Abstract
Triple-negative breast cancer (TNBC) represents a particularly aggressive and heterogeneous subtype of breast cancer, associated with poor prognosis and limited treatment options. This review delves into the rising incidence of TNBC, particularly among younger women, and explores the significant demographic disparities that contribute to variations in incidence, treatment access and survival outcomes. We provide a discussion of TNBC’s molecular and genetic landscape, including key pathways revolving around TP53, BRCA1/2 mutations, and PI3K/AKT signaling, which have informed the development of targeted therapies. Recent practice-changing studies are highlighted, which have resulted in the integration of immune checkpoint inhibitors in both early-stage and metastatic settings, the application of PARP inhibitors for BRCA-mutated TNBC and the introduction of antibody-drug conjugates as valuable new therapeutic options. We also review the role of neoadjuvant chemotherapy, novel biomarkers such as tumor-infiltrating lymphocytes, and advancements in diagnostic tools, including machine learning-based imaging and spatial transcriptomics, which are all driving shifts towards personalized approaches. This review synthesizes emerging research and major changes in clinical practice to provide a concise overview of the recent innovations and upcoming trends in TNBC diagnosis and therapy.
Keywords: TNBC, oncology, diagnosis, therapeutics, medicine, genomics, immunotherapy
Introduction
Breast cancer remains one of the most common malignancies affecting women worldwide, representing a major portion of cancer-related morbidity and mortality.1 Among the various subtypes, triple negative breast cancer (TNBC) stands out due to its aggressive behavior and poor prognosis. The American Society of Clinical Oncology and College of American Pathologists guidelines define TNBC as cancers where estrogen receptor (ER) and progesterone receptor (PR) are less than 1% by immunohistochemistry (IHC), and human epidermal growth factor receptor 2 (HER2) is either 0 to 1+ by IHC, or IHC 2+ with negative fluorescence in situ hybridization (FISH).2,3 These cancers represent approximately 10–15% of all breast cancers.4 Localized or early-stage cancers of all subtypes can be approached with curative intent by surgical resection and postoperative radiation if lumpectomy is performed, typically followed by systemic therapy. Metastatic breast cancer is treated with systemic therapy according to subtype, with the goal of preventing further growth or spread. Unlike other subtypes that can benefit from hormonal or HER2-directed therapies, TNBC lacks these specific targets, which for many years made conventional chemotherapy regimens the only practical treatment options. This aggressive subtype is associated with lower rates of cure and poorer overall survival compared to ER/PR-positive or HER2-positive breast cancers.5,6 The heterogeneity of TNBC also contributes to its complexity, with varying molecular subtypes influencing prognosis and treatment response.7,8 Advancements in the understanding of TNBC’s unique biology have brought about promising new treatment strategies. Molecular profiling has revealed distinct subtypes within TNBC, opening opportunities for more personalized approaches.9 Targeted therapies and immunotherapies have in recent years emerged as practice-changing avenues, offering new hope for patients with this daunting subtype. This review will explore the genomic and epidemiologic landscapes of TNBC, examine recent breakthroughs, highlight disparities in incidence and treatment outcomes and discuss future directions in research and clinical practice.
Rising Incidence of TNBC
Despite improvements in rates of breast cancer survival, recent epidemiologic evidence points to a concerning increase in the incidence of multiple forms of breast cancer, including TNBC. This recent surge is notable among younger women, reflecting broader trends in cancer incidence in adolescents and young adults.10–14 Studies highlight that women born after 1980 have higher rates of breast cancer, and that a higher proportion of these represent TNBC, compared to those born before 1950.15,16 TNBC thus constitutes a larger proportion of breast cancers among younger women as compared to other age groups, emphasizing the need for targeted prevention and consideration of alternative early detection strategies in this demographic. International trends also show a rise in breast cancer among adolescents and young adults worldwide.12,17–20
Genotypic Associations and Racial Disparities
Genetic and molecular heterogeneity of TNBC present major challenges for suitable therapeutics. Genomic studies have identified several key mutations and pathways associated with TNBC, including alterations in TP53, BRCA1 and BRCA2 genes, as well as cellular pathways including PI3K/AKT, MYC and DNA repair mechanisms.21–24 These genetic insights have fueled the development of targeted therapies and personalized approaches that have reached clinical practice and remain under investigation in clinical trials, as discussed below. A particularly striking aspect of TNBC is that significant racial and ethnic disparities exist in its incidence and outcomes. Black women are disproportionately affected by TNBC, exhibiting incidence rates nearly double that of White counterparts.4 This disparity is thought to be driven by a complex interplay of genetic, socioeconomic and healthcare access factors. Genetic studies have revealed that certain mutations, such as those in the BRCA1 gene are more prevalent in Black women.24 Another recent study revealed a distinct DNA methylation pattern present in Black women diagnosed before age 50, suggesting a distinct epigenetic profile among this group.25 Regardless of these genetic factors, a robust body of work underscores that Black women experience higher breast cancer mortality rates due to later stage diagnosis, limited access to adequate screening, and financial barriers to completing optimal treatment.26–33 Consequently, survival rates are lower among Black women at every stage and every breast cancer subtype as compared to White women. Addressing these gaps is a critical health priority that demands urgent multidisciplinary advocacy and action. The medical and scientific communities must extend opportunities for patient education and engagement, facilitate accessible screening for all, and actively involve minoritized populations in medical research studies, and support the inclusion of specialized race-congruent staff at all levels.34
Therapeutic Advances in Early-Stage TNBC
Remarkable progress has been made in the management of early-stage TNBC with the addition of neoadjuvant chemotherapy and immunotherapy. Neoadjuvant therapy, which is administered before surgical resection, has become a cornerstone in the treatment of this aggressive subtype. This approach not only helps reduce tumor size, making surgery more effective, but also provides an early indication of how the tumor responds to systemic therapy, which is crucial for tailoring subsequent treatment strategies. Neoadjuvant chemotherapy has become a standard treatment pathway in eligible patients with triple-negative tumors that measure 2 cm or greater or involve lymph nodes, and can also be employed for early HER2-positive and advanced ER/PR-positive tumors.
