Open Access

Advances in predicting breast cancer driver mutations: Tools for precision oncology (Review)

  • This article is part of the special Issue: Significance of molecular analyses in the era of personalized tumor therapy
  • Authors:
    • Wenhui Hao
    • Barani Kumar Rajendran
    • Tingting Cui
    • Jiayi Sun
    • Yingchun Zhao
    • Thirunavukkarasu Palaniyandi
    • Masilamani Selvam
  • View Affiliations

  • Published online on: October 24, 2024     https://doi.org/10.3892/ijmm.2024.5447
  • Article Number: 6
  • Copyright: © Hao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the modern era of medicine, prognosis and treatment, options for a number of cancer types including breast cancer have been improved by the identification of cancer‑specific biomarkers. The availability of high‑throughput sequencing and analysis platforms, the growth of publicly available cancer databases and molecular and histological profiling facilitate the development of new drugs through a precision medicine approach. However, only a fraction of patients with breast cancer with few actionable mutations typically benefit from the precision medicine approach. In the present review, the current development in breast cancer driver gene identification, actionable breast cancer mutations, as well as the available therapeutic options, challenges and applications of breast precision oncology are systematically described. Breast cancer driver mutation‑based precision oncology helps to screen key drivers involved in disease development and progression, drug sensitivity and the genes responsible for drug resistance. Advances in precision oncology will provide more targeted therapeutic options for patients with breast cancer, improving disease‑free survival and potentially leading to significant successes in breast cancer treatment in the near future. Identification of driver mutations has allowed new targeted therapeutic approaches in combination with standard chemo‑ and immunotherapies in breast cancer. Developing new driver mutation identification strategies will help to define new therapeutic targets and improve the overall and disease‑free survival of patients with breast cancer through efficient medicine.

Introduction

Cancer initiation and progression is a long and complex biological phenomenon caused by any significant alterations in the genome, proteome and chromatin or in any other cellular levels. In total, 10-30% of breast cancer cases are genetically inherited, 5-10% of cases are strongly correlated with hereditary factors and nearly 5% of cases are caused by high penetrance gene mutations such as BRCA1, BRCA2, TP53, CDH1, STK11 and LKB1 (1,2). Among these high-penetrance genes, BRCA1 and BRCA2 are the most crucial genes involved in the regulation of DNA repair, transcription and the cell cycle. Somatic/germline mutations in these two genes are associated with breast cancer and are considered the strongest susceptibility markers that have been identified, with a 45-80% life-time risk in breast cancer in various ethnic and generalized population levels (3-5). A number of these mutations are largely from somatic cells and the majority are neutral/passenger mutations, while certain mutations are more harmful (driver mutations) and give specific cellular advantage, leading to cell proliferation (6-8). Due to the increasing prevalence of high-throughput next-generation sequencing (whole genome, exome and targeted sequencing), the genomic information of thousands of tumors from various cancer types can help researchers to identify and characterize cancer samples in an easier and more robust way (9-11). Besides, increasing the amount of cancer sequencing data is also helpful to find ways to treat patients using multiple approaches. One such approach is driver gene mutation identification and treatment. To date, mutational recurrence in patients is a highly reliable gene marker for driver gene identification (12).

Most driver genes are cancer or subtype specific, so identification of specific cancer drivers is an important step in cancer therapy (13). Additionally, these driver mutations lead to structural and functional consequences that could lead to tumor heterogeneity and drug resistance (14-16). Thus, identifying key driver mutations is a prominent method for disease diagnosis and management. However, identifying those key players is cumbersome with insufficient tumor information (including low coverage and sequence bias), a complex tumor microenvironment, intra/inter-tumor heterogeneity and other biological issues (17). In recent years, several dedicated cancer biology studies have made numerous notable contributions including large cancer sequence depositories such as The Cancer Genome Atlas (TCGA; https://portal.gdc.cancer.gov/), COSMIC database (https://cancer.sanger.ac.uk/cosmic) and International Cancer Genome Consortium (ICGC; https://icgc.org/), and several versatile sequence analysis tools and servers (18). However, conventional treatments and their outcomes are highly limited due to the diversity of patient genome profiles (19,20). Hence, identification of patient-specific treatment plans (precision oncology) is in urgent need for cancer therapy (21,22).

A pharmacogenomics-based treatment strategy is the most advanced and effective. Genetic testing (including DNA sequencing technology) can identify specific mutational alterations related to cancer, which is most likely to be helpful in the development of a patient-specific treatment plan when the patient does not respond to standard therapy. However, chemotherapeutic agents with a narrow therapeutic window and adverse drug toxicities are life-threatening (23). Breast cancer targeted therapies generally target a specific gene or protein and show an improved biological response to the disease with minimal side effects. Precision cancer therapy makes clinical decisions based on the identification of targets using genomic/proteomics data (24). Therefore, cancer treatment will be improved by increasing the amount of tumor genomic data, including mutation, methylation and expression data (25). One of the standard precision oncology approaches is treating patients with cancer based on subtyping (26). Besides, targeting the most actionable, identified and reported driver gene mutations in several cancer samples will help to treat patients in the new paradigm of breast cancer precision medicine (27-29). Furthermore, several additional metrics are needed to further identify driver genes for understanding precision oncology treatment (Fig. 1). In the present review, several breast cancer-associated driver genes, existing strategies in driver gene identification, actionable targets, various existing challenges and applications of precision oncology in breast cancer prognosis and treatment are covered.

Computational identification of breast cancer driver mutations

The identification of breast cancer drivers is the initial step in targeted therapy. Cancer driver identification strategies are evolving, and several tools are being developed, including sequence-based cancer driver prediction tools such as MutSigCV (30), Mutation Set Enrichment Analysis (31), OncodriveFML (32), OncodriveCLUST (33), MuSiC2 (34) and ActiveDriver (35) Similarly, several tools have been developed to predict mutation consequences at the protein level including Sorting Intolerant From Tolerant (SIFT) (36), Polymorphism Phenotyping v2 (PolyPhen-2) (37), CanDrA (38), CHASM (39) and MutationAssessor (40). Several breast cancer drivers are identified and classified based on their occurrence in cancer, their histological and molecular functions and regulatory properties (41). Most cancer drivers have an oncogene, tumor suppressor or dual gene role (42). Even a single mutation in a driver gene may cause diverse effects and show differential tumorigenic and drug response potentials in patients with cancer (43).

Additionally, several computational programs and servers contribute to identifying key driver mutations for precision oncology. Along with germline mutations, numerous somatic variants are being detected by several efficient tools including MuTect (https://github.com/broadinstitute/mutect), VarScan (http://varscan.sourceforge.net/), GATK variant calling pipeline (https://software.broadinstitute.org/gatk/), Torrent variant caller (http://coolgenes.cahe.wsu.edu/ion-docs/Torrent-Variant-Caller-Plugin.html), DeepVariant (https://github.com/google/deepvariant) and Strelka (https://github.com/Illumina/strelka). These tools facilitate the identification of somatic mutations in a robust manner. HotSpot3D (https://github.com/ding-lab/hotspot3d), Cancer3D (http://cancer3d.org/), AlloDriver (https://mdl.shsmu.edu.cn/ALD/), SGDriver (44), CLUMPs (45) and 20/20+ (46) are being used to predict mutational impacts at the 3D structural and conformational level.

Actionable breast cancer driver mutations and the available drugs

Genome and proteome level data are screened for mutations and their corresponding protein level impacts are assessed using high-throughput technologies. However, only <10% of mutations are actionable, hence targeting only actionable mutations may not be beneficial in certain patients and thus lead to a poor response to therapy (47). Several public oncogenomic databases such as TCGA (48), IGC and cBioportal (www.cbioportal.org) provide large scale multi-level information that can be used in research to facilitate disease prognosis, prevention and drug discovery (49). Several targetable breast cancer mutations including oncogenic, truncating, amplifications and fusions have been identified, and these mutations and their corresponding U.S. Food and Drug Administration (FDA)-approved drugs are listed in Table I and detailed information about key gene mutations and targeted drugs can be accessed using OncoKB, a data resource for precision oncology (www.oncokb.org).

