Abstract

Advances in DNA and RNA sequencing revealed substantially greater genomic complexity in breast cancer than simple models of a few driver mutations would suggest. Only very few, recurrent mutations or copy-number variations in cancer-causing genes have been identified. The two most common alterations in breast cancer are TP53 (affecting the majority of triple-negative breast cancers) and PIK3CA (affecting almost half of estrogen receptor-positive cancers) mutations, followed by a long tail of individually rare mutations affecting <1%–20% of cases. Each cancer harbors from a few dozen to a few hundred potentially high-functional impact somatic variants, along with a much larger number of potentially high-functional impact germline variants. It is likely that it is the combined effect of all genomic variations that drives the clinical behavior of a given cancer. Furthermore, entirely new classes of oncogenic events are being discovered in the noncoding areas of the genome and in noncoding RNA species driven by errors in RNA editing. In light of this complexity, it is not unexpected that, with the exception of HER2 amplification, no robust molecular predictors of benefit from targeted therapies have been identified. In this review, we summarize the current genomic portrait of breast cancer, focusing on genetic aberrations that are actively being targeted with investigational drugs.

Implications for Practice:

Next-generation sequencing is now widely available in the clinic, but interpretation of the results is challenging, and its impact on treatment selection is often limited. This work provides an overview of frequently encountered molecular abnormalities in breast cancer and discusses their potential therapeutic implications. This review emphasizes the importance of administering investigational targeted therapies, or off-label use of approved targeted drugs, in the context of a formal clinical trial or registry programs to facilitate learning about the clinical utility of tumor target profiling.

Introduction

Breast cancer represents a complex and heterogeneous disease, encompassing several molecularly and clinically distinct entities that can be classified according to gene expression profiles and clinicopathological features [1, 2]. Hereditary disease accounts only for a small percentage of all breast tumors, whereas the majority of breast cancers are sporadic, resulting from the accumulation of acquired somatic alterations [3]. Recent advances in sequencing-based technologies have increased our understanding of genetic aberrations and dysregulated oncogenic pathways, involving growth signaling, stress response, metabolism, and cell-to-cell communication, which affect breast cancer development and progression. These somatic alterations, together with the host response to cancer, determine the clinical course of the disease. A large number of often individually rare, but potentially functionally important, molecular alterations are encountered in unique combinations in every cancer. How to best exploit the genomic anatomy of cancer for therapeutic advantage remains an important unanswered challenge [47]. In this review, we provide a portrait of currently known genomic alterations and related pathways in breast cancer.

Materials and Methods

We conducted a review of the literature using PubMed, Web of Science, and Embase databases from 2000 to May 2016 using the search terms “breast cancer,” “genetic mutations,” “genetic alterations,” “biomarkers,” “response to therapy,” “tumor heterogeneity,” and “targeted therapies.” Additional studies were identified through the references listed in review publications. Recent emerging clinical findings presented at scientific meetings in the form of abstracts were also reviewed and incorporated. We note that the reported frequency for gene-level mutations depends on platform, sequencing depth, sample size, and definition of actionability, which vary from study to study. In this review, we use the frequency numbers from the largest studies that we could identify. We also searched clinicaltrials.gov to identify relevant ongoing phase II–III clinical trials evaluating targeted agents in patients with breast cancer.

The Genomic Landscape of Breast Cancers

The genetic basis of susceptibility to breast cancer is related to multiple germline variations at different loci. These alleles have broadly variable frequencies and confer different levels of risk, leading to a classification as high-, moderate-, and low-penetrance alleles (Fig. 1). Even though the major rare high-penetrance genes, BRCA1 and BRCA2, which are involved in DNA repair processes, account for approximately 50% and 30% of familial breast cancers, respectively, further efforts are needed to unravel the impact of variants of unknown pathological significance or other susceptibility genes [3].

Major cancer susceptibility loci identified in breast cancer. Each breast cancer susceptibility locus is mapped on their chromosomal location. The risk conferred by each specific allele is indicated by color. Chromosomes are not drawn to scale.
Figure 1

Major cancer susceptibility loci identified in breast cancer. Each breast cancer susceptibility locus is mapped on their chromosomal location. The risk conferred by each specific allele is indicated by color. Chromosomes are not drawn to scale.

Sporadic breast cancers develop through the acquisition of novel malignant traits that confer a selective advantage to tumor cells. Although the accumulation of such genomic alterations is a requisite for the expansion and progression of a transformed cell population, recent insights that have emerged from comprehensive genomic studies have revealed additional heterogeneity in the genomic landscape of breast cancer [812]. The diversity of genetic pattern among breast tumors may explain the wide differences in biological features, clinical behavior, and response to therapies. The analysis of primary breast tumors has demonstrated that missense mutations are more common in estrogen receptor (ER)-positive/luminal and human epidermal growth factor receptor 2 (HER2)-positive subtypes, whereas triple-negative breast cancers (TNBCs) are enriched for nonsense, frameshift, and complex mutations (Fig. 2A) [13]. Similarly, a higher rate of genomic rearrangements was found in HER2-positive breast cancers and TNBCs [14]. Breast cancer subtypes also differ regarding the pattern of copy-number alterations (CNAs) (Table 1) [11]. Novel data on genomic spectra, signaling pathways, and gene expression profiles are leading to the refinement of the molecular classification of breast cancer (Fig. 2B; Table 1) [812, 1418].

Mutation spectrum in breast cancer subtypes. (A): Percentage of somatic mutations types in breast cancer overall and molecular subtypes according to COSMIC database. The ER-positive/luminal group includes luminal A and B subtypes. (B): Frequencies of the most commonly mutated cancer-related genes across breast cancer subtypes, according to the Cancer Genome Atlas [9].
Figure 2

Mutation spectrum in breast cancer subtypes. (A): Percentage of somatic mutations types in breast cancer overall and molecular subtypes according to COSMIC database. The ER-positive/luminal group includes luminal A and B subtypes. (B): Frequencies of the most commonly mutated cancer-related genes across breast cancer subtypes, according to the Cancer Genome Atlas [9].

Abbreviations: ER, estrogen receptor; HER2, human epidermal growth factor receptor 2.

Table 1

Most common gene mutations and copy number alterations in different molecular subtypes of primary breast cancer

Table 1

Most common gene mutations and copy number alterations in different molecular subtypes of primary breast cancer

Estrogen Receptor-Positive/Luminal Breast Cancers

ER-positive/luminal breast cancers have the highest number of recurrent mutations (Fig. 2B; Table 1) [9, 14]. In luminal A cancers, the most frequently mutated gene is PIK3CA (45%), followed by GATA3 (14%), MAP3K1 (13%), TP53 (12%), CDH1 (9%), MLL3 (8%), MAP2K4 (7%), NCOR1 (5%), and RUNX1 (5%) [9]. The PIK3CA and GATA3 mutations appear mutually exclusive, with the much rarer mutations in AKT1 (4%) and FOXA1 (2%), respectively [9, 19]. Luminal B tumors have a lower frequency of PIK3CA mutations (29%) and a slightly higher frequency of GATA3 mutations (15%) (Fig. 2B; Table 1) [9]. Conversely, the frequency of TP53 mutations (29%) is higher in luminal B cancers and is associated with worse outcome [20]. There is also higher frequency of ATM loss and CCND1, CDK4, CDK6, and MDM2 amplifications in luminal B cancers [9]. ER-positive cancers have relatively few recurrent CNAs compared with TNBCs (Table 1) [9, 10, 21, 22]. A common and potentially clinically relevant event in luminal B tumors is the amplification of fibroblast growth factor receptors (FGFRs) [9, 22].

