Next Article in Journal
Current Research on the Pathogenesis of NAFLD/NASH and the Gut–Liver Axis: Gut Microbiota, Dysbiosis, and Leaky-Gut Syndrome
Previous Article in Journal
Mechanisms Underlining Inflammatory Pain Sensitivity in Mice Selected for High and Low Stress-Induced Analgesia—The Role of Endocannabinoids and Microglia
Previous Article in Special Issue
Pancancer Analysis of the Oncogenic and Prognostic Role of NOL7: A Potential Target for Carcinogenesis and Survival
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Insight into Molecular Targets of Breast Cancer Brain Metastasis

1
Department of Pharmacology, Faculty of Pharmacy, Dadasaheb Balpande College of Pharmacy, Nagpur 440037, India
2
Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
3
Department of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
4
Department of Biology, College of Science and Arts at Alkamil, University of Jeddah, Jeddah 23218, Saudi Arabia
5
Department of Kuliyate Tib, National Institute of Unani Medicine, Kottigepalya, Bengaluru 560091, India
6
Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
7
Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
8
King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11687; https://doi.org/10.3390/ijms231911687
Submission received: 15 August 2022 / Revised: 27 September 2022 / Accepted: 28 September 2022 / Published: 2 October 2022
(This article belongs to the Special Issue Genome Organization and Regulation of Its Activity in Cancer)

Abstract

:
Brain metastasis is one of the major reasons of death in breast cancer (BC) patients, significantly affecting the quality of life, physical activity, and interdependence on several individuals. There is no clear evidence in scientific literature that depicts an exact mechanism relating to brain metastasis in BC patients. The tendency to develop breast cancer brain metastases (BCBMs) differs by the BC subtype, varying from almost half with triple-negative breast cancer (TNBC) (HER2 ER PR), one-third with HER2+ (human epidermal growth factor receptor 2-positive, and around one-tenth with luminal subclass (ER+ (estrogen positive) or PR+ (progesterone positive)) breast cancer. This review focuses on the molecular pathways as possible therapeutic targets of BCBMs and their potent drugs under different stages of clinical trial. In view of increased numbers of clinical trials and systemic studies, the scientific community is hopeful of unraveling the underlying mechanisms of BCBMs that will help in designing an effective treatment regimen with multiple molecular targets.

1. Introduction

Cancer has a common characteristic of dysregulated cell growth composed of a heterogeneous group of ailments [1,2]. It evolved from an amalgamation of genetic and epigenetic aberrations that cause the switch-off/on of tumor suppressor genes (TSGs) and oncogenes [2,3]. Among different cancer types, breast cancer (BC) is a heterogeneous ailment that consists of specific biological sub-divisions [4]. Nearly half a million people die annually due to BC worldwide. The global incidence of cancer has increased to 19.3 million newly identified cases and 10 million cancer-associated mortalities per year [5,6,7]. BC is the most common type of cancer worldwide and the fifth leading cause of cancer-associated deaths amongst females, with an estimated incidence of 2.3 million (11.7%) new cases and 684,996 (6.9%) deaths in 2020 [7].
BC is further classified into several subclasses, characterized by immunohistochemical staining {(e.g., ER, PR, HER2 (ERBB2)}, proliferation marker protein Ki-67 (MKI67), genomic markers (e.g., BRCA1, BRCA2, and PIK3CA), and immunomarkers (e.g., tumor-infiltrating lymphocytes and PD-L1) [8]. Conventionally, the treatment of BC is based on the expression of the progesterone receptor (PR), estrogen receptor (ER), and human epidermal growth factor receptor 2 (HER2). The presence of these biomarkers has facilitated the development of effective and targeted treatments. Moreover, chemotherapy is the only therapeutic option for tumors with triple-negative breast cancer (TNBC), which lacks expression of ER, PR, and overexpression of HER2 [2,9,10].
One of the deadliest diseases that affect people is brain tumors, which have a high relapse rate and only a moderate to poor prognosis. It can be divided into two main categories: primary brain tumors that begin in the brain and secondary brain tumors that are produced by cancer cells that have spread from tumors and are established in other regions of the body. Primary brain tumors can originate from various types of brain cells, the meninges, and nerves. Gliomas, which develop from the brain’s glial tissue, are the most characteristic primary tumor type [11].
The blood-brain barrier (BBB) is one of the major challenging factors in treating brain tumors [12]. The BBB comprises various molecular parts and transport mechanisms, which produce machinery for drug efflux or obstructions to drug entry into the brain. Therefore, to tackle such types of challenges in treating brain tumors, efficient drug delivery alteration and innovative therapeutic approaches are required, in addition to traditional strategies [12].
Secondary brain tumors or brain metastases cause barrier degradation, culminating in a significant rise in membrane permeability throughout and outside the tumor mass [12]. The aggregation of circulating tumor cells within the brain microvasculature largely initiates brain metastases. The distinct microenvironment of brain cells further restricts the entry of systemic therapies and promotes tumor growth. Due to BBB breakdown, the highly proliferating metastatic cancerous cell enters the CNS mainly through the bloodstream [13]. These tumor cells then rapidly divide and cause local invasion, inflammation, displacement, and edema. The cancerous cells are concentrated in the brain areas with a greater blood flow supply, while the distribution of tumor cells among different brain regions varies with the histological subtypes [14,15].

2. Brain Metastases (BMs)

The most common malignant tumors of the central nervous system (CNS) are brain metastases [16]. Brain metastases (BMs) are ten times more prevalent than primary brain tumors, affecting 10% to 20% of adult cancer patients [17]. The most frequent causes of BMs are lung adenocarcinoma, breast carcinoma, and melanoma [18]. Brain metastasis in breast cancer is frequently identified in patients with the advanced-stage illness. It has a poor prognosis because the blood-brain barrier (BBB) hinders the delivery of various medications in the CNS [19]. Some leading causes of brain metastases result in renal cell carcinoma (melanoma), breast, colorectal, and lung cancer, which are associated with poor survival [20]. The metastatic property of breast carcinoma cells is not restricted to the brain. It could affect several other organs, too, such as the liver, distant lymph nodes, lungs, and bones [21]. Approximately 10–30% of women with metastatic breast cancer develop BMs [21]. However, people with subtypes of breast cancer commonly experience metastasis of cancer cells in the brain. Patients with HER2-positive or triple-negative breast cancer are at a higher risk of developing brain metastases [21].
The incidence of brain metastases seems to have increased in recent years, likely due to the prolonged survival of patients receiving effective treatments and the availability of better imaging techniques that lead to improved detection of brain metastases [19,22]. Table 1 summarizes different risk factors, their identification, and chances of BM in BC patients.

3. Breast Cancer Brain Metastases (BCBM)

Breast cancer brain metastases (BCBM) are the second most frequent type of brain metastases and one of the most typical breast cancer metastases [31,32]. Scientific studies have identified that patients with breast cancer had a 5.1% rate of BCBM incidence. Moreover, among patients with any metastatic disease, 14.2% developed BCBM during the therapeutic phase of the illness [32]. Most BM occurs in patients with HER2-positive and ER-negative metastatic BC. Among these, HER2-positive BC patients have a higher rate of survival. Due to the therapeutic challenges, BCBM requires an integrated therapy approach for its management [33].
Three therapeutic approaches are available for treating metastatic brain tumors, including anticancer agents, surgery, and radiation therapy [19,21,32]. Metastasis represents the primary cause of death in BC patients suffering from BMs, which gradually progresses into a more advanced stage [34]. Metastasized breast cancers or circulating tumor cells evade the BBB, and upon reaching the specified cranial cells (astrocytes), they initiate tumorigenesis leading to tumor formation. The initial growth of BMs in the brain is associated with the entry of cancerous cells into the bloodstream and different locations of the brain, where they grow and multiply rapidly [21]. Hence, further insight into breast cancer brain metastasis mechanisms is expected to provide a mode of management or inhibition of these cancer types. Figure 1 depicts the formation of breast cancer brain tumors.

4. Challenges of BM

Despite several available multimodal treatments and more advanced systemic therapies, conventional treatments of BM include surgery, radiotherapy, chemotherapy, and immunotherapy [17]. The rapid development of BMs contributes significantly to overall cancer mortality, mainly in the advanced stage, and is also linked to poor prognosis. Several factors, such as subtype, are a strong prognostic factor for the long-term survival of BC with a high grade of BM [35]. The patient’s survival rate is very low (approx. 3 to 5 months) in the case of the triple-negative cancer grade type. Metastasis of aggressive TNBC has higher apparent diffusion coefficient values than the less aggressive hormone-positive group. An increase in apparent diffusion coefficient values indicates a poor prognosis in patients with brain metastases due to BC [36].
Patients’ age also affects the progression rate, and age > 45 is related to a smaller survival from the first tumor relapse time [23] (American Cancer Society, 2019). In addition, the presence of some extracranial disease leads to the worst case of BM progression in breast cancer patients, representing the overall burden of brain metastases. Hence, tumor grade, and prognostic factor, is an important tool for evaluating progression rate and is defined as graded prognostic assessment (GPA) [37]. Karnofsky’s performance status score, extracranial metastases, and GPA include age and number of tumors in BC brain metastases [32,38]. Recently, surgery and radiotherapy have been the two commonly used treatment methods for metastatic brain cancers [32]. Other systemic methods are still in the developmental stage and are restricted to regulating extracranial malady. However, newer imaging techniques (e.g., response assessment in neuro-oncology brain metastasis) must be employed for evaluation consistency [39]. Diffusion tensor imaging (DTI) plays a crucial role in setting up a suitable treatment methodology for low diffusion coefficient brain metastases due to BC.

