Skip to main content

Main menu

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Other Publications
    • cbm

User menu

  • My alerts

Search

  • Advanced search
Cancer Biology & Medicine
  • Other Publications
    • cbm
  • My alerts
Cancer Biology & Medicine

Advanced Search

 

  • Home
  • About
    • About CBM
    • Editorial Board
    • Announcement
  • Articles
    • Ahead of print
    • Current Issue
    • Archive
    • Collections
    • Cover Story
  • For Authors
    • Instructions for Authors
    • Resources
    • Submit a Manuscript
  • For Reviewers
    • Become a Reviewer
    • Instructions for Reviewers
    • Resources
    • Outstanding Reviewer
  • Subscription
  • Alerts
    • Email Alerts
    • RSS Feeds
    • Table of Contents
  • Contact us
  • Follow cbm on Twitter
  • Visit cbm on Facebook
Review ArticleReview
Open Access

The treatment of breast cancer in the era of precision medicine

Jingwen Bai, Yiyang Gao and Guojun Zhang
Cancer Biology & Medicine April 2025, 22 (4) 322-347; DOI: https://doi.org/10.20892/j.issn.2095-3941.2024.0510
Jingwen Bai
1The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yiyang Gao
2Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
3Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Guojun Zhang
1The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
2Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
3Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Guojun Zhang
  • For correspondence: [email protected]
  • Article
  • Figures & Data
  • Info & Metrics
  • References
  • PDF
Loading

Abstract

The management of breast cancer, one of the most common and heterogeneous malignancies, has transformed with the advent of precision medicine. This review explores current developments in genetic profiling, molecular diagnostics, and targeted therapies that have revolutionized breast cancer treatment. Key innovations, such as cyclin-dependent kinases 4/6 (CDK4/6) inhibitors, antibody-drug conjugates (ADCs), and immune checkpoint inhibitors (ICIs), have improved outcomes for hormone receptor-positive (HR+), HER2-positive (HER2+), and triple-negative breast cancer (TNBC) subtypes remarkably. Additionally, emerging treatments, such as PI3K inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, and mRNA-based therapies, offer new avenues for targeting specific genetic mutations and improving treatment response, particularly in difficult-to-treat breast cancer subtypes. The integration of liquid biopsy technologies provides a non-invasive approach for real-time monitoring of tumor evolution and treatment response, thus enabling dynamic adjustments to therapy. Molecular imaging and artificial intelligence (AI) are increasingly crucial in enhancing diagnostic precision, personalizing treatment plans, and predicting therapeutic outcomes. As precision medicine continues to evolve, it has the potential to significantly improve survival rates, decrease recurrence, and enhance quality of life for patients with breast cancer. By combining cutting-edge diagnostics, personalized therapies, and emerging treatments, precision medicine can transform breast cancer care by offering more effective, individualized, and less invasive treatment options.

keywords

  • Breast cancer
  • precision medicine
  • diagnostic precision
  • personalized therapy

Introduction

Breast cancer is among the most prevalent and biologically diverse malignancies globally, and accounts for a substantial portion of cancer diagnoses and deaths among women1. Despite decades of research and advancements in screening and treatment, breast cancer remains a leading cause of cancer-related mortality in women1. The disease is characterized by its heterogeneity, including various subtypes with differing tumor biology, prognosis, and response to treatment. In 2000, Perou et al.2 introduced molecular subtyping, classifying breast cancer into luminal-like, HER2-enriched, basal-like, and normal-like types. This landmark discovery facilitated the development of subtype-specific therapies, such as endocrine therapy (ET) for luminal-like tumors and anti-HER2 therapy for HER2-enriched tumors. Subsequently, gene expression analysis by Sørlie et al.3 further refined the luminal-like subtype by dividing it into 2 distinct subgroups, luminal A and luminal B, on the basis of differences in prognosis and treatment response. The 2011 St. Gallen Conference built on these findings by establishing an immunohistochemical classification system for streamlining clinical application, on the basis of markers such as estrogen receptor (ER), progesterone receptor (PR), HER2, and Ki-674. Historically, breast cancer treatment was relatively uniform, relying on standard protocols such as surgery, chemotherapy (ChT), and radiotherapy, with limited consideration of individual tumor biology. This “one-size-fits-all” approach often resulted in suboptimal outcomes, particularly for patients with aggressive or advanced disease.

The advent of precision medicine has revolutionized breast cancer treatment by offering a personalized approach5–7. Precision medicine involves tailoring treatments according to the genetic, molecular, and environmental factors unique to each patient’s cancer8. This approach transcends the traditional “one-size-fits-all” approach to therapy, by using specific biomarkers, genetic testing, and advanced technologies to guide treatment decisions. The shift from conventional treatment paradigms to precision-based strategies has significantly improved patient outcomes, particularly regarding survival and quality of life9. Advances in molecular diagnostics10 and genetic profiling11 have been central to this transformation, by enabling the identification of key biomarkers and actionable mutations that can guide treatment decisions. In particular, the ability to classify tumors according to specific genetic and molecular features, such as HER2-low expression12 and mutations in BRCA1/213–15, has expanded therapeutic options and enabled more targeted interventions.

Recent breakthroughs in technologies such as next-generation sequencing (NGS) have facilitated deeper understanding of tumor biology and enabled the identification of novel molecular targets16. These discoveries led to the development of targeted therapies, including cyclin-dependent kinases 4/6 (CDK4/6) inhibitors17, antibody-drug conjugates (ADCs)18, and immune checkpoint inhibitors (ICIs)19, which have shown considerable promise in overcoming treatment resistance and improving outcomes in advanced and metastatic breast cancer. For example, CDK4/6 inhibitors have been shown to delay disease progression in hormone receptor-positive (HR+) breast cancer20. Simultaneously, ADCs such as trastuzumab deruxtecan have demonstrated efficacy in HER2-low and HER2-ultralow breast cancer21,22, a subtype that previously had limited treatment options. ICIs, particularly in triple-negative breast cancer (TNBC), are also opening new avenues for treatment23,24 and offering hope for patients with a subtype historically associated with poor prognosis.

The integration of diagnostic tools, such as genetic profiling and liquid biopsies, has markedly enhanced the precision medicine approach in breast cancer care. Genetic profiling assays, including Oncotype DX25,26 and MammaPrint27,28, offer valuable insights into recurrence risk and potential ChT benefits, and empower clinicians to make more informed treatment decisions. Liquid biopsies provide critical information through analysis of circulating tumor cells (CTCs)29 and circulating tumor-derived materials such as circulating tumor DNA (ctDNA)30, circulating miRNA31, and extracellular vesicles32. Together, these tools enhance personalized treatment strategies, support the early detection of therapeutic resistance, and increase the likelihood of timely and effective interventions.

This review explores the current state of precision medicine and therapies for female breast cancer, focusing on advanced treatments and diagnostics and future developments in this field.

Precision medicine in early breast cancer (EBC)

In recent years, breast cancer mortality rates have declined in many countries, particularly among younger populations, primarily because of advancements in earlier detection and precision therapy strategies33. However, despite these improvements, breast cancer remains the leading cause of cancer-related deaths among women worldwide1, including in China34. Most patients with breast cancer are diagnosed in early stages (stage I–III) and undergo potentially curative treatments. These treatments typically include surgery followed by systemic treatments such as neoadjuvant and/or adjuvant ChT, ET, radiotherapy, or combinations of these approaches. The Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) conducted a comprehensive analysis of patients with EBC diagnosed between 1990 and 2009, on the basis of their extensive database35. The risk of distant recurrence among patients with EBC diagnosed after the year 2000 was approximately 20% lower than the risk of those diagnosed in the 1990s. This improvement is attributed to advancements in diagnostic and treatment methods, particularly the development of adjuvant therapies. Next, we discuss recent advancements in systemic treatment for EBC.

HR+/HER2-negative (HER2–) EBC

HR+/HER2− breast cancer, the most common subtype, represents approximately 60%–70% of all breast cancer cases36. This subtype responds well to ET, and ChT may be used in high-risk patients. In the past decade, substantial advancements in precision medicine have reshaped the treatment landscape for HR+/HER2− EBC by providing new therapeutic options aligned with patients’ tumor biology and individual risk factors. These options include extended ET, CDK4/6 inhibitors, and poly (ADP-ribose) polymerase (PARP) inhibitors, all of which decrease recurrence and improve overall survival (OS)37.

ET is the essential treatment for HR+/HER2− EBC

ET, a core treatment approach for HR+/HER2− EBC, leverages the hormone dependence of these tumors to curb growth and decrease recurrence risk. This therapy targets hormonal pathways that drive tumor proliferation and has been found to significantly improve long-term outcomes for patients38.

For premenopausal women with HR+/HER2− EBC at low risk, the ER selective modulator tamoxifen is often used as the primary ET without ChT39–41. However, for high-risk patients with HR+/HER2− EBC, tamoxifen alone might have diminished effectiveness in patients who have retained ovarian function, because their ovaries continue to produce estrogen. In these patients, ovarian function suppression (OFS) with gonadotropin-releasing hormone analogues, such as leuprolide, can help decrease estrogen production and enhance tamoxifen’s effects42,43. The Suppression of Ovarian Function Trial (SOFT) and Tamoxifen and Exemestane Trial (TEXT) evaluated OFS combined with tamoxifen or exemestane in premenopausal women44,45. The SOFT trial found that tamoxifen with OFS, compared with tamoxifen alone, improved outcomes, particularly in high-risk women. The TEXT trial showed that exemestane plus OFS achieved better outcomes than tamoxifen plus OFS among high-risk premenopausal women, thereby supporting the use of aromatase inhibitors (AIs) in this group. For postmenopausal women, ET often includes AIs such as anastrozole, letrozole, and exemestane. AIs block aromatase, an enzyme responsible for converting androgens into estrogen, and thereby effectively decrease estrogen levels in the body and consequently the stimulation of HR+ breast cancer cells, which depend on estrogen for growth46. AIs therapy has been shown to decrease recurrence rates, particularly in postmenopausal women with high recurrence risk, and to have greater efficacy than tamoxifen47,48.

Extending ET beyond the standard 5 years has been another key strategy for decreasing long-term recurrence risk in patients with HR+/HER2− EBC. Trials such as MA.1R have shown that extending AIs therapy beyond 5 years significantly decreases recurrence, particularly in high-risk patients, such as those with lymph node involvement49. However, this benefit must be weighed against the potential for adverse effects, including bone density loss and increased fracture risk. Consequently, extended ET is considered on a case-by-case basis, through shared decision-making between clinicians and patients to balance the benefits and risks. Interestingly, a multigene prognostic genomic assay, Breast Cancer Index (BCI), helps predict recurrence risk and guide the duration of ET in patients with HR+ EBC with high hormone receptor/insulin-like growth factor (H/I) ratios, who significantly benefit from extended treatment50.

Ongoing studies are also exploring selective estrogen receptor degraders51, such as fulvestrant52 and elacestrant53,54, which have shown promise in metastatic HR+ breast cancer and are currently being tested in high-risk early-stage disease.

These advancements highlight the importance of personalized ET in improving outcomes in patients with in HR+/HER2− EBC by tailoring treatments to specific tumor characteristics and patient profiles.

CDK4/6 inhibitors improve prognosis for HR+/HER2− EBC with high recurrence risk

Despite the effectiveness of ET, many patients with HR+/HER2− EBC experience recurrence, particularly those with high-risk features, such as large tumors or multiple lymph node involvement55. Genetic profiling has revealed that these patients almost always maintain retinoblastoma function; therefore, the primary pathway targeted by CDK4/6 inhibitors remains intact56. In addition, CCND1, which encodes cyclin D1, is a direct target gene of the ER and consequently is frequently overexpressed in ER-positive cancers57. Therefore, the addition of CDK4/6 inhibitors has been a major advancement. CDK4/6 inhibitors, primarily palbociclib, abemaciclib, ribociclib, and dalpiciclib, inhibit CDK4/6’s activity and halt cancer cell growth at a critical point in the cell cycle, thus preventing proliferation.

Palbociclib

The first CDK4/6 inhibitor approved for HR+/HER2− advanced breast cancer (ABC), palbociclib, has become standard in metastatic settings20,37. However, in the early-stage setting, the PALLAS trial, evaluating palbociclib with ET in stage II or III HR+/HER2− EBC, has indicated that this therapy has no significant benefit over ET alone in preventing recurrence58 (Table 1). Similarly, the PENELOPE-B trial, assessing palbociclib plus ET in patients with high-risk HR+/HER2− EBC with residual disease after neoadjuvant ChT, compared with ET alone, did not improve invasive disease-free survival (iDFS)59 (Table 1). Despite these early-stage setbacks, palbociclib remains promising in metastatic settings, and ongoing research is exploring its full potential in EBC.

View this table:
  • View inline
  • View popup
Table 1

Trials on CDK4/6 inhibitors in patients with HR+/HER2– EBC

Ribociclib

Ribociclib has shown efficacy in both metastatic and early-stage HR+/HER2− breast cancer. The NATALEE trial demonstrated significant improvements in iDFS with the addition of ribociclib to ET for early-stage HR+/HER2− breast cancer, including node-positive cases61 (Table 1). With a favorable safety profile, including low rates of neutropenia and gastrointestinal adverse effects, ribociclib has broad applicability, including for patients with low-risk disease, and therefore may benefit a wide population.

Abemaciclib

The most recent CDK4/6 inhibitor approved for adjuvant treatment in HR+/HER2− EBC, abemaciclib, has achieved significant improvements in iDFS for high-risk patients. In the MONARCH-E trial, abemaciclib decreased the risk of distant recurrence by 30.4%, and was found to benefit patients with high-risk features such as elevated Ki-67 levels or large tumors60,64,65 (Table 1). Continuous administration of abemaciclib, in contrast to other CDK4/6 inhibitors, enhances treatment efficacy but also increases the incidence of gastrointestinal adverse effects, notably diarrhea, which requires careful management64,66. Despite these challenges, abemaciclib remains a critical treatment option for patients with high-risk HR+/HER2− EBC, particularly those with node-positive or aggressive disease.

Dalpiciclib

Dalpiciclib, although widely used in regions such as Asia for advanced HR+/HER2− breast cancer, is not currently approved for EBC treatment in many areas. Studies in metastatic settings have demonstrated its efficacy in prolonging progression-free survival (PFS) when combined with ET, similarly to the other CDK4/6 inhibitors67,68. Its potential use in EBC remains under exploration in clinical trials aimed at determining its efficacy in preventing recurrence and improving outcomes in patients with high-risk EBC62,63 (Table 1). The distinct characteristics of dalpiciclib, such as its lower rates of gastrointestinal adverse effects than abemaciclib, may make it a future candidate for broader applications in metastatic and early-stage settings.

Each CDK4/6 inhibitor offers unique benefits and challenges in treating HR+/HER2− breast cancer. Abemaciclib is currently a leading candidate because of its effectiveness in patients with high-risk early-stage tumors, particularly those with aggressive biology. Ribociclib provides a favorable balance of efficacy and safety, and therefore is suitable for a broader patient population. Although palbociclib and dalpiciclib remain under evaluation in early-stage disease, both remain crucial in the management of metastatic HR+/HER2− breast cancer.

Exploration of immunotherapy for high-risk HR+/HER2− EBC

Immunotherapy, which was traditionally focused on treating TNBC, is currently being explored in HR+/HER2− breast cancer, particularly for high-risk patients. ICIs are being tested with standard therapies to improve immune responses in patients with HR+/HER2− tumors, which typically exhibit relatively low immune activity.

