Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 1.
Published in final edited form as: Cancer J. 2021 Jan-Feb;27(1):76–82. doi: 10.1097/PPO.0000000000000503

Triple-negative breast cancer central nervous system metastases from the laboratory to the clinic

Alexandra S Zimmer 1
PMCID: PMC7839270  NIHMSID: NIHMS1652706  PMID: 33475296

Abstract

Triple-negative breast cancer accounts for 15-20% of breast cancers and has an incidence as high as 50% of brain metastases once patients develop advanced disease. The lack of targeted and effective therapies, characteristic of this subtype of breast cancer, is especially evident once CNS metastases occur. Compared to other subtypes of breast cancer, TNBC patients have the shorter interval from diagnosis to development of brain metastases and the shorter overall survival once they occur, a median of 4-6months. Preclinical studies of TNBC and CNS microenvironment are actively ongoing, clarifying mechanisms and orienting more effective approaches to therapy. While the first drugs have been specifically approved for use in metastatic TNBC, data on their CNS effect is still awaited.

Keywords: TNBC, brain metastasis, CNS involvement, PARP inhibitors, sacituzumab-govitecan, immune checkpoint inhibitors

Introduction

Breast cancer is the most prevalent cancer among women and the second most common cause of brain metastases 1. Up to 30% of breast cancer patients develop brain metastases and the risk is subtype specific, being higher in human epidermal growth factor receptor 2 (HER2)-positive and triple-negative breast cancer (TNBC) 2,3. TNBC subtype is defined as hormone-receptor and HER2-negative, and accounts for 15-20% of breast cancers overall. In the genomic or molecular classification analysis, TNBC overlaps ~80% with the basal-like subtype 4. Once metastatic, half of the patients in this subgroup will develop brain metastases 5. Moreover, the overall incidence of breast cancer brain metastases is reportedly increasing, likely due to multiple factors such as better imaging techniques, increased screening and enhanced long term survival with improved cancer therapy 6,7. While in the HER2-positive subtype several options of targeted therapy provide a good systemic control and progressive improvement in overall survival (OS) despite brain metastases, in the TNBC subtype, the aggressiveness and difficult systemic control of the metastatic disease are possibly related to the high incidence of central nervous system (CNS) involvement. Consequently, understanding the pathology and improving treatment of brain metastases in TNBC is of utmost importance.

Epidemiology and natural history

TNBC has appeared in medical literature as a new entity since around 2005, characterized by its overall aggressiveness and lack of available targeted therapy, and accounting for 12-17% of breast cancer cases 8. Soon, cohort evaluations and comparative studies identified a different profile in terms first recurrence sites and metastatic disease for TNBC, with 40% of cases involving lungs and 30% involving CNS 9-11. A single center analysis of 116 patients with metastatic TNBC, treated between 2000 and 2006, showed that up to 46% developed CNS metastases at some point before death, with 14% (16/116) presenting CNS metastases as first site of metastatic disease 5. Evaluating a larger group of patients, an analysis of the Surveillance, Epidemiology, and End Results (SEER) 18 registry selected 206,913 breast cancer patients from 2010 to 2014, with 11.8% characterized as TNBC, and confirmed an incidence of 30.8% brain metastases in TNBC patients with advanced disease, associated to a poorer median overall survival of 6 months (p<0.001) when compared to other subtypes 12. A possible correlation with stage at diagnosis and brain metastases as first site of metastatic disease was demonstrated in a retrospective analysis of 2448 TNBC patients diagnosed with early stage breast cancer and followed between 1990 and 2010 13. Patients with stage III disease had a significant increase in the risk of developing brain metastases as a first site of recurrence (hazards ratio = 3.51; 95% CI = 1.85 – 6.67; p =.0001) compared to patients with stage I disease, in a multivariate analysis.

When compared to other subtypes in various retrospective analysis, TNBC seems to have typically a shorter interval between initial breast cancer diagnosis and development of brain metastases than other subtypes of breast cancer 14-17. The shorter timeline is also corroborated by the smaller number of lines of therapy TNBC patients received before their first diagnosis of brain metastases in comparison to other subtypes 18.

Multiple retrospective analysis have repeatedly reported the median OS after diagnosis of TNBC brain metastases to be dismal, mostly around 4-6 months 5,17,19-21, with two publications showing extremes of 2.9 in one22 and 8.4 months in other 23. The final cause of death was directly related to brain metastases in more than half of those patients 16-19,22,24. Importantly, in all breast cancer subtypes, the OS after brain metastases diagnosis was better in patients that received systemic therapy as opposed to those who did not, even if slightly so in TNBC 19,23.

Particular issues with CNS metastases

Historically, the general effect of systemic therapy has been unimpressive in breast cancer brain metastases (BCBM). More often than not, intracranial metastases present in setting of active extracranial disease and responses to systemic treatment are frequently different in those sites. Understanding the unique aspects of CNS metastases has been target of active research and has corroborated clear obstacles to an effective therapy.

One of the anticipated issues reside in the known protective effect and very particular structure of blood-brain barrier (BBB). Yet, once a brain metastases forms, the BBB is modified and becomes what is called the blood-tumor barrier (BBT) 25. The BBT presents some, but still very heterogeneous permeability to drugs. This was well demonstrated by preclinical evaluation of marked paclitaxel and doxorubicin uptake in breast cancer mice models, and then confirmed clinically by the heterogeneous uptake of capecitabine and lapatinib administered to patients before the surgical resection of their BCBM 26,27.

Brain metastases cells have to change and adapt to the CNS microenvironment to establish and grow. An analysis of gene expression in mice models of brain metastases showed that the brain microenvironment induces complete reprogramming of metastasized cancer cells and leads to gain of neuronal cell characteristics 28. In the same lines, analysis of resected TNBC and HER2-positive BCBM from patients found that breast-to-brain metastatic tissue and cells presented an acquired GABAergic phenotype similar to the neuronal cells 29. In fact, whole exome analysis of different primary tumor types and their matched brain metastases, which included BCBM, revealed the clear presence of a genomic evolution pattern in the brain metastases 30. Importantly, clinically actionable mutations were found in those brain metastases that would not otherwise been demonstrated by sole analysis of extracranial metastases.