Table I

Key actionable breast cancer mutations and their FDA approved drugs.

Table I

Key actionable breast cancer mutations and their FDA approved drugs.

GenesMutationsFDA approved drugs
NTRK1G595RLarotrectinib
NTRK3G623RLarotrectinib
BRAFK601, L597, G464, G469A, G469R and G469VPLX8394
CDK12Truncating mutationsCemiplimab, nivolumab and pembrolizumab
CDKN2AOncogenic mutationsRibociclib, palbociclib and abemaciclib
FGFR1Oncogenic mutationsBGJ398, AZD4547, erdafitinib and Debio1347
FGFR2Oncogenic mutationsErdafitinib, BGJ398, AZD4547 and Debio1347
FGFR3Oncogenic mutationsErdafitinib, Debio1347, BGJ398 and AZD4547
KRASOncogenic mutationsCobimetinib, trametinib and binimetinib
METFusions Crizotinib
MTOROncogenic mutationsTemsirolimus and everolimus
NF1Oncogenic mutationsTrametinib and cobimetinib
PTENOncogenic mutationsAZD8186 and GSK2636771
AKT1E17KAZD5363
ERBB2Oncogenic mutationsNeratinib
ESR1Oncogenic mutationsFulvestrant and AZD9496
PIK3CAOncogenic mutationsFulvestrant + copanlisib
PIK3CAOncogenic mutationsGDC-0077
BRCA1Oncogenic mutationsTalazoparib
BRCA1Oncogenic mutationsOlaparib
BRCA2Oncogenic mutationsTalazoparib
BRCA2Oncogenic mutationsOlaparib
ERBB2AmplificationTrastuzumab + lapatinib (or each as a monotherapy), neratinib, ado-trastuzumab emtansine, pertuzumab + trastuzumab, trastuzumab + tucatinib + capecitabine and trastuzumab deruxtecan
NTRK1FusionsLarotrectinib and entrectinib
NTRK2FusionsLarotrectinib and entrectinib
NTRK3FusionsLarotrectinib and entrectinib
PIK3CAOncogenic mutationsAlpelisib + ulvestrant

[i] FDA, U.S. Food and Drug Administration.

Trastuzumab is a widely tested drug against breast cancer and understanding the action and resistance of this drug will help to develop new viable therapeutic approaches (50). Several key pathways such as the AKT, mTOR, PIK3CA and cell regulation pathways, and numerous variants of tyrosine kinase receptors are targeted for drug discovery and development. A study has demonstrated that the resistance mechanism of trastuzumab is potentially caused by the insulin-like growth factor I (IGF-1) receptor and mutation of IGF-1 shows a significant level of drug resistance against trastuzumab (51). A high percentage of somatic mutations in TP53, PI3KCA, PTEN and AKT have been identified in breast cancer. Several large level mutational landscape studies have paved the way to identify subgroup-specific sensitivities in these pathways (52,53). Of the hotspot mutations identified in the most commonly mutated gene, PIK3CA (~25%), 80-90% of mutations occur in exon9 (E545K/E542K) and three hotspot mutations occur in exon 20 (54).

Targetable kinase family driver mutations and multi-kinase inhibitors in breast cancer precision therapy

The epidermal growth factor receptor (EGFR) is another proven significant target in several cancer types, and a number of inhibitors are designed and used for EGFR-specific gene mutations including gefitinib, erlotinib, trastuzumab and afatinib (55-58). Similarly, in breast cancer, the tyrosine-kinase activity domain of HER2 is prone to pathogenic mutations. Several key HER2 activating somatic mutations have been identified, including G309A, D769H/Y, V777L, P780ins, V842i and R896C (59). Tumors harboring the T798M HER2 mutation can be treated with kinase inhibitors such as lapatinib or trastuzumab (60). Similarly, other HER2 mutations including the L755S, T798I and L869R mutations and several duplication events including S310, V777 and Y772-A775dup are treated with neratinib (61-63). By contrast, a known EGFR inhibitor, gefitinib, showed differential response based on EGFR heterogeneity in triple negative breast cancer (TNBC) (64). CDK4 and CDK6 inhibitors are administered to treat hormone receptor positive breast cancer. A study showed that, cyclin E1 gene amplification and RB transcriptional corepressor 1 (RB1) loss in T47D cell lines results in resistance to CDK4/6 inhibitors, and multiple mutations in RB1 in metastatic breast cancer show resistance to CDK4/6 inhibitors (65,66).

Precision oncology approaches for DNA repair defect mutations in breast cancer

Identification of DNA repair defects and their mutational events could help to identify personalized treatment strategies and targets. One such example is BRCA1/BRCA2 mutations or loss, which lead to deficiency in homologous recombination and genomic instability (1,67). BRCA1 mutations are proportionately higher in TNBC subtypes with several crucial gene mutations considered to be a major risk in young women and crucial for the scientific community for disease prevention and treatment (68). Therefore, identifying and characterizing BRCA1/2 functions and mutations may help to design personalized approaches to treat patients with breast cancer. Poly-(ADP ribose) polymerase 1 (PARP1) functions as a DNA damage sensor for both single and double-stranded DNA breaks and PARP2 is also responsible for base-excision DNA repair through homo and heterodimerization with PARP1; thus, these two proteins play a significant role in maintaining genomic stability through DNA repair mechanisms (69,70). Deleterious mutations in BRCA genes are highly sensitive to PARP1 inhibitors and DNA alkylating agents (71). PARP1 inhibitors intensely reduce DNA single and double-stranded breaks in BRCA1/2-deficient tumors, resulting in improved sensitivity to DNA damaging agents such as cisplatin and PARP1 inhibitors, which are typically administered in BRCA mutation-associated breast and ovarian cancer (72).

Trabectedin is another inhibitor recently approved in Europe and North Korea for the treatment of soft tissue sarcomas including breast, ovarian, prostate and other solid tumors. Trabectedin functions by targeting the minor grooves of DNA, bending the DNA toward the major grooves through which it increases therapeutic efficiency by blocking transcription coupled nucleotide excision repair machinery, leading to cell death (73-75). A previous study demonstrated that the PARP1 inhibitor, olaparib, combined with cediranib potentially inhibits homology-directed DNA repair via BRCA1/2 and RAD51 downregulation and significantly improves progression-free survival (76,77). A list of drugs used for DNA repair defects at various levels of clinical trials are listed in Table II (74,78-88). These inhibitors mainly target DNA repair pathways in BRCA1/2 mutant/deficient breast cancer.

Table II

List of clinical studies in DNA repair defects mutations and their outcomes.

Table II

List of clinical studies in DNA repair defects mutations and their outcomes.