Key trials, including KEYNOTE-756 and CheckMate 7FL, have shown promising results. KEYNOTE-756, a phase III trial evaluating pembrolizumab combined with neoadjuvant ET and ChT, has demonstrated improved pathological complete response (pCR) rates69 (Table 2). Similarly, the CheckMate 7FL trial, investigating nivolumab with neoadjuvant ChT followed by adjuvant ET, has shown improvements in pCR and decreases in residual cancer burden, and offered hope for patients with high-risk HR+/HER2− tumors70 (Table 2). Although immunotherapy for HR+/HER2− breast cancer remains in its early stages, these trials suggest that ICIs might enhance the effectiveness of existing therapies; therefore, further research is warranted to identify the optimal combinations and the patient populations that would benefit most.

View this table:
  • View inline
  • View popup
Table 2

Trials on ICIs in early breast cancer

PARP inhibitors are recommended for high-risk BRCA1/2-mutated EBC

For patients with germline BRCA1 or BRCA2 mutations, PARP inhibitors, such as olaparib, provide an effective adjuvant treatment option. The OlympiA trial has demonstrated that olaparib decreases recurrence risk and improves OS in patients with high-risk BRCA-mutated EBC, thus reaffirming its role in precision medicine for this subgroup76–78.

HER2-positive (HER2+) EBC

Approximately 15%–20% of breast cancers are classified as HER2+79. This subtype is more aggressive than the HER2− subtype. In recent years, breast cancer subtypes have been found to exist not only within the traditional HER2+ and HER2− categories but also within a spectrum including HER2-low and HER2-ultra-low cancers80. These distinctions are aiding in further refinement of treatment strategies, particularly through the use of precision medicine-tailored treatments based on tumor molecular characteristics. Key elements of precision medicine for HER2+ EBC are described below.

Dual HER2 blockade has superior efficacy to single HER2 blockade

A significant breakthrough in treating HER2+ EBC has been dual HER2 blockade combining trastuzumab and pertuzumab. The APHINITY trial highlighted that adding pertuzumab to trastuzumab and ChT significantly improves iDFS, particularly in patients with high-risk, lymph node-positive tumors81. At 6-year follow-up, patients treated with this dual blockade experienced 4.5% greater iDFS and 24% lower recurrence risk or death rates than those who did not receive pertuzumab (HR 0.76)82. Furthermore, the third interim OS of the APHINITY trial, updated in 202483, showed that pertuzumab addition resulted in an absolute 4.9% improvement in 8-year iDFS in the node-positive cohort, whereas no iDFS benefit from pertuzumab was observed in the node-negative cohort. Additionally, no evidence of late-onset cardiac toxicity associated with pertuzumab addition was found, thus providing critical clinical insights for the management of patients with HER2+ EBC.

Additionally, the FDChina study further refined dual HER2 blockade by evaluating a fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection84. This approach achieved the same efficacy as intravenous administration in total pCR rates while simplifying the treatment process, decreasing patient burden, and maintaining a similar safety profile84. This innovation offers patients a more convenient treatment option without compromising outcomes.

ADCs are recommended for patients with HER2+ EBC who do not achieve pCR

Trastuzumab emtansine (T-DM1) has become a foundational therapy for patients with HER2+ EBC who do not achieve pCR after neoadjuvant treatment (Table 3). The critical KATHERINE trial has shown that T-DM1, compared with trastuzumab, significantly decreases the risk of invasive disease recurrence or death by 50.0%, achieving a 3-year iDFS rate of 88.3% for T-DM1 vs. 77.0% for trastuzumab alone85. In 2024, 8.4-year follow-up data further confirmed T-DM1’s sustained benefit, with an iDFS rate of approximately 81.0%, compared with 67.0% for trastuzumab86. This benefit extends across various subgroups, including patients with differing nodal and HR status. Additionally, updated OS data have shown a 34% decrease in mortality risk of patients treated with T-DM186.

View this table:
  • View inline
  • View popup
Table 3

Currently approved ADCs for HER2-positive breast cancer

Moreover, trastuzumab deruxtecan (T-DXd) has shown substantial potential in HER2-low EBC. The TALENT trial, designed to evaluate the neoadjuvant use of T-DXd, either alone or in combination with ET, reported highly promising early results in 202321. The overall response rate was 75% in patients receiving T-DXd monotherapy and 63.2% in those receiving T-DXd combined with anastrozole21. Additionally, neoadjuvant administration of T-DXd is under evaluation in the DESTINY-Breast11 trial in locally advanced or inflammatory HER2+ breast cancer (NCT05113251). Adjuvant use of T-DXd is currently being explored in the ongoing DESTINY-Breast05 trial, in which its effectiveness is being compared with that of T-DM1 in patients with residual invasive HER2+ breast cancer after neoadjuvant therapy (NCT04622319). Furthermore, the SHAMROCK study will assess the use of neoadjuvant T-DXd in early-stage HER2+ breast cancer by incorporating strategies for both escalating and de-escalating therapy87.

These advances in ADCs, both in HER2+ and HER2-low breast cancer, reflect the growing influence of precision medicine in providing more tailored and effective treatments that improve long-term outcomes for patients across the HER2 spectrum.

Tyrosine kinase inhibitors (TKIs) are recommended for patients with HER2+ EBC at high recurrence risk after completion of standard HER2-targeted therapies

TKIs, such as lapatinib, tucatinib, and neratinib, are used primarily in the adjuvant setting for HER2+ breast cancer. However, they have also been explored in the neoadjuvant setting in some trials. Neratinib is approved for extended adjuvant therapy in HER2+ EBC, particularly in patients who have completed trastuzumab-based treatment. The ExteNET trial has demonstrated that 1 year of neratinib significantly improves iDFS, particularly in HR+ patients, and decreases the risk of distant recurrence88. This treatment is recommended after completion of trastuzumab in the adjuvant setting, to minimize recurrence. Although TKIs are less frequently used in the neoadjuvant setting, some studies have investigated their potential. For example, lapatinib has been explored as a neoadjuvant option in combination with trastuzumab and ChT in HER2+ breast cancer. The NeoALTTO trial has demonstrated that adding lapatinib to trastuzumab before surgery (neoadjuvant therapy) achieves a significantly greater pCR rate than trastuzumab alone89. However, owing to mixed results and adverse effects, lapatinib has not gained widespread use in the neoadjuvant setting.

TNBC EBC

TNBC lacks ER, PR, and HER2 expression90, thus limiting the effectiveness of hormonal therapies and HER2-targeted agents widely used in other breast cancer subtypes. Consequently, TNBC has been associated with poor prognosis91,92, high risk of early recurrence93,94, and limited treatment options. However, recent advances in targeted therapies, immunotherapy, and personalized medicine are beginning to shift this paradigm, particularly for patients with high-risk early-stage TNBC. This movement toward individualized care, focusing on the specific biological traits of the tumor and the patient’s genetic profile, has been instrumental in improving outcomes in early TNBC95. The rapid evolution of precision medicine in TNBC offers hope to patients with this aggressive and traditionally challenging subtype.

Immunotherapy improves pCR, EFS, and OS rates in early TNBC

Immunotherapy has emerged as a promising treatment option for TNBC. The primary focus of immunotherapy in early TNBC is on ICIs, particularly anti-PD-1 and anti-PD-L1 therapies, to enhance the immune system’s ability to recognize and destroy cancer cells96.

Pembrolizumab (Keytruda)

Pembrolizumab is among the most studied ICIs in early TNBC. The KEYNOTE-522 trial (Table 2), a phase III study, evaluated pembrolizumab in combination with neoadjuvant ChT in early-stage, high-risk TNBC. This treatment significant improved pCR rates (64.8% in the pembrolizumab group vs. 51.2% in the ChT-only group)71. The trial also demonstrated improved event-free survival (EFS)72; consequently, pembrolizumab has become a cornerstone of early TNBC treatment. The final OS results of the KEYNOTE-522 trial were presented in 202473. In patients with early-stage TNBC receiving neoadjuvant therapy, adding pembrolizumab to ChT significantly improved the 5-year OS, from 81.7% to 86.6%. Moreover, long-term benefits of EFS were also observed. These findings further support the efficacy of pembrolizumab.

Atezolizumab (Tecentriq)

Another checkpoint inhibitor, atezolizumab, was evaluated in the IMpassion031 trial (Table 2), in combination with nab-paclitaxel, followed by doxorubicin plus cyclophosphamide as neoadjuvant treatment in early TNBC. Addition of atezolizumab achieved a higher pCR rate (58.0%) than placebo plus ChT (41.0%)74. Consequently, atezolizumab has been established as a potential neoadjuvant option for early-stage TNBC.

Avelumab (Bavencio)

The A-BRAVE trial (Table 2), a phase III study, examined avelumab in the adjuvant setting for patients with early TNBC at high recurrence risk. Although avelumab did not significantly improve DFS, it achieved a meaningful improvement in OS by decreasing the risk of death and distant metastases in high-risk patients. These findings support its potential use in selected patients’ post-neoadjuvant therapy75.

ICIs are part of a growing arsenal against early TNBC. These therapies have shown promise both in neoadjuvant and adjuvant settings, and herald a major shift toward personalized immunotherapy improving outcomes in a subtype of breast cancer with previously limited therapeutic options.

PARP inhibitors are recommended in patients with high-risk early-stage BRCA-mutated TNBC

In 2005, 2 groundbreaking studies showed that synthetic lethality can be exploited in BRCA1/2-deficient tumors: cells lacking homologous recombination are selectively killed when PARP is inhibited97,98. In 2009, the phase I trial of the PARP inhibitor olaparib demonstrated objective responses in patients with advanced breast, ovarian, and prostate cancers bearing germline BRCA1/2 mutations99. Subsequent trials also indicated substantial promise of PARP inhibitors in treating patients with early TNBC with BRCA1/2 mutations. The OlympiA trial demonstrated that 1 year of olaparib treatment significantly improved both iDFS and OS in patients with high-risk early-stage BRCA-mutated TNBC77. Olaparib decreased the risk of disease recurrence and death, and was the first PARP inhibitor demonstrated to achieve an OS benefit in this setting. The trial results emphasize the importance of PARP inhibitors in personalizing treatment for patients with TNBC.

Precision medicine in early TNBC has transformed the treatment landscape by incorporating immunotherapy, PARP inhibitors, and ADCs, and surpassing traditional ChT in improving survival and recurrence. These therapies, which target the unique biology of TNBC and subgroups, are moving the field closer to personalized treatment options that extend beyond standard ChT. Although challenges remain, such as understanding resistance mechanisms and identifying optimal combination therapies, these advances provide a more hopeful outlook for patients with TNBC, particularly those at high recurrence risk.

Continued research on the tumor microenvironment, genetic mutations, and emerging biomarkers should further refine these approaches and enable greater personalization in the future. For patients with early TNBC, these precision medicine strategies signify a shift toward more effective, targeted care aimed at decreasing recurrence, improving survival, and profoundly affecting treatment for this challenging breast cancer subtype.

Precision medicine in ABC

HR+ ABC

HR+ ABC typically responds well to ETs, which are foundational in its management. However, because resistance mechanisms frequently develop over time, additional targeted therapies are frequently required.

ETs provide an initial therapy for ABC

In clinical practice, ETs are favored for their ability to manage advanced disease with a relatively favorable adverse effect profile, in contrast to ChT. These treatments are valuable for patients requiring long-term management, because they can be administered over extended periods to control disease progression.

ET is often used as a monotherapy in patients with indolent disease, or in combination with targeted therapies such as CDK4/6 inhibitors in patients with high-risk features or those who exhibit resistance to first-line therapies. Extending the duration of ET in patients who continue to benefit is a common approach, provided that the patient tolerates the treatment well.

Despite the broad application of ETs, clinical challenges arise from the development of resistance, particularly in patients with mutations, such as ESR1100. For such cases, novel therapies, such as oral selective estrogen receptor degraders (e.g., fulvestrant52 and elacestrant53,54), provide new treatment avenues when traditional endocrine therapies fail. The EMERALD trial has demonstrated that elacestrant significantly improves PFS in patients with ESR1-mutant ABC53. Therefore, this treatment may provide an essential option in cases in which tumors no longer respond to standard ETs101.

CDK4/6 inhibitors combined with ET are the standard treatment for HR+ ABC

CDK4/6 inhibitors represent a major advancement in treating HR+ ABC, particularly for patients who experience disease progression under ET. When combined with ET, these inhibitors—palbociclib, ribociclib, abemaciclib, and the emerging dalpiciclib—have achieved substantial improvements in PFS and OS102 (Table 4).

View this table:
  • View inline
  • View popup
Table 4

Trials on CDK4/6 inhibitors in patients with hormone receptor-positive, HER2-negtive ABC

Palbociclib, ribociclib, and abemaciclib are combined with ETs, particularly for patients with high-risk disease or significant tumor burdens. Clinical trials including PALOMA-2105,106 and MONALEESA-2109,110 have demonstrated that adding CDK4/6 inhibitors to letrozole extends PFS by 10–11 months beyond ET alone. These inhibitors have become standard care for advanced HR+ breast cancer.

Dalpiciclib, a newer addition to the CDK4/6 inhibitor class, has shown promise in improving outcomes for patients with HR+ ABC. Clinical studies such as the DAWNA-1 trial have demonstrated that, when combined with ET, this treatment significantly prolongs PFS, particularly in patients with endocrine-resistant or heavily pretreated disease67. Additionally, dalpiciclib exhibits a manageable safety profile, in which neutropenia is a notable but manageable adverse effect67. As ongoing trials evaluate its efficacy and potential applications63,68, dalpiciclib offers another valuable option that broadens the available therapeutic strategies for managing HR+ ABC.

PI3K/AKT/mTOR inhibitors are effective options for endocrine-resistant HR+ ABC

Targeting the PI3K/AKT/mTOR pathway is critical in managing HR+ ABC, particularly for patients who develop resistance to ETs because of mutations in the PIK3CA gene119,120. This pathway is a central component in cellular growth, metabolism, and survival, and its dysregulation is associated with resistance mechanisms in HR+ breast cancer, thus contributing to disease progression despite standard treatments121.

PI3K inhibitors, including alpelisib and inavolisib, have emerged as key therapeutic agents for overcoming resistance in HR+ breast cancer driven by PIK3CA mutations122. Alpelisib, a selective PI3K inhibitor, has notable efficacy in PIK3CA-mutant HR+ breast cancer, and its combination with ET has become a common treatment approach123. In the seminal SOLAR-1 trial, patients with PIK3CA-mutant HR+ breast cancer who received alpelisib plus fulvestrant experienced a significant improvement in PFS over that observed in patients receiving ET alone124. Similarly, inavolisib, another selective PI3K inhibitor, has shown efficacy in combination with aromatase inhibitors, by significantly extending PFS in patients with PIK3CA-mutant tumors125. These inhibitors provide essential options for patients with endocrine-resistant disease, and offer an alternative strategy for targeting the PI3K pathway and delaying disease progression.

AKT inhibitors, such as capivasertib, offer a distinct approach targeting the PI3K/AKT/mTOR pathway126. Capivasertib has shown promising results in combination with ET, by improving PFS in patients with disease progression under prior treatments127,128. As an AKT inhibitor, capivasertib directly targets a downstream component of the PI3K pathway, thus providing an alternative therapeutic option for patients with HR+ breast cancer resistant to other treatments.

