• HOME
  • NEWS
  • EXPLORE
    • CAREER
      • Companies
      • Jobs
    • EVENTS
    • iGEM
      • News
      • Team
    • PHOTOS
    • VIDEO
    • WIKI
  • BLOG
  • COMMUNITY
    • FACEBOOK
    • INSTAGRAM
    • TWITTER
Tuesday, November 25, 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

Aldometanib Extends Lifespan in Liver Cancer Mice

Bioengineer by Bioengineer
November 25, 2025
in Health
Reading Time: 5 mins read
0
Share on FacebookShare on TwitterShare on LinkedinShare on RedditShare on Telegram

In a groundbreaking advance that could redefine cancer therapy, a newly developed glucose starvation mimetic, aldometanib, has demonstrated an extraordinary ability to overcome immune resistance in mice suffering from hepatocellular carcinoma (HCC). This discovery promises not only to extend survival but potentially enable afflicted mice to reach normal lifespan benchmarks, an unprecedented milestone in oncology research. The findings, recently published in Cell Research, outline a novel approach targeting tumor metabolism that dismantles immune barriers and reactivates the body’s natural defenses against one of the most formidable liver cancers.

Hepatocellular carcinoma, a primary malignancy of the liver, notoriously evades immune destruction by creating an immunosuppressive microenvironment, which shields tumor cells from immune surveillance and therapeutics alike. Traditional modalities, including chemotherapy and immunotherapy, have shown limited efficacy in significantly altering survival outcomes in advanced HCC. The work led by Hu, Wang, and Lan and their colleagues reveals that metabolic stress, specifically glucose deprivation mimicked pharmacologically by aldometanib, can disrupt this tumor immune evasion, opening a potent avenue for cancer control.

This study introduces aldometanib as a pharmacological agent designed to simulate the effects of glucose starvation within the tumor microenvironment without causing systemic hypoglycemia. By selectively targeting tumor cell metabolism, aldometanib induces a state that hinders the energetic and biosynthetic capacity of cancer cells, simultaneously modulating key immune cells that have been co-opted by the tumor. The mimicry of glucose scarcity impairs the tumor’s ability to sustain its immune suppressive tactics, thereby reinvigorating antitumor immunity.

In mechanistic detail, aldometanib operates through interference with glycolysis pathways that tumors heavily rely upon for energy production and survival. Cancer cells exhibit an elevated glucose uptake to meet their high metabolic demands—a phenomenon known as the Warburg effect. Aldometanib exploits this metabolic vulnerability by disrupting crucial enzymatic activities downstream of glucose metabolism, which not only weakens tumor cells but also alters the metabolic programming of immune cells within the tumor microenvironment, such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs).

One of the pivotal breakthroughs reported is the restoration of effective cytotoxic T lymphocyte (CTL) function. Normally incapacitated in the context of dense immune barriers erected by the tumor, CTLs regain their activation and infiltration capacities following aldometanib treatment. This reactivation circumvents one of the major hurdles in current immunotherapeutic approaches where the immune system is present but functionally paralyzed. Consequently, the tumor is subjected to renewed immune pressure, leading to suppressed growth and reduced metastatic potential.

The experimental model employed—mice genetically predisposed to develop hepatocellular carcinoma—provides highly relevant preclinical insights. Notably, these mice, treated with aldometanib, were observed to survive to normal lifespan expectations without evident toxicity or adverse metabolic effects. This outcome contrasts sharply with conventional treatments, which often extend life only modestly and with considerable side effects. The extended survival and preserved quality of life highlight the therapeutic window and safety of targeting metabolic checkpoints in liver cancer.

Importantly, the study addresses the complex interplay between tumor metabolism and immune regulation. Tumor cells manipulate the metabolic landscape to create niches of nutrient deprivation for immune cells, resulting in energy-starved lymphocytes with compromised effector functions. Aldometanib counteracts this metabolic sabotage by normalizing nutrient availability for immune cells and disrupting the metabolic crosstalk that favors tumor survival. This metabolic therapeutic strategy represents a paradigm shift from direct cytotoxicity toward immune modulation through metabolic intervention.

The research further delineates how aldometanib influences signaling pathways involved in immune cell trafficking and activation. Increased expression of chemokines and cytokines supportive of effector T cell recruitment was observed, while suppressive signals were markedly diminished. This reprogramming of the tumor microenvironment promotes immune infiltration and sustained antitumor activity, providing a comprehensive assault on tumor-mediated immune escape phenomena.

Moreover, aldometanib’s specificity for tumor metabolic pathways reduces collateral damage to normal tissues—a notable advancement compared to earlier glycolysis inhibitors whose systemic toxicity limited clinical applications. The nuanced modulation of metabolism allows the preservation of physiological functions while selectively incapacitating cancer cells and immune suppressive elements. This selectivity paves the way for combining aldometanib with existing immunotherapies, potentially enhancing their efficacy through synergistic mechanisms.