Furthermore, an issue very specific to BCBM is the subtype discordance or switching between primary tumor and the brain metastases. Most BCBM are not biopsied or resected given the specific risk-benefit of a CNS surgical intervention. Consequently, comparative studies are limited mostly by sample size. In spite of that, several groups have now repeatedly confirmed the existence of discordance of subtypes 31-34.The largest analysis involved 219 patients and showed an overall 36.3% incidence of discordance for estrogen receptor (ER), progesterone receptor (PR) or HER2, leading to a change of subtype in 22.8% of cases as a result of receptor alteration. Gain in HER2 expression in the brain metastases was more common than loss (8.0% vs 2.5%, respectively), while loss of ER or PR was more common in the brain metastases than gain (14.8% vs 1.9% and 22.4% vs 2.9%, respectively). Generally, cases with discordance between the primary tumor and brain metastases also had discordances between intra- and extracranial metastases 34.

All these findings indicate the need for organ-specific therapy, considering the tumor specific CNS metastases characteristics, and the need for validation of non-invasive methods to improve precision diagnosis with minimal risk.

Preclinical data

Genomics

In TNBC, tumor biology and brain metastases connection still needs elucidation. A couple of different analysis of paired primary breast cancer samples with respective metastases, showed that the overall gene expression phenotype was mostly maintained in the metastases, even in brain lesions 35,36. Nevertheless, clear presence of genetic branching evolution has also been demonstrated in whole-exome sequencing analysis of paired tumors and brain metastases, including breast cancer cases 30. While brain metastases shared common genetic ancestry aspects with the primary tumor, they also presented new potentially oncogenic alterations that could contribute to a divergence in response to therapy frequently seen in brain metastases versus other areas of disease. Moreover, genetic selection pressure applied by the effect of previous lines of treatment has to be also taken into account when comparing different studies.

In a deep genomic profiling integrating gene copy number, gene expression and DNA methylation datasets on a collection of breast brain metastases, basal-like tumors were one of the most commonly represented subtypes in brain metastases cohorts 37. Independent of subtypes, brain metastases were associated with frequent large chromosomal gains (1q, 5p, 8q, 11q, and 20q), deletions (8p, 17p, 21p and Xq), amplifications and overexpressed genes (ATAD2, BRAF, DERL1, DNMTRB and NEK2A), and enrichment in cell cycle and G2/M transition pathways (AURKA, AURKB and FOXM1) was also demonstrated 37. However, while overall methylation levels were increased in breast cancer brain metastasis, basal-like brain metastases were associated with significantly lower levels of methylation, which tends to lead to defects in cell migration and adhesion and contributes to a more metastatic pattern.

Looking for a specific genetic profile linked to brain metastases predisposition inside TNBC, a retrospective analysis investigated expression of 29,369 gene transcripts in primary TNBC tumor samples from 119 patients, including patients with symptomatic brain metastases 38. No gene expression was found to be associated with the occurrence of symptomatic brain metastases, but there were many valid criticisms to the study, including the lack of brain metastases samples limiting subtypes classification and genetic analysis only of primary breast tumor, the possible “contamination” of the non-brain metastases pool by asymptomatic brain metastases patients and limitation of utilized technique.

In a different approach, transcription factors highly and specifically expressed in primary TNBCs were selected and analyzed 39. Among those, engrailed 1 (EN1) was of specific interest because of its known roles in brain and dermomyotome development 40,41. Preclinical evaluation showed that genes involved in WNT and Hedgehog signaling, neurogenesis, and axonal guidance were direct EN1 transcriptional targets. Additionally, expression analysis was performed on METABRIC dataset where high expression of EN1 correlated with increased risk of developing brain metastases in TNBC patients, as well, as with shorter overall survival 39.

Preclinical models

Preclinical models are essential to elucidate the complexity of CNS metastases and perform initial tests of new therapies. Different models to study brain metastases in laboratory include cell lines and mice models. Several TNBC cell lines and models are available. The models, methods of development and analysis are extensively reviewed in references 42 and 43.

Microenvironment and molecular pathways

Tumor microenvironment has an important role in metastases establishment and growth. Evaluation of the microenvironment in TNBC brain metastases are mostly limited to preclinical models with the function of generating hypothesis that need validation in the clinical setting.

Brain-derived neurotrophic factor (BDNF), for example, is widely expressed in the brain and has the function to stimulate neurons growth and synapses 44,45. In TNBC brain metastases models, BDNF was demonstrated to perform autocrinal regulation of BDNF-tumor cell tropomyosin kinase receptor B (TrkB) gene expression with consequent increase in cell migratory activity, which is important for metastases development. Extending the analysis to protein expression levels in tumor tissue arrays, a correlation was found with patient prognosis and TrkB expression, but not of BDNF 46. Furthermore, a more detailed evaluation identified estradiol (E2) as causing upregulation of BDNF in estrogen receptor positive astrocytes 47, which activated TrkB signaling and increased invasiveness and tumor initiating capacity of TNBC cells in vitro 48. These findings provide and interesting basis for the clinical evaluation of estrogen depletion therapy and/or TrkB inhibitors in the prevention of brain metastases.

The mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) pathway also seems to be activated in TNBC and brain metastases, promoting tumor colonization, survival, and growth 49,50. Utilizing clinically available brain-penetrant inhibitors in intracranial TNBC tumors mice models, treatment with MEK, PI3K, or platelet derived growth factor receptor (PDGFR) inhibitors was evaluated. Combination treatments with selumetinib (MEK inhibitor) and buparlisib (PIK3 inhibitor) or selumetinib and pazopanib (PDGFR inhibitor) were synergistic in vitro but improved survival only in in two of the four TNBC intracranial metastases models 51. Though these represent rational combinations for treatment of TNBC brain metastases, its clinical development needs to be carefully considered in view of the significant toxicity observed both in this preclinical setting and also known from experiences in the context of other tumors clinical treatment.