Another agent, everolimus, is an mTOR inhibitor that targets a downstream component of the PI3K/AKT/mTOR pathway, thereby offering an alternative mechanism to combat resistance to CDK4/6 inhibitor and ET129,130. Everolimus is frequently used alongside exemestane in advanced HR+ breast cancer, particularly in patients with disease progression under prior ETs. The BOLERO-2 trial has emphasized the clinical benefits of adding everolimus, which significantly extended PFS, particularly in heavily pretreated patients who would otherwise have had limited therapeutic options131. This combination is valuable for patients with advanced disease, because it not only helps manage resistance but also maintains quality of life by delaying progression.

These targeted therapies provide essential options for managing HR+ breast cancer and addressing the critical need for effective treatments in patients with endocrine-resistant disease driven by PI3K pathway mutations.

ADCs provide promising treatment options for advanced HR+ breast cancer after multiple therapies fail

For patients whose disease progresses after multiple lines of endocrine and targeted therapies, ADCs provide a new approach132. These therapies deliver cytotoxic ChT directly to tumor cells and consequently minimize systemic exposure133.

T-DXd, initially developed for HER2+ breast cancer, has shown efficacy in HR+ breast cancer with low HER2 expression (Table 3). The DESTINY-Breast04 trial has demonstrated significant improvements in PFS for patients with advanced HR+ disease who have exhausted other treatment options22.

Sacituzumab govitecan (SG), targeting Trop-2, has also shown promising results in heavily pretreated HR+ patients with breast cancer. The ASCENT trial has indicated extended OS and consequently brought hope to patients with few remaining therapeutic options134,135.

ETs remain a fundamental part of managing advanced HR+ breast cancer, particularly in patients with relatively less aggressive disease or those who would benefit from long-term treatment with fewer adverse effects. When ETs are combined with targeted treatments such as CDK4/6 inhibitors, PI3K/AKT/mTOR pathway inhibitors, and ADCs, the outcomes improve significantly. New therapies, such as doruciclib and elacestrant, provide promising solutions for patients with resistant disease, by making precision medicine an essential component of ABC care. As research progresses, refining these therapies and addressing resistance mechanisms will be critical to further improving survival and quality of life for advanced patients with breast cancer.

HER2+ ABC

Since the discovery of HER2/neu136, precision medicine for advanced HER2+ breast cancer continues to make major strides, primarily through the development of targeted therapies such as monoclonal antibodies, ADCs, and small molecule TKIs. These treatments are highly tailored to the molecular characteristics of HER2+ tumors, and have been found to improve survival rates and address resistance mechanisms.

Trastuzumab and pertuzumab provide an initial therapy for metastatic HER2+ breast cancer

Monoclonal antibodies, such as trastuzumab, have been foundational in advanced HER2-positive breast cancer treatment137. Trastuzumab blocks HER2 receptor signaling, and consequently prevents tumor growth and engages the immune system to destroy cancer cells138. Pertuzumab, a monoclonal antibody that blocks HER2 dimerization, is often combined with trastuzumab for dual HER2 blockade139. The CLEOPATRA trial has demonstrated that this combination, along with ChT, significantly improves PFS and OS in patients with metastatic HER2+ breast cancer140. This regimen remains the first-line standard of care for most patients.

ADCs provide a second-line treatment for HER2+ ABC

ADCs represent a major advancement in precision medicine by combining targeted HER2 inhibition with ChT. T-DM1, the first ADC for HER2+ breast cancer, links trastuzumab to a cytotoxic agent that specifically kills HER2+ cancer cells141. The EMILIA trial established T-DM1 as the preferred second-line treatment after trastuzumab, by showing significant improvements in PFS and overall safety142 (Table 3).

More recently, T-DXd, a more potent ADC, has shown superior efficacy. The DESTINY-Breast03 trial has demonstrated that T-DXd achieves a median PFS longer than 25 months, as compared with 7 months for T-DM1; therefore, this treatment is the preferred second-line option143. Furthermore, the DESTINY-Breast04 trial extended T-DXd’s use to patients with HER2-low expression, thus broadening its applicability beyond traditional HER2+ breast cancer22,144. The DESTINY-Breast06 trial evaluated T-DXd in patients with HER2-ultralow breast cancer, and might enable expansion of this potent ADC to a broader patient population145 (Table 3).

TKIs enhance treatment for HER2+ ABC, including brain metastases

In HER2+ ABC, TKIs have become a crucial component of treatment, particularly for patients resistant to traditional HER2-targeted therapies, such as trastuzumab or pertuzumab. TKIs are small molecules that specifically inhibit the tyrosine kinase activity of the HER2 receptor, a protein that drives tumor cell growth and proliferation in HER2+ breast cancer146. By blocking this activity, TKIs effectively disrupt the signaling pathways essential for tumor survival and consequently provide a more targeted therapeutic option.

Lapatinib was among the first TKIs approved for HER2+ breast cancer and is often used in combination with capecitabine147. However, newer TKIs, such as tucatinib, neratinib, and pyrotinib, have since expanded the treatment landscape by providing more potent options with enhanced specificity and efficacy. Tucatinib has shown particularly impressive results in patients with brain metastases common and challenging complications in HER2+ breast cancer, because of its ability to cross the blood-brain barrier more effectively than other HER2-targeted drugs148,149. The HER2CLIMB trial has demonstrated that the combination of tucatinib with trastuzumab and capecitabine significantly improves PFS and OS in patients with HER2+ breast cancer with or without brain metastases150.

Pyrotinib is another potent HER2-targeted TKI that inhibits HER1 and HER4, and consequently broadly disrupts HER family signaling pathways151. Clinical trials such as the PERMEATE152 and PHOEBE153 trials have demonstrated that pyrotinib in combination with capecitabine improves PFS and response rates in patients whose disease previously progressed under other HER2-targeted therapies. This broad-spectrum activity makes pyrotinib a promising option, particularly for patients with brain metastases or those who have experienced disease progression on traditional HER2-targeted agents.

Although TKIs provide potent options, they can have adverse effects, such as diarrhea, rash, and liver enzyme elevations, which often require monitoring and management154. However, the clinical benefits of TKIs, particularly their ability to address CNS involvement and enhance outcomes in heavily pretreated patients, underscore their value in the HER2+ breast cancer treatment landscape. As research continues to explore new TKIs and combination strategies, TKIs are anticipated to play an increasingly prominent role in treating HER2+ ABC, by extending survival and improving quality of life for many patients.

Advanced TNBC

ChT remains a foundational treatment for advanced TNBC155. However, precision medicine is transforming this field by tailoring treatment strategies to the unique molecular characteristics of individual tumors. Unlike other breast cancers, TNBC lacks common therapeutic targets, and its historically restricted treatment options have contributed to its aggressive progression and higher relapse rates. Recent advances in genomics and molecular profiling have led to the discovery of distinct TNBC subtypes, thus facilitating the development of targeted therapies, immunotherapies, and novel drug delivery systems156. These approaches are reshaping TNBC management and bringing new hope for improved outcomes in advanced and metastatic stages.

Immunotherapy has transformed TNBC treatment

Immunotherapy has transformed TNBC treatment, particularly in tumors expressing PD-L1. ICIs, such as pembrolizumab and atezolizumab, enable the immune system to attack cancer cells by blocking the PD-1/PD-L1 pathway157. In the KEYNOTE-355 trial, pembrolizumab combined with ChT has been found to significantly improve PFS in PD-L1-positive metastatic TNBC (9.7 vs. 5.6 months)23 (Table 5). Similarly, in the IMpassion130 trial, atezolizumab combined with nab-paclitaxel has been found to improve PFS in patients with PD-L1-positive TNBC (7.5 vs. 5.0 months)24. These findings further established immunotherapy as a critical tool for TNBC treatment (Table 5).

View this table:
  • View inline
  • View popup
Table 5

Trials on ICIs in ABC

Preliminary results from Future-C-PLUS have indicated promising increases in overall response rate and PFS with this therapy compared with ICI monotherapy, particularly in difficult-to-treat cases (Table 5)158. This combination strategy highlights the potential of “immune-priming” approaches to enhance checkpoint inhibitor efficacy across TNBC subtypes and to expand the benefits of immunotherapy to a broader patient population. The study exemplifies the evolving role of combination therapies in precision medicine for TNBC, and underscores the need for continued research into optimizing and individualizing immunotherapy strategies in this aggressive cancer type.

ADCs are advancing TNBC treatment, improving outcomes in advanced, resistant cases

ADCs, which combine monoclonal antibodies with ChT, have become a promising treatment for TNBC. SG, targeting Trop-2, has been particularly effective159. In the ASCENT trial, SG has been found to improve PFS (5.6 vs. 1.7 months) and OS (12.1 vs. 6.7 months) in patients with heavily pretreated metastatic TNBC134. ADCs, such as trastuzumab deruxtecan, initially used for HER2+ cancer, are being explored for HER2-low TNBC, and have shown promising early results160 that may open new avenues for treatment.

Identifying new targets is essential for advanced TNBC

A key component of precision medicine in TNBC is identifying molecular subtypes that exhibit unique biological characteristics and therapeutic vulnerabilities. The Fudan Classification divides TNBC into 4 primary subtypes: basal-like immune-suppressed, basal-like immune-activated, mesenchymal, and luminal androgen receptor161. Each subtype has distinct gene expression profiles and pathways that can inform tailored treatment approaches. For example, basal-like immune-activated tumors show signs of immune activation with increased immune cell infiltration and therefore are likely to respond to immunotherapies161. In contrast, luminal androgen receptor subtypes express androgen receptor signaling and therefore are candidates for anti-androgen therapies161. By aligning treatments with the molecular characteristics of each subtype, precision medicine can maximize efficacy while minimizing unnecessary adverse effects.

Targeted therapies have become a cornerstone of precision medicine in TNBC. Drugs inhibiting specific signaling pathways, such as the PI3K/AKT/mTOR pathway, are being developed for TNBC subgroups with mutations in genes, such as PIK3CA and PTEN162. For instance, PI3K inhibitors, such as alpelisib, are being studied in patients with TNBC bearing these genetic alterations163. Additionally, PARP inhibitors, including olaparib164,165 and talazoparib166,167, have shown success in patients with TNBC with BRCA1 or BRCA2 mutations. In the OlympiAD trial, olaparib, compared with standard ChT, has been found to significantly improve PFS in patients with BRCA-mutated TNBC (7.0 vs. 4.2 months)165. Similarly, in the EMBRACA trial, talazoparib extended PFS in this subgroup (8.6 vs. 5.6 months), thereby confirming the value of PARP inhibitors in BRCA-mutated TNBC167. These trials underscore the potential of targeted therapies in precision medicine to extend survival and decrease disease burden for specific TNBC subtypes.

Advancements in precision medicine have transformed the treatment of advanced TNBC, by overcoming historical limitations arising from a lack of therapeutic targets. Immunotherapy, remarkably ICIs, have improved survival in PD-L1-positive TNBC, particularly in combination with ChT. ADCs, such as SG, offer promising options for resistant or metastatic disease. Molecular profiling, such as the Fudan classification, enables tailored treatments with targeted therapies, such as PARP and PI3K inhibitors for specific subgroups. These innovations highlight the growing value of precision medicine in optimizing TNBC management and improving outcomes. Ongoing research will be critical to expanding and refining these strategies.

In conclusion, the precision diagnosis and treatment of breast cancer have made remarkable strides in recent decades (Figure 1). Key milestones include the discovery of ER in 196738, the approval of tamoxifen in 197740, and the identification of HER2 in 1984136. The 1990s brought notable breakthroughs, such as the discovery of BRCA1/BRCA2 (1994–1996)13,14 and the approval of trastuzumab (Herceptin) in 1998137. The early 2000s saw the proposal of molecular subtypes in 20002, followed by the launch of multi-gene testing tools, such as MammaPrint in 200227. In the 2010s, advancements continued with the approval of therapies such as olaparib in 2014168 and T-DXd in 2020144, whereas the use of NGS technology16 and pembrolizumab71 further personalized treatment strategies. More recently, the standardization of liquid biopsy technology in 2021169 marked an important step in non-invasive diagnostics. In the future, emerging technologies including artificial intelligence (AI), multi-omics, molecular imaging, and cell therapy are poised to push breast cancer diagnosis and treatment to even more precise and individualized levels.

Figure 1
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1

Key milestones in precision medicine for breast cancer.

The future and emerging frontiers of precision medicine in breast cancer

Although current advancements are reshaping breast cancer diagnosis and treatment, the full potential of precision medicine continues to unfold. Emerging technologies—such as large-scale omics studies170, AI171, molecular imaging172, cell-based therapies173, and mRNA innovations174—are poised to drive transformative breakthroughs that may lead to the next generation of personalized therapies (Figure 2). Below, we explore key directions for the evolution of precision medicine in breast cancer and the exciting frontiers that could become the next breakthrough of cancer treatment.

Figure 2
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2

Precision medicine for breast cancer. This figure illustrates a personalized treatment framework that integrates diagnostic and therapeutic strategies for patients with breast cancer, regardless of metastasis presence. Tissue and blood samples are analyzed with immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), multi-omics approaches (genomics, proteomics, and transcriptomics), and liquid biopsy techniques (ctDNA/CTCs). Molecular imaging is used to visualize tumor markers, whereas artificial intelligence (AI) aids in data interpretation and decision-making. On the basis of the molecular subtypes, personalized therapies are tailored, including endocrine therapies with or without CDK4/6 inhibitors for HR+/HER2− tumors, HER2-targeted therapies (e.g., trastuzumab) for HER2+ tumors, ChT or immunotherapy for TNBC, and innovative cell/mRNA-based therapies.

The next breakthrough of breast cancer treatment in the era of precision medicine

Precision medicine has already revolutionized breast cancer treatment, but the next major breakthrough is likely to emerge from integrating large-scale omics data and advanced AI. Analysis of comprehensive molecular, genetic, and phenotypic data should provide unprecedented understanding of breast cancer biology and enable more precise, personalized interventions.

Large-scale omics studies

  • Genomics: whole-genome sequencing and whole-exome sequencing are expected to become routinely used to uncover rare mutations, identify new driver genes, and enhance understanding of how genetic alterations (e.g., TP53, BRCA1/2, and PIK3CA) drive cancer progression175. These insights should fuel the development of highly targeted therapies that are more effective and less toxic.

  • Proteomics and transcriptomics: mapping of protein expression176–178, post-translational modifications179, and gene expression profiles180 is expected to further reveal breast cancer’s heterogeneity and enable the creation of personalized treatment plans considering the dynamic proteome of a patient’s cancer, rather than its genetic makeup alone.

  • Metabolomics: investigating metabolic reprogramming in breast cancer cells, such as shifts in glycolysis and oxidative phosphorylation, should lead to the identification of new therapeutic targets and precision therapies tailored to the metabolic vulnerabilities of each tumor181–184.

AI and machine learning integration

AI is expected to play a crucial role in analyzing the vast data from omics technologies, medical imaging, and patient histories, thereby providing deeper insights into cancer behavior. Key applications include the following:

  • Predicting treatment response: machine learning models can predict how breast cancer subtypes will respond to specific treatments, thus enabling earlier optimization of therapy and minimizing adverse effects185–187.

  • Identifying novel targets: AI can identify novel drug targets and biomarkers by analyzing complex genetic188,189 and proteomic data190, and can suggest drug repurposing opportunities for specific subtypes of breast cancer191,192.

Emerging therapies: cell therapies and mRNA innovations

Two of the most exciting frontiers in precision medicine are cell-based therapies and mRNA treatments, particularly for advanced and metastatic breast cancer.

