tumors with FGFR/FGF alterations also harbor alterations in other cancer genes, which may contribute to drug resistance as well (Supplementary Fig. S5; Supplementary Table S5). Despite these caveats, with the evidence from unbiased screens, genomic evidence in relevant patient samples, and confirmatory experimental models, the FGFR pathway clearly emerges as a clinically important resistance mechanism for SERDs and CDK4/6 inhibitors.

Strategies to target the FGFR pathway in breast cancer patients with FGFR alterations are currently being assessed in clinical trials (51–56). The combination of FGFR inhibitors and endocrine therapy is also being clinically investigated (51). As FGFR pathway activation also results in resistance to CDK4/6 inhibitors, a triple combination with the addition of CDK4/6 inhibitors may also be considered. One challenge for the use of FGFR inhibitors is to identify reliable biomarkers. Our results suggest focal and high-level amplifications, clonal activating mutations or high expression levels of FGFR and FGF genes, particularly in the metastatic setting, may be used to guide the clinical use of FGFR inhibitors. Activating alterations in FGFR2, which are rare in primary treatment naïve breast cancer but appear to be clonally acquired in a subset of patients with resistant ER+ MBC, may be a particularly good biomarker for the development of FGFR inhibitors.

Our work also highlights that the effective clinical use of FGFR inhibitors needs to consider the variable drug sensitivity of different FGFR2 mutations, which were acquired in some patients following endocrine therapy. The two irreversible pan-FGFR kinase inhibitors, FIIN-2 and FIIN-3, had superior efficacy in targeting all FGFR2 mutants including N550K when compared with other FGFR inhibitors. Both FIIN compounds exhibits good overall kinase selectivity at the concentration of 1 μmol/L, although FIIN-3 can also target wild-type and several mutant EGFR (36). The in vivo efficacy, other off-target effects and toxicity of FIIN compounds still warrant further investigation.

Alterations in FGFR1 and FGFR2 activated the MAPK pathway, and MEK inhibition was able to overcome the resistance conferred by FGFR pathway to some degree. We previously demonstrated acquired ERBB2 mutations resulted in a reprogrammed ER signature and an elevated MAPK transcriptional signature (11). In this study, the transcriptional analysis results suggest that MAPK activation resulting from FGFR/FGF overexpression is more pronounced than mTOR activation. This is consistent with our experimental findings, which demonstrated that MAPK inhibition is particularly effective in cells expressing FGFR2-activating alterations. Furthermore, increased frequency of alterations in MAPK pathway genes have been found in tumors posthormonal therapy, including EGFR, ERBB2, and NF1 (14). The fact that multiple mechanisms of resistance to ER-directed therapies and/or CDK4/6 inhibitors activate the MAPK pathway suggests that this may be an important node of resistance in ER+ MBC. Thus, combining endocrine therapy and CDK4/6 inhibitors with agents that target MAPK pathway, such as MEK inhibitors and/or SHP2 inhibitors (57, 58), may be a unifying strategy to overcome or prevent resistance resulting from multiple genetic aberrations that lead to resistance in ER+ MBC. Further in vivo validation is needed to establish the translational implication of the combinational strategies proposed herein.

In summary, the integration of a functional genomic screen and genomic analysis of pre- and posttreatment biopsies revealed the FGFR pathway as an important resistance mechanism for endocrine therapy and CDK4/6 inhibitors in ER+ breast cancer. With the increasing use of SERDs and CDK4/6 inhibitors in the clinic, we anticipate that the prevalence of FGFR/FGF alterations might increase in the future. Targeting the FGFR pathway with FGFR inhibitors or agents that target downstream MAPK signaling may improve clinical outcomes in patients with aberrations in FGFR/FGF genes. Furthermore, our study highlights the need to sequence metastatic biopsy or blood biopsies at the time of resistance to identify patients with these alterations who may benefit from targeting the FGFR pathway.

