• HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
Saturday, November 15, 2025
BIOENGINEER.ORG
No Result
View All Result
  • Login
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
  • HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
        • Lecturer
        • PhD Studentship
        • Postdoc
        • Research Assistant
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
No Result
View All Result
Bioengineer.org
No Result
View All Result
Home NEWS Science News Health

Sulindac Sulfide Blocks Cancer via let-7b-K-Ras Pathway

Bioengineer by Bioengineer
November 15, 2025
in Health
Reading Time: 4 mins read
0
Share on FacebookShare on TwitterShare on LinkedinShare on RedditShare on Telegram

In an era where cancer research continuously pushes the boundaries of therapeutic innovation, a groundbreaking study has emerged spotlighting the potential of sulindac sulfide, a non-steroidal anti-inflammatory drug (NSAID) metabolite, in suppressing oncogenic transformation. This novel investigation, spearheaded by researchers Liang, Z., Ma, R., Yi, B., and colleagues, elucidates a sophisticated molecular interplay involving let-7b microRNA and the notorious K-Ras signaling pathway, a driver implicated in various malignancies. Published recently in Cell Death Discovery, the study illuminates mechanisms by which sulindac sulfide curtails cancerous progression, marking a significant stride in targeted cancer therapy development.

Crucial to the study is the role of let-7b, a member of the let-7 family of microRNAs, widely recognized for its tumor-suppressive properties. The let-7 family intricately regulates gene expression post-transcriptionally, and let-7b in particular has garnered attention for its ability to modulate proto-oncogenes. The researchers strategically focused on how sulindac sulfide influences let-7b to inhibit aberrant cell transformation. Their findings reveal that administration of sulindac sulfide elevates let-7b expression levels, which in turn exerts a potent repressive effect on K-Ras signaling, a pathway frequently hyperactivated in a spectrum of human cancers.

K-Ras, a small GTPase protein, serves as a pivotal molecular switch modulating cell proliferation, differentiation, and survival. Mutations in K-Ras represent some of the most common genetic aberrations in oncogenesis, conferring aggressive growth and therapeutic resistance. However, directly targeting K-Ras has historically been clinically challenging due to its structural and functional complexities. The mechanism uncovered by this research illustrates an indirect yet robust approach: enhancing let-7b levels to suppress K-Ras expression and downstream oncogenic signaling, thereby impeding cancer cell transformation without the need for direct K-Ras blockade.

The investigative team employed a comprehensive array of molecular and cellular biology techniques to delineate this pathway. Using oncogenic transformation models and sophisticated gene expression assays, they quantified the upregulation of let-7b in response to sulindac sulfide treatment. Concurrently, they measured a concomitant decrease in K-Ras protein levels, confirming the translational repression orchestrated by let-7b microRNA binding to the 3′ untranslated region of K-Ras mRNA. This transcriptional interference effectively diminished the oncogenic signaling cascade, leading to a suppression of tumorigenic phenotypes.

Beyond in vitro assays, the study extended its scope to in vivo models, underscoring the translational potential of sulindac sulfide. Animal models with induced K-Ras-driven tumors exhibited significantly reduced tumor growth and improved histopathological features upon treatment with sulindac sulfide. This hints at the drug’s efficacy in real-world biological contexts, imparting hope for therapeutic application in patients whose cancers harbor K-Ras mutations or depend on aberrant K-Ras signaling for progression.

One particularly striking aspect of this research is the therapeutic repurposing of sulindac sulfide, a metabolite of a well-characterized NSAID with a long history of clinical use for inflammatory conditions. The safety profile of such NSAIDs is well-documented, potentially expediting the transition of sulindac sulfide into oncological clinical trials. This repositioning could mitigate the protracted timelines typically associated with novel drug development, offering a faster roadmap to targeted cancer therapy.

The study also delves into the broader implications of microRNA modulation in oncology. MicroRNAs like let-7b serve as master regulators, capable of orchestrating complex gene networks involved in cell fate determination. By leveraging microRNAs to indirectly target difficult-oncology proteins such as K-Ras, the work pioneers a promising paradigm shift in cancer treatment strategies, where small RNA molecules become central therapeutic nodes.

Intriguingly, the upregulation of let-7b by sulindac sulfide involves epigenetic modification dynamics not fully elucidated here but warranting future investigation. The potential interplay between the drug and chromatin remodeling enzymes or DNA methylation states could further enhance the precision of therapeutic interventions aimed at reinstituting tumor suppressor microRNAs.

Moreover, the researchers identify a reduction in downstream effectors of K-Ras signaling, including those involved in the MAPK/ERK and PI3K/AKT pathways, which are critical conduits for cell proliferation and survival in cancerous tissues. This multifaceted downregulation underscores the potency of let-7b-mediated repression in dismantling the oncogenic network at various nodes, culminating in comprehensive growth inhibition of transformed cells.

Considering the challenge of resistance in cancer therapies, this microRNA-based mechanism offers a new vantage point, as targeting K-Ras indirectly via let-7b may circumvent common resistance mutations that emerge against direct inhibitors. This provides a durable therapeutic strategy by exploiting the endogenous regulatory machinery of cells to maintain oncogenic suppression.

Notably, the researchers emphasize the specificity of sulindac sulfide’s action in elevating let-7b among the let-7 family members and the subsequent selective repression of K-Ras. Such specificity reduces the risk of off-target effects and underscores the precision that can be achieved through modulating microRNA expression, an aspect critical for minimizing toxicity in clinical use.

While sulindac sulfide shows compelling promise, the study also recognizes the importance of further clinical validation. Dosage optimization, pharmacokinetic profiling, and long-term toxicity studies are necessary to fully harness this compound’s therapeutic potential. The groundwork laid here will fuel multi-disciplinary efforts to translate these bench-side discoveries to bedside treatments.

The discovery also sparks considerations about combinatorial regimens. Leveraging sulindac sulfide alongside existing chemotherapeutic or targeted agents could enhance therapeutic outcomes by attacking cancer cells through distinct yet complementary molecular pathways. Such strategies could potentiate responses and delay resistance further.

In conclusion, the work by Liang and colleagues represents a landmark advance by revealing the role of sulindac sulfide in suppressing oncogenic transformation through a let-7b-mediated repression of K-Ras signaling. It shines a spotlight on microRNA-based therapeutics as a promising frontier in oncology, emphasizing the utility of repurposing established drugs to combat some of the most challenging oncogenic drivers. This study adds a vital piece to the complex puzzle of K-Ras-targeted cancer therapy, setting the stage for a new era of precision oncology.

As research continues to unravel the sophisticated molecular crosstalk underlying cancer, findings such as these amplify optimism that targeted, effective, and safer cancer treatments are within reach. Sulindac sulfide and let-7b together could reshape therapeutic landscapes, transforming incurable cancers into manageable conditions, and heralding a future where oncogenic signaling pathways are no longer insurmountable barriers but actionable targets.

Subject of Research: Investigation into how sulindac sulfide suppresses oncogenic transformation via let-7b-mediated repression of K-Ras signaling.

Article Title: Sulindac sulfide suppresses oncogenic transformation through let-7b-mediated repression of K-Ras signaling.

Article References:
Liang, Z., Ma, R., Yi, B. et al. Sulindac sulfide suppresses oncogenic transformation through let-7b-mediated repression of K-Ras signaling. Cell Death Discov. 11, 530 (2025). https://doi.org/10.1038/s41420-025-02858-2

Image Credits: AI Generated

DOI: 14 November 2025

Tags: cancer progression curtailmentcancer research innovationsgene expression regulation in cancerK-Ras signaling pathway inhibitionlet-7b microRNA rolemolecular mechanisms of cancer treatmentnon-steroidal anti-inflammatory drugsoncogenic transformation suppressionsulindac sulfide cancer therapysulindac sulfide mechanism of actiontargeted cancer therapy advancementstumor-suppressive microRNAs

Share12Tweet8Share2ShareShareShare2

Related Posts

Unveiling Microplastics: Extraction and Analysis Techniques

November 15, 2025

Protein Biomarkers: A New Era in Pulmonary Hypertension

November 15, 2025

Depression and Death Anxiety in Elderly Quality of Life

November 15, 2025

Assessing Midwives’ Workload in Free Maternal Care

November 15, 2025

POPULAR NEWS

  • blank

    Stinkbug Leg Organ Hosts Symbiotic Fungi That Protect Eggs from Parasitic Wasps

    318 shares
    Share 127 Tweet 80
  • ESMO 2025: mRNA COVID Vaccines Enhance Efficacy of Cancer Immunotherapy

    210 shares
    Share 84 Tweet 53
  • New Research Unveils the Pathway for CEOs to Achieve Social Media Stardom

    201 shares
    Share 80 Tweet 50
  • New Study Suggests ALS and MS May Stem from Common Environmental Factor

    142 shares
    Share 57 Tweet 36

About

We bring you the latest biotechnology news from best research centers and universities around the world. Check our website.

Follow us

Recent News

Unveiling Microplastics: Extraction and Analysis Techniques

MD Anderson Unveils $2.5 Billion “Only Possible Here” Campaign to Transform Cancer Research and Treatment

Protein Biomarkers: A New Era in Pulmonary Hypertension

Subscribe to Blog via Email

Enter your email address to subscribe to this blog and receive notifications of new posts by email.

Join 69 other subscribers
  • Contact Us

Bioengineer.org © Copyright 2023 All Rights Reserved.

Welcome Back!

Login to your account below

Forgotten Password?

Retrieve your password

Please enter your username or email address to reset your password.

Log In
No Result
View All Result
  • Homepages
    • Home Page 1
    • Home Page 2
  • News
  • National
  • Business
  • Health
  • Lifestyle
  • Science

Bioengineer.org © Copyright 2023 All Rights Reserved.