The advent of immune checkpoint inhibitors marked a major breakthrough in the treatment of early-stage TNBC. The Phase 3 KEYNOTE-522 trial assessed the efficacy of pembrolizumab, a programmed death-1 (PD-1) inhibitor, versus placebo in combination with neoadjuvant chemotherapy. This trial showed that addition of neoadjuvant and adjuvant pembrolizumab to sequential neoadjuvant chemotherapy significantly improved pathological complete response (pCR) rates (64.8 vs 51.2%).35 This also yielded the benefit of avoiding morbid complete axillary lymph node dissections in more patients. Subsequent analysis also demonstrated benefits in 3-year event-free survival (EFS, 84.5 vs 76.8%) and 5-year overall survival (OS, 86.6 vs 81.7%).36 This regimen has been widely accepted into clinical practice with follow-up studies generally supporting its use in this setting.37–39 Some groups have observed significant limitations in implementing this regimen into practice, often related to significant immune toxicity or disparities in access to care.37–43 The ongoing phase 3 OptimICE-PCR trial aims to assess whether 1 year of adjuvant pembrolizumab significantly influences outcomes after obtaining a pCR, and this may help avoid unneeded exposure to immunotherapy in this group.44 Patients with residual disease after neoadjuvant therapy and surgery can continue on 6 to 8 cycles of adjuvant capecitabine, based on results of the CREATE-X trial, which found PFS and OS benefits over control in patients who did not achieve a pCR after neoadjuvant chemotherapy.45 These patients are often offered to complete 1 year of pembrolizumab concurrently with capecitabine, though data are limited regarding combined treatment.
Targeted therapy has also made a recent stride in the treatment of early-stage TNBC. The phase 3 OlympiA trial randomized patients with HER2-negative early breast cancer and BRCA1 or BRCA2 germline pathogenic variants to receive adjuvant olaparib.46 Treatment with the poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor, which targets defective homologous recombination repair of DNA damage, resulted in an increase in invasive disease-free survival of 85.9 vs 77.1%. The Phase 2–3 PARTNER trial later found no significant difference in pCR, EFS or OS in TNBC patients with wildtype germline BRCA1 and BRCA2 randomized to receive neoadjuvant olaparib or placebo in addition to chemoimmunotherapy.47 Results for the germline mutated BRCA1/2 cohort are still pending.
Surgical Management in Early-Stage and Recurrent TNBC
Surgical strategies for triple-negative breast cancer (TNBC) are increasingly guided by tumor biology, treatment response, and recurrence patterns. Evidence supports that breast-conserving surgery (BCS), when combined with adjuvant radiotherapy, is a safe and effective option for many patients with TNBC. A meta-analysis involving over 19,000 patients demonstrated that BCS with radiotherapy provides comparable or better outcomes than mastectomy, with significantly lower odds of locoregional recurrence (LRR), distant metastasis (DM), and all-cause mortality.48 Notably, patients with smaller tumors were more likely to undergo BCS, highlighting the importance of careful patient selection at this stage. Similarly, a retrospective study found that BCS with radiotherapy achieved oncological outcomes equivalent to mastectomy across various tumor and nodal risk subclasses, even in cases with larger tumors where oncoplastic techniques were utilized to optimize local control.49 Emerging work has also explored alternatives to traditional surgical approaches. A phase 2 trial evaluated the omission of surgery in patients achieving a pathological complete response (pCR) to neoadjuvant systemic therapy, confirmed by vacuum-assisted core biopsy. Among the 50 enrolled patients (42% with TNBC), no ipsilateral breast tumor recurrences were observed in the 31 patients who achieved pCR and thus received radiotherapy alone.50 In cases of ipsilateral breast cancer recurrence (IBCR), salvage mastectomy has traditionally been the standard approach. However, a recent retrospective study indicates that a second BCS may yield better distant disease-free survival (DDFS), OS, and breast cancer-specific survival (BCSS) compared to mastectomy for patients with small recurrent TNBC tumors.51 These results are largely in line with a systematic review including patients with various breast cancer subtypes, which demonstrated that repeat BCS for IBCR is associated with a higher rate of second local recurrence compared to salvage mastectomy but offers a small overall survival benefit.52 Together these studies all support the expanding role of BCS in TNBC and highlight the importance of careful patient selection, with tumor biology and response to prior treatment serving as critical determinants of the surgical approach.