P. Mao reports grants from AACR, Stand Up to Cancer, and The V Foundation during the conduct of the study. S.A. Wander reports personal fees from Foundation Medicine (consulting), InfiniteMD (consulting), and Puma Biotechnology (ad board) outside the submitted work. A.G. Waks reports other from Genentech (institutional research funding) and other from MacroGenics (institutional research funding) outside the submitted work. J.H. Chung reports personal fees from Foundation Medicine/Roche (employment, stock) during the conduct of the study. S.S. Freeman reports a patent for methods for determining tumor fraction of cell-free DNA (cfDNA) as well as cfDNA whole exome sequencing (WO2017161175A1), assigned to the Broad Institute and not licensed. O. Rozenblatt-Rosen reports other from Broad Institute (funding to the KCO from the Klarman Family Foundation) during the conduct of the study. V.A. Miller reports other from EQRX (employee) and other from Revolution Medicines (board of directors, stockholder) during the conduct of the study; other from ROCHE/FMI (stockholder) and other from Mirati Therapeutics (stockholder) outside the submitted work; as well as a patent, (Methods and compositions for detecting a drug resistant EGFR mutant) US8501413B2 licensed to MolecularMD, assignee Sloan Kettering Institute for Cancer Research. D.E. Root reports grants from Abbvie, Janssen, Merck, and Vir outside the submitted work. A. Regev reports personal fees from Syros, ThermoFisher Scientific, Celsius Therapeutics, Immunitas, Asimov, and Neogene Therapeutics during the conduct of the study; A. Regev also reports employment with Genentech that began on August 1, 2020. E.P. Winer reports personal fees from Carrick Therapeutics, G1 Therapeutics, Genentech/Roche, Genomic Health, GSK, Jounce, Leap, Lilly, Novartis, Seattle Genetics, and Syros outside the submitted work. N.U. Lin reports grants from Genentech, Merck, and Pfizer, grants and personal fees from Seattle Genetics, personal fees from Daichii Sankyo, Denali, California Institute for Regenerative Medicine, and Puma outside the submitted work. N. Wagle reports grants from Department of Defense, American Association for Cancer Research (AACR), NCI, Susan G. Komen, The V Foundation, The Breast Cancer Alliance, The Cancer Couch Foundation, Twisted Pink, Hope Scarves, Stand Up to Cancer, and National Science foundation (NSF) during the conduct of the study; grants and personal fees from Novartis, other from Foundation Medicine, personal fees from Eli Lilly, grants from Puma Biotechnology, and personal fees and other from Relay Therapeutics outside the submitted work. No potential conflicts of interest were disclosed by the other authors.

P. Mao: Conceptualization, formal analysis, validation, investigation, methodology, writing-original draft, writing-review and editing. O. Cohen: Conceptualization, data curation, software, formal analysis, methodology, writing-original draft, writing-review and editing. K.J. Kowalski: Validation, investigation, methodology. J.G. Kusiel: Validation, investigation, methodology. J.E. Buendia-Buendia: Data curation, formal analysis, methodology. M.S. Cuoco: Methodology. P. Exman: Data curation. S.A. Wander: Data curation, writing-review and editing. A.G. Waks: Data curation. U. Nayar: Resources. J. Chung: Resources, data curation. S. Freeman: Methodology. O. Rozenblatt-Rosen: Resources. V.A. Miller: Resources, data curation. F. Piccioni: Resources, methodology. D.E. Root: Resources, methodology. A. Regev: Formal analysis. E.P. Winer: Supervision. N.U. Lin: Resources, supervision. N. Wagle: Conceptualization, formal analysis, supervision, funding acquisition, writing-original draft, writing-review and editing.

This work was supported by the Department of Defense W81XWH-13-1-0032 (N. Wagle), NCI Breast Cancer SPORE at DF/HCC #P50CA168504 (N. Wagle, N.U. Lin, and E.P. Winer), Susan G. Komen CCR15333343 (N. Wagle), The V Foundation (N. Wagle), The Breast Cancer Alliance (N. Wagle), The Cancer Couch Foundation (N. Wagle), Twisted Pink (N. Wagle), Hope Scarves (N. Wagle), Breast Cancer Research Foundation (N.U. Lin and E.P. Winer), ACT NOW (to Dana-Farber Cancer Institute Breast Oncology Program), Fashion Footwear Association of New York (to Dana-Farber Cancer Institute Breast Oncology Program), Friends of Dana-Farber Cancer Institute (to N.U. Lin), Stand Up to Cancer (N. Wagle), National Science Foundation (N. Wagle), and SU2C-TVF Convergence Scholarship (P. Mao). Research supported by the 2013 Landon Foundation-AACR INNOVATOR Award for Research in Personalized Cancer Medicine, Grant Number 13-60-27-WAGL (N. Wagle). Research supported by the 2017 AACR Basic Cancer Research Fellowship, Grant Number 17-40-01-MAP (P. Mao). We thank Qaren Quartey, Christian Kapstad and Gabriela Johnson for technical assistance; Karla Helvie, Laura Dellostritto, Lori Marini, Nelly Oliver, Shreevidya Periyasamy, Colin Mackichan, Max Lloyd, and Mahmoud Charif for assistance with patient sample collection and annotation; and Flora Luo, Tinghu Zhang and Nathanael Gray for providing reagents. We thank Jorge Gómez Tejeda Zañudo for helpful discussions and comments on the manuscript. We are grateful to all the patients who volunteered for our tumor biopsy protocol and generously provided the tissue analyzed in this study.