InterventionsTrial IDStudy nameStatusCancer type/subtype
Cisplatin + rucaparibNCT01074970PARP Inhibition for Triple Negative Breast Cancer (ER/PR/HER2)With BRCA1/2 MutationsCompletedBreast Cancer
Gemcitabine + carboplatin + BSI-201NCT00813956A Phase 2 Study of Standard Chemotherapy Plus BSI-201 (a PARP Inhibitor) in the Neoadjuvant Treatment of Triple Negative Breast CancerCompletedTriple negative breast cancer
AZ2281 + carboplatinNCT01445418AZD2281 Plus Carboplatin to Treat Breast and Ovarian CancerCompletedBreast and ovarian Cancer
AZD2171 + fulvestrantNCT00454805AZD2171 in Addition to Fulvestrant in Patients With Advanced Breast CancerCompletedAdvanced breast cancer
PARP inhibitor 2X-121NCT03562832Investigation of Anti-tumour Effect and Tolerability of the PARP Inhibitor 2X-121 in Patients With Metastatic Breast Cancer Selected by the 2X-121 DRPRecruitingMetastatic breast cancer
TalazoparibNCT03990896Evaluation of Talazoparib, a PARP Inhibitor, in Patients With Somatic BRCA Mutant Metastatic Breast Cancer: Genotyping Based Clinical TrialRecruitingBreast cancer
RucaparibNCT03911453Window of Opportunity Trial, PARP Inhibitor Rucaparib Affect on PD-L1 Expression in Triple Negative Breast TumorsRecruitingBreast cancer
Talazoparib + Sacituzumab GovitecanNCT04039230Study to Evaluate Sacituzumab Govitecan in Combination With Talazoparib in Patients With Metastatic Breast CancerRecruitingBreast cancer
Niraparib + TrastuzumabNCT03368729Niraparib in Combination With Trastuzumab in Metastatic HER2+ Breast CancerRecruitingMetastatic breast+ cancer and HER2 breast carcinoma
Olaparib + Paclitaxel and CarboplatinNCT03150576Platinum and Polyadenosine 5′Diphosphoribose Polymerisation (PARP) Inhibitor for Neoadjuvant Treatment of Triple Negative Breast Cancer (TNBC) and/or Germline BRCA (gBRCA) Positive Breast CancerRecruitingBreast cancer
LynparzaNCT04041128PARP Inhibition During Pre-surgical Window in Breast/Ovary CancerRecruitingOvarian and breast cancer

Targeting breast cancer driver mutations by immunotherapy

Several breast and other cancer drivers can be treated using different strategies, including combination therapy (double or triple combination), by targeting more than one genetic event (mutations/mutations plus copy number events or mutations plus upregulation), which improves antitumor potential (89-91). The efficacy of immunotherapies are tested with positive outcomes in both primary and metastatic tumors and are the most potent alternatives to the cytotoxic chemo- and radiotherapies (92). Immunotherapy enhances both progression-free and overall survival and prevents disease recurrence in patients with breast cancer by targeting specific genes or pathways. Checkpoint inhibition is a known approach used in cancer treatment, which targets certain checkpoint molecules such as programmed cell death protein 1, programmed death-ligand 1 (PD-L1) and CTLA4 (93,94). Atezolizumab is an FDA approved PD-L1 antibody for the treatment of metastatic TNBC along with other cancer types (95). Trastuzumab is the first antibody used for the treatment of metastatic breast cancer with a gene amplification or upregulation of CD340 and HER2 (96). At present, several anti-HER2 inhibitors including afatinib, lapatinib, gefitinib and neratinib are used alone or in combination with several monoclonal antibodies and chemotherapeutic agents (97). A list of monoclonal antibodies and combined treatments administered for several breast cancer subtypes are listed in Table SI (88,98-122). In recent years, resistance against a number of monoclonal and combination therapies has been observed, hence antibody-drug conjugates (ADCs) have been established to overcome this drug resistance. A T-cell bispecific antibodies approach and an ADC-based FDA-approved drug combination (ado-trastuzumab emtansine) are the most constructive approaches for the treatment of patients with breast cancer (123).

Challenges and applications of precision oncology in breast cancer

Overall, ~10% of mutations in breast cancer are deemed actionable, highlighting a significant challenge in the realm of precision oncology. Several vital factors determine tumor growth, immune escape and survival. The T-cell response is the most crucial for identifying tumor cells from the normal cell population to produce antitumor immunity (124,125). This immunogenic potentials may vary from one breast cancer subtype to another (126). Drug efficacy is influenced not only by targeted genes but also by various factors, including genetic variability, individual drug performance and mutations that affect drug metabolism. For instance, cytochrome P450 (CYP) pathway members (including CYP3A4, CYP19A and CYP2D6) have been associated with metabolizing anticancer drugs (127). A recent study revealed that HER2+ breast cancer is more responsive to immunotherapy, but estrogen receptor-negative and HER2+ breast cancer has more immunogenic potential (128). Higher expression of estrogen may lower interferon-γ signaling and human leukocyte antigen gene complex-II expression, which facilitates tumor escape from immune action (129). Besides, estrogens are known to be a risk factor for breast cancer by enhancing several key oncogenic growth factors including EGF, IGF, vascular endothelial growth factor, fibroblast growth factor and their corresponding receptors. An estrogen-high tumor microenvironment plays an important immunosuppressive role for the survival of tumor cells in weak immunogenic tumor cells (98,130,131). Hence, targeting these genes and their active mutations may improve breast cancer prognosis and treatment. Similarly, the use of anti-estrogen therapies combined with aromatase inhibitors could be a better approach to improve the further response to immunotherapies (132).

Existing driver mutation prediction approaches and their challenges

The identification of somatic driver genes from germline variants is a crucial step in genomic oncology. In addition to several known germline variants, a growing number of vital somatic variants are being identified. Those somatic variants are validated through modern computational strategies and functional annotation resources including SIFT (36), Polyphen-2 (37), CHASM (39), Mutation Assessor (40), DbNSFP (133) and Mutation Taster (134). However, recent developments in high-throughput techniques and potential computational resources/tools have resulted in very few mutations being clinically actionable. There are major difficulties in differentiating driver from passenger mutations, a lack of strategies to validate genomic variants and challenges associating the clinical relevance of these mutations. Apart from single nucleotide polymorphism, several copy number variations, including copy-number gains and amplifications, and copy-number loss have been identified in breast cancer (135). BRCA1 is a well-known tumor suppressor gene in breast cancer and identifying the key driver genes in BRCA1-associated tumorigenesis will help to predict the road map of this cancer type. In a public sequence repository (cBioportal), ~80 BRCA1-mutated/deficient breast cancer types were found, and the majority of mutations belong to deleterious single nucleotide variations and copy number events, including homozygous deletions or amplifications. Among these mutations TP53 and MYC are the most commonly copy number altered driver genes in BRCA1-associated tumorigenesis and contributing to over 65 and 40% of cases respectively, highlighting their significant roles in cancer progression and potential for targeted therapies (136). The highest number of MYC driver mutations identified in BRCA1-associated tumors was in the TNBC subtype. Additionally, the amplification of MYC along with the copy number amplification of PIK3CA and the loss of copy number in RB1 and PTEN, supports MYC amplification and promotes breast tumorigenesis (137,138). However, due to a low number of cases in this cohort, it is challenging to determine the outcomes of these drivers.

Role of driver genes in breast cancer prognosis and the tumor microenvironment

Along with genomic data for the prediction of breast cancer driver genes, mRNA expression data plays a crucial role in predicting drivers in disease prognosis and their involvement in the tumor microenvironment (139). A recent study revealed a list of differentially expressed breast cancer driver genes to help predict disease prognosis and overall survival (140). The mRNA expression levels of the most enriched driver genes including DDX3X, BRD7, CCR7 and UBE2A are associated with a higher hazard ratio. Several key breast cancer drivers, in conjunction with the tumor microenvironment, significantly influence treatment response in patients with breast cancer. These drivers are responsible for tumor heterogeneity and for varied responses to drug (141). In precision oncology, high-throughput sequencing data including genomics, transcriptomics and proteomics data helps to predict the characteristics of patients and the tumor behavior at the genome/proteome level. Tumor heterogeneity is a prime cause for overall patient survival, disease-free survival and response to chemo- or immunotherapy.