Cell therapies

  • Personalized immunotherapy: CAR-T cell therapy, which involves genetically modifying a patient’s T cells to target tumor-specific antigens, has revolutionized treatment of hematological cancers193. For breast cancer, particularly TNBC, tumor-infiltrating lymphocyte (TIL) therapies have substantial promise194. Personalized TIL therapy has the potential to dramatically improve outcomes, particularly in resistant or metastatic cases195.

  • Oncolytic virus therapy: Genetically modified viruses that selectively infect and kill cancer cells are another promising approach. These oncolytic viruses can be engineered to enhance immune responses and can be paired with ICIs, thus providing a multi-pronged strategy to eliminate breast cancer cells195–198.

mRNA therapies

The success of mRNA vaccines for COVID-19 has paved the way to treatments for cancers including breast cancer.

  • mRNA cancer vaccines: These vaccines can be designed to encode tumor-specific antigens that stimulate the immune system to target and destroy cancer cells199. Tailoring vaccines to each patient’s tumor profile offers a form of personalized immunotherapy200,201.

  • mRNA as therapeutic agents: Beyond vaccines, mRNA-based therapies can directly target cancer by delivering genetic instructions to cells, and enabling them to produce proteins that either inhibit tumor growth or trigger cancer cell death202. Although still in the early stages, this approach has substantial promise for treating HR+ and HER2+ breast cancer.

The road to personalized medicine: when and how will it arrive?

Although precision medicine is already influencing breast cancer treatment, fully personalized medicine might require 1 or 2 decades to mature. Key milestones include the following.

Comprehensive genomic profiling

Routine genetic testing, such as NGS of both tumor and normal tissue, is expected to become standard. This testing should help tailor therapies based on somatic mutations and germline mutations, which can influence treatment responses and predispose individuals to cancer203.

Liquid biopsies

Advances in liquid biopsy technology, which analyzes cancer DNA in blood, should allow for non-invasive monitoring of tumor evolution and therapy response169. This monitoring would enable real-time treatment adjustments and early recurrence detection, even before clinical symptoms appear204.

AI-powered personalized treatment plans

AI systems are expected to integrate genetic, molecular, and clinical data to produce fully personalized treatment plans optimizing efficacy and minimizing toxicity205,206. This approach could combine multiple therapies—e.g., ChT, immunotherapy, targeted therapy, and hormone therapy—according to each patient’s unique tumor profile.

Molecular imaging

As precision medicine evolves, molecular imaging is expected to become more integrated into breast cancer diagnosis and treatment207. Advanced imaging technologies would allow for real-time visualization of tumor molecular features, such as specific receptors208–212, mutations213, and metabolic activity214. Techniques such as PET scans, magnetic resonance spectroscopy, and optical imaging are expected to enable early detection of tumors, treatment response monitoring, and tumor evolution tracking.

Precision medicine is shaping the future of breast cancer treatment, driven by breakthroughs in omics studies, AI, molecular imaging, and novel therapies, such as cell-based immunotherapies and mRNA treatments. In the coming years, therapies are expected to become increasingly personalized, through tailoring to each individual’s genetic, molecular, and immune profile. Although the timeline for widespread personalized medicine is uncertain, we are on the cusp of a revolution promising more targeted, effective, and individualized treatments leading to better outcomes and fewer adverse effects for patients.

Conclusions

Precision medicine has fundamentally transformed breast cancer management by recognizing its molecular heterogeneity and tailoring therapies to distinct disease subtypes215. This shift has led to the identification of key mutations, such as BRCA1/2216 and PIK3CA217, driving the development of highly targeted therapies that are more effective and less toxic. Advances in genetic profiling, biomarkers, and technologies, such as liquid biopsies, have significantly enhanced diagnostic accuracy and treatment personalization. Simultaneously, insights into the tumor microenvironment have informed the growth of immunotherapy strategies. These innovations have markedly improved survival rates and quality of life, particularly for patients with challenging subtypes, such as triple-negative and HER2+ breast cancer.

Key advancements in areas including CDK4/6 inhibitors, PARP inhibitors, and ADCs have precisely targeted oncogenic pathways, and improved DFS and OS. The integration of ICIs underscores the increasing importance of immunotherapy in precision oncology. However, challenges such as therapy resistance and the need for reliable predictive biomarkers remain major hurdles in fully harnessing the potential of precision medicine.

Emerging technologies are expected to drive the next wave of breakthroughs. Large-scale omics studies, including genomics, proteomics, and metabolomics, offer comprehensive understanding of breast cancer at the molecular level, and can uncover rare mutations and metabolic pathways that may become therapeutic targets. AI is expected to complement these studies by analyzing complex datasets to predict treatment responses, identify new drug targets, and personalize care for individual patients. Moreover, cell-based therapies, such as CAR-T and TIL therapies, are advancing immunotherapy, particularly for resistant or metastatic cases. Simultaneously, oncolytic viruses offer novel ways to target and destroy cancer cells directly. Moreover, mRNA innovations, inspired by the success of COVID-19 vaccines, promise breakthroughs in personalized cancer vaccines and therapeutic applications, by enabling the immune system to recognize and eliminate tumor-specific antigens218.

Molecular imaging is likely to become increasingly important in precision medicine, by allowing real-time monitoring of tumor behavior and treatment responses207. This monitoring would enable dynamic treatment adjustments and enhanced personalization of therapies, particularly in detecting resistance early and tracking tumor evolution219. By integrating AI with molecular imaging, clinicians can create more accurate, individualized treatment plans that optimize efficacy while minimizing adverse effects.

In summary, precision medicine has become a cornerstone of modern breast cancer treatment, by offering more effective, less invasive, and highly personalized care. With the rapid evolution of omics studies, AI, cell-based therapies, mRNA technologies and molecular imaging, the field promises to deliver transformative outcomes that improve survival rates and quality of life for patients at every stage of the disease.

Conflict of interest statement

No potential conflicts of interest are disclosed.

Author contributions

Conceived and designed the review: Guojun Zhang.

Collected the data: Jingwen Bai, Yiyang Gao.

Wrote the paper: Jingwen Bai, Yiyang Gao.

Reviewed and revised the paper: Guojun Zhang.

Footnotes

  • ↵*These authors contributed equally to this work.

  • Received November 8, 2024.
  • Accepted March 5, 2025.
  • Copyright: © 2025, The Authors

This work is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License.