The authors emphasize that beyond hepatocellular carcinoma, this metabolic approach may have wide-reaching implications for other cancers characterized by immune exclusion and metabolic dysregulation. Tumors that employ similar immune evasion strategies through metabolic rewiring could be rendered susceptible to analogous interventions, broadening the impact of this research. Future studies will be essential to validate these findings across different cancer types and to optimize dosing strategies for maximal therapeutic benefit.

Importantly, this discovery aligns with the growing appreciation of cancer metabolism as a therapeutic frontier. Metabolic checkpoints are emerging as critical regulators of tumor-immune interactions, and agents like aldometanib exemplify how targeting metabolism can transcend traditional boundaries of oncology. This innovative approach underscores the value of integrating metabolic science with immunology to devise multidimensional cancer therapies.

The translational potential of aldometanib is underscored by its favorable pharmacokinetic properties observed in preclinical evaluations. Oral bioavailability, metabolic stability, and minimal off-target effects were all reported, enhancing prospects for clinical development. These attributes will facilitate the progression to human trials, where the promise of extending survival in patients with aggressive liver cancer could dramatically alter clinical practice.

The implications extend beyond survival statistics; by restoring immune competence, aldometanib represents a step toward durable cancer remission and possibly cure. Current immune checkpoint inhibitors have transformed cancer care but are often limited by resistance mechanisms and incomplete immune reactivation. The glucose starvation mimetic’s ability to remove foundational immune barriers suggests a complementary role that could enhance and sustain responses over time.

This work illustrates how a deep understanding of tumor biology at the intersection of metabolism and immunity can yield novel therapeutic insights. By combining cutting-edge metabolic inhibitors with immune modulators, researchers are carving out new strategies that harness the body’s own defenses against cancer. The success in murine HCC models offers hope for similarly impactful innovations in human oncology.

Looking forward, clinical trials will be pivotal in confirming efficacy and safety in human populations, identifying biomarkers predictive of response, and determining the best combinational regimens. If successful, aldometanib could become a cornerstone agent in precision oncology, transforming lives and expanding the horizon of cancer therapeutics beyond current limits.

This landmark study reaffirms that overcoming cancer is not solely a battle of drugs versus tumor cells but a sophisticated engagement of metabolic and immune networks. The glucose starvation mimetic aldometanib embodies this principle, unlocking pathways to long-term survival and immune restoration in one of the deadliest cancers. As researchers continue to unravel metabolic-immune interactions, the future of cancer therapy looks increasingly bright and hopeful.

Subject of Research: The use of a glucose starvation mimetic, aldometanib, to remove immune barriers and extend survival in mice with hepatocellular carcinoma.

Article Title: Glucose starvation mimetic aldometanib removes immune barriers permitting mice with hepatocellular carcinoma to live to normal ages.

Article References:
Hu, HH., Wang, X., Lan, B. et al. Glucose starvation mimetic aldometanib removes immune barriers permitting mice with hepatocellular carcinoma to live to normal ages. Cell Res (2025). https://doi.org/10.1038/s41422-025-01195-4

Image Credits: AI Generated

DOI: https://doi.org/10.1038/s41422-025-01195-4

Tags: advancements in hepatocellular carcinoma researchAldometanib for liver cancerextending lifespan in cancer modelsglucose starvation mimetic in oncologyhepatocellular carcinoma treatment breakthroughsimmunosuppressive microenvironment in liver cancermetabolic stress and tumor controlnovel cancer therapies and survival ratesovercoming immune resistance in HCCpharmacological agents in cancer treatmentreactivating natural defenses against cancertumor metabolism targeting in cancer therapy

Share12Tweet7Share2ShareShareShare1

Related Posts

Maternal Gut Drives Newborn Antibiotic-Resistant Bacteria

November 25, 2025

IGF2BP3/IL6ST/STAT3 Loop Accelerates Colorectal Cancer Progression

November 25, 2025

Unlocking Biomarkers for Platinum Resistance in Ovarian Cancer

November 25, 2025

Thyroid Hormone Sensitivity Linked to Frailty in Seniors

November 25, 2025

POPULAR NEWS

  • New Research Unveils the Pathway for CEOs to Achieve Social Media Stardom

    New Research Unveils the Pathway for CEOs to Achieve Social Media Stardom

    203 shares
    Share 81 Tweet 51
  • Scientists Uncover Chameleon’s Telephone-Cord-Like Optic Nerves, A Feature Missed by Aristotle and Newton

    119 shares
    Share 48 Tweet 30
  • Neurological Impacts of COVID and MIS-C in Children

    93 shares
    Share 37 Tweet 23
  • Scientists Create Fast, Scalable In Planta Directed Evolution Platform

    98 shares
    Share 39 Tweet 25

About

BIOENGINEER.ORG

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

Follow us

Recent News

Maternal Gut Drives Newborn Antibiotic-Resistant Bacteria

IGF2BP3/IL6ST/STAT3 Loop Accelerates Colorectal Cancer Progression

Unlocking Biomarkers for Platinum Resistance in Ovarian Cancer

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.