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.
Chen
WY
,
Colditz
GA
. 
Risk factors and hormone-receptor status: epidemiology, risk-prediction models and treatment implications for breast cancer
.
Nat Clin Pract Oncol
2007
;
4
:
415
23
.
2.
Osborne
CK
,
Schiff
R
. 
Mechanisms of endocrine resistance in breast cancer
.
Annu Rev Med
2011
;
62
:
233
47
.
3.
Lei
JT
,
Gou
X
,
Ellis
MJ
. 
ESR1 fusions drive endocrine therapy resistance and metastasis in breast cancer
.
Mol Cell Oncol
2018
;
5
:
e1526005
.
4.
Robinson
DR
,
Wu
YM
,
Vats
P
,
Su
F
,
Lonigro
RJ
,
Cao
X
, et al
Activating ESR1 mutations in hormone-resistant metastatic breast cancer
.
Nat Genet
2013
;
45
:
1446
51
.
5.
Toy
W
,
Shen
Y
,
Won
H
,
Green
B
,
Sakr
RA
,
Will
M
, et al
ESR1 ligand-binding domain mutations in hormone-resistant breast cancer
.
Nat Genet
2013
;
45
:
1439
45
.
6.
Merenbakh-Lamin
K
,
Ben-Baruch
N
,
Yeheskel
A
,
Dvir
A
,
Soussan-Gutman
L
,
Jeselsohn
R
, et al
D538G mutation in estrogen receptor-alpha: a novel mechanism for acquired endocrine resistance in breast cancer
.
Cancer Res
2013
;
73
:
6856
64
.
7.
Jeselsohn
R
,
Yelensky
R
,
Buchwalter
G
,
Frampton
G
,
Meric-Bernstam
F
,
Gonzalez-Angulo
AM
, et al
Emergence of constitutively active estrogen receptor-alpha mutations in pretreated advanced estrogen receptor-positive breast cancer
.
Clin Cancer Res
2014
;
20
:
1757
67
.
8.
Garcia-Becerra
R
,
Santos
N
,
Diaz
L
,
Camacho
J
. 
Mechanisms of resistance to endocrine therapy in breast cancer: focus on signaling pathways, miRNAs and genetically based resistance
.
Int J Mol Sci
2012
;
14
:
108
45
.
9.
Miller
TW
,
Perez-Torres
M
,
Narasanna
A
,
Guix
M
,
Stal
O
,
Perez-Tenorio
G
, et al
Loss of Phosphatase and Tensin homologue deleted on chromosome 10 engages ErbB3 and insulin-like growth factor-I receptor signaling to promote antiestrogen resistance in breast cancer
.
Cancer Res
2009
;
69
:
4192
201
.
10.
Massarweh
S
,
Osborne
CK
,
Jiang
S
,
Wakeling
AE
,
Rimawi
M
,
Mohsin
SK
, et al
Mechanisms of tumor regression and resistance to estrogen deprivation and fulvestrant in a model of estrogen receptor-positive, HER-2/neu-positive breast cancer
.
Cancer Res
2006
;
66
:
8266
73
.
11.
Nayar
U
,
Cohen
O
,
Kapstad
C
,
Cuoco
MS
,
Waks
AG
,
Wander
SA
, et al
Acquired HER2 mutations in ER(+) metastatic breast cancer confer resistance to estrogen receptor-directed therapies
.
Nat Genet
2019
;
51
:
207
16
.
12.
Croessmann
S
,
Formisano
L
,
Kinch
LN
,
Gonzalez-Ericsson
PI
,
Sudhan
DR
,
Nagy
RJ
, et al
Combined blockade of activating ERBB2 mutations and ER results in synthetic lethality of ER+/HER2 mutant breast cancer
.