References

  1. 1.↵
    1. Bray F,
    2. Laversanne M,
    3. Sung H,
    4. Ferlay J,
    5. Siegel RL,
    6. Soerjomataram I, et al.
    Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024; 74: 229–63.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Perou CM,
    2. Sørlie T,
    3. Eisen MB,
    4. van de Rijn M,
    5. Jeffrey SS,
    6. Rees CA, et al.
    Molecular portraits of human breast tumours. Nature. 2000; 406: 747–52.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Sørlie T,
    2. Perou CM,
    3. Tibshirani R,
    4. Aas T,
    5. Geisler S,
    6. Johnsen H, et al.
    Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A. 2001; 98: 10869–74.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Goldhirsch A,
    2. Wood WC,
    3. Coates AS,
    4. Gelber RD,
    5. Thürlimann B,
    6. Senn HJ, et al.
    Strategies for subtypes – dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol. 2011; 22: 1736–47.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Lloyd MR,
    2. Jhaveri K,
    3. Kalinsky K,
    4. Bardia A,
    5. Wander SA.
    Precision therapeutics and emerging strategies for HR-positive metastatic breast cancer. Nat Rev Clin Oncol. 2024; 21: 743–61.
    OpenUrlPubMed
  6. 6.
    1. Goutsouliak K,
    2. Veeraraghavan J,
    3. Sethunath V,
    4. De Angelis C,
    5. Osborne CK,
    6. Rimawi MF, et al.
    Towards personalized treatment for early stage HER2-positive breast cancer. Nat Rev Clin Oncol. 2020; 17: 233–50.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Criscitiello C,
    2. Azim HA Jr.,
    3. Schouten PC,
    4. Linn SC,
    5. Sotiriou C.
    Understanding the biology of triple-negative breast cancer. Ann Oncol. 2012; 23: vi13–8.
    OpenUrlCrossRefPubMed
  8. 8.↵
    Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012; 490: 61–70.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Tsimberidou AM,
    2. Fountzilas E,
    3. Nikanjam M,
    4. Kurzrock R.
    Review of precision cancer medicine: evolution of the treatment paradigm. Cancer Treat Rev. 2020; 86: 102019.
  10. 10.↵
    1. Rakha EA,
    2. Pareja FG.
    New advances in molecular breast cancer pathology. Semin Cancer Biol. 2021; 72: 102–13.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. MacConaill LE.
    Existing and emerging technologies for tumor genomic profiling. J Clin Oncol. 2013; 31: 1815–24.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Tarantino P,
    2. Hamilton E,
    3. Tolaney SM,
    4. Cortes J,
    5. Morganti S,
    6. Ferraro E, et al.
    HER2-low breast cancer: pathological and clinical landscape. J Clin Oncol. 2020; 38: 1951–62.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Futreal PA,
    2. Liu Q,
    3. Shattuck-Eidens D,
    4. Cochran C,
    5. Harshman K,
    6. Tavtigian S, et al.
    BRCA1 mutations in primary breast and ovarian carcinomas. Science. 1994; 266: 120–2.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Miki Y,
    2. Katagiri T,
    3. Kasumi F,
    4. Yoshimoto T,
    5. Nakamura Y.
    Mutation analysis in the BRCA2 gene in primary breast cancers. Nat Genet. 1996; 13: 245–7.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Tung NM,
    2. Garber JE.
    BRCA1/2 testing: therapeutic implications for breast cancer management. Br J Cancer. 2018; 119: 141–52.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Goodwin S,
    2. McPherson JD,
    3. McCombie WR.
    Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016; 17: 333–51.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. O’Leary B,
    2. Finn RS,
    3. Turner NC.
    Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016; 13: 417–30.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Dumontet C,
    2. Reichert JM,
    3. Senter PD,
    4. Lambert JM,
    5. Beck A.
    Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov. 2023; 22: 641–61.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Gaynor N,
    2. Crown J,
    3. Collins DM.
    Immune checkpoint inhibitors: key trials and an emerging role in breast cancer. Semin Cancer Biol. 2022; 79: 44–57.
    OpenUrlPubMed
  20. 20.↵
    1. Turner NC,
    2. Ro J,
    3. Andre F,
    4. Loi S,
    5. Verma S,
    6. Iwata H, et al.
    Palbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med. 2015; 373: 209–19.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Bardia A,
    2. Hurvitz S,
    3. Press MF,
    4. Wang LS,
    5. McAndrew NP,
    6. Chan D, et al.
    TRIO-US B-12 TALENT: neoadjuvant trastuzumab deruxtecan with or without anastrozole for HER2-low, HR+ early stage breast cancer. Cancer Res. 2023; 83: GS2–03.
    OpenUrl
  22. 22.↵
    1. Modi S,
    2. Jacot W,
    3. Yamashita T,
    4. Sohn J,
    5. Vidal M,
    6. Tokunaga E, et al.
    Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N Engl J Med. 2022; 387: 9–20.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Cortes J,
    2. Rugo HS,
    3. Cescon DW,
    4. Im SA,
    5. Yusof MM,
    6. Gallardo C, et al.
    Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022; 387: 217–26.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Schmid P,
    2. Adams S,
    3. Rugo HS,
    4. Schneeweiss A,
    5. Barrios CH,
    6. Iwata H, et al.
    Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018; 379: 2108–21.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Paik S,
    2. Shak S,
    3. Tang G,
    4. Kim C,
    5. Baker J,
    6. Cronin M, et al.
    A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004; 351: 2817–26.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Kalinsky K,
    2. Barlow WE,
    3. Gralow JR,
    4. Meric-Bernstam F,
    5. Albain KS,
    6. Hayes DF, et al.
    21-Gene assay to inform chemotherapy benefit in node-positive breast cancer. N Engl J Med. 2021; 385: 2336–47.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. van ‘t Veer LJ,
    2. Dai H,
    3. van de Vijver MJ,
    4. He YD,
    5. Hart AA,
    6. Mao M, et al.
    Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002; 415: 530–6.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Venetis K,
    2. Pescia C,
    3. Cursano G,
    4. Frascarelli C,
    5. Mane E,
    6. De Camilli E, et al.
    The evolving role of genomic testing in early breast cancer: implications for diagnosis, prognosis, and therapy. Int J Mol Sci. 2024; 25: 5717.
    OpenUrlPubMed
  29. 29.↵
    1. Zhang Q,
    2. Cai Z,
    3. Gerratana L,
    4. Davis AA,
    5. D’Amico P,
    6. Chawla A, et al.
    Early evaluation of risk stratification and clinical outcomes for patients with advanced breast cancer through combined monitoring of baseline circulating tumor cells and DNA. Clin Cancer Res. 2024; 30: 3470–80.
    OpenUrlPubMed
  30. 30.↵
    1. Garcia-Murillas I,
    2. Cutts R,
    3. Abbott C,
    4. Boyle SM,
    5. Pugh J,
    6. Chen R, et al.
    Ultra-sensitive ctDNA mutation tracking to identify molecular residual disease and predict relapse in patients with early breast cancer. Am Soc Clin Oncol. 2024; 42: 1010.
    OpenUrl
  31. 31.↵
    1. Padroni L,
    2. De Marco L,
    3. Fiano V,
    4. Milani L,
    5. Marmiroli G,
    6. Giraudo MT, et al.
    Identifying microRNAs suitable for detection of breast cancer: a systematic review of discovery phases studies on microRNA expression profiles. Int J Mol Sci. 2023; 24: 15114.
  32. 32.↵
    1. Su X,
    2. Shan Z,
    3. Duan S.
    Harnessing extracellular vesicles using liquid biopsy for cancer diagnosis and monitoring: highlights from AACR Annual Meeting 2024. J Hematol Oncol. 2024; 17: 55.
    OpenUrlPubMed
  33. 33.↵
    1. Allemani C,
    2. Weir HK,
    3. Carreira H,
    4. Harewood R,
    5. Spika D,
    6. Wang XS, et al.
    Global surveillance of cancer survival 1995-2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (CONCORD-2). Lancet. 2015; 385: 977–1010.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Han B,
    2. Zheng R,
    3. Zeng H,
    4. Wang S,
    5. Sun K,
    6. Chen R, et al.
    Cancer incidence and mortality in China, 2022. J Natl Cancer Cent. 2024; 4: 47–53.
    OpenUrlCrossRefPubMed
  35. 35.↵
    Early Breast Cancer Trialists’ Collaborative Group. Reductions in recurrence in women with early breast cancer entering clinical trials between 1990 and 2009: a pooled analysis of 155 746 women in 151 trials. Lancet. 2024; 404: 1407–18.
    OpenUrlPubMed
  36. 36.↵
    1. André F,
    2. Ciruelos E,
    3. Rubovszky G,
    4. Campone M,
    5. Loibl S,
    6. Rugo HS, et al.
    Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019; 380: 1929–40.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Gradishar WJ,
    2. Moran MS,
    3. Abraham J,
    4. Abramson V,
    5. Aft R,
    6. Agnese D, et al.
    Breast Cancer, Version 3.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2024; 22: 331–57.
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. Jensen EV,
    2. DeSombre ER.
    Estrogen-receptor interaction. Science. 1973; 182: 126–34.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Johnson KA,
    2. Brown PH.
    Drug development for cancer chemoprevention: focus on molecular targets. Semin Oncol. 2010; 37: 345–58.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Davies C,
    2. Pan H,
    3. Godwin J,
    4. Gray R,
    5. Arriagada R,
    6. Raina V, et al.
    Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet. 2013; 381: 805–16.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Fisher B,
    2. Dignam J,
    3. Wolmark N,
    4. Wickerham DL,
    5. Fisher ER,
    6. Mamounas E, et al.
    Tamoxifen in treatment of intraductal breast cancer: National Surgical Adjuvant Breast and Bowel Project B-24 randomised controlled trial. Lancet. 1999; 353: 1993–2000.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Francis PA,
    2. Regan MM,
    3. Fleming GF,
    4. Láng I,
    5. Ciruelos E,
    6. Bellet M, et al.
    Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med. 2015; 372: 436–46.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Pagani O,
    2. Regan MM,
    3. Walley BA,
    4. Fleming GF,
    5. Colleoni M,
    6. Láng I, et al.
    Adjuvant exemestane with ovarian suppression in premenopausal breast cancer. N Engl J Med. 2014; 371: 107–18.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Burstein HJ,
    2. Lacchetti C,
    3. Anderson H,
    4. Buchholz TA,
    5. Davidson NE,
    6. Gelmon KE, et al.
    Adjuvant endocrine therapy for women with hormone receptor-positive breast cancer: American Society of Clinical Oncology clinical practice guideline update on ovarian suppression. J Clin Oncol. 2016; 34: 1689–701.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    1. Francis PA,
    2. Regan MM,
    3. Fleming GF.
    Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med. 2015; 372: 1673.
    OpenUrlCrossRef
  46. 46.↵
    1. Chumsri S,
    2. Howes T,
    3. Bao T,
    4. Sabnis G,
    5. Brodie A.
    Aromatase, aromatase inhibitors, and breast cancer. J Steroid Biochem Mol Biol. 2011; 125: 13–22.
    OpenUrlCrossRefPubMed
  47. 47.↵
    1. Cuzick J,
    2. Sestak I,
    3. Baum M,
    4. Buzdar A,
    5. Howell A,
    6. Dowsett M, et al.
    Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 10-year analysis of the ATAC trial. Lancet. Oncol. 2010; 11: 1135–41.
    OpenUrl
  48. 48.↵
    1. Gibson L,
    2. Lawrence D,
    3. Dawson C,
    4. Bliss J.
    Aromatase inhibitors for treatment of advanced breast cancer in postmenopausal women. Cochrane Database Syst Rev. 2009; 2009: CD003370.
  49. 49.↵
    1. Baker H.
    Extending aromatase-inhibitor adjuvant therapy to 10 years. Lancet Oncol. 2016; 17: e275.
  50. 50.↵
    1. Noordhoek I,
    2. Treuner K,
    3. Putter H,
    4. Zhang Y,
    5. Wong J,
    6. Meershoek-Klein Kranenbarg E, et al.
    Breast cancer index predicts extended endocrine benefit to individualize selection of patients with HR+ early-stage breast cancer for 10 years of endocrine therapy. Clin Cancer Res. 2021; 27: 311–19.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    1. Gheysen M,
    2. Punie K,
    3. Wildiers H,
    4. Neven P.
    Oral SERDs changing the scenery in hormone receptor positive breast cancer, a comprehensive review. Cancer Treat Rev. 2024; 130: 102825.
  52. 52.↵
    1. Robertson JFR,
    2. Bondarenko IM,
    3. Trishkina E,
    4. Dvorkin M,
    5. Panasci L,
    6. Manikhas A, et al.
    Fulvestrant 500 mg versus anastrozole 1 mg for hormone receptor-positive advanced breast cancer (FALCON): an international, randomised, double-blind, phase 3 trial. Lancet. 2016; 388: 2997–3005.
    OpenUrlCrossRefPubMed
  53. 53.↵
    1. Bidard FC,
    2. Kaklamani VG,
    3. Neven P,
    4. Streich G,
    5. Montero AJ,
    6. Forget F, et al.
    Elacestrant (oral selective estrogen receptor degrader) versus standard endocrine therapy for estrogen receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: results from the randomized phase III EMERALD trial. J Clin Oncol. 2022; 40: 3246–56.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Bardia A,
    2. Kaklamani V,
    3. Wilks S,
    4. Weise A,
    5. Richards D,
    6. Harb W, et al.
    Phase I study of elacestrant (RAD1901), a novel selective estrogen receptor degrader, in ER-positive, HER2-negative advanced breast cancer. J Clin Oncol. 2021; 39: 1360–70.
    OpenUrlPubMed
  55. 55.↵
    1. Pan H,
    2. Gray R,
    3. Braybrooke J,
    4. Davies C,
    5. Taylor C,
    6. McGale P, et al.
    20-Year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years. N Engl J Med. 2017; 377: 1836–46.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Finn RS,
    2. Dering J,
    3. Conklin D,
    4. Kalous O,
    5. Cohen DJ,
    6. Desai AJ, et al.
    PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 2009; 11: R77.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Pernas S,
    2. Tolaney SM,
    3. Winer EP,
    4. Goel S.
    CDK4/6 inhibition in breast cancer: current practice and future directions. Ther Adv Med Oncol. 2018; 10: 1758835918786451.
  58. 58.↵
    1. Gnant M,
    2. Dueck AC,
    3. Frantal S,
    4. Martin M,
    5. Burstein HJ,
    6. Greil R, et al.
    Adjuvant palbociclib for early breast cancer: the PALLAS trial results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol. 2022; 40: 282–93.
    OpenUrlPubMed
  59. 59.↵
    1. Loibl S,
    2. Marmé F,
    3. Martin M,
    4. Untch M,
    5. Bonnefoi H,
    6. Kim SB, et al.
    Palbociclib for residual high-risk invasive HR-positive and HER2-negative early breast cancer-The Penelope-B trial. J Clin Oncol. 2021; 39: 1518–30.
    OpenUrlCrossRefPubMed
  60. 60.↵
    1. Johnston SRD,
    2. Harbeck N,
    3. Hegg R,
    4. Toi M,
    5. Martin M,
    6. Shao ZM, et al.
    Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2−, node-positive, high-risk, early breast cancer (monarchE). J Clin Oncol. 2020; 38: 3987–98.
    OpenUrlCrossRefPubMed
  61. 61.↵
    1. Slamon D,
    2. Lipatov O,
    3. Nowecki Z,
    4. McAndrew N,
    5. Kukielka-Budny B,
    6. Stroyakovskiy D, et al.
    Ribociclib plus endocrine therapy in early breast cancer. N Engl J Med. 2024; 390: 1080–91.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Zhang L,
    2. Geng C,
    3. Liu Y,
    4. Han J,
    5. Han M,
    6. Huang R, et al.
    A phase II trial comparing dalpiciclib in combination with letrozole versus standard chemotherapy as neoadjuvant therapy in patients with high-risk HR-positive HER-2 negative breast cancer: DARLING-02. J Clin Oncol. 2024; 42: TPS626.
  63. 63.↵
    1. Zhang L,
    2. Yang C,
    3. Ma J,
    4. Li Y,
    5. Luo R,
    6. Han J, et al.
    Abstract PO2-02-05: CDK4/6 inhibitor dalpiciclib combined with letrozole as neoadjuvant therapy in postmenopausal patients with hormone receptor-positive, HER2-negative stage II-III breast cancer: a single-arm exploratory trial. Cancer Res. 2024; 84: PO2-02–05.
    OpenUrl
  64. 64.↵
    1. Johnston SRD,
    2. Toi M,
    3. O’Shaughnessy J,
    4. Rastogi P,
    5. Campone M,
    6. Neven P, et al.
    Abemaciclib plus endocrine therapy for hormone receptor-positive, HER2-negative, node-positive, high-risk early breast cancer (monarchE): results from a preplanned interim analysis of a randomised, open-label, phase 3 trial. Lancet Oncol. 2023; 24: 77–90.
    OpenUrlCrossRefPubMed
  65. 65.↵
    1. Harbeck N,
    2. Rastogi P,
    3. Martin M,
    4. Tolaney SM,
    5. Shao ZM,
    6. Fasching PA, et al.
    Adjuvant abemaciclib combined with endocrine therapy for high-risk early breast cancer: updated efficacy and Ki-67 analysis from the monarchE study. Ann Oncol. 2021; 32: 1571–81.
    OpenUrlCrossRefPubMed
  66. 66.↵
    1. Rugo HS,
    2. Vitko A,
    3. Thoele K.
    An overview of adverse event (AE) management for patients (pts) receiving abemaciclib. J Clin Oncol. 2022; 40: 239–39.
    OpenUrl
  67. 67.↵
    1. Xu B,
    2. Zhang Q,
    3. Zhang P,
    4. Hu X,
    5. Li W,
    6. Tong Z, et al.
    Dalpiciclib or placebo plus fulvestrant in hormone receptor-positive and HER2-negative advanced breast cancer: a randomized, phase 3 trial. Nat Med. 2021; 27: 1904–9.
    OpenUrlCrossRefPubMed
  68. 68.↵
    1. Zhang P,
    2. Zhang Q,
    3. Tong Z,
    4. Sun T,
    5. Li W,
    6. Ouyang Q, et al.
    Dalpiciclib plus letrozole or anastrozole versus placebo plus letrozole or anastrozole as first-line treatment in patients with hormone receptor-positive, HER2-negative advanced breast cancer (DAWNA-2): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2023; 24: 646–57.
    OpenUrlPubMed
  69. 69.↵
    1. Cardoso F,
    2. McArthur HL,
    3. Schmid P,
    4. Cortés J,
    5. Harbeck N,
    6. Telli ML, et al.
    LBA21 KEYNOTE-756: phase III study of neoadjuvant pembrolizumab (pembro) or placebo (pbo) + chemotherapy (chemo), followed by adjuvant pembro or pbo + endocrine therapy (ET) for early-stage high-risk ER+/HER2– breast cancer. Ann Oncol. 2023; 34: S1254–335.
    OpenUrl
  70. 70.↵
    1. Loi S,
    2. Curigliano G,
    3. Salgado RF,
    4. Romero Diaz RI,
    5. Delaloge S,
    6. Rojas C, et al.
    LBA20 A randomized, double-blind trial of nivolumab (NIVO) vs placebo (PBO) with neoadjuvant chemotherapy (NACT) followed by adjuvant endocrine therapy (ET) ± NIVO in patients (pts) with high-risk, ER+ HER2− primary breast cancer (BC). Ann Oncol. 2023; 34: S1259–60.
    OpenUrlCrossRef
  71. 71.↵
    1. Schmid P,
    2. Cortes J,
    3. Pusztai L,
    4. McArthur H,
    5. Kümmel S,
    6. Bergh J, et al.
    Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020; 382: 810–21.
    OpenUrlCrossRefPubMed
  72. 72.↵
    1. Schmid P,
    2. Cortes J,
    3. Dent R,
    4. Pusztai L,
    5. McArthur H,
    6. Kümmel S, et al.
    Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022; 386: 556–67.
    OpenUrlCrossRefPubMed
  73. 73.↵
    1. Schmid P,
    2. Cortes J,
    3. Dent R,
    4. McArthur H,
    5. Pusztai L,
    6. Kümmel S, et al.
    Overall survival with pembrolizumab in early-stage triple-negative breast cancer. N Engl J Med. 2024; 391: 1981–91.
    OpenUrlCrossRefPubMed
  74. 74.↵
    1. Mittendorf EA,
    2. Zhang H,
    3. Barrios CH,
    4. Saji S,
    5. Jung KH,
    6. Hegg R, et al.
    Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): a randomised, double-blind, phase 3 trial. Lancet. 2020; 396: 1090–100.
    OpenUrlCrossRefPubMed
  75. 75.↵
    1. Conte PF,
    2. Dieci MV,
    3. Bisagni G,
    4. Schmid P,
    5. Fotia V,
    6. Piacentini F, et al.