Metastatic TNBC
In patients who experience a recurrence of their cancer after surgery/radiation or distant spread is noted at diagnosis, single-agent chemotherapy regimens incorporating anthracyclines or taxanes have been the mainstay of first-line therapy for many years. Incorporation of immune checkpoint inhibitors as initial treatment for metastatic TNBC came with results of KEYNOTE-355. This phase 3 trial found an overall survival benefit with addition of pembrolizumab to chemotherapy for patients with PD-L1 combined positive score (CPS) of 10 or greater, whereas the benefit was not seen for tumors with lower PD-L1 expression.53 This strategy and PD-L1 testing has now become standard practice as first-line therapy for metastatic TNBC. Pembrolizumab monotherapy did not show a statistically significant improvement in metastatic TNBC outcomes over single-agent chemotherapy in the phase 3 KEYNOTE-119 trial.54 In cases where PD-L1 CPS is less than 10 with a germline BRCA1/2 mutation, PARP inhibitors olaparib and talazoparib have shown benefits in progression-free survival and can be recommended as first-line therapy.55,56
The other notable advance in this setting came with the antibody-drug conjugate sacituzumab govitecan, which was studied in the phase 3 ASCENT trial. This agent is composed of an antibody targeting trophoblast cell surface antigen (TROP)2 coupled to the irinotecan active metabolite SN-38, essentially facilitating targeted chemotherapy delivery.57 TROP2 is a protein present in high levels in various cancers, including over 90% of breast cancer cells. Delivery of the cell-permeable drug SN-38 has also been shown to elicit antitumor effects on neighboring tumor cells through a process termed bystander effect. Similarly, the phase 3 DESTINY-Breast04 trial advanced trastuzumab deruxtecan as another second-line option for patients with triple-negative HER2-low disease.58 Unfortunately, despite these advances, survival outcomes remain quite limited for metastatic disease and further investigative efforts are needed for this vulnerable population.
Emerging Diagnostics and Future Directions
Preclinical and clinical phase investigations continue to identify potential new avenues to diagnose, treat and predict response to therapy in TNBC. Imaging tools play a critical role in the initial detection of cancer and ongoing refinement in modalities, including optimal screening and clinical course prediction, will be essential to maximize benefit to the most patients. Ultrasound, magnetic resonance imaging (MRI) and positron emission tomography (PET) are the most widely employed imaging modalities in breast cancer diagnosis, and significant interest has developed in understanding features of TNBC as an adjunct to traditional pathologic analysis or even independently.59 With recent advances in machine learning algorithms and applicability, these models are more commonly being applied to provide highly accurate predictions of outcomes such as likelihood of pCR and are likely to provide significant insights in the realms of TNBC and oncology at large.60–62
The notion and potential of tumor-infiltrating lymphocytes (TILs) in various cancers have been postulated for several years, however their applicability as a prognostic or therapeutic has been unclear. Retrospective studies have recently provided evidence that TIL abundance can predict overall survival in early-stage TNBC.63,64 Similarly, multiple preclinical and early-stage clinical investigations are underway studying the suitability of various forms of intratumoral immunotherapy, including viral vector-based, antibodies or innate immune agonists.65 Technical progress in single cell RNA sequencing and spatial transcriptomics have also enormously expanded our ability to visualize and probe gene expression shifts in fascinating multi-dimensional representations, and gain mechanistic understanding behind treatment response or unresponsiveness.66,67 These studies will propel oncologic diagnosis and treatment towards a much needed, highly personalized approach for patients with TNBC. Developments in molecular profiling and gene signature assays can also provide indications of likely tumor trajectories and responses to therapy.68,69 Likewise, deeper basic science interrogations of the genomic and epigenomic landscapes of various cancers including TNBC and viral infections have dissected intricacies of chromosomal instability, DNA damage sensing and interferon signaling, which may provide explanations for responsiveness to immunotherapy and other new lines of treatment.70–74