Clin Cancer Res
2019
;
25
:
277
89
.
13.
Sokol
ES
,
Feng
YX
,
Jin
DX
,
Basudan
A
,
Lee
AV
,
Atkinson
JM
, et al
Loss of function of NF1 is a mechanism of acquired resistance to endocrine therapy in lobular breast cancer
.
Ann Oncol
2019
;
30
:
115
23
.
14.
Razavi
P
,
Chang
MT
,
Xu
G
,
Bandlamudi
C
,
Ross
DS
,
Vasan
N
, et al
The genomic landscape of endocrine-resistant advanced breast cancers
.
Cancer Cell
2018
;
34
:
427
38
.
15.
Johannessen
CM
,
Johnson
LA
,
Piccioni
F
,
Townes
A
,
Frederick
DT
,
Donahue
MK
, et al
A melanocyte lineage program confers resistance to MAP kinase pathway inhibition
.
Nature
2013
;
504
:
138
42
.
16.
Wilson
FH
,
Johannessen
CM
,
Piccioni
F
,
Tamayo
P
,
Kim
JW
,
Van Allen
EM
, et al
A functional landscape of resistance to ALK inhibition in lung cancer
.
Cancer Cell
2015
;
27
:
397
408
.
17.
Le
X
,
Antony
R
,
Razavi
P
,
Treacy
DJ
,
Luo
F
,
Ghandi
M
, et al
Systematic functional characterization of resistance to PI3K inhibition in breast cancer
.
Cancer Discov
2016
;
6
:
1134
47
.
18.
Gonzalez-Malerva
L
,
Park
J
,
Zou
L
,
Hu
Y
,
Moradpour
Z
,
Pearlberg
J
, et al
High-throughput ectopic expression screen for tamoxifen resistance identifies an atypical kinase that blocks autophagy
.
Proc Natl Acad Sci U S A
2011
;
108
:
2058
63
.
19.
Fox
EM
,
Miller
TW
,
Balko
JM
,
Kuba
MG
,
Sanchez
V
,
Smith
RA
, et al
A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer
.
Cancer Res
2011
;
71
:
6773
84
.
20.
Mendes-Pereira
AM
,
Sims
D
,
Dexter
T
,
Fenwick
K
,
Assiotis
I
,
Kozarewa
I
, et al
Genome-wide functional screen identifies a compendium of genes affecting sensitivity to tamoxifen
.
Proc Natl Acad Sci U S A
2012
;
109
:
2730
5
.
21.
Joseph
JD
,
Darimont
B
,
Zhou
W
,
Arrazate
A
,
Young
A
,
Ingalla
E
, et al
The selective estrogen receptor downregulator GDC-0810 is efficacious in diverse models of ER+ breast cancer
.
eLife
2016
;
5
:
e15828
.
22.
Yang
X
,
Boehm
JS
,
Yang
X
,
Salehi-Ashtiani
K
,
Hao
T
,
Shen
Y
, et al
A public genome-scale lentiviral expression library of human ORFs
.
Nat Methods
2011
;
8
:
659
61
.
23.
Adalsteinsson
VA
,
Ha
G
,
Freeman
SS
,
Choudhury
AD
,
Stover
DG
,
Parsons
HA
, et al
Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors
.
Nat Commun
2017
;
8
:
1324
.
24.
Ulz
P
,
Belic
J
,
Graf
R
,
Auer
M
,
Lafer
I
,
Fischereder
K
, et al
Whole-genome plasma sequencing reveals focal amplifications as a driving force in metastatic prostate cancer
.
Nat Commun
2016
;
7
:
12008
.
25.
Wander
SA
,
Cohen
O
,
Gong
X
,
Johnson
GN
,
Buendia-Buendia
JE
,
Lloyd
MR
, et al
The genomic landscape of intrinsic and acquired resistance to cyclin-dependent kinase 4/6 inhibitors in patients with hormone receptor positive metastatic breast cancer
.
Cancer Discov
2020
;
10
:
1174
93
.
26.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
2012
;
483
:
603
7
.
27.
Rangel
N
,
Villegas
VE
,
Rondon-Lagos
M
. 
Profiling of gene expression regulated by 17beta-estradiol and tamoxifen in estrogen receptor-positive and estrogen receptor-negative human breast cancer cell lines
.
