DiosMetin Targets INF2: New Colorectal Therapy
Colorectal cancer (CRC) remains one of the most formidable challenges in oncology, ranking as the third most prevalent malignancy within the gastrointestinal tract and occupying the position of the second leading cause of cancer-related mortality worldwide. For decades, researchers have pursued the identification of molecular targets that could enable the development of efficacious, precision therapies. […]

Colorectal cancer (CRC) remains one of the most formidable challenges in oncology, ranking as the third most prevalent malignancy within the gastrointestinal tract and occupying the position of the second leading cause of cancer-related mortality worldwide. For decades, researchers have pursued the identification of molecular targets that could enable the development of efficacious, precision therapies. The intrinsic complexity of CRC, exacerbated by the heterogeneity of tumor cell populations and the non-specific expression of many biomarkers across diverse cell types, has historically impeded efforts to create targeted therapeutic strategies with minimal off-target effects. However, a recent breakthrough study published in BMC Cancer in 2025 reveals a promising new avenue for CRC treatment through the precise targeting of a novel biomarker, INF2, utilizing a natural compound known as DiosMetin 7-O-β-D-Glucuronide.
The crux of this research centers around INF2, a formin family protein characterized by its involvement in actin cytoskeleton remodeling—vital for numerous cellular processes, including cell division and motility. Using cutting-edge single-cell RNA sequencing technologies coupled with advanced machine learning algorithms, the investigators meticulously mapped INF2 expression patterns within CRC tissues. Their analyses revealed that INF2 is not only significantly overexpressed in colorectal tumors but its levels positively correlate with disease progression, marking it as an unequivocal prognostic biomarker in CRC. This represents a pivotal leap forward, as INF2’s distinct elevation in cancerous cells compared to normal tissue offers a tangible target for therapeutic intervention.
Delving deeper into the functional role of INF2, the research team employed a series of in vitro assays involving CRC cell lines with high INF2 expression. Genetic knockdown experiments elucidated that silencing INF2 substantially curtailed the proliferation and migratory capabilities of these malignant cells, underscoring INF2’s essential contribution to tumor growth dynamics and metastatic potential. This functional validation not only confirms the biomarker’s clinical relevance but also substantiates the rationale for pursuing INF2 inhibition as a therapeutic strategy.
.adsslot_blkfTN19Ie{ width:728px !important; height:90px !important; }
@media (max-width:1199px) { .adsslot_blkfTN19Ie{ width:468px !important; height:60px !important; } }
@media (max-width:767px) { .adsslot_blkfTN19Ie{ width:320px !important; height:50px !important; } }
ADVERTISEMENT
Having established the foundation for INF2 as a viable target, the investigators embarked on an innovative screening effort to identify compounds capable of selectively inhibiting INF2 activity. This pursuit led them to DiosMetin 7-O-β-D-Glucuronide, a glucuronidated metabolite of the flavonoid diosmetin, which is naturally abundant in several plant species and known for its bioactive properties. Through computational docking studies, the compound demonstrated high binding affinity to INF2’s functional domains, suggesting a direct inhibitory mechanism. Biochemical assays corroborated these findings, showing that DiosMetin 7-O-β-D-Glucuronide effectively impairs INF2-mediated actin polymerization in CRC cells.
Importantly, the therapeutic window of DiosMetin 7-O-β-D-Glucuronide was rigorously evaluated, revealing a striking selective cytotoxicity profile. While INF2-high CRC cells experienced marked suppression in proliferation and migration upon treatment, normal colorectal epithelial cells exhibited minimal adverse effects, emphasizing the compound’s specificity and potential safety in clinical scenarios. This selectivity is a critical hallmark for any prospective anticancer agent, especially given the notorious toxicity associated with conventional chemotherapies.
Further clinical relevance was substantiated through immunohistochemical analyses of CRC patient tissue samples. Consistently higher INF2 staining was observed in late-stage tumors, aligning well with transcriptomic data and solidifying INF2’s role as a marker of disease severity. This confluence of data from molecular, cellular, and tissue levels delivers a comprehensive picture of INF2 as not only a biomarker but also a functional driver of colorectal carcinogenesis.
The implications of targeting INF2 with DiosMetin 7-O-β-D-Glucuronide transcend the immediate therapeutic potential. This approach exemplifies the broader paradigm shift in cancer treatment — leveraging precision medicine powered by deep molecular insights and natural product pharmacology. By integrating computational tools, single-cell omics, and traditional biochemical methods, the study embodies a multi-disciplinary strategy that modern oncology desperately requires.