    A-BRAVE trial: a phase III randomized trial with avelumab in early triple-negative breast cancer with residual disease after neoadjuvant chemotherapy or at high risk after primary surgery and adjuvant chemotherapy. J Clin Oncol. 2024; 42: LBA500.
  76. 76.↵
    1. Geyer CE Jr.,
    2. Garber JE,
    3. Gelber RD,
    4. Yothers G,
    5. Taboada M,
    6. Ross L, et al.
    Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann Oncol. 2022; 33: 1250–68.
    OpenUrlCrossRefPubMed
  77. 77.↵
    1. Tutt ANJ,
    2. Garber JE,
    3. Kaufman B,
    4. Viale G,
    5. Fumagalli D,
    6. Rastogi P, et al.
    Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med. 2021; 384: 2394–405.
    OpenUrlCrossRefPubMed
  78. 78.↵
    1. Ganz PA,
    2. Bandos H,
    3. Španić T,
    4. Friedman S,
    5. Müller V,
    6. Kuemmel S, et al.
    Patient-reported outcomes in OlympiA: a phase III, randomized, placebo-controlled trial of adjuvant olaparib in gBRCA1/2 mutations and high-risk human epidermal growth factor receptor 2-negative early breast cancer. J Clin Oncol. 2024; 42: 1288–300.
    OpenUrlPubMed
  79. 79.↵
    1. Giaquinto AN,
    2. Sung H,
    3. Miller KD,
    4. Kramer JL,
    5. Newman LA,
    6. Minihan A, et al.
    Breast cancer statistics, 2022. CA Cancer J Clin. 2022; 72: 524–41.
    OpenUrlCrossRefPubMed
  80. 80.↵
    1. Wolff AC,
    2. Somerfield MR,
    3. Dowsett M,
    4. Hammond MEH,
    5. Hayes DF,
    6. McShane LM, et al.
    Human epidermal growth factor receptor 2 testing in breast cancer: ASCO-College of American Pathologists Guideline Update. J Clin Oncol. 2023; 41: 3867–72.
    OpenUrlCrossRefPubMed
  81. 81.↵
    1. von Minckwitz G,
    2. Procter M,
    3. de Azambuja E,
    4. Zardavas D,
    5. Benyunes M,
    6. Viale G, et al.
    Adjuvant pertuzumab and trastuzumab in early HER2-positive breast cancer. N Engl J Med. 2017; 377: 122–31.
    OpenUrlCrossRefPubMed
  82. 82.↵
    1. Piccart M,
    2. Procter M,
    3. Fumagalli D,
    4. de Azambuja E,
    5. Clark E,
    6. Ewer MS, et al.
    Adjuvant pertuzumab and trastuzumab in early HER2-positive breast cancer in the APHINITY trial: 6 years’ follow-up. J Clin Oncol. 2021; 39: 1448–57.
    OpenUrlCrossRefPubMed
  83. 83.↵
    1. Loibl S,
    2. Jassem J,
    3. Sonnenblick A,
    4. Parlier D,
    5. Winer E,
    6. Bergh J, et al.
    Adjuvant pertuzumab and trastuzumab in early human epidermal growth factor receptor 2-positive breast cancer in the APHINITY trial: third interim overall survival analysis with efficacy update. J Clin Oncol. 2024; 42: 3643–51.
    OpenUrlPubMed
  84. 84.↵
    1. Shao Z,
    2. Huang T,
    3. Fan Z,
    4. Wang Y,
    5. Yan X,
    6. Yang H, et al.
    1MO The fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection (PH FDC SC) in Chinese patients (pts) with HER2-positive early breast cancer (EBC): primary analysis of the phase III, randomised FDChina study. Ann Oncol. 2022; 33: S1431.
  85. 85.↵
    1. von Minckwitz G,
    2. Huang CS,
    3. Mano MS,
    4. Loibl S,
    5. Mamounas EP,
    6. Untch M, et al.
    Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019; 380: 617–28.
    OpenUrlCrossRefPubMed
  86. 86.↵
    1. Loibl S,
    2. Mano MS,
    3. Untch M,
    4. Huang C-S,
    5. Mamounas E,
    6. Wolmark N, et al.
    Phase III study of adjuvant ado-trastuzumab emtansine vs trastuzumab for residual invasive HER2-positive early breast cancer after neoadjuvant chemotherapy and HER2-targeted therapy: KATHERINE final IDFS and updated OS analysis. Cancer Res. 2024; 84: GS03–12.
    OpenUrlCrossRef
  87. 87.↵
    1. Dowling GP,
    2. Toomey S,
    3. Bredin P,
    4. Parker I,
    5. Mulroe E,
    6. Marron J, et al.
    Neoadjuvant trastuzumab deruxtecan (T-DXd) with response-directed definitive therapy in early stage HER2-positive breast cancer: a phase II study protocol (SHAMROCK study). BMC Cancer. 2024; 24: 91.
    OpenUrlPubMed
  88. 88.↵
    1. Chan A,
    2. Delaloge S,
    3. Holmes FA,
    4. Moy B,
    5. Iwata H,
    6. Harvey VJ, et al.
    Neratinib after trastuzumab-based adjuvant therapy in patients with HER2-positive breast cancer (ExteNET): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2016; 17: 367–77.
    OpenUrlCrossRefPubMed
  89. 89.↵
    1. Baselga J,
    2. Bradbury I,
    3. Eidtmann H,
    4. Di Cosimo S,
    5. de Azambuja E,
    6. Aura C, et al.
    Lapatinib with trastuzumab for HER2-positive early breast cancer (NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. Lancet. 2012; 379: 633–40.
    OpenUrlCrossRefPubMed
  90. 90.↵
    1. Wolff AC,
    2. Hammond ME,
    3. Hicks DG,
    4. Dowsett M,
    5. McShane LM,
    6. Allison KH, et al.
    Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013; 31: 3997–4013.
    OpenUrlAbstract/FREE Full Text
  91. 91.↵
    1. Dent R,
    2. Trudeau M,
    3. Pritchard KI,
    4. Hanna WM,
    5. Kahn HK,
    6. Sawka CA, et al.
    Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007; 13: 4429–34.
    OpenUrlAbstract/FREE Full Text
  92. 92.↵
    1. Lin NU,
    2. Claus E,
    3. Sohl J,
    4. Razzak AR,
    5. Arnaout A,
    6. Winer EP.
    Sites of distant recurrence and clinical outcomes in patients with metastatic triple-negative breast cancer: high incidence of central nervous system metastases. Cancer. 2008; 113: 2638–45.
    OpenUrlCrossRefPubMed
  93. 93.↵
    1. Zhang L,
    2. Fang C,
    3. Xu X,
    4. Li A,
    5. Cai Q,
    6. Long X.
    Androgen receptor, EGFR, and BRCA1 as biomarkers in triple-negative breast cancer: a meta-analysis. Biomed Res Int. 2015; 2015: 357485.
  94. 94.↵
    1. Gluz O,
    2. Liedtke C,
    3. Gottschalk N,
    4. Pusztai L,
    5. Nitz U,
    6. Harbeck N.
    Triple-negative breast cancer--current status and future directions. Ann Oncol. 2009; 20: 1913–27.
    OpenUrlCrossRefPubMed
  95. 95.↵
    1. Derakhshan F,
    2. Reis-Filho JS.
    Pathogenesis of triple-negative breast cancer. Annu Rev Pathol. 2022; 17: 181–204.
    OpenUrlPubMed
  96. 96.↵
    1. Emens LA.
    Breast cancer immunotherapy: facts and hopes. Clin Cancer Res. 2018; 24: 511–20.
    OpenUrlAbstract/FREE Full Text
  97. 97.↵
    1. Farmer H,
    2. McCabe N,
    3. Lord CJ,
    4. Tutt AN,
    5. Johnson DA,
    6. Richardson TB, et al.
    Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005; 434: 917–21.
    OpenUrlCrossRefPubMed
  98. 98.↵
    1. Bryant HE,
    2. Schultz N,
    3. Thomas HD,
    4. Parker KM,
    5. Flower D,
    6. Lopez E, et al.
    Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005; 434: 913–7.
    OpenUrlCrossRefPubMed
  99. 99.↵
    1. Fong PC,
    2. Boss DS,
    3. Yap TA,
    4. Tutt A,
    5. Wu P,
    6. Mergui-Roelvink M, et al.
    Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009; 361: 123–34.
    OpenUrlCrossRefPubMed
  100. 100.↵
    1. Jeselsohn R,
    2. Buchwalter G,
    3. De Angelis C,
    4. Brown M,
    5. Schiff R.
    ESR1 mutations—a mechanism for acquired endocrine resistance in breast cancer. Nat Rev Clin Oncol. 2015; 12: 573–83.
    OpenUrlCrossRefPubMed
  101. 101.↵
    1. Shah M,
    2. Lingam H,
    3. Gao X,
    4. Gittleman H,
    5. Fiero MH,
    6. Krol D, et al.
    US Food and Drug Administration approval summary: elacestrant for estrogen receptor-positive, human epidermal growth factor receptor 2-negative, ESR1-mutated advanced or metastatic breast cancer. J Clin Oncol. 2024; 42: 1193–201.
    OpenUrlPubMed
  102. 102.↵
    1. Gao JJ,
    2. Cheng J,
    3. Bloomquist E,
    4. Sanchez J,
    5. Wedam SB,
    6. Singh H, et al.
    CDK4/6 inhibitor treatment for patients with hormone receptor-positive, HER2-negative, advanced or metastatic breast cancer: a US Food and Drug Administration pooled analysis. Lancet Oncol. 2020; 21: 250–60.
    OpenUrlCrossRefPubMed
  103. 103.
    1. Finn RS,
    2. Crown JP,
    3. Lang I,
    4. Boer K,
    5. Bondarenko IM,
    6. Kulyk SO, 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.
    OpenUrlCrossRefPubMed
  104. 104.
    1. Finn RS,
    2. Boer K,
    3. Bondarenko I,
    4. Patel R,
    5. Pinter T,
    6. Schmidt M, et al.
    Overall survival results from the randomized phase 2 study of palbociclib in combination with letrozole versus letrozole alone for first-line treatment of ER+/HER2− advanced breast cancer (PALOMA-1, TRIO-18). Breast Cancer Res Treat. 2020; 183: 419–28.
    OpenUrlCrossRefPubMed
  105. 105.↵
    1. Finn RS,
    2. Martin M,
    3. Rugo HS,
    4. Jones S,
    5. Im SA,
    6. Gelmon K, et al.
    Palbociclib and letrozole in advanced breast cancer. N Engl J Med. 2016; 375: 1925–36.
    OpenUrlCrossRefPubMed
  106. 106.↵
    1. Finn RS,
    2. Rugo HS,
    3. Dieras VC,
    4. Harbeck N,
    5. Im S-A,
    6. Gelmon KA, et al.
    Overall survival (OS) with first-line palbociclib plus letrozole (PAL+LET) versus placebo plus letrozole (PBO+LET) in women with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer (ER+/HER2− ABC): analyses from PALOMA-2. J Clin Oncol. 2022; 40: LBA1003.
  107. 107.
    1. Cristofanilli M,
    2. Turner NC,
    3. Bondarenko I,
    4. Ro J,
    5. Im SA,
    6. Masuda N, et al.
    Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 2016; 17: 425–39.
    OpenUrlCrossRefPubMed
  108. 108.
    1. Cristofanilli M,
    2. Rugo HS,
    3. Im SA,
    4. Slamon DJ,
    5. Harbeck N,
    6. Bondarenko I, et al.
    Overall survival with palbociclib and fulvestrant in women with HR+/HER2− ABC: updated exploratory analyses of PALOMA-3, a double-blind, phase III randomized study. Clin Cancer Res. 2022; 28: 3433–42.
    OpenUrlPubMed
  109. 109.↵
    1. Hortobagyi GN,
    2. Stemmer SM,
    3. Burris HA,
    4. Yap YS,
    5. Sonke GS,
    6. Paluch-Shimon S, et al.
    Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2-negative advanced breast cancer. Ann Oncol. 2018; 29: 1541–47.
    OpenUrlCrossRefPubMed
  110. 110.↵
    1. Hortobagyi GN,
    2. Stemmer SM,
    3. Burris HA,
    4. Yap YS,
    5. Sonke GS,
    6. Hart L, et al.
    Overall survival with ribociclib plus letrozole in advanced breast cancer. N Engl J Med. 2022; 386: 942–50.
    OpenUrlCrossRefPubMed
  111. 111.
    1. Slamon DJ,
    2. Neven P,
    3. Chia S,
    4. Fasching PA,
    5. De Laurentiis M,
    6. Im SA, et al.
    Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J Clin Oncol. 2018; 36: 2465–72.
    OpenUrlCrossRefPubMed
  112. 112.
    1. Slamon DJ,
    2. Neven P,
    3. Chia S,
    4. Fasching PA,
    5. De Laurentiis M,
    6. Im SA, et al.
    Overall survival with ribociclib plus fulvestrant in advanced breast cancer. N Engl J Med. 2020; 382: 514–24.
    OpenUrlCrossRefPubMed
  113. 113.
    1. Tripathy D,
    2. Im SA,
    3. Colleoni M,
    4. Franke F,
    5. Bardia A,
    6. Harbeck N, et al.
    Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial. Lancet Oncol. 2018; 19: 904–15.
    OpenUrlCrossRefPubMed
  114. 114.
    1. Lu YS,
    2. Im SA,
    3. Colleoni M,
    4. Franke F,
    5. Bardia A,
    6. Cardoso F, et al.
    Updated overall survival of ribociclib plus endocrine therapy versus endocrine therapy alone in pre- and perimenopausal patients with HR+/HER2− advanced breast cancer in MONALEESA-7: a phase III randomized clinical trial. Clin Cancer Res. 2022; 28: 851–59.
    OpenUrlPubMed
  115. 115.
    1. Sledge GW Jr.,
    2. Toi M,
    3. Neven P,
    4. Sohn J,
    5. Inoue K,
    6. Pivot X, et al.
    MONARCH 2: abemaciclib in combination with fulvestrant in women with HR+/HER2− advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol. 2017; 35: 2875–84.
    OpenUrlCrossRefPubMed
  116. 116.
    1. Llombart-Cussac A,
    2. Sledge G,
    3. Toi M,
    4. Neven P,
    5. Sohn JH,
    6. Inoue K, et al.
    Abstract PD13-11: PD13-11 final overall survival analysis of Monarch 2: a phase 3 trial of abemaciclib plus fulvestrant in patients with hormone receptor-positive, HER2-negative advanced breast cancer. Cancer Res. 2023; 83: PD13–11.
    OpenUrl
  117. 117.
    1. Patnaik A,
    2. Rosen LS,
    3. Tolaney SM,
    4. Tolcher AW,
    5. Goldman JW,
    6. Gandhi L, et al.
    Efficacy and safety of abemaciclib, an inhibitor of CDK4 and CDK6, for patients with breast cancer, non-small cell lung cancer, and other solid tumors. Cancer Discov. 2016; 6: 740–53.
    OpenUrlAbstract/FREE Full Text
  118. 118.
    1. Hu X,
    2. Zhang Q,
    3. Sun T,
    4. Yin Y,
    5. Li H,
    6. Yan M, et al.
    Abemaciclib plus non-steroidal aromatase inhibitor or fulvestrant in women with HR+/HER2− advanced breast cancer: final results of the randomized phase III MONARCH plus trial. Chin Med J (Engl). 2024. doi:10.1097/CM9.0000000000003151.
    OpenUrlCrossRef
  119. 119.↵
    1. Tokunaga E,
    2. Kimura Y,
    3. Mashino K,
    4. Oki E,
    5. Kataoka A,
    6. Ohno S, et al.
    Activation of PI3K/Akt signaling and hormone resistance in breast cancer. Breast Cancer. 2006; 13: 137–44.
    OpenUrlCrossRefPubMed
  120. 120.↵
    1. Nunnery SE,
    2. Mayer IA.
    Targeting the PI3K/AKT/mTOR pathway in hormone-positive breast cancer. Drugs. 2020; 80: 1685–97.
    OpenUrlPubMed
  121. 121.↵
    1. Glaviano A,
    2. Foo ASC,
    3. Lam HY,
    4. Yap KCH,
    5. Jacot W,
    6. Jones RH, et al.
    PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 2023; 22: 138.
    OpenUrlCrossRefPubMed
  122. 122.↵
    1. Zheng Y,
    2. Zhang Z,
    3. Li D,
    4. Huang R,
    5. Ning S.
    Breaking through therapeutic barriers: insights into CDK4/6 inhibition resistance in hormone receptor-positive metastatic breast cancer. Biochim Biophys Acta Rev Cancer. 2024; 1879: 189174.
  123. 123.↵
    1. Markham A.
    Alpelisib: first global approval. Drugs. 2019; 79: 1249–53.
    OpenUrlCrossRefPubMed
  124. 124.↵
    1. André F,
    2. Ciruelos EM,
    3. Juric D,
    4. Loibl S,
    5. Campone M,
    6. Mayer IA, et al.
    Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: final overall survival results from SOLAR-1. Ann Oncol. 2021; 32: 208–17.
    OpenUrlCrossRefPubMed
  125. 125.↵
    1. Turner NC,
    2. Im SA,
    3. Saura C,
    4. Juric D,
    5. Loibl S,
    6. Kalinsky K, et al.
    Inavolisib-based therapy in PIK3CA-mutated advanced breast cancer. N Engl J Med. 2024; 391: 1584–96.
    OpenUrlPubMed
  126. 126.↵
    1. Pervanidis KA,
    2. D’Angelo GD,
    3. Weisner J,
    4. Brandherm S,
    5. Rauh D.
    Akt inhibitor advancements: from capivasertib approval to covalent-allosteric promises. J Med Chem. 2024; 67: 6052–63.
    OpenUrlPubMed
  127. 127.↵
    1. Oliveira M,
    2. Rugo HS,
    3. Howell SJ,
    4. Dalenc F,
    5. Cortes J,
    6. Gomez HL, et al.
    Capivasertib and fulvestrant for patients with hormone receptor-positive, HER2-negative advanced breast cancer (CAPItello-291): patient-reported outcomes from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2024; 25: 1231–44.
    OpenUrlPubMed
  128. 128.↵
    1. Turner NC,
    2. Oliveira M,
    3. Howell SJ,
    4. Dalenc F,
    5. Cortes J,
    6. Gomez Moreno HL, et al.
    Capivasertib in hormone receptor-positive advanced breast cancer. N Engl J Med. 2023; 388: 2058–70.
    OpenUrlCrossRefPubMed
  129. 129.↵
    1. Raheem F,
    2. Karikalan SA,
    3. Batalini F,
    4. El Masry A,
    5. Mina L.
    Metastatic ER+ breast cancer: mechanisms of resistance and future therapeutic approaches. Int J Mol Sci. 2023; 24: 16198.
  130. 130.↵
    1. Papadimitriou MC,
    2. Pazaiti A,
    3. Iliakopoulos K,
    4. Markouli M,
    5. Michalaki V,
    6. Papadimitriou CA.
    Resistance to CDK4/6 inhibition: mechanisms and strategies to overcome a therapeutic problem in the treatment of hormone receptor-positive metastatic breast cancer. Biochim Biophys Acta Mol Cell Res. 2022; 1869: 119346.
  131. 131.↵
    1. Baselga J,
    2. Campone M,
    3. Piccart M,
    4. Burris HA,
    5. Rugo HS,
    6. Sahmoud T, et al.
    Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. New Engl J Med. 2012; 366: 520–29.
    OpenUrlCrossRefPubMed
  132. 132.↵
    1. Chang HL,
    2. Schwettmann B,
    3. McArthur HL,
    4. Chan IS.
    Antibody-drug conjugates in breast cancer: overcoming resistance and boosting immune response. J Clin Invest. 2023; 133: e172156.
  133. 133.↵
    1. Drago JZ,
    2. Modi S,
    3. Chandarlapaty S.
    Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol. 2021; 18: 327–44.
    OpenUrlCrossRefPubMed
  134. 134.↵
    1. Bardia A,
    2. Hurvitz SA,
    3. Tolaney SM,
    4. Loirat D,
    5. Punie K,
    6. Oliveira M, et al.
    Sacituzumab govitecan in metastatic triple-negative breast cancer. N Engl J Med. 2021; 384: 1529–41.
    OpenUrlCrossRefPubMed
  135. 135.↵
    1. Bardia A,
    2. Rugo HS,
    3. Tolaney SM,
    4. Loirat D,
    5. Punie K,
    6. Oliveira M, et al.
    Final results from the randomized phase III ASCENT Clinical Trial in metastatic triple-negative breast cancer and association of outcomes by human epidermal growth factor receptor 2 and trophoblast cell surface antigen 2 expression. J Clin Oncol. 2024; 42: 1738–44.
    OpenUrlPubMed
  136. 136.↵
    1. Schechter AL,
    2. Stern DF,
    3. Vaidyanathan L,
    4. Decker SJ,
    5. Drebin JA,
    6. Greene MI, et al.
    The neu oncogene: an erb-B-related gene encoding a 185,000-Mr tumour antigen. Nature. 1984; 312: 513–6.
    OpenUrlCrossRefPubMed
  137. 137.↵
    1. Baselga J.
    Clinical trials of Herceptin® (trastuzumab). Eur J Cancer. 2001; 37: 18–24.
    OpenUrlPubMed
  138. 138.↵
    1. Spector NL,
    2. Blackwell KL.
    Understanding the mechanisms behind trastuzumab therapy for human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2009; 27: 5838–47.
    OpenUrlAbstract/FREE Full Text
  139. 139.↵
    1. Metzger-Filho O,
    2. Winer EP,
    3. Krop I.
    Pertuzumab: optimizing HER2 blockade. Clin Cancer Res. 2013; 19: 5552–6.
    OpenUrlAbstract/FREE Full Text
  140. 140.↵
    1. Swain SM,
    2. Baselga J,
    3. Kim S-B,
    4. Ro J,
    5. Semiglazov V,
    6. Campone M, et al.
    Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015; 372: 724–34.
    OpenUrlCrossRefPubMed
  141. 141.↵
    1. García-Alonso S,
    2. Ocaña A,
    3. Pandiella A.
    Trastuzumab emtansine: mechanisms of action and resistance, clinical progress, and beyond. Trends Cancer. 2020; 6: 130–46.
    OpenUrlPubMed
  142. 142.↵
    1. Verma S,
    2. Miles D,
    3. Gianni L,
    4. Krop IE,
    5. Welslau M,
    6. Baselga J, et al.
    Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012; 367: 1783–91.
    OpenUrlCrossRefPubMed
  143. 143.↵
    1. Hurvitz SA,
    2. Hegg R,
    3. Chung WP,
    4. Im SA,
    5. Jacot W,
    6. Ganju V, et al.
    Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 2023; 401: 105–17.
    OpenUrlCrossRefPubMed
  144. 144.↵
    1. Modi S,
    2. Park H,
    3. Murthy RK,
    4. Iwata H,
    5. Tamura K,
    6. Tsurutani J, et al.
    Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer: results from a phase Ib study. J Clin Oncol. 2020; 38: 1887–96.
    OpenUrlCrossRefPubMed
  145. 145.↵
    1. Bardia A,
    2. Barrios C,
    3. Dent R,
    4. Hu X,
    5. O’Shaughnessy J,
    6. Yonemori K, et al.
    Abstract OT-03-09: Trastuzumab deruxtecan (T-DXd; DS-8201) vs investigator’s choice of chemotherapy in patients with hormone receptor-positive (HR+), HER2 low metastatic breast cancer whose disease has progressed on endocrine therapy in the metastatic setting: a randomized, global phase 3 trial (DESTINY-Breast06). Cancer Res. 2021; 81: OT-03–09.
    OpenUrl
  146. 146.↵
    1. Kurokawa H,
    2. Arteaga CL.
