The genomic revolution has fundamentally changed our perception of breast cancer. It is now apparent from DNA-based massively parallel sequencing data that at the genomic level, every breast cancer is unique and shaped by the mutational processes to which it was exposed during its lifetime. More than 90 breast cancer driver genes have been identified as recurrently mutated, and many occur at low frequency across the breast cancer population. Certain cancer genes are associated with traditionally defined histologic subtypes, but genomic intertumoral heterogeneity exists even between cancers that appear the same under the microscope. Most breast cancers contain subclonal populations, many of which harbor driver alterations, and subclonal structure is typically remodeled over time, across metastasis and as a consequence of treatment interventions. Genomics is deepening our understanding of breast cancer biology, contributing to an accelerated phase of targeted drug development and providing insights into resistance mechanisms. Genomics is also providing tools necessary to deliver personalized cancer medicine, but a number of challenges must still be addressed. Clin Cancer Res; 23(11); 2630–9. ©2017 AACR.

See all articles in this CCR Focus section, “Breast Cancer Research: From Base Pairs to Populations.”

Technical and analytic developments underpin major advances in our understanding of breast cancer biology over the last two decades. The application of gene expression arrays in the early 2000s led to a refined molecular classification, the development of prognostic assays, and a better characterization of the contribution of the tumor microenvironment (1, 2). First steps toward translating molecularly defined breast cancer subsets came from efforts using gene expression–based signatures, which have led to multiple commercially available and clinically useful assays in hormone receptor–positive breast cancer, and are continuing to be developed for multiple clinical/translational applications today. Discussing RNA-based assays is beyond the scope of the current review but was the topic of a recent CCR 20th Anniversary Commentary (2). The genomic revolution later saw the emergence of commercially available massively parallel sequencing approaches that permit the comprehensive profiling of an entire cancer's genome, all coding genes (exome), or a selection of genes (targeted sequencing). DNA sequencing technologies offer the potential to deliver personalized medicine by matching appropriate targeted therapeutics with unique molecular aberrations within an individual's cancer. Furthermore, massively parallel sequencing can be used to identify biomarkers associated with response to experimental targeted therapies, both in clinical trials and in vitro.

Here we consider five practical applications of the genomic revolution, summarizing in each case what we have learned thus far and considering the challenges that remain for future endeavors (Fig. 1). Key messages include the following: (i) the driver landscape of breast cancer includes many rare cancer genes and an unexpected level of inter- and intratumoral driver mutation heterogeneity; (ii) understanding the mutational processes operating throughout a cancer's lifetime may identify therapeutic vulnerabilities; (iii) many genomic features align with traditional molecular and histologic subtypes that can aid classification; (iv) cancers demonstrate subclonal diversification that may underlie primary and acquired treatment resistance; and (v) a range of patterns of metastatic dissemination are observed reflecting clonal evolution.

Figure 1.

Applications of the genomic revolution in breast cancer. Each of the five segments represents an application of the genomic revolution, main findings are pictured within the inner, white panel, and outstanding challenges are summarized in the outer, blue panel. Histologic subtypes: invasive ductal cancer (IDC) and invasive lobular cancer (ILC).

Figure 1.

Applications of the genomic revolution in breast cancer. Each of the five segments represents an application of the genomic revolution, main findings are pictured within the inner, white panel, and outstanding challenges are summarized in the outer, blue panel. Histologic subtypes: invasive ductal cancer (IDC) and invasive lobular cancer (ILC).

Close modal

To date, almost 100 high-probability breast cancer driver genes have been identified through the application of computational approaches to massively parallel sequence data generated from more than 4,000 primary breast cancer samples (Supplementary Table S1; refs. 3–18). Cancer genes have been identified based upon somatic mutation recurrence rates that exceed expectation after taking into account features such as background mutation rate and spectrum, nucleotide context, enrichment in phosphorylation-specific regions or tertiary protein structures, and the ratio of nonsynonymous to synonymous mutations (19–24). Various alternative methodologies exist such as those that adopt subnetwork analyses to identify groups of potential cancer genes by incorporating pathway or gene interaction information, and these may be particularly useful for identifying very rare driver genes (25–28).

Saturation analyses predict that based on the number of samples now sequenced, most cancer genes implicated in 2% or more breast cancers will have been identified (29). However, we expect that many more low-frequency cancer genes remain to be discovered, because there is emerging evidence, as discussed below, that metastatic, pretreated, male, and special histopathologic subtypes of cancers are genomically distinct from the “general” breast cancer population (Tables 1 and 2; Supplementary Table S1; refs. 6, 10–18, 30–37). Rare “populations” are often the most clinically challenging to treat, and further focused driver discovery exercises in these groups are warranted.

Table 1.

Overview of the genomic findings and distribution of the PAM50 intrinsic molecular subtypes in the general breast cancer population, according to the estrogen receptor (ER) status

Statistics genomic alterations: median (min, max)Recurrent drivers (subs/indels)Recurrent drivers (CNAs/SVs)Clinical associationsPrognostic associationsPAM50 subtypes
ER positive 
  • Subs: 2,637 (507–76,097)

  • Indels: 180 (34–19,436)

  • SVs: 50 (0–1,221)

  • Drivers: 3 (0–14)

 
  • PIK3CA (36%)

  • TP53 (20%)

  • GATA3 (15%)

  • MAP3K1 (9%)

  • CDH1 (8%)

  • KMT2C (6%)

  • PTEN (6%)

 
  • CCND1 amp (22%)

  • MYC amp (16%)

  • ZNF703/FGFR1 amp (15%)

  • HER2 amp (11%)

  • ZNF217 amp (9%)

  • MAP2K4 del or SV (8%)

 
  • PIK3CA, MAP3K1, KMT2C, and CBFB mutations with low grade

  • TP53 mutations with high grade

  • GATA3 and CBFB mutations with younger ages

  • CDH1 and SF3B1 mutations with older ages

 
  • TP53, SMAD4, and USP9X mutations with worse survival

  • GATA3 and MAP3K1 with longer survival

 
  • HER2 (n = 2,123): Lum A 47%, Lum B 28%, HER2-E 5%, basal 6%, normal-like 13%

  • HER2+ (n = 250): Lum A 26%, Lum B 26%, HER2-E 37%, basal 6%, normal-like 4%

 
ER negative 
  • Subs: 6,924 (722–93,102)

  • Indels: 313 (19–66,764)

  • SVs: 176 (0–1,221)

  • Drivers: 3 (0–11)

 
  • TP53 (77%)

  • PIK3CA (13%)

  • RB1 (10%)

  • PTEN (6%)

  • KMT2C (6%)

  • PIK3R1 (4%)

 
  • MYC amp (25%)

  • PTEN del or SV (25%)

  • HER2 amp (16%)

  • RB1 del or SV (14%)

  • ARID1B SV (7%)

  • CCND3 amp (7%)

 
  • TP53 mutations with high grade

  • CDH1 and HER2 mutations with low grade

  • KMT2C mutations with older ages

 
  • PIK3CA and NF1 mutations with worse survival

 
  • HER2 (n = 453): Lum A 2%, Lum B 0%, HER2-E 8%, basal-like 84%, normal-like 6%

  • HER2+ (n = 154): Lum A 0%, Lum B 1%, HER2-E 69%, basal-like 25%, normal-like 5%

 
Statistics genomic alterations: median (min, max)Recurrent drivers (subs/indels)Recurrent drivers (CNAs/SVs)Clinical associationsPrognostic associationsPAM50 subtypes
ER positive 
  • Subs: 2,637 (507–76,097)

  • Indels: 180 (34–19,436)

  • SVs: 50 (0–1,221)

  • Drivers: 3 (0–14)

 
  • PIK3CA (36%)

  • TP53 (20%)

  • GATA3 (15%)

  • MAP3K1 (9%)

  • CDH1 (8%)

  • KMT2C (6%)

  • PTEN (6%)

 
  • CCND1 amp (22%)

  • MYC amp (16%)

  • ZNF703/FGFR1 amp (15%)

  • HER2 amp (11%)

  • ZNF217 amp (9%)

  • MAP2K4 del or SV (8%)