Conclusions and future perspectives

The identification of driver mutations has allowed for new targeted therapeutic approaches in combination with standard chemo- and immunotherapies in breast cancer. Existing drugs for the identified actionable mutations in breast cancer are also used to treat other cancer types; however, whether these drugs are beneficial to other cancer types is still unclear. For example, trastuzumab, which targets HER2 amplification/upregulation, is beneficial to both breast and gastric cancer, while it shows no significant results in lung and ovarian cancer (142,143). Even with the developing modern applications in clinical trial design, challenges continue, including tumor cellularity, intra- and inter-tumor heterogeneity and the tumor microenvironment. Hence, identifying new strategies to overcome these challenges and identifying new therapeutic targets/biomarkers will help to improve the overall and disease-free survival of patients through efficient breast cancer medicine. The present review connects the current strategies with future approaches for identifying novel breast cancer drivers, aiming to aid researchers and ultimately benefit patients. Differential drug responses among breast cancer subtypes influence overall efficacy. Therefore, identifying new driver genes, novel susceptibility regions or loci, and alternative pathways will expedite the discovery of new therapeutic targets. The ultimate goal of breast cancer precision oncology is to identify more therapeutic targets and to increase the drug efficacy while reducing toxicity for patients.

Supplementary Data

Availability of data and materials

Not applicable.

Authors' contributions

WH and BKR designed this study. WH generated the figure. BKR, WH, TP, JS, YZ, MMS and TC performed the background research. BKR and WH drafted and revised the manuscript. All authors contributed to editorial changes in the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This research was supported by the Excellent Young Scientist Foundation of Xinjiang Uyghur Autonomous Region of China (grant no. 2022D01E52) and the Natural Science Foundation of Xinjiang Uygur Autonomous Region (grant no. 2023D01C39).

References

1 

Harvey-Jones E, Raghunandan M, Robbez-Masson L, Magraner-Pardo L, Alaguthurai T, Yablonovitch A, Yen J, Xiao H, Brough R, Frankum J, et al: Longitudinal profiling identifies co-occurring BRCA1/2 reversions, TP53BP1, RIF1 and PAXIP1 mutations in PARP inhibitor resistant advanced breast cancer. Ann Oncol. 35:364–380. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Waghela BN, Vaidya FU, Ranjan K, Chhipa AS, Tiwari BS and Pathak C: AGE-RAGE synergy influences programmed cell death signaling to promote cancer. Mol Cell Biochem. 476:585–598. 2021. View Article : Google Scholar

3 

Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, et al: IFN-ү-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 127:2930–2940. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Bhaskaran SP, Huang T, Rajendran BK, Guo M, Luo J, Qin Z, Zhao B, Chian J, Li S and Wang SM: Ethnic-specific BRCA1/2 variation within Asia population: evidence from over 78 000 cancer and 40 000 non-cancer cases of Indian, Chinese, Korean and Japanese populations. J Med Genet. 58:752–759. 2021. View Article : Google Scholar

5 

Yuan H, Xiu L, Li N, Li Y, Wu L and Yao H: PARPis response and outcome of ovarian cancer patients with BRCA1/2 germline mutation and a history of breast cancer. J Gynecol Oncol. 35:e512024. View Article : Google Scholar : PubMed/NCBI

6 

Ranjan K and Pathak C: Expression of cFLIPL Determines the Basal Interaction of Bcl-2 With Beclin-1 and Regulates p53 Dependent Ubiquitination of Beclin-1 During Autophagic Stress. J Cell Biochem. 117:1757–1768. 2016. View Article : Google Scholar

7 

Ranjan K, Hedl M, Sinha S, Zhang X and Abraham C: Ubiquitination of ATF6 by disease-associated RNF186 promotes the innate receptor-induced unfolded protein response. J Clin Invest. 131:e1454722021. View Article : Google Scholar : PubMed/NCBI

8 

Salvadores M and Supek F: Cell cycle gene alterations associate with a redistribution of mutation risk across chromosomal domains in human cancers. Nat Cancer. 5:330–346. 2024. View Article : Google Scholar : PubMed/NCBI

9 

Xie F, Guo W, Wang X, Zhou K, Guo S, Liu Y, Sun T, Li S, Xu Z, Yuan Q, et al: Mutational profiling of mitochondrial DNA reveals an epithelial ovarian cancer-specific evolutionary pattern contributing to high oxidative metabolism. Clin Transl Med. 14:e15232024. View Article : Google Scholar : PubMed/NCBI

10 

Zhu J, Yang J, Chen X, Wang Y, Wang X, Zhao M, Li G, Wang Y, Zhu Y, Yan F, et al: Integrated Bulk and Single-cell RNA sequencing data constructs and validates a prognostic model for non-small cell lung cancer. J Cancer. 15:796–808. 2024. View Article : Google Scholar : PubMed/NCBI

11 

Zhao H, Yu L and Wang L, Yin X, Liu K, Liu W, Lin S and Wang L: Integrated analysis of single-cell and bulk RNA sequencing data reveals immune-related lncRNA-mRNA prognostic signature in triple-negative breast cancer. Genes Dis. 11:571–574. 2024. View Article : Google Scholar

12 

Brown AL, Li M, Goncearenco A and Panchenko AR: Finding driver mutations in cancer: Elucidating the role of background mutational processes. PLoS Comput Biol. 15:e10069812019. View Article : Google Scholar : PubMed/NCBI

13 

Li F, Gao L, Wang P and Hu Y: Identifying cancer specific driver modules using a network-based method. Molecules. 23:11142018. View Article : Google Scholar : PubMed/NCBI

14 

Pala L, Sala I, Pagan E, De Pas T, Zattarin E, Catania C, Cocorocchio E, Rossi G, Laszlo D, Ceresoli G, et al: 'Heterogeneity of treatment effect on patients' long-term outcome according to pathological response type in neoadjuvant RCTs for breast cancer.'. Breast. 73:1036722024. View Article : Google Scholar

15 

Schade E: A differentform for the certification of cause of death. Ned Tijdschr Geneeskd. 130:2310–2312. 1986.In Dutch.

16 

Dagogo-Jack I and Shaw AT: Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 15:81–94. 2018. View Article : Google Scholar

17 

Akinpelu A, Akinsipe T, Avila LA, Arnold RD and Mistriotis P: The impact of tumor microenvironment: unraveling the role of physical cues in breast cancer progression. Cancer Metastasis Rev. 43:823–844. 2024. View Article : Google Scholar : PubMed/NCBI

18 

Pabinger S, Dander A, Fischer M, Snajder R, Sperk M, Efremova M, Krabichler B, Speicher MR, Zschocke J and Trajanoski Z: A survey of tools for variant analysis of next-generation genome sequencing data. Brief Bioinform. 15:256–278. 2014. View Article : Google Scholar :

19 

Phillips KA, Deverka PA, Sox HC, Khoury MJ, Sandy LG, Ginsburg GS, Tunis SR, Orlando LA and Douglas MP: Making genomic medicine evidence-based and patient-centered: A structured review and landscape analysis of comparative effectiveness research. Genet Med. 19:1081–1091. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Ranjan K, Hedl M and Abraham C: The E3 ubiquitin ligase RNF186 and RNF186 risk variants regulate innate receptor-induced outcomes. Proc Natl Acad Sci USA. 118:e20135001182021. View Article : Google Scholar : PubMed/NCBI

21 

Krebs K and Milani L: Translating pharmacogenomics into clinical decisions: Do not let the perfect be the enemy of the good. Hum Genomics. 13:392019. View Article : Google Scholar : PubMed/NCBI

22 

Ding RB, Chen P, Rajendran BK, Lyu X, Wang H, Bao J, Zeng J, Hao W, Sun H, Wong AH, et al: Molecular landscape and subtype-specific therapeutic response of nasopharyngeal carcinoma revealed by integrative pharmacogenomics. Nat Commun. 12:30462021. View Article : Google Scholar : PubMed/NCBI

23 

Nain P, Seth L, Bell AS, Raval P, Sharma G, Bethel M, Sharma G and Guha A: Chemotherapy in Pregnancy: Assessing the safety of adriamycin administration in pregnancy complicated by breast cancer. JACC Case Rep. 28:1021412023. View Article : Google Scholar

