Publications

Resolution of innate immune cells with proresolving lipid mediators in idiopathic pulmonary fibrosis

Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive incurable lung disease characterized by chronic inflammation and fibrosis, with innate immune cells such as neutrophils and macrophages playing central roles in its pathogenesis. This review explores the involvement of these immune cells in the inflammatory process of IPF, focusing on their contribution to disrupted tissue repair and impaired resolution. The balance between host defense mechanisms, including leukocyte recruitment, and the release of proresolving mediators is crucial for maintaining healthy tissue function and returning to preinflammatory states. We highlight the importance of inflammation resolution to prevent an overactive immune response, which can lead to irreversible fibrosis. Specialized proresolving mediators, including lipoxins, resolvins, protectins, and maresins, are discussed in terms of their regulatory effects on neutrophils and macrophages in IPF. These mediators exhibit potent anti-inflammatory actions, which can modulate the immune response and promote the resolution of inflammation. Overall, this review underscores the significance of immune modulation in IPF, with a focus on the therapeutic potential of specialized proresolving mediators in controlling the inflammatory response and preventing fibrosis progression. Future research into the antifibrotic properties of SPMs and their impact on innate immune cell regulation holds promise for novel therapeutic strategies in IPF treatment.

Full Article: https://academic.oup.com/jleukbio/article-abstract/117/7/qiaf100/8182905?redirectedFrom=fulltext

Resolution of innate immune cells with proresolving lipid mediators in idiopathic pulmonary fibrosis Read More »

Exploring experiences of the regulatory toxicology system: system-level promoters and inhibitors of new approach methodologies

 

Abstract

The transition from traditional animal-based approaches and assessments to New Approach Methodologies (NAMs) marks a scientific revolution in regulatory toxicology, with the potential of enhancing human and environmental protection. However, implementing the effective use of NAMs in regulatory toxicology has proven to be challenging, and so far, efforts to facilitate this change frequently focus on singular technical, psychological or economic inhibitors. This article takes a system-thinking approach to these challenges, a holistic framework for describing interactive relationships between the components of a system of interest. In this case, the regulatory toxicology system. We do so by analysing and interpreting a very large qualitative data set of experts’ observations, collected in a 3-day interactive workshop and three follow-up online workshops with a heterogeneous sample of experts representing major actors from the global regulatory toxicology system. We identified leverage points (where a small change within a system can have a disproportionately large effect) in the six core aspects—infrastructure, processes, culture, technology, goals, and actors—in the regulatory toxicology system to facilitate the effective use of NAMs. Identified systematic leverage points include the need for a functioning incentive structure for effectively discovering, developing, validating and using NAMs within academia, regulation, and industry; and measures that prevent or mitigate unwanted effects of using NAMs that acknowledge clashes between scientific, regulatory, political and social processes. The results serve as a basis for follow-up activities that reflect on the actual effectiveness of these levers and that develop measures for the regulatory toxicology system.

Exploring experiences of the regulatory toxicology system: system-level promoters and inhibitors of new approach methodologies Read More »

Apoptosis-targeting BH3 mimetics: transforming treatment for patients with acute myeloid leukaemia

Abstract

Acute myeloid leukaemia (AML) remains a challenging haematological malignancy, with most patients developing resistance to standard-of-care (SOC) treatments. This resistance is often attributed to the overexpression of anti-apoptotic BCL-2 family proteins, which regulate the intrinsic apoptotic pathway by inhibiting pro-apoptotic effector proteins such as BAX and BAK. AML cells exploit this imbalance to evade apoptosis and sustain survival, necessitating the development of novel therapeutic strategies. BH3 mimetics are small-molecule inhibitors targeting the pro-survival BCL-2 family proteins and have emerged as promising agents in patients with AML who are unable to receive high-intensity induction chemotherapy. Co-treatment with the BCL-2-specific inhibitor venetoclax and various SOC therapies has been proven effective, with several combinations now approved by the US Food and Drug Administration for adults with AML who are ≥75 years of age and/or are ineligible for intensive induction chemotherapy, on the basis of improved response rates and survival outcomes compared with the previous SOC. In this Review, we highlight the transformative potential of BH3 mimetics in AML therapy, including ongoing studies investigating novel combination regimens and efforts to further refine treatment strategies, with the ultimate goal of improving outcomes for patients with AML.