Current management

Local therapy

The current treatment of brain metastases is multidisciplinary, although mainly based on combination of local therapeutics including surgical resection, stereotactic radiosurgery (SRS) and/or whole brain radiation (WBRT) 52. Notably, the fundamental trials establishing local therapy for brain metastases evaluated cohorts mainly composed of patients with non-small cell lung cancer and with smaller proportions of breast cancer patients. Overall, the addition of WBRT to surgery or SRS improves local progression free survival but does not affect OS 53-55. However, worsening quality of life and neurocognitive decline have been demonstrated with WBRT 56-58. Therefore, SRS is the preferred treatment for limited number of brain metastases 59, while WBRT tends to be mainly indicated for treatment of diffuse brain metastases. In cases with solitary brain lesion and controlled systemic disease, the combination of surgical resection and postoperative SRS has reportedly improved survival and should be the preferred approach 60,61.

Systemic therapy

Systemic therapy has been generally less effective in treatment of BCBM than of non-CNS metastatic disease, as documented by several clinical trials 62-66. Nevertheless, retrospective analysis repeatedly demonstrate improvement of overall survival when systemic therapy is offered to brain metastases patients as opposed to no systemic therapy at all 21. Contrasting with the HER2-positive subtype, in which anti-HER2 agents have generated some intra-cranial response 67-70, TNBC treatment has been mostly based on the systemic chemotherapy agents reaching very limited effect in CNS.

In recent years, at least three classes of drugs or regimens have shown more targeted benefits in the treatment of subgroups of metastatic TNBC: immune checkpoint inhibitors (ICI), poly (ADP-ribose) polymerase (PARP) inhibitors, and an antibody drug conjugate (ADC) targeting Trop-2 71-74.

PARP inhibitors

BRCA1/2 mutation carriers have an elevated risk of developing breast cancer. Overall, up to 20% of TNBC have a germline BRCA mutation, and BRCA1 carriers most likely will develop TNBC 75,76. Analysis of different cohorts of breast cancer patients with germline BRCA mutation have demonstrated high incidences of brain metastases (44%-53%) in the BRCA1 carriers, most of them with TNBC (84%-73%) 77,78; and the risk of brain metastases seems to be more strongly associated with tumor subtype than with the mutation 77.

PARP inhibitors (PARPi) are a class of drugs that inhibit double-stranded DNA repair. This repair mechanism is already deficient in BRCA mutated cells, which makes PARPi an effective therapy in breast cancer patients affected by BRCA mutation 79. Two PARPi are currently approved for treatment of metastatic BRCA1/2 mutated breast cancer based on PFS improvement when compared to chemotherapy, olaparib (7.0 versus 4.2 months; 0.58, 95% CI 0.43-0.80; p<0.001), and talazoparib (8.6 versus 5.6 months; HR 0.54, 95% CI 0.41-0.71; p<0.001) 72,80-82. In the phase III trial evaluating talazoparib, patients with treated and stable CNS disease were enrolled 80, but no patient with active CNS disease or evaluation of CNS endpoints was performed in both trials. Nevertheless, the PARPi veliparib presented CNS penetrance and survival benefit when combined to carboplatin for treatment of BRCA mutated TNBC in preclinical models of intracranial metastases 83. A phase II trial (NCT02595905) is currently ongoing and evaluating the use of cisplatin plus or minus veliparib in TNBC metastatic breast cancer patients with or without BRCA mutation, also including patients with brain metastases. Initial results of the study showed significant benefit in PFS with the addition of veliparib only in the BRCA-like TNBC cohort (5.9 versus 4.2 months, HR, 0.53; 95% CI 0.34-0.83; p=0 .006), and CNS endpoints are yet to be reported 84.

Immune checkpoint inhibitors (ICI)

TNBC are very immunogenic tumors, presenting high number of tumor mutations, more tumor infiltrating lymphocytes (TIL), and high PDL-1 expression both in in tumor and immune cells around them 85-88. Immune checkpoints are proteins, such as PDL-1, produced by some of the immune cells and tumor cells and control negatively the immune response. ICI are agents that block those immune checkpoints and enhance the immune response. Two of those agents, atezolizumab (anti-PDL1) and pembrolizumab (anti-PD1) are currently approved for initial treatment of PDL1 positive TNBC in combination with chemotherapy 89.

The Impassion130 trial randomized 902 previously untreated metastatic TNBC patients between nab-paclitaxel plus or minus atezolizumab and showed progression free survival (PFS) benefit with addition of atezolizumab (7.2 versus 5.5 months, HR 0.80, 95% CI 0.69–0.92, p=0.0021). An OS benefit, however, was observed only in the TNBC subgroup presenting PDL1 positive immune cells, in an exploratory analysis (25 versus 18 months, HR 0.71, 95% CI 0.54 – 0.94). Only 61 patients with treated and asymptomatic CNS disease were enrolled and presented the same PFS benefit as the overall trial population 89. No specific CNS endpoints were evaluated.

In the Keynote355 trial, 847 previously untreated metastatic TNBC patients were randomized between chemotherapy plus or minus pembrolizumab. Adding pembrolizumab improved PFS (9.7 versus 5.6 months, HR 0.65, 95% CI 0.49 – 0.86, p=0.0012) in patients with PDL1 combined positive score (CPS) ≥ 10 90. Once more, only patients with treated and stable brain metastases were enrolled and no specific CNS endpoints were reported.

Much remains to be understood regarding the CNS and brain metastases immunological environment. The CNS used to be considered as having an absolute immune privilege, but that assumption has been questioned and studies demonstrate changes in blood-brain barrier and environment once primary or metastatic tumors are present 91. ICI therapy has even led to intracranial responses in melanoma and non-small cell lung cancer 92,93. In melanoma, selected aspects of brain metastases environment such as immune infiltrate, hemorrhage, necrosis have prognostic significance 94. An analysis of 203 BCBM tissue biobank looked into these same histologic aspects. Necrosis was higher in TNBC in comparison to other subtypes and associated with worse prognosis. Meanwhile, gliosis was linked to a better prognosis in TNBC 95. The analysis, however, was not made in relation to a specific therapy.