Breast Cancer
2017
;
9
:
537
50
.
28.
Cohen
O
,
Kim
D
,
Oh
C
,
Waks
A
,
Oliver
N
,
Helvie
K
, et al
Abstract S1-01: whole exome and transcriptome sequencing of resistant ER+ metastatic breast cancer
.
Cancer Res
2017
;
77
:
S1
01
.
29.
The Cancer Genome Atlas Network
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
30.
Gallo
LH
,
Nelson
KN
,
Meyer
AN
,
Donoghue
DJ
. 
Functions of Fibroblast Growth Factor Receptors in cancer defined by novel translocations and mutations
.
Cytokine Growth Factor Rev
2015
;
26
:
425
49
.
31.
Jeselsohn
R
,
Buchwalter
G
,
De Angelis
C
,
Brown
M
,
Schiff
R
. 
ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer
.
Nat Rev Clin Oncol
2015
;
12
:
573
83
.
32.
Turner
N
,
Pearson
A
,
Sharpe
R
,
Lambros
M
,
Geyer
F
,
Lopez-Garcia
MA
, et al
FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer
.
Cancer Res
2010
;
70
:
2085
94
.
33.
Shee
K
,
Yang
W
,
Hinds
JW
,
Hampsch
RA
,
Varn
FS
,
Traphagen
NA
, et al
Therapeutically targeting tumor microenvironment-mediated drug resistance in estrogen receptor-positive breast cancer
.
J Exp Med
2018
;
215
:
895
910
.
34.
Reintjes
N
,
Li
Y
,
Becker
A
,
Rohmann
E
,
Schmutzler
R
,
Wollnik
B
. 
Activating somatic FGFR2 mutations in breast cancer
.
PLoS One
2013
;
8
:
e60264
.
35.
Byron
SA
,
Chen
H
,
Wortmann
A
,
Loch
D
,
Gartside
MG
,
Dehkhoda
F
, et al
The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors
.
Neoplasia
2013
;
15
:
975
88
.
36.
Tan
L
,
Wang
J
,
Tanizaki
J
,
Huang
Z
,
Aref
AR
,
Rusan
M
, et al
Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors
.
Proc Natl Acad Sci U S A
2014
;
111
:
E4869
77
.
37.
Yu
M
,
Bardia
A
,
Aceto
N
,
Bersani
F
,
Madden
MW
,
Donaldson
MC
, et al
Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility
.
Science
2014
;
345
:
216
20
.
38.
Sergushichev
AA
. 
An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation
.
bioRxiv
2016
:
060012
.
39.
Ahmed
TA
,
Adamopoulos
C
,
Karoulia
Z
,
Wu
X
,
Sachidanandam
R
,
Aaronson
SA
, et al
SHP2 drives adaptive resistance to ERK signaling inhibition in molecularly defined subsets of ERK-dependent tumors
.
Cell Rep
2019
;
26
:
65
78
.
40.
Fedele
C
,
Ran
H
,
Diskin
B
,
Wei
W
,
Jen
J
,
Geer
MJ
, et al
SHP2 inhibition prevents adaptive resistance to MEK inhibitors in multiple cancer models
.
Cancer Discov
2018
;
8
:
1237
49
.
41.
Ruess
DA
,
Heynen
GJ
,
Ciecielski
KJ
,
Ai
J
,
Berninger
A
,
Kabacaoglu
D
, et al
Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase
.
Nat Med
2018
;
24
:
954
60
.
42.
Formisano
L
,
Lu
Y
,
Servetto
A
,
Hanker
AB
,
Jansen
VM
,
Bauer
JA
, et al
Aberrant FGFR signaling mediates resistance to CDK4/6 inhibitors in ER+ breast cancer
.
Nat Commun
2019
;
10
:
1373
.
43.
Drago
JZ
,
Formisano
L
,
Juric
D
,
Niemierko
A
,
Servetto
A
,
Wander
SA
, et al
FGFR1 gene amplification mediates endocrine resistance but retains TORC sensitivity in metastatic hormone receptor positive (HR+) breast cancer
.
Clin Cancer Res
2019
;
25
:
6443
51
.
44.
Guan
J
,
Zhou
W
,
Hafner
M
,
Blake
RA
,
Chalouni
C
,
Chen
IP
, et al
Therapeutic ligands antagonize estrogen receptor function by impairing its mobility
.
