toplogo
Sign In

Dual Targeting of Histone Deacetylases and MYC as a Promising Therapeutic Approach for H3-K27M Pediatric Gliomas


Core Concepts
Combining inhibition of histone deacetylases (HDACi) and the MYC pathway using a PIN1 inhibitor (Sulfopin) shows synergistic anti-tumor effects in H3-K27M pediatric glioma models, by downregulating prominent oncogenic pathways such as mTOR.
Abstract

This study explores a novel therapeutic strategy for treating H3-K27M-mutant diffuse midline gliomas (DMGs), an aggressive and fatal pediatric brain tumor. The authors demonstrate that dual targeting of histone deacetylases (HDACi) and the MYC pathway using a PIN1 inhibitor (Sulfopin) leads to synergistic inhibition of tumor cell viability in patient-derived H3-K27M glioma cultures.

Key highlights:

  • Sulfopin, a PIN1 inhibitor, downregulates MYC target genes and reduces viability of H3-K27M glioma cells.
  • Combining Sulfopin with the HDACi Vorinostat results in an additive effect on cell viability, more pronounced in H3.3-K27M compared to H3.1-K27M cells.
  • The combined treatment leads to robust downregulation of prominent oncogenic pathways, including mTOR signaling, both in vitro and in vivo.
  • Mechanistically, the combined treatment attenuates the HDACi-mediated increase in H3K27ac levels on promoters and enhancers of key oncogenes.
  • In an orthotopic xenograft model, the combined treatment significantly reduces tumor growth compared to single agents or control.

These findings highlight the therapeutic potential of simultaneously targeting epigenetic and transcriptional vulnerabilities in H3-K27M gliomas, and provide a rationale for further clinical development of this combination approach.

edit_icon

Customize Summary

edit_icon

Rewrite with AI

edit_icon

Generate Citations

translate_icon

Translate Source

visual_icon

Generate MindMap

visit_icon

Visit Source

Stats
Sulfopin treatment led to a mild reduction in cell viability across eight patient-derived DMG cultures. Combination of Sulfopin (10 μM) and Vorinostat (1 μM) reduced cell viability by 80% in SU-DIPG13 cells. The combination treatment significantly reduced tumor growth in an orthotopic xenograft mouse model compared to control or single agent treatments.
Quotes
"Sulfopin dependent inhibition of PIN1 was shown to downregulate the expression of MYC target genes in several cancer cell lines." "Genes downregulated by Sulfopin were also enriched for MYC-bound genes identified in several non-glioma cell lines, supporting a role for PIN1 in activation of the MYC pathway also in these glioma cells." "Sulfopin treatment resulted in reduction of H3K27me3 signal over CpG islands, perhaps contributing to Sulfopin-dependent growth inhibition in these cells."

Deeper Inquiries

What are the potential mechanisms by which the combination of Sulfopin and Vorinostat leads to synergistic downregulation of the mTOR pathway in H3-K27M gliomas?

The combination of Sulfopin and Vorinostat likely exerts synergistic downregulation of the mTOR pathway in H3-K27M gliomas through multiple mechanisms: MYC Inhibition: Sulfopin, as a PIN1 inhibitor, downregulates MYC target genes, which are known to be involved in the regulation of mTOR signaling. By inhibiting MYC activity, Sulfopin indirectly affects the expression of genes within the mTOR pathway. Epigenetic Regulation: Vorinostat, as an HDAC inhibitor, leads to increased histone acetylation, including H3K27ac, which is associated with active promoters and enhancers. The combination treatment attenuates the accumulation of H3K27ac on promoters and enhancers of oncogenes within the mTOR pathway, resulting in their decreased expression. Transcriptional Disruption: The combined treatment alters the transcriptional programs of H3-K27M glioma cells, leading to the downregulation of genes associated with the mTOR pathway. This transcriptional disruption may involve the direct or indirect regulation of key genes within the mTOR signaling cascade. Additive Effect: The additive effect of Sulfopin and Vorinostat on cell viability and gene expression profiles suggests a synergistic interaction between the two drugs in targeting oncogenic pathways, including mTOR. This synergistic effect may involve complementary mechanisms of action that converge on the regulation of mTOR signaling.

How could the observed differences in sensitivity between H3.3-K27M and H3.1-K27M cells to the combined treatment be leveraged to develop personalized therapeutic approaches?

The observed differences in sensitivity between H3.3-K27M and H3.1-K27M cells to the combined treatment with Sulfopin and Vorinostat provide an opportunity to develop personalized therapeutic approaches for H3-K27M gliomas: Biomarker Identification: The differential response of H3.3-K27M and H3.1-K27M cells to the combination treatment suggests that specific molecular markers, such as MYC expression levels, could be used to predict individual responses. By profiling these markers in patient tumors, clinicians can stratify patients based on their predicted sensitivity to the treatment. Tailored Treatment Plans: Patients with H3.3-K27M mutations, which show higher sensitivity to the combination therapy, could be prioritized for this specific treatment regimen. Conversely, alternative therapeutic strategies may be considered for patients with H3.1-K27M mutations, taking into account their lower sensitivity to the combined treatment. Precision Medicine: Leveraging the differences in sensitivity between H3.3-K27M and H3.1-K27M cells allows for the implementation of precision medicine approaches in the treatment of H3-K27M gliomas. By customizing treatment plans based on the molecular characteristics of individual tumors, clinicians can optimize therapeutic outcomes and minimize potential side effects.

Given the limitations of the xenograft model in recapitulating the full complexity of the tumor microenvironment, what additional in vivo studies could be conducted to further validate the therapeutic potential of this combination strategy?

To further validate the therapeutic potential of the combination strategy of Sulfopin and Vorinostat in H3-K27M gliomas, additional in vivo studies could be conducted, including: Patient-Derived Xenograft Models: Utilizing patient-derived xenograft (PDX) models derived from H3-K27M glioma patients can better recapitulate the heterogeneity and complexity of human tumors. Testing the combination treatment in PDX models can provide more clinically relevant insights into its efficacy and safety. Immunocompetent Models: Employing immunocompetent mouse models, such as genetically engineered mouse models (GEMMs) or syngeneic models, can assess the impact of the combination therapy on the immune response within the tumor microenvironment. This can help evaluate potential immunomodulatory effects of the treatment. Combination with Immunotherapies: Investigating the combination of Sulfopin and Vorinostat with immunotherapies, such as immune checkpoint inhibitors or CAR-T cell therapy, can explore synergistic effects and potential enhanced anti-tumor immune responses. This approach can address the limitations of the xenograft model in capturing immune interactions. Long-Term Survival Studies: Conducting long-term survival studies in animal models treated with the combination therapy can provide insights into the durability of the treatment response and potential for disease recurrence. Monitoring survival outcomes over an extended period can inform the clinical relevance of the treatment strategy. By incorporating these additional in vivo studies, researchers can further validate the therapeutic potential of the combination strategy and enhance the translational relevance of the findings for future clinical applications.
0
star