In a retrospective evaluation of 84 breast cancer brain metastases, PD-L1 and PD-L2 were analyzed by immunohistochemistry and found to be present in 53 % and 36 % of cases, respectively, and not related to the brain metastasis phenotype 96. In the same analysis, PD-1 expression on TILs correlated positively with CD4+ and CD8+ TILs. Overall, these findings suggest a promising landscape for use of immune checkpoint inhibitors, if we assume immune functions in CNS mimic the systemic ones. Conversely, in an analysis of 46 paired samples from primary breast cancers and respective brain metastases, the percentage of TILs was significantly lower in the metastatic lesions. The numbers of CD4/CD8/Foxp3-positive cells were also significantly higher in primary lesions, but there was no clear difference in PD-L1 positivity between primary tumors and brain metastases 97. Clinical trials evaluating immunological therapeutics alone or combined to other systemic agents or radiation are currently ongoing.

Antibody drug conjugates

Antibody drug conjugates (ADCs) are a class of drugs that deliver treatment in a targeted way, combining the active compound to a specific antibody. Sacituzumab-govitecan is an antibody–drug conjugate in which an active metabolite of irinotecan (SN-38) is linked to the humanized antitrophoblast cell-surface antigen 2 (Trop-2) monoclonal antibody 98. Trop-2 is detected in triple-negative breast cancer cells amongst other tumors 99.

In a phase I/II trial, 108 TNBC patients previously treated with a median of 3 lines for metastatic disease, received treatment with Sacituzumab-govitecan and presented a response rate of 33.3% (95% CI, 24.6 to 43.1), with median duration of response of 7.7 months (95% CI, 4.9 to 10.8), with main adverse events being hematologic toxicity related 74. Based on these results, the ADC has been approved for use in metastatic TNBC patients that received at least 2 prior therapies in metastatic setting. The phase III ASCENT trial compared sacituzumab govitecan with single-agent chemotherapy per physician’s choice (capecitabine, eribulin, vinorelbine or gemcitabine) in 468 patients with metastatic TNBC and showed improvement in PFS (5.6 versus 1.7 months; HR 0.41; p<0.0001) and OS (12.1 versus 6.7 months; HR 0.48; p<0.0001) with the use of the ADC 100. Both trials were non-informative regarding possible CNS effects of the drug since patients with active CNS disease were not eligible. However, in a phase 0 trial (NCT03995706), Sacituzumab-govitecan was given as single pre-operative dose to 19 patients with CNS primary tumors or metastases undergoing resection with continued treatment after surgery until progression. Sacituzumab-govitecan delivered 150-fold (BC brain metastases) and 40-fold (glioblastomas) the 50% inhibitory concentration (IC50) of SN38, and produced partial responses in both cohorts per preliminary reports 101. These are stimulating results and should lead to further investigation of this ADC activity in TNBC brain metastases. A phase II trial with Sacituzumab-govitecan in patients with HER2-negative BCBM is expected to be launched by the Southwestern Oncology Group (SWOG) soon.

Table 1 presents currently available clinical trials for TNBC with CNS metastases.

Table 1.

Available clinical trials for TNBC with brain metastases

NCT Title Intervention Eligibility
03994796 (Phase II) Genomically-Guided Treatment Trial in Brain Metastases Palbociclib or GDC-0084 or entrectinib, pending on presence of gene mutation
  • clinically actionable alteration in NTRK, ROS1, or CDK pathway or PI3K pathway

  • at least one prior TNBC directed therapy in the metastatic setting

03449238 (Phase I/II) Pembrolizumab and Stereotactic Radiosurgery (SRS) of Selected Brain Metastases in Breast Cancer Patients Pembrolizumab plus SRS
  • at least 2 intracranial untreated metastases

  • exclusion: current systemic therapy, previous WBRT or anti-PD1/PD1 or PDL2 agents

03483012 (Phase II) Atezolizumab and Stereotactic Radiation in Triple-negative Breast Cancer and Brain Metastasis Atezolizumab plus SRS
  • up to 5 intracranial untreated metastases with indication for SRS

  • exclusion: previous treatment with anti-PD1/PD1 or PDL2 agents

02886585 (Phase II) Pembrolizumab In Central Nervous System Metastases Pembrolizumab
  • measurable CNS metastases

04434560 (Phase II) Neoadjuvant Immunotherapy in Brain Metastases Nivolumab and Ipilimumab
  • 1 to 3 previously untreated CNS metastases, at least one resectable, plan for SRS

  • TNBC that is PD-L1 positive

03328884 (Phase II) Evaluation of the Efficacy and Safety of Nal-IRI for Progressing Brain Metastases in Breast Cancer Patients Irinotecan Hydrochloride
  • measurable CNS metastases

  • exclusion: previous treatment with irinotecan

04039230 (Phase IB/II) Phase 1b/2 Study to Evaluate Antibody-Drug Conjugate Sacituzumab Govitecan in Combination with PARP Inhibitor Talazoparib in Patients with Metastatic Breast Cancer Sacituzumab-govitecan and Talazoparib
  • only stable CNS disease allowed

03952325 (Phase II) Tesetaxel Plus 3 Different PD-(L)1 Inhibitors in Patients with Metastatic TNBC and Tesetaxel Monotherapy in Patients with HER2 Negative MBC (CONTESSA TRIO) Tesetaxel plus either Nivolumab, Pembrolizumab or Atezolizumab
  • known metastases to the CNS are permitted but not required

  • exclusion: previous treatment with anti-PD1/PD1 or PDL2 agents

Looking forward

Many aspects remain to be clarified to improve the treatment of TNBC brain metastases. While information on CNS effect of the currently therapies specifically approved for TNBC treatment is eagerly awaited, a couple of innovative clinical trials is considering different approaches to the problem.

Based on the demonstrated presence of genomic branched evolution in brain metastases that may generate specific targetable mutations in the brain lesions 30, a genomically guided phase II trial (Alliance A071701; NCT03994796) has been developed. Patients with TNBC and brain metastases are also eligible. Each patient will receive the specific treatment pending on actionable mutations found in their brain metastases: abemaciclib for CDK alterations, dual mTOR/PI3K inhibitor paxalisib/GDC-0084 for mTOR/AKT/PI3K alterations, and entrectinib for NTRK/ROS1 fusions (specific for lung brain metastases). The primary endpoint of the trial is overall CNS response rate. This approach represents the next steps in the precision medicine strategy.