Finally, the following phase 2 trials modeled as components of I-SPY2.2 engaged a novel neoadjuvant sequential therapy platform, previously employed in I-SPY2,75 as a means to efficiently identify therapies likely to succeed in phase 3 trials.76 Randomization was stratified into 8 subtypes, defined by receptor status and a 70-gene signature based on a commercially available assay. The trials evaluated efficacy of datopotamab-deruxtecan (Dato-DXd), an antibody-drug conjugate targeting TROP2, both alone76 and in combination with durvalumab,77 a PD-L1 inhibitor, in patients with high-risk stage 2/3 breast cancer. The results showed that Dato-DXd alone did not meet the prespecified criteria for advancing to phase 3 trials in any breast cancer subtype, but the combination treatment graduated in the “immune-positive” subtype. Dato-DXd is now recommended as an option for second-line treatment of patients with metastatic ER/PR-positive HER2-negative breast cancer based on results of the TROPION-Breast01 trial but has not yet received an indication in TNBC.78 Additional trials are ongoing to assess the use of Dato-DXd in locally recurrent inoperable or metastatic TNBC (TROPION-Breast02), in conjunction with durvalumab for residual TNBC after neoadjuvant therapy at time of surgery (TROPION-Breast03) and as neoadjuvant therapy for TNBC in conjunction with durvalumab (TROPION-Breast04).79–81 Perhaps most notably, the I-SPY2.2 trial design incorporates up to 3 blocks of distinct treatments, allowing real-time adjustments based on MRI and biopsy results to optimize patient outcomes and minimize exposure to ineffective agents.82
Conclusion
The landscape of TNBC is rapidly evolving with significant advancements in both our understanding of its molecular underpinnings and the development of novel therapeutic strategies. Integration of immune checkpoint inhibitors and targeted therapies has opened new avenues for treatment, offering hope for improved outcomes in this challenging subtype. However, the persistence of health disparities and real-world challenges in treatment administration underscore the need for a more personalized and equitable approach to care. Future molecular insights, such as deeper understanding of tumor heterogeneity, dynamic genomic changes and immune microenvironment interactions, have the potential to revolutionize TNBC treatment algorithms. Ongoing research into mechanisms of immune interactions with the use of single-cell sequencing, epigenomic profiling and spatial transcriptomics could enable further therapeutic personalization and may result in development of novel immune cell therapies applicable across a variety of cancer types. Continued biomedical research and clinical trials are essential to refine these strategies, enhance diagnostic accuracy, and ultimately improve survival rates for all patients with TNBC. The future of TNBC management depends on the integration of cutting-edge science, precision oncology approaches and a commitment to eliminating health inequities.
Disclosure
The authors report no conflicts of interest in this work.
References
- 1.Siegel RL, Kratzer TB, Giaquinto AN, Sung H, Jemal A. Cancer statistics, 2025. CA Cancer J Clin. 2025;75(1):10–45. doi: 10.3322/caac.21871 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Wolff AC, Somerfield MR, Dowsett M, et al. Human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med. 2023;147(9):993–1000. doi: 10.5858/arpa.2023-0950-SA [DOI] [PubMed] [Google Scholar]
- 3.Allison KH, Hammond MEH, Dowsett M, et al. Estrogen and progesterone receptor testing in breast cancer: ASCO/CAP guideline update. J Clin Oncol. 2020;38(12):1346–1366. doi: 10.1200/JCO.19.02309 [DOI] [PubMed] [Google Scholar]
- 4.Giaquinto AN, Sung H, Newman LA, et al. Breast cancer statistics 2024. CA Cancer J Clin. 2024;74(6):477–495. doi: 10.3322/caac.21863 [DOI] [PubMed] [Google Scholar]
- 5.Li X, Yang J, Peng L, et al. Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res Treat. 2017;161(2):279–287. doi: 10.1007/s10549-016-4059-6 [DOI] [PubMed] [Google Scholar]
- 6.Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321(3):288–300. doi: 10.1001/jama.2018.19323 [DOI] [PubMed] [Google Scholar]
- 7.Lehmann BD, Jovanovic B, Chen X, et al. Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS One. 2016;11(6):e0157368. doi: 10.1371/journal.pone.0157368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Garrido-Castro AC, Lin NU, Polyak K. Insights into molecular classifications of triple-negative breast cancer: improving patient selection for treatment. Cancer Discov. 2019;9(2):176–198. doi: 10.1158/2159-8290.CD-18-1177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bianchini G, De Angelis C, Licata L, Gianni L. Treatment landscape of triple-negative breast cancer — expanded options, evolving needs. Nat Rev Clin Oncol. 2022;19(2):91–113. doi: 10.1038/s41571-021-00565-2 [DOI] [PubMed] [Google Scholar]
