[HTML][HTML] Molecular profiling reveals a hypoxia signature in breast implant-associated anaplastic large cell lymphoma

N Oishi, T Hundal, JL Phillips, S Dasari, G Hu… - …, 2021 - ncbi.nlm.nih.gov
N Oishi, T Hundal, JL Phillips, S Dasari, G Hu, DS Viswanatha, R He, M Mai, HK Jacobs…
Haematologica, 2021ncbi.nlm.nih.gov
Breast implant-associated anaplastic large cell lymphoma (BIAALCL) is a recently
characterized T-cell malignancy that has raised significant patient safety concerns and led to
worldwide impact on the implants used and clinical management of patients undergoing
reconstructive or cosmetic breast surgery. Molecular signatures distinguishing BIA-ALCL
from other anaplastic large cell lymphomas have not been fully elucidated and classification
of BIA-ALCL as a World Health Organization entity remains provisional. We performed RNA …
Abstract
Breast implant-associated anaplastic large cell lymphoma (BIAALCL) is a recently characterized T-cell malignancy that has raised significant patient safety concerns and led to worldwide impact on the implants used and clinical management of patients undergoing reconstructive or cosmetic breast surgery. Molecular signatures distinguishing BIA-ALCL from other anaplastic large cell lymphomas have not been fully elucidated and classification of BIA-ALCL as a World Health Organization entity remains provisional. We performed RNA sequencing and gene set enrichment analysis comparing BIA-ALCL to non-BIAALCL and identified dramatic upregulation of hypoxia signaling genes including the hypoxia-associated biomarker CA9 (carbonic anyhydrase-9). Immunohistochemistry validated CA9 expression in all BIA-ALCL, with only minimal expression in non-BIA-ALCL. Growth induction in BIA-ALCL-derived cell lines cultured under hypoxic conditions was proportional to upregulation of CA9 expression, and RNA sequencing demonstrated induction of the same gene signature observed in BIAALCL tissue samples compared to non-BIA-ALCL. CA9 silencing blocked hypoxia-induced BIA-ALCL cell growth and cell cycle-associated gene expression, whereas CA9 overexpression in BIA-ALCL cells promoted growth in a xenograft mouse model. Furthermore, CA9 was secreted into BIA-ALCL cell line supernatants and was markedly elevated in human BIA-ALCL seroma samples. Finally, serum CA9 concentrations in mice bearing BIA-ALCL xenografts were significantly elevated compared to those in control serum. Together, these findings characterize BIA-ALCL as a hypoxia-associated neoplasm, likely attributable to the unique microenvironment in which it arises. These data support classification of BIA-ALCL as a distinct entity and uncover opportunities for investigating hypoxia-related proteins such as CA9 as novel biomarkers and therapeutic targets in this disease.
ncbi.nlm.nih.gov