In setting of high-risk for brain metastases development and poor prognosis once they are established, a preventative approach would also make sense. Based on its high CNS penetrance, temozolomide was evaluated in TNBC brain metastases mice models and found to be highly effective in preventing brain metastases development 102. No effect was seen in treating established brain metastases lesions though. Currently, a phase I/II clinical trial for secondary prevention of brain metastases with temozolomide (NCT03190967) is opened for HER2-positive breast cancer patients 103. A phase I/II brain metastases prevention clinical trial with temozolomide for patients with TNBC is currently under development with plans to open in the soon. Eligible patients should have received local therapy for brain metastases close to enrollment and will then receive temozolomide in combination with an active systemic treatment for extracranial TNBC based on physician’s choice.

Conclusion

Brain metastases are frequent and confer an even poorer prognosis to patients with TNBC. Different than HER2-positive breast cancer, systemic therapeutic options have just recently started to be available for patients with metastatic TNBC, and data on specific CNS action of those agents is still unclear. There is, however, an increased awareness of brain metastases being an unmet need in this subtype of breast cancer. CNS biology is knowingly complex and has a distinctive microenvironment that needs to be studied in association with the specific nature of TNBC metastases, guiding the development of brain permeable therapeutic options. Associated with that, clinical trials design has to be reconsidered to include patients with CNS active metastases, mirroring the reality of our clinics and patients’ lives. Effectively connecting laboratory evidences to our clinical research is the only promising pathway to improve the lives of our patients with TNBC and CNS metastases.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. Ca-a Cancer Journal for Clinicians 2020;70:7–30. [DOI] [PubMed] [Google Scholar]
  • 2.Arvold ND, Oh KS, Niemierko A, et al. Brain metastases after breast-conserving therapy and systemic therapy: incidence and characteristics by biologic subtype. Breast Cancer Research and Treatment 2012;136:153–60. [DOI] [PubMed] [Google Scholar]
  • 3.Kennecke H, Yerushalmi R, Woods R, et al. Metastatic Behavior of Breast Cancer Subtypes. Journal of Clinical Oncology 2010;28:3271–7. [DOI] [PubMed] [Google Scholar]
  • 4.Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proceedings of the National Academy of Sciences of the United States of America 2001;98:10869–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lin NU, Claus E, Sohl J, Razzak AR, Arnaout A, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Nayak L, Lee EQ, Wen PY. Epidemiology of Brain Metastases. Current Oncology Reports 2012;14:48–54. [DOI] [PubMed] [Google Scholar]
  • 7.Thulin A, Ronnerman E, Zhang C, et al. Clinical outcome of patients with brain metastases from breast cancer - A population based study over 21 years. Breast 2020;50:113–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Foulkes WD, Smith IE, Reis-Filho JS. Triple-Negative Breast Cancer. New England Journal of Medicine 2010;363:1938–48. [DOI] [PubMed] [Google Scholar]
  • 9.Dent R, Hanna WM, Trudeau M, Rawlinson E, Sun P, Narod SA. Pattern of metastatic spread in triple-negative breast cancer. Breast Cancer Research and Treatment 2009;115:423–8. [DOI] [PubMed] [Google Scholar]
  • 10.Rodriguez-Pinilla SM, Sarrio D, Honrado E, et al. Prognostic significance of basal-like phenotype and fascin expression in node-negative invasive breast carcinomas. Clinical Cancer Research 2006;12:1533–9. [DOI] [PubMed] [Google Scholar]
  • 11.Liedtke C, Mazouni C, Hess KR, et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. Journal of Clinical Oncology 2008;26:1275–81. [DOI] [PubMed] [Google Scholar]
  • 12.Kim Y-J, Kim J-S, Kim IA. Molecular subtype predicts incidence and prognosis of brain metastasis from breast cancer in SEER database. Journal of Cancer Research and Clinical Oncology 2018;144:1803–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dawood S, Lei X, Litton JK, Buchholz TA, Hortobagyi GN, Gonzalez-Angulo AM. Incidence of brain metastases as a first site of recurrence among women with triple receptor-negative breast cancer. Cancer 2012;118:4652–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Saraf A, Grubb CS, Hwang ME, et al. Breast cancer subtype and stage are prognostic of time from breast cancer diagnosis to brain metastasis development. Journal of Neuro-Oncology 2017;134:453–63. [DOI] [PubMed] [Google Scholar]
  • 15.Matsuo S, Watanabe J, Mitsuya K, Hayashi N, Nakasu Y, Hayashi M. Brain metastasis in patients with metastatic breast cancer in the real world: a single-institution, retrospective review of 12-year follow-up. Breast Cancer Research and Treatment 2017;162:169–79. [DOI] [PubMed] [Google Scholar]
  • 16.Anders CK, Deal AM, Miller CR, et al. The Prognostic Contribution of Clinical Breast Cancer Subtype, Age, and Race Among Patients With Breast Cancer Brain Metastases. Cancer 2011;117:1602–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Eichler AF, Kuter I, Ryan P, Schapira L, Younger J, Henson JW. Survival in patients with brain metastases from breast cancer. Cancer 2008;112:2359–67. [DOI] [PubMed] [Google Scholar]
  • 18.Niikura N, Hayashi N, Masuda N, et al. Treatment outcomes and prognostic factors for patients with brain metastases from breast cancer of each subtype: a multicenter retrospective analysis. Breast Cancer Research and Treatment 2014;147:103–12. [DOI] [PubMed] [Google Scholar]
  • 19.Kaplan MA, Isikdogan A, Koca D, et al. Biological Subtypes and Survival Outcomes in Breast Cancer Patients with Brain Metastases (Study of the Anatolian Society of Medical Oncology). Oncology 2012;83:141–50. [DOI] [PubMed] [Google Scholar]