Cell
2019
;
178
:
949
63
.
45.
AlFakeeh
A
,
Brezden-Masley
C
. 
Overcoming endocrine resistance in hormone receptor-positive breast cancer
.
Curr Oncol
2018
;
25
:
S18
S27
.
46.
Loh
YN
,
Hedditch
EL
,
Baker
LA
,
Jary
E
,
Ward
RL
,
Ford
CE
. 
The Wnt signalling pathway is upregulated in an in vitro model of acquired tamoxifen resistant breast cancer
.
BMC Cancer
2013
;
13
:
174
.
47.
Won
HS
,
Lee
KM
,
Oh
JE
,
Nam
EM
,
Lee
KE
. 
Inhibition of beta-catenin to overcome endocrine resistance in tamoxifen-resistant breast cancer cell line
.
PLoS One
2016
;
11
:
e0155983
.
48.
Vallabhaneni
S
,
Nair
BC
,
Cortez
V
,
Challa
R
,
Chakravarty
D
,
Tekmal
RR
, et al
Significance of ER-Src axis in hormonal therapy resistance
.
Breast Cancer Res Treat
2011
;
130
:
377
85
.
49.
Yates
LR
,
Knappskog
S
,
Wedge
D
,
Farmery
JHR
,
Gonzalez
S
,
Martincorena
I
, et al
Genomic evolution of breast cancer metastasis and relapse
.
Cancer Cell
2017
;
32
:
169
84
.
50.
O'Leary
B
,
Cutts
RJ
,
Liu
Y
,
Hrebien
S
,
Huang
X
,
Fenwick
K
, et al
The genetic landscape and clonal evolution of breast cancer resistance to palbociclib plus fulvestrant in the PALOMA-3 trial
.
Cancer Discov
2018
;
8
:
1390
403
.
51.
Musolino
A
,
Campone
M
,
Neven
P
,
Denduluri
N
,
Barrios
CH
,
Cortes
J
, et al
Phase II, randomized, placebo-controlled study of dovitinib in combination with fulvestrant in postmenopausal patients with HR(+), HER2(−) breast cancer that had progressed during or after prior endocrine therapy
.
Breast Cancer Res
2017
;
19
:
18
.
52.
Andre
F
,
Bachelot
T
,
Campone
M
,
Dalenc
F
,
Perez-Garcia
JM
,
Hurvitz
SA
, et al
Targeting FGFR with dovitinib (TKI258): preclinical and clinical data in breast cancer
.
Clin Cancer Res
2013
;
19
:
3693
702
.
53.
Smyth
EC
,
Turner
NC
,
Pearson
A
,
Peckitt
C
,
Chau
I
,
Watkins
DJ
, et al
Phase II study of AZD4547 in FGFR amplified tumours: Gastroesophageal cancer (GC) cohort pharmacodynamic and biomarker results
.
J Clin Oncol
2016
;
34
:
154
.
54.
Babina
IS
,
Turner
NC
. 
Advances and challenges in targeting FGFR signalling in cancer
.
Nat Rev Cancer
2017
;
17
:
318
32
.
55.
Chae
YK
,
Ranganath
K
,
Hammerman
PS
,
Vaklavas
C
,
Mohindra
N
,
Kalyan
A
, et al
Inhibition of the fibroblast growth factor receptor (FGFR) pathway: the current landscape and barriers to clinical application
.
Oncotarget
2017
;
8
:
16052
74
.
56.
Nogova
L
,
Sequist
LV
,
Perez Garcia
JM
,
Andre
F
,
Delord
JP
,
Hidalgo
M
, et al
Evaluation of BGJ398, a fibroblast growth factor receptor 1-3 kinase inhibitor, in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors: results of a global phase I, dose-escalation and dose-expansion study
.
J Clin Oncol
2017
;
35
:
157
65
.
57.
Matalkah
F
,
Martin
E
,
Zhao
H
,
Agazie
YM
. 
SHP2 acts both upstream and downstream of multiple receptor tyrosine kinases to promote basal-like and triple-negative breast cancer
.
Breast Cancer Res
2016
;
18
:
2
.
58.
Chen
YN
,
LaMarche
MJ
,
Chan
HM
,
Fekkes
P
,
Garcia-Fortanet
J
,
Acker
MG
, et al
Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases
.
Nature
2016
;
535
:
148
52
.