- 10.Sung H, Siegel RL, Rosenberg PS, Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. 2019;4(3):e137–e147. doi: 10.1016/S2468-2667(18)30267-6 [DOI] [PubMed] [Google Scholar]
- 11.American Cancer Society. Special Section: Cancer in Adolescents and Young Adults. Atlanta: American Cancer Society; 2020. [Google Scholar]
- 12.Ugai T, Sasamoto N, Lee H-Y, et al. Is early-onset cancer an emerging global epidemic? Current evidence and future implications. Nat Rev Clin Oncol. 2022;19(10):656–673. doi: 10.1038/s41571-022-00672-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Koh B, Tan DJH, Ng CH, et al. Patterns in cancer incidence among people younger than 50 years in the US, 2010 to 2019. JAMA Network Open. 2023;6(8):e2328171. doi: 10.1001/jamanetworkopen.2023.28171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.American Cancer Society. Cancer Facts & Figures 2025. Atlanta: American Cancer Society; 2025. [Google Scholar]
- 15.Di Martino E, Smith L, Bradley SH, et al. Incidence trends for twelve cancers in younger adults-a rapid review. Br J Cancer. 2022;126(10):1374–1386. doi: 10.1038/s41416-022-01704-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sung H, Jiang C, Bandi P, et al. Differences in cancer rates among adults born between 1920 and 1990 in the USA: an analysis of population-based cancer registry data. Lancet Public Health. 2024;9(8):e583–e593. doi: 10.1016/S2468-2667(24)00156-7 [DOI] [PubMed] [Google Scholar]
- 17.Ghoncheh M, Pournamdar Z, Salehiniya H. Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev. 2016;17(S3):43–46. doi: 10.7314/apjcp.2016.17.s3.43 [DOI] [PubMed] [Google Scholar]
- 18.Almansour NM. Triple-negative breast cancer: a brief review about epidemiology, risk factors, signaling pathways, treatment and role of artificial intelligence. Front Mol Biosci. 2022;9:836417. doi: 10.3389/fmolb.2022.836417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kim J, Harper A, McCormack V, et al. Global patterns and trends in breast cancer incidence and mortality across 185 countries. Nature Med. 2025;31(4):1154–1162. doi: 10.1038/s41591-025-03502-3 [DOI] [PubMed] [Google Scholar]
- 20.Tang DD, Ye ZJ, Liu WW, et al. Survival feature and trend of female breast cancer: a comprehensive review of survival analysis from cancer registration data. Breast. 2025;79:103862. doi: 10.1016/j.breast.2024.103862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ademuyiwa FO, Chen I, Luo J, et al. Immunogenomic profiling and pathological response results from a clinical trial of docetaxel and carboplatin in triple-negative breast cancer. Breast Cancer Res Treat. 2021;189(1):187–202. doi: 10.1007/s10549-021-06307-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Galappaththi SPL, Smith KR, Alsatari ES, et al. The genomic and biologic landscapes of breast cancer and racial differences. Int J Mol Sci. 2024;25(23):13165. doi: 10.3390/ijms252313165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Khan MA, Jain VK, Rizwanullah M, Ahmad J, Jain K. PI3K/AKT/mTOR pathway inhibitors in triple-negative breast cancer: a review on drug discovery and future challenges. Drug Discovery Today. 2019;24(11):2181–2191. doi: 10.1016/j.drudis.2019.09.001 [DOI] [PubMed] [Google Scholar]
- 24.Pop L-A, Cojocneanu-Petric R-M, Pileczki V, et al. Genetic alterations in sporadic triple negative breast cancer. Breast. 2018;38:30–38. doi: 10.1016/j.breast.2017.11.006 [DOI] [PubMed] [Google Scholar]
- 25.Ensenyat-Mendez M, Solivellas-Pieras M, Llinas-Arias P, et al. Epigenetic profiles of triple-negative breast cancers of African American and white females. JAMA Network Open. 2023;6(10):e2335821. doi: 10.1001/jamanetworkopen.2023.35821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cho B, Han Y, Lian M, et al. Evaluation of racial/ethnic differences in treatment and mortality among women with triple-negative breast cancer. JAMA Oncol. 2021;7(7):1016–1023. doi: 10.1001/jamaoncol.2021.1254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jatoi I, Sung H, Jemal A. The emergence of the racial disparity in U.S. breast-cancer mortality. N Engl J Med. 2022;386(25):2349–2352. doi: 10.1056/NEJMp2200244 [DOI] [PubMed] [Google Scholar]