    Inhibition of erbB receptor (HER) tyrosine kinases as a strategy to abrogate antiestrogen resistance in human breast cancer. Clin Cancer Res. 2001; 7: 4436s–42s; discussion 11s-12s.
    OpenUrlAbstract/FREE Full Text
  147. 147.↵
    1. Geyer CE,
    2. Forster J,
    3. Lindquist D,
    4. Chan S,
    5. Romieu CG,
    6. Pienkowski T, et al.
    Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006; 355: 2733–43.
    OpenUrlCrossRefPubMed
  148. 148.↵
    1. Moulder SL,
    2. Borges VF,
    3. Baetz T,
    4. McSpadden T,
    5. Fernetich G,
    6. Murthy RK, et al.
    Phase I study of ONT-380, a HER2 inhibitor, in patients with HER2+-advanced solid tumors, with an expansion cohort in HER2+ metastatic breast cancer (MBC). Clin Cancer Res. 2017; 23: 3529–36.
    OpenUrlAbstract/FREE Full Text
  149. 149.↵
    1. Olson DJ,
    2. Kulukian A,
    3. Taylor JD,
    4. Zaval MC,
    5. Nesterova A,
    6. Hensley KM, et al.
    Abstract 1962: Preclinical characterization of tucatinib in HER2-amplified xenograft and CNS implanted tumors. Cancer Res. 2020; 80: 1962.
    OpenUrlCrossRef
  150. 150.↵
    1. Curigliano G,
    2. Mueller V,
    3. Borges V,
    4. Hamilton E,
    5. Hurvitz S,
    6. Loi S, et al.
    Tucatinib versus placebo added to trastuzumab and capecitabine for patients with pretreated HER2+ metastatic breast cancer with and without brain metastases (HER2CLIMB): final overall survival analysis. Ann Oncol. 2022; 33: 321–29.
    OpenUrlCrossRefPubMed
  151. 151.↵
    1. Yu Y,
    2. Yang Y,
    3. Li H,
    4. Fan Y.
    Targeting HER2 alterations in non-small cell lung cancer: therapeutic breakthrough and challenges. Cancer Treat Rev. 2023; 114: 102520.
  152. 152.↵
    1. Yan M,
    2. Ouyang Q,
    3. Sun T,
    4. Niu L,
    5. Yang J,
    6. Li L, et al.
    Pyrotinib plus capecitabine for patients with human epidermal growth factor receptor 2-positive breast cancer and brain metastases (PERMEATE): a multicentre, single-arm, two-cohort, phase 2 trial. Lancet Oncol. 2022; 23: 353–61.
    OpenUrlPubMed
  153. 153.↵
    1. Xu B,
    2. Yan M,
    3. Ma F,
    4. Hu X,
    5. Feng J,
    6. Ouyang Q, et al.
    Pyrotinib plus capecitabine versus lapatinib plus capecitabine for the treatment of HER2-positive metastatic breast cancer (PHOEBE): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2021; 22: 351–60.
    OpenUrlCrossRefPubMed
  154. 154.↵
    1. Shyam Sunder S,
    2. Sharma UC,
    3. Pokharel S.
    Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther. 2023; 8: 262.
    OpenUrlPubMed
  155. 155.↵
    1. Leon-Ferre RA,
    2. Goetz MP.
    Advances in systemic therapies for triple negative breast cancer. BMJ. 2023; 381: e071674.
  156. 156.↵
    1. Asleh K,
    2. Riaz N,
    3. Nielsen TO.
    Heterogeneity of triple negative breast cancer: current advances in subtyping and treatment implications. J Exp Clin Cancer Res. 2022; 41: 265.
    OpenUrlCrossRefPubMed
  157. 157.↵
    1. Postow MA,
    2. Callahan MK,
    3. Wolchok JD.
    Immune checkpoint blockade in cancer therapy. J Clin Oncol. 2015; 33: 1974–82.
    OpenUrlAbstract/FREE Full Text
  158. 158.↵
    1. Chen L,
    2. Jiang YZ,
    3. Wu SY,
    4. Wu J,
    5. Di GH,
    6. Liu GY, et al.
    Famitinib with camrelizumab and nab-paclitaxel for advanced immunomodulatory triple-negative breast cancer (FUTURE-C-Plus): an open-label, single-arm, phase II trial. Clin Cancer Res. 2022; 28: 2807–17.
    OpenUrlPubMed
  159. 159.↵
    1. Starodub AN,
    2. Ocean AJ,
    3. Shah MA,
    4. Guarino MJ,
    5. Picozzi VJ Jr.,
    6. Vahdat LT, et al.
    First-in-human trial of a novel anti-trop-2 antibody-SN-38 conjugate, sacituzumab govitecan, for the treatment of diverse metastatic solid tumors. Clin Cancer Res. 2015; 21: 3870–8.
    OpenUrlAbstract/FREE Full Text
  160. 160.↵
    1. Tarantino P,
    2. Tolaney SM,
    3. Curigliano G.
    Trastuzumab deruxtecan (T-DXd) in HER2-low metastatic breast cancer treatment. Ann Oncol. 2023; 34: 949–50.
    OpenUrlPubMed
  161. 161.↵
    1. Jiang Y-Z,
    2. Liu Y,
    3. Xiao Y,
    4. Hu X,
    5. Jiang L,
    6. Zuo W-J, et al.
    Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial. Cell Res. 2021; 31: 178–86.
    OpenUrlCrossRefPubMed
  162. 162.↵
    1. Pascual J,
    2. Turner NC.
    Targeting the PI3-kinase pathway in triple-negative breast cancer. Ann Oncol. 2019; 30: 1051–60.
    OpenUrlPubMed
  163. 163.↵
    1. Savas P,
    2. Lo LL,
    3. Luen SJ,
    4. Blackley EF,
    5. Callahan J,
    6. Moodie K, et al.
    Alpelisib monotherapy for PI3K-altered, pretreated advanced breast cancer: a phase II study. Cancer Discov. 2022; 12: 2058–73.
    OpenUrlPubMed
  164. 164.↵
    1. Eikesdal HP,
    2. Yndestad S,
    3. Elzawahry A,
    4. Llop-Guevara A,
    5. Gilje B,
    6. Blix ES, et al.
    Olaparib monotherapy as primary treatment in unselected triple negative breast cancer. Ann Oncol. 2021; 32: 240–49.
    OpenUrlCrossRefPubMed
  165. 165.↵
    1. Robson M,
    2. Im SA,
    3. Senkus E,
    4. Xu B,
    5. Domchek SM,
    6. Masuda N, et al.
    Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017; 377: 523–33.
    OpenUrlCrossRefPubMed
  166. 166.↵
    1. Turner NC,
    2. Telli ML,
    3. Rugo HS,
    4. Mailliez A,
    5. Ettl J,
    6. Grischke EM, et al.
    A phase II study of talazoparib after platinum or cytotoxic nonplatinum regimens in patients with advanced breast cancer and germline BRCA1/2 mutations (ABRAZO). Clin Cancer Res. 2019; 25: 2717–24.
    OpenUrlAbstract/FREE Full Text
  167. 167.↵
    1. Litton JK,
    2. Rugo HS,
    3. Ettl J,
    4. Hurvitz SA,
    5. Gonçalves A,
    6. Lee KH, et al.
    Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018; 379: 753–63.
    OpenUrlCrossRefPubMed
  168. 168.↵
    1. Buchtel KM,
    2. Vogel Postula KJ,
    3. Weiss S,
    4. Williams C,
    5. Pineda M,
    6. Weissman SM.
    FDA approval of PARP inhibitors and the impact on genetic counseling and genetic testing practices. J Genet Couns. 2018; 27: 131–39.
    OpenUrlPubMed
  169. 169.↵
    1. Deveson IW,
    2. Gong B,
    3. Lai K,
    4. LoCoco JS,
    5. Richmond TA,
    6. Schageman J, et al.
    Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology. Nat Biotechnol. 2021; 39: 1115–28.
    OpenUrlPubMed
  170. 170.↵
    1. Brooks TG,
    2. Lahens NF,
    3. Mrčela A,
    4. Grant GR.
    Challenges and best practices in omics benchmarking. Nat Rev Genet. 2024; 25: 326–39.
    OpenUrlCrossRefPubMed
  171. 171.↵
    1. Haue AD,
    2. Hjaltelin JX,
    3. Holm PC,
    4. Placido D,
    5. Brunak SR.
    Artificial intelligence-aided data mining of medical records for cancer detection and screening. Lancet Oncol. 2024; 25: e694–703.
    OpenUrlPubMed
  172. 172.↵
    1. Yang RQ,
    2. Lou KL,
    3. Wang PY,
    4. Gao YY,
    5. Zhang YQ,
    6. Chen M, et al.
    Surgical navigation for malignancies guided by near-infrared-II fluorescence imaging. Small Methods. 2021; 5: e2001066.
  173. 173.↵
    1. Wardell CM,
    2. Boardman DA,
    3. Levings MK.
    Harnessing the biology of regulatory T cells to treat disease. Nat Rev Drug Discov. 2024; 24: 93–111.
    OpenUrlPubMed
  174. 174.↵
    1. Tani H.
    Recent advances and prospects in RNA drug development. Int J Mol Sci. 2024; 25: 12284.
  175. 175.↵
    1. Ganatra H,
    2. Tan JK,
    3. Simmons A,
    4. Bigogno CM,
    5. Khurana V,
    6. Ghose A, et al.
    Applying whole-genome and whole-exome sequencing in breast cancer: a review of the landscape. Breast Cancer. 2024; 31: 999–1009.
    OpenUrlPubMed
  176. 176.↵
    1. Xu G,
    2. Yu J,
    3. Lyu J,
    4. Zhan M,
    5. Xu J,
    6. Huang M, et al.
    Proteogenomic landscape of breast ductal carcinoma reveals tumor progression characteristics and therapeutic targets. Adv Sci (Weinh). 2024; 11: e2401041.
  177. 177.
    1. Moreno-Ulloa A,
    2. Zárate-Córdova VL,
    3. Ramírez-Sánchez I,
    4. Cruz-López JC,
    5. Perez-Ortiz A,
    6. Villarreal-Garza C, et al.
    Evaluation of a proteomics-guided protein signature for breast cancer detection in breast tissue. J Proteome Res. 2024; 23: 4907–23.
    OpenUrlPubMed
  178. 178.↵
    1. Johnston LE,
    2. Randall J,
    3. Chouraichi S,
    4. Luu M,
    5. Hunt AL,
    6. Mauro L, et al.
    Proteomics based selection achieves complete response to HER2 therapy in HER2 IHC 0 breast cancer. NPJ Precis Oncol. 2024; 8: 203.
    OpenUrlPubMed
  179. 179.↵
    1. Lusby R,
    2. Demirdizen E,
    3. Inayatullah M,
    4. Kundu P,
    5. Maiques O,
    6. Zhang Z, et al.
    Pan-cancer drivers of metastasis. Mol Cancer. 2025; 24: 2.
    OpenUrlPubMed
  180. 180.↵
    1. Jia G,
    2. Chen Z,
    3. Ping J,
    4. Cai Q,
    5. Tao R,
    6. Li C, et al.
    Refining breast cancer genetic risk and biology through multi-ancestry fine-mapping analyses of 192 risk regions. Nat Genet. 2025; 57: 80–7.
    OpenUrlCrossRefPubMed
  181. 181.↵
    1. Vaida M,
    2. Arumalla KK,
    3. Tatikonda PK,
    4. Popuri B,
    5. Bux RA,
    6. Tappia PS, et al.
    Identification of a novel biomarker panel for breast cancer screening. Int J Mol Sci. 2024; 25: 11835.
  182. 182.
    1. Jiménez-Franco A,
    2. Jiménez-Aguilar JM,
    3. Canela-Capdevila M,
    4. García-Pablo R,
    5. Castañé H,
    6. Martínez-Navidad C, et al.
    Preliminary metabolomics study suggests favorable metabolic changes in the plasma of breast cancer patients after surgery and adjuvant treatment. Biomedicines. 2024; 12: 2196.
    OpenUrlPubMed
  183. 183.
    1. Wu HC,
    2. Lai Y,
    3. Liao Y,
    4. Deyssenroth M,
    5. Miller GW,
    6. Santella RM, et al.
    Plasma metabolomics profiles and breast cancer risk. Breast Cancer Res. 2024; 26: 141.
    OpenUrlPubMed
  184. 184.↵
    1. Arumalla KK,
    2. Haince JF,
    3. Bux RA,
    4. Huang G,
    5. Tappia PS,
    6. Ramjiawan B, et al.
    Metabolomics-based machine learning models accurately predict breast cancer estrogen receptor status. Int J Mol Sci. 2024; 25: 13029.
  185. 185.↵
    1. Wang T,
    2. Wang S,
    3. Li Z,
    4. Xie J,
    5. Chen H,
    6. Hou J.
    Machine learning-informed liquid-liquid phase separation for personalized breast cancer treatment assessment. Front Immunol. 2024; 15: 1485123.
  186. 186.
    1. Gerratana L,
    2. Davis AA,
    3. Foffano L,
    4. Reduzzi C,
    5. Rossi T,
    6. Medford A, et al.
    Integrating machine learning-predicted circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in metastatic breast cancer: a proof of principle study on endocrine resistance profiling. Cancer Lett. 2025; 609: 217325.
  187. 187.↵
    1. Gao Y,
    2. Ventura-Diaz S,
    3. Wang X,
    4. He M,
    5. Xu Z,
    6. Weir A, et al.
    An explainable longitudinal multi-modal fusion model for predicting neoadjuvant therapy response in women with breast cancer. Nat Commun. 2024; 15: 9613.
    OpenUrlPubMed
  188. 188.↵
    1. Frascarelli C,
    2. Venetis K,
    3. Marra A,
    4. Mane E,
    5. Ivanova M,
    6. Cursano G, et al.
    Deep learning algorithm on H&E whole slide images to characterize TP53 alterations frequency and spatial distribution in breast cancer. Comput Struct Biotechnol J. 2024; 23: 4252–59.
    OpenUrlPubMed
  189. 189.↵
    1. Rashid MM,
    2. Selvarajoo K.
    Advancing drug-response prediction using multi-modal and -omics machine learning integration (MOMLIN): a case study on breast cancer clinical data. Brief Bioinform. 2024; 25: bbae300.
  190. 190.↵
    1. Ma X,
    2. Sun P,
    3. Zhang ZY.
    An integrative framework for protein interaction network and methylation data to discover epigenetic modules. IEEE/ACM Trans Comput Biol Bioinform. 2019; 16: 1855–66.
    OpenUrl
  191. 191.↵
    1. Llaguno-Roque JL,
    2. Barrientos-Martínez RE,
    3. Acosta-Mesa HG,
    4. Barranca-Enríquez A,
    5. Mezura-Montes E,
    6. Romo-González T.
    Use and comparison of machine learning techniques to discern the protein patterns of autoantibodies present in women with and without breast pathology. J Proteome Res. 2025; 24: 289–302.
    OpenUrlPubMed
  192. 192.↵
    1. Zhang Z,
    2. Fu L,
    3. Yun B,
    4. Wang X,
    5. Wang X,
    6. Wu Y, et al.
    Differentially localized protein identification for breast cancer based on deep learning in immunohistochemical images. Commun Biol. 2024; 7: 935.
    OpenUrlPubMed
  193. 193.↵
    1. Tao Z,
    2. Chyra Z,
    3. Kotulová J,
    4. Celichowski P,
    5. Mihályová J,
    6. Charvátová S, et al.
    Impact of T cell characteristics on CAR-T cell therapy in hematological malignancies. Blood Cancer J. 2024; 14: 213.
    OpenUrlPubMed
  194. 194.↵
    1. Serrano García L,
    2. Jávega B,
    3. Llombart Cussac A,
    4. Gión M,
    5. Pérez-García JM,
    6. Cortés J, et al.
    Patterns of immune evasion in triple-negative breast cancer and new potential therapeutic targets: a review. Front Immunol. 2024; 15: 1513421.
  195. 195.↵
    1. Han E,
    2. Choi HY,
    3. Kwon HJ,
    4. Chung YR,
    5. Shin HC,
    6. Kim EK, et al.
    Characterization of tumor-infiltrating lymphocytes and their spatial distribution in triple-negative breast cancer. Breast Cancer Res. 2024; 26: 180.
    OpenUrlPubMed
  196. 196.
    1. Chen XY,
    2. Liu Y,
    3. Zhu WB,
    4. Li SH,
    5. Wei S,
    6. Cai J, et al.
    Arming oncolytic M1 virus with gasdermin E enhances antitumor efficacy in breast cancer. iScience. 2024; 27: 111148.
  197. 197.
    1. Kim HS,
    2. Youn YH,
    3. Kim HJ,
    4. Koo YH,
    5. Lee J,
    6. Kwon IK, et al.
    Enhanced antitumor efficacy of oncolytic vaccinia virus therapy through keratin-mediated delivery in triple-negative breast cancer. Int J Mol Sci. 2024; 25: 11470.
  198. 198.↵
    1. Tang S,
    2. Lyles KV,
    3. Wang Y,
    4. Fan D,
    5. Luo M.
    Enhancing the efficacy of breast cancer immunotherapy using a Smac-armed oncolytic virus. Cancers. 2024; 16: 3248.
    OpenUrlPubMed
  199. 199.↵
    1. Shariati A,
    2. Khani P,
    3. Nasri F,
    4. Afkhami H,
    5. Khezrpour A,
    6. Kamrani S, et al.
    mRNA cancer vaccines from bench to bedside: a new era in cancer immunotherapy. Biomark Res. 2024; 12: 157.
    OpenUrlPubMed
  200. 200.↵
    1. Li R,
    2. Hu JC,
    3. Rong L,
    4. He Y,
    5. Wang X,
    6. Lin X, et al.
    The guided fire from within: intratumoral administration of mRNA-based vaccines to mobilize memory immunity and direct immune responses against pathogen to target solid tumors. Cell Discov. 2025; 10: 127.
    OpenUrlPubMed
  201. 201.↵
    1. Zhang M,
    2. Wang Y,
    3. Li B,
    4. Yang B,
    5. Zhao M,
    6. Li B, et al.
    STING-activating polymers boost lymphatic delivery of mRNA vaccine to potentiate cancer immunotherapy. Adv Mater. 2024; e2412654.
  202. 202.↵
    1. Shi Y,
    2. Shi M,
    3. Wang Y,
    4. You J.
    Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther. 2024; 9: 322.
    OpenUrlPubMed
  203. 203.↵
    1. Zhang Z,
    2. Ma X,
    3. La Y,
    4. Guo X,
    5. Chu M,
    6. Bao P, et al.
    Advancements in the application of scRNA-seq in breast research: a review. Int J Mol Sci. 2024; 25: 13706.
  204. 204.↵
    1. Hitchen N,
    2. Shahnam A,
    3. Tie J.
    Circulating tumor DNA: a pan-cancer biomarker in solid tumors with prognostic and predictive value. Annu Rev Med. 2024; 76: 207–23.
    OpenUrl
  205. 205.↵
    1. McCaffrey C,
    2. Jahangir C,
    3. Murphy C,
    4. Burke C,
    5. Gallagher WM,
    6. Rahman A.
    Artificial intelligence in digital histopathology for predicting patient prognosis and treatment efficacy in breast cancer. Expert Rev Mol Diagn. 2024; 24: 363–77.
    OpenUrlPubMed
  206. 206.↵
    1. Zhong R,
    2. Zhang Y,
    3. Qiu W,
    4. Zhang K,
    5. Feng Q,
    6. Cao X, et al.
    Quantitative histopathology analysis based on label-free multiphoton imaging for breast cancer diagnosis and neoadjuvant immunotherapy response assessment. Int J Biol Sci. 2025; 21: 363–81.
    OpenUrlPubMed
  207. 207.↵
    1. Bai JW,
    2. Qiu SQ,
    3. Zhang GJ.
    Molecular and functional imaging in cancer-targeted therapy: current applications and future directions. Signal Transduct Target Ther. 2023; 8: 89.
    OpenUrlPubMed
  208. 208.↵
    1. Chen W,
    2. Zhang Y,
    3. Zhang L,
    4. Luo X,
    5. Yang X,
    6. Zhu Y, et al.
    Intraoperative evaluation of tumor margins using a TROP2 near-infrared imaging probe to enable human breast-conserving surgery. Sci Transl Med. 2024; 16: eado2461.
  209. 209.
    1. Zhu YY,
    2. Song L,
    3. Zhang YQ,
    4. Liu WL,
    5. Chen WL,
    6. Gao WL, et al.
    Development of a rare earth nanoprobe enables in vivo real-time detection of sentinel lymph node metastasis of breast cancer using NIR-IIb imaging. Cancer Res. 2023; 83: 3428–41.
    OpenUrlPubMed
  210. 210.
    1. Zhang YQ,
    2. Liu WL,
    3. Luo XJ,
    4. Shi JP,
    5. Zeng YZ,
    6. Chen WL, et al.
    Novel self-assembled multifunctional nanoprobes for second-near-infrared-fluorescence-image-guided breast cancer surgery and enhanced radiotherapy efficacy. Adv Sci (Weinh). 2023; 10: e2205294.
  211. 211.
    1. Yang RQ,
    2. Wang PY,
    3. Lou KL,
    4. Dang YY,
    5. Tian HN,
    6. Li Y, et al.
    Biodegradable nanoprobe for NIR-II fluorescence image-guided surgery and enhanced breast cancer radiotherapy efficacy. Adv Sci (Weinh). 2022; 9: e2104728.
  212. 212.↵
    1. Liu JJ,
    2. Wang Z,
    3. Nie LM,
    4. Zhu YY,
    5. Li G,
    6. Lin LL, et al.
    RGD-functionalised melanin nanoparticles for intraoperative photoacoustic imaging-guided breast cancer surgery. Eur J Nucl Med Mol Imaging. 2022; 49: 847–60.
    OpenUrlPubMed
  213. 213.↵
    1. Deng H,
    2. Lei Q,
    3. Wang C,
    4. Wang Z,
    5. Chen H,
    6. Wang G, et al.
    A fluorogenic probe for predicting treatment response in non-small cell lung cancer with EGFR-activating mutations. Nat Commun. 2022; 13: 6944.
    OpenUrlPubMed
  214. 214.↵
    1. Wei M,
    2. Bai J,
    3. Shen X,
    4. Lou K,
    5. Gao Y,
    6. Lv R, et al.
    Glutathione-exhausting nanoprobes for NIR-II fluorescence imaging-guided surgery and boosting radiation therapy efficacy via ferroptosis in breast cancer. ACS Nano. 2023; 17: 11345–61.
    OpenUrlPubMed
  215. 215.↵
    1. Banerjee S,
    2. Booth CM,
    3. Bruera E,
    4. Büchler MW,
    5. Drilon A,
    6. Fry TJ, et al.
    Two decades of advances in clinical oncology – lessons learned and future directions. Nat Rev Clin Oncol. 2024; 21: 771–80.
    OpenUrlPubMed
  216. 216.↵
    1. Cheng HH,
    2. Shevach JW,
    3. Castro E,
    4. Couch FJ,
    5. Domchek SM,
    6. Eeles RA, et al.
    BRCA1, BRCA2, and associated cancer risks and management for male patients: a review. JAMA Oncol. 2024; 10: 1272–81.
    OpenUrlPubMed
  217. 217.↵
    1. Miricescu D,
    2. Totan A,
    3. Stanescu-Spinu II,
    4. Badoiu SC,
    5. Stefani C,
    6. Greabu M.
    PI3K/AKT/mTOR signaling pathway in breast cancer: from molecular landscape to clinical aspects. Int J Mol Sci. 2020; 22: 173.
    OpenUrlCrossRefPubMed
  218. 218.↵
    1. Sayour EJ,
    2. Boczkowski D,
    3. Mitchell DA,
    4. Nair SK.
    Cancer mRNA vaccines: clinical advances and future opportunities. Nat Rev Clin Oncol. 2024; 21: 489–500.
    OpenUrlCrossRefPubMed
  219. 219.↵
    1. Gao YY,
    2. Yang RQ,
    3. Lou KL,
    4. Dang YY,
    5. Dong YY,
    6. He YY, et al.
    In vivo visualization of fluorescence reflecting CDK4 activity in a breast cancer mouse model. MedComm. 2022; 3: e136.
PreviousNext
Back to top