24 

Dey N, Williams C, Leyland-Jones B and De P: Mutation matters in precision medicine: A future to believe in. Cancer Treat Rev. 55:136–149. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Rajendran BK and Deng CX: A comprehensive genomic meta-analysis identifies confirmatory role of OBSCN gene in breast tumorigenesis. Oncotarget. 8:102263–102276. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Tsuchida J, Rothman J, McDonald KA, Nagahashi M, Takabe K and Wakai T: Clinical target sequencing for precision medicine of breast cancer. Int J Clin Oncol. 24:131–140. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Ramage KS, Lock A, White JM, Ekins MG, Kiefel MJ, Avery VM and Davis RA: Semisynthesis and Cytotoxic Evaluation of an Ether Analogue Library Based on a Polyhalogenated Diphenyl Ether Scaffold Isolated from a Lamellodysidea Sponge. Mar Drugs. 22:332024. View Article : Google Scholar : PubMed/NCBI

28 

Hyman DM, Taylor BS and Baselga J: Implementing Genome-Driven Oncology. Cell. 168:584–599. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Ranjan K and Pathak C: Expression of FADD and cFLIPL balances mitochondrial integrity and redox signaling to substantiate apoptotic cell death. Mol Cell Biochem. 422:135–150. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES and Getz G: Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 505:495–501. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Jia P, Wang Q, Chen Q, Hutchinson KE, Pao W and Zhao Z: MSEA: Detection and quantification of mutation hotspots through mutation set enrichment analysis. Genome Biol. 15:4892014. View Article : Google Scholar : PubMed/NCBI

32 

Mularoni L, Sabarinathan R, Deu-Pons J, Gonzalez-Perez A and Lopez-Bigas N: OncodriveFML: A general framework to identify coding and non-coding regions with cancer driver mutations. Genome Biol. 17:1282016. View Article : Google Scholar : PubMed/NCBI

33 

Tamborero D, Gonzalez-Perez A and Lopez-Bigas N: OncodriveCLUST: Exploiting the positional clustering of somatic mutations to identify cancer genes. Bioinformatics. 29:2238–2244. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Dees ND, Zhang Q, Kandoth C, Wendl MC, Schierding W, Koboldt DC, Mooney TB, Callaway MB, Dooling D, Mardis ER, et al: MuSiC: Identifying mutational significance in cancer genomes. Genome Res. 22:1589–1598. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Reimand J and Bader GD: Systematic analysis of somatic mutations in phosphorylation signaling predicts novel cancer drivers. Mol Syst Biol. 9:6372013. View Article : Google Scholar : PubMed/NCBI

36 

Ng PC and Henikoff S: Accounting for human polymorphisms predicted to affect protein function. Genome Res. 12:436–446. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Adzhubei I, Jordan DM and Sunyaev SR: Predicting functional effect of human missense mutations using PolyPhen-2. Curr Protoc Hum Genet Chapter. 7:Unit7 20. 2013.

38 

Carter H, Douville C, Stenson PD, Cooper DN and Karchin R: Identifying Mendelian disease genes with the variant effect scoring tool. BMC Genomics. 14(Suppl 3): S32013. View Article : Google Scholar : PubMed/NCBI

39 

Wong WC, Kim D, Carter H, Diekhans M, Ryan MC and Karchin R: CHASM and SNVBox: Toolkit for detecting biologically important single nucleotide mutations in cancer. Bioinformatics. 27:2147–2148. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Reva B, Antipin Y and Sander C: Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 39:e1182011. View Article : Google Scholar : PubMed/NCBI

41 

Rajendran BK and Deng CX: Characterization of potential driver mutations involved in human breast cancer by computational approaches. Oncotarget. 8:50252–50272. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Shen L, Shi Q and Wang W: Double agents: Genes with both oncogenic and tumor-suppressor functions. Oncogenesis. 7:252018. View Article : Google Scholar : PubMed/NCBI

43 

Dong X, Huang D, Yi X, Zhang S, Wang Z, Yan B, Chung Sham P, Chen K and Jun Li M: Diversity spectrum analysis identifies mutation-specific effects of cancer driver genes. Commun Biol. 3:62020. View Article : Google Scholar : PubMed/NCBI

44 

Zhao J, Cheng F and Zhao Z: SGDriver: A novel structural genomics-based approach to prioritize cancer related and potentially druggable somatic mutations. BMC Bioinformatics. 16(suppl 15): P212015. View Article : Google Scholar :

45 

Kamburov A, Lawrence MS, Polak P, Leshchiner I, Lage K, Golub TR, Lander ES and Getz G: Comprehensive assessment of cancer missense mutation clustering in protein structures. Proc Natl Acad Sci USA. 112:E5486–E5495. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Tokheim CJ, Papadopoulos N, Kinzler KW, Vogelstein B and Karchin R: Evaluating the evaluation of cancer driver genes. Proc Natl Acad Sci USA. 113:14330–14335. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Ipe J, Swart M, Burgess KS and Skaar TC: High-Throughput assays to assess the functional impact of genetic variants: A road towards genomic-driven medicine. Clin Transl Sci. 10:67–77. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar

49 

Leyens L, Reumann M, Malats N and Brand A: Use of big data for drug development and for public and personal health and care. Genet Epidemiol. 41:51–60. 2017. View Article : Google Scholar

50 

Pierobon M, Ramos C, Wong S, Hodge KA, Aldrich J, Byron S, Anthony SP, Robert NJ, Northfelt DW, Jahanzeb M, et al: Enrichment of PI3K-AKT-mTOR pathway activation in hepatic metastases from breast cancer. Clin Cancer Res. 23:4919–4928. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Stratton MR, Campbell PJ and Futreal PA: The cancer genome. Nature. 458:719–724. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Korkaya H and Wicha M: Reprogramming of normal stem cells and cancer stem cells by the tumor microenvironment. Nat Rev Cancer. 13:763–776. 2013.

54 

Pipek O, Alpar D, Rusz O, Bodor C, Udvarnoki Z, Medgyes-Horvath A, Csabai I, Szallasi Z, Madaras L, Kahan Z, et al: Genomic Landscape of Normal and Breast Cancer Tissues in a Hungarian Pilot Cohort. Int J Mol Sci. 24:85532023. View Article : Google Scholar : PubMed/NCBI

55 

Nakai K, Hung MC and Yamaguchi H: A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am J Cancer Res. 6:1609–1623. 2016.PubMed/NCBI

56 

Zhao S, Ma Y, Liu L, Fang J, Ma H, Feng G, Xie B, Zeng S, Chang J, Ren J, et al: Ningetinib plus gefitinib in EGFR-mutant non-small-cell lung cancer with MET and AXL dysregulations: A phase 1b clinical trial and biomarker analysis. Lung Cancer. 188:1074682024. View Article : Google Scholar : PubMed/NCBI

57 

Wu G, Chen Q, Lv D, Lin L and Huang J: Pulmonary Adenocarcinoma Patient with Complex Mutations on EGFR benefits from furmonertinib after acquiring gefitinib resistance: A case report. Recent Pat Anticancer Drug Discov. 19:247–252. 2024. View Article : Google Scholar : PubMed/NCBI

58 

Lewis GD, Li G, Guo J, Yu SF, Fields CT, Lee G, Zhang D, Dragovich PS, Pillow T, Wei B, et al: The HER2-directed antibody-drug conjugate DHES0815A in advanced and/or metastatic breast cancer: Preclinical characterization and phase 1 trial results. Nat Commun. 15:4662024. View Article : Google Scholar : PubMed/NCBI

59 

Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, Goel N, Aronson AB, Li S, et al: Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 3:224–237. 2013. View Article : Google Scholar :