- 28.Ko NY, Hong S, Winn RA, Calip GS. Association of insurance status and racial disparities with the detection of early-stage breast cancer. JAMA Oncol. 2020;6(3):385–392. doi: 10.1001/jamaoncol.2019.5672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lubarsky M, Hernandez AE, Collie BL, et al. Does structural racism impact receipt of NCCN guideline-concordant breast cancer treatment? Breast Cancer Res Treat. 2024;206(3):509–517. doi: 10.1007/s10549-024-07245-6 [DOI] [PubMed] [Google Scholar]
- 30.Miller-Kleinhenz JM, Collin LJ, Seidel R, et al. Racial disparities in diagnostic delay among women with breast cancer. J Am College Radiol. 2021;18(10):1384–1393. doi: 10.1016/j.jacr.2021.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Saka AH, Giaquinto AN, McCullough LE, et al. Cancer statistics for African American and Black people, 2025. CA Cancer J Clin. 2025;75(2):111–140. doi: 10.3322/caac.21874 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Siddharth S, Sharma D. Racial disparity and triple-negative breast cancer in African-American women: a multifaceted affair between obesity, biology, and socioeconomic determinants. Cancers. 2018;10(12):514. doi: 10.3390/cancers10120514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Warnecke RB, Campbell RT, Vijayasiri G, Barrett RE, Rauscher GH. Multilevel examination of health disparity: the role of policy implementation in neighborhood context. Patient Resources, and in Healthcare Facilities on Later Stage of Breast Cancer Diagnosis Cancer Epidemiol Biomarkers Prev. 2019;28(1):59–66. doi: 10.1158/1055-9965.EPI-17-0945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Pleasant V. A public health emergency: breast cancer among black communities in the United States. Obstetrics and Gynecology Clinics of North America. 2024;51(1):69–103. doi: 10.1016/j.ogc.2023.11.001 [DOI] [PubMed] [Google Scholar]
- 35.Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382(9):810–821. doi: 10.1056/NEJMoa1910549 [DOI] [PubMed] [Google Scholar]
- 36.Schmid P, Cortes J, Dent R, et al. Overall survival with pembrolizumab in early-stage triple-negative breast cancer. N Engl J Med. 2024;391(21):1981–1991. doi: 10.1056/NEJMoa2409932 [DOI] [PubMed] [Google Scholar]
- 37.LeVee A, Wong M, Flores S, et al. Impact of neoadjuvant pembrolizumab adherence on pathologic complete response in triple-negative breast cancer: a real-world analysis. Oncologist. 2024;29(7):566–574. doi: 10.1093/oncolo/oyae064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Villacampa G, Navarro V, Matikas A, et al. Neoadjuvant immune checkpoint inhibitors plus chemotherapy in early breast cancer: a systematic review and meta-analysis. JAMA Oncol. 2024;10(10):1331–1341. doi: 10.1001/jamaoncol.2024.3456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Connors C, Valente SA, ElSherif A, et al. Real-world outcomes with the KEYNOTE-522 regimen in early-stage triple-negative breast cancer. Ann Surg Oncol. 2025;32(2):912–921. doi: 10.1245/s10434-024-16390-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Downs-Canner S, Mittendorf EA. Preoperative immunotherapy combined with chemotherapy for triple-negative breast cancer: perspective on the KEYNOTE-522 study. Ann Surg Oncol. 2023;30(6):3166–3169. doi: 10.1245/s10434-023-13267-z [DOI] [PubMed] [Google Scholar]
- 41.Kok M, Gielen RJ, Adams S, et al. Academic uphill battle to personalize treatment for patients with stage II/III triple-negative breast cancer. J Clin Oncol. 2024;42(30):3523–3529. doi: 10.1200/JCO.24.00372 [DOI] [PubMed] [Google Scholar]
- 42.Woriax HE, Thomas SM, Plichta JK, et al. Racial/ethnic disparities in pathologic complete response and overall survival in patients with triple-negative breast cancer treated with neoadjuvant chemotherapy. J Clin Oncol. 2024;42(14):1635–1645. doi: 10.1200/JCO.23.01199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Rizzo A, Schipilliti FM, Di Costanzo F, et al. Discontinuation rate and serious adverse events of chemoimmunotherapy as neoadjuvant treatment for triple-negative breast cancer: a systematic review and meta-analysis. ESMO Open. 2023;8(6):102198. doi: 10.1016/j.esmoop.2023.102198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tolaney S, Ballman K, Perou CM, et al. Abstract PO4-19-05: optimICE-pCR: de-escalation of therapy in early-stage TNBC patients who achieve pCR after neoadjuvant chemotherapy with checkpoint inhibitor therapy (Alliance A012103). Cancer Res. 2024;84(9_Supplement):PO4–19–05–PO4–19–05. doi: 10.1158/1538-7445.Sabcs23-po4-19-05 [DOI] [Google Scholar]
- 45.Masuda N, Lee SJ, Ohtani S, et al. Adjuvant capecitabine for breast cancer after preoperative chemotherapy. N Engl J Med. 2017;376(22):2147–2159. doi: 10.1056/NEJMoa1612645 [DOI] [PubMed] [Google Scholar]