  • 20.Oehrlich NE, Spineli LM, Papendorf F, Park-Simon T-W. Clinical outcome of brain metastases differs significantly among breast cancer subtypes. Oncology Letters 2017;14:194–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gao C, Wang F, Suki D, et al. Effects of systemic therapy and local therapy on outcomes of 873 breast cancer patients with metastatic breast cancer to brain:MDAnderson Cancer Center experience. International Journal of Cancer 2020. [DOI] [PubMed] [Google Scholar]
  • 22.Dawood S, Broglio K, Esteva FJ, et al. Survival among women with triple receptor-negative breast cancer and brain metastases. Annals of Oncology 2009;20:621–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Miller JA, Kotecha R, Ahluwalia MS, et al. Overall Survival and the Response to Radiotherapy Among Molecular Subtypes of Breast Cancer Brain Metastases Treated With Targeted Therapies. Cancer 2017;123:2283–93. [DOI] [PubMed] [Google Scholar]
  • 24.Nam B-H, Kim SY, Han H-S, et al. Breast cancer subtypes and survival in patients with brain metastases. Breast Cancer Research 2008;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nature Reviews Cancer 2020;20:26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lockman PR, Mittapalli RK, Taskar KS, et al. Heterogeneous Blood-Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer. Clinical Cancer Research 2010;16:5664–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Morikawa A, Peereboom DM, Thorsheim HR, et al. Capecitabine and lapatinib uptake in surgically resected brain metastases from metastatic breast cancer patients: a prospective study. Neuro-Oncology 2015;17:289–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Park ES, Kim SJ, Kim SW, et al. Cross-species hybridization of microarrays for studying tumor transcriptome of brain metastasis. Proceedings of the National Academy of Sciences of the United States of America 2011;108:17456–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Neman J, Termini J, Wilczynski S, et al. Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proceedings of the National Academy of Sciences of the United States of America 2014;111:984–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Brastianos PK, Carter SL, Santagata S, et al. Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets. Cancer Discovery 2015;5:1164–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Duchnowska R, Dziadziuszko R, Trojanowski T, et al. Conversion of epidermal growth factor receptor 2 and hormone receptor expression in breast cancer metastases to the brain. Breast Cancer Research 2012;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jung J, Lee SH, Park M, et al. Discordances in ER, PR, and HER2 between primary breast cancer and brain metastasis. Journal of Neuro-Oncology 2018;137:295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Priedigkeit N, Hartmaier RJ, Chen Y, et al. Intrinsic Subtype Switching and Acquired ERBB2/HER2 Amplifications and Mutations in Breast Cancer Brain Metastases. Jama Oncology 2017;3:666–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kaidar-Person O, Meattini I, Jain P, et al. Discrepancies between biomarkers of primary breast cancer and subsequent brain metastases: an international multicenter study. Breast Cancer Research and Treatment 2018;167:479–83. [DOI] [PubMed] [Google Scholar]
  • 35.Harrell JC, Prat A, Parker JS, et al. Genomic analysis identifies unique signatures predictive of brain, lung, and liver relapse. Breast Cancer Research and Treatment 2012;132:523–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Echeverria GV, Powell E, Seth S, et al. High-resolution clonal mapping of multi-organ metastasis in triple negative breast cancer. Nature Communications 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Salhia B, Kiefer J, Ross JTD, et al. Integrated Genomic and Epigenomic Analysis of Breast Cancer Brain Metastasis. Plos One 2014;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Duchnowska R, Jarzab M, Zebracka-Gala J, et al. Brain Metastasis Prediction by Transcriptomic Profiling in Triple-Negative Breast Cancer. Clinical Breast Cancer 2017;17:E65–E75. [DOI] [PubMed] [Google Scholar]
  • 39.Peluffo G, Subedee A, Harper NW, et al. EN1 Is a Transcriptional Dependency in Triple-Negative Breast Cancer Associated with Brain Metastasis. Cancer Research 2019;79:4173–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fuchs J, Stettler O, Alvarez-Fischer D, Prochiantz A, Moya KL, Joshi RL. Engrailed signaling in axon guidance and neuron survival. European Journal of Neuroscience 2012;35:1837–45. [DOI] [PubMed] [Google Scholar]
  • 41.Atit R, Sgaier SK, Mohamed OA, et al. beta-catenin activation is necessary and sufficient to specify the dorsal dermal fate in the mouse. Developmental Biology 2006;296:164–76. [DOI] [PubMed] [Google Scholar]
  • 42.Zimmer AS, Van Swearingen AED, Anders CK. HER2-positivebreast cancer brain metastasis: A new and exciting landscape. Cancer Reports 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Valiente M, Van Swearingen AED, Anders CK, et al. Brain Metastasis Cell Lines Panel: A Public Resource of Organotropic Cell Lines. Cancer Research 2020;80:4314–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Acheson A, Conover JC, Fandl JP, et al. A BDNF AUTOCRINE LOOP IN ADULT SENSORY NEURONS PREVENTS CELL DEATH. Nature 1995;374:450–3. [DOI] [PubMed] [Google Scholar]
  • 45.Huang EJ, Reichardt LF. Neurotrophins: Roles in neuronal development and function. Annual Review of Neuroscience 2001;24:677–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tsai Y-F, Tseng L-M, Hsu C-Y, Yang M-H, Chiu J-H, Shyr Y-M. Brain-derived neurotrophic factor (BDNF) - TrKB signaling modulates cancer-endothelial cells interaction and affects the outcomes of triple negative breast cancer. Plos One 2017;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sartorius CA, Hanna CT, Gril B, et al. Estrogen promotes the brain metastatic colonization of triple negative breast cancer cells via an astrocyte-mediated paracrine mechanism. Oncogene 2016;35:2881–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Contreras-Zarate MJ, Day NL, Ormond DR, et al. Estradiol induces BDNF/TrkB signaling in triple-negative breast cancer to promote brain metastases. Oncogene 2019;38:4685–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Koboldt DC, Fulton RS, McLellan MD, et al. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Da Silva L, Simpson PT, Smart CE, et al. HER3 and downstream pathways are involved in colonization of brain metastases from breast cancer. Breast Cancer Research 2010;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Van Swearingen AED, Sambade MJ, Siegel MB, et al. Combined kinase inhibitors of MEK1/2 and either PI3K or PDGFR are efficacious in intracranial triple-negative breast cancer. Neuro-Oncology 2017;19:1481–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tsao MN, Rades D, Wirth A, et al. Radiotherapeutic and surgical management for newly diagnosed brain metastasis(es): An American Society for Radiation Oncology evidence-based guideline. Practical radiation oncology 2012;2:210–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet 2004;363:1665–72. [DOI] [PubMed] [Google Scholar]
