1. Academic Validation
  2. Bifunctional inhibitor reveals NEK2 as a therapeutic target and regulator of oncogenic pathways in lymphoma

Bifunctional inhibitor reveals NEK2 as a therapeutic target and regulator of oncogenic pathways in lymphoma

  • Mol Cancer Ther. 2023 Oct 11. doi: 10.1158/1535-7163.MCT-23-0299.
Mason McCrury 1 Kennith Swafford 1 Sydnye L Shuttleworth 1 Syed Hassan Mehdi 1 Baku Acharya 1 Debasmita Saha 1 Kevin Naceanceno 1 Stephanie D Byrum 2 Aaron J Storey 2 Ying-Zhi Xu 1 Claire Doshier 1 Vijay Patel 1 Ginell R Post 1 Annick De Loose 3 Analiz Rodriguez 3 Leonard D Shultz 4 Fenghuang Zhan 3 Donghoon Yoon 1 Brendan Frett 5 Samantha Kendrick 1
Affiliations

Affiliations

  • 1 University of Arkansas for Medical Sciences, Little Rock, AR, United States.
  • 2 U of Arkansas for Medical Sciences, Little Rock, AR, United States.
  • 3 University of Arkansas for Medical Sciences, Little Rock, United States.
  • 4 Jackson Laboratory, United States.
  • 5 University of Arizona, United States.
Abstract

Expression of the serine/threonine kinase NEK2 is essential for entry into mitosis via its role in facilitating centrosome separation. Its overactivity can lead to tumorigenesis and drug resistance through the activation of several oncogenic pathways including Akt. While the cancer-enabling activities of NEK2 are documented in many malignancies, including correlations with poor survival in myeloma, breast, and non-small cell lung Cancer, little is known about the role of NEK2 in lymphoma. Here, in tumors from patients with diffuse large B cell lymphoma (DLBCL), the most common, aggressive non-Hodgkin lymphoma, we found a high abundance of NEK2 mRNA and protein associated with an inferior overall survival. Using our recently developed NEK2 inhibitor, NBI-961, we discovered DLBCL cell lines and patient-derived cells exhibit a dependency on NEK2 for their viability. This compromised cell fitness was directly attributable to efficient NEK2 inhibition and proteasomal degradation by NBI-961. In a subset of particularly sensitive DLBCL cells, NBI-961 induced G2/mitosis arrest and Apoptosis. In contrast, an existing indirect NEK2 inhibitor, INH154, did not prevent NEK2 autophosphorylation, induce NEK2 proteasomal degradation, or affect cell viability. Global proteomics and phospho-proteomics revealed that NEK2 orchestrates cell-cycle and apoptotic pathways through regulation of both known and new signaling molecules. We show the loss of NEK2 sensitized DLBCL to the chemotherapy agents doxorubicin and vincristine, and effectively suppressed tumor growth in mice. These studies establish the oncogenic activity of NEK2 in DLBCL and set the foundation for development of anti-NEK2 therapeutic strategies in this frequently refractory and relapse-prone Cancer.

Figures
Products