5. The Blood-Brain Barrier (BBB) and Blood-Tumor Barrier (BTB): A Physiological Barrier in Brain Cancer Treatment

The BBB maintains the CNS structure and function by creating an organized neurovascular unit (NVU), which contains endothelial cells (ECs), pericytes, and astrocytic end feet [40,41]. These structures also prevent the pathway of the drug into brain tumors. The BTB is formed as a result of tumor development and BBB interruption. Scientific studies reported that suboptimal therapies in brain tumors are due to the lesser permeability of BTB than BBB to small and large molecules [42,43].
The BBB has a special structure surrounded by the basal lamina that contains glycoproteins cleaved to perform different functions [44]. In addition, multiple ligands also contribute to its function through distinct signaling cells, viz. ECs, pericytes, and astrocytes [45]. ATP-binding cassette transporters’ (ABC transporters) polarization can regulate the active transport through the BBB [46]. Astrocytes are the most abundant cell type in the brain, and their metabolic sensors play an integral part in BBB development and function [47]. The astrocytic end feet dislocation from an endothelial cell by a single invading tumor cell causes local BBB breaching [48]. Astrocytes and pericytes play important roles in BBB function during development, adulthood, and disease progression. Vascular functions, such as vessel remodeling and neuroinflammation, are regulated by pericytes, sited at the abluminal side of the endothelium. Similarly, the microglia, an innate immune cell, can influence CNS vasculogenesis development and contribute to BBB function [49,50]. GABA-ergic, cholinergic, noradrenergic, and serotonergic neurons can affect the CNS endothelium to maintain blood flow, neurovascular coupling, and BBB permeability at synaptic endings [51].
Some diseases, such as brain cancer, stroke, and autoimmune deficiency syndrome, can modulate the structure and function of BBB [52,53]. The alterations in the BBB are not correlated with tumor size, type, or anatomic location [54]. The endothelial cell-specific mitogen, known as vascular endothelial growth factor (VEGF), is a potent inducer of vascular permeability. VEGF may mediate endothelial cell proliferation and vascular permeability in glial tumors. This association has significance for therapeutic applications, including evaluation of the administration of water-soluble medications, edema treatment, and anti-angiogenesis therapy [55,56]. Compared to the surrounding normal brain, where the BBB is typically intact, high-grade gliomas have a disturbed BTB that allows enhanced drug delivery of the drugs to the tumor core [12].
The BTB comprises three distinct micro-vessel accumulations, i.e., continuous, and non-fenestrated capillaries, continuous and fenestrated capillaries, and capillaries containing inter-endothelial gaps [57]. Drug efflux transporters are found in both BBB and BTB endothelial cells [58]. The advanced/invasive glioma cells are spread outside areas of disrupted BTB and inside normal brain regions [59]. In low- and high-grade gliomas, the BBB and BTB barriers thwart the delivery of sufficient therapies. The BTB is formed by brain tumor capillaries and comprises a barrier different from the BBB. The hypoxic areas resulting from metabolic demands of developed glioma lead to high expression of VEGF and angiogenesis, resulting in irregular vessel formation and disruption of BTB function [60,61]. The overexpression of receptors on brain tumor capillaries could be exploited to enhance drug delivery to tumor tissues [62,63]. An illustrative image depicting the BBB/BTB affecting brain cancer treatment has been provided in the Figure 2.

6. Treatment Modalities and Challenges

Highly chemotherapy-sensitive primary tumors are only eligible for systemic treatments of BMs. Systemic therapies are recommended in cases where therapeutic options have been exhausted [64]. Hence, the efficacious treatment for BCBMs is still lacking.

7. Complete Systemic Targeted Therapy

Systemic targeted therapy designed for BCBM patients is chemotherapy-based cytotoxicity [65]. To obtain a better outcome from targeted therapy, it is crucial to consider the tumor subtype and its prognosis rate.

8. Local Therapy Modalities

Patients with a solitary or small number of lesions should preferably go for a surgical line of handling and resection of the BM, specifically, at the time the brain syndrome is fine and the BM is showing symptoms. Except for the limitation of the anatomic location of the metastatic lesion, surgical resection has capability for instant development of intracranial hypertension and establishes histological analysis for patients in which there is no other site of metastasis [23]. In a learning study, 50 patients’ performance at the beginning of BM were studied. Several major tumors were randomized in which additional surgical resection of BMs followed via whole-brain radiation targeting (WBRT), or needle biopsy following WBRT [66]. The recurrence of BMs is less general within the surgical practice set than the emission collection [67]. Meanwhile, surgical procedures or radiotherapy are considered common in the current management plan of BCBMs.

9. Surgical Approaches

Systemic medical therapy significantly enhances the survival rate of stage 4 de novo BC patients, as evident from the literature [68,69]. It also shows the effectiveness of primary tumor surgery for a better prognosis at an early stage under specific clinical conditions. On the other hand, multiple surgical approaches provide a better opportunity to reduce the anesthesiologic conditions and unnecessary surgical trauma [70].

10. Radiation Targeted Therapy

Whole-brain radiation targeted treatment is an extremely vital line of a BC cure, especially for multiple targeted lesions in the brain. The two major goals of WBRT are the eradication of microscopic seeding of the brain and controlling macroscopic metastases [66]. The benefit of this therapy was well received in 95 randomized trials with various age groups of patients with BMBC. It was observed that the radiation-treated group had a small recurrence at the operation site and in some other areas of the brain, however, the complete survival rate was not increased [71].

11. Stereotactic Radiotherapy

This has been suggested as a substitute management option for patients with some degree of BM or lesion at difficult anatomic locations [72]. This method provides high-precision photon radiation in smaller volumes targeting and carefully avoiding the major parts of brain tissues. WBRT causes toxicity leading to neurocognitive decline, while stereotactic radiotherapy does not show such effect [35]. In the below-mentioned sections, we have covered different molecular pathways involved in regulating BCBM. Because of their significant role in BCBM, these pathways could be potential therapeutic targets for BCBM treatment. The molecular interactions of different pathways involved in BCBM have been depicted in Figure 3.

12. Molecular Pathways Involved in the Regulation of BCBM

12.1. TGFβ/SMAD Signaling Pathway

It is well-known that epithelial to mesenchymal transition (EMT) proteins such as Slug, Snail, and Zeb1 are expressed through transforming growth factor β (TGFβ), SMAD, and PI3K signaling pathways (Figure 3) [73]. The EMT proteins promote this transition by decreasing the expression of E-cadherin and increasing the expression of N-cadherin [74]. EMT plays a significant role in cancer by increasing invasiveness and metastasis, resulting in poor prognosis and survival [75]. The EMT proteins suppress the expression of CDH1 [76]. The CDH1 gene translates to E-cadherin, which plays a critical role in cell adhesion and is involved in cell attachment to other cells and the extracellular matrix (ECM) [77]. Without E-cadherin, the breast cancer cells are detached from the breast tissue, forming circulating tumor cells (CTCs) that can metastasize to other tissues, including the lungs and brain [24]. Under normal physiological conditions, the BBB selectively regulates materials that go into the brain compartment by preventing the paracellular diffusion of compounds. This causes an obstacle for breast CTCs to pass through the BBB; however, in BCBM, the breast CTCs diffuse through the endothelial cell junctions [24,78]. The endothelial cell junctions are the part of the BBB that is modified during BM formation. Slug, Snail, Zeb1, VEGFA, and CD44 contribute to BM formation by enhancing the trans-endothelial migration of tumor cells via downregulation of endothelial integrity, enabling the breast CTCs to pass the BBB [79] (Mittal, 2018). Targeting the SMAD protein in the TGFβ/SMAD signaling pathway has been suggested to attenuate brain metastasis in BCBM patients [80]. The lipoprotein receptor-related protein 1 (LRP-1) inhibitors (ANG1005 and GRN1005) bind to LRP-1, leading to LRP-1 receptor-mediated transcytosis or endocytosis across the BBB, resulting in tumor growth arrest and apoptosis [81,82].

12.2. PI3K/mTOR Signaling Pathway

The phosphoinositide 3-kinase (PI3K) signaling pathway is a central pathway involved in cellular processes such as cell survival, cell proliferation, cell metabolism, and angiogenesis (Figure 3) [83,84,85]. It also plays a significant role in BCBM with approximately 77% of patients having been noted to have an activated PI3K signaling pathway [86,87]. The activation of the PI3K signaling pathway is associated with increased expression of metastatic and immunosuppressive genes, which include CTLA4, PD-L1, CSF1R, and CSF1 in the tumor microenvironment of metastasized brain cells [21,88]. The loss of function of phosphatase and tensin homolog (PTEN), a tumor suppressor and a negative regulator of PI3K signaling, is detected in 25–71% of BCBM patients, with the highest percentage in TNBC cases [89]. Overexpression of PTEN in astrocytes suppresses invasiveness and cell migration, suggesting PTEN as a promising therapeutic target for BCBM treatment [87]. The mTOR is a serine/threonine protein kinase, a downstream protein of PI3K, and Akt plays a significant role in several cancer types [84,90]. Simultaneous mTOR and PI3K protein inhibition have been reported to attenuate BCBM [91]. Everolimus and Buparlisib (BKM120), mTOR and PI3K inhibitors, are used to treat BCBM in combination with other anticancer drugs such as trastuzumab and vinorelbine [38].

12.3. HER2/Epidermal Growth Factor Receptor (EGFR) Signaling Pathway

EGFR is a transmembrane protein that activates the EGFR signaling pathway through homo/hetero-dimerization and auto-phosphorylation in response to ligand binding [92]. EGFR forms heterodimer with HER2, activating the PI3K/AKT signaling cascade. HER2+ breast cancer is susceptible to brain metastasis due to its link with PI3K signaling pathway (Figure 3) [93]. The HER2 protein dimerizes with another similar protein called HER3, triggering cell proliferation and survival. One study based on immune-histochemistry reported that HER3 is over-expressed in around 60% of BCBM patients [94].
HER2 signaling is a master regulator of many pro-inflammatory, proliferative, and pro-metastatic pathways, the most notable of which is the cyclo-oxygenase 2 (COX2) [95,96]. The HER2/EGFR signaling pathway is directly or indirectly associated with COX2 upregulation, which has shown to induce specified brain metastasis. Because BCBM patients have high expression of both HER2 and EGFR, several drugs (some approved and others in clinical trials) are used to target various stages of the HER2/EGFR signaling pathway [97,98]. Some of the drugs that target HER2/EGFR are Lapatinib (targets HER2 receptor), Trastuzumab (targets HER2 receptor), KD019 (targets HER2, Src, and EGFR), ARRY-380 (targets HER2 receptor), HKI-272 (targets HER1, HER2, and HER4 receptors), Afatinib (targets EGFR1, EGFR2, and EGFR4 receptors), and tucatinib (targets HER2 receptor). It is worth mentioning that tucatinib, a tyrosine kinase inhibitor, combined with trastuzumab and capecitabine, was approved by the USFDA on April 17, 2020, as a BCBM treatment regimen [38,99,100].

