Advertisement
100th Anniversary Viewpoints
Open Access | 10.1172/JCI187520
1Section of Hematology and Oncology, Department of Medicine and
2Center for Clinical Cancer Genetics & Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
Address correspondence to: Olufunmilayo I. Olopade, 5841 S. Maryland Avenue, MC 2115, Chicago, Illinois 60637, USA. Phone: 773.702.1632; Email: folopade@medicine.bsd.uchicago.edu.
Find articles by Wu, S. in: JCI | PubMed | Google Scholar
1Section of Hematology and Oncology, Department of Medicine and
2Center for Clinical Cancer Genetics & Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
Address correspondence to: Olufunmilayo I. Olopade, 5841 S. Maryland Avenue, MC 2115, Chicago, Illinois 60637, USA. Phone: 773.702.1632; Email: folopade@medicine.bsd.uchicago.edu.
Find articles by Zheng, Y. in: JCI | PubMed | Google Scholar
1Section of Hematology and Oncology, Department of Medicine and
2Center for Clinical Cancer Genetics & Global Health, Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
Address correspondence to: Olufunmilayo I. Olopade, 5841 S. Maryland Avenue, MC 2115, Chicago, Illinois 60637, USA. Phone: 773.702.1632; Email: folopade@medicine.bsd.uchicago.edu.
Find articles by Olopade, O. in: JCI | PubMed | Google Scholar
Published November 1, 2024 - More info
Breast cancer is the most diagnosed cancer among women worldwide, with an estimated 2.3 million new cases and 670,000 deaths reported in 2022 (1). Advances in genomic research have heralded a new era in precision medicine, enabling personalized treatment and risk assessment based on molecular profiles. Next-generation sequencing and deep-learning (DL) algorithms have transformed breast cancer care by facilitating the analysis of complex and extensive datasets, with the potential to democratize access to omics-informed clinical trials.
A comprehensive understanding of the heterogeneity and complexity of breast cancer across diverse populations, along with intricate disease mechanisms, is fueling innovations aimed at personalizing breast cancer care and reducing global disparities in outcomes.
For decades, the treatment of breast cancer has been based on classification according to estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status (2–5). However, advances in genomics technologies have underscored the critical role of molecular profiling in determining prognosis and guiding treatment strategies. For patients with early stage breast cancer, genomic information enables more accurate predictions of recurrence and metastasis risk, identifies molecular subtypes of the disease, and helps prevent overtreatment by refining traditional therapeutic approaches (Table 1). Furthermore, discovering pathogenic variants in BRCA1 DNA repair associated (BRCA1) and BRCA2 genes has been pivotal to our understanding of both hereditary and sporadic breast cancers. These variants largely increase breast and ovarian cancer risks and identify patients who may benefit from targeted therapies like poly (ADP-ribose) polymerase (PARP) inhibitors. The OlympiA trials have shown that olaparib effectively reduces recurrence risk in patients with BRCA-mutated breast cancer, an example of enhanced outcomes through personalized care for high-risk individuals (6). For hormone receptor–positive (HR+) breast cancers, the addition of CDK4/6 inhibitors to hormonal therapies offers a more potent treatment, particularly for patients with high disease burden or progression on prior endocrine therapy. Selective ER degraders are a class of drugs representing a substantial advance in treating HR+ breast cancers by effectively degrading ER and inhibiting tumor growth. In contrast, HER2-positive breast cancers, known for their aggressive behavior, respond favorably to targeted therapies such as trastuzumab, pertuzumab, and tyrosine kinase inhibitors.
Recent innovations in antibody-drug conjugates (ADCs), such as trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have notably improved survival outcomes for HER2-positive patients (7, 8). The FDA approval of T-DXd for unresectable or metastatic HER2-positive cases underscores the critical role of genomic and molecular profiling in treatment decisions. While these advances herald a promising future for breast cancer therapy, especially for a recently defined subset of HER2-low breast cancer, the challenge of broadening access to these drugs for rare subtypes of breast cancer remains. Triple-negative breast cancer (TNBC) is a particularly aggressive disease with limited treatment options. Immunotherapeutic agents like pembrolizumab have expanded treatment possibilities for PD-(L)1-positive TNBC, offering new hope for patients, though further research is needed to fully evaluate their impact (9). The development of Trop2-based ADCs also holds promise for improving TNBC outcomes (10). Applying artificial intelligence (AI) and DL algorithms to analyze the tumor microenvironment (TME) can substantially enhance ADC development by identifying therapeutic targets and optimizing antibody design. Integrating genetic, pharmacogenomic, and histopathologic data can further improve the selection of antibody targets and cytotoxic payloads, making ADCs more versatile therapeutic options across multiple cancer types.