  • 54.Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases - A randomized controlled trial. Jama-Journal of the American Medical Association 2006;295:2483–91. [DOI] [PubMed] [Google Scholar]
  • 55.Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant Whole-Brain Radiotherapy Versus Observation After Radiosurgery or Surgical Resection of One to Three Cerebral Metastases: Results of the EORTC 22952-26001 Study. Journal of Clinical Oncology 2011;29:134–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Soffietti R, Kocher M, Abacioglu UM, et al. A European Organisation for Research and Treatment of Cancer Phase III Trial of Adjuvant Whole-Brain Radiotherapy Versus Observation in Patients With One to Three Brain Metastases From Solid Tumors After Surgical Resection or Radiosurgery: Quality-of-Life Results. Journal of Clinical Oncology 2013;31:65–72. [DOI] [PubMed] [Google Scholar]
  • 57.Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncology 2009;10:1037–44. [DOI] [PubMed] [Google Scholar]
  • 58.Brown PD, Asher AL, Ballman KV, et al. NCCTG N0574 (Alliance): A phase III randomized trial of whole brain radiation therapy (WBRT) in addition to radiosurgery (SRS) in patients with 1 to 3 brain metastases. Journal of Clinical Oncology 2015;33. [Google Scholar]
  • 59.Serizawa T, Yamamoto M, Sato Y, et al. Gamma Knife surgery as sole treatment for multiple brain metastases: 2-center retrospective review of 1508 cases meeting the inclusion criteria of the JLGK0901 multi-institutional prospective study. Journal of Neurosurgery 2010;113:48–52. [DOI] [PubMed] [Google Scholar]
  • 60.Patchell RA, Tibbs PA, Walsh JW, et al. A RANDOMIZED TRIAL OF SURGERY IN THE TREATMENT OF SINGLE METASTASES TO THE BRAIN. New England Journal of Medicine 1990;322:494–500. [DOI] [PubMed] [Google Scholar]
  • 61.Hatiboglu MA, Wildrick DM, Sawaya R. The role of surgical resection in patients with brain metastases. Ecancermedicalscience 2013;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Boogerd W, Dalesio O, Bais EM, van der Sande JJ. Response of brain metastases from breast cancer to systemic chemotherapy. Cancer 1992;69:972–80. [DOI] [PubMed] [Google Scholar]
  • 63.Peereboom DM. Chemotherapy in brain metastases. Neurosurgery 2005;57:54–65. [DOI] [PubMed] [Google Scholar]
  • 64.Freilich RJ, Seidman AD, Deangelis LM. CENTRAL-NERVOUS-SYSTEM PROGRESSION OF METASTATIC BREAST-CANCER IN PATIENTS TREATED WITH PACLITAXEL. Cancer 1995;76:232–6. [DOI] [PubMed] [Google Scholar]
  • 65.Rosner D, Nemoto T, Lane WW. CHEMOTHERAPY INDUCES REGRESSION OF BRAIN METASTASES IN BREAST-CARCINOMA. Cancer 1986;58:832–9. [DOI] [PubMed] [Google Scholar]
  • 66.Walbert T, Gilbert MR. The role of chemotherapy in the treatment of patients with brain metastases from solid tumors. International Journal of Clinical Oncology 2009;14:299–306. [DOI] [PubMed] [Google Scholar]
  • 67.Bachelot T, Romieu G, Campone M, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncology 2013;14:64–71. [DOI] [PubMed] [Google Scholar]
  • 68.Freedman RA, Gelman RS, Anders CK, et al. TBCRC 022: A Phase II Trial of Neratinib and Capecitabine for Patients With Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. Journal of Clinical Oncology 2019;37:1081-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lin NU, Borges V, Anders C, et al. Intracranial Efficacy and Survival With Tucatinib Plus Trastuzumab and Capecitabine for Previously Treated HER2-Positive Breast Cancer With Brain Metastases in the HER2CLIMB Trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2020:JCO2000775-JCO. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Montemurro F, Delaloge S, Barrios CH, et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial☆. Annals of oncology : official journal of the European Society for Medical Oncology 2020. [DOI] [PubMed] [Google Scholar]
  • 71.Schmid P, Adams S, Rugo HS, et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. The New England journal of medicine 2018. [DOI] [PubMed] [Google Scholar]
  • 72.Robson M, Im S-A, Senkus E, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. New England Journal of Medicine 2017;377:523–33. [DOI] [PubMed] [Google Scholar]
  • 73.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;379:753–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bardia A, Mayer IA, Vahdat LT, et al. Sacituzumab Govitecan-hziy in Refractory Metastatic Triple-Negative Breast Cancer. N Engl J Med 2019;380:741–51. [DOI] [PubMed] [Google Scholar]
  • 75.Mavaddat N, Barrowdale D, Andrulis IL, et al. Pathology of Breast and Ovarian Cancers among BRCA1 and BRCA2 Mutation Carriers: Results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiology Biomarkers & Prevention 2012;21:134–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gonzalez-Angulo AM, Timms KM, Liu SY, et al. Incidence and Outcome of BRCA Mutations in Unselected Patients with Triple Receptor-Negative Breast Cancer. Clinical Cancer Research 2011;17:1082–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Song Y, Barry WT, Seah DS, Tung NM, Garber JE, Lin NU. Patterns of recurrence and metastasis in BRCA1/BRCA2-associated breast cancers. Cancer 2020;126:271–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Garber H, Lehner M, Raghavendra AS, et al. The incidence and impact of brain metastasis in patients with hereditary BRCA1/2 mutated invasive breast cancer in a prospectively followed cohort. Journal of Clinical Oncology 2020;38. [Google Scholar]
