Breast Cancer and SLC7A11: A Comprehensive Analysis and Future Prospects

Significance 

Breast cancer is the leading malignancy in women, it is a heterogeneous disease manifesting in various biological subtypes. These subtypes exhibit distinct morphological, biological, and clinical behaviors, influencing their response to treatment. Central to cancer progression and therapeutic failure is metabolic reprogramming, where tumors adapt their metabolism to meet the demands of rapid growth and proliferation. This altered metabolism, especially in the context of glutamine metabolism, is critical for cancer cell survival and proliferation. The sodium-independent, chloride-dependent anionic L-cystine/L-glutamate antiporter Solute Carrier Family 7 Member 11 (SLC7A11) has emerged as a key player in this process. SLC7A11’s role in facilitating cystine uptake in exchange for glutamate, and its subsequent contribution to the synthesis of glutathione, a major antioxidant, makes it a potential target for cancer therapy, particularly in breast cancer.

Recent studies have shown a significant association between high SLC7A11 mRNA and protein expression and poor prognostic factors in breast cancer. Intriguingly, SLC7A11 expression varies across breast cancer subtypes, with notable increases in basal-like/triple-negative (TN) and luminal B tumors. This differential expression suggests a subtype-specific role of SLC7A11 in breast cancer progression. Furthermore, the study of SLC7A11’s DNA methylation patterns and its association with immune cell infiltrates offers new insights into its potential role in cancer metabolism and tumor immunity.

In a new study published in  Journal Cancer Biology & Therapy led by Professor Andrew  Green  and conducted by Preyanka Natha, Lutfi  Alfarsi, Rokaya El-Ansari, Brendah  Masisi, Busra Erkan, Ali Fakroun, Ian   Ellis, Emad  Rakha & from the School of Medicine at University of Nottingham , the researchers conducted a series of comprehensive experiments to analyze the role of SLC7A11 in breast cancer. The team used the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1,980) and Breast Cancer Gene Expression Miner (n = 4,712) databases to assess SLC7A11 gene copy number and mRNA expression levels. They found high SLC7A11 mRNA expression was significantly associated with high tumor grades. SLC7A11 copy number gain was particularly noted in HER2+ tumors, suggesting a subtype-specific pattern. They also evaluated protein levels of SLC7A11 using immunohistochemistry in a large BC cohort (n = 1,981) and found high SLC7A11 protein expression was predominantly observed in Estrogen Receptor (ER)-negative and Triple Negative (TN) BC subtypes, indicating its potential role in these aggressive cancer forms.

The researchers used the Cancer Genome Atlas (TCGA) dataset to study SLC7A11 DNA methylation patterns and found specific CpG islands within SLC7A11 showed high levels of methylation, hinting at a potential epigenetic regulation mechanism in breast cancer. When the authors examined the SLC7A11 mRNA expression’s relationship with immune infiltrates using the TIMER dataset (n = 1,100), they found a positive association was observed between SLC7A11 gene expression and neutrophil and macrophage infiltration, suggesting a potential link between SLC7A11 and tumor immunity. Moreover, the correlation between SLC7A11 and other amino acid transporters and enzymes involved in glutamine metabolism was explored where they found SLC7A11 to be positively correlated with other amino acid transporters and enzymes associated with glutamine metabolism, indicating a coordinated role in cancer cell metabolic regulation.

The study also evaluated the relationship between SLC7A11 expression and patient outcomes. High SLC7A11 mRNA and protein expressions were associated with poorer patient outcomes, particularly in terms of breast cancer specific survival. These experiments collectively suggest that SLC7A11 plays a significant role in BC metabolism, showing differential expression across subtypes and associations with poor patient outcomes. The study underscores the importance of metabolic reprogramming in cancer progression and highlights SLC7A11 as a potential therapeutic target, especially in aggressive breast cancer subtypes.

The study findings indicates that SLC7A11 copy number gain is significantly associated with HER2+ breast cancers, while high mRNA and protein expressions are observed in more aggressive subtypes like TN and ER-negative cancers. This association suggests a potential role of SLC7A11 in these subtypes’ aggressive behavior and poor prognosis. Additionally, SLC7A11 is positively correlated with other amino acid transporters and enzymes involved in glutamine metabolism, underlining a coordinated role in metabolic regulation. A significant finding of the study is the positive association of SLC7A11 gene expression with neutrophil and macrophage infiltration, indicating a possible link between SLC7A11 and tumor immunity. This correlation suggests that SLC7A11 could influence the tumor microenvironment, potentially affecting cancer progression and response to therapy.

The authors demonstrated SLC7A11 as a significant factor in breast cancer metabolism, with differential expression across subtypes and associations with poor patient outcomes. Its role in the altered metabolic needs of tumors, particularly in aggressive breast cancer subtypes, makes it a promising candidate for targeted therapy. Further functional studies are needed to unravel the precise mechanisms by which SLC7A11 contributes to breast cancer progression and to explore its potential as a therapeutic target. In conclusion, the comprehensive analysis of SLC7A11 in breast cancer subtypes presents new avenues for understanding the metabolic heterogeneity of this disease. Its significant associations with poor prognostic factors and immune cell infiltration shed light on its potential role in breast cancer progression. As a potential therapeutic target, SLC7A11 offers a promising avenue for developing novel treatment strategies, particularly for aggressive breast cancer subtypes where current treatment options are limited. Further research into the functional mechanisms of SLC7A11 in breast cancer will be pivotal in translating these findings into clinical practice.

Breast Cancer and SLC7A11: A Comprehensive Analysis and Future Prospects - Medicine Innovates
Image Credit: Cancer Biol Ther. 2024 Dec 31;25(1):2291855. Figure showcase SLC7A11 protein expression in representative invasive breast cancer tissue microarray cores determined using immunohistochemistry showing a) high, and b) negative expression.

About the author

Andrew R Green

Associate Professor, Faculty of Medicine & Health Sciences
Nottingham University

My main research expertise is within breast cancer molecular pathology using techniques such as immunohistochemistry, tissue microarrays (TMAs), laser microdissection and gene expression profiling,

Reference

Nath P, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol Ther. 2024;25(1):2291855. doi: 10.1080/15384047.2023.2291855.

Go To Cancer Biol Ther.