Precision oncology integrated with germline genetic testing has transformed breast cancer care by revealing the genetic underpinnings of disease development, recurrence risk, and treatment resistance. These advances facilitate more personalized cancer treatments tailored to each patient’s unique genetic and molecular profiles.
A milestone in the molecular characterization of breast cancer was the discovery of the BRCA1 and BRCA2 genes. BRCA1 pathogenic variants, enriched in basal-like/TNBC, and the epigenetic silencing of BRCA1, which impairs DNA repair, play significant roles in cancer progression (11). The advancement of polygenic risk scores (PRSs) based on breast cancer genetic variants identified by GWASs allows for more personalized breast cancer screening, which supports early detection and efficient prevention strategies (12–14). Integrating genomic, epigenomic, and pseudogene expression data with clinical features holds promise for developing precise prognostic models to personalize treatment and accelerate progress in prevention and early detection, with potential to reduce disparities in breast cancer outcomes when appropriately deployed for population health management (15).
Given the paucity of omics data from non-European ancestry populations, large datasets from initiatives such as The Cancer Genome Atlas and the International Cancer Genome Consortium have provided comprehensive catalogs of genomic aberrations that inform clinical management and precision medicine. The genomic landscape of breast cancer is notably heterogeneous, including mutations, copy number variations, and structural alterations that affect tumor progression and treatment response (16). Advances in high-throughput sequencing and computational biology have identified differences in genomic alterations associated with aggressive breast cancer progression and metastasis in diverse populations. Notable findings in the mutational landscape include the mutually exclusive presence of GATA3 and TP53/PIK3CA pathogenic variants, higher levels of intratumoral heterogeneity, and genomic instability in breast cancer among Nigerian women, as well as the discovery of a unique breast cancer subtype defined by early clonal pathogenic variants in GATA3 and a younger age at diagnosis (17). The TP53 gene is the most frequently mutated in breast cancer, particularly in TNBC and HER2-positive subtypes. Patients with TP53-mutant breast cancer typically face a higher risk of recurrence, poorer overall survival, and a greater likelihood of metastasis compared with those with wild-type TP53. There is an urgent need to identify upstream lifestyle and environmental risk factors for specific mutational signatures. Various clinical trials currently explore WEE1 and MDM2/MDMX inhibitors for TP53-mutated cancers. Additionally, PIK3CA pathogenic variants, common in HR+ breast cancers, activate the PI3K/AKT/mTOR pathway and are effectively targeted by alpelisib combined with endocrine therapy. Mutations in other genes, such as PTEN, AKT1, and ESR1, guide the development of novel inhibitors and combination therapies to overcome resistance mechanisms.
While genomics offers insights into mutations and genetic variability, proteomics translates these changes into the biological mechanisms that drive cancer progression, drug resistance, and treatment response. Initiatives like the Clinical Proteomic Tumor Analysis Consortium significantly enhance our understanding of genetic alterations at the protein level. Integrative approaches, including digital pathology, provide a comprehensive view of the molecular landscape, fostering the creation of highly effective, individualized treatment strategies. Modern techniques now allow for the simultaneous assessment of multiple biomarkers, revealing complex interactions within the TME. These advances will be crucial for developing personalized treatments, deepening our understanding of cellular diversity and refining prognostic and predictive assessments, ultimately improving therapies and biomarker development in the future (18).
AI-based imaging algorithms and DL models enhance the accuracy and efficiency of cancer detection, assisting radiologists in identifying breast lesions (19). AI also automates breast-density assessments, enabling earlier diagnosis and intervention. By integrating imaging data with patient-specific omics profiles, computational methods can aid in developing personalized treatment plans (20). AI could offer high accuracy in classifying cancerous tissues, underscoring its critical role in diagnostic precision and tailored treatments (21). Furthermore, radiogenomics links imaging features with genetic mutations to predict molecular pathway activity and optimize treatment. For example, tumor texture heterogeneity on imaging can indicate ERBB2 amplification, guiding the use of trastuzumab or pertuzumab and highlighting the importance of the TME.
The TME, composed of a diverse array of cell types in a dynamic and intricate structure surrounding the tumor, plays a critical role in immune modulation, making it a crucial target for advancing more effective immunotherapeutic strategies. Alterations in the TME can influence the release of tumor-derived materials, including circulating tumor cells, cell-free DNA, and exosomes, into the bloodstream. Analyzing these components provides valuable insights into tumor dynamics and treatment response. Liquid biopsy, utilizing advanced cell identification and single-cell sequencing, serves as a noninvasive tool to track disease progression and detect resistance mutations with high sensitivity and specificity. For example, detecting ESR1 mutations in HR+ breast cancer can predict resistance to aromatase inhibitors, guiding a switch to therapies such as fulvestrant.