12.4. JAK/Signal Transducer and Activator of Transcription 3 (STAT3) Signaling Pathway

The JAK-STAT pathway is known for regulating the expression of growth factors and cytokines [101]. Some of the genes involved in the JAK-STAT pathway include PD-L1, VEGFA, and CTLA4, which play a crucial role in the survival of BCBM by escaping from immunosurveillance (Figure 3) [102]. STAT3 is critical for astrocytic scar formation and is involved in axon regeneration [103] (Anderson et al., 2016). Most astrocytes in BMs are expressed as an activated form of STAT3, the phosphorylated STAT3 (pSTAT3) [104] (Priego et al., 2018). The pSTAT3+ cancerous astrocytes bypass immunosurveillance by expressing escape-promoting genes, such as PD-L1, CTLA4, VEGFA, and TIMP-1 [105]. Therefore, STAT3 in BMs could be a potential therapeutic target for BCBM treatment. Nivolumab, an approved anticancer drug, targets PD-1 in BCBM, thereby preventing the binding of PD-L1 to PD-1. Similarly, using nivolumab and other treatment regimens helps cancer immunotherapy [106,107].

13. Role of Oncogenes Regulating Brain Metastasis through Breast Cancer Cells Growth Inhibition

13.1. Role of CXCR4 Gene

Gene therapy signifies a potential method for the treatment of BCBM. A nano-sized poly(lactone-co-β-amino ester) has been reported to deliver the CXC chemokine receptor 4 (CXCR4) gene to target BCBM via surface conjugation of AMD3100 in the cancer microenvironment [108]. Promelittin (proMel) is delineated to manifest secretory protein and discharges cytolytic melittin due to MMP-2 stimulation gathered at tumor sites. The release of proMel efficiently impedes cancer development through the AMD3100-conjugated nanoparticles in a BCBM mouse model. This is an innovative strategy for treating BCBM via targeted delivery of promelittin-led gene therapy [109].

13.2. Role of SOX2 Gene

SOX2 is overexpressed in cancer cells, helping in the adhesion of cancerous cells to the endothelial (microvascular) cells. It promotes trans-endothelial migration and BBB permeability, while the silencing of SOX2 prevents these incidents. These functions of SOX2 are possibly due to the upregulation of HBEGF and FSCN1, involving AKT and β-catenin signaling pathways. A recent in vivo study reported that SOX2 enhances the growth of BCBM [110].

13.3. Role of BRCA1/2 Gene

Approximately 5-10% of breast cancer cases are associated with the hereditary mutation of BRCA1 and BRCA1 genes [111]. The induction of BMs is common among females with BRCA2/BRCA1 mutations, especially those suffering from metastatic BC. Moreover, carrier-mediated mutation at BRCA2 shows a considerable rate of recurrence of CNS metastasis than the non-carrier type [112].
The crucial process of single-strand break repair in DNA is absent in BRCA1 and BRCA2 mutations, dramatically increasing breast/ovarian cancer chances. An early study reported a higher rate of BCBM in patients with BRCA1 mutations [113]. Although no statistical correlation was noted between BRCA1 mutation carriers and non-carriers (p = 0.06), BRCA1 mutations (58% vs. 24%) are believed to be a common mutation in BCBM patients [114]. A recent study reported that patients with BRCA1 and BRCA2 mutations have significantly higher rates of CNS metastases than non-carriers (BRCA1: 53% and BRCA2: 50% vs. non-carriers: 25%, respectively) [112].

14. Emerging Trends of Therapeutics to Treat BMs

Neratinib is an irreversible chloroanilino-quinazoline-derived inhibitor of HER2 [115]. Neratinib, combined with capecitabine, has shown its effectiveness, mainly for managing and treating HER2+ metastatic breast cancer (MBC) patients. On the other hand, pyrotinib is more effective and promising against HER2+ MBC, a pan-ErbB receptor TKI (tyrosine kinase inhibitor), binds irreversibly, and is orally given [116,117,118,119]. This drug showed encouraging results in intracranial control [93].

15. Forecast for BCBM Cure

Bevacizumab, a drug for cancer chemotherapy, has been reported to be effective in glioblastomas and has the potential to block VEGF-like growth factors, which is vital for the cure of BCBM [120]. Two studies supported the above observation. The first study observed 65% CNS responses for etoposide, cisplatin, and bevacizumab, and the second study noted 61% responses for the same drug [121]. However, based on the overall survival rate of brain metastatic cancer patients taking these drugs, it was withdrawn from Food and Drug Administration approval.
Capecitabine along with lapatinib and everolimus, in addition to one more combination of vinorelbine with trastuzumab and everolimus, were evaluated in HER2-positive BCBM patients [122,123]. This study showed a high level of brain metastases in triple-negative BC and HER2-positive patients, highlighting the requirement of cost-effective strategies against brain metastases risk groups and their subgroups [124]. One study reported that the expansion time of BCBM patients through triple-negative and HER2-positive is not comparable [125].
Pertuzumab is regarded as a systemic therapy for BCBM patients, which is a humanized monoclonal antibody that prevents HER2 dimerization ([126]. In patients with HER2+ brain metastases, a phase II clinical trial (NCT02536339) is underway to measure the efficacy and safety of a combination of pertuzumab and high doses of trastuzumab. Preliminary data of this finding were published by Lin et al. (2017).
A monoclonal antibody, a trastuzumab emtansine (T-DM1), is an antibody-drug conjugate made up of trastuzumab, associated with DM1 (a cytotoxic substance maytansine derivative) [127]. Some researchers reported beneficial case studies that the administration of T-DM1 was a well-tolerated therapeutic approach for patients with HER2+ BCBM [127,128]. Trastuzumab deruxtecan is an antibody-drug combination containing trastuzumab and a derivative of exatecan (topoisomerase I inhibitor), used for treating BCBM. For HER2+ metastatic breast cancer (MBC) that had received T-DM1 treatment previously, the phase II trial DESTINY-Breast01 examined the effectiveness and safety of this combination (NCT03248492) [129,130,131]. Several signaling cascades involved in BCBM, therapeutic targets, and their potential drugs have been depicted in Figure 4.
Tucatinib is a tyrosine kinase inhibitor, highly selective for the HER2 protein and can slightly inhibit the epidermal growth factor receptor [132]. Trastuzumab and capecitabine, in combination with tucatinib, demonstrated improved progression-free survival and enhanced CNS response in the BCBM treatment regimen [38,99,100,132]. Some recent studies also reported mTOR and PI3K inhibitors (Everolimus and Buparlisib (BKM120)) for the treatment of BCBM in combination with other anticancer drugs, such as trastuzumab and vinorelbine [38,133]. Pembrolizumab is a humanized monoclonal IgG4-antibody with high affinity and great selectivity against PD-1 [134]. PD-L1 inhibitors have been evaluated as targeted therapies for advanced BC in various trials. Pembrolizumab exhibited sufficient activity and safety for advanced TNBC, as shown by the KEYNOTE-012 trial (NCT01848834) [32,134]. Stereotactic radiosurgery (SRS) and atezolizumab are combined to manage TNB with BCBM in the phase II atezolizumab trial (NCT03483012) [135]. In addition, pembrolizumab and SRS are being tested in phase I/II trials for patients with BCBM to determine their effectiveness and safety (NCT03449238). In another clinical trial, patients with BCBM are assessed for SRS following nivolumab in phase I (NCT03807765) research [32]. Table 2 summarizes current/clinical trial drugs and their mechanism of action against BCBM.

16. Conclusions

Brain metastasis (BM) is common in breast cancer patients with poor prognosis and remains a life-threatening disease. There is a pressing need for effective therapy to cure breast cancers and breast cancer brain metastases. A thorough and detailed understanding of the CNS sequence process requires robust analytical data to manage primary and secondary BM. As the biology of BM is delicate, the challenges associated with BBB need to be handled to treat BCBMs effectively. Moreover, there is a greater risk of BC treatment at the advanced stage due to the more complex tumor microenvironment and deregulatory pathways, ultimately leading to BM. Targeting the mTOR pathway is one of the effective and commonly used treatments for BC patients. However, there is no well-defined treatment plan for brain metastases. Presently, several studies on BCBM are underway, which are expected to help scientists to chalk out an effective treatment strategy against this life threatening disease. In addition, there is an urgent need for multidimensional in vitro and in vivo studies involving the epigenetic players so that the cancer cells can be arrested in the G2/M phase. Furthermore, downregulation of the oncogenes (EGFR, VEGFR, PDGFR, HER2) accompanied by upregulation of the tumor suppressor genes (p53, p73, p21, RB, BRCA1/BRCA2, PTEN gene) can also orchestrate effective apoptosis in a tumor cell.

Funding

This research work was funded by Institutional Fund Projects under grant no. (IFPDP-51-22). Therefore, the authors gratefully acknowledge technical and financial support from the Ministry of Education and Deanship of Scientific Research (DSR), King Abdulaziz University (KAU), Jeddah, Saudi Arabia.