 
  • PIK3CA, MAP3K1, KMT2C, and CBFB mutations with low grade

  • TP53 mutations with high grade

  • GATA3 and CBFB mutations with younger ages

  • CDH1 and SF3B1 mutations with older ages

 
  • TP53, SMAD4, and USP9X mutations with worse survival

  • GATA3 and MAP3K1 with longer survival

 
  • HER2 (n = 2,123): Lum A 47%, Lum B 28%, HER2-E 5%, basal 6%, normal-like 13%

  • HER2+ (n = 250): Lum A 26%, Lum B 26%, HER2-E 37%, basal 6%, normal-like 4%

 
ER negative 
  • Subs: 6,924 (722–93,102)

  • Indels: 313 (19–66,764)

  • SVs: 176 (0–1,221)

  • Drivers: 3 (0–11)

 
  • TP53 (77%)

  • PIK3CA (13%)

  • RB1 (10%)

  • PTEN (6%)

  • KMT2C (6%)

  • PIK3R1 (4%)

 
  • MYC amp (25%)

  • PTEN del or SV (25%)

  • HER2 amp (16%)

  • RB1 del or SV (14%)

  • ARID1B SV (7%)

  • CCND3 amp (7%)

 
  • TP53 mutations with high grade

  • CDH1 and HER2 mutations with low grade

  • KMT2C mutations with older ages

 
  • PIK3CA and NF1 mutations with worse survival

 
  • HER2 (n = 453): Lum A 2%, Lum B 0%, HER2-E 8%, basal-like 84%, normal-like 6%

  • HER2+ (n = 154): Lum A 0%, Lum B 1%, HER2-E 69%, basal-like 25%, normal-like 5%

 

NOTE: The data from the first three columns were extracted from the “560 genomes” series (9), the clinical and prognostic associations were taken from the results on the METABRIC series (10), whereas the distribution of the intrinsic subtypes was based on the combined METABRIC (10) and The Cancer Genome Atlas (TCGA; ref. 6) series (C. Perou; personal communication).

Abbreviations: amp, amplification; CNA, copy number aberrations; del, deletion; HER2-E, HER2-enriched; Lum A, Luminal A; Lum B, Luminal B; Subs, substitutions; SV, structural variants.

Table 2.

Key genomic findings in rare histologic subtypes (massively parallel sequencing)

Histologic subtypeFrequencyNumber of cases evaluated by MPSKey genomic findings
Mostly ER and HER2 
Adenoid cystic (32) <1% 12 
  • Mutation burden and landscape of these cases more concordant with those from adenoid cystic carcinomas of the salivary gland than triple-negative breast cancers (TNBC).

  • Harbor the typical MYB–NFIB fusion gene and lack TP53 and PIK3CA mutations.

 
Apocrine (9, 12, 15) ∼1% 15 
  • All cases with PIK3CA or PTEN alterations, less TP53 (∼30%) compared with other TNBC.

  • Genomic features consistent with luminal subtype.

 
Medullary (10) <5% 34 
  • TP53 and PIK3CA mutations present in 87% and 9% of the cases, respectively.

 
Metaplastic (9, 11, 18, 33) 0.2%–5% 25 
  • TP53 mutations present in ∼70% of the cases, similar to other TNBCs.

  • PI3K/AKT/mTOR pathway and Wnt pathway–related genes each altered in >50% of the cases.

 
  • Mostly ER+ and HER2

 
Lobular (6, 10, 13, 17, 18) 5%–15% >1,200 
  • CDH1 mutated in ∼60% and LOH in ∼90%.

  • Alterations in PI3K pathway (PIK3CA, PTEN, AKT1) present in >half of the tumors, with Akt/mTOR activation in 45% of the cases.

  • AKT1, FOXA1, HER2, HER3, PTEN, and TBX3 alterations more frequent in ILC than IDC.

  • HER2 and AKT1 mutations associated with increased risk of early relapse.

  • Histologic subtype–specific associations: ESR1 gains in solid subtype, HER2 mutations in mixed nonclassic, and TP53 mutations in both.

 
Micropapillary (9, 18) 3%–6% 11 
  • Profile similar to common breast cancers—PIK3CA and MAP3K1 mutations present in 45%, GATA3 in 27%, and PTEN in 18%. TP53 alteration observed in only one sample.

 
Mucinous (9–11) ∼2% 61 (22 from metastatic pts) 
  • Characterized by increased frequency of GATA3 (23%) mutations, and decreased frequency of PIK3CA (8%) and TP53 (8%) alterations compared to IDC.

  • One third of primary tumors do not present any reported driver mutation.

  • Sequencing of 22 metastatic cases (for half of the patients, samples originated from the primary tumor and for the remaining, from metastases) revealed higher frequency of ZNF703/FGFR1 amplification (36%), CCND1 amplification (27%), HER2 alterations (23%), and PIK3CA mutations (23%) compared with nonmetastatic mucinous breast cancer.

 
Neuroendocrine (16) 2%–5% 18 
  • Genomics are similar to neuroendocrine tumors from other sites.

  • Compared with other more common types of ER+/HER2 breast cancers, mutation frequency in TP53 and PIK3CA is lower, but ARID1A, FOXA1, and TBX3 is higher.

 
Histologic subtypeFrequencyNumber of cases evaluated by MPSKey genomic findings
Mostly ER and HER2 
Adenoid cystic (32) <1% 12 
  • Mutation burden and landscape of these cases more concordant with those from adenoid cystic carcinomas of the salivary gland than triple-negative breast cancers (TNBC).

  • Harbor the typical MYB–NFIB fusion gene and lack TP53 and PIK3CA mutations.

 
Apocrine (9, 12, 15) ∼1% 15 
  • All cases with PIK3CA or PTEN alterations, less TP53 (∼30%) compared with other TNBC.

  • Genomic features consistent with luminal subtype.

 
Medullary (10) <5% 34 
  • TP53 and PIK3CA mutations present in 87% and 9% of the cases, respectively.

 
Metaplastic (9, 11, 18, 33) 0.2%–5% 25 
  • TP53 mutations present in ∼70% of the cases, similar to other TNBCs.

  • PI3K/AKT/mTOR pathway and Wnt pathway–related genes each altered in >50% of the cases.

 
  • Mostly ER+ and HER2

 
Lobular (6, 10, 13, 17, 18) 5%–15% >1,200 
  • CDH1 mutated in ∼60% and LOH in ∼90%.

  • Alterations in PI3K pathway (PIK3CA, PTEN, AKT1) present in >half of the tumors, with Akt/mTOR activation in 45% of the cases.

  • AKT1, FOXA1, HER2, HER3, PTEN, and TBX3 alterations more frequent in ILC than IDC.

  • HER2 and AKT1 mutations associated with increased risk of early relapse.

  • Histologic subtype–specific associations: ESR1 gains in solid subtype, HER2 mutations in mixed nonclassic, and TP53 mutations in both.

 
Micropapillary (9, 18) 3%–6% 11 
  • Profile similar to common breast cancers—PIK3CA and MAP3K1 mutations present in 45%, GATA3 in 27%, and PTEN in 18%. TP53 alteration observed in only one sample.

 
Mucinous (9–11) ∼2% 61 (22 from metastatic pts) 
  • Characterized by increased frequency of GATA3 (23%) mutations, and decreased frequency of PIK3CA (8%) and TP53 (8%) alterations compared to IDC.

  • One third of primary tumors do not present any reported driver mutation.

  • Sequencing of 22 metastatic cases (for half of the patients, samples originated from the primary tumor and for the remaining, from metastases) revealed higher frequency of ZNF703/FGFR1 amplification (36%), CCND1 amplification (27%), HER2 alterations (23%), and PIK3CA mutations (23%) compared with nonmetastatic mucinous breast cancer.

 
Neuroendocrine (16) 2%–5% 18 
  • Genomics are similar to neuroendocrine tumors from other sites.

  • Compared with other more common types of ER+/HER2 breast cancers, mutation frequency in TP53 and PIK3CA is lower, but ARID1A, FOXA1, and TBX3 is higher.

 

Abbreviations: IDC, invasive ductal cancer; ILC, invasive lobular cancer; MPS, massively parallel sequencing; pts, patients.

Driver alterations refer to events that are predicted to inactivate tumor suppressor genes or activate oncogenes—common mechanisms in the former include homozygous deletions, truncating point mutations, or disrupting rearrangements and in the latter, focal amplifications and activating missense mutations, typically within mutational hotspots. A recent genome-wide analysis of 560 breast cancers (9) reported that the average cancer contains three such driver alterations (range = 0–14) and, consistent with other series, confirmed that only a handful of cancer genes are mutated in more than 10% of breast cancers (3–10). The most frequently implicated cancer genes (and their frequencies) across the 560 breast cancers are TP53 (41%), PIK3CA (30%), MYC (20%), PTEN (16%), CCND1 (16%), ERBB2 (13%), FGFR1 (11%), and GATA3 (10%; ref. 9). Because most cancer genes are mutated in less than 5% of cancers, considerable interpatient heterogeneity exists (4, 7). By example, whole-exome sequencing of 100 breast cancers identified driver point mutations and copy number changes in 40 genes in 73 combinations, indicating that at the driver gene level, most primary breast cancers are distinct (4). The wide range of possible driver combinations within an individual cancer will demand systematic approaches to facilitate target prioritization and guide combination therapy approaches if we reach a position where a large number of effective therapeutic targets are available.

The majority of driver events detected in a primary tumor are clonal, being present in all tumor cells. However, subclones are common, and these can harbor driver events that alter some of the most common and most frequently clonal breast cancer genes including TP53, MYC, PIK3CA, and HER2, in addition to rarer cancer genes such as FGFR2 and RUNX1 (3, 35, 36, 38, 39). In a multiregion sampling study, we identified subclonal, potentially targetable alterations in 13 of 50 primary breast cancers and demonstrated that subclones can be spatially separated, posing a challenge to representative sampling (36). In a quarter of these cases, we identified parallel evolution, with the same cancer gene mutated multiple times in different subclones in the same tumor, indicating that particular subclonal aberrations within an individual cancer may be under strong selection (36). Highlighting the clinical relevance of such a finding, in one case of metastatic breast cancer, parallel genetic loss of PTEN in multiple metastatic sites was documented after treatment with the PI(3)Kα inhibitor BYL719 (40). In this case, the emergence of a convergent PTEN-null treatment–resistant phenotype provided strong rationale for targeting the subclonal alterations; however, in general, it is not yet clear when and if we should target subclonal alterations or if a greater benefit will be derived from targeting clonal alterations that are present in every cancer cell.

Determining the range of driver events operating within an individual breast cancer depends, to some extent, on the sequencing technology employed (Fig. 2). Targeted approaches can provide (at lower cost than whole-genome sequencing) high-depth sequence coverage, high sensitivity, and accurate quantification of point mutations, overcoming, to some extent, technical challenges such as low tumor cellularity and driver mutation subclonality (ref. 41; Fig. 2). However, targeted approaches make prior assumptions about the range of relevant cancer genes. Furthermore, breast cancers are characterized by frequent and often complex structural variation, and the precise nature of structural variation can only be captured using genome-wide sequencing (Fig. 2; ref. 42). For example, a recent study revealed that rearrangement breakpoints disrupt tumor suppressor gene footprints more frequently than expected, contributing to the recessive cancer gene landscape, with the most frequently affected genes being PTEN, RB1, and ARID1B in 7%, 5%, and 4% of the cases, respectively (9). The same study revealed that recurrent fusion genes and noncoding driver mutations are uncommon in primary breast cancers (9). The full contribution of structural variation to the driver landscape of breast cancers remains to be determined. Selecting the most appropriate technology for incorporation in clinico-genomic trials is of paramount importance.

Figure 2.

Insights into breast cancer genomes by applying different massively parallel sequencing (MPS) approaches. Comparison of whole-genome sequencing (WGS), whole-exome sequencing (WES), and targeted sequencing (TS) as approaches for exploring the breast cancer genome. Two typical but contrasting examples of breast cancer genomes are presented. WGS reveals all genomic mutations types as presented within the two Circos plots. Colored dots in the outer ring represent base substitutions (Subs), copy number changes are reported in the second ring, whereas the colored lines in the center represent structural variant (SV), otherwise known as rearrangements. WES and TS identify only a fraction of mutations and do not necessarily reflect the stark differences between the two genomic landscapes pictured. The large numbers of mutations identified by WGS permit mutation signature detection. WES identifies typically 10 to 300 mutations, and signature analysis is unreliable at the lower end of this range. Notably, the homologous recombination deficiency (HRD) substitution signature is relatively subtle, and confident detection usually demands several hundred mutations. Although not represented in this figure, the high depth afforded by TS offers the additional advantage of exploring subclonality by comparing different mutations' variant allele fractions within a tumor, providing accurate copy number can be determined. Note the reported numbers are estimates only for illustration purposes. NA, not available.

Figure 2.

Insights into breast cancer genomes by applying different massively parallel sequencing (MPS) approaches. Comparison of whole-genome sequencing (WGS), whole-exome sequencing (WES), and targeted sequencing (TS) as approaches for exploring the breast cancer genome. Two typical but contrasting examples of breast cancer genomes are presented. WGS reveals all genomic mutations types as presented within the two Circos plots. Colored dots in the outer ring represent base substitutions (Subs), copy number changes are reported in the second ring, whereas the colored lines in the center represent structural variant (SV), otherwise known as rearrangements. WES and TS identify only a fraction of mutations and do not necessarily reflect the stark differences between the two genomic landscapes pictured. The large numbers of mutations identified by WGS permit mutation signature detection. WES identifies typically 10 to 300 mutations, and signature analysis is unreliable at the lower end of this range. Notably, the homologous recombination deficiency (HRD) substitution signature is relatively subtle, and confident detection usually demands several hundred mutations. Although not represented in this figure, the high depth afforded by TS offers the additional advantage of exploring subclonality by comparing different mutations' variant allele fractions within a tumor, providing accurate copy number can be determined. Note the reported numbers are estimates only for illustration purposes. NA, not available.

Close modal

Defining the targetable landscape of breast cancer remains in its infancy. High-throughput functional validation approaches are indicated to determine the significance of potential cancer genes and driver mutations (43, 44). Importantly, the clinical viability of even some of the most common cancer genes (present in more than 10% of cancers) as therapeutic targets remains as yet unproven, with only ERBB2 and PIK3CA (45) associated with objective responses to targeted therapies and only the former demonstrating validated and independent prognostic and predictive value (46). TP53, GATA3, and MYC have diverse transcriptional activities, and attempts to target them both directly and indirectly have proved difficult for various reasons, including lack of specificity and efficiency (47–51). These targets remain an intense area of research. For example, clinical trials assessing the activity of PIM kinase inhibitors in MYC-amplified triple-negative breast cancers are now keenly awaited following promising preclinical work that identified a targetable synthetic lethal interaction between PIM1 and MYC (52).

It is important to consider that the presence of an apparent target does not necessarily predict for response. Recent phase III studies confirmed a survival advantage associated with the use of mTOR inhibitors (53) and CDK4/6 inhibitors (54) in advanced estrogen receptor (ER)–positive breast cancers; however, to date, biomarkers to predict response to these agents have not been identified. In the PALOMA 1 trial, CCND1 amplifications or loss of p16 failed to predict for a response to the CDK4/6 inhibitor palbociclib (55). In a retrospective analysis of the BOLERO-2 trial, progression-free survival benefit was observed in association with the mTOR inhibitor everolimus irrespective of the presence or absence of genomic alterations within PIK3CA, CCND1, and FGFR1 (56). However, the extent of benefit from everolimus varied according to the exon specific mutation (exon 20 versus exon 9), indicating that the definition of a target may be more nuanced than initially appreciated.

Overcoming the significant challenges of defining and ultimately targeting the driver landscape of breast cancer will demand large-scale, international, novel clinico-genomic screening and clinical trial strategies, as has been discussed comprehensively elsewhere (57). It is also clear that, since many driver alterations are ultarare in breast cancer, studies from single cases or small case series will also be extremely valuable, particularly for exploring extreme response or therapeutic resistance (58).

Whole-genome sequencing identifies thousands of mutations within each cancer, and although the majority of them are passenger events, the patterns that they form, termed “mutational signatures,” can reveal the mutational processes that shaped that cancer's evolution (59). A recent analysis of 560 breast cancer whole genomes identified 20 distinct mutational signatures (9). Typically, multiple mutational processes are identified within each individual breast cancer, and their relative contributions may vary over time (36, 60).

A comprehensive overview of mutational processes in breast cancer is presented in another article in this CCR Focus section (61). In the context of “practical applications,” the fundamental principle is that understanding the processes that generate mutational diversity, the substrate of natural selection, could ultimately result in strategies to oppose cancer development and progression (62). However, for most mutational signatures, both etiology and direct clinical utility are at this time unclear. At present, the mutation signatures with the greatest potential clinical benefit are those that identify deficiencies in the homologous recombination repair pathway and may predict for sensitivity to platinum salt chemotherapies and PARP inhibitors (63–68). Such signatures could extend the number of patients for whom such agents may confer benefit, beyond those with germline or somatic mutations or promoter methylation within BRCA1/2 and other BRCA-related genes (9, 69, 70), and has been used to confirm that an ER-positive tumor arising in a BRCA1 carrier is related to the germline lesion rather than being a sporadic cancer (71).

Despite considerable intertumoral heterogeneity, certain genomic patterns align with the molecular and histologic subtypes. For example, luminal-like/ER-positive breast cancers typically harbor mutations in PIK3CA, GATA3, or MAP3K1 (6–8). The most commonly mutated cancer gene in basal-like/triple-negative breast cancers is TP53, and this usually contains truncating (frameshift or nonsense) mutations (72). In contrast, when TP53 mutations occur in luminal-like cancers, they are usually missense mutations (8, 10, 73). Characteristic genomic changes also underlie histologic subtypes. Recently, several consortia have genomically characterized more than 1,225 invasive lobular breast cancers (6, 10, 13, 17, 18). Lobular tumors typically express ER and lack HER2 amplification, and represent the second most frequent histologic subtype, accounting for up to 15% of all invasive breast carcinomas (74). Lobular tumors display a distinct mutational pattern compared with their ER-positive/HER2-negative ductal counterparts, including a higher frequency of CDH1 and PTEN loss and AKT1, ERBB2, ERBB3, and TBX3 mutations (6, 10, 13, 17). Furthermore, ER-positive tumors of lobular and ductal type have a strong tendency toward FOXA1 or GATA3 mutations, respectively, indicating that distinct genomic mechanisms of ER activation may exist in these distinct pathologies (6, 17, 75). Distinct genomic patterns are also characteristic of rare tumor types, including adenoid cystic, apocrine, medullary, mucinous, metaplastic, micropapillary, and neuroendocrine tumors, and are detailed in Table 1 and Supplementary Table S1 (10–12, 15, 16, 18, 32, 33, 76).

We continue to extrapolate from global breast cancer populations to make clinical treatment decisions for special histologic subtypes. Now, there is growing evidence of molecular differences between these subtypes. We should, therefore, acknowledge both molecular and histologic subtypes as relevant pathologic covariates in future clinical and translational research. In addition, because certain potential therapeutic targets are enriched in some subtypes, such as ERBB2 and ERBB3 mutations in lobular carcinomas, one could imagine that conventional pathology could be used to enhance mutational screening for clinical trial enrollment.

Clinically overt metastasis to distant organs is currently incurable. The most informative sequencing approaches exploring this fatal transition compare primary tumor and metastatic deposits from the same individual (Supplementary Table S1). Studies focused on coding mutations reveal that most of the driver events within the primary tumor are shared with associated metastatic deposits (77–80). This indicates that the primary tumor may be a good surrogate for the relapse-seeding clone, which is the target for adjuvant therapies that have the potential to improve cancer cure rates (79, 80). For example, in a study of single metastatic deposits from 33 individuals, 87% of driver mutations and 86% of copy number aberrations were also detected in the primary tumor (79). A recent single-cell sequencing study of disseminated tumor cells supported this linear model of late dissemination from the primary tumor (81). Metastatic deposits generally harbor additional private mutations, often including known driver mutations, reflecting ongoing subclonal diversification after dissemination (36, 77–80, 82).

A handful of published studies have started to unravel the genomic basis of breast cancer progression and define the temporal ordering of mutations through the analysis of the primary tumor and multiple metastatic deposits acquired during “rapid” autopsy (Supplementary Table S1; refs. 40, 77–80, 82–84). In this context, a recent survey conducted in Australia to evaluate patients' attitudes toward rapid autopsy and tissue donation revealed that 87% were willing to donate tissue at death (85). So far, multiple tumor deposits from a total of 17 breast cancer patients who underwent autopsy have been investigated at the mutational and copy number level (40, 80, 83, 84). Although each patient reported in these studies has her own history of treatment and disease evolution, several messages emerge. First, given the observed primary/metastasis and intermetastasis genomic heterogeneity, there is no single tumor sample that recapitulates the complete disease, that is, all the genomic alterations present in all samples from the patient (40, 80, 82–84). Second, both linear and parallel evolution from the primary tumor are observed (Fig. 1; refs. 80, 83, 84). In some patients, there is only one successful seeding event that gives rise to a so-called metastatic precursor that then seeds the other metastases, possibly in a cascading manner (Fig. 1). In other patients, multiple successful seeding events arise from the primary tumor. Finally, evidence is accumulating that metastatic cross-seeding occurs between established metastases (83, 84).

A potential limitation of the studies that investigate metastasis using a targeted sequencing approach is that the cancer genes present in these panels were derived almost exclusively from the study of treatment-naïve primary tumors and, consequently, may fail to identify genes enriched or specific to metastatic, relapsed, or pretreated cancers. Some preliminary evidence from metastatic tumors suggests indeed that different drivers may be associated with metastasis, and that these are drawn from an even wider range of cancer genes than in primary tumors (Supplementary Table S1; refs. 30, 34, 79, 86). It is conceivable that some genes only act as drivers in nuanced situations, for example, in resisting a specific anticancer therapy, and in specific cancer subtypes, so studies should be powered to identify rare resistance mutations, a theme that will be explored in further detail later in this review (34, 87–89).

Extensive work remains within this field to determine when and what driver mutations are most relevant to the application of personalized medicine. Exhaustive sampling of all metastatic deposits is not practical, so if metastasis-specific mutations are clinically important, representative sampling strategies need to be developed. Options include sampling areas of progressive disease after treatment interventions, particularly when a mixed response is seen, or alternatively, using “liquid biopsies” (refs. 90–92; Supplementary Table S1). In one study, sequencing of circulating tumor DNA (ctDNA) successfully identified changes in the levels of subclonal mutations as the relevant metastatic sites responded or progressed, providing proof of principle for the role of a liquid biopsy to monitor treatment response (90). Such an approach can also use ctDNA to predict minimal residual disease and relapse, potentially extending the window of opportunity for applying adjuvant therapies to further improve cancer cure rates (93).

Changes in subclonal structure have been demonstrated across treatment interventions using cytotoxic and endocrine therapy and targeted therapies (35–37, 40, 94). Several genomic mechanisms of resistance to various therapies have recently been uncovered, with mutations in ESR1, the gene encoding the ER, being the most described and studied in the context of endocrine resistance (34, 87, 95–98). Mutations cluster in the ligand-binding domain of ESR1 and activate ER in the absence of the estrogen ligand. These mutations have been detected almost exclusively in the context of prior endocrine therapy, typically with aromatase inhibitors, and more frequently where sensitivity to prior endocrine therapy was observed (98), suggesting that these mutations are associated with acquired, but not primary, endocrine resistance. A recent large-scale initiative, the Metastatic Breast Cancer Project, characterized over 140 metastatic biopsies from endocrine therapy–treated patients and identified several acquired mutations, in addition to ESR1, that may be involved in endocrine resistance, including AKT1, ERBB2, KRAS, and RB1 alterations (99). There is also preliminary evidence from cell-line and patient-derived xenograft models that CDK4/6-inhibitor resistance may be mediated by loss of RB1 or amplification of CCNE1 or CDK6 (100, 101).

Treatment resistance might be complicated by heterogeneity, for example, multiple metastatic deposits from an individual may exhibit parallel evolution of genomic resistance, whereas other metastases may not have any genomic mark of resistance, potentially explaining the heterogeneous response to treatment observed in the clinic (40, 83, 96–98, 102).

Interrogating ctDNA represents an attractive option for identifying multiple mechanisms of resistance in different metastases. This is the strategy adopted by the recent UK plasmaMATCH trial (ISRCTN16945804), a primary aim of which is to determine whether plasma-derived ctDNA is an acceptable alternative to tumor biopsies for mutation identification in metastatic breast cancer. A secondary aim is to determine whether there is a clinical benefit from therapeutic targeting of mutations identified in ctDNA. However, open questions remain regarding the contribution of tumor deposits to ctDNA. This contribution could be dependent on the metastatic organ, cancer proliferation rate, histology, size of the metastasis, and response to therapy. For instance, it has been demonstrated that brain metastases do not or poorly contribute to ctDNA in plasma and that ctDNA from cerebrospinal fluid would offer a better representation of the genomic alterations of brain metastases (103).

It is also important to perform comprehensive assessment of the various pathways activated within an individual cancer rather than seeking a single mutation in isolation (104). For example, only a fraction of patients receiving selective PI3Kα inhibitors respond, and nonresponders tend to demonstrate an increase in ER-dependent transcription that can be overcome in some cases by combining the PI3Kα inhibitor with an anti-estrogen such as tamoxifen or fulvestrant (105).

Although the genomic revolution has profoundly broadened our understanding of the number and nature of somatic alterations within breast cancer genomes, the clinical benefit of these discoveries remains modest. The feasibility of DNA sequencing to direct the management of metastatic breast cancer through a personalized cancer medicine approach has been examined in two prospective molecular screening studies: SAFIR01/UNICANCER and MOSCATO, in the years 2011 to 2012 (106, 107). The two studies included 423 and 129 patients, and identified potentially targetable alterations in 40% and 46% of patients with the ability to personalize therapy in 13% and 23% of cases, respectively. Of the patients who received targeted therapy, 9% and 20% had a partial response and 21% and 56% had stable disease, respectively. Therefore, 3% and 15% of patients originally screened in these two studies derived clinical benefit from a targeted therapy, but in the absence of a control arm, we cannot infer if the benefit exceeded that of standard of care. It is worth noting that the SHIVA trial, which randomized 293 patients with potentially targetable alterations to molecularly targeted versus standard therapy and found no improvement in outcome but higher toxicity, involved last-generation targeted agents (108). The effect of a personalized cancer medicine approach on survival or, indeed, any other outcome remains to be determined within randomized trials using validated biomarkers and modern targeted drugs.

Although this review has focused on genomic sequence data, it is clear that tackling breast cancer, a complex set of diseases, will require integrated assessment of a wide range of data types, including pathologic characterization (such as histologic subtyping and assessment of the cellular composition), transcriptomics, epigenetics, and proteomics, in addition to genomics (6, 10, 109).

Most large driver discovery studies published to date include primary, treatment-naïve cancers (Supplementary Table S1), and around 80% of these are likely to have been cured using existing treatment strategies. Moving forward, focus should be placed on understanding the mechanisms of both primary and acquired treatment resistance and the genomics of metastasis that is almost universally fatal. This should include using genomics to identify primary breast cancers most at risk of relapse and to characterize disseminated tumor cells that are capable of metastasis formation.

We believe that these goals can be achieved through the joint results from many large-scale, prospective initiatives that aim to better characterize metastases and link molecular findings with meticulously annotated clinical and treatment information. These initiatives include the AURORA program, the Metastatic Breast Cancer Project (https://www.mbcproject.org/users/new), the UK plasmaMATCH (ISRCTN16945804), as well as the various rapid autopsy programs (99, 110, 111). However, given the extensive intertumoral heterogeneity of breast cancer, we must not dismiss the power of “n of 1” studies and small case series that could deliver key insights into ultrarare alterations and associated treatment resistance and response.

A large amount of genomic data from several thousand breast cancers has already been generated, and further value can almost certainly be extracted from this. For example, a combined analysis, although challenging on account of the heterogeneity of the datasets, would have much higher power to detect rare mutations than any individual study to date. Supporting such a collaborative approach is the AACR Project Genomics Evidence Neoplasia Information Exchange (GENIE; http://www.aacr.org/Research/Research/Pages/aacr-project-genie.aspx#.WH6TMnCFNOC), an international data-sharing project that aims to collate genomic and clinical data from tens of thousands of patients in multiple institutions.

Great enthusiasm accompanied the initial years of the genomic revolution as a flood of novel insights into the breast cancer genome were presented in quick succession. We now have a clearer idea not only of the potential clinical applications of genomics but also of the scale and nature of the challenges that must be surmounted to bring maximum benefit to breast cancer patients. It is clear that delivering personalized breast cancer treatments to all patients in the future will only be possible if we focus on collaborative and integrative approaches now.

No potential conflicts of interest were disclosed.

This work was supported by a grant from Les Amis de Bordet (to C. Desmedt).

1.
Weigelt
B
,
Pusztai
L
,
Ashworth
A
,
Reis-Filho
JS
. 
Challenges translating breast cancer gene signatures into the clinic
.
Nat Rev Clin Oncol
2011
;
9
:
58
64
.
2.
Gingras
I
,
Desmedt
C
,
Ignatiadis
M
,
Sotiriou
C
. 
CCR 20th Anniversary Commentary: gene-expression signature in breast cancer–where did it start and where are we now?
Clin Cancer Res
2015
;
21
:
4743
6
.
3.
Shah
SP
,
Roth
A
,
Goya
R
,
Oloumi
A
,
Ha
G
,
Zhao
Y
, et al
The clonal and mutational evolution spectrum of primary triple-negative breast cancers
.
Nature
2012
;
486
:
395
9
.
4.
Stephens
PJ
,
Tarpey
PS
,
Davies
H
,
Van Loo
P
,
Greenman
C
,
Wedge
DC
, et al
The landscape of cancer genes and mutational processes in breast cancer
.
Nature
2012
;
486
:
400
4
.
5.
Banerji
S
,
Cibulskis
K
,
Rangel-Escareno
C
,
Brown
KK
,
Carter
SL
,
Frederick
AM
, et al
Sequence analysis of mutations and translocations across breast cancer subtypes
.
Nature
2012
;
486
:
405
9
.
6.
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
.
7.
Ellis
MJ
,
Ding
L
,
Shen
D
,
Luo
J
,
Suman
VJ
,
Wallis
JW
, et al
Whole-genome analysis informs breast cancer response to aromatase inhibition
.
Nature
2012
;
486
:
353
60
.
8.
The Cancer Genome Atlas Network
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
9.
Nik-Zainal
S
,
Davies
H
,
Staaf
J
,
Ramakrishna
M
,
Glodzik
D
,
Zou
X
, et al
Landscape of somatic mutations in 560 breast cancer whole-genome sequences
.
Nature
2016
;
534
:
47
54
.
10.
Pereira
B
,
Chin
SF
,
Rueda
OM
,
Vollan
HK
,
Provenzano
E
,
Bardwell
HA
, et al
The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes
.
Nat Commun
2016
;
7
:
11479
.
11.
Ross
JS
,
Badve
S
,
Wang
K
,
Sheehan
CE
,
Boguniewicz
AB
,
Otto
GA
, et al
Genomic profiling of advanced-stage, metaplastic breast carcinoma by next-generation sequencing reveals frequent, targetable genomic abnormalities and potential new treatment options
.
Arch Pathol Lab Med
2015
;
139
:
642
9
.
12.
Vranic
S
,
Marchio
C
,
Castellano
I
,
Botta
C
,
Scalzo
MS
,
Bender
RP
, et al
Immunohistochemical and molecular profiling of histologically defined apocrine carcinomas of the breast
.
Hum Pathol
2015
;
46
:
1350
9
.
13.
Michaut
M
,
Chin
SF
,
Majewski
I
,
Severson
TM
,
Bismeijer
T
,
de Koning
L
, et al
Integration of genomic, transcriptomic and proteomic data identifies two biologically distinct subtypes of invasive lobular breast cancer
.
Sci Rep
2016
;
6
:
18517
.
14.
Piscuoglio
S
,
Ng
CK
,
Murray
MP
,
Guerini-Rocco
E
,
Martelotto
LG
,
Geyer
FC
, et al
The genomic landscape of male breast cancers
.
Clin Cancer Res
2016
;
22
:
4045
56
.
15.
Weisman
PS
,
Ng
CK
,
Brogi
E
,
Eisenberg
RE
,
Won
HH
,
Piscuoglio
S
, et al
Genetic alterations of triple negative breast cancer by targeted next-generation sequencing and correlation with tumor morphology
.
Mod Pathol
2016
;
29
:
476
88
.
16.
Marchio
C
,
Geyer
FC
,
Ng
CK
,
Piscuoglio
S
,
De Filippo
MR
,
Cupo
M
, et al
The genetic landscape of breast carcinomas with neuroendocrine differentiation
.
J Pathol
2016
;
241
:
405
19
.
17.
Desmedt
C
,
Zoppoli
G
,
Gundem
G
,
Pruneri
G
,
Larsimont
D
,
Fornili
M
, et al
Genomic characterization of primary invasive lobular breast cancer
.
J Clin Oncol
2016
;
34
:
1872
81
.
18.
Dieci
MV
,
Smutna
V
,
Scott
V
,
Yin
G
,
Xu
R
,
Vielh
P
, et al
Whole exome sequencing of rare aggressive breast cancer histologies
.
Breast Cancer Res Treat
2016
;
156
:
21
32
.
19.
Lawrence
MS
,
Stojanov
P
,
Polak
P
,
Kryukov
GV
,
Cibulskis
K
,
Sivachenko
A
, et al
Mutational heterogeneity in cancer and the search for new cancer-associated genes
.
Nature
2013
;
499
:
214
8
.
20.
Gonzalez-Perez
A
,
Perez-Llamas
C
,
Deu-Pons
J
,
Tamborero
D
,
Schroeder
MP
,
Jene-Sanz
A
, et al
IntOGen-mutations identifies cancer drivers across tumor types
.
Nat Methods
2013
;
10
:
1081
2
.
21.
Reimand
J
,
Bader
GD
. 
Systematic analysis of somatic mutations in phosphorylation signaling predicts novel cancer drivers
.
Mol Syst Biol
2013
;
9
:
637
.
22.
Dees
ND
,
Zhang
Q
,
Kandoth
C
,
Wendl
MC
,
Schierding
W
,
Koboldt
DC
, et al
MuSiC: identifying mutational significance in cancer genomes
.
Genome Res
2012
;
22
:
1589
98
.
23.
Martincorena
I
,
Roshan
A
,
Gerstung
M
,
Ellis
P
,
Van Loo
P
,
McLaren
S
, et al
Tumor evolution. High burden and pervasive positive selection of somatic mutations in normal human skin
.
Science
2015
;
348
:
880
6
.
24.
Meyer
MJ
,
Lapcevic
R
,
Romero
AE
,
Yoon
M
,
Das
J
,
Beltran
JF
, et al
mutation3D: cancer gene prediction through atomic clustering of coding variants in the structural proteome
.
Hum Mutat
2016
;
37
:
447
56
.
25.
Zhao
J
,
Zhang
S
,
Wu
LY
,
Zhang
XS
. 
Efficient methods for identifying mutated driver pathways in cancer
.
Bioinformatics
2012
;
28
:
2940
7
.
26.
Ciriello
G
,
Cerami
E
,
Sander
C
,
Schultz
N
. 
Mutual exclusivity analysis identifies oncogenic network modules
.
Genome Res
2012
;
22
:
398
406
.
27.
Leiserson
MD
,
Blokh
D
,
Sharan
R
,
Raphael
BJ
. 
Simultaneous identification of multiple driver pathways in cancer
.
PLoS Comput Biol
2013
;
9
:
e1003054
.
28.
Hou
JP
,
Ma
J
. 
DawnRank: discovering personalized driver genes in cancer
.
Genome Med
2014
;
6
:
56
.
29.
Lawrence
MS
,
Stojanov
P
,
Mermel
CH
,
Robinson
JT
,
Garraway
LA
,
Golub
TR
, et al
Discovery and saturation analysis of cancer genes across 21 tumour types
.
Nature
2014
;
505
:
495
501
.
30.
Lefebvre
C
,
Bachelot
T
,
Filleron
T
,
Pedrero
M
,
Campone
M
,
Soria
JC
, et al
Mutational profile of metastatic breast cancers: a retrospective analysis
.
PLoS Med
2016
;
13
:
e1002201
.
31.
Kehr
EL
,
Jorns
JM
,
Ang
D
,
Warrick
A
,
Neff
T
,
Degnin
M
, et al
Mucinous breast carcinomas lack PIK3CA and AKT1 mutations
.
Hum Pathol
2012
;
43
:
2207
12
.
32.
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
.
33.
Ng
CKY
,
Piscuoglio
S
,
Geyer
FC
,
Burke
KA
,
Pareja
F
,
Eberle
CA
, et al
The landscape of somatic genetic alterations in metaplastic breast carcinomas
.
Clin Cancer Res
2017
Feb 2. [Epub ahead of print].
34.
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
.
35.
Miller
CA
,
Gindin
Y
,
Lu
C
,
Griffith
OL
,
Griffith
M
,
Shen
D
, et al
Aromatase inhibition remodels the clonal architecture of estrogen-receptor-positive breast cancers
.
Nat Commun
2016
;
7
:
12498
.
36.
Yates
LR
,
Gerstung
M
,
Knappskog
S
,
Desmedt
C
,
Gundem
G
,
Van Loo
P
, et al
Subclonal diversification of primary breast cancer revealed by multiregion sequencing
.
Nat Med
2015
;
21
:
751
9
.
37.
Balko
JM
,
Giltnane
JM
,
Wang
K
,
Schwarz
LJ
,
Young
CD
,
Cook
RS
, et al
Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets
.
Cancer Discov
2014
;
4
:
232
45
.
38.
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
.
39.
Martelotto
LG
,
Baslan
T
,
Kendall
J
,
Geyer
FC
,
Burke
KA
,
Spraggon
L
, et al
Whole-genome single-cell copy number profiling from formalin-fixed paraffin-embedded samples
.
Nat Med
2017
;
23
:
376
85
.
40.
Juric
D
,
Castel
P
,
Griffith
M
,
Griffith
OL
,
Won
HH
,
Ellis
H
, et al
Convergent loss of PTEN leads to clinical resistance to a PI(3)Kalpha inhibitor
.
Nature
2015
;
518
:
240
4
.
41.
Goodwin
S
,
McPherson
JD
,
McCombie
WR
. 
Coming of age: ten years of next-generation sequencing technologies
.
Nat Rev Genet
2016
;
17
:
333
51
.
42.
Navin
N
,
Kendall
J
,
Troge
J
,
Andrews
P
,
Rodgers
L
,
McIndoo
J
, et al
Tumour evolution inferred by single-cell sequencing
.
Nature
2011
;
472
:
90
4
.
43.
Niu
B
,
Scott
AD
,
Sengupta
S
,
Bailey
MH
,
Batra
P
,
Ning
J
, et al
Protein-structure-guided discovery of functional mutations across 19 cancer types
.
Nat Genet
2016
;
48
:
827
37
.
44.
Katigbak
A
,
Cencic
R
,
Robert
F
,
Senecha
P
,
Scuoppo
C
,
Pelletier
J
. 
A CRISPR/Cas9 functional screen identifies rare tumor suppressors
.
Sci Rep
2016
;
6
:
38968
.
45.
Janku
F
,
Wheler
JJ
,
Westin
SN
,
Moulder
SL
,
Naing
A
,
Tsimberidou
AM
, et al
PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations
.
J Clin Oncol
2012
;
30
:
777
82
.
46.
Dawood
S
,
Broglio
K
,
Buzdar
AU
,
Hortobagyi
GN
,
Giordano
SH
. 
Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review
.
J Clin Oncol
2010
;
28
:
92
8
.
47.
Patel
JH
,
Loboda
AP
,
Showe
MK
,
Showe
LC
,
McMahon
SB
. 
Analysis of genomic targets reveals complex functions of MYC
.
Nat Rev Cancer
2004
;
4
:
562
8
.
48.
Prochownik
EV
,
Vogt
PK
. 
Therapeutic targeting of Myc
.
Genes Cancer
2010
;
1
:
650
9
.
49.
Muller
PA
,
Vousden
KH
. 
Mutant p53 in cancer: new functions and therapeutic opportunities
.
Cancer Cell
2014
;
25
:
304
17
.
50.
Ellis
MJ
,
Perou
CM
. 
The genomic landscape of breast cancer as a therapeutic roadmap
.
Cancer Discov
2013
;
3
:
27
34
.
51.
Posternak
V
,
Cole
MD
. 
Strategically targeting MYC in cancer
.
F1000Res
2016
;
5
.
52.
Horiuchi
D
,
Camarda
R
,
Zhou
AY
,
Yau
C
,
Momcilovic
O
,
Balakrishnan
S
, et al
PIM1 kinase inhibition as a targeted therapy against triple-negative breast tumors with elevated MYC expression
.
Nat Med
2016
;
22
:
1321
9
.
53.
Baselga
J
,
Campone
M
,
Piccart
M
,
Burris
HA
 III
,
Rugo
HS
,
Sahmoud
T
, et al
Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer
.
N Engl J Med
2012
;
366
:
520
9
.
54.
Turner
NC
,
Ro
J
,
Andre
F
,
Loi
S
,
Verma
S
,
Iwata
H
, et al
Palbociclib in hormone-receptor-positive advanced breast cancer
.
N Engl J Med
2015
;
373
:
209
19
.
55.
Finn
RS
,
Crown
JP
,
Ettl
J
,
Schmidt
M
,
Bondarenko
IM
,
Lang
I
, et al
Efficacy and safety of palbociclib in combination with letrozole as first-line treatment of ER-positive, HER2-negative, advanced breast cancer: expanded analyses of subgroups from the randomized pivotal trial PALOMA-1/TRIO-18
.
Breast Cancer Res
2016
;
18
:
67
.
56.
Hortobagyi
GN
,
Chen
D
,
Piccart
M
,
Rugo
HS
,
Burris
HA
 III
,
Pritchard
KI
, et al
Correlative analysis of genetic alterations and everolimus benefit in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: results from BOLERO-2
.
J Clin Oncol
2016
;
34
:
419
26
.
57.
Arnedos
M
,
Vicier
C
,
Loi
S
,
Lefebvre
C
,
Michiels
S
,
Bonnefoi
H
, et al
Precision medicine for metastatic breast cancer–limitations and solutions
.
Nat Rev Clin Oncol
2015
;
12
:
693
704
.
58.
Hyman
DM
,
Puzanov
I
,
Subbiah
V
,
Faris
JE
,
Chau
I
,
Blay
JY
, et al
Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations
.
N Engl J Med
2015
;
373
:
726
36
.
59.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Aparicio
SA
,
Behjati
S
,
Biankin
AV
, et al
Signatures of mutational processes in human cancer
.
Nature
2013
;
500
:
415
21
.
60.
Nik-Zainal
S
,
Alexandrov
LB
,
Wedge
DC
,
Van Loo
P
,
Greenman
CD
,
Raine
K
, et al
Mutational processes molding the genomes of 21 breast cancers
.
Cell
2012
;
149
:
979
93
.
61.
Nik-Zainal
S
,
Morganella
S
. 
Mutational signatures in breast cancer: the problem at the DNA level
.
Clin Cancer Res
2017
;
23
:
2617
29
.
62.
Helleday
T
,
Eshtad
S
,
Nik-Zainal
S
. 
Mechanisms underlying mutational signatures in human cancers
.
Nat Rev Genet
2014
;
15
:
585
98
.
63.
Telli
ML
,
Timms
KM
,
Reid
J
,
Hennessy
B
,
Mills
GB
,
Jensen
KC
, et al
Homologous recombination deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer
.
Clin Cancer Res
2016
;
22
:
3764
73
.
64.
Watkins
JA
,
Irshad
S
,
Grigoriadis
A
,
Tutt
AN
. 
Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers
.
Breast Cancer Res
2014
;
16
:
211
.
65.
Abkevich
V
,
Timms
KM
,
Hennessy
BT
,
Potter
J
,
Carey
MS
,
Meyer
LA
, et al
Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer
.
Br J Cancer
2012
;
107
:
1776
82
.
66.
Birkbak
NJ
,
Wang
ZC
,
Kim
JY
,
Eklund
AC
,
Li
Q
,
Tian
R
, et al
Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents
.
Cancer Discov
2012
;
2
:
366
75
.
67.
Popova
T
,
Manie
E
,
Rieunier
G
,
Caux-Moncoutier
V
,
Tirapo
C
,
Dubois
T
, et al
Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation
.
Cancer Res
2012
;
72
:
5454
62
.
68.
Glodzik
D
,
Morganella
S
,
Davies
H
,
Simpson
PT
,
Li
Y
,
Zou
X
, et al
A somatic-mutational process recurrently duplicates germline susceptibility loci and tissue-specific super-enhancers in breast cancers
.
Nat Genet
2017
;
49
:
341
8
.
69.
Antoniou
AC
,
Casadei
S
,
Heikkinen
T
,
Barrowdale
D
,
Pylkas
K
,
Roberts
J
, et al
Breast-cancer risk in families with mutations in PALB2
.
N Engl J Med
2014
;
371
:
497
506
.
70.
Davies
H
,
Glodzik
D
,
Morganella
S
,
Yates
LR
,
Staaf
J
,
Zou
X
, et al
HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures
.
Nat Med
2017
;
23
:
517
25
.
71.
Natrajan
R
,
Mackay
A
,
Lambros
MB
,
Weigelt
B
,
Wilkerson
PM
,
Manie
E
, et al
A whole-genome massively parallel sequencing analysis of BRCA1 mutant oestrogen receptor-negative and -positive breast cancers
.
J Pathol
2012
;
227
:
29
41
.
72.
The Cancer Genome Atlas Network
. 
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
73.
Silwal-Pandit
L
,
Vollan
HK
,
Chin
SF
,
Rueda
OM
,
McKinney
S
,
Osako
T
, et al
TP53 mutation spectrum in breast cancer is subtype specific and has distinct prognostic relevance
.
Clin Cancer Res
2014
;
20
:
3569
80
.
74.
Lakhani
SR
,
International Agency for Research on Cancer
,
World Health Organization
.
WHO classification of tumours of the breast
.
Lyon, France
:
International Agency for Research on Cancer
; 
2012
.
75.
Theodorou
V
,
Stark
R
,
Menon
S
,
Carroll
JS
. 
GATA3 acts upstream of FOXA1 in mediating ESR1 binding by shaping enhancer accessibility
.
Genome Res
2013
;
23
:
12
22
.
76.
Ross
JS
,
Gay
LM
,
Nozad
S
,
Wang
K
,
Ali
SM
,
Boguniewicz
A
, et al
Clinically advanced and metastatic pure mucinous carcinoma of the breast: a comprehensive genomic profiling study
.
Breast Cancer Res Treat
2016
;
155
:
405
13
.
77.
Shah
SP
,
Morin
RD
,
Khattra
J
,
Prentice
L
,
Pugh
T
,
Burleigh
A
, et al
Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution
.
Nature
2009
;
461
:
809
13
.
78.
Ding
L
,
Ellis
MJ
,
Li
S
,
Larson
DE
,
Chen
K
,
Wallis
JW
, et al
Genome remodelling in a basal-like breast cancer metastasis and xenograft
.
Nature
2010
;
464
:
999
1005
.
79.
Meric-Bernstam
F
,
Frampton
GM
,
Ferrer-Lozano
J
,
Yelensky
R
,
Perez-Fidalgo
JA
,
Wang
Y
, et al
Concordance of genomic alterations between primary and recurrent breast cancer
.
Mol Cancer Ther
2014
;
13
:
1382
9
.
80.
Hoadley
KA
,
Siegel
MB
,
Kanchi
KL
,
Miller
CA
,
Ding
L
,
Zhao
W
, et al
Tumor evolution in two patients with basal-like breast cancer: a retrospective genomics study of multiple metastases
.
PLoS Med
2016
;
13
:
e1002174
.
81.
Demeulemeester
J
,
Kumar
P
,
Moller
EK
,
Nord
S
,
Wedge
DC
,
Peterson
A
, et al
Tracing the origin of disseminated tumor cells in breast cancer using single-cell sequencing
.
Genome Biol
2016
;
17
:
250
.
82.
Brastianos
PK
,
Carter
SL
,
Santagata
S
,
Cahill
DP
,
Taylor-Weiner
A
,
Jones
RT
, et al
Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets
.
Cancer Discov
2015
;
5
:
1164
77
.
83.
Savas
P
,
Teo
ZL
,
Lefevre
C
,
Flensburg
C
,
Caramia
F
,
Alsop
K
, et al
The subclonal architecture of metastatic breast cancer: results from a prospective community-based rapid autopsy program "CASCADE"
.
PLoS Med
2016
;
13
:
e1002204
.
84.
Brown
D
,
Smeets
D
,
Székely
B
,
Larsimont
D
,
Szász
AM
,
Adnet
PY
, et al
Phylogenetic analysis of metastatic progression in breast cancer using somatic mutations and copy number aberrations
.
Nat Commun
2017
;
8
:
14944
.
85.
Achkar
T
,
Wilson
J
,
Simon
J
,
Rosenzweig
M
,
Puhalla
S
. 
Metastatic breast cancer patients: attitudes toward tissue donation for rapid autopsy
.
Breast Cancer Res Treat
2016
;
155
:
159
64
.
86.
Yates
L
. 
Abstract 1804 The driver landscape of breast cancer metastasis and relapse
.
Eur J Cancer
2015
;
51
:
S266
.
87.
Li
S
,
Shen
D
,
Shao
J
,
Crowder
R
,
Liu
W
,
Prat
A
, et al
Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts
.
Cell Rep
2013
;
4
:
1116
30
.
88.
Wang
T
,
Liu
JH
,
Zhang
J
,
Wang
L
,
Chen
C
,
Dai
PG
. 
A multiplex allele-specific real-time PCR assay for screening of ESR1 mutations in metastatic breast cancer
.
Exp Mol Pathol
2015
;
98
:
152
7
.
89.
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
.
90.
Murtaza
M
,
Dawson
SJ
,
Pogrebniak
K
,
Rueda
OM
,
Provenzano
E
,
Grant
J
, et al
Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer
.
Nat Commun
2015
;
6
:
8760
.
91.
Dawson
SJ
,
Tsui
DW
,
Murtaza
M
,
Biggs
H
,
Rueda
OM
,
Chin
SF
, et al
Analysis of circulating tumor DNA to monitor metastatic breast cancer
.
N Engl J Med
2013
;
368
:
1199
209
.
92.
De Mattos-Arruda
L
,
Weigelt
B
,
Cortes
J
,
Won
HH
,
Ng
CK
,
Nuciforo
P
, et al
Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle
.
Ann Oncol
2014
;
25
:
1729
35
.
93.
Garcia-Murillas
I
,
Schiavon
G
,
Weigelt
B
,
Ng
C
,
Hrebien
S
,
Cutts
RJ
, et al
Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer
.
Sci Transl Med
2015
;
7
:
302ra133
.
94.
Gellert
P
,
Segal
CV
,
Gao
Q
,
Lopez-Knowles
E
,
Martin
LA
,
Dodson
A
, et al
Impact of mutational profiles on response of primary oestrogen receptor-positive breast cancers to oestrogen deprivation
.
Nat Commun
2016
;
7
:
13294
.
95.
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
.
96.
Schiavon
G
,
Hrebien
S
,
Garcia-Murillas
I
,
Cutts
RJ
,
Pearson
A
,
Tarazona
N
, et al
Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer
.
Sci Transl Med
2015
;
7
:
313ra182
.
97.
Spoerke
JM
,
Gendreau
S
,
Walter
K
,
Qiu
J
,
Wilson
TR
,
Savage
H
, et al
Heterogeneity and clinical significance of ESR1 mutations in ER-positive metastatic breast cancer patients receiving fulvestrant
.
Nat Commun
2016
;
7
:
11579
.
98.
Fribbens
C
,
O'Leary
B
,
Kilburn
L
,
Hrebien
S
,
Garcia-Murillas
I
,
Beaney
M
, et al
Plasma ESR1 mutations and the treatment of estrogen receptor-positive advanced breast cancer
.
J Clin Oncol
2016
;
34
:
2961
8
.
99.
Cohen
O
Oh
C
,
Waks
A
,
Oliver
N
,
Helvie
K
,
Marini
L
, et al
Whole exome and transcriptome sequencing of resistant ER+ metastatic breast cancer [abstract]
.
In: Proceedings of the 39th Annual San Antonio Breast Cancer Symposium; 2016 Dec 6–10
;
San Antonio, TX
.
SABCS
; 
2016
.
Abstract nr S1-01
.
100.
Herrera-Abreu
MT
,
Palafox
M
,
Asghar
U
,
Rivas
MA
,
Cutts
RJ
,
Garcia-Murillas
I
, et al
Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer
.
Cancer Res
2016
;
76
:
2301
13
.
101.
Yang
C
,
Li
Z
,
Bhatt
T
,
Dickler
M
,
Giri
D
,
Scaltriti
M
, et al
Acquired CDK6 amplification promotes breast cancer resistance to CDK4/6 inhibitors and loss of ER signaling and dependence
.
Oncogene
2017
;
36
:
2255
64
.
102.
Chandarlapaty
S
,
Chen
D
,
He
W
,
Sung
P
,
Samoila
A
,
You
D
, et al
Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial
.
JAMA Oncol
2016
;
2
:
1310
5
.
103.
De Mattos-Arruda
L
,
Mayor
R
,
Ng
CK
,
Weigelt
B
,
Martinez-Ricarte
F
,
Torrejon
D
, et al
Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma
.
Nat Commun
2015
;
6
:
8839
.
104.
Saini
KS
,
Loi
S
,
de Azambuja
E
,
Metzger-Filho
O
,
Saini
ML
,
Ignatiadis
M
, et al
Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer
.
Cancer Treat Rev
2013
;
39
:
935
46
.
105.
Bosch
A
,
Li
Z
,
Bergamaschi
A
,
Ellis
H
,
Toska
E
,
Prat
A
, et al
PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor-positive breast cancer
.
Sci Transl Med
2015
;
7
:
283ra51
.
106.
Andre
F
,
Bachelot
T
,
Commo
F
,
Campone
M
,
Arnedos
M
,
Dieras
V
, et al
Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER)
.
Lancet Oncol
2014
;
15
:
267
74
.
107.
Hollebecque
A
,
Massard
C
,
De Baere
T
,
Auger
N
,
Lacroix
L
,
Koubi-Pick
V
, et al
Molecular screening for cancer treatment optimization (MOSCATO 01): a prospective molecular triage trial-interim results
.
J Clin Oncol
31
, 
2013
(
suppl; abstr 2512
).
108.
Le Tourneau
C
,
Delord
JP
,
Gonçalves
A
,
Gavoille
C
,
Dubot
C
,
Isambert
N
, et al
Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial
.
Lancet Oncol
2015
;
16
:
1324
34
.
109.
Klevebring
D
,
Lindberg
J
,
Rockberg
J
,
Hilliges
C
,
Hall
P
,
Sandberg
M
, et al
Exome sequencing of contralateral breast cancer identifies metastatic disease
.
Breast Cancer Res Treat
2015
;
151
:
319
24
.
110.
Zardavas
D
,
Irrthum
A
,
Swanton
C
,
Piccart
M
. 
Clinical management of breast cancer heterogeneity
.
Nat Rev Clin Oncol
2015
;
12
:
381
94
.
111.
Alsop
K
,
Thorne
H
,
Sandhu
S
,
Hamilton
A
,
Mintoff
C
,
Christie
E
, et al
A community-based model of rapid autopsy in end-stage cancer patients
.
Nat Biotechnol
2016
;
34
:
1010
4
.