Breast cancer presents a complex landscape for precision medicine, with challenges arising from the disease’s intricacies and the advanced technologies required to address them. Precision medicine, grounded in genetic insights, offers promise, through integrating germline and somatic profiling to guide treatment decisions. Additionally, genetic testing raises ethical concerns, such as incidental findings and potential privacy issues, requiring education for healthcare providers and an informed public to participate in innovative clinical trials.
Despite these challenges, AI-based integrative research and advanced computational methods offer potential approaches to identify novel biomarkers and optimize breast cancer treatment regimens. Innovative clinical trial designs, such as platform and basket studies, enable validation of these biomarkers. Technologies such as single-cell RNA sequencing provide deeper insights into TME heterogeneity and resistance mechanisms. The convergence of AI, genomics, immunotherapy, and digital pathology is set to revolutionize breast cancer treatment, paving the way for more personalized and effective therapies. Key to our success will be high quality cancer care powered by affordable access to health insurance, innovative clinical trial design, and community-engaged research so that every person has a fighting chance to survive and thrive after a diagnosis of breast cancer, the most common female cancer in the world.
This work was supported by an NIH grant (R01 MD013452 to OIO) and a Susan G. Komen grant (SAC210203 to OIO).
Address correspondence to: Olufunmilayo I. Olopade, 5841 S. Maryland Avenue, MC 2115, Chicago, Illinois 60637, USA. Phone: 773.702.1632; Email: folopade@medicine.bsd.uchicago.edu.
Conflict of interest: OIO serves on the advisory boards for CancerIQ and Tempus.
Copyright: © 2024, Wu et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.
Reference information:J Clin Invest. 2024;134(21):e187520. https://doi.org/10.1172/JCI187520.
Solid organ transplantation: solid but not yet spectacularLaurence A. Turka
Leptin physiology and pathophysiology: knowns and unknowns 30 years after its discoveryJeffrey S. Flier et al.
The GLP-1 journey: from discovery science to therapeutic impactDaniel J. Drucker
CAR T cells for hematological malignanciesBarbara Savoldo et al.
Clinical investigation of hypoxia-inducible factors: getting thereGregg L. Semenza
Pancreatic β cell function versus insulin resistance: application of the hyperbolic law of glucose toleranceRichard N. Bergman
Nitric oxide in vascular biology: elegance in complexityJoseph Loscalzo
How the JCI’s most-cited paper sparked the field of lipoprotein researchMichael S. Brown et al.
SGLT2 inhibitors: cardiorenal metabolic drugs for the agesRalph A. DeFronzo
G protein–coupled receptors: from radioligand binding to cellular signalingHoward A. Rockman et al.
Bisphosphonates for osteoporosis: from bench to clinicTeresita Bellido
Pandemics past, present, and future: progress and persistent risksArturo Casadevall
Major breakthroughs in hematopoietic stem cell transplantation and future challenges in clinical implementationLeslie S. Kean et al.
Life-saving effect of pulmonary surfactant in premature babiesJ. Usha Raj et al.
Advancing chemokine research: the molecular function of CXCL8Yiran Hou et al.
Navigating an enigma: the continuing journey of autoimmunity discoveriesMariana J. Kaplan
Defining and targeting mechanisms of eosinophilic inflammation in a new era of severe asthma treatmentJoshua A. Boyce et al.
The other pandemic: lessons from 40 years of HIV researchMary E. Klotman et al.
A half-century of VEGFA: from theory to practiceSusan E. Quaggin
Two decades of advances in preeclampsia research: molecular mechanisms and translational studiesS. Ananth Karumanchi
The forgotten pandemic: how understanding cholera illuminated mechanisms of chloride channels in multiple diseasesQais Al-Awqati
Is it time to rethink the relationship between adipose inflammation and insulin resistance?Evan D. Rosen et al.
Checkpoint therapy in cancer treatment: progress, challenges, and future directionsMesude Bicak et al.
The arc of discovery, from the description of cystic fibrosis to effective treatmentsMichael J. Welsh
Intertwining clonality and resistance: Staphylococcus aureus in the antibiotic eraHenry F. Chambers et al.
The expanding application of antisense oligonucleotides to neurodegenerative diseasesCharlotte J. Sumner et al.
Toward a better understanding of chronic hepatitis B virus infectionBarbara Rehermann