Acknowledgments

The authors greatly appreciate the research facility provided by the King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BBBBlood–brain barrier
BCBreast cancer
BCBMBreast cancer brain metastasis
bHER2-ATCBiparatopic anti-HER2 antibody-tubulysin conjugate
BMsBrain metastases
BMsBCBrain metastases breast cancer
BRCA1Breast cancer type 1
cANGPTL4Angiopoietin-like 4 fibrinogen-like domain
CNSCentral nervous system
COX-2/MMP1Cyclooxygenase 2/Matrix metalloproteinase 1
CXCR4CXC chemokine receptor 4
DTIDiffusion tensor imaging
EREstrogen
GPAGraded prognostic assessment
HDACsHistone deacetylases
HER2Human epidermal growth factor receptor 2
IGF1RType 1 insulin-like growth factor receptor
LncRNALong noncoding RNA
MBCMetastatic breast cancer
MEF2Myocyte enhancer factor 2
miRNAsMicroRNAs
MMP-2Matrix metalloproteinase-2
mTORMammalian target of rapamycin
NPNTNephronectin
PCIProphylactic cranial irradiation
PDGFPlatelet-derived growth factor
PDGFRβPlatelet-derived growth factor receptor-beta
PD-1Programmed death receptor 1
PDXsPatient-derived xenografts
PI3K/AKTPhosphatidylinositol-3-kinase/Protein kinase B
PRProgesterone
RRM2Ribonucleotide reductase M2
SRSStereotactic radiosurgery
T-DM1Trastuzumab emtansine
TGLI1Truncated glioma-associated oncogene homolog 1
TNBCTriple-negative breast cancer
VEGFVascular endothelial growth factor
WBRTWhole-brain radiation targeting

