bims-midomi Biomed News
on MDM2 and mitochondria
Issue of 2025–09–14
two papers selected by
Gavin McStay, Liverpool John Moores University



  1. Bioorg Med Chem Lett. 2025 Sep 04. pii: S0960-894X(25)00303-8. [Epub ahead of print]129 130394
      Murine double minute 2 (MDM2, also known as human double minute 2 or HDM2) is a negative regulator of the tumor suppressor protein p53 and is overexpressed in many cancers. Over the past two decades, substantial progress has been made in developing inhibitors of the MDM2-p53 interaction, thereby allowing the p53 protein to exert antitumor effects through cell apoptosis and cycle arrest. While there are currently no FDA-approved MDM2 inhibitors available, several small molecule MDM2 inhibitors and a stapled peptide inhibitor of the MDM2-p53 interaction are in clinical development. Availability of these clinical candidates, representing a diverse array of chemical scaffolds that bind to the same p53 binding site on the MDM2 protein with low nanomolar inhibition potency (IC50), presents a significant opportunity for developing molecular imaging probes for MDM2 in parallel. This review summarizes the MDM2 inhibitors that have been evaluated in clinical trials, which could serve as starting leads for imaging probe development, and recent progress in developing radiotracers for MDM2 and for evaluating MDM2 expression levels in tumors noninvasively using the highly sensitive, molecular imaging techniques positron emission tomography (PET) or single-photon emission computed tomography (SPECT).
    Keywords:  MDM2; MDM2 inhibitor; Molecular imaging; PET; SPECT; cancer; p53
    DOI:  https://doi.org/10.1016/j.bmcl.2025.130394
  2. Int J Mol Sci. 2025 Sep 05. pii: 8648. [Epub ahead of print]26(17):
      Acute lymphoblastic leukemia (ALL) remains a formidable therapeutic challenge, particularly within high-risk cohorts. Advances in next-generation sequencing have elucidated critical mutations that significantly influence prognosis and therapeutic decision-making. Tyrosine kinase inhibitors (TKIs) have significantly improved treatment outcomes in Philadelphia chromosome-positive (Ph+) ALL. Meanwhile, emerging therapies such as monoclonal antibodies and chimeric antigen receptor (CAR) T-cell therapies show promise for B-cell ALL, although they are associated with considerable toxicities. These developments underscore the persistent need for alternative therapeutic strategies that can benefit a wider range of patients. In this study, human ALL cell lines-characterized by either wild-type or mutant tumor protein p53 (TP53) status-were treated with RG7388 (an MDM2 (mouse double minute 2 homolog) inhibitor) and BBI608 (a STAT3 (signal transducer and activator of transcription 3) inhibitor), both as single agents and in combination. Cell viability was quantified using XTT assays, while apoptosis was assessed via flow cytometry. Additionally, immunoblotting and qRT-PCR were employed to evaluate changes in protein and gene expression, respectively. RG7388 demonstrated potent growth inhibition in the majority of ALL cell lines, with p53-mutant cell lines exhibiting resistance. BBI608 reduced cell viability across all tested cell lines, though with variable sensitivity. Notably, the combination of RG7388 and BBI608 elicited synergistic anti-proliferative effects in p53 wild-type and partially functional p53-mutant cells, enhancing apoptosis and stabilizing p53 protein levels. In contrast, MOLT-4 cells, which harbor concurrent TP53 and STAT3 mutations, did not benefit from the combination treatment, indicating an inherent resistance phenotype within this subset. Collectively, these findings highlight the therapeutic potential of combined MDM2 and STAT3 inhibition in ALL, particularly in p53 wild-type and partially functional p53-mutant contexts. This combinatorial approach augments apoptosis and tumor growth suppression, offering a promising avenue for expanding treatment options for a broader patient population. Further investigation is warranted to validate these preclinical findings and to explore translational implications in genetically diverse ALL subsets.
    Keywords:  BBI608 (napabucasin); MDM2; RG7388 (idasanutlin); STAT3; acute lymphoblastic leukemia (ALL); targeted cancer therapies
    DOI:  https://doi.org/10.3390/ijms26178648