60 

Rexer BN, Ghosh R, Na rasanna A, Estrada MV, Chakrabarty A, Song Y, Engelman JA and Arteaga CL: Human breast cancer cells harboring a gatekeeper T798M mutation in HER2 overexpress EGFR ligands and are sensitive to dual inhibition of EGFR and HER2. Clin Cancer Res. 19:5390–5401. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Ben-Baruch NE, Bose R, Kavuri SM, Ma CX and Ellis MJ: HER2-Mutated Breast Cancer Responds to Treatment With Single-Agent Neratinib, a Second-Generation HER2/EGFR Tyrosine Kinase Inhibitor. J Natl Compr Canc Netw. 13:1061–1064. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Hanker AB, Brewer MR, Sheehan JH, Koch JP, Sliwoski GR, Nagy R, Lanman R, Berger MF, Hyman DM, Solit DB, et al: An Acquired HER2(T798I) Gatekeeper Mutation Induces Resistance to Neratinib in a Patient with HER2 mutant-driven breast cancer. Cancer Discov. 7:575–585. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Hyman DM, Piha-Paul SA, Won H, Rodon J, Saura C, Shapiro GI, Juric D, Quinn DI, Moreno V, Doger B, et al: HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature. 554:189–194. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Savage P, Blanchet-Cohen A, Revil T, Badescu D, Saleh SMI, Wang YC, Zuo D, Liu L, Bertos NR, Munoz-Ramos V, et al: A Targetable EGFR-Dependent tumor-initiating program in breast cancer. Cell Rep. 21:1140–1149. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Herrera-Abreu MT, Palafox M, Asghar U, Rivas MA, Cutts RJ, Garcia-Murillas I, Pearson A, Guzman M, Rodriguez O, Grueso J, et al: Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor-Positive Breast Cancer. Cancer Res. 76:2301–2313. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Condorelli R, Spring L, O'Shaughnessy J, Lacroix L, Bailleux C, Scott V, Dubois J, Nagy RJ, Lanman RB, Iafrate AJ, et al: Polyclonal RB1 mutations and acquired resistance to CDK 4/6 inhibitors in patients with metastatic breast cancer. Ann Oncol. 29:640–645. 2018. View Article : Google Scholar

67 

Woodward ER, Lalloo F, Forde C, Pugh S, Burghel GJ, Schlecht H, Harkness EF, Howell A, Howell SJ, Gandhi A and Evans DG: Germline testing of BRCA1, BRCA2, PALB2 and CHEK2 c.1100delC in 1514 triple negative familial and isolated breast cancers from a single centre, with extended testing of ATM, RAD51C and RAD51D in over 400. J Med Genet. 61:385–391. 2023. View Article : Google Scholar : PubMed/NCBI

68 

Belli C, Duso BA, Ferraro E and Curigliano G: Homologous recombination deficiency in triple negative breast cancer. Breast. 45:15–21. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Miao K, Lei JH, Valecha MV, Zhang A, Xu J, Wang L, Lyu X, Chen S, Miao Z, Zhang X, et al: NOTCH1 activation compensates BRCA1 deficiency and promotes triple-negative breast cancer formation. Nat Commun. 11:32562020. View Article : Google Scholar : PubMed/NCBI

70 

McCann KE and Hurvitz SA: Advances in the use of PARP inhibitor therapy for breast cancer. Drugs Context. 7:2125402018. View Article : Google Scholar : PubMed/NCBI

71 

Caron MC, Sharma AK, O'Sullivan J, Myler LR, Ferreira MT, Rodrigue A, Coulombe Y, Ethier C, Gagne JP, Langelier MF, et al: Poly(ADP-ribose) polymerase-1 antagonizes DNA resection at double-strand breaks. Nat Commun. 10:29542019. View Article : Google Scholar : PubMed/NCBI

72 

Bailly C, Thuru X and Quesnel B: Combined cytotoxic chemotherapy and immunotherapy of cancer: Modern times. NAR Cancer. 2:zcaa0022020. View Article : Google Scholar : PubMed/NCBI

73 

van Kesteren Ch, de Vooght MM, Lopez-Lazaro L, Mathot RA, Schellens JH, Jimeno JM and Beijnen JH: Yondelis (trabectedin, ET-743): The development of an anticancer agent of marine origin. Anticancer Drugs. 14:487–502. 2003. View Article : Google Scholar : PubMed/NCBI

74 

Zelek L, Yovine A, Brain E, Turpin F, Taamma A, Riofrio M, Spielmann M, Jimeno J and Misset JL: A phase II study of Yondelis (trabectedin, ET-743) as a 24-h continuous intravenous infusion in pretreated advanced breast cancer. Br J Cancer. 94:1610–1614. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Le Cesne A, Martin-Broto J and Grignani G: A review of the efficacy of trabectedin as second-line treatment of advanced soft tissue sarcoma. Future Oncol. 18(30s): 5–11. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, Delaloge S, Li W, Tung N, Armstrong A, et al: Olaparib for metastatic breast cancer in patients with a Germline BRCA Mutation. N Engl J Med. 377:523–533. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Pujade-Lauraine E, Ledermann JA, Selle F, Gebski V, Penson RT, Oza AM, Korach J, Huzarski T, Poveda A, Pignata S, et al: Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 18:1274–1284. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Kalra M, Tong Y, Jones DR, Walsh T, Danso MA, Ma CX, Silverman P, King MC, Badve SS, Perkins SM and Miller KD: Cisplatin +/− rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer. NPJ Breast Cancer. 7:292021. View Article : Google Scholar

79 

Kaplan AR, Gueble SE, Liu Y, Oeck S, Kim H, Yun Z and Glazer PM: Cediranib suppresses homology-directed DNA repair through down-regulation of BRCA1/2 and RAD51. Sci Transl Med. 11:eaav45082019. View Article : Google Scholar : PubMed/NCBI

80 

Telli ML, Jensen KC, Vinayak S, Kurian AW, Lipson JA, Flaherty PJ, Timms K, Abkevich V, Schackmann EA, Wapnir IL, et al: Phase II study of gemcitabine, carboplatin, and iniparib as neoadjuvant therapy for triple-negative and BRCA1/2 mutation-associated breast cancer with assessment of a tumor-based measure of genomic instability: PrECOG 0105. J Clin Oncol. 33:1895–1901. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Shamseddine AI and Farhat FS: Platinum-based compounds for the treatment of metastatic breast cancer. Chemotherapy. 57:468–487. 2011. View Article : Google Scholar

82 

Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, et al: Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 434:917–921. 2005. View Article : Google Scholar : PubMed/NCBI

83 

Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ and Helleday T: Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 434:913–917. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Hyams DM, Chan A, de Oliveira C, Snyder R, Vinholes J, Audeh MW, Alencar VM, Lombard J, Mookerjee B, Xu J, et al: Cediranib in combination with fulvestrant in hormone-sensitive metastatic breast cancer: A randomized Phase II study. Invest New Drugs. 31:1345–1354. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Litton JK, Rugo HS, Ettl J, Hurvitz SA, Goncalves A, Lee KH, Fehrenbacher L, Yerushalmi R, Mina LA, Martin M, et al: Talazoparib in patients with advanced breast cancer and a germline BRCA Mutation. N Engl J Med. 379:753–763. 2018. View Article : Google Scholar : PubMed/NCBI

86 

Ettl J, Quek RGW, Lee KH, Rugo HS, Hurvitz S, Goncalves A, Fehrenbacher L, Yerushalmi R, Mina LA, Martin M, et al: Quality of life with talazoparib versus physician's choice of chemotherapy in patients with advanced breast cancer and germline BRCA1/2 mutation: patient-reported outcomes from the EMBRACA phase III trial. Ann Oncol. 29:1939–1947. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Bindra RS, Gibson SL, Meng A, Westermark U, Jasin M, Pierce AJ, Bristow RG, Classon MK and Glazer PM: Hypoxia-induced down-regulation of BRCA1 expression by E2Fs. Cancer Res. 65:11597–11604. 2005. View Article : Google Scholar : PubMed/NCBI

88 

Kumar M, Ranjan K, Singh V, Pathak C, Pappachan A and Singh DD: Hydrophilic Acylated Surface Protein A (HASPA) of Leishmania donovani: Expression, Purification and Biophysico-Chemical Characterization. Protein J. 36:343–351. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Liu ZB, Zhang L, Bian J and Jian J: Combination strategies of checkpoint immunotherapy in metastatic breast cancer. Onco Targets Ther. 13:2657–2666. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Kroemer G and Zitvogel L: Cancer immunotherapy in 2017: The breakthrough of the microbiota. Nat Rev Immunol. 18:87–88. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, Seliger B and Marincola FM: Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer. 81:116–129. 2017. View Article : Google Scholar : PubMed/NCBI

92 

Wang Y, Xu Z, Wu KL, Yu L, Wang C, Ding H, Gao Y, Sun H, Wu YH, Xia M, et al: Siglec-15/sialic acid axis as a central glyco-immune checkpoint in breast cancer bone metastasis. Proc Natl Acad Sci USA. 121:e23129291212024. View Article : Google Scholar : PubMed/NCBI

93 

Krasniqi E, Barchiesi G, Pizzuti L, Mazzotta M, Venuti A, Maugeri-Sacca M, Sanguineti G, Massimiani G, Sergi D, Carpano S, et al: Immunotherapy in HER2-positive breast cancer: state of the art and future perspectives. J Hematol Oncol. 12:1112019. View Article : Google Scholar : PubMed/NCBI

94 

Pardoll DM: The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 12:252–264. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Sharmni Vishnu K, Win TT, Aye SN and Basavaraj AK: Combined atezolizumab and nab-paclitaxel in the treatment of triple negative breast cancer: A meta-analysis on their efficacy and safety. BMC Cancer. 22:11392022. View Article : Google Scholar : PubMed/NCBI

96 

Darvin P, Toor SM, Sasidharan Nair V and Elkord E: Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp Mol Med. 50:1–11. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Garcia-Aranda M and Redondo M: Immunotherapy: A challenge of breast cancer treatment. Cancers (Basel). 11:18222019. View Article : Google Scholar : PubMed/NCBI

98 

Garcia-Aranda M and Redondo M: Protein kinase targets in breast cancer. Int J Mol Sci. 18:25432017. View Article : Google Scholar : PubMed/NCBI

99 

Treilleux I, Blay JY, Bendriss-Vermare N, Ray-Coquard I, Bachelot T, Guastalla JP, Bremond A, Goddard S, Pin JJ, Barthelemy-Dubois C and Lebecque S: Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res. 10:7466–7474. 2004. View Article : Google Scholar : PubMed/NCBI

100 

Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL and Banham AH: Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol. 24:5373–5380. 2006. View Article : Google Scholar : PubMed/NCBI

101 

Gobert M, Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, Biota C, Doffin AC, Durand I, Olive D, et al: Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res. 69:2000–2009. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Mackall CL, Fleisher TA, Brown MR, Magrath IT, Shad AT, Horowitz ME, Wexler LH, Adde MA, McClure LL and Gress RE: Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood. 84:2221–2228. 1994. View Article : Google Scholar : PubMed/NCBI

103 

Guckel B, Stumm S, Rentzsch C, Marme A, Mannhardt G and Wallwiener D: A CD80-transfected human breast cancer cell variant induces HER-2/neu-specific T cells in HLA-A*02-matched situations in vitro as well as in vivo. Cancer Immunol Immunother. 54:129–140. 2005. View Article : Google Scholar

104 

Morse MA, Chaudhry A, Gabitzsch ES, Hobeika AC, Osada T, Clay TM, Amalfitano A, Burnett BK, Devi GR, Hsu DS, et al: Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother. 62:1293–1301. 2013. View Article : Google Scholar : PubMed/NCBI

105 

Kouloulias VE, Dardoufas CE, Kouvaris JR, Gennatas CS, Polyzos AK, Gogas HJ, Sandilos PH, Uzunoglu NK, Malas EG and Vlahos LJ: Liposomal doxorubicin in conjunction with reirradiation and local hyperthermia treatment in recurrent breast cancer: A phase I/II trial. Clin Cancer Res. 8:374–382. 2002.PubMed/NCBI

106 

Morse MA, Hobeika AC, Osada T, Serra D, Niedzwiecki D, Lyerly HK and Clay TM: Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood. 112:610–618. 2008. View Article : Google Scholar : PubMed/NCBI

107 

Meredith R, Torgue J, Shen S, Fisher DR, Banaga E, Bunch P, Morgan D, Fan J and Straughn JM Jr: Dose escalation and dosimetry of first-in-human α radioimmunotherapy with 212Pb-TCMC-trastuzumab. J Nucl Med. 55:1636–1642. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Bernal-Estevez DA, Garcia O, Sanchez R and Parra-Lopez CA: Monitoring the responsiveness of T and antigen presenting cell compartments in breast cancer patients is useful to predict clinical tumor response to neoadjuvant chemotherapy. BMC Cancer. 18:772018. View Article : Google Scholar : PubMed/NCBI

109 

Wiseman C, Presant C, Rao R and Smith J: Clinical responses to intralymphatic whole-cell melanoma vaccine augmented by in vitro incubation with alpha-interferon. Ann N Y Acad Sci. 690:388–391. 1993. View Article : Google Scholar : PubMed/NCBI

110 

Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, et al: Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 17:4550–4557. 2011. View Article : Google Scholar : PubMed/NCBI

111 

Adams S, Kozhaya L, Martiniuk F, Meng TC, Chiriboga L, Liebes L, Hochman T, Shuman N, Axelrod D, Speyer J, et al: Topical TLR7 agonist imiquimod can induce immune-mediated rejection of skipn metastases in patients with breast cancer. Clin Cancer Res. 18:6748–6757. 2012. View Article : Google Scholar : PubMed/NCBI

112 

Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, Nisenbaum H, Pasha T, Xu M, Fox KR, et al: Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res. 67:1842–1852. 2007. View Article : Google Scholar : PubMed/NCBI

113 

Koski GK, Koldovsky U, Xu S, Mick R, Sharma A, Fitzpatrick E, Weinstein S, Nisenbaum H, Levine BL, Fox K, et al: A novel dendritic cell-based immunization approach for the induction of durable Th1-polarized anti-HER-2/neu responses in women with early breast cancer. J Immunother. 35:54–65. 2012. View Article : Google Scholar

114 

Sharma A, Koldovsky U, Xu S, Mick R, Roses R, Fitzpatrick E, Weinstein S, Nisenbaum H, Levine BL, Fox K, et al: HER-2 pulsed dendritic cell vaccine can eliminate HER-2 expression and impact ductal carcinoma in situ. Cancer. 118:4354–4362. 2012. View Article : Google Scholar : PubMed/NCBI

115 

Garnett CT, Schlom J and Hodge JW: Combination of docetaxel and recombinant vaccine enhances T-cell responses and antitumor activity: Effects of docetaxel on immune enhancement. Clin Cancer Res. 14:3536–3544. 2008. View Article : Google Scholar : PubMed/NCBI

116 

Mohebtash M, Tsang KY, Madan RA, Huen NY, Poole DJ, Jochems C, Jones J, Ferrara T, Heery CR, Arlen PM, et al: A pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancer. Clin Cancer Res. 17:7164–7173. 2011. View Article : Google Scholar : PubMed/NCBI

117 

Hodge JW, Sabzevari H, Yafal AG, Gritz L, Lorenz MG and Schlom J: A triad of costimulatory molecules synergize to amplify T-cell activation. Cancer Res. 59:5800–5807. 1999.PubMed/NCBI

118 

Berinstein NL, Karkada M, Morse MA, Nemunaitis JJ, Chatta G, Kaufman H, Odunsi K, Nigam R, Sammatur L, MacDonald LD, et al: First-in-man application of a novel therapeutic cancer vaccine formulation with the capacity to induce multi-functional T cell responses in ovarian, breast and prostate cancer patients. J Transl Med. 10:1562012. View Article : Google Scholar : PubMed/NCBI

119 

Robbins PF, Eggensperger D, Qi CF and Schlom J: Definition of the expression of the human carcinoembryonic antigen and non-specific cross-reacting antigen in human breast and lung carcinomas. Int J Cancer. 53:892–897. 1993. View Article : Google Scholar : PubMed/NCBI

120 

Madan RA, Arlen PM and Gulley JL: PANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther. 7:543–554. 2007. View Article : Google Scholar : PubMed/NCBI

121 

Kwa M, Li X, Novik Y, Oratz R, Jhaveri K, Wu J, Gu P, Meyers M, Muggia F, Speyer J, et al: Serial immunological parameters in a phase II trial of exemestane and low-dose oral cyclophosphamide in advanced hormone receptor-positive breast cancer. Breast Cancer Res Treat. 168:57–67. 2018. View Article : Google Scholar

122 

Rios-Doria J, Durham N, Wetzel L, Rothstein R, Chesebrough J, Holoweckyj N, Zhao W, Leow CC and Hollingsworth R: Doxil synergizes with cancer immunotherapies to enhance antitumor responses in syngeneic mouse models. Neoplasia. 17:661–670. 2015. View Article : Google Scholar : PubMed/NCBI

123 

Nejadmoghaddam MR, Minai-Tehrani A, Ghahremanzadeh R, Mahmoudi M, Dinarvand R and Zarnani AH: Antibody-Drug Conjugates: Possibilities and Challenges. Avicenna J Med Biotechnol. 11:3–23. 2019.PubMed/NCBI

124 

Vonderheide RH, Domchek SM and Clark AS: Immunotherapy for breast cancer: What are we missing? Clin Cancer Res. 23:2640–2646. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Zhang X, Kim S, Hundal J, Herndon JM, Li S, Petti AA, Soysal SD, Li L, McLellan MD, Hoog J, et al: Breast cancer neoantigens can induce CD8(+) T-Cell responses and antitumor immunity. Cancer Immunol Res. 5:516–523. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Ayoub NM, Al-Shami KM and Yaghan RJ: Immunotherapy for HER2-positive breast cancer: recent advances and combination therapeutic approaches. Breast Cancer (Dove Med Press). 11:53–69. 2019.PubMed/NCBI

127 

Olopade OI, Grushko TA, Nanda R and Huo D: Advances in breast cancer: Pathways to personalized medicine. Clin Cancer Res. 14:7988–7999. 2008. View Article : Google Scholar : PubMed/NCBI

128 

Uma K and Jan FS: HER2 in breast cancer: A review and update. Adv Anat Pathol. 21:100–107. 2014. View Article : Google Scholar

129 

Mostafa AA, Codner D, Hirasawa K, Komatsu Y, Young MN, Steimle V and Drover S: Activation of ERα signaling differentially modulates IFN-ү induced HLA-class II expression in breast cancer cells. PLoS One. 9:e873772014. View Article : Google Scholar

130 

Rothenberger NJ, Somasundaram A and Stabile LP: The role of the estrogen pathway in the tumor microenvironment. Int J Mol Sci. 19:6112018. View Article : Google Scholar : PubMed/NCBI

131 

Makhoul I, Atiq M, Alwbari A and Kieber-Emmons T: Breast cancer immunotherapy: An update. Breast Cancer (Auckl). 12:11782234187748022018. View Article : Google Scholar : PubMed/NCBI

132 

Johnston SR, Martin LA, Leary A, Head J and Dowsett M: Clinical strategies for rationale combinations of aromatase inhibitors with novel therapies for breast cancer. J Steroid Biochem Mol Biol. 106:180–186. 2007. View Article : Google Scholar : PubMed/NCBI

133 

Liu X, Li C, Mou C, Dong Y and Tu Y: dbNSFP v4: A comprehensive database of transcript-specific functional predictions and annotations for human nonsynonymous and splice-site SNVs. Genome Med. 12:1032020. View Article : Google Scholar : PubMed/NCBI

134 

Steinhaus R, Proft S, Schuelke M, Cooper DN, Schwarz JM and Seelow D: MutationTaster2021. Nucleic Acids Res. 49(W1): W446–W451. 2021. View Article : Google Scholar : PubMed/NCBI

135 

Shahrouzi P, Forouz F, Mathelier A, Kristensen VN and Duijf PHG: Copy number alterations: A catastrophic orchestration of the breast cancer genome. Trends Mol Med. 30:750–764. 2024. View Article : Google Scholar : PubMed/NCBI

136 

Annunziato S, de Ruiter JR, Henneman L, Brambillasca CS, Lutz C, Vaillant F, Ferrante F, Drenth AP, van der Burg E, Siteur B, et al: Comparative oncogenomics identifies combinations of driver genes and drug targets in BRCA1-mutated breast cancer. Nat Commun. 10:3972019. View Article : Google Scholar : PubMed/NCBI

137 

Kaysudu I, Gungul TB, Atici S, Yilmaz S, Bayram E, Guven G, Cizmecioglu NT, Sahin O, Yesiloz G, Haznedaroglu BZ and Cizmecioglu O: Cholesterol biogenesis is a PTEN-dependent actionable node for the treatment of endocrine therapy-refractory cancers. Cancer Sci. 114:4365–4375. 2023. View Article : Google Scholar : PubMed/NCBI

138 

Lu Y, Dong K, Yang M and Liu J: Network pharmacology-based strategy to investigate the bioactive ingredients and molecular mechanism of Evodia rutaecarpa in colorectal cancer. BMC Complement Med Ther. 23:4332023. View Article : Google Scholar : PubMed/NCBI

139 

Pranav P, Palaniyandi T, Baskar G, Ravi M, Rajendran BK, Sivaji A and Ranganathan M: Gene expressions and their significance in organoid cultures obtained from breast cancer patient-derived biopsies. Acta Histochem. 124:1519102022. View Article : Google Scholar : PubMed/NCBI

140 

Du XW, Li G, Liu J, Zhang CY, Liu Q, Wang H and Chen TS: Comprehensive analysis of the cancer driver genes in breast cancer demonstrates their roles in cancer prognosis and tumor microenvironment. World J Surg Oncol. 19:2732021. View Article : Google Scholar : PubMed/NCBI

141 

Liu X, Jin G, Qian J, Yang H, Tang H, Meng X and Li Y: Digital gene expression profiling analysis and its application in the identification of genes associated with improved response to neoadjuvant chemotherapy in breast cancer. World J Surg Oncol. 16:822018. View Article : Google Scholar : PubMed/NCBI

142 

Martin V, Cappuzzo F, Mazzucchelli L and Frattini M: HER2 in solid tumors: More than 10 years under the microscope; where are we now? Future Oncol. 10:1469–1486. 2014. View Article : Google Scholar : PubMed/NCBI

143 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2025
Volume 55 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hao W, Rajendran BK, Cui T, Sun J, Zhao Y, Palaniyandi T and Selvam M: Advances in predicting breast cancer driver mutations: Tools for precision oncology (Review). Int J Mol Med 55: 6, 2025.
APA
Hao, W., Rajendran, B.K., Cui, T., Sun, J., Zhao, Y., Palaniyandi, T., & Selvam, M. (2025). Advances in predicting breast cancer driver mutations: Tools for precision oncology (Review). International Journal of Molecular Medicine, 55, 6. https://doi.org/10.3892/ijmm.2024.5447
MLA
Hao, W., Rajendran, B. K., Cui, T., Sun, J., Zhao, Y., Palaniyandi, T., Selvam, M."Advances in predicting breast cancer driver mutations: Tools for precision oncology (Review)". International Journal of Molecular Medicine 55.1 (2025): 6.
Chicago
Hao, W., Rajendran, B. K., Cui, T., Sun, J., Zhao, Y., Palaniyandi, T., Selvam, M."Advances in predicting breast cancer driver mutations: Tools for precision oncology (Review)". International Journal of Molecular Medicine 55, no. 1 (2025): 6. https://doi.org/10.3892/ijmm.2024.5447