References

  1. Alitheen, N.B.; Yeap, S.K.; Faujan, N.H.; Ho, W.Y.; Beh, B.K.; Mashitoh, A.R. Leukemia and therapy. Am. J. Immunol. 2011, 7, 54–61. [Google Scholar] [CrossRef] [Green Version]
  2. Kaleem, M.; Perwaiz, M.; Nur, S.M.; Abdulrahman, A.O.; Ahmad, W.; Al-Abbasi, F.A.; Kumar, V.; Kamal, M.A.; Anwar, F. Epigenetics of Triple-Negative Breast Cancer via Natural Compounds. Curr. Med. Chem. 2021, 28, 1436–1458. [Google Scholar] [CrossRef] [PubMed]
  3. Alhosin, M.; Sharif, T.; Mousli, M.; Etienne-Selloum, N.; Fuhrmann, G.; Schini-Kerth, V.B.; Bronner, C. Down-regulation of UHRF1, associated with re-expression of tumor suppressor genes, is a common feature of natural compounds exhibiting anti-cancer properties. J. Exp. Clin. Cancer Res. 2011, 30, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sharma, M.; Sharma, J.D.; Sarma, A.; Ahmed, S.; Kataki, A.C.; Saxena, R.; Sharma, D. Triple negative breast cancer in people of North East India: Critical insights gained at a regional cancer centre. Asian Pac. J. Cancer Prev. 2014, 15, 4507–4511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jabbarzadeh Kaboli, P.; Salimian, F.; Aghapour, S.; Xiang, S.; Zhao, Q.; Li, M.; Wu, X.; Du, F.; Zhao, Y.; Shen, J.; et al. Akt-targeted therapy as a promising strategy to overcome drug resistance in breast cancer—A comprehensive review from chemotherapy to immunotherapy. Pharmacol. Res. 2020, 156, 104806. [Google Scholar] [CrossRef]
  6. Michniak-Kohn, B.B.; Deol, P.K.; Kotta, S.; Aldawsari, H.M.; Badr-Eldin, S.M.; Nair, A.B.; Kaleem, M.; Dalhat, M.H. Thermosensitive Hydrogels Loaded with Resveratrol Nanoemulsion: Formulation Optimization by Central Composite Design and Evaluation in MCF-7 Human Breast Cancer Cell Lines. Gels 2022, 8, 450. [Google Scholar] [CrossRef]
  7. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  8. Loibl, S.; Gianni, L. HER2-positive breast cancer. Lancet 2017, 389, 2415–2429. [Google Scholar] [CrossRef]
  9. Ensenyat-Mendez, M.; Llinàs-Arias, P.; Orozco, J.I.J.; Íñiguez-Muñoz, S.; Salomon, M.P.; Sesé, B.; DiNome, M.L.; Marzese, D.M. Current Triple-Negative Breast Cancer Subtypes: Dissecting the Most Aggressive Form of Breast Cancer. Front. Oncol. 2021, 11, 681476. [Google Scholar] [CrossRef]
  10. Tsang, J.Y.S.; Tse, G.M. Molecular Classification of Breast Cancer. Adv. Anat. Pathol. 2020, 27, 27–35. [Google Scholar] [CrossRef]
  11. Aldape, K.; Brindle, K.M.; Chesler, L.; Chopra, R.; Gajjar, A.; Gilbert, M.R.; Gottardo, N.; Gutmann, D.H.; Hargrave, D.; Holland, E.C.; et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol. 2019, 16, 509–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Belykh, E.; Shaffer, K.V.; Lin, C.; Byvaltsev, V.A.; Preul, M.C.; Chen, L. Blood-Brain Barrier, Blood-Brain Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain Tumors. Front. Oncol. 2020, 10, 739. [Google Scholar] [CrossRef] [PubMed]
  13. Krizbai, I.A.; Nyúl-Tóth, Á.; Bauer, H.H.; Farkas, A.E.; Traweger, A.; Haskó, J.; Wilhelm, I.; Bauer, H.C. Pharmaceutical Targeting of the Brain. Curr. Pharm. Des. 2016, 22, 5442–5462. [Google Scholar] [CrossRef] [PubMed]
  14. Quattrocchi, C.C.; Errante, Y.; Gaudino, C.; Mallio, C.A.; Giona, A.; Santini, D.; Tonini, G.; Zobel, B.B. Spatial brain distribution of intra-axial metastatic lesions in breast and lung cancer patients. J. Neurooncol. 2012, 110, 79–87. [Google Scholar] [CrossRef]
  15. Takano, K.; Kinoshita, M.; Takagaki, M.; Sakai, M.; Tateishi, S.; Achiha, T.; Hirayama, R.; Nishino, K.; Uchida, J.; Kumagai, T.; et al. Different spatial distributions of brain metastases from lung cancer by histological subtype and mutation status of epidermal growth factor receptor. Neur. Oncol. 2016, 18, 716–724. [Google Scholar] [CrossRef] [Green Version]
  16. Izutsu, N.; Kinoshita, M.; Ozaki, T.; Sakai, M.; Nakanishi, K.; Nakayama, T.; Tamaki, Y.; Kishima, H. Cerebellar preference of luminal a and b type and basal ganglial preference of her2-positive type breast cancer-derived brain metastases. Mol. Clin. Oncol. 2021, 15, 175. [Google Scholar] [CrossRef]
  17. Achrol, A.S.; Rennert, R.C.; Anders, C.; Soffietti, R.; Ahluwalia, M.S.; Nayak, L.; Peters, S.; Arvold, N.D.; Harsh, G.R.; Steeg, P.S.; et al. Brain metastases. Nat. Rev. Dis. Prim. 2019, 5, 5. [Google Scholar] [CrossRef]
  18. Matzenauer, M.; Vrana, D.; Melichar, B. Treatment of brain metastases. Biomed. Pap. 2016, 160, 484–490. [Google Scholar] [CrossRef] [Green Version]
  19. Bollig-Fischer, A.; Michelhaugh, S.; Ali-Fehmi, R.; Mittal, S. The molecular genomics of metastatic brain tumours. OA Mol. Oncol. 2013, 1, 6. [Google Scholar] [CrossRef] [Green Version]
  20. Hosonaga, M.; Saya, H.; Arima, Y. Molecular and cellular mechanisms underlying brain metastasis of breast cancer. Cancer Metastasis Rev. 2020, 39, 711–720. [Google Scholar] [CrossRef]
  21. Franchino, F.; Rudà, R.; Soffietti, R. Mechanisms and therapy for cancer metastasis to the brain. Front. Oncol. 2018, 8, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Knier, N.N.; Hamilton, A.M.; Foster, P.J. Comparing the fate of brain metastatic breast cancer cells in different immune compromised mice with cellular magnetic resonance imaging. Clin. Exp. Metastasis 2020, 37, 465–475. [Google Scholar] [CrossRef] [PubMed]
  23. American Cancer Society. Breast Cancer Facts & Figures 2019–2020. Am. Cancer Soc. 2019, 1–44. [Google Scholar]
  24. Zhang, L.; Ridgway, L.D.; Wetzel, M.D.; Ngo, J.; Yin, W.; Kumar, D.; Goodman, J.C.; Groves, M.D.; Marchetti, D. The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci. Transl. Med. 2013, 5, 180ra48. [Google Scholar] [CrossRef] [Green Version]
  25. Harbeck, N.; Gnant, M. Breast cancer. Lancet 2017, 389, 1134–1150. [Google Scholar] [CrossRef]
  26. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast cancer. Nat. Rev. Dis. Prim. 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  27. Tsuji, W.; Plock, J.A. Breast Cancer Metastasis. Introd. Cancer Metastasis 2017, 2017, 13–31. [Google Scholar] [CrossRef]
  28. Chia, S.K.; Ellard, S.L.; Mates, M.; Welch, S.; Mihalcioiu, C.; Miller, W.H.; Gelmon, K.; Lohrisch, C.; Kumar, V.; Taylor, S.; et al. A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer. Breast Cancer Res. 2017, 19, 54. [Google Scholar] [CrossRef] [Green Version]
  29. Venur, V.A.; Leone, J.P. Targeted therapies for brain metastases from breast cancer. Int. J. Mol. Sci. 2016, 17, 1543. [Google Scholar] [CrossRef]
  30. Cameron, D.; Piccart-Gebhart, M.J.; Gelber, R.D.; Procter, M.; Goldhirsch, A.; de Azambuja, E.; Castro, G.; Untch, M.; Smith, I.; Gianni, L.; et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: Final analysis of the HERceptin Adjuvant (HERA) trial. Lancet 2017, 389, 1195–1205. [Google Scholar] [CrossRef] [Green Version]
  31. Friedrich, M.; Swords, D.; Terjung Thill, M.; Baum, S.; Bischoff, J. Breast cancer and metastases of the central nervous system. Eur. J. Gynaecol. Oncol. 2017, 38, 653–656. [Google Scholar] [CrossRef]
  32. Watase, C.; Shiino, S.; Shimoi, T.; Noguchi, E.; Kaneda, T.; Yamamoto, Y.; Yonemori, K.; Takayama, S.; Suto, A. Breast cancer brain metastasis—Overview of disease state, treatment options and future perspectives. Cancers 2021, 13, 1078. [Google Scholar] [CrossRef] [PubMed]
  33. Bailleux, C.; Eberst, L.; Bachelot, T. Treatment strategies for breast cancer brain metastases. Br. J. Cancer 2021, 124, 142–155. [Google Scholar] [CrossRef] [PubMed]
  34. Redig, A.J.; McAllister, S.S. Breast cancer as a systemic disease: A view of metastasis. J. Intern. Med. 2013, 274, 113–126. [Google Scholar] [CrossRef] [Green Version]
  35. Soffietti, R.; Abacioglu, U.; Baumert, B.; Combs, S.E.; Kinhult, S.; Kros, J.M.; Marosi, C.; Metellus, P.; Radbruch, A.; Freixa, S.S.V.; et al. Diagnosis and treatment of brain metastases from solid tumors: Guidelines from the European Association of neuro-oncology (EANO). Neur. Oncol. 2017, 19, 162–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Yurtsever, I.; Sari, L.; Gultekin, M.A.; Toprak, H.; Turk, H.M.; Aliyev, A.; Peker, A.A.; Yabaci, A.; Alkan, A. Diffusion Tensor Imaging of Brain Metastases in Patients with Breast Cancer According to Molecular Subtypes. Curr. Med. Imaging Former. Curr. Med. Imaging Rev. 2020, 17, 120–128. [Google Scholar] [CrossRef]
  37. Harris, J.R.; Lippman, M.E.; Veronesi, U.; Willett, W. Breast cancer. N. Engl. J. Med. 1992, 327, 319–328. [Google Scholar] [CrossRef]
  38. Riecke, K.; Müller, V.; Weide, R.; Schmidt, M.; Park-simon, T.W.; Möbus, V.; Mundhenke, C.; Polasik, A.; Lübbe, K.; Hesse, T.; et al. Predicting prognosis of breast cancer patients with brain metastases in the BMBC registry—Comparison of three different GPA prognostic scores. Cancers 2021, 13, 844. [Google Scholar] [CrossRef]
  39. Goss, G.; Tsai, C.M.; Shepherd, F.A.; Ahn, M.J.; Bazhenova, L.; Crinò, L.; de Marinis, F.; Felip, E.; Morabito, A.; Hodge, R.; et al. CNS response to osimertinib in patients with T790M-positive advanced NSCLC: Pooled data from two phase II trials. Ann. Oncol. 2018, 29, 687–693. [Google Scholar] [CrossRef]
  40. Abbott, N.J. Blood—brain barrier structure and function and the challenges for CNS drug delivery. J. Inherit. Metab. Dis. 2013, 36, 437–449. [Google Scholar] [CrossRef]
  41. O’Brown, N.M.; Pfau, S.J.; Gu, C. Bridging barriers: A comparative look at the blood—brain barrier across organisms. Genes Dev. 2018, 32, 466–478. [Google Scholar] [CrossRef]
  42. Hobbs, S.K.; Monsky, W.L.; Yuan, F.; Roberts, W.G.; Griffith, L.; Torchilin, V.P.; Jain, R.K. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc. Natl. Acad. Sci. USA 1998, 95, 4607–4612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K.; et al. Is the blood—brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neur. Oncol. 2018, 20, 184–191. [Google Scholar] [CrossRef] [PubMed]
  44. Thomsen, M.S.; Routhe, L.J.; Moos, T. The vascular basement membrane in the healthy and pathological brain. J. Cereb. Blood Flow Metab. 2017, 37, 3300–3317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood--brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [Green Version]
  46. Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef]
  47. Giaume, C.; Koulakoff, A.; Roux, L.; Holcman, D.; Rouach, N. Astroglial networks: A step further in neuroglial and gliovascular interactions. Nat. Rev. Neurosci. 2010, 11, 87–99. [Google Scholar] [CrossRef]