HER2-Positive Breast Cancers

A core set of genomic regions consistently affected by CNAs has been identified in HER2-positive breast cancers (Table 1) [23]. In addition to the common amplification at 17q12 (containing the HER2 oncogene), other recurrent CNAs included a large number of gains, losses, and amplifications [24]. A source of genomic heterogeneity within HER2-positive tumors is dependent on the size of the HER2 amplicon, because multiple genes can be coamplified with HER2 [11]. Beyond the core of the amplicon, which includes at least HER2-C17orf37-GRB7 genes, coamplifications of other genes have been described [24, 25]. Particularly, the amplification of the HER2/TOP2A region has been correlated with the overexpression of several additional genes, such as CASC3, CDC6, RARA, and SMARCE1 [26]. It is worth noting that 20%–40% of HER2-positive cancers are TOP2A-deleted and that alterations in other growth factor receptors have been associated with the HER2 status, including EGFR, FGFR, and HER3 [9, 17, 27].

Triple-Negative Breast Cancers

TNBCs represent a molecularly highly heterogeneous group with substantial transcriptional and DNA copy number variability [28]. These cancers have the highest frequency of TP53 disabling mutations (up to 80%) [9, 20]. Mutations and/or deletions in RB1 (retinoblastoma 1; 20%) and mutations in PIK3CA (9%), MLL3 (5%), and GATA3 (2%), as well as amplification of the CCNE1 gene (9%), occur at relatively low frequencies (Fig. 2B) [9]. Other repeatedly observed, but rather rare, genetic aberrations include mutations in ATR, COL6A3, FBXW7, MYO3A, PARK2, SYNE1/2, and USH2A genes [17, 29]. Despite the overall low prevalence of PIK3CA mutations, the activation of the phosphatidylinositol 3-kinase (PI3K) signaling has been found in specific TNBC subtypes, especially because of loss of PTEN and INPP4B, or PIK3R1 mutations [9, 30]. The genomic landscape of TNBCs is characterized by frequent and numerous DNA copy number losses and gains, suggesting a form of genomic instability (Table 1) [9, 11, 3133]. Gains/amplifications and overexpression of BRAF, CDK6, CDKN2A, CCNE1, E2F, EGFR, FGFR1, FGFR2, IGFR1, KIT, MET, MYC, PDGFRA, and PIK3CA are also often encountered in TNBC [9, 32, 33].

Intratumor Genomic Heterogeneity and Implications for Treatment

Tumor heterogeneity creates a potential framework to explain several clinical features of cancer [3438]. Clinicians have long noticed variable tumor responses in different metastatic sites, suggesting variable treatment sensitivity in different locations. Within-tumor clonal heterogeneity and variable clonal composition of different metastatic lesions provide a simple explanation for this clinical phenomenon and is supported by recent results from tumor sequencing of metastatic sites and matching primary tumors. Clonal selection in response to therapy has also been demonstrated in several different cancer types and provides explanation for the development of treatment resistance over time [37, 3941]. The evolutionary relatedness of primary tumors and multiple metastases from the same patient suggests multiple different ways that metastasis can arise (Fig. 3A) [42, 43]. Most metastatic lesions show large degrees of similarity to the primary tumor, indicating direct seeding from the primary tumor followed by modest genetic drift, whereas other lesions are more similar to one or more other metastatic lesions, suggesting that a metastatic site could give rise to further metastatic lesions (Fig. 3A) [42, 43]. Our understanding of how clinical drug resistance emerges during therapy has also evolved (Fig. 3B; Table 2) [44]. Some forms of resistance emerge through selection of intrinsically resistant subclones present in the cancer as a minority clone from the beginning (Fig. 3B) [12, 69, 7375]. Other forms of resistance arise through de novo mutations that create new clones with drug resistance, not originally present in the cancer (Fig. 3B) [74, 76]. Several studies showed that appearance of new mutations, undetectable in the primary tumor, can arise after neoadjuvant chemotherapy (PTEN and TP53), HER2-targeted therapies (PIK3CA), and during endocrine therapy (ESR1) for metastatic breast cancer [5054, 7779]. Even though the relative proportion of drug-resistant subclones is generally higher in the post-treatment specimens, retreating cancers with drugs on which they previously progressed often continues to produce tumor response. Selectively removing a drug-sensitive subpopulation can lead to a temporary collapse of the entire population, but eventual recovery happens through new subclones stepping in to provide the critical function for the entire population [80]. Considering cancer as a society of neoplastic cells will allow for exploring new therapeutic strategies that could target between-cell communication or interrupt the complementary functions cells provide to each another.

Biological and clinical consequences of tumor heterogeneity. (A): Tumor dissemination may occur at earlier stages of cancer progression, resulting in substantial divergences between primary and metastatic lesions. Alternatively, cell subpopulations in the primary tumor evolve by acquiring multiple genetic aberrations and aggressive phenotypes, ultimately leading to a late-stage metastatic dissemination. (B): Distinct subclones may harbor constitutive alterations that confer different drug sensitivity. Cancer therapy may trigger clonal evolution and increase intratumoral heterogeneity, allowing for the selection of cells with advantageous genetic aberrations. Further evolution of the resistant clone in response to therapy may lead to relapse after initial treatment response.
Figure 3

Biological and clinical consequences of tumor heterogeneity. (A): Tumor dissemination may occur at earlier stages of cancer progression, resulting in substantial divergences between primary and metastatic lesions. Alternatively, cell subpopulations in the primary tumor evolve by acquiring multiple genetic aberrations and aggressive phenotypes, ultimately leading to a late-stage metastatic dissemination. (B): Distinct subclones may harbor constitutive alterations that confer different drug sensitivity. Cancer therapy may trigger clonal evolution and increase intratumoral heterogeneity, allowing for the selection of cells with advantageous genetic aberrations. Further evolution of the resistant clone in response to therapy may lead to relapse after initial treatment response.

Table 2

The most relevant and frequent gene mutations associated with therapy response and biological mechanisms in different breast cancer subtypes

Table 2

The most relevant and frequent gene mutations associated with therapy response and biological mechanisms in different breast cancer subtypes

The evolutionary relatedness of primary tumors and multiple metastases from the same patient suggests multiple different ways that metastasis can arise.

Molecular Predictors of Response to Targeted Therapy in Breast Cancer Subtypes

The fundamental premise behind “precision medicine” and tumor target profiling is that the targetable molecular abnormalities that are detected in a cancer will serve as predictors of treatment response to the appropriate targeted therapy. Several clinical trials suggest that the relationship between benefit from targeted therapies and molecular defects is more complex than hoped for.

For example, the predictive value of the PI3K pathway activation (i.e., PIK3CA mutations and loss of PTEN) in HER2-overexpressing tumors has been analyzed in different neoadjuvant clinical trials, generating variable results, leaving HER2 amplification as the only biomarker associated with anti-HER2 therapies benefit [6468, 81, 82]. It is noteworthy that recent data have demonstrated the presence of HER2 mutations in nonamplified HER2 tumors, potentially advancing the development of novel anti-HER2 drugs in this clinical setting [83].

ER-positive tumors are also highly dependent on the PI3K signaling for cell growth and survival [84]. Everolimus, an mTOR (mammalian target of rapamycin) inhibitor that interrupts the PI3K-mediated signaling, is approved in combination with exemestane for the treatment of advanced postmenopausal ER-positive/HER2-negative breast cancer patients, but results from the BOLERO-2 trial showed that PIK3CA mutations did not predict benefit from everolimus. However, the PI3K assessment was performed on primary tumors rather than metastatic biopsies [85]. Conversely, in the same trial, circulating ESR1 mutations showed a potential predictive efficacy of the addition of everolimus to standard endocrine therapy [54, 85]. The addition of the AKT inhibitor MK-2206 to anastrozole also did not confer significant benefit to ER-positive/HER2-negative patients with PIK3CA mutations [86]. However, preliminary results from the phase III BELLE-2 trial indicated that only patients with circulating PIK3CA mutations benefited from a combination of the PI3K inhibitor BKM120 and fulvestrant [87]. Several larger phase II and III trials with α-specific PIK3CA inhibitors are still under way to further define the predictive function, and therapeutic target value, of PIK3CA mutations [8890].