- 46.Tutt ANJ, Garber JE, Kaufman B, et al. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med. 2021;384(25):2394–2405. doi: 10.1056/NEJMoa2105215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Abraham JE, Pinilla K, Dayimu A, et al. The PARTNER trial of neoadjuvant olaparib with chemotherapy in triple-negative breast cancer. Nature. 2024;629(8014):1142–1148. doi: 10.1038/s41586-024-07384-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fancellu A, Houssami N, Sanna V, Porcu A, Ninniri C, Marinovich ML. Outcomes after breast-conserving surgery or mastectomy in patients with triple-negative breast cancer: meta-analysis. Br J Surg. 2021;108(7):760–768. doi: 10.1093/bjs/znab145 [DOI] [PubMed] [Google Scholar]
- 49.Di Leone A, Franco A, Zotta F, et al. Local treatment of triple-negative breast cancer: is mastectomy superior to breast-conserving surgery? J Pers Med. 2023;13(5):865. doi: 10.3390/jpm13050865 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kuerer HM, Smith BD, Krishnamurthy S, et al. Eliminating breast surgery for invasive breast cancer in exceptional responders to neoadjuvant systemic therapy: a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2022;23(12):1517–1524. doi: 10.1016/S1470-2045(22)00613-1 [DOI] [PubMed] [Google Scholar]
- 51.Gentile D, Sagona A, Spoto R, et al. Salvage mastectomy is not the treatment of choice for aggressive subtypes of ipsilateral breast cancer recurrence: a single-institution retrospective study. Eur J Breast Health. 2022;18(4):315–322. doi: 10.4274/ejbh.galenos.2022.2022-5-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Tollan CJ, Pantiora E, Valachis A, Karakatsanis A, Tasoulis MK. A systematic review and meta-analysis on the role of repeat breast-conserving surgery for the management of ipsilateral breast cancer recurrence. Ann Surg Oncol. 2022;29(10):6440–6453. doi: 10.1245/s10434-022-12197-6 [DOI] [PubMed] [Google Scholar]
- 53.Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022;387(3):217–226. doi: 10.1056/NEJMoa2202809 [DOI] [PubMed] [Google Scholar]
- 54.Winer EP, Lipatov O, Im S-A, et al. Pembrolizumab versus investigator-choice chemotherapy for metastatic triple-negative breast cancer (KEYNOTE-119): a randomised, open-label, phase 3 trial. Lancet Oncol. 2021;22(4):499–511. doi: 10.1016/S1470-2045(20)30754-3 [DOI] [PubMed] [Google Scholar]
- 55.Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523–533. doi: 10.1056/NEJMoa1706450 [DOI] [PubMed] [Google Scholar]
- 56.Litton JK, Rugo HS, Ettl J, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379(8):753–763. doi: 10.1056/NEJMoa1802905 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bardia A, Hurvitz SA, Tolaney SM, et al. Sacituzumab govitecan in metastatic triple-negative breast cancer. N Engl J Med. 2021;384(16):1529–1541. doi: 10.1056/NEJMoa2028485 [DOI] [PubMed] [Google Scholar]
- 58.Modi S, Jacot W, Yamashita T, et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N Engl J Med. 2022;387(1):9–20. doi: 10.1056/NEJMoa2203690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Adrada BE, Moseley TW, Kapoor MM, et al. Triple-negative breast cancer: histopathologic features, genomics, and treatment. Radiographics. 2023;43(10):e230034. doi: 10.1148/rg.230034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Corredor G, Bharadwaj S, Pathak T, Viswanathan VS, Toro P, Madabhushi A. A review of AI-based radiomics and computational pathology approaches in triple-negative breast cancer: current applications and perspectives. Clin Breast Cancer. 2023;23(8):800–812. doi: 10.1016/j.clbc.2023.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Guo J, Hu J, Zheng Y, Zhao S, Ma J. Artificial intelligence: opportunities and challenges in the clinical applications of triple-negative breast cancer. Br J Cancer. 2023;128(12):2141–2149. doi: 10.1038/s41416-023-02215-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Yin H-L, Jiang Y, Xu Z, Jia -H-H, Lin G-W. Combined diagnosis of multiparametric MRI-based deep learning models facilitates differentiating triple-negative breast cancer from fibroadenoma magnetic resonance BI-RADS 4 lesions. J Cancer Res Clin Oncol. 2023;149(6):2575–2584. doi: 10.1007/s00432-022-04142-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Leon-Ferre RA, Jonas SF, Salgado R, et al. Tumor-infiltrating lymphocytes in triple-negative breast cancer. JAMA. 2024;331(13):1135–1144. doi: 10.1001/jama.2024.3056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Obeagu EI, Obeagu GU. Lymphocyte infiltration in breast cancer: a promising prognostic indicator. Medicine. 2024;103(49):e40845. doi: 10.1097/MD.0000000000040845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mantooth SM, Abdou Y, Saez-Ibanez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol. 2024;15:1385484. doi: 10.3389/fimmu.2024.1385484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Shiao SL, Gouin KH, Ing N, et al. Single-cell and spatial profiling identify three response trajectories to pembrolizumab and radiation therapy in triple negative breast cancer. Cancer Cell. 2024;42(1):70–84e8. doi: 10.1016/j.ccell.2023.12.012 [DOI] [PubMed] [Google Scholar]