  48. Watkins, S.; Robel, S.; Kimbrough, I.F.; Robert, S.M.; Ellis-Davies, G.; Sontheimer, H. Disruption of astrocyte—Vascular coupling and the blood—Brain barrier by invading glioma cells. Nat. Commun. 2014, 5, 4196. [Google Scholar] [CrossRef] [Green Version]
  49. Lannes, N.; Eppler, E.; Etemad, S.; Yotovski, P.; Filgueira, L. Microglia at center stage: A comprehensive review about the versatile and unique residential macrophages of the central nervous system. Oncotarget 2017, 8, 114393. [Google Scholar] [CrossRef] [Green Version]
  50. Shemer, A.; Erny, D.; Jung, S.; Prinz, M. Microglia plasticity during health and disease: An immunological perspective. Trends Immunol. 2015, 36, 614–624. [Google Scholar] [CrossRef]
  51. Iadecola, C. The neurovascular unit coming of age: A journey through neurovascular coupling in health and disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef] [PubMed]
  52. de Vries, H.E.; Kooij, G.; Frenkel, D.; Georgopoulos, S.; Monsonego, A.; Janigro, D. Inflammatory events at blood—Brain barrier in neuroinflammatory and neurodegenerative disorders: Implications for clinical disease. Epilepsia 2012, 53, 45–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Groothuis, D.R.; Molnar, P.; Blasberg, R.G. Regional blood flow and blood-to-tissue transport in five brain tumor models. Brain Tumor Biol. 1984, 27, 132–153. [Google Scholar]
  55. Gerstner, E.R.; Duda, D.G.; di Tomaso, E.; Ryg, P.A.; Loeffler, J.S.; Sorensen, A.G.; Ivy, P.; Batchelor, T.T. VEGF inhibitors in the treatment of cerebral edema in patients with brain cancer. Nat. Rev. Clin. Oncol. 2009, 6, 229–236. [Google Scholar] [CrossRef]
  56. Machein, M.R.; Kullmer, J.; Fiebich, B.L.; Plate, K.H.; Warnke, P.C. Vascular endothelial growth factor expression, vascular volume, and capillary permeability in human brain tumors. Neurosurgery 1999, 44, 732–741. [Google Scholar] [CrossRef]
  57. Blasberg, R.G.; Nakagawa, H.; Bourdon, M.A.; Groothuis, D.R.; Patlak, C.S.; Bigner, D.D. Regional localization of a glioma-associated antigen defined by monoclonal antibody 81C6 in vivo: Kinetics and implications for diagnosis and therapy. Cancer Res. 1987, 47, 4432–4443. [Google Scholar]
  58. Takamiya, Y.; Abe, Y.; Tanaka, Y.; Tsugu, A.; Kazuno, M.; Oshika, Y.; Maruo, K.; Ohnishi, Y.; Sato, O.; Yamazaki, H.; et al. Murine P-glycoprotein on stromal vessels mediates multidrug resistance in intracerebral human glioma xenografts. Br. J. Cancer 1997, 76, 445–450. [Google Scholar] [CrossRef]
  59. la Fougere, C.; Suchorska, B.; Bartenstein, P.; Kreth, F.-W.; Tonn, J.-C. Molecular imaging of gliomas with PET: Opportunities and limitations. Neur. Oncol. 2011, 13, 806–819. [Google Scholar] [CrossRef] [Green Version]
  60. Hardee, M.E.; Zagzag, D. Mechanisms of glioma-associated neovascularization. Am. J. Pathol. 2012, 181, 1126–1141. [Google Scholar] [CrossRef] [Green Version]
  61. Plate, K.H.; Scholz, A.; Dumont, D.J. Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited. Acta Neuropathol. 2012, 124, 763–775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Juillerat-Jeanneret, L. The targeted delivery of cancer drugs across the blood—Brain barrier: Chemical modifications of drugs or drug-nanoparticles? Drug Discov. Today 2008, 13, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  63. Ningaraj, N.S.; Rao, M.; Hashizume, K.; Asotra, K.; Black, K.L. Regulation of blood-brain tumor barrier permeability by calcium-activated potassium channels. J. Pharmacol. Exp. Ther. 2002, 301, 838–851. [Google Scholar] [CrossRef]
  64. Nussbaum, E.S.; Djalilian, H.R.; Cho, K.H.; Hall, W.A. Brain metastases: Histology, multiplicity, surgery, and survival. Cancer 1996, 78, 1781–1788. [Google Scholar] [CrossRef]
  65. Chamberlain, M.C.; Baik, C.S.; Gadi, V.K.; Bhatia, S.; Chow, L.Q.M. Systemic therapy of brain metastases: Non-small cell lung cancer, breast cancer, and melanoma. Neur. Oncol. 2017, 19, i1–i24. [Google Scholar] [CrossRef] [Green Version]
  66. Halasz, L.M.; Uno, H.; Hughes, M.; D’Amico, T.; Dexter, E.U.; Edge, S.B.; Hayman, J.A.; Niland, J.C.; Otterson, G.A.; Pisters, K.M.W.; et al. Comparative effectiveness of stereotactic radiosurgery versus whole-brain radiation therapy for patients with brain metastases from breast or non–small cell lung cancer. Cancer 2016, 122, 2091–2100. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, Q.; Chen Jian Yu, X.; Cai, G.; Yang, Z.; Cao, L.; Hu, C.; Guo, X.; Sun, J.; Chen, J. Survival benefit of anti-HER2 therapy after whole-brain radiotherapy in HER2-positive breast cancer patients with brain metastasis. Breast Cancer 2016, 23, 732–739. [Google Scholar] [CrossRef]
  68. Lee, J.S.; Toktas, O.; Soran, A. Role of Locoregional Treatment in De Novo Stage IV Breast Cancer. Clin. Med. Insights Oncol. 2020, 14, 1–5. [Google Scholar] [CrossRef]
  69. Yoshimura, M. Radiation therapy for primary tumor of de novo stage IV breast cancer. Transl. Cancer Res. 2020, 9, 5108. [Google Scholar] [CrossRef]
  70. Santori, F.; Vanni, G.; Buonomo, O.C.; De Majo, A.; Rho, M.; Granai, A.V.; Pellicciaro, M.; Cotesta, M.; Assogna, M.; D’Angelillo, R.M.; et al. Ulcerated breast cancer with single brain metastasis: A combined surgical approach. Clinical presentation at one year follow up—A case report. Int. J. Surg. Case Rep. 2020, 73, 75–78. [Google Scholar] [CrossRef]
  71. Pérez, M.; Schootman, M.; Hall, L.E.; Jeffe, D.B. Accelerated partial breast irradiation compared with whole breast radiation therapy: A breast cancer cohort study measuring change in radiation side-effects severity and quality of life. Breast Cancer Res. Treat. 2017, 162, 329–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Possanzini, M.; Greco, C. Stereotactic radiotherapy in metastatic breast cancer. Breast 2018, 41, 57–66. [Google Scholar] [CrossRef] [PubMed]
  73. Ma, J.; Sanchez-Duffhues, G.; Goumans, M.J.; ten Dijke, P. TGF-β-Induced Endothelial to Mesenchymal Transition in Disease and Tissue Engineering. Front. Cell Dev. Biol. 2020, 8, 260. [Google Scholar] [CrossRef] [PubMed]
  74. Chow, A.; Arteaga, C.L.; Wang, S.E. When tumor suppressor TGFβ meets the HER2 (ERBB2) oncogene. J. Mammary Gland. Biol. Neoplasia 2011, 16, 81–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Grusch, M.; Petz, M.; Metzner, T.; Ozturk, D.; Schneller, D.; Mikulits, W. The crosstalk of RAS with the TGF-β family during carcinoma progression and its implications for targeted cancer therapy. Curr. Cancer Drug Targets 2010, 10, 849–857. [Google Scholar] [CrossRef] [PubMed]
  76. Demirkan, B. The Roles of Epithelial-to-Mesenchymal Transition (EMT) and Mesenchymal-to-Epithelial Transition (MET) in Breast Cancer Bone Metastasis: Potential Targets for Prevention and Treatment. J. Clin. Med. 2013, 2, 264–282. [Google Scholar] [CrossRef] [Green Version]
  77. Liu, F.; Gu, L.N.; Shan, B.E.; Geng, C.Z.; Sang, M.X. Biomarkers for EMT and MET in breast cancer: An update (review). Oncol. Lett. 2016, 12, 4869–4876. [Google Scholar] [CrossRef] [Green Version]
  78. Witzel, I.; Oliveira-Ferrer, L.; Pantel, K.; Müller, V.; Wikman, H. Breast cancer brain metastases: Biology and new clinical perspectives. Breast Cancer Res. 2016, 18, 8. [Google Scholar] [CrossRef] [Green Version]
  79. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. Mech. Dis. 2018, 13, 395–412. [Google Scholar] [CrossRef]
  80. Krizbai, I.A.; Gasparics, Á.; Nagyőszi, P.; Fazakas, C.; Molnár, J.; Wilhelm, I.; Bencs, R.; Rosivall, L.; Sebe, A. Endothelial-mesenchymal transition of brain endothelial cells: Possible role during metastatic extravasation. PLoS ONE 2015, 10, e0119655. [Google Scholar] [CrossRef] [Green Version]
  81. Drappatz, J.; Brenner, A.; Wong, E.T.; Eichler, A.; Schiff, D.; Groves, M.D.; Mikkelsen, T.; Rosenfeld, S.; Sarantopoulos, J.; Meyers, C.A.; et al. Phase I study of GRN1005 in recurrent malignant glioma. Clin. Cancer Res. 2013, 19, 1567–1576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Li, F.; Tang, S.C. Targeting metastatic breast cancer with ANG1005, a novel peptide-paclitaxel conjugate that crosses the blood-brain-barrier (BBB). Genes Dis. 2017, 4, 1–3. [Google Scholar] [CrossRef] [PubMed]
  83. Islam, B.U.; Khan, M.S.; Husain, F.M.; Rehman, M.T.; Zughaibi, T.A.; Abuzenadah, A.M.; Urooj, M.; Kamal, M.A.; Tabrez, S. mTOR Targeted Cancer Chemoprevention by Flavonoids. Curr. Med. Chem. 2020, 8, 8068–8082. [Google Scholar] [CrossRef]
  84. ul Islam, B.; Suhail, M.; Khan, M.S.; Ahmad, A.; Zughaibi, T.A.; Husain, F.M.; Rehman, M.T.; Tabrez, S. Flavonoids and PI3K/Akt/mTOR Signaling Cascade: A Potential Crosstalk in Anticancer Treatment. Curr. Med. Chem. 2021, 28, 8083–8097. [Google Scholar] [CrossRef]
  85. Zughaibi, T.A.; Suhail, M.; Tarique, M.; Tabrez, S. Targeting pi3k/akt/mtor pathway by different flavonoids: A cancer chemopreventive approach. Int. J. Mol. Sci. 2021, 22, 12455. [Google Scholar] [CrossRef]
  86. Blazquez, R.; Wlochowitz, D.; Wolff, A.; Seitz, S.; Wachter, A.; Perera-Bel, J.; Bleckmann, A.; Beißbarth, T.; Salinas, G.; Riemenschneider, M.J.; et al. PI3K: A master regulator of brain metastasis-promoting macrophages/microglia. Glia 2018, 66, 2438–2455. [Google Scholar] [CrossRef]
  87. Hohensee, I.; Chuang, H.-N.N.; Grottke, A.; Werner, S.; Schulte, A.; Horn, S.; Lamszus, K.; Bartkowiak, K.; Witzel, I.; Westphal, M.; et al. PTEN mediates the cross talk between breast and glial cells in brain metastases leading to rapid disease progression. Oncotarget 2017, 8, 6155–6168. [Google Scholar] [CrossRef] [Green Version]
  88. Corti, C.; Antonarelli, G.; Criscitiello, C.; Lin, N.U.; Carey, L.A.; Cortés, J.; Poortmans, P.; Curigliano, G. Targeting brain metastases in breast cancer. Cancer Treat. Rev. 2022, 103, 102324. [Google Scholar] [CrossRef]
  89. Schmit, F.; Utermark, T.; Zhang, S.; Wang, Q.; Von, T.; Roberts, T.M.; Zhao, J.J. PI3K isoform dependence of PTEN-deficient tumors can be altered by the genetic context. Proc. Natl. Acad. Sci. USA 2014, 111, 6395–6400. [Google Scholar] [CrossRef] [Green Version]
  90. Ni, J.; Ramkissoon, S.H.; Xie, S.; Goel, S.; Stover, D.G.; Guo, H.; Luu, V.; Marco, E.; Ramkissoon, L.A.; Kang, Y.J.; et al. Combination inhibition of PI3K and mTORC1 yields durable remissions in mice bearing orthotopic patient-derived xenografts of HER2-positive breast cancer brain metastases. Nat. Med. 2016, 22, 723–726. [Google Scholar] [CrossRef]
