1. Academic Validation
  2. Paralog-specific signaling by IRAK1/4 maintains MyD88-independent functions in MDS/AML

Paralog-specific signaling by IRAK1/4 maintains MyD88-independent functions in MDS/AML

  • Blood. 2023 May 12;blood.2022018718. doi: 10.1182/blood.2022018718.
Joshua Ruina Bennett 1 Chiharu Ishikawa 1 Puneet Agarwal 1 Jennifer Yeung 1 Avery Sampson 1 Emma Uible 1 Eric Vick 2 Lyndsey Bolanos 3 Kathleen M Hueneman 1 Mark Wunderlich 1 Amal Kolt 4 Kwangmin Choi 1 Andrew G Volk 1 Kenneth D Greis 2 Jan S Rosenbaum 4 Scott Hoyt 5 Craig J Thomas 6 Daniel T Starczynowski 1
Affiliations

Affiliations

  • 1 Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States.
  • 2 University of Cincinnati, Cincinnati, Ohio, United States.
  • 3 Cincinnati Children's Hospital Medical Center, Cincinnati, USA, Cincinnati, Ohio, United States.
  • 4 Kurome Therapeutics, Cincinnati, Ohio, United States.
  • 5 National Institutes of Health, Rockville, Maryland, United States.
  • 6 National Institutes of Health, Bethesda, Maryland, United States.
Abstract

Dysregulation of innate immune signaling is a hallmark of hematologic malignancies. Recent therapeutic efforts to subvert aberrant innate immune signaling in MDS and AML have focused on the kinase IRAK4. IRAK4 inhibitors have achieved promising, though moderate, responses in pre-clinical studies and in clinical trials for MDS and AML. The reasons underlying the limited responses to IRAK4 inhibitors remain unknown. Here, we reveal that inhibiting IRAK4 in leukemic cells elicits functional complementation and compensation by its paralog, IRAK1. Using genetic approaches, we demonstrate that co-targeting IRAK1 and IRAK4 is required to suppress leukemic stem/progenitor cell (LSPC) function and induce differentiation in cell lines and patient-derived cells. While IRAK1 and IRAK4 are presumed to function primarily downstream of the proximal adapter MyD88, we found that complimentary and compensatory IRAK1 and IRAK4 dependencies in MDS/AML occur via non-canonical MyD88-independent pathways. Genomic and proteomic analyses revealed that IRAK1 and IRAK4 preserve the undifferentiated state of MDS/AML LSPCs by coordinating a network of pathways, including ones that converge on the PRC2 complex and JAK-STAT signaling. To translate these findings, we implemented a structure-based design of a potent and selective dual IRAK1 and IRAK4 Inhibitor KME-2780. MDS/AML cell lines and patient-derived samples showed significant suppression of LSPCs in vitro and in xenograft studies when treated with KME-2780 as compared to selective IRAK4 inhibitors. Our results provide a mechanistic basis and rationale for co-targeting IRAK1 and IRAK4 for the treatment of cancers, including MDS/AML.

Figures
Products