- 67.Wang XQ, Danenberg E, Huang C-S, et al. Spatial predictors of immunotherapy response in triple-negative breast cancer. Nature. 2023;621(7980):868–876. doi: 10.1038/s41586-023-06498-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Denkert C, Schneeweiss A, Rey J, et al. Molecular adaptation to neoadjuvant immunotherapy in triple-negative breast cancer. Cell Rep Med. 2024;5(11):101825. doi: 10.1016/j.xcrm.2024.101825 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dugo M, Huang C-S, Egle D, et al. The immune-related 27-gene signature determaio predicts response to neoadjuvant atezolizumab plus chemotherapy in triple-negative breast cancer. Clin Cancer Res. 2024;30(21):4900–4909. doi: 10.1158/1078-0432.CCR-24-0149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Agelidis A, Suryawanshi RK, Patil CD, Campeau A, Gonzalez DJ, Shukla D. Dissociation of DNA damage sensing by endoglycosidase HPSE. iScience. 2021;24(3):102242. doi: 10.1016/j.isci.2021.102242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Cho M-G, Kumar RJ, Lin -C-C, et al. MRE11 liberates cGAS from nucleosome sequestration during tumorigenesis. Nature. 2024;625(7995):585–592. doi: 10.1038/s41586-023-06889-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Funnell T, O’Flanagan CH, Williams MJ, et al. Single-cell genomic variation induced by mutational processes in cancer. Nature. 2022;612(7938):106–115. doi: 10.1038/s41586-022-05249-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Houlahan KE, Mangiante L, Sotomayor-Vivas C, et al. Complex rearrangements fuel ER(+) and HER2(+) breast tumours. Nature. 2025:638(8050):510–518. doi: 10.1038/s41586-024-08377-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li J, Hubisz MJ, Earlie EM, et al. Non-cell-autonomous cancer progression from chromosomal instability. Nature. 2023;620(7976):1080–1088. doi: 10.1038/s41586-023-06464-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Harrington D, Parmigiani G. I-SPY 2 — a glimpse of the future of phase 2 drug development? N Engl J Med. 2016;375(1):7–9. doi: 10.1056/NEJMp1602256 [DOI] [PubMed] [Google Scholar]
- 76.Khoury K, Meisel JL, Yau C, et al. Datopotamab-deruxtecan in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial. Nat Med. 2024;30(12):3728–3736. doi: 10.1038/s41591-024-03266-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shatsky RA, Trivedi MS, Yau C, et al. Datopotamab–deruxtecan plus durvalumab in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial. Nat Med. 2024;30(12):3737–3747. doi: 10.1038/s41591-024-03267-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bardia A, Jhaveri K, Im SA, et al. Datopotamab deruxtecan versus chemotherapy in previously treated inoperable/metastatic hormone receptor-positive human epidermal growth factor receptor 2-negative breast cancer: primary results from TROPION-breast01. J Clin Oncol. 2025;43(3):285–296. doi: 10.1200/JCO.24.00920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dent RA, Cescon DW, Bachelot T, et al. TROPION-Breast02: datopotamab deruxtecan for locally recurrent inoperable or metastatic triple-negative breast cancer. Future Oncol. 2023;19(35):2349–2359. doi: 10.2217/fon-2023-0228 [DOI] [PubMed] [Google Scholar]
- 80.Bardia A, Pusztai L, Albain K, et al. TROPION-Breast03: a randomized Phase III global trial of datopotamab deruxtecan ± durvalumab in patients with triple-negative breast cancer and residual invasive disease at surgical resection after neoadjuvant therapy. Ther Adv Med Oncol. 2024;16:17588359241248336. doi: 10.1177/17588359241248336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.McArthur HL, Tolaney SM, Dent R, et al. TROPION-Breast04: a randomized phase III study of neoadjuvant datopotamab deruxtecan (Dato-DXd) plus durvalumab followed by adjuvant durvalumab versus standard of care in patients with treatment-naïve early-stage triple negative or HR-low/HER2- breast cancer. Ther Adv Med Oncol. 2025;17:17588359251316176. doi: 10.1177/17588359251316176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Perez-Garcia J, Antonarelli G, Gion M, Llombart-Cussac A, Cortes J. Moving toward response-adapted trials in oncology. Nat Med. 2024;30(12):3426–3428. doi: 10.1038/s41591-024-03346-3 [DOI] [PubMed] [Google Scholar]