  91. Elmenier, F.M.; Lasheen, D.S.; Abouzid, K.A.M. Phosphatidylinositol 3 kinase (PI3K) inhibitors as new weapon to combat cancer. Eur. J. Med. Chem. 2019, 183, 111718. [Google Scholar] [CrossRef] [PubMed]
  92. Kodack, D.P.; Askoxylakis, V.; Ferraro, G.B.; Sheng, Q.; Badeaux, M.; Goel, S.; Qi, X.; Shankaraiah, R.; Cao, Z.A.; Ramjiawan, R.R.; et al. The brain microenvironment mediates resistance in luminal breast cancer to PI3K inhibition through HER3 activation. Sci. Transl. Med. 2017, 9, eaal4682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lin, Y.; Lin, M.; Zhang, J.; Wang, B.; Tao, Z.; Du, Y.; Zhang, S.; Cao, J.; Wang, L.; Hu, X. Real-World Data of Pyrotinib-Based Therapy in Metastatic HER2-Positive Breast Cancer: Promising Efficacy in Lapatinib-Treated Patients and in Brain Metastasis. Cancer Res Treat. 2020, 52, 1059–1066. [Google Scholar] [CrossRef] [PubMed]
  94. Da Silva, L.; Simpson, P.T.; Smart, C.E.; Cocciardi, S.; Waddell, N.; Lane, A.; Morrison, B.J.; Vargas, A.C.; Healey, S.; Beesley, J.; et al. HER3 and downstream pathways are involved in colonization of brain metastases from breast cancer. Breast Cancer Res. 2010, 12, R46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Giri, D.K.; Ali-Seyed, M.; Li, L.-Y.; Lee, D.-F.; Ling, P.; Bartholomeusz, G.; Wang, S.-C.; Hung, M.-C. Endosomal Transport of ErbB-2: Mechanism for Nuclear Entry of the Cell Surface Receptor. Mol. Cell. Biol. 2005, 25, 11005–11018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Wang, S.-C.C.; Lien, H.-C.C.; Xia, W.; Chen, I.-F.F.; Lo, H.-W.W.; Wang, Z.; Ali-Seyed, M.; Lee, D.-F.F.; Bartholomeusz, G.; Ou-Yang, F.; et al. Binding at and transactivation of the COX-2 promoter by nuclear tyrosine kinase receptor ErbB-2. Cancer cell 2004, 6, 251–261. [Google Scholar] [CrossRef] [Green Version]
  97. Bryan, S.; Witzel, I.; Borgmann, K.; Oliveira-Ferrer, L. Molecular mechanisms associated with brain metastases in her2-positive and triple negative breast cancers. Cancers 2021, 13, 4137. [Google Scholar] [CrossRef]
  98. Sirkisoon, S.R.; Carpenter, R.L.; Rimkus, T.; Miller, L.; Metheny-Barlow, L.; Lo, H.W. EGFR and HER2 signaling in breast cancer brain metastasis. Front. Biosci. (Elite Ed.) 2016, 8, 245–263. [Google Scholar] [CrossRef] [Green Version]
  99. Balch, S.M.; Vaz-Luis, I.; Li, T.; Tayob, N.; Jain, E.; Helvie, K.; Buendia-Buendia, J.E.; Shannon, E.; Isakoff, S.J.; Tung, N.M.; et al. A phase II study of efficacy, toxicity, and the potential impact of genomic alterations on response to eribulin mesylate in combination with trastuzumab and pertuzumab in women with human epidermal growth factor receptor 2 (HER2)+ metastatic breast cancer. Breast Cancer Res. Treat. 2021, 189, 411–423. [Google Scholar] [CrossRef]
  100. Batra, A.; Kong, S.; Cheung, W.Y. Eligibility of real-world patients with metastatic breast cancer for clinical trials. Breast 2020, 54, 171–178. [Google Scholar] [CrossRef]
  101. Lo, H.-W.W.; Hsu, S.-C.C.; Xia, W.; Cao, X.; Shih, J.-Y.Y.; Wei, Y.; Abbruzzese, J.L.; Hortobagyi, G.N.; Hung, M.-C.C. Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res. 2007, 67, 9066–9076. [Google Scholar] [CrossRef] [Green Version]
  102. Heiland, D.H.; Ravi, V.M.; Behringer, S.P.; Frenking, J.H.; Wurm, J.; Joseph, K.; Garrelfs, N.W.C.; Strähle, J.; Heynckes, S.; Grauvogel, J.; et al. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma. Nat. Commun. 2019, 10, 2541. [Google Scholar] [CrossRef] [Green Version]
  103. Anderson, M.A.; Burda, J.E.; Ren, Y.; Ao, Y.; O’Shea, T.M.; Kawaguchi, R.; Coppola, G.; Khakh, B.S.; Deming, T.J.; Sofroniew, M.V. Astrocyte scar formation AIDS central nervous system axon regeneration. Nature 2016, 532, 195–200. [Google Scholar] [CrossRef]
  104. Priego, N.; Zhu, L.; Monteiro, C.; Mulders, M.; Wasilewski, D.; Bindeman, W.; Doglio, L.; Martinez-Saez, E.; y Cajal, S.R.; Megías, D. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. 2018, 24, 1024–1035. [Google Scholar] [CrossRef]
  105. Kim, S.-J.; Kim, J.-S.; Park, E.S.; Lee, J.-S.; Lin, Q.; Langley, R.R.; Maya, M.; He, J.; Kim, S.-W.; Weihua, Z.; et al. Astrocytes upregulate survival genes in tumor cells and induce protection from chemotherapy. Neoplasia 2011, 13, 286–298. [Google Scholar] [CrossRef] [Green Version]
  106. Crinò, L.; Bronte, G.; Bidoli, P.; Cravero, P.; Minenza, E.; Cortesi, E.; Garassino, M.C.; Proto, C.; Cappuzzo, F.; Grossi, F.; et al. Nivolumab and brain metastases in patients with advanced non-squamous non-small cell lung cancer. Lung Cancer 2019, 129, 35–40. [Google Scholar] [CrossRef] [Green Version]
  107. Lorger, M.; Andreou, T.; Fife, C.; James, F. Immune Checkpoint Blockade—How Does It Work in Brain Metastases? Front. Mol. Neurosci. 2019, 12, 282. [Google Scholar] [CrossRef]
  108. Guo, X.; Deng, G.; Liu, J.; Zou, P.; Du, F.; Liu, F.; Chen, A.T.; Hu, R.; Li, M.; Zhang, S.; et al. Thrombin-responsive, brain-targeting nanoparticles for improved stroke therapy. ACS Nano 2018, 12, 8723–8732. [Google Scholar] [CrossRef]
  109. Zhou, Y.; Zhang, S.; Chen, Z.; Bao, Y.; Chen, A.T.; Sheu, W.C.; Liu, F.; Jiang, Z.; Zhou, J. Targeted Delivery of Secretory Promelittin via Novel Poly(lactone-co-β-amino ester) Nanoparticles for Treatment of Breast Cancer Brain Metastases. Adv. Sci. 2020, 7, 1901866. [Google Scholar] [CrossRef] [Green Version]
  110. Xiao, W.; Zheng, S.; Xie Xinhua Li, X.; Zhang, L.; Yang, A.; Wang, J.; Tang, H.; Xie, X. SOX2 Promotes Brain Metastasis of Breast Cancer by Upregulating the Expression of FSCN1 and HBEGF. Mol. Ther. Oncolytics 2020, 17, 118–129. [Google Scholar] [CrossRef]
  111. Claus, E.B.; Schildkraut, J.M.; Thompson, W.D.; Risch, N.J. The genetic attributable risk of breast and ovarian cancer. Cancer 1996, 77, 2318–2324. [Google Scholar] [CrossRef]
  112. Song, Y.; Barry, W.T.; Seah, D.S.; Tung, N.M.; Garber, J.E.; Lin, N.U. Patterns of recurrence and metastasis in BRCA1/BRCA2-associated breast cancers. Cancer 2020, 126, 271–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Albiges, L.; André, F.; Balleyguier, C.; Gomez-Abuin, G.; Chompret, A.; Delaloge, S. Spectrum of breast cancer metastasis in BRCA1 mutation carriers: Highly increased incidence of brain metastases. Ann. Oncol. 2005, 16, 1846–1847. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, L.J.; Alexander, B.; Schnitt, S.J.; Comander, A.; Gallagher, B.; Garber, J.E.; Tung, N. Clinical outcome of triple negative breast cancer in BRCA1 mutation carriers and non-carriers. Cancer 2011, 117, 3093–3100. [Google Scholar] [CrossRef] [Green Version]
  115. Tsou, H.-R.; Overbeek-Klumpers, E.G.; Hallett, W.A.; Reich, M.F.; Floyd, M.B.; Johnson, B.D.; Michalak, R.S.; Nilakantan, R.; Discafani, C.; Golas, J.; et al. Optimization of 6, 7-disubstituted-4-(arylamino) quinoline-3-carbonitriles as orally active, irreversible inhibitors of human epidermal growth factor receptor-2 kinase activity. J. Med. Chem. 2005, 48, 1107–1131. [Google Scholar] [CrossRef]
  116. Jiang, Z.; Yan, M.; Hu, X.; Zhang, Q.; Ouyang, Q.; Feng, J.; Yin, Y.; Sun, T.; Tong, Z.; Wang, X.; et al. Pyrotinib combined with capecitabine in women with HER2+ metastatic breast cancer previously treated with trastuzumab and taxanes: A randomized phase III study. J. Clin. Oncol. 2019, 37, 1001. [Google Scholar] [CrossRef]
  117. Li, X.; Yang, C.; Wan, H.; Zhang, G.; Feng, J.; Zhang Lei Chen, X.; Zhong, D.; Lou, L.; Tao, W.; Zhang, L. Discovery and development of pyrotinib: A novel irreversible EGFR/HER2 dual tyrosine kinase inhibitor with favorable safety profiles for the treatment of breast cancer. Elsevier 2017, 110, 51–61. [Google Scholar] [CrossRef]
  118. Ma, F.; Li, Q.; Chen, S.; Zhu, W.; Fan, Y.; Wang, J.; Luo, Y.; Xing, P.; Lan, B.; Li, M.; et al. Phase I study and biomarker analysis of pyrotinib, a novel irreversible Pan-ERBB receptor tyrosine kinase inhibitor, in patients with human epidermal growth factor receptor 2—positive metastatic breast cancer. J. Clin. Oncol. 2017, 35, 3105–3112. [Google Scholar] [CrossRef]
  119. Ma, F.; Ouyang, Q.; Li, W.; Jiang, Z.; Tong, Z.; Liu, Y.; Li, H.; Yu, S.; Feng, J.; Wang, S.; et al. Pyrotinib or lapatinib combined with capecitabine in HER2–positive metastatic breast cancer with prior taxanes, anthracyclines, and/ or trastuzumab: A randomized, phase II study. J. Clin. Oncol. 2019, 37, 2610–2619. [Google Scholar] [CrossRef]
  120. Lindholm, E.M.; Krohn, M.; Iadevaia, S.; Kristian, A.; Mills, G.B.; Mælandsmo, G.M.; Engebraaten, O. Proteomic characterization of breast cancer xenografts identifies early and late bevacizumab-induced responses and predicts effective drug combinations. Clin. Cancer Res. 2014, 20, 404–412. [Google Scholar] [CrossRef] [Green Version]
  121. Karaman, S.; Leppänen, V.M.; Alitalo, K. Vascular endothelial growth factor signaling in development and disease. Development 2018, 145, dev151019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Voigtlaender, M.; Schneider-Merck, T.; Trepel, M. Lapatinib. Recent Results Cancer Res. 2018, 211, 19–44. [Google Scholar] [CrossRef]
  123. Fares, J.; Kanojia, D.; Rashidi, A.; Ulasov, I.; Lesniak, M.S. Landscape of combination therapy trials in breast cancer brain metastasis. Int J Cancer 2020, 147, 1939–1952. [Google Scholar] [CrossRef]
  124. Amiri-Kordestani, L.; Blumenthal, G.M.; Xu, Q.C.; Zhang, L.; Tang, S.W.; Ha, L.; Weinberg, W.C.; Chi, B.; Candau-Chacon, R.; Hughes, P.; et al. FDA approval: Ado-trastuzumab emtansine for the treatment of patients with HER2-positive metastatic breast cancer. Clin. Cancer Res. 2014, 20, 4436–4441. [Google Scholar] [CrossRef] [Green Version]
  125. Hecht, J.R.; Bang, Y.J.; Qin, S.K.; Chung, H.C.; Xu, J.M.; Park, J.O.; Jeziorski, K.; Shparyk, Y.; Hoff, P.M.; Sobrero, A.; et al. Lapatinib in combination with capecitabine plus oxaliplatin in human epidermal growth factor receptor 2-positive advanced or metastatic gastric, esophageal, or gastroesophageal adenocarcinoma: TRIO-013/LOGiC—A randomized phase III trial. J. Clin. Oncol. 2016, 34, 443–451. [Google Scholar] [CrossRef] [Green Version]
