bims-p53act Biomed News
on p53 mutations and anti-cancer therapy response
Issue of 2025–11–30
nine papers selected by
Toni Martínez Bernabé, Universitat de les Illes Balears



  1. bioRxiv. 2025 Oct 09. pii: 2025.10.06.680794. [Epub ahead of print]
      Loss of the normal tumor suppressive functions of p53, the "guardian of the genome," is common in advanced cancers. Mutations in TP53 occur in various ways which can result in p53 null tumor cells as well as in tumor cells highly expressing missense, gain of oncogenic function p53 proteins. Gain of function p53 proteins not only lose wild type p53 pro-apoptotic and DNA repair functions, but gain novel oncogenic functions. We hypothesize that directly targeting missense mutant p53 instead of downstream pathways has substantial advantages. Analogues of curcumin have been reported to bind mutant p53 to accomplish this goal. In this report, we developed a series of novel molecules derived from the curcumin backbone. We identified a particularly active curcumin analogue, AKT-100, and demonstrate binding to mutant p53 proteins to reinstate p53 wild type functionality. This agent exhibits impressive cell killing in multiple ovarian and serous endometrial cancer cells at concentrations ranging from 100-300 nM. AKT-100 is also synergistic with the PARP inhibitor olaparib and with chemotherapy by inhibiting alternative DNA repair mechanisms associated with resistance to standard therapies. RNA sequencing confirms that AKT-100 reactivates wild type p53-driven expression of genes associated with normal cell cycle regulation (induction of CDKN1A encoding p21 and GADD45A ), apoptosis (induction of PMAIP1 encoding Noxa and DR5 encoding Death Receptor 5), and inhibits DNA replication and repair in cancer cells. Thus, p53 reactivators under development, such as AKT-100, hold promise as novel therapeutic agents to directly target missense mutant, gain of oncogenic function p53.
    DOI:  https://doi.org/10.1101/2025.10.06.680794
  2. Int J Mol Sci. 2025 Nov 18. pii: 11134. [Epub ahead of print]26(22):
      Colorectal cancer (CRC) remains one of the most lethal malignancies in the United States, with African American (AA) patients experiencing disproportionately higher incidence and mortality compared to Caucasian Americans (CAs). These disparities have been linked to tumor-intrinsic genomic differences, including microsatellite instability (MSI) and p53 mutation status, which may influence chemotherapeutic response, particularly to the standard-of-care agent 5-fluorouracil (5-FU). However, mechanistic insights have been limited by the lack of racially diverse preclinical models. Here, we evaluated the efficacy of F10 (a novel fluoropyrimidine polymer) vs. 5-FU using AA- and CA-derived CRC cell lines with distinct MSI and p53 profiles. MTT assays revealed that MSI status, more than racial origin, predicted 5-FU sensitivity. Transcriptomics uncovered distinct gene expression patterns associated with MSI status and racial background, particularly in drug metabolism pathways. F10 demonstrated superior potency and consistency vs. 5-FU across all cell lines, independent of race, MSI, or p53 status. Additionally, in silico docking and immunofluorescence suggest that the dietary triterpene lupeol enhances F10 efficacy, perhaps through stabilization of the Fas apoptosis pathway. These findings underscore the therapeutic potential of F10 and the importance of integrating diverse tumor models with dietary adjuvants to inform more effective and inclusive CRC treatment strategies.
    Keywords:  5-fluorouracil; colorectal cancer; fluoropyrimidine; lupeol; racial health disparity
    DOI:  https://doi.org/10.3390/ijms262211134
  3. bioRxiv. 2025 Nov 09. pii: 2025.11.06.687028. [Epub ahead of print]
      The tumor suppressor TP53 gene (p53) is mutated in most human malignancies; however, existing treatment options are largely ineffective, lack selectivity, and cause toxic side effects. To address these clinical problems, we developed a sequential triple-drug strategy for p53 mutant cancer cells. Here we show that a combination of a thymidine analogue (TAS102) plus PARP inhibitor (PARPi) promotes formation of DNA double-strand breaks (DSBs) and G2-arrest specifically in p53 mutant cancer cells. Transcriptome analysis revealed that TAS102-PARPi treatment of p53 mutant cells did not repress DNA replication but activated DSB repair and blocked the mitotic program, consistent with G2-arrest. In contrast, TAS102-PARPi treatment of normal p53 wild-type cells resulted in a temporal G1-arrest and rapid recovery of cell cycle capacity after drug withdrawal. In p53 mutant cancer cells, subsequent blocking of a G2-checkpoint kinase, such as WEE1, released these G2-arrested cells into mitosis, leading to massive cell death. Delayed administration of a G2-kinase inhibitor provides time for p53 wild-type cells to repair DNA, thereby minimizing toxicity to normal tissues. This sequential triple-drug strategy exhibited robust efficacy in preclinical models of colorectal and pancreatic cancers and was well tolerated in mice. Together, our findings illustrate a promising triple-drug strategy for targeting p53 mutant malignancies.
    DOI:  https://doi.org/10.1101/2025.11.06.687028
  4. bioRxiv. 2025 Nov 04. pii: 2025.11.02.686136. [Epub ahead of print]
      Glucosylceramide synthase (GCS) catalyzes ceramide glycosylation in response to cell stress that produces glucosylceramide and other glycosphingolipids. GCS overexpression is a cause of drug resistance and enriches cancer stem cells (CSCs) during cancer chemotherapy. Previous studies showed that GCS modulates expression of p53 mutants and oncogenic gain-of-function (GOF) in heterozygous knock-in cell models ( TP53 R273H -/+ ). However, it is unclear whether GCS can modulate the effects of homozygous p53 mutations, which are common in many cancer cases. We report herewith that inhibition of GCS, via UGCG-knockout and using new inhibitor (Genz-161), effectively re-sensitizes drug resistance and diminishes CSCs in colon cancer cells carrying the homozygous p53 R273H mutation. In aggressive WiDr cells carrying TP53 R273H mutation, knockout of UGCG gene using CRISPR/Cas9 editing or inhibition of GCS with Genz-161 sensitized cancer cells to oxaliplatin, irinotecan and paclitaxel. With decreased ceramide glycosylation in lipidomic profiling, both UGCG-knockout and Genz-161 treatments substantially decreased wound healing, and diminished CSCs and tumor growth under chemotherapy. Interestingly, inhibition of RNA m 6 A methylation by neplanocin A reactivated p53 function and reversed drug resistance. Mechanistic investigation revealed that GCS inhibition downregulated METTL3 expression and repressed RNA-m 6 A modification on mutant p53 R273H effects. Altogether, our findings demonstrate that ceramide glycosylation promotes METTL3 expression and RNA m 6 A methylation in response to drug-induced stress, thereby promoting mutant p53 expression and associated GOF. Conversely, inhibition of GCS can diminish CSCs and drug resistance via reduction of m 6 A modification and reactivation of p53 function. GCS inhibition is an achievable approach for mutant cancer treatment.
    DOI:  https://doi.org/10.1101/2025.11.02.686136
  5. bioRxiv. 2025 Nov 05. pii: 2025.10.27.684888. [Epub ahead of print]
      The BCL2 inhibitor venetoclax has therapeutic activity in several hematological malignancies. In acute myeloid leukemia (AML), venetoclax combined with hypomethylating agents is the standard of care for patients unfit for intensive chemotherapy, but intrinsic and acquired resistance are common. Loss of p53 function is strongly associated with venetoclax resistance, and adding venetoclax to 5-azacitidine provides no overall survival benefit in TP53 -mutant AML. Other frequent mechanisms of venetoclax resistance in AML include FLT3 mutations, MCL-1 upregulation, and altered mitochondrial metabolism. Unfortunately, it has been challenging to develop agents that target these mechanisms directly and combinatorially. Here we report that pitavastatin, an inhibitor of HMG-CoA-reductase, promotes apoptosis and overcomes several venetoclax resistance mechanisms in human AML cells. At clinically achievable concentrations, pitavastatin treatment has potent cytotoxic activity in cells with mutations in TP53 or FLT3 . The apoptotic mechanism involves p53-independent PUMA upregulation and reduced MCL-1 expression. Pitavastatin also suppresses mitochondrial gene expression and oxidative metabolism. The pro-apoptotic actions of pitavastatin depend on depletion of geranylgeranyl pyrophosphate (GGPP) and can be recapitulated by inhibiting GGPP synthase or geranylgeranyltransferase-1 enzymes. These results provide a mechanistic rationale for adding pitavastatin to AML regimens to prevent or overcome venetoclax resistance.
    DOI:  https://doi.org/10.1101/2025.10.27.684888
  6. Adv Protein Chem Struct Biol. 2025 ;pii: S1876-1623(25)00069-0. [Epub ahead of print]148 179-228
      Cancer appears to be a significant global public health concern and most prevalent leading cause of death worldwide. Delays in the diagnosis and treatment may lead to an increase in the prevalence of advanced-stage disease and death. Therefore, creating innovative diagnostic instruments and treatments that demonstrate high effectiveness is imperative. The majority of malignancies have dysfunction in the p53 pathway. In addition, p53 becomes dysfunctional and tends to undergo misfolding and aggregation, resulting in the creation of amyloid aggregates. Efforts are underway to investigate methods for reinstating the regular functioning and manifestation of p53. In this study, we have investigated Heat shock proteins (HSPs), which are molecular chaperones that play a significant role in various cellular processes such as intercellular transportation, formation or disintegration of complex protein, stabilisation or degradation of aggregated or misfolded proteins and protein folding. HSP40, also known as JDPs, are distinguished by their highly conserved J-domains. These domains facilitate the ability to bind to HSP70 and as a co-chaperone enhance the activity of ATPase. Emerging evidence indicates that HSP70/JDPs can influence the levels and/or functions of both wild-type and mutant p53. Only a small number of HSP40/JDPs, including C7, C2, B9, B1, A3, and DNAJA1, have been seen to influence the functions of both WT-p53 and Mut-p53. However, out of the sixteen members, only these handful are implicated in the advancement of cancer. Therefore, studying other HSP40/JDPs that are involved in the advancement of cancer and the activities of p53 (both mutant and wild type), together with their related processes, would enhance our understanding of how cancer progresses, we might potentially speed up the development of innovative treatments for cancer. It is expected that pharmacological molecules and their analogues that specifically target p53 aggregation might be utilised with other anticancer drugs to address the issue of p53 aggregation.
    Keywords:  Cancer; Chaperons; Heat shock proteins; Misfolding; Mutant p53; Mutations; Wild type p53
    DOI:  https://doi.org/10.1016/bs.apcsb.2025.08.011
  7. Adv Protein Chem Struct Biol. 2025 ;pii: S1876-1623(25)00073-2. [Epub ahead of print]148 255-298
      The concept of tumors as prion-like diseases similar to neurodegenerative disorders has gained attraction in recent years. p53, the most well-known tumor suppressor, has been extensively studied for its expression, mutations, and functions in various cancers. Recent findings reveal that p53 undergoes prion-like aggregation in tumors, leading to pathological amyloid fibril formation, functional alterations, and tumor progression. The mechanisms of p53 aggregation involve mutations, structural domains, isoforms, and external factors such as Zn²+ concentrations, pH, temperature, and chaperone abnormalities. While the role of p53 aggregation in tumors is increasingly recognized, controversies remain regarding its precise pathogenic mechanisms. This chapter reviews the structural features of p53 amyloid fibrils, its aggregation characteristics and effects, and the molecular mechanisms driving this phenomenon. Additionally, this chapter summarizes current therapeutic approaches targeting p53 aggregation and prion-like behavior, including small molecules and peptides designed to inhibit aggregation and restore p53's tumor suppressive function. By illuminating these aspects, this chapter aims to deepen our comprehension of how p53 aggregation disrupts its physiological functions. It also highlights the potential of targeting these aggregates as a novel therapeutic strategy in cancer treatment.
    Keywords:  Amyloid fibrils; P53 Aggregation; P53 Mutations; P53 Structural dynamics; Peptide-based p53 therapies; Protein misfolding
    DOI:  https://doi.org/10.1016/bs.apcsb.2025.08.015
  8. Front Immunol. 2025 ;16 1684089
       Objective: To investigate the impact of co-mutations of EGFR with TP53 or KRAS on the prognosis of non-small cell lung cancer (NSCLC) patients, and the efficacy of platinum-based doublet chemotherapy plus immunotherapy after EGFR-TKI resistance.
    Methods: This was a retrospective study that included 168 patients with locally advanced or advanced NSCLC who had next-generation sequencing (NGS) performed at our institution between January 1, 2021, and October 31, 2023. Based on their genomic profiles, patients were categorized into three groups: EGFR single mutation, EGFR/TP53 co-mutation, and EGFR/KRAS co-mutation. Baseline clinical data were collected, including gender, age, smoking history, histological subtype, clinical stage, ECOG performance status, gene testing results, and treatment regimens. All patients were treated with EGFR tyrosine kinase inhibitors (TKIs) as first-line therapy, including first-, second-, or third-generation agents. Upon disease progression, patients received platinum-based doublet chemotherapy plus immunotherapy as second-line treatment. The primary endpoint was progression-free survival (PFS). Survival curves were generated using the Kaplan-Meier method and compared by log-rank test. Baseline characteristics among the three groups were compared using the chi-square test. Multivariate Cox regression analysis was performed to evaluate independent prognostic factors for PFS by incorporating all baseline clinical variables and gene mutation status into the model.
    Results: A total of 168 patients were included in the analysis: 36 with EGFR single mutation, 80 with EGFR/TP53 co-mutation, and 52 with EGFR/KRAS co-mutation. There were no statistically significant differences among the three groups with respect to baseline characteristics, including gender, age, smoking history, histological type, clinical stage, and ECOG performance status (P > 0.05). Immune-related marker expression was significantly different between the EGFR single mutation group and the two co-mutation groups (P < 0.05), while no significant difference was observed between the co-mutation groups (P = 0.945). Following first-line EGFR-TKI therapy, the EGFR single mutation group showed a significantly longer median PFS compared with the EGFR/TP53 and EGFR/K-RAS co-mutation groups (P < 0.0001). No significant difference in PFS was observed between the two co-mutation groups (P = 0.174). Following progression on EGFR-TKIs, all patients received platinum-based doublet chemotherapy plus immunotherapy. In second-line treatment, the median PFS in the EGFR single-mutation group, which was shorter than in the EGFR/TP53 and EGFR/KRAS co-mutation groups (overall log-rank P < 0.0001), with no significant difference between the two co-mutation cohorts (P = 0.174). However, in multivariable Cox models adjusting for age, sex, smoking history, clinical stage, histology, and ECOG performance status, both EGFR/TP53 and EGFR/KRAS co-mutations were independently associated with a higher hazard of progression. ECOG PS ≥2 was associated with a numerically higher hazard that did not reach statistical significance. No significant associations were observed for other covariates (age, sex, smoking history, clinical stage, histology; all P>0.05).
    Conclusion: In the first-line setting, patients with an EGFR single mutation treated with EGFR-TKIs had a longer median PFS than those with EGFR/TP53 and EGFR/KRAS co-mutations (14.1 vs 10.4 and 10.9 months, respectively; both P < 0.0001), whereas no statistically significant difference was observed between the two co-mutation subgroups (P = 0.174). Following the development of resistance, all patients received platinum-based doublet chemotherapy plus immunotherapy; in the second-line setting, median PFS was modestly longer in the co-mutation groups compared with the single-mutation group (EGFR/TP53: 5.2 months; EGFR/KRAS: 5.0 months; EGFR single mutation: 3.9 months; overall log-rank P < 0.0001), with no significant difference between the TP53 and KRAS subgroups (P = 0.174). These associations were evident on Kaplan-Meier curves (with numbers at risk) and log-rank testing, and were supported by multivariable Cox models adjusted for age, sex, smoking history, clinical stage, histology, and ECOG performance status.
    Keywords:  EGFR-TKI; co-mutation; immunotherapy; non-small cell lung cancer; progression-free survival
    DOI:  https://doi.org/10.3389/fimmu.2025.1684089
  9. Mol Med. 2025 Nov 29.
      Multiple myeloma (MM) with high-risk (HR) genetic abnormalities has poor prognosis, despite the use of novel therapeutic agents. However, the individual contribution of specific HR genetic abnormalities or their co-occurrence to poor outcomes, especially in the era of novel agents, remains unclear. This study evaluated the impact of multi-hit TP53 (del(17p) and TP53 mutation or ≥ 2 TP53 mutations) compared with other HR abnormalities on progression-free survival (PFS), overall survival (OS) and blood signature in a real-world cohort of 204 patients with MM treated with novel agents (median follow-up 28 months). Patients with multi-hit TP53 (10.4%) had the shortest PFS and OS compared with those with single HR abnormalities (p ≤ 0.011) or with co-occurrence of ≥ 2 other HR abnormalities (p ≤ 0.002), regardless of therapy line. The relative risk of early progression in patients with multi-hit TP53 was almost three times higher than that of patients with other HR abnormalities. The prevalence of TP53 alterations increased in later disease stages: multi-hit TP53 was detected in 7.6% of patients with ≤ 1 prior therapy line and in 36.4% of patients with ≥ 2 prior lines. Patients with multi-hit TP53 also differed in blood signature, particularly in counts of white blood cells, lymphocytes, serum creatinine and β2-microglobulin levels compared with those with other HR abnormalities. In conclusion, multi-hit TP53 is associated with the poorest survival among all HR subgroups in MM. Considering that TP53 alterations accumulate during MM progression and are associated with drug resistance even in the context of novel therapies, our study further emphasizes the need for routine evaluation of both del(17p) and TP53 mutations. Patients with multi-hit TP53 should be prioritized for inclusion in trials of novel therapeutic strategies.
    Keywords:  High-risk Myeloma; Multi-hit TP53 ; Multiple Myeloma; Next-Generation Sequencing
    DOI:  https://doi.org/10.1186/s10020-025-01392-2