Unlike many current therapeutic agents that indiscriminately attack rapidly dividing cells, risking substantial collateral damage, INF2 inhibition promises a more refined attack on tumor cells that rely heavily on cytoskeletal dynamics to invade and disseminate. The modulation of actin remodeling through INF2 interference represents a novel mode of action distinct from classical chemotherapeutic targets such as DNA synthesis inhibitors or microtubule disruptors.
Another remarkable aspect of this research is the utilization of DiosMetin 7-O-β-D-Glucuronide, which taps into the vast and relatively underexploited reservoir of natural compounds for anticancer drug development. The compound’s natural derivation and demonstrated specificity could potentially translate to fewer side effects and improved patient compliance, addressing some of the major limitations of existing therapies. Moreover, the metabolite’s known pharmacokinetic properties could facilitate its optimization for oral administration and systemic delivery.
Looking ahead, the research team envisions several avenues for translating these findings into clinical practice. Preclinical studies in animal models are anticipated to assess in vivo efficacy, biodistribution, and toxicity profiles. Should these prove favorable, early-phase clinical trials could elucidate the therapeutic index of DiosMetin 7-O-β-D-Glucuronide in human CRC patients, particularly those exhibiting high INF2 expression in tumor biopsies.
From a diagnostic perspective, the establishment of INF2 as a predictive biomarker could revolutionize patient stratification. Liquid biopsy techniques, bioinformatics-driven pathology, and immunohistochemical scoring systems may converge to guide tailored therapeutic regimens, ensuring that only patients likely to benefit from INF2-targeted therapy receive such interventions, thus enhancing treatment efficacy and minimizing unnecessary exposure.
This study also raises intriguing questions about the broader biological role of INF2 in cancer biology. While its involvement in CRC is now more clearly defined, exploration into its functions in other malignancies and its interplay with other cellular pathways might uncover further therapeutic targets or synergistic drug combinations.
Furthermore, the integration of machine learning algorithms to dissect single-cell transcriptomes sets a methodological benchmark for future cancer biomarker discovery. This approach can be adapted to other cancer types, enhancing the granularity of tumor profiling and enabling the identification of highly specific molecular vulnerabilities.
The identification and validation of DiosMetin 7-O-β-D-Glucuronide as a selective INF2 inhibitor heralds a promising leap toward a new class of targeted therapeutics in colorectal cancer. This breakthrough underscores the power of harnessing natural compounds and advanced computational biology to overcome longstanding challenges in oncology.
As the oncology landscape evolves, strategies such as the one outlined in this study will be instrumental in shifting from broadly cytotoxic treatments toward more precise, effective, and less toxic therapeutic interventions. The marriage of biomarker identification and natural product pharmacology represented here may serve as a blueprint for future drug discovery efforts.
Ultimately, this novel INF2-centered approach offers hope for the millions of patients worldwide battling colorectal cancer. With continued research and clinical development, DiosMetin 7-O-β-D-Glucuronide could emerge as a cornerstone in the arsenal against this devastating disease, transforming patient outcomes and redefining therapeutic paradigms in the 21st century.
Subject of Research: Colorectal cancer therapy targeting the biomarker INF2 using a natural compound inhibitor.
Article Title: Targeting INF2 with DiosMetin 7-O-β-D-Glucuronide: a new stratagem for colorectal cancer therapy.
Article References:
Zeng, Z., Ke, Y., Huang, F. et al. Targeting INF2 with DiosMetin 7-O-β-D-Glucuronide: a new stratagem for colorectal cancer therapy. BMC Cancer 25, 982 (2025). https://doi.org/10.1186/s12885-025-14357-9
Image Credits: Scienmag.com
DOI: https://doi.org/10.1186/s12885-025-14357-9
Tags: actin cytoskeleton remodelingbreakthroughs in colorectal therapycancer-related mortality statisticscolorectal cancer treatmentDiosMetin 7-O-β-D-GlucuronideINF2 biomarker researchmachine learning in oncologynovel natural compounds in cancer therapyprecision oncology advancementssingle-cell RNA sequencing in cancertargeted therapies for CRCtumor heterogeneity in colorectal cancer
What's Your Reaction?