  126. Swain, S.M.; Baselga, J.; Miles, D.; Im, Y.H.; Quah, C.; Lee, L.F.; Cortés, J. Incidence of central nervous system metastases in patients with HER2-positive metastatic breast cancer treated with pertuzumab, trastuzumab, and docetaxel: Results from the randomized phase III study CLEOPATRA. Ann. Oncol. 2014, 25, 1116–1121. [Google Scholar] [CrossRef]
  127. Bartsch, R.; Berghoff, A.S.; Vogl, U.; Rudas, M.; Bergen, E.; Dubsky, P.; Dieckmann, K.; Pinker, K.; Bago-Horvath, Z.; Galid, A.; et al. Activity of T-DM1 in Her2-positive breast cancer brain metastases. Clin. Exp. Metastasis. 2015, 32, 729–737. [Google Scholar] [CrossRef]
  128. Jacot, W.; Pons, E.; Frenel, J.S.; Guiu, S.; Levy, C.; Heudel, P.E.; Bachelot, T.; D’Hondt, V.; Darlix, A.; Firmin, N.; et al. Efficacy and safety of trastuzumab emtansine (T-DM1) in patients with HER2-positive breast cancer with brain metastases. Breast Cancer Res. Treat. 2016, 157, 307–318. [Google Scholar] [CrossRef]
  129. Jerusalem, G.; Park, Y.H.; Yamashita, T.; Hurvitz, S.A.; Chen, S.; Cathcart, J.; Lee, C.; Perrin, C. 138O CNS metastases in HER2-positive metastatic breast cancer treated with trastuzumab deruxtecan: DESTINY-Breast01 subgroup analyses. Ann. Oncol. 2020, 31, S63–S64. [Google Scholar] [CrossRef]
  130. Modi, S. Trastuzumab deruxtecan in previously treated HER2-positive metastatic breast cancer: Plain language summary of the DESTINY-Breast01 study. Futur. Oncol. 2021, 17, 3415–3423. [Google Scholar] [CrossRef]
  131. Modi, S.; Saura, C.; Yamashita, T.; Park, Y.H.; Kim, S.-B.; Tamura, K.; Andre, F.; Iwata, H.; Ito, Y.; Tsurutani, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N. Engl. J. Med. 2020, 382, 610–621. [Google Scholar] [CrossRef]
  132. Murthy, R.K.; Loi, S.; Okines, A.; Paplomata, E.; Hamilton, E.; Hurvitz, S.A.; Lin, N.U.; Borges, V.; Abramson, V.; Anders, C.; et al. Tucatinib, Trastuzumab, and Capecitabine for HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2020, 382, 597–609. [Google Scholar] [CrossRef]
  133. Di Leo, A.; Johnston, S.; Lee, K.S.; Ciruelos, E.; Lønning, P.E.; Janni, W.; O’Regan, R.; Mouret-Reynier, M.A.; Kalev, D.; Egle, D.; et al. Buparlisib plus fulvestrant in postmenopausal women with hormone-receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2018, 19, 87–100. [Google Scholar] [CrossRef]
  134. Nanda, R.; Chow, L.Q.M.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in patients with advanced triple-negative breast cancer: Phase Ib keynote-012 study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef]
  135. Syriac, A.K.; Nandu, N.S.; Leone, J.P. Central Nervous System Metastases from Triple-Negative Breast Cancer: Current Treatments and Future Prospective. Breast Cancer (Dove Med. Press) 2022, 14, 1–13. [Google Scholar] [CrossRef]
  136. Hussain, M.H.A.; Powles, T.; Albers, P.; Castellano, D.; Daneshmand, S.; Gschwend, J.; Nishiyama, H.; Oudard, S.; Tayama, D.; Davarpanah, N.N.; et al. IMvigor010: Primary analysis from a phase III randomized study of adjuvant atezolizumab (atezo) versus observation (obs) in high-risk muscle-invasive urothelial carcinoma (MIUC). J. Clin. Oncol. 2020, 38, 5000. [Google Scholar] [CrossRef]
  137. Régina, A.; Demeule, M.; Ché, C.; Lavallée, I.; Poirier, J.; Gabathuler, R.; Béliveau, R.; Castaigne, J.P. Antitumour activity of ANG1005, a conjugate between paclitaxel and the new brain delivery vector Angiopep-2. Br. J. Pharmacol. 2008, 155, 185–197. [Google Scholar] [CrossRef] [Green Version]
  138. Goetz, M.P.; Toi, M.; Campone, M.; Trédan, O.; Bourayou, N.; Sohn, J.; Park, I.H.; Paluch-Shimon, S.; Huober, J.; Chen, S.C.; et al. MONARCH 3: Abemaciclib as initial therapy for advanced breast cancer. J. Clin. Oncol. 2017, 35, 3638–3646. [Google Scholar] [CrossRef]
  139. Kourie, H.R.; Chaix, M.; Gombos, A.; Aftimos, P.; Awada, A. Pharmacodynamics, pharmacokinetics and clinical efficacy of neratinib in HER2-positive breast cancer and breast cancer with HER2 mutations. Expert Opin. Drug Metab. Toxicol. 2016, 12, 947–957. [Google Scholar] [CrossRef]
  140. Yardley, D.A.; Hart, L.L.; Ward, P.J.; Wright, G.L.; Shastry, M.; Finney, L.; DeBusk, L.M.; Hainsworth, J.D. Cabazitaxel Plus Lapatinib as Therapy for HER2 + Metastatic Breast Cancer with Intracranial Metastases: Results of a Dose-finding Study. Clin. Breast Cancer 2018, 18, E781–E787. [Google Scholar] [CrossRef]
  141. White, J.; Moughan, J.; Kim, I.; Peereboom, D.; De Los Santos, J.; Sperduto, P.; Mehta, M. Abstract OT1-04-02: NRG oncology/RTOG 1119: Phase II randomized study of whole brain radiotherapy with concurrent lapatinib in patients with brain met from HER2-positive breast cancer—A collaborative study of RTOG & KROG (NCT01622868). Cancer Res. 2017, 77, OT1-04-02. [Google Scholar] [CrossRef]
  142. Pistilli, B.; Pluard, T.; Urruticoechea, A.; Farci, D.; Kong, A.; Bachelot, T.; Chan, S.; Han, H.S.; Jerusalem, G.; Urban, P.; et al. Phase II study of buparlisib (BKM120) and trastuzumab in patients with HER2+ locally advanced or metastatic breast cancer resistant to trastuzumab-based therapy. Breast Cancer Res. Treat. 2018, 168, 357–364. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Formation of breast cancer-associated brain tumors.
Figure 1. Formation of breast cancer-associated brain tumors.
Ijms 23 11687 g001
Figure 2. An illustrative image depicting the blood-brain barrier and blood-tumor barrier.
Figure 2. An illustrative image depicting the blood-brain barrier and blood-tumor barrier.
Ijms 23 11687 g002
Figure 3. Molecular interactions of different pathways involved in the regulation of BCBM. The epithelial to mesenchymal transition (EMT) genes inhibit cadherin, promoting metastasis. PI3K/AKT/mTOR and RAS/RAF/ERK pathways activate cellular processes, such as cell proliferation, survival, migration, and angiogenesis. PI3K/AKT/mTOR and JAK/STAT pathways help BCBMs escape immunosurveillance. COX2 aids in prostaglandin synthesis and inflammation activation. Histone deacetylation of growth factors and protein glycosylation aid in bypassing the BBB.
Figure 3. Molecular interactions of different pathways involved in the regulation of BCBM. The epithelial to mesenchymal transition (EMT) genes inhibit cadherin, promoting metastasis. PI3K/AKT/mTOR and RAS/RAF/ERK pathways activate cellular processes, such as cell proliferation, survival, migration, and angiogenesis. PI3K/AKT/mTOR and JAK/STAT pathways help BCBMs escape immunosurveillance. COX2 aids in prostaglandin synthesis and inflammation activation. Histone deacetylation of growth factors and protein glycosylation aid in bypassing the BBB.
Ijms 23 11687 g003
Figure 4. Several signaling cascades involved in BCBM therapeutic targets and their potential drugs.
Figure 4. Several signaling cascades involved in BCBM therapeutic targets and their potential drugs.
Ijms 23 11687 g004
Table 1. Risk factors, identification, and chances of BM in BC patients.
Table 1. Risk factors, identification, and chances of BM in BC patients.
Risk FactorsIdentificationBrain Metastasis RiskReferences
Lymph nodesHistopathologyPositive lymphnodes-4, HR = 2.5, p = 0.029[23]
Tumor gradeHistopathologyThe grade of the tumor is 3; Rate is 7.9% after 10-years completion of follow-up[24]
Tumor sizeHistopathologyThe size of the tumor is 2 cm, after 10 years with a 7% rate. Size of tumor greater than 2 cm have a high risk of BM[25]
Luminal A and B
subtypes
Molecular biologyOver-expresses of epidermal growth factor of a human determined as HER-2[26]
AlphaB-crystallin (CRYAB)Genetic biomarkerOccurrence of BM[27]
Phosphatidylinositol 3-kinase (PI3K) and mitogen-activated
protein kinase (MAPK/ERK)
Molecular biomarkerOverexpression of HER 3 receptor MAPK signaling pathway preferentially
activated in the BM of cancer patients
[23]
VEGF and CXCR-4Molecular biomarkerDisrupt the BBB with migration in the
parenchyma region
[28]
A2,6-sialyltransferaseGenetic driverCancer cell extravasations in the course of the BBB[29]
Adjuvant
trastuzumab trials
Targeted therapy for HER2-positiveBM risk was increased with a range of 1.32 to 1.9[30]
Table 2. Current/clinical trial drugs and their mechanism of action against brain metastatic disease in BC.
Table 2. Current/clinical trial drugs and their mechanism of action against brain metastatic disease in BC.
Target DrugTreatment TypeTumor TypeMode of ActionReferences
EP versus ICChemotherapyBCBM subgroupTopoisomerase-I inhibitor-polymer conjugate[136]
Ang-1005ChemotherapyBCBM with
leptomeningeal
carcinomatosis
Peptide-paclitaxel
conjugate
[137]
AbemaciclibTargeted therapyHER2-BCCDK 4 and 6 inhibitor[138]
Neratinib
monotherapy
Targeted therapyHER2+ progression within CNSPan-HER inhibitor[139]
Cabazitaxel +
lapatinib
ChemotherapyHER2+ progressionHER2 positive[140]
Lapatinib + WBRTChemotherapyEffective therapeutic EGFR family targetHER2[141]
BKM120 +
capecitabine
Triple-negativeCapability toward penetration to the BBBPI3K[142]
PertuzumabSystemic therapyHER2+ BCBMMonoclonal antibody[93,126]
Everolimus and
Buparlisib
BCBMmTOR and PI3K
inhibitors
[133,38]
Trastuzumab
emtansine
HER2+ BCBMMonoclonal antibody[127,128]
Trastuzumab
deruxtecan
Systemic therapyBCBM HER2+ metastatic breast cancerTopoisomerase I
inhibitor
[129,130,131]
Trastuzumab, capecitabine, with tucatinibTargeted therapyHER2+ BCBMTyrosine kinase inhibitor[99,132]
PembrolizumabHigh affinity and great
selectivity
Advance TNBCMonoclonal IgG4-
antibody
[32,134]
Stereotactic
radiosurgery (SRS) and atezolizumab
TNB with BCBM [32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaleem, M.; Dalhat, M.H.; Azmi, L.; Asar, T.O.; Ahmad, W.; Alghanmi, M.; Almostadi, A.; Zughaibi, T.A.; Tabrez, S. An Insight into Molecular Targets of Breast Cancer Brain Metastasis. Int. J. Mol. Sci. 2022, 23, 11687. https://doi.org/10.3390/ijms231911687

AMA Style

Kaleem M, Dalhat MH, Azmi L, Asar TO, Ahmad W, Alghanmi M, Almostadi A, Zughaibi TA, Tabrez S. An Insight into Molecular Targets of Breast Cancer Brain Metastasis. International Journal of Molecular Sciences. 2022; 23(19):11687. https://doi.org/10.3390/ijms231911687

Chicago/Turabian Style

Kaleem, Mohammed, Mahmood Hassan Dalhat, Lubna Azmi, Turky Omar Asar, Wasim Ahmad, Maimonah Alghanmi, Amal Almostadi, Torki A. Zughaibi, and Shams Tabrez. 2022. "An Insight into Molecular Targets of Breast Cancer Brain Metastasis" International Journal of Molecular Sciences 23, no. 19: 11687. https://doi.org/10.3390/ijms231911687

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop