Your browser doesn't support javascript.
loading
E3 ubiquitin ligase ASB8 promotes selinexor-induced proteasomal degradation of XPO1.
Kwanten, Bert; Deconick, Tine; Walker, Christopher; Wang, Feng; Landesman, Yosef; Daelemans, Dirk.
Afiliação
  • Kwanten B; KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy (Rega Institute), Leuven, Belgium.
  • Deconick T; KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy (Rega Institute), Leuven, Belgium.
  • Walker C; Karyopharm Therapeutics, Newton, MA 02459, USA.
  • Wang F; Karyopharm Therapeutics, Newton, MA 02459, USA.
  • Landesman Y; Karyopharm Therapeutics, Newton, MA 02459, USA.
  • Daelemans D; KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy (Rega Institute), Leuven, Belgium. Electronic address: dirk.daelemans@kuleuven.be.
Biomed Pharmacother ; 160: 114305, 2023 Apr.
Article em En | MEDLINE | ID: mdl-36731340
ABSTRACT
Selinexor (KPT-330), a small-molecule inhibitor of exportin-1 (XPO1, CRM1) with potent anticancer activity, has recently been granted FDA approval for treatment of relapsed/refractory multiple myeloma and diffuse large B-cell lymphoma (DLBCL), with a number of additional indications currently under clinical investigation. Since selinexor has often demonstrated synergy when used in combination with other drugs, notably bortezomib and dexamethasone, a more comprehensive approach to uncover new beneficial interactions would be of great value. Moreover, stratifying patients, personalizing therapeutics and improving clinical outcomes requires a better understanding of the genetic vulnerabilities and resistance mechanisms underlying drug response. Here, we used CRISPR-Cas9 loss-of-function chemogenetic screening to identify drug-gene interactions with selinexor in chronic myeloid leukemia, multiple myeloma and DLBCL cell lines. We identified the TGFß-SMAD4 pathway as an important mediator of resistance to selinexor in multiple myeloma cells. Moreover, higher activity of this pathway correlated with prolonged progression-free survival in multiple myeloma patients treated with selinexor, indicating that the TGFß-SMAD4 pathway is a potential biomarker predictive of therapeutic outcome. In addition, we identified ASB8 (ankyrin repeat and SOCS box containing 8) as a shared modulator of selinexor sensitivity across all tested cancer types, with both ASB8 knockout and overexpression resulting in selinexor hypersensitivity. Mechanistically, we showed that ASB8 promotes selinexor-induced proteasomal degradation of XPO1. This study provides insight into the genetic factors that influence response to selinexor treatment and could support both the development of predictive biomarkers as well as new drug combinations.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfoma Difuso de Grandes Células B / Mieloma Múltiplo Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Biomed Pharmacother Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfoma Difuso de Grandes Células B / Mieloma Múltiplo Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Biomed Pharmacother Ano de publicação: 2023 Tipo de documento: Article