The CDK4/6 inhibitor PD-0332991 (palbociclib) was recently approved for the treatment of metastatic ER-positive breast cancer in combination with endocrine therapy, and several other CDK-targeted drugs are in clinical trials [91, 92]. Amplification of CDK4/6 and CCND1 and loss of the CDK4/6 inhibitors CDKN2A and CDKN2C are often detected in ER-positive cancers [9]. However, results from the PALOMA-1 trial showed that these genetic aberrations are not predictive for the efficacy of treatment with palbociclib [93].

For TNBC, there are no targeted therapies specifically approved [47]. However, recent data suggest a significant clinical activity of poly(ADP-ribose) polymerase (PARP) inhibitors in germline BRCA mutant breast cancer with dysfunctional homologous recombination DNA repair [4749].

Potential New Therapeutic Targets and Treatment Strategies Derived From Genomic Analysis of Breast Cancer

Despite very rare driver mutations that have been found in directly targetable genes (e.g., kinase genes), limiting the development of genotype targeted-therapies in breast cancer, several agents are currently used in the clinic, whereas others are not yet approved for clinical practice or are still under evaluation (Table 3). Furthermore, results derived from next-generation sequencing (NGS) studies have demonstrated novel genetic aberrations and pathways dysregulation that may represent future therapeutic targets (Table 4).

Table 3

Targeted anticancer agents under investigation for breast cancer currently in phase II/III clinical trials

Table 3

Targeted anticancer agents under investigation for breast cancer currently in phase II/III clinical trials

Table 4

Potential therapeutic gene targets in breast cancer

Table 4

Potential therapeutic gene targets in breast cancer

ESR1 mutations, leading to ligand-independent signaling, commonly arise under the selective pressure of endocrine therapy during the treatment of metastatic disease [53]. Several novel selective estrogen receptor degraders are in phase I (i.e., GDC0927, AZD9496, and RAD1901) and phase II (GDC0810) clinical trials that may turn out to be effective new therapies for these ESR1 mutant cancers (Table 3). Similar mutations conferring ligand-independent activity to HER2, in the absence of HER2 amplification, were also reported, and several clinical trials are testing the efficacy of HER2 kinase inhibitors in these patients, who are currently considered “HER2 normal” by immunohistochemistry of fluorescence in situ hybridization assay.

It is also increasingly recognized that PIK3CA inhibitors alone may be not sufficient to keep this important pathway inhibited because of redundancies, feedback loops, and simultaneous mutations in downstream pathway members. Indeed, concurrent perturbations of the PI3K/AKT/mTOR and the mitogen-activated protein kinase (MAPK) cascades are frequently observed in tumors. Several inhibitors of the Raf/MEK/ERK pathway are currently under clinical evaluation alone and in combination with PIK3CA/mTOR inhibitors (Table 3) [9, 58, 94, 95]. Furthermore, PI3K inhibition has been shown to boost the ERK pathway through the enhancement of the epidermal growth factor receptor (EGFR)/HER2/human epidermal growth factor receptor 3 (HER3) signaling, and HER3 may in turn promote the reactivation of the PI3K/AKT and MAPK pathways [96, 97]. The modest activity of EGFR inhibitors in TNBC may also be due to crosstalk between signaling pathways and compensatory feedback loops [98101]. Thus, combination trials with other drugs, including NOTCH and AXL inhibitors, warrant further clinical investigation [99101]. Amplifications of FGFR1 and the activation of related pathways have been observed in TNBC [9]. Dovitinib and lucitanib showed antitumor activity, and clinical trials in fibroblast growth factor (FGF)-deregulated breast cancer are ongoing (Table 3). However, the therapeutic potential of FGFR inhibition in breast cancer requires further evaluation, in light of uncertain clinical results, and the key role of FGFRs in ordinary cell physiology [102].

An emerging success story in targeted therapies is the significant clinical activity of PARP inhibitors in germline BRCA mutant breast cancer [45, 103, 104]. Several trials showed single-agent activity in BRCA mutant breast cancers, and numerous trials, including a large adjuvant trial with olaparib, are under way (Table 3).

APOBEC mutations can lead to instability and mutagenesis in breast cancer [105, 106]. The APOBEC3-mediated mutagenesis results in a specific pattern of nucleic acid changes, called APOBEC mutation signature, which has been found to be enriched in HER2-overexpressed, basal-like, and luminal B subtypes. This evidence suggests that APOBECs could be crucial factors for targeted therapies aiming to block mechanisms producing widespread genomic alterations that drive tumor development, progression, and likely therapy resistance [105, 106]. Besides APOBEC enzymes, other mutational processes involving specific DNA repair pathways (i.e., mismatch repair and homologous recombination) lead to distinctive DNA damage [107, 108]. The identification of different genomic signatures arising from distinct defective pathways can provide a powerful means of revealing the somatic mutational basis of breast cancer [107, 108].

Recurrent mutations of TP53 make this gene a target of particular interest in TNBC. Considering that the direct targeting of TP53 is challenging, an alternative option may be to focus on activated signaling molecules downstream of TP53. For instance, CHEK1/2, ATM, and ATR are emerging as promising targets (Tables 3, 4) [109]. TP53-deficient cells rely on ATR/CHEK1 to arrest cell-cycle progression after DNA-damaging chemotherapy. Thus, the inhibition of CHEK1 and/or ATR in TP53-deficient cancers may lead to mitotic catastrophe and apoptosis [103, 104]. Several other TP53-associated kinases, such as polo-like kinases, aurora kinases, and, recently, the MPS1 kinase, have shown promising results in vitro and in vivo, suggesting their potential as suitable candidates for further development toward clinical studies (Table 4) [110]. The TP53-associated pathway is also defective in an important quote of luminal B tumors because of TP53 mutations, amplification of MDM2, or ATM loss [9]. Several molecules that inhibit the ubiquitin ligase MDM2, or that disrupt the MDM2-TP53 interaction, are currently in early clinical evaluation in patients with advanced malignancies other than breast cancer [111]. Other proteins involved in the ubiquitin-proteasome system, such as the tumor suppressor FBXW7 and the SKP1 ligase, have been found to be mutated or dysregulated in breast cancer (especially TNBC/basal-like), affecting a complex network of oncogenic processes, including cell differentiation and genomic stability [17, 112, 113]. In particular, loss of SKP1 has been shown to deregulate multiple targets, including EGFR and SRC tyrosine kinase in basal-like breast cancer [113]. An integrated genomic-proteomic approach has recently allowed the identification of novel phosphosite markers in PIK3CA- (i.e., EIF2AK4 and RPS6KA5) and TP53- (i.e., CHEK2, EEF2K, and MASTL) mutated tumors, and other important amplicon-associated phosphorylated kinases (i.e., CDK12, PAK1, PTK2, RIPK2 and TLK2) (Table 4) [113]. Overall, the analysis of activated pathways and the understanding of dysregulated signaling circuitries can provide novel insights into the functional consequences of somatic mutations and help identify novel therapeutic targets in breast cancer.

Overall, the analysis of activated pathways and the understanding of dysregulated signaling circuitries can provide novel insights into the functional consequences of somatic mutations and help identify novel therapeutic targets in breast cancer.

Genes identified as oncogenic drivers in other tumor types may also represent potential therapeutic targets. For instance, tumors carrying activating mutations and amplification in BRAF, KRAS, and MET genes might be sensitive to specific inhibitors. Furthermore, other potential driver mutations in genes that are involved in chromatin remodeling and epigenetic regulation of gene expression have been reported in breast cancer [8, 9, 17, 114]. Patients with such mutations may be potentially enrolled in clinical trials using drugs that target epigenetic modifications, particularly DNA methyltransferase and histone deacetylase inhibitors (Tables 3, 4).

Tumor cells dynamically interact with a heterogeneous microenvironment through a bidirectional signaling network. An outstanding ability of the immune system, especially adaptive immunity, is to recognize tumor-specific antigen. Different gene mutations generate new antigenic peptides that could be recognized by T lymphocytes and trigger antitumor immune responses [115]. The identification of specific antigens resulting from mutations in driver genes, as well as chromosomal translocations and gene amplifications, could provide the rationale for the development of novel immunotherapeutic strategies for patients with breast cancers [116]. Remarkably, the majority of mutated antigens contributing to the immunogenicity of human tumors results from alterations in nonrecurrent genes, suggesting that infrequent mutations identified from large-scale sequencing studies may represent tumor-specific antigens and hold promise for use in cancer immunotherapy (Table 4) [115].

Discussion

Identifying the functional relevance of individually often-rare variants is a daunting challenge for laboratory investigators and clinical trialists. Studying the functional interaction between the multiple somatic alterations and inherited germline polymorphisms is going to be key for the formulation of a more comprehensive model of carcinogenesis. The importance of interaction between the host immune system and the cancer is also increasingly recognized as an important determinant of clinical outcome. Somatic mutations, even without functional impact on biological pathways, can represent novel antigens that could activate adaptive immunity in the tumor microenvironment. A major challenge in the implementation of molecular target-directed therapeutic trials is the rarity and diversity of potentially actionable mutations in breast cancer, so that very large numbers of patients need to be screened to find the 20–100 patients required for a typical phase II trial [117119].

The presence of multiple potentially targetable alterations in most cancers suggests that individualized combination therapies may be the most effective strategy, which presents a fundamental challenge to any traditional clinical trial design and raises unprecedented regulatory issues. Furthermore, the repertoire of biologically and therapeutically important mutations may change during the evolution of a cancer or perhaps even from metastatic site to site. Although several studies have demonstrated the existence of “private” mutations (i.e., mutations unique to a particular tumor site while absent in other sites), the biological and therapeutic relevance of these observations is unknown.

Molecular alterations are also being investigated as response predictors to targeted therapies. However, no single gene alterations emerged as clinically useful predictors to any therapy (other than HER2 amplification for HER2-targeted therapies). There is only weak, or no, association between PIK3CA mutations and response to mTOR or PI3K inhibitors in clinical trials. Similarly, amplifications in the CDK genes were not predictive of benefit from CDK4/6 inhibitors, and FGFR inhibitors also had only modest activity in FGFR1-amplified tumors. Conversely, activating mutations in HER2 emerged as a potential new marker and target to extend the use of anti-HER2 kinase inhibitor therapy. Several ongoing clinical trials are examining the therapeutic target value of this abnormality. ESR1 mutations that render the receptor constitutively active are also frequently encountered in metastatic biopsies, whereas this mutation is very rare in primary cancers. Efforts are under way to develop therapies that target the mutant ER.

Some, but not all, of the somatic mutations present in a cancer can also be detected as circulating-free tumor DNA. It is hoped that sequencing of circulating tumor DNA that can easily be obtained from blood may provide a tool to monitor risk of recurrence after potentially curative therapy, detect early recurrence, and monitor response to therapy [120124].

Besides the importance of variants in protein-coding regions, the majority of the alterations occur in noncoding portions of the genome [125]. Different noncoding sequence variants, ranging from single-nucleotide variants to small insertions and deletions, to large structural variants have been demonstrated in human cancer [125127]. Somatic variations can lead to gain of transcription factor-binding sites responsible of tumorigenesis. Fusion events because of genomic rearrangements, variations in noncoding RNAs and their binding sites, and variants in pseudogenes modulating gene expression have been linked to cancer [125]. Like somatic variants, germline noncoding affects gene expression through several mechanisms (e.g., promoter mutations, single-nucleotide polymorphisms in enhancers and noncoding RNAs and their binding sites, and variants in introns) [125]. Recently, recurrent mutations have been found in the promoter of TBC1D12 and WDR74, as well as in the long noncoding RNAs MALAT1 and NEAT1 in breast cancer [108]. However, the identification of noncoding drivers is complex and requires additional studies. Overall, it is evident that cancer results from a more complex interplay of inherited germline and acquired somatic variants, including also noncoding region alterations. As for breast cancer, which seems to harbor more alterations in the noncoding regions compared with other tumors, the identification of noncoding driver mutations/alterations remains crucial to enable therapeutic approaches that target the specific linked proteins. The constant evolution of technologies allows the ever-increasing identification of novel mutated cancer genes (e.g., FOXP1, MED23, MLLT4, XBP1, and ZFP36L1) [108]. This ongoing update of cancer genes and noncoding regions’ alterations progresses toward a comprehensive understanding of the source and consequences of somatic mutations, ultimately enabling the development of novel targeted therapies for breast cancer.

Conclusion

Recent insights from comprehensive genomic characterizations of cancer revealed large-scale heterogeneity in the genomic landscape of breast cancers. The diversity of genomic aberrations and the resulting dysregulation of a broad range of biological pathways help to explain the varied clinical behavior of breast tumors. Better understanding the biological effects of these aberrations remains a major focus of research and may pave the way for rational combination targeted therapies.

For Further Reading: Juliann Chmielecki, Jeffrey S. Ross, Kai Wang et al. Oncogenic Alterations in ERBB2/HER2 Represent Potential Therapeutic Targets Across Tumors From Diverse Anatomic Sites of Origin. The Oncologist 2015;20:7–12.

Implications for Practice: Tumors with amplification or overexpression of ERBB2/HER2 have a high likelihood of being sensitive to ERBB2/HER2 inhibitors, based on evidence from published studies. Current clinical practice investigates amplification or overexpression of ERBB2/HER2 in breast, gastric, and gastroesophageal cancers. However, recent studies suggest that mutations can also activate this gene, and these alterations may be similarly sensitive to ERBB2/HER2 inhibitors. Our data identified activation of ERBB2/HER2 (either amplification or activating mutation) in 27 different tumor types. Consequently, more comprehensive molecular profiling of multiple tumor types has the potential to identify additional patients who may derive clinical benefit from ERBB2/HER2 inhibitors.

Acknowledgments

This work was supported by Associazione Italiana per la Ricerca sul Cancro Grant 6251 to L.S.; Fondazione Italiana per la Ricerca sul Cancro Fellowship 18328 to G.B.; and the Breast Cancer Research Foundation to L.P.

Author Contributions

Conception/Design: Libero Santarpia, Lajos Pusztai

Provision of study material or patients: Libero Santarpia, Giulia Bottai, Catherine M. Kelly, Balázs Győrffy, Borbala Székely, Lajos Pusztai

Collection and/or assembly of data: Libero Santarpia, Giulia Bottai, Catherine M. Kelly, Balázs Győrffy, Lajos Pusztai

Data analysis and interpretation: Libero Santarpia, Giulia Bottai, Catherine M. Kelly, Balázs Győrffy, Borbala Székely, Lajos Pusztai

Manuscript writing: Libero Santarpia, Lajos Pusztai

Final approval of manuscript: Libero Santarpia, Giulia Bottai, Catherine M. Kelly, Balázs Győrffy, Borbala Székely, Lajos Pusztai

Disclosures

The authors indicated no financial relationships.

References

1

Anderson
 
WF
,
Rosenberg
 
PS
,
Prat
 
A
 et al. .
How many etiological subtypes of breast cancer: Two, three, four, or more?
.
J Natl Cancer Inst
.
2014
;
106
:
dju165
.

2

Dawson
 
SJ
,
Rueda
 
OM
,
Aparicio
 
S
 et al. .
A new genome-driven integrated classification of breast cancer and its implications
.
EMBO J
.
2013
;
32
:
617
628
.

3

Couch
 
FJ
,
Nathanson
 
KL
,
Offit
 
K
.
Two decades after BRCA: Setting paradigms in personalized cancer care and prevention
.
Science
.
2014
;
343
:
1466
470
.

4

Fadoukhair
 
Z
,
Zardavas
 
D
,
Chad
 
MA
 et al. .
Evaluation of targeted therapies in advanced breast cancer: The need for large-scale molecular screening and transformative clinical trial designs
.
Oncogene
.
2016
;
35
:
1743
1749
.

5

Redig
 
AJ
,
Jänne
 
PA
.
Basket trials and the evolution of clinical trial design in an era of genomic medicine
.
J Clin Oncol
.
2015
;
33
:
975
977
.

6

Lacombe
 
D
,
Tejpar
 
S
,
Salgado
 
R
 et al. .
European perspective for effective cancer drug development
.
Nat Rev Clin Oncol
.
2014
;
11
:
492
498
.

7

Kelly
 
CM
,
Buzdar
 
AU
.
Using multiple targeted therapies in oncology: Considerations for use, and progress to date in breast cancer
.
Drugs
.
2013
;
73
:
505
515
.

8

Stephens
 
PJ
,
Tarpey
 
PS
,
Davies
 
H
 et al. .
The landscape of cancer genes and mutational processes in breast cancer
.
Nature
.
2012
;
486
:
400
404
.

9

Cancer Genome Atlas Network
.
Comprehensive molecular portraits of human breast tumours
.
Nature
.
2012
;
490
:
61
70
.

10

Curtis
 
C
,
Shah
 
SP
,
Chin
 
SF
 et al. .
The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups
.
Nature
.
2012
;
486
:
346
352
.

11

Bergamaschi
 
A
,
Kim
 
YH
,
Wang
 
P
 et al. .
Distinct patterns of DNA copy number alteration are associated with different clinicopathological features and gene-expression subtypes of breast cancer
.
Genes Chromosomes Cancer
.
2006
;
45
:
1033
1040
.

12

Yates
 
LR
,
Gerstung
 
M
,
Knappskog
 
S
 et al. .
Subclonal diversification of primary breast cancer revealed by multiregion sequencing
.
Nat Med
.
2015
;
21
:
751
759
.

13

Forbes
 
SA
,
Bindal
 
N
,
Bamford
 
S
 et al. .
COSMIC: Mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer
.
Nucleic Acids Res
.
2011
;
39
:
D945
D950
.

14

Banerji
 
S
,
Cibulskis
 
K
,
Rangel-Escareno
 
C
 et al. .
Sequence analysis of mutations and translocations across breast cancer subtypes
.
Nature
.
2012
;
486
:
405
409
.

15

Stephens
 
PJ
,
McBride
 
DJ
,
Lin
 
ML
 et al. .
Complex landscapes of somatic rearrangement in human breast cancer genomes
.
Nature
.
2009
;
462
:
1005
1010
.

16

Santarpia
 
L
,
Iwamoto
 
T
,
Di Leo
 
A
 et al. .
DNA repair gene patterns as prognostic and predictive factors in molecular breast cancer subtypes
.
The Oncologist
.
2013
;
18
:
1063
1073
.

17

Santarpia
 
L
,
Qi
 
Y
,
Stemke-Hale
 
K
 et al. .
Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers
.
Breast Cancer Res Treat
.
2012
;
134
:
333
343
.

18

Asmann
 
YW
,
Necela
 
BM
,
Kalari
 
KR
 et al. .
Detection of redundant fusion transcripts as biomarkers or disease-specific therapeutic targets in breast cancer
.
Cancer Res
.
2012
;
72
:
1921
1928
.

19

Ciriello
 
G
,
Sinha
 
R
,
Hoadley
 
KA
 et al. .
The molecular diversity of Luminal A breast tumors
.
Breast Cancer Res Treat
.
2013
;
141
:
409
420
.

20

Silwal-Pandit
 
L
,
Vollan
 
HK
,
Chin
 
SF
 et al. .
TP53 mutation spectrum in breast cancer is subtype specific and has distinct prognostic relevance
.
Clin Cancer Res
.
2014
;
20
:
3569
3580
.

21

Adélaïde
 
J
,
Finetti
 
P
,
Bekhouche
 
I
 et al. .
Integrated profiling of basal and luminal breast cancers
.
Cancer Res
.
2007
;
67
:
11565
11575
.

22

Ades
 
F
,
Zardavas
 
D
,
Bozovic-Spasojevic
 
I
 et al. .
Luminal B breast cancer: Molecular characterization, clinical management, and future perspectives
.
J Clin Oncol
.
2014
;
32
:
2794
2803
.

23

Staaf
 
J
,
Jönsson
 
G
,
Ringnér
 
M
 et al. .
Landscape of somatic allelic imbalances and copy number alterations in HER2-amplified breast cancer
.
Breast Cancer Res
.
2011
;
13
:
R129
.

24

Sircoulomb
 
F
,
Bekhouche
 
I
,
Finetti
 
P
 et al. .
Genome profiling of ERBB2-amplified breast cancers
.
BMC Cancer
.
2010
;
10
:
539
.

25

Staaf
 
J
,
Jönsson
 
G
,
Ringnér
 
M
 et al. .
High-resolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer
.
Breast Cancer Res
.
2010
;
12
:
R25
.

26

Arriola
 
E
,
Marchio
 
C
,
Tan
 
DS
 et al. .
Genomic analysis of the HER2/TOP2A amplicon in breast cancer and breast cancer cell lines
.
Lab Invest
.
2008
;
88
:
491
503
.

27

Press
 
MF
,
Sauter
 
G
,
Buyse
 
M
 et al. .
Alteration of topoisomerase II-alpha gene in human breast cancer: Association with responsiveness to anthracycline-based chemotherapy
.
J Clin Oncol
.
2011
;
29
:
859
867
.

28

Lehmann
 
BD
,
Bauer
 
JA
,
Chen
 
X
 et al. .
Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies
.
J Clin Invest
.
2011
;
121
:
2750
2767
.

29

Shah
 
SP
,
Roth
 
A
,
Goya
 
R
 et al. .
The clonal and mutational evolution spectrum of primary triple-negative breast cancers
.
Nature
.
2012
;
486
:
395
399
.

30

Abramson
 
VG
,
Lehmann
 
BD
,
Ballinger
 
TJ
 et al. .
Subtyping of triple-negative breast cancer: implications for therapy
.
Cancer
.
2015
;
121
:
8
16
.

31

Andre
 
F
,
Job
 
B
,
Dessen
 
P
 et al. .
Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array
.
Clin Cancer Res
.
2009
;
15
:
441
451
.

32

Natrajan
 
R
,
Weigelt
 
B
,
Mackay
 
A
 et al. .
An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers
.
Breast Cancer Res Treat
.
2010
;
121
:
575
589
.

33

Turner
 
N
,
Lambros
 
MB
,
Horlings
 
HM
 et al. .
Integrative molecular profiling of triple negative breast cancers identifies amplicon drivers and potential therapeutic targets
.
Oncogene
.
2010
;
29
:
2013
2023
.

34

McFarland
 
CD
,
Korolev
 
KS
,
Kryukov
 
GV
 et al. .
Impact of deleterious passenger mutations on cancer progression
.
Proc Natl Acad Sci USA
.
2013
;
110
:
2910
2915
.

35

Wood
 
LD
,
Parsons
 
DW
,
Jones
 
S
 et al. .
The genomic landscapes of human breast and colorectal cancers
.
Science
.
2007
;
318
:
1108
1113
.

36

Navin
 
NE
.
Tumor evolution in response to chemotherapy: Phenotype versus genotype
.
Cell Reports
.
2014
;
6
:
417
419
.

37

Wang
 
Y
,
Waters
 
J
,
Leung
 
ML
 et al. .
Clonal evolution in breast cancer revealed by single nucleus genome sequencing
.
Nature
.
2014
;
512
:
155
160
.

38

Almendro
 
V
,
Cheng
 
YK
,
Randles
 
A
 et al. .
Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity
.
Cell Reports
.
2014
;
6
:
514
527
.

39

Fisher
 
R
,
Pusztai
 
L
,
Swanton
 
C
.
Cancer heterogeneity: Implications for targeted therapeutics
.
Br J Cancer
.
2013
;
108
:
479
485
.

40

Negrini
 
S
,
Gorgoulis
 
VG
,
Halazonetis
 
TD
.
Genomic instability—an evolving hallmark of cancer
.
Nat Rev Mol Cell Biol
.
2010
;
11
:
220
228
.

41

Metzger-Filho
 
O
,
Tutt
 
A
,
De Azambuja
 
E
 et al. .
Dissecting the heterogeneity of triple-negative breast cancer
.
J Clin Oncol
.
2012
;
30
:
1879
1887
.

42

Shah
 
SP
,
Morin
 
RD
,
Khattra
 
J
 et al. .
Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution
.
Nature
.
2009
;
461
:
809
813
.

43

Ding
 
L
,
Ellis
 
MJ
,
Li
 
S
 et al. .
Genome remodelling in a basal-like breast cancer metastasis and xenograft
.
Nature
.
2010
;
464
:
999
1005
.

44

Burrell
 
RA
,
Swanton
 
C
.
Tumour heterogeneity and the evolution of polyclonal drug resistance
.
Mol Oncol
.
2014
;
8
:
1095
1111
.

45

Lord
 
CJ
,
Ashworth
 
A
.
Mechanisms of resistance to therapies targeting BRCA-mutant cancers
.
Nat Med
.
2013
;
19
:
1381
1388
.

46

Pinto
 
CA
,
Widodo
 
E
,
Waltham
 
M
 et al. .
Breast cancer stem cells and epithelial mesenchymal plasticity—implications for chemoresistance
.
Cancer Lett
.
2013
;
341
:
56
62
.

47

Badve
 
S
,
Dabbs
 
DJ
,
Schnitt
 
SJ
 et al. .
Basal-like and triple-negative breast cancers: A critical review with an emphasis on the implications for pathologists and oncologists
.
Mod Pathol
.
2011
;
24
:
157
167
.

48

Tutt
 
A
,
Ellis
 
P
,
Kilburn
 
L
 et al. .
The TNT trial: A randomized phase III trial of carboplatin (C) compared with docetaxel (D) for patients with metastatic or recurrent locally advanced triple negative or BRCA1/2 breast cancer (CRUK/07/012)
.
Paper presented at San Antonio Breast Cancer Symposium
; December 9–13,
2014
;
San Antonio, TX
. doi:10.1158/1538-7445.SABCS14-S3-01

49

Tutt
 
A
,
Robson
 
M
,
Garber
 
JE
 et al. .
Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: A proof-of-concept trial
.
Lancet
.
2010
;
376
:
235
244
.

50

Toy
 
W
,
Shen
 
Y
,
Won
 
H
 et al. .
ESR1 ligand-binding domain mutations in hormone-resistant breast cancer
.
Nat Genet
.
2013
;
45
:
1439
1445
.

51

Jeselsohn
 
R
,
Yelensky
 
R
,
Buchwalter
 
G
 et al. .
Emergence of constitutively active estrogen receptor-α mutations in pretreated advanced estrogen receptor-positive breast cancer
.
Clin Cancer Res
.
2014
;
20
:
1757
1767
.

52

Jeselsohn
 
R
,
Buchwalter
 
G
,
De Angelis
 
C
 et al. .
ESR1 mutations—a mechanism for acquired endocrine resistance in breast cancer
.
Nat Rev Clin Oncol
.
2015
;
12
:
573
583
.

53

Schiavon
 
G
,
Hrebien
 
S
,
Garcia-Murillas
 
I
 et al. .
Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer
.
Sci Transl Med
.
2015
;
7
:
313ra182
.

54

Chandarlapaty
 
S
,
Sung
 
P
,
Chen
 
D
 et al. et al.
cfDNA analysis from BOLERO-2 plasma samples identifies a high rate of ESR1 mutations: Exploratory analysis for prognostic and predictive correlation of mutations reveals different efficacy outcomes of endocrine therapy–based regimens
.
Paper presented at: San Antonio Breast Cancer Symposium
; December 8-12,
2015
;
San Antonio, TX
. doi:10.1158/1538-7445.SABCS15-S2-07

55

André
 
F
,
Cortés
 
J
.
Rationale for targeting fibroblast growth factor receptor signaling in breast cancer
.
Breast Cancer Res Treat
.
2015
;
150
:
1
8
.

56

Ellis
 
MJ
,
Ding
 
L
,
Shen
 
D
 et al. .
Whole-genome analysis informs breast cancer response to aromatase inhibition
.
Nature
.
2012
;
486
:
353
360
.

57

Rexer
 
BN
,
Arteaga
 
CL
.
Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications
.
Crit Rev Oncog
.
2012
;
17
:
1
16
.

58

De Luca
 
A
,
Maiello
 
MR
,
D’Alessio
 
A
 et al. .
The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: Role in cancer pathogenesis and implications for therapeutic approaches
.
Expert Opin Ther Targets
.
2012
;
16
 
suppl 2
:
S17
S27
.

59

Zardavas
 
D
,
Phillips
 
WA
,
Loi
 
S
.
PIK3CA mutations in breast cancer: Reconciling findings from preclinical and clinical data
.
Breast Cancer Res
.
2014
;
16
:
201
.

60

Loi
 
S
,
Michiels
 
S
,
Lambrechts
 
D
 et al. .
Somatic mutation profiling and associations with prognosis and trastuzumab benefit in early breast cancer
.
J Natl Cancer Inst
.
2013
;
105
:
960
967
.

61

Stemke-Hale
 
K
,
Gonzalez-Angulo
 
AM
,
Lluch
 
A
 et al. .
An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer
.
Cancer Res
.
2008
;
68
:
6084
6091
.

62

Sabine
 
VS
,
Crozier
 
C
,
Brookes
 
CL
 et al. .
Mutational analysis of PI3K/AKT signaling pathway in tamoxifen exemestane adjuvant multinational pathology study
.
J Clin Oncol
.
2014
;
32
:
2951
2958
.

63

Di Nicolantonio
 
F
,
Arena
 
S
,
Tabernero
 
J
 et al. .
Deregulation of the PI3K and KRAS signaling pathways in human cancer cells determines their response to everolimus
.
J Clin Invest
.
2010
;
120
:
2858
2866
.

64

Dave
 
B
,
Migliaccio
 
I
,
Gutierrez
 
MC
 et al. .
Loss of phosphatase and tensin homolog or phosphoinositol-3 kinase activation and response to trastuzumab or lapatinib in human epidermal growth factor receptor 2-overexpressing locally advanced breast cancers
.
J Clin Oncol
.
2011
;
29
:
166
173
.

65

Baselga
 
J
,
Cortés
 
J
,
Im
 
SA
 et al. .
Biomarker analyses in CLEOPATRA: A phase III, placebo-controlled study of pertuzumab in human epidermal growth factor receptor 2-positive, first-line metastatic breast cancer
.
J Clin Oncol
.
2014
;
32
:
3753
3761
.

66

Loibl
 
S
,
Von Minckwitz
 
G
,
Schneeweiss
 
A
 et al. .
PIK3CA mutations are associated with lower rates of pathologic complete response to anti-human epidermal growth factor receptor 2 (her2) therapy in primary HER2-overexpressing breast cancer
.
J Clin Oncol
.
2014
;
32
:
3212
3220
.

67

Majewski
 
IJ
,
Nuciforo
 
P
,
Mittempergher
 
L
 et al. .
PIK3CA mutations are associated with decreased benefit to neoadjuvant human epidermal growth factor receptor 2-targeted therapies in breast cancer
.
J Clin Oncol
.
2015
;
33
:
1334
1339
.

68

Nuciforo
 
PG
,
Aura
 
C
,
Holmes
 
E
 et al. .
Benefit to neoadjuvant anti-human epidermal growth factor receptor 2 (HER2)-targeted therapies in HER2-positive primary breast cancer is independent of phosphatase and tensin homolog deleted from chromosome 10 (PTEN) status
.
Ann Oncol
.
2015
;
26
:
1494
1500
.

69

Pandolfi
 
PP
.
Breast cancer–loss of PTEN predicts resistance to treatment
.
N Engl J Med
.
2004
;
351
:
2337
2338
.

70

Bertheau
 
P
,
Turpin
 
E
,
Rickman
 
DS
 et al. .
Exquisite sensitivity of TP53 mutant and basal breast cancers to a dose-dense epirubicin-cyclophosphamide regimen
.
PLoS Med
.
2007
;
4
:
e90
.

71

Bonnefoi
 
H
,
Piccart
 
M
,
Bogaerts
 
J
 et al. .
TP53 status for prediction of sensitivity to taxane versus non-taxane neoadjuvant chemotherapy in breast cancer (EORTC 10994/BIG 1-00): A randomised phase 3 trial
.
Lancet Oncol
.
2011
;
12
:
527
539
.

72

Kandioler-Eckersberger
 
D
,
Ludwig
 
C
,
Rudas
 
M
v et al. .
TP53 mutation and p53 overexpression for prediction of response to neoadjuvant treatment in breast cancer patients
.
Clin Cancer Res
.
2000
;
6
:
50
56
.

73

Yap
 
TA
,
Gerlinger
 
M
,
Futreal
 
PA
 et al. .
Intratumor heterogeneity: Seeing the wood for the trees
.
Sci Transl Med
.
2012
;
4
:
127ps10
.

74

Zardavas
 
D
,
Irrthum
 
A
,
Swanton
 
C
 et al. .
Clinical management of breast cancer heterogeneity
.
Nat Rev Clin Oncol
.
2015
;
12
:
381
394
.

75

Turner
 
NC
,
Reis-Filho
 
JS
.
Genetic heterogeneity and cancer drug resistance
.
Lancet Oncol
.
2012
;
13
:
e178
e185
.

76

Swanton
 
C
.
Intratumor heterogeneity: Evolution through space and time
.
Cancer Res
.
2012
;
72
:
4875
4882
.

77

Balko
 
JM
,
Giltnane
 
JM
,
Wang
 
K
 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
245
.

78

Jiang
 
YZ
,
Yu
 
KD
,
Bao
 
J
 et al. .
Favorable prognostic impact in loss of TP53 and PIK3CA mutations after neoadjuvant chemotherapy in breast cancer
.
Cancer Res
.
2014
;
74
:
3399
3407
.

79

Janiszewska
 
M
,
Liu
 
L
,
Almendro
 
V
 et al. .
In situ single-cell analysis identifies heterogeneity for PIK3CA mutation and HER2 amplification in HER2-positive breast cancer
.
Nat Genet
.
2015
;
47
:
1212
1219
.

80

Axelrod
 
R
,
Axelrod
 
DE
,
Pienta
 
KJ
.
Evolution of cooperation among tumor cells
.
Proc Natl Acad Sci USA
.
2006
;
103
:
13474
13479
.

81

Gianni
 
L
,
Bianchini
 
G
,
Kiermaier
 
A
 et al. .
Neoadjuvant pertuzumab (P) and trastuzumab (H): Biomarker analyses of a 4-arm randomized phase II study (NeoSphere) in patients (pts) with HER2-positive breast cancer (BC)
.
Cancer Res
.
2011
;
71
 
suppl 24
:
S5-1a
.

82

Contreras
 
A
,
Herrera
 
S
,
Wang
 
T
 et al. .
PIK3CA mutations and/or low PTEN predict resistance to combined anti-HER2 therapy with lapatinib and trastuzumab and without chemotherapy in TBCRC006, a neoadjuvant trial of HER2-positive breast cancer patients
.
Cancer Res
.
2013
;
73
 
suppl 24
:
PD1-2a
.

83

Bose
 
R
,
Kavuri
 
SM
,
Searleman
 
AC
 et al. .
Activating HER2 mutations in HER2 gene amplification negative breast cancer
.
Cancer Discov
.
2013
;
3
:
224
237
.

84

Mohamed
 
A
,
Krajewski
 
K
,
Cakar
 
B
 et al. .
Targeted therapy for breast cancer
.
Am J Pathol
.
2013
;
183
:
1096
1112
.

85

Hortobagyi
 
GN
,
Chen
 
D
,
Piccart
 
M
 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
 
2015

86

Ma
 
CX
,
Suman
 
VJ
,
Goetz
 
M
 et al. .
A phase II neoadjuvant trial of MK-2206, an AKT inhibitor, in combination with anastrozole for clinical stage 2 or 3 PIK3CA mutant estrogen receptor positive HER2 negative (ER+HER2-) breast cancer (BC)
.
Paper presented at San Antonio Breast Cancer Symposium
; December 8–12,
2015
;
San Antonio, TX
.

87

Baselga
 
J
,
Im
 
S-A
,
Iwata
 
H
 et al. .
PIK3CA status in circulating tumor DNA (ctDNA) predicts efficacy of buparlisib (BUP) plus fulvestrant (FULV) in postmenopausal women with endocrine-resistant HR+/HER2– advanced breast cancer (BC): First results from the randomized, phase III BELLE-2 trial
.
Paper presented at San Antonio Breast Cancer Symposium
; December 8–12,
2015
;
San Antonio, TX
.

88

Juric
 
D
,
Rodon
 
J
,
Gonzalez-Angulo
 
AM
 et al. ,.
BYL719, a next generation PI3K alpha specific inhibitor: Preliminary safety, PK, and efficacy results from the first-in-human study
.
Philadelphia, PA
:
AACR
.
2012
. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research, Chicago, IL, 31 March–4 April 2012.

89

Juric
 
D
,
Krop
 
I
,
Ramanathan
 
RK
 et al. ,.
GDC-0032, a beta isoform-sparing PI3K inhibitor: Results of a first-in-human phase Ia dose escalation study
.
Philadelphia, PA
:
AACR
.
2013
. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research, Washington, DC, 6–10 April 2013.

90

Arnedos
 
M
,
Vicier
 
C
,
Loi
 
S
 et al. .
Precision medicine for metastatic breast cancer—limitations and solutions
.
Nat Rev Clin Oncol
.
2015
;
12
:
693
704
.

91

Turner
 
NC
,
Ro
 
J
,
André
 
F
 et al. .
Palbociclib in hormone-receptor-positive advanced breast cancer
.
N Engl J Med
.
2015
;
373
:
209
219
.

92

Morrison
 
C
.
Pfizer’s CDK4/6 inhibitor approved for advanced breast cancer
.
Nat Biotechnol
.
2015
;
33
:
323
324
.

93

Finn
 
RS
,
Crown
 
JP
,
Lang
 
I
 et al. .
The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study
.
Lancet Oncol
.
2015
;
16
:
25
35
.

94

Davis
 
NM
,
Sokolosky
 
M
,
Stadelman
 
K
 et al. .
Deregulation of the EGFR/PI3K/PTEN/Akt/mTORC1 pathway in breast cancer: possibilities for therapeutic intervention
.
Oncotarget
.
2014
;
5
:
4603
4650
.

95

Blair
 
BG
,
Wu
 
X
,
Zahari
 
MS
 et al. .
A phosphoproteomic screen demonstrates differential dependence on HER3 for MAP kinase pathway activation by distinct PIK3CA mutations
.
Proteomics
.
2015
;
15
:
318
326
.

96

Serra
 
V
,
Scaltriti
 
M
,
Prudkin
 
L
 et al. .
PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer
.
Oncogene
.
2011
;
30
:
2547
2557
.

97

Gala
 
K
,
Chandarlapaty
 
S
.
Molecular pathways: HER3 targeted therapy
.
Clin Cancer Res
.
2014
;
20
:
1410
1416
.

98

Carey
 
LA
,
Rugo
 
HS
,
Marcom
 
PK
 et al. .
TBCRC 001: Randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer
.
J Clin Oncol
.
2012
;
30
:
2615
2623
.

99

Meyer
 
AS
,
Miller
 
MA
,
Gertler
 
FB
 et al. .
The receptor AXL diversifies EGFR signaling and limits the response to EGFR-targeted inhibitors in triple-negative breast cancer cells
.
Sci Signal
.
2013
;
6
:
ra66
.

100

Elkabets
 
M
,
Pazarentzos
 
E
,
Juric
 
D
 et al. .
AXL mediates resistance to PI3Kα inhibition by activating the EGFR/PKC/mTOR axis in head and neck and esophageal squamous cell carcinomas
.
Cancer Cell
.
2015
;
27
:
533
546
.

101

Jiao
 
X
,
Wood
 
LD
,
Lindman
 
M
 et al. .
Somatic mutations in the Notch, NF-KB, PIK3CA, and Hedgehog pathways in human breast cancers
.
Genes Chromosomes Cancer
.
2012
;
51
:
480
489
.

102

Brooks
 
AN
,
Kilgour
 
E
,
Smith
 
PD
.
Molecular pathways: Fibroblast growth factor signaling: a new therapeutic opportunity in cancer
.
Clin Cancer Res
.
2012
;
18
:
1855
1862
.

103

Turner
 
N
,
Moretti
 
E
,
Siclari
 
O
 et al. .
Targeting triple negative breast cancer: Is p53 the answer?
.
Cancer Treat Rev
.
2013
;
39
:
541
550
.

104

Guha
 
M
.
PARP inhibitors stumble in breast cancer
.
Nat Biotechnol
.
2011
;
29
:
373
374
.

105

Roberts
 
SA
,
Lawrence
 
MS
,
Klimczak
 
LJ
 et al. .
An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers
.
Nat Genet
.
2013
;
45
:
970
976
.

106

Nik-Zainal
 
S
,
Wedge
 
DC
,
Alexandrov
 
LB
 et al. .
Association of a germline copy number polymorphism of APOBEC3A and APOBEC3B with burden of putative APOBEC-dependent mutations in breast cancer
.
Nat Genet
.
2014
;
46
:
487
491
.

107

Morganella
 
S
,
Alexandrov
 
LB
,
Glodzik
 
D
 et al. .
The topography of mutational processes in breast cancer genomes
.
Nat Commun
.
2016
;
7
:
11383
.

108

Nik-Zainal
 
S
,
Davies
 
H
,
Staaf
 
J
 et al. .
Landscape of somatic mutations in 560 breast cancer whole-genome sequences
.
Nature
.
2016
;
534
:
4754
.

109

Curtin
 
NJ
.
DNA repair dysregulation from cancer driver to therapeutic target
.
Nat Rev Cancer
.
2012
;
12
:
801
817
.

110

Győrffy
 
B
,
Bottai
 
G
,
Lehmann-Che
 
J
 et al. .
TP53 mutation-correlated genes predict the risk of tumor relapse and identify MPS1 as a potential therapeutic kinase in TP53-mutated breast cancers
.
Mol Oncol
.
2014
;
8
:
508
519
.

111

Wade
 
M
,
Li
 
YC
,
Wahl
 
GM
.
MDM2, MDMX and p53 in oncogenesis and cancer therapy
.
Nat Rev Cancer
.
2013
;
13
:
83
96
.

112

Davis
 
RJ
,
Welcker
 
M
,
Clurman
 
BE
.
Tumor suppression by the Fbw7 ubiquitin ligase: Mechanisms and opportunities
.
Cancer Cell
.
2014
;
26
:
455
464
.

113

Mertins
 
P
,
Mani
 
DR
,
Ruggles
 
KV
 et al. .
Proteogenomics connects somatic mutations in signalling in breast cancer
.
Nature
.
2016
;
534
:
55
62
.

114

Prensner
 
JR
,
Chinnaiyan
 
AM
.
Metabolism unhinged: IDH mutations in cancer
.
Nat Med
.
2011
;
17
:
291
293
.

115

Coulie
 
PG
,
Van den Eynde
 
BJ
,
Van der Bruggen
 
P
.
Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy
.
Nat Rev Cancer
.
2014
;
14
:
135
146
.

116

Tran
 
E
,
Turcotte
 
S
,
Gros
 
A
 et al. .
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer
.
Science
.
2014
;
344
:
641
645
.

117

Zardavas
 
D
,
Maetens
 
M
,
Irrthum
 
A
 et al. .
The AURORA initiative for metastatic breast cancer
.
Br J Cancer
.
2014
;
111
:
1881
1887
.

118

Breast Cancer Research Foundation. Founder’s fund
. Available at http://www.bcrfcure.org/founders-fund. Accessed Dec 19, 2015.

119

Stahel
 
R
,
Bogaerts
 
J
,
Ciardiello
 
F
 et al. .
Optimising translational oncology in clinical practice: Strategies to accelerate progress in drug development
.
Cancer Treat Rev
.
2015
;
41
:
129
135
.

120

De Mattos-Arruda
 
L
,
Cortes
 
J
,
Santarpia
 
L
 et al. .
Circulating tumour cells and cell-free DNA as tools for managing breast cancer
.
Nat Rev Clin Oncol
.
2013
;
10
:
377
389
.

121

De Mattos-Arruda
 
L
,
Weigelt
 
B
,
Cortes
 
J
 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
1735
.

122

Murtaza
 
M
,
Dawson
 
SJ
,
Pogrebniak
 
K
 et al. .
Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer
.
Nat Commun
.
2015
;
6
:
8760
.

123

Dawson
 
SJ
,
Tsui
 
DW
,
Murtaza
 
M
 et al. .
Analysis of circulating tumor DNA to monitor metastatic breast cancer
.
N Engl J Med
.
2013
;
368
:
1199
1209
.

124

Garcia-Murillas
 
I
,
Schiavon
 
G
,
Weigelt
 
B
 et al. .
Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer
.
Sci Transl Med
.
2015
;
7
:
302ra133
.

125

Khurana
 
E
,
Fu
 
Y
,
Chakravarty
 
D
 et al. .
Role of non-coding sequence variants in cancer
.
Nat Rev Genet
.
2016
;
17
:
93
108
.

126

Wahlestedt
 
C
.
Targeting long non-coding RNA to therapeutically upregulate gene expression
.
Nat Rev Drug Discov
.
2013
;
12
:
433
446
.

127

Ling
 
H
,
Vincent
 
K
,
Pichler
 
M
 et al. .
Junk DNA and the long non-coding RNA twist in cancer genetics
.
Oncogene
.
2015
;
34
:
5003
5011
.

Author notes

Disclosures of potential conflicts of interest may be found at the end of this article.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)