In this issue

Cancer Biology & Medicine: 22 (4)
Cancer Biology & Medicine
Vol. 22, Issue 4
15 Apr 2025
  • Table of Contents
  • Index by author
Print
Download PDF
Email Article

Thank you for your interest in spreading the word on Cancer Biology & Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The treatment of breast cancer in the era of precision medicine
(Your Name) has sent you a message from Cancer Biology & Medicine
(Your Name) thought you would like to see the Cancer Biology & Medicine web site.
Citation Tools
The treatment of breast cancer in the era of precision medicine
Jingwen Bai, Yiyang Gao, Guojun Zhang
Cancer Biology & Medicine Apr 2025, 22 (4) 322-347; DOI: 10.20892/j.issn.2095-3941.2024.0510

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
The treatment of breast cancer in the era of precision medicine
Jingwen Bai, Yiyang Gao, Guojun Zhang
Cancer Biology & Medicine Apr 2025, 22 (4) 322-347; DOI: 10.20892/j.issn.2095-3941.2024.0510
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Precision medicine in early breast cancer (EBC)
    • Precision medicine in ABC
    • The future and emerging frontiers of precision medicine in breast cancer
    • Conclusions
    • Conflict of interest statement
    • Author contributions
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • References
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • Senescent macrophages in cancer: roles in tumor progression and treatment opportunities
  • Advances in strategies to improve the immunotherapeutic efficacy of chimeric antigen receptor-T cell therapy for lymphoma
  • Application and future prospects of bispecific antibodies in the treatment of non-small cell lung cancer
Show more Review

Similar Articles

Keywords

  • Breast cancer
  • precision medicine
  • diagnostic precision
  • personalized therapy

Navigate

  • Home
  • Current Issue

More Information

  • About CBM
  • About CACA
  • About TMUCIH
  • Editorial Board
  • Subscription

For Authors

  • Instructions for authors
  • Journal Policies
  • Submit a Manuscript

Journal Services

  • Email Alerts
  • Facebook
  • RSS Feeds
  • Twitter

 

© 2025 Cancer Biology & Medicine

Powered by HighWire