Full Article: https://www.nature.com/articles/s41571-025-01068-0

Apoptosis-targeting BH3 mimetics: transforming treatment for patients with acute myeloid leukaemia Read More »

In vivo CRISPR screening identifies POU3F3 as a novel regulator of ferroptosis resistance in hepatocellular carcinoma via retinoic acid signaling

Abstract

Background

Sorafenib, a ferroptosis agonist, is a first-line treatment for advanced hepatocellular carcinoma (HCC). However, its clinical efficacy is limited due to drug resistance, resulting in modest improvements in patient survival. Hence, the present study has been designed to identify critical molecular targets associated with sorafenib resistance and investigate the potential inhibitors in overcoming this therapeutic challenge.

Methods

In vivo whole-genome CRISPR/Cas9 library screens were conducted to identify resistance factors to ferroptosis agonists, such as RSL3 and sorafenib, in HCC. The effects and underlying molecular mechanisms of these resistance factors were investigated in HCC cells using ferroptosis detection assays, xenograft tumor models, chromatin immunoprecipitation (ChIP), and dual-luciferase reporter assays. Potential inhibitors targeting these factors were evaluated through computer-aided virtual screening, molecular dynamics simulations, surface plasmon resonance analysis, and functional evaluations.

Results

A retinoic acid metabolism gene cluster, including ADH4, ALDH1A1, ALDH1A3, FABP5, RBP1, and RDH10, was found demonstrating upregulation in HCC cells treated with ferroptosis agonist, sorafenib. This gene cluster contributes to the ferroptosis resistance by producing the strong reducing agent retinoic acid. The transcription factor POU3F3 was identified as a key regulator for the retinoic acid metabolism gene cluster, which simultaneously binds to their promoters, increasing their transcription and promoting retinoic acid production. Knockdown of POU3F3 significantly enhanced the pro-ferroptotic and inhibitory effects of sorafenib on HCC cells by suppressing retinoic acid metabolism. Furthermore, rosarin was identified as a POU3F3 inhibitor, with an equilibrium dissociation constant of 7.57 µM, and demonstrated a synergistic effect with sorafenib against HCC cells both in vitro and in vivo.

Conclusions

According to the results, POU3F3 acts as a protective regulator against sorafenib-induced ferroptosis in HCC cells by enhancing the transcription of multiple retinoic acid metabolism genes and promoting retinoic acid production. The POU3F3 inhibitor, rosarin, shows potential as an ideal candidate for overcoming sorafenib resistance in HCC.

Full Article: https://biosignaling.biomedcentral.com/articles/10.1186/s12964-025-02285-x

In vivo CRISPR screening identifies POU3F3 as a novel regulator of ferroptosis resistance in hepatocellular carcinoma via retinoic acid signaling Read More »

A Metabolism-Oriented Strategy to Directly Generate Photosensitizer-Engineered Extracellular Vesicles from Cancer Cells

Abstract

Extracellular vesicles (EVs) hold great potential for delivering cancer therapy drugs. However, limited efficiency and sophisticated drug encapsulation procedures have hindered their effectiveness. Herein, β-D-glucose is modified with the synthesized photosensitizer (1-(4-carboxybutyl)-4-(7-(4-(diphenylamino)phenyl)benzo[c][1,2,5] thiadiazol-4-yl)pyridin-1-ium, named TB) via amide bond to form a glucose-conjugated photosensitizer, referred to as TBG, which is further utilized as a metabolic substrate for cancer cells. Through simple co-incubation with TBG, cancer cells directly generate TBG-engineered EVs in situ via a metabolism-driven process, in which glucose transporters play a critical role. Notably, a higher yield of engineered EVs is observed in TBG-treated cells compared to the TB-treated group. This enhancement could be attributed to increased glucose transporter activity and adenosine triphosphate (ATP) synthesis, highlighting the significance of glucose-modified chemicals. Remarkably, this metabolism-driven strategy has been successfully validated across three cell lines, highlighting its versatility and broad applicability. The extracted TBG-EVs maintain a strong targeting ability toward cancer cells and demonstrate enhanced efficacy in photodynamic therapy for tumor ablation. The study offers an alternative strategy to efficiently produce cargo-loading EVs via direct biological metabolism.

Full Article: https://doi.org/10.1002/adma.202505726

A Metabolism-Oriented Strategy to Directly Generate Photosensitizer-Engineered Extracellular Vesicles from Cancer Cells Read More »

The application of organoids in treatment decision-making for digestive system cancers: progress and challenges

Abstract

Digestive system cancers—including gastric, liver, colorectal, esophageal, and pancreatic malignancies—remain leading causes of cancer death, with treatment resistance posing major challenges in advanced disease. Patient-derived cancer organoids (PDCOs), 3D mini-tumors grown from patient biopsies, have revolutionized personalized oncology by faithfully replicating tumor biology and enabling predictive drug testing for chemotherapy, radiotherapy, targeted therapy, and immunotherapy. While demonstrating good predictive accuracy, current limitations include incomplete tumor microenvironments, variable establishment rates, and lengthy processing times. Emerging technologies like AI, organ-on-chip systems, and 3D bioprinting are addressing these challenges, while clinical trials explore applications in neoadjuvant therapy and real-time treatment guidance. This Review highlights key advances in PDCO technology and its transformative potential for treatment decision-making in digestive system cancers, bridging laboratory research with clinical care to enable truly personalized therapeutic strategies tailored to individual tumor biology.

Full Article: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-025-02429-0

The application of organoids in treatment decision-making for digestive system cancers: progress and challenges Read More »

Unlocking therapeutic potential: Exploring nuclear receptors in brain cancer treatment

Abstract

Brain cancer remains among the most lethal malignancies worldwide, with approximately 321,476 new cases and 248,305 deaths reported globally in 2022. The treatment of malignant brain tumors presents substantial clinical challenges, primarily due to their resistance to standard therapeutic approaches. Despite decades of intensive research, effective treatment strategies for brain cancer are still lacking. Nuclear receptors (NRs), a superfamily of ligand-activated transcription factors, regulate a broad range of physiological processes including metabolism, immunity, stress response, reproduction, and cellular differentiation. Increasing evidence highlights the involvement of NRs in oncogenesis, with several members demonstrating altered expression and function in brain tumors. Aberrations in NR signaling, encompassing receptors such as androgen receptors, estrogen receptors, estrogen-related receptors, glucocorticoid receptors, NR subfamily 4 group A, NR subfamily 1 group D member 2, NR subfamily 5 group A member 2, NR subfamily 2 group C member 2, liver X receptors, peroxisome-proliferator activated receptors, progesterone receptors, retinoic acid receptors, NR subfamily 2 group E member 1, thyroid hormone receptors, vitamin D receptors, and retinoid X receptors, have been implicated in promoting hallmark malignant phenotypes, including enhanced survival, proliferation, invasion, migration, metastasis, and resistance to therapy. This review aims to explore the roles of key NRs in brain cancer, with an emphasis on their prognostic significance, and to evaluate the therapeutic potential of targeting these receptors using selective agonists or antagonists.

Unlocking therapeutic potential: Exploring nuclear receptors in brain cancer treatment Read More »

Next-generation immunotherapeutic approaches for blood cancers: Exploring the efficacy of CAR-T and cancer vaccines

Abstract

Recent advancements in immunotherapy, particularly Chimeric antigen receptor (CAR)-T cell therapy and cancer vaccines, have significantly transformed the treatment landscape for leukemia. CAR-T cell therapy, initially promising in hematologic cancers, faces notable obstacles in solid tumors due to the complex and immunosuppressive tumor microenvironment. Challenges include the heterogeneous immune profiles of tumors, variability in antigen expression, difficulties in therapeutic delivery, T cell exhaustion, and reduced cytotoxic activity at the tumor site. Additionally, the physical barriers within tumors and the immunological camouflage used by cancer cells further complicate treatment efficacy. To overcome these hurdles, ongoing research explores the synergistic potential of combining CAR-T cell therapy with cancer vaccines and other therapeutic strategies such as checkpoint inhibitors and cytokine therapy. This review describes the various immunotherapeutic approaches targeting leukemia, emphasizing the roles and interplay of cancer vaccines and CAR-T cell therapy. In addition, by discussing how these therapies individually and collectively contribute to tumor regression, this article aims to highlight innovative treatment paradigms that could enhance clinical outcomes for leukemia patients. This integrative approach promises to pave the way for more effective and durable treatment strategies in the oncology field. These combined immunotherapeutic strategies hold great promise for achieving more complete and lasting remissions in leukemia patients. Future research should prioritize optimizing treatment sequencing, personalizing therapeutic combinations based on individual patient and tumor characteristics, and developing novel strategies to enhance T cell persistence and function within the tumor microenvironment. Ultimately, these efforts will advance the development of more effective and less toxic immunotherapeutic interventions, offering new hope for patients battling this challenging disease.

Full Article: https://doi.org/10.1186/s40164-025-00662-3

Next-generation immunotherapeutic approaches for blood cancers: Exploring the efficacy of CAR-T and cancer vaccines Read More »

TGF-β-driven EMT in cancer progression and drug resistance

Abstract

The carcinogenesis and drug resistance can be accelerated by TGF-β, primarily by enhancing epithelial-mesenchymal transition (EMT). This review examines the complex mechanisms by which TGF-β drives EMT across different tumors, highlighting its function in increasing cellular plasticity, promoting metastasis, and contributing to therapy resistance. TGF-β activates both canonical Smad-dependent and non-canonical signaling, leading to profound changes in cell morphology, motility, and stemness. This review highlights recent discoveries on how TGF-β regulates cancer stem cells and contributes to drug resistance, including resistance to both conventional chemotherapy and targeted treatments. In addition, it examines the intricate interaction between TGF-β and the key molecular pathways controlling EMT, such as PI3K/AKT, MAPK, and epigenetic regulators. It also examines potential therapeutic approaches aimed at TGF-β-induced EMT, emphasizing promising preclinical results from novel compounds and combination therapies—including natural products, small-molecule inhibitors, and epigenetic regulators—that interfere with TGF-β receptor activation or downstream signaling pathways. Understanding these complex interactions provides valuable insights for developing more effective cancer therapies. The review concludes by identifying key research gaps as well as suggesting future directions for investigating TGF-β’s role in cancer biology and treatment resistance.

Full Article: https://doi.org/10.1016/j.cytogfr.2025.05.004

TGF-β-driven EMT in cancer progression and drug resistance Read More »

Autophagy paradox: Genetic and epigenetic control of autophagy in cancer progression

 

Abstract

Autophagy is a highly regulated, evolutionarily conserved process of self-digestion controlled by autophagy-related (ATG) genes. It involves the lysosomal degradation of cargoes, including cytoplasmic organelles, misfolded proteins, and toxic aggregates, to enrich cellular nutrient pools and reduce oxidative stress. In normal cells, basal autophagy occurs to maintain cellular homeostasis, which changes during tumor initiation, progression, and malignant transformation. The alteration in autophagy in cancer remains unclear and under-explored. Research indicates that genetic regulations, such as gene mutations, gene polymorphisms, or epigenetic modifications, including DNA methylation, histone modification, microRNAs (miRNAs), and long non-coding RNAs (lncRNAs), regulate ATGs, orchestrating the fluctuating nature of autophagy in cancer. Many studies describe the paradoxical role of autophagy in cancer, portraying it as a double-edged sword depending on the context, oscillating between promoting cell survival and inducing cell death-the dual roles in preventing tumor initiation and supporting tumor progression place autophagy at the centre of controversy. Recent findings suggest that autophagy is regulated at the intrinsic cellular level and within the tumor microenvironment. Thus, identifying the molecules, mediators, and mechanisms associated with the regulation of autophagy during tumor development, maintenance, therapy resistance, and dormancy could open new research avenues to enhance the efficacy of cancer therapeutics. Furthermore, this review encompasses preclinical studies and clinical trials, highlighting the effectiveness of modulating autophagy in cancer therapy.

Autophagy paradox: Genetic and epigenetic control of autophagy in cancer progression Read More »

Scroll to Top