  • 79.Zimmer AS, Gillard M, Lipkowitz S, Lee JM. Update on PARP Inhibitors in Breast Cancer. Current Treatment Options in Oncology 2018;19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Litton JK, Rugo HS, Ettl J, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. New England Journal of Medicine 2018;379:753–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Robson ME, Tung N, Conte P, et al. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician's choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Annals of Oncology 2019;30:558–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jennifer K Litton SAH, Lida A Mina, Rugo Hope S., Lee Kyung-Hun, Gonçalves Anthony, Diab Sami, Woodward Natasha, Goodwin Annabel, Yerushalmi Rinat, Roché Henri, Im Young-Hyuck, Eiermann Wolfgang, Quek Ruben G. W., Usari Tiziana, Lanzalone Silvana, Czibere Akos, Blum Joanne L., Martin Miguel, Ettl Johannes. Talazoparib (TALA) in germline BRCA1/2 (gBRCA1/2)-mutated human epidermal growth factor receptor 2 negative (HER2-) advanced breast cancer (ABC): Final overall survival (OS) results from randomized Phase 3 EMBRACA trial AACR Virtual Meeting II 2020. [Google Scholar]
  • 83.Karginova O, Siegel MB, Van Swearingen AED, et al. Efficacy of Carboplatin Alone and in Combination with ABT888 in Intracranial Murine Models of BRCA-Mutated and BRCA-Wild-Type Triple-Negative Breast Cancer. Molecular Cancer Therapeutics 2015;14:920–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sharma P, Rodler E, Barlow WE, et al. Results of a phase II randomized trial of cisplatin +/− veliparib in metastatic triple-negative breast cancer (TNBC) and/or germline BRCA-associated breast cancer (SWOG S1416). Journal of Clinical Oncology 2020;38. [Google Scholar]
  • 85.Denkert C, von Minckwitz G, Darb-Esfahani S, et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncology 2018;19:40–50. [DOI] [PubMed] [Google Scholar]
  • 86.Mittendorf EA, Philips AV, Meric-Bernstam F, et al. PD-L1 Expression in Triple-Negative Breast Cancer. Cancer Immunology Research 2014;2:361–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gatalica Z, Snyder C, Maney T, et al. Programmed Cell Death 1 (PD-1) and Its Ligand (PD-L1) in Common Cancers and Their Correlation with Molecular Cancer Type. Cancer Epidemiology Biomarkers & Prevention 2014;23:2965–70. [DOI] [PubMed] [Google Scholar]
  • 88.Luen S, Virassamy B, Savas P, Salgado R, Loi S. The genomic landscape of breast cancer and its interaction with host immunity. Breast 2016;29:241–50. [DOI] [PubMed] [Google Scholar]
  • 89.Schmid P, Rugo HS, Adams S, et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncology 2020;21:44–59. [DOI] [PubMed] [Google Scholar]
  • 90.Cortes J, Cescon DW, Rugo HS, et al. Randomized, double-blind, phase III study of pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer. Journal of Clinical Oncology 2020;38. [DOI] [PubMed] [Google Scholar]
  • 91.Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune privilege of the CNS. Nature Immunology 2017;18:123–31. [DOI] [PubMed] [Google Scholar]
  • 92.Tawbi HA, Forsyth PA, Algazi A, et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. New England Journal of Medicine 2018;379:722–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Goldberg SB, Schalper KA, Gettinger SN, et al. Pembrolizumab for management of patients with NSCLC and brain metastases: long-term results and biomarker analysis from a non -randomised, open -label, phase 2 trial. Lancet Oncology 2020;21:655–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hamilton R, Krauze M, Romkes M, et al. Pathologic and gene expression features of metastatic melanomas to the brain. Cancer 2013;119:2737–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sambade MJ, Prince G, Deal AM, et al. Examination and prognostic implications of the unique microenvironment of breast cancer brain metastases. Breast Cancer Research and Treatment 2019;176:321–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Duchnowska R, Peksa R, Radecka B, et al. Immune response in breast cancer brain metastases and their microenvironment: the role of the PD-1/PD-L axis. Breast Cancer Research 2016;18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ogiya R, Niikura N, Kumaki N, et al. Comparison of immune microenvironments between primary tumors and brain metastases in patients with breast cancer. Oncotarget 2017;8:103671–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Starodub AN, Ocean AJ, Shah MA, 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. Clinical Cancer Research 2015;21:3870–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Huang HY, Groth J, Sossey-Alaoui K, Hawthorn L, Beall S, Geradts J. Aberrant expression of novel and previously described cell membrane markers in human breast cancer cell lines and tumors. Clinical Cancer Research 2005;11:4357–64. [DOI] [PubMed] [Google Scholar]
  • 100.Bardia SMT A, Loirat D, Punie K, Oliveira M, Rugo HS, Brufsky A, Kalinsky K, Cortés J, O'Shaughnessy J, Dieras VC, Carey LA, Gianni L, Piccart M, Loibl S, Goldenberg D, Hong Q, Olivo MS, Itri LM, Hurvitz SA. ASCENT: A randomized phase III study of sacituzumab govitecan (SG) vs treatment of physician’s choice (TPC) in patients (pts) with previously treated metastatic triple-negative breast cancer (mTNBC). Annals of Oncology 2020;31:S1142–S215. [Google Scholar]
  • 101.Brenner RP AJ, Chiou J, Floyd J, Garcia M, Surapaneni P, Kaklamani V, Lathrop K, Crownover R, Caron JL, Tiziani S. Delivery and activity of SN-38 by sacituzumab govitecan in CNS tumours. Annals of Oncology 2020;31:S396–S408. [Google Scholar]
  • 102.Palmieri D, Duchnowska R, Woditschka S, et al. Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner. Clinical Cancer Research 2014;20:2727–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zimmer AS, Steinberg SM, Smart DD, et al. Temozolomide in secondary prevention of HER2-positive breast cancer brain metastases. Future oncology (London, England) 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES