• All Solutions All Solutions Caret
    • Editage

      One platform for all researcher needs

    • Paperpal

      AI-powered academic writing assistant

    • R Discovery

      Your #1 AI companion for literature search

    • Mind the Graph

      AI tool for graphics, illustrations, and artwork

    • Journal finder

      AI-powered journal recommender

    Unlock unlimited use of all AI tools with the Editage Plus membership.

    Explore Editage Plus
  • Support All Solutions Support
    discovery@researcher.life
Discovery Logo
Paper
Search Paper
Cancel
Ask R Discovery Chat PDF
Explore

Feature

  • menu top paper My Feed
  • library Library
  • translate papers linkAsk R Discovery
  • chat pdf header iconChat PDF
  • audio papers link Audio Papers
  • translate papers link Paper Translation
  • chrome extension Chrome Extension

Content Type

  • preprints Preprints
  • conference papers Conference Papers
  • journal articles Journal Articles

More

  • resources areas Research Areas
  • topics Topics
  • resources Resources

Immunotherapy Resistance Research Articles

  • Share Topic
  • Share on Facebook
  • Share on Twitter
  • Share on Mail
  • Share on SimilarCopy to clipboard
Follow Topic R Discovery
By following a topic, you will receive articles in your feed and get email alerts on round-ups.
Overview
1336 Articles

Published in last 50 years

Related Topics

  • Therapeutic Resistance
  • Therapeutic Resistance
  • Therapy Resistance
  • Therapy Resistance
  • Acquired Resistance
  • Acquired Resistance

Articles published on Immunotherapy Resistance

Authors
Select Authors
Journals
Select Journals
Duration
Select Duration
1057 Search results
Sort by
Recency
Tumor-intrinsic ENO1 inhibition promotes antitumor immune response and facilitates the efficacy of anti-PD-L1 immunotherapy in bladder cancer

Abstract Immunotherapy has revolutionized cancer treatment, yet understanding immunotherapy resistance mechanisms remains challenging. Here, a CRISPR cas9 screening in vivo and an RNA-sequencing for clinical immunotherapy resistance BC samples identified enolase 1 (ENO1) as a potent regulator of anti-PD-L1 treatment efficacy. Investigation of clinical BC samples demonstrated a correlation between ENO1 overexpression and immune evasion in BC, evidenced by reduced CD8+ T cell infiltration and resistance to anti-PD-L1 therapy. Increased CD8+ T cell infiltration and function were indicative of antitumor immunity, which was elicited by ENO1 knockdown, which also suppressed carcinogenesis. Single-cell RNA sequencing demonstrated that wild-type (WT) and ENO1 knockout (KO) tumors have different immune cell compositions with the latter preferring an immunostimulatory microenvironment. Mechanistically, ENO1 regulated CD8+ T cell function and tumor-associated macrophage (TAM) polarization via the SPP1-ITGA4/ITGB1 pathway in the TME. Importantly, genetic and pharmacological inhibition of ENO1 sensitizes tumors to anti-tumor immunity and synergizes with anti-PD-L1 therapy. The results highlight tumor-intrinsic ENO1 as a critical regulator of tumor immune evasion in BC. Targeting ENO1 enhance the efficacy of immune checkpoint blockade therapy by promoting antitumor immunity.

Read full abstract
  • Journal IconJournal of Experimental & Clinical Cancer Research
  • Publication Date IconJul 15, 2025
  • Author Icon Chengquan Shen + 5
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Abstract B045: Deep learning–guided spatial dissection of melanoma uncovers compartmentalized tumor states associated with response and resistance to immunotherapy and its combination with MAPK inhibitors

Abstract Purpose: Intratumoral heterogeneity is a key determinant of immunotherapy resistance, yet current biomarkers largely rely on single-modality features—such as CD8+ T cell infiltration, IFNG expression, or checkpoint ligand staining—without accounting for the spatial complexity of the tumor microenvironment. Composite biomarkers that integrate both tumor-intrinsic programs and their spatial organization remain underdeveloped. To address this, we combined machine learning–based molecular classification with spatial transcriptomics to uncover how transcriptional tumor states are physically structured within the tumor architecture and how this organization governs response or resistance to immunotherapy. Methods: We leveraged non-negative matrix factorization (NMF) and tumor microenvirnoment metabolism signatures with bulk RNA-seq data from >700 melanoma tumors across multiple phase I–III clinical trials. These programs were mapped onto spatial transcriptomics data using Tangram 2.0, a deep learning model that aligns single-cell or deconvolved bulk data to spatial coordinates. We analyzed cell–cell interactions, tumor-stromal patterning, and radial gene gradients to interpret functional spatial architecture. Results: Spatial mapping revealed that melanoma tumors often harbor multiple transcriptional programs arranged in discrete, non-overlapping tissue regions. Notably, undifferentiated (UR) and differentiated (DC) programs—associated with distinct therapeutic responses—co-existed within the same tumor but remained compartmentalized. Only spatial domains with DC-like features exhibited MAPK inhibitor–induced MHC-I expression and immune cell infiltration, whereas UR regions remained immune-excluded and were enriched for fibroblasts and extracellular matrix remodeling. These findings suggest that spatial segregation of tumor states, rather than overall tumor composition, may predict response to immunotherapy. Conclusions: This study presents a machine learning–enabled spatial framework that links transcriptional identity to physical tissue structure in melanoma. By uncovering how spatial architecture constrains therapeutic response, our work highlights the value of AI-driven models like Tangram for interpreting spatial data and guiding precision immuno-oncology. Citation Format: Kalpit Shah. Deep learning–guided spatial dissection of melanoma uncovers compartmentalized tumor states associated with response and resistance to immunotherapy and its combination with MAPK inhibitors [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Artificial Intelligence and Machine Learning; 2025 Jul 10-12; Montreal, QC, Canada. Philadelphia (PA): AACR; Clin Cancer Res 2025;31(13_Suppl):Abstract nr B045.

Read full abstract
  • Journal IconClinical Cancer Research
  • Publication Date IconJul 10, 2025
  • Author Icon Kalpit Shah
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

CPLX1 is a novel prognostic biomarker in CRC correlating with immunotherapy resistance and ferroptosis

BackgroundColorectal cancer (CRC) remains a predominant contributor to cancer-related mortality globally, with its resistance to immunotherapeutic strategies presenting a formidable challenge in patient management. Recent investigations have illuminated the prospective involvement of ferroptosis, a regulated form of cell death, in both cancer progression and the development of resistance to therapeutic interventions.ObjectiveThis study aims to elucidate the prognostic significance of CPLX1 in CRC, specifically its correlation with immunotherapy resistance and its association with ferroptosis, thereby contributing to a deeper understanding of tumor biology and therapeutic vulnerability.MethodsWe conducted an integrative analysis of RNA-seq datasets from the TCGA-COAD and TCGA-READ projects, along with the GEO GSE156451 dataset, to discern differentially expressed genes. Expression levels of CPLX1 were evaluated utilizing the TIMER 2.0 database, and survival analyses were performed via Kaplan-Meier plots and Cox regression modeling to assess prognostic implications. Additionally, mutational analyses through cBioPortal and COSMIC datasets were employed to identify CPLX1 mutations in COAD. Co-expression and functional enrichment analyses, alongside Gene Set Enrichment Analysis (GSEA), were also conducted to delineate pathways impacted by CPLX1.ResultsOur findings indicate that high expression levels of CPLX1 are significantly correlated with poor prognostic outcomes in CRC patients. Through immune infiltration analyses employing ssGSEA, we observed notable associations between CPLX1 expression and specific immune cell populations. Furthermore, the interaction between CPLX1 and ferroptosis-related genes suggests a potential mechanistic linkage that could underpin therapeutic resistance.ConclusionCPLX1 is identified as a novel prognostic biomarker in CRC, exhibiting clear correlations with both immunotherapy resistance and ferroptosis. These findings indicate that targeting CPLX1 may provide novel therapeutic strategies to ameliorate treatment resistance in CRC.

Read full abstract
  • Journal IconFrontiers in Immunology
  • Publication Date IconJul 9, 2025
  • Author Icon Canyu Liu + 7
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Hypoxia-induced tumor cell-intrinsic PLAU activation drives immunotherapy resistance in collagenic lung adenocarcinoma.

Hypoxia-induced tumor cell-intrinsic PLAU activation drives immunotherapy resistance in collagenic lung adenocarcinoma.

Read full abstract
  • Journal IconInternational immunopharmacology
  • Publication Date IconJul 9, 2025
  • Author Icon Li Wan + 12
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Targeting p-FGFR1Y654 Enhances CD8+ T Cells Infiltration and Overcomes Immunotherapy Resistance in Esophageal Squamous Cell Carcinoma by Regulating the CXCL8–CXCR2 Axis

Background: Esophageal squamous cell carcinoma (ESCC) is a fatal malignant tumor. Several studies have demonstrated that immune checkpoint inhibitors can provide clinical benefits to patients with ESCC. However, the single-agent efficacy of these agents remains limited. Although combination therapies (e.g., radiotherapy, chemotherapy) can help to overcome immunotherapy resistance in ESCC, their severe side effects limit clinical application. This study aimed to explore new resistance mechanisms to immunotherapy in ESCC and identify novel molecular targets to overcome immunotherapy resistance. Methods: We employed immunohistochemistry staining to examine the p-FGFR1Y654 in tumor samples obtained from 103 patients with ESCC, in addition to evaluating CD8+ T cell infiltration. In vitro expression, western blotting, CCK-8, 5-bromo-2′-deoxyuridine incorporation assays, and migration assays were used to confirm the impact of AZD4547 on p-FGFR1Y654 expression and the proliferation and migration in ESCC cell lines. Through RNA sequencing analysis, databases such as the Cancer Genome Atlas (TCGA) and Gene Set Cancer Analysis (GSCA), and the reconstruction of transgenic mice using the humanized immune system, we validated the correlation between the expression of p-FGFR1Y654 and CD8+ T cell infiltration. We also explored how p-FGFR1Y654 recruits myeloid-derived suppressor cells (MDSCs) through the CXCL8–CXCR2 axis to suppress the therapeutic efficacy of immunotherapy in ESCC. Finally, the tumor-suppressive effects of AZD4547 combined with immunotherapy were confirmed in vivo in tumor-bearing mice with a humanized immune system. Results: We found that the inhibition of p-FGFR1Y654 expression in ESCC can enhance CD8+ T cell infiltration by suppressing the CXCL8-–XCR2 recruitment of MDSCs. AZD4547, combined with immunotherapy, further promotes immunotherapeutic efficacy in ESCC. Conclusions: In conclusion, our study presents a promising model for combination therapy in ESCC immunotherapy.

Read full abstract
  • Journal IconBiomedicines
  • Publication Date IconJul 8, 2025
  • Author Icon Hong Luo + 15
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Why Cancer Immunotherapy Fails?: Mechanisms of Resistance and Emerging Reversal Strategies

Cancer immunotherapy has revolutionized the field of oncology by enabling the immune system to target and destroy cancer cells. Therapies such as immune checkpoint inhibitors, including PD-1/PD-L1 and CTLA-4 blockade, and chimeric antigen receptor T (CAR-T) cells have shown unprecedented clinical efficacy, especially in melanoma, non-small cell lung cancer, and hematologic malignancies. These advances have transformed treatment paradigms, leading to significant improvements in outcomes for some patients. However, a major challenge remains: a substantial proportion of patients exhibit primary resistance or relapse after initial response, limiting the broader success of these therapies. In this review, we highlight cancer-intrinsic, immune microenvironment-related, and host-related mechanisms that underlie immunotherapy resistance, including tumor antigen loss, MHC downregulation, immunosuppressive microenvironments, and host factors such as microbiome composition. We also discuss emerging strategies to overcome resistance, such as combination therapies (e.g., checkpoint inhibitors with targeted agents or chemotherapy), novel immune checkpoint targets (e.g., LAG-3, TIM-3, TIGIT), and personalized immunotherapies including neoantigen vaccines. By breaking down the complex biology of resistance and highlighting innovative approaches under development, this review aims to guide future research and clinical efforts toward more durable and widely applicable immunotherapy responses.

Read full abstract
  • Journal IconJournal of Cancer and Tumor International
  • Publication Date IconJul 8, 2025
  • Author Icon Muhydeen Damilola Badru + 8
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Tumor-associated bacteria activate PRDX1-driven glycolysis to promote immune evasion and PD-1 antibody resistance in hepatocellular carcinoma

BackgroundRecent studies have highlighted the presence of intratumoral bacteria in hepatocellular carcinoma (HCC), yet their contribution to immunotherapy resistance remains largely unexplored. This study investigates the mechanisms by which bacterial infection reshapes tumor metabolism to undermine the efficacy of anti-PD-1 therapy.MethodsWe conducted 16S rRNA gene sequencing on 29 HCC clinical samples and integrated the data with single-cell RNA sequencing of 12,487 cells to map microbial, metabolic, and immune interactions within the tumor microenvironment. Functional validation was performed using orthotopic HCC mouse models (n = 8 per group), coupled with flow cytometry-based immune profiling.ResultsEnrichment of Streptococcaceae was strongly associated with upregulation of key glycolytic enzymes (LDHA, PKM2; p < 0.001) and dysfunction of natural killer cells (reduced CD56dim/CD16bright populations; hazard ratio = 2.15, 95% CI: 1.34–3.42). Mechanistically, bacterial colonization induced peroxiredoxin 1 (PRDX1) expression via the NF-κB pathway. This led to excessive lactate production, which suppressed CD8+ T cell cytotoxicity (p = 0.003) and increased the expression of immune checkpoint molecules (TIM-3: 2.7-fold; LAG-3: 1.9-fold). In vivo, bacterial infection decreased the antitumor efficacy of PD-1 blockade by 43% (tumor volume vs. control; p = 0.008), an effect that was reversed upon PRDX1 inhibition.ConclusionOur findings identify PRDX1 as a central node in bacteria-driven metabolic reprogramming that facilitates immune evasion and resistance to PD-1 therapy in HCC. These findings provide the first evidence linking intratumoral bacteria to PD-1 resistance via redox-regulated metabolism, proposing dual targeting of PRDX1 and gut microbiota as a novel combinatorial immunotherapy strategy.

Read full abstract
  • Journal IconFrontiers in Microbiology
  • Publication Date IconJul 7, 2025
  • Author Icon Heng Zhang + 8
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Recent advances in tumour microenvironment impact immunotherapy resistance in gastric cancer.

Recent advances in tumour microenvironment impact immunotherapy resistance in gastric cancer.

Read full abstract
  • Journal IconCritical reviews in oncology/hematology
  • Publication Date IconJul 4, 2025
  • Author Icon Qiuhong Sun + 4
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Engineered Bacteria-Nanoparticle Conjugate Reprograms Immunosuppressive Niche via Dendritic Cell-Centric Innate-Adaptive Immune Coupling.

The immunosuppressive tumor microenvironment (TME) represents a critical barrier to effective T cell activation, contributing to immunotherapy resistance. Dendritic cells (DCs), essential initiators of T cell-mediated adaptive immunity, frequently display both quantitative defects and functional impairments within the TME. Compounding this challenge, emerging evidence highlights the indispensable role of innate immunity in sustaining T cell activity and establishing durable immunological memory. To address these limitations, we engineered M-CHNP/D: a bacteria-nanoparticle hybrid platform integrating Escherichia coli with acid-responsive calcium carbonate nanoparticles encapsulating the programmed cell death ligand-1 blocking peptide DPPA-1. Leveraging the motility and targeting capabilities of bacteria, M-CHNP/D achieves a deep tumor penetration and neutralizes the acidic TME. M-CHNP/D induced tumor-cell-derived CCL3 upregulation, driving DC recruitment and spatial redistribution within the tumor parenchyma. This intervention enhanced DCs' antigen-presenting capacity, ultimately potentiating adaptive immune responses. Furthermore, M-CHNP/D administration significantly increased the population of innate immune cells and induced their phenotypic reprogramming toward antitumor functional states. By reshaping the immune "hot" network, M-CHNP/D combined with radiotherapy effectively inhibited tumor growth and recurrence. M-CHNP/D demonstrates significant potential in modulating both innate and adaptive immunity, offering a robust strategy for improving cancer immunotherapy outcomes.

Read full abstract
  • Journal IconACS nano
  • Publication Date IconJul 2, 2025
  • Author Icon Xiaoxi Wang + 15
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

NGR-modified curcumin nanovesicles reverse immunotherapy resistance in triple-negative breast cancer via TLR9 and mTOR pathway modulation

Curcumin (Cur), a natural bioactive compound extracted from Curcuma longa, has garnered extensive interest due to its modulation of inflammation, antioxidant, and anti-tumor properties. However, its therapeutic translation remains constrained by limited systemic bioavailability. Triple-negative breast cancer (TNBC), an aggressive variant of breast malignancies, exhibits strong resistance to conventional therapies and poor prognosis. The present study was designed to clarify the mechanism through which NGR-modified nanovesicles loaded with Cur (NGR-NVs@Cur) reverse immunotherapy resistance in TNBC. Using transcriptomic and network pharmacology analysis, we identified key genes involved in TNBC development and immunotherapy resistance to determine the targets of Cur. In vitro experiments, including SA-β-gal staining, flow cytometry, and glycolysis analysis, validated that TNBC cells induce glycolysis and CD8+ T cell senescence. NGR-NVs@Cur were successfully constructed and marked by transmission electron microscopy (TEM), dynamic light scattering (DLS), pH-responsive release, and cellular uptake assays. Further cell-based studies demonstrated that NGR-NVs@Cur suppressed TNBC cell proliferation, migration, glycolysis, and reversed CD8+ T cell senescence. In vivo, both subcutaneous xenograft and adoptive T cell transfer models were developed to evaluate the therapeutic effects of NGR-NVs@Cur in combination with immune checkpoint inhibitors (ICIs, e.g., J43). The results revealed that Cur inhibited TNBC cell glycolysis and T cell senescence by activating TLR9 and suppressing the mTOR pathway, and that NGR-NVs@Cur enhanced targeted Cur delivery and effectively reversed immunotherapy resistance. This study demonstrated a novel strategy by which Cur, delivered via tumor-targeted nanovesicles, modulates glycolysis and CD8+ T cell senescence through the TLR9–mTOR axis, offering promising insights into overcoming immune resistance in TNBC.

Read full abstract
  • Journal IconCell Biology and Toxicology
  • Publication Date IconJul 1, 2025
  • Author Icon Shuo Wang + 6
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Tumor site influences efficacy of MEKi and immunotherapy combinations in pre-clinical model of cholangiocarcinoma.

Although chemotherapy and anti-PD-L1 antibodies are the standard of care for cholangiocarcinoma (CCA), resistance is common and limits durable benefits for patients. This hurdle underscores the urgent need to innovate combination approaches that promote durable immunity in patients. Mek inhibitors (MEKi) have shown potential to enhance immunotherapy in CCA models, but early clinical trials combining MEKi with anti-PD-L1 therapy have yielded suboptimal results. We hypothesized that addition of CD27 agonist would salvage MEKi-induced T cell impairment and reduce CCA burden in vivo. We show CD27 agonism potentiates T cell activation in the presence of MEKi in vitro. Further, triple therapy with CD27 agonist, anti-PD-L1, and MEKi elicit efficacy in a subcutaneous CCA model, accompanied by increases in tumor-infiltrating CD8+ T cells with memory phenotypes. Although triple therapy enhanced CD8+ T cell infiltration in mice with orthotopic CCA liver tumors, its impact on overall survival was less pronounced. Further investigation revealed orthotopic CCA tumors harbored more CD11b+Gr-1+ cells when compared to either subcutaneous CCA tumors, or to normal liver. Finally, in mice with orthotopic CCA tumors treated with triple therapy, depletion of Gr-1+ cells triggered severe toxicity, characterized by rapid weight loss and uncontrolled systemic and hepatic inflammation. These findings identify myeloid cells as key mediators of both immunotherapy resistance and toxicity in CCA, highlighting the critical importance of tumor site and use of physiologically relevant pre-clinical models when evaluating immunotherapy strategies.

Read full abstract
  • Journal IconHepatology (Baltimore, Md.)
  • Publication Date IconJul 1, 2025
  • Author Icon Jacklyn Hammons + 19
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment.

Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment.

Read full abstract
  • Journal IconBiochimica et biophysica acta. Reviews on cancer
  • Publication Date IconJul 1, 2025
  • Author Icon Lu Liu + 2
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Riplet promotes lipid metabolism changes associated with CD8 T cell exhaustion and anti-PD-1 resistance in hepatocellular carcinoma.

The overall response rate to immunotherapy is modest in hepatocellular carcinoma (HCC), and immunotherapy resistance mechanisms are incompletely understood. We report that the E3 ubiquitin ligase Riplet is universally silenced by promoter hypermethylation in HCC. Loss of Riplet modulates fatty acid metabolism to promote terminal exhaustion of CD8 T cells. Riplet loss impedes K48-linked polyubiquitination of fatty acid synthase (FASN), consequently accelerating fatty acid production in HCC. Tumor cell-derived free fatty acids, especially palmitic acid (PA/C16:0), activate STAT3 (signal transducers and activators of transcription 3) by enhancing its palmitoylation in T cells, consequently triggering terminal CD8 T cell exhaustion. HCC cells with Riplet deficiency are resistant to anti-PD-1 therapy, and treatment with an FASN inhibitor overcomes resistance. Our study shows how Riplet can alter lipid metabolism and induce CD8 T cell exhaustion and anti-PD-1 resistance, thus suggesting avenues for combined therapies for treating patients with Riplet-deficient HCC.

Read full abstract
  • Journal IconScience immunology
  • Publication Date IconJun 27, 2025
  • Author Icon Junnan Liang + 22
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Targeting Annexin A2 to reactivate tumor-associated antigens presentation and relieve immune tolerance in liver cancer.

Tumor cells manipulate the tumor-associated antigens presentation to escape immune surveillance; however, the molecular mechanism is not exactly clear and the measure to intervene is missing. Annexin A2 was knockout by the CRISPR-Cas9 or blocked by the small-molecule matrine, PY60, and hexapeptide. Chemically and genetically induced primary liver cancer models, and the orthotopically implanted liver tumor model were used. Tumor immune environment was analyzed by single-cell sequencing. Annexin A2-interacted proteins and tumor-associated antigens were identified by co-immunoprecipitation coupled with liquid chromatography with tandem mass spectrometry. Tumor cells killing effects were evaluated by co-culture of tumor cells and CD8+ T cells. Targeting Annexin A2 effectively suppressed the progression of liver cancer. The immunosuppressive microenvironment was improved by Annexin A2 inhibition in tumor tissues. The CD8+ T cells were increased and activated by targeting Annexin A2. Mechanistically, targeting Annexin A2 inhibited its combination with HSP90. The HSP90-mediated tumor-associated antigens presentation was recovered, and the major histocompatibility complex I-presented short peptides were changed, increasing the tumor cells killing by CD8+ T cells. Interestingly, Annexin A2 was increased in liver cancer tissues and the overall survival was significantly reduced in patients with high expression. However, Annexin A2 was positively correlated with immune cell infiltration in liver cancer, implying that Annexin A2 was used by tumor cells for immune escape and immunotherapy resistance. Hence, we further confirmed that blocking Annexin A2 increased the therapeutic effects of anti-programmed cell death protein-1 both in vitro and in vivo. Taken together, our results identified the role of Annexin A2 in the tumor-associated antigens presentation and immune evasion, which could be an actionable target in cancer immunotherapy.

Read full abstract
  • Journal IconJournal for immunotherapy of cancer
  • Publication Date IconJun 26, 2025
  • Author Icon Yuxiao Tang + 17
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

CAF-derived LRRC15 orchestrates macrophage polarization and limits PD-1 immunotherapy efficacy in glioblastoma.

The effectiveness of PD-1/PD-L1 immune checkpoint blockade therapy in glioblastoma (GBM) is limited due to the tumor immunosuppressive microenvironment (TIME). Therefore, strategies of reprogramming TIME to a proinflammatory state offers a promising therapeutic approach. We applied bioinformatics analysis of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (stRNA-seq) to identify a significant accumulation of a cancer-associated fibroblasts (CAFs) subcluster with elevated LRRC15 expression in the nonresponders to anti-PD-1 therapy. Molecular mechanism of LRRC15 were functionally validated in vitro and in vivo. These CAFs subcluster drive the infiltration of macrophages (Mφ) into the tumor microenvironment and promote their polarization toward the M2 phenotype. Deletion of Lrrc15 in CAFs significantly restrained tumor growth and prolonged survival in mouse models. Mechanistically, LRRC15 in CAFs promotes IL8 expression by activating the downstream FAK/SRC/NF-κB pathways, leading to Mφ migration and M2-like polarization. In turn, M2-like Mφs secrete TGF-β, which induces LRRC15 expression in CAFs via SMAD2-dependent transcriptional activation. Targeting CAFs subcluster with elevated LRRC15 expression in combination with anti-PD-1 treatment enhanced antitumor efficacy. Our findings suggest that targeting LRRC15 may provide a novel strategy to augment anti-PD-1 therapy and overcome immunotherapy resistance in GBM.

Read full abstract
  • Journal IconNeuro-oncology
  • Publication Date IconJun 26, 2025
  • Author Icon Feifei Luo + 10
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Multi-omic analysis of gallbladder cancer identifies distinct tumor microenvironments associated with disease progression.

Gallbladder carcinoma (GBC) is the most aggressive biliary tract cancer and is associated with a high mortality rate. Treatment of GBC faces therapeutic challenges owing to the elusive nature of in situ drivers within the local tumor microenvironment that drive its progression. Here, we created a single-cell atlas of 1,117,245 cells and a mutational landscape from 102 patients, which unveiled spatial-temporal characterizations of cellular constitutions, spatial interplays and molecular functions, and generalized five local ecosystems stratifying clinical outcomes. An integrated epithelial program, AI-EPI, combined with spatial transcriptome analysis, revealed the concurrent localization of a highly malignant tumor subtype (GM16) and AREG+ T cell, B cell, dendritic cell and macrophage subtypes within the pro-metastatic niche of primary adenocarcinomas. In vitro and in vivo experiments suggest that in addition to promoting metastasis, AREG facilitates CXCL5 expression in tumor cells through EGFR-pERK-EGR1 signaling, leading to increased neutrophil infiltration and impeding the effectiveness of immunotherapy. Our study provides a spatial-temporal landscape of the GBC microenvironment and sheds light on potential strategies for preventing immunotherapy resistance.

Read full abstract
  • Journal IconNature genetics
  • Publication Date IconJun 26, 2025
  • Author Icon Tao Zhou + 20
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Constitutive expression of the transcriptional co-activator IκBζ promotes melanoma growth and immunotherapy resistance

IκBζ, a rather unknown co-regulator of NF-κB, can either activate or repress a subset of NF-κB target genes. While its role as an inducibly expressed, transcriptional regulator of cytokines and chemokines in immune cells is established, IκBζ’s function in solid cancer remains unclear. Here we show that IκBζ protein is constitutively expressed in a subfraction of melanoma cell lines, and around 30% of all melanoma cases, independently of its mRNA levels or known mutations. Deleting IκBζ in melanoma abrogates the activity and chromatin association of STAT3 and NF-κB, thereby reducing the expression of the pro-proliferative cytokines IL-1β and IL-6, thus impairing melanoma cell growth. Additionally, IκBζ suppresses Cxcl9, Cxcl10, and Ccl5 expression via HDAC3 and EZH2, which impairs the recruitment of NK and CD8+ T cells into the tumor, causing resistance to α-PD-1 immunotherapy in mice. Thus, tumor-derived IκBζ may serve as a therapeutic target and prognostic marker for melanoma with high tumor cell proliferation, cytotoxic T- and NK-cell exclusion, and unfavorable immunotherapy responses.

Read full abstract
  • Journal IconNature Communications
  • Publication Date IconJun 25, 2025
  • Author Icon Antonia Kolb + 19
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Leveraging liquid biopsy to uncover resistance mechanisms and guide personalized immunotherapy.

Leveraging liquid biopsy to uncover resistance mechanisms and guide personalized immunotherapy.

Read full abstract
  • Journal IconTranslational oncology
  • Publication Date IconJun 24, 2025
  • Author Icon Zeinab Dalloul + 4
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

Exploring the Role of DPF1 in Hepatocellular Carcinoma: Implications for Prognosis and Therapy.

Hepatocellular carcinoma (HCC) is a life-threatening cancer with rising incidence and mortality rates. Identifying new prognostic biomarkers is crucial for improving HCC management. This study investigates the role of Double PHD Fingers 1 (DPF1) in hepatocellular carcinoma (HCC), exploring its potential as a prognostic indicator and therapeutic target. We analyzed DPF1 expression in 374 hepatocellular carcinoma (HCC) tissues and 50 normal tissues from the TCGA-HCC database, as well as in 240 HCC tissues and 202 normal tissues from the ICGC-HCC repository. We examined the correlation between DPF1 expression and clinical parameters, immune cell infiltration, drug response profiles, cancer stem cell (CSC) characteristics, and its diagnostic/prognostic potential using various bioinformatics tools and statistical analyses. Validation was performed using the ICGC and HPA databases, and qRT-PCR was used to confirm DPF1 expression in HCC cell lines. DPF1 exhibited abnormal expression in HCC and several other malignancies. Elevated DPF1 levels were significantly associated with higher Alpha-fetoprotein (AFP) levels (p = 0.043) and poorer clinical outcomes, including diminished overall survival (OS) (p = 0.002), progression-free survival (PFS) (p = 0.018), and disease-specific survival (DSS) (p = 0.001). DPF1 expression was also linked to immune cell infiltration, immune checkpoint gene expression, drug sensitivity, and CSC characteristics. Notably, DPF1 was significantly overexpressed in HCC tissues and cell lines at both transcriptional and translational levels. Our study reveals that DPF1 is a novel prognostic biomarker in HCC, with potential implications for immunotherapy and drug resistance. Elevated DPF1 expression is associated with adverse clinical outcomes and may serve as a target for future therapeutic interventions in HCC.

Read full abstract
  • Journal IconCurrent medicinal chemistry
  • Publication Date IconJun 20, 2025
  • Author Icon Fan Yang + 6
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

NDRG1-Driven Lactate Accumulation Promotes Lung Adenocarcinoma Progression Through the Induction of an Immunosuppressive Microenvironment.

Lung adenocarcinoma (LUAD) is a leading cause of cancer-related mortality, with the tumor microenvironment (TME) playing a critical role in its progression. Metabolic reprogramming, particularly lactate accumulation, drives immune suppression within the TME. Utilizing single-cell RNA sequencing (scRNA-seq) of 30 LUAD samples, genome-wide association studies (GWAS) involving 29,863 patients and 55,586 controls, and clinical data from 220 LUAD patients, we identified N-Myc downstream-regulated gene 1 (NDRG1) as a key pathogenic gene in LUAD, strongly associated with tumor progression and poor prognosis. Mechanistic studies revealed that NDRG1 stabilizes lactate dehydrogenase A (LDHA) by inhibiting its ubiquitination, thereby enhancing glycolysis and promoting lactate accumulation. This process fosters immune suppression by inducing M2 macrophage polarization, impairing CD8+ T cell function, and upregulating immunosuppressive genes. Furthermore, histone H3K18 lactylation in macrophages exacerbates this immunosuppressive state. Clinically, elevated NDRG1 expression correlates with increased PD-L1 levels, a higher abundance of immunosuppressive macrophages, and reduced CD8+ T cell infiltration, contributing to immunotherapy resistance. Conversely, low NDRG1 expression is associated with enhanced CD8+ T cell infiltration and improved therapeutic outcomes. Preclinical studies demonstrated targeting NDRG1 suppresses tumor growth, alleviates immune suppression, and boosts anti-PD-L1 efficacy. These findings establish NDRG1 as a critical LUAD regulator and a promising immunotherapy target.

Read full abstract
  • Journal IconAdvanced science (Weinheim, Baden-Wurttemberg, Germany)
  • Publication Date IconJun 20, 2025
  • Author Icon Gujie Wu + 14
Just Published Icon Just Published
Cite IconCite
Chat PDF IconChat PDF
Save

  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • .
  • .
  • .
  • 10
  • 1
  • 2
  • 3
  • 4
  • 5

Popular topics

  • Latest Artificial Intelligence papers
  • Latest Nursing papers
  • Latest Psychology Research papers
  • Latest Sociology Research papers
  • Latest Business Research papers
  • Latest Marketing Research papers
  • Latest Social Research papers
  • Latest Education Research papers
  • Latest Accounting Research papers
  • Latest Mental Health papers
  • Latest Economics papers
  • Latest Education Research papers
  • Latest Climate Change Research papers
  • Latest Mathematics Research papers

Most cited papers

  • Most cited Artificial Intelligence papers
  • Most cited Nursing papers
  • Most cited Psychology Research papers
  • Most cited Sociology Research papers
  • Most cited Business Research papers
  • Most cited Marketing Research papers
  • Most cited Social Research papers
  • Most cited Education Research papers
  • Most cited Accounting Research papers
  • Most cited Mental Health papers
  • Most cited Economics papers
  • Most cited Education Research papers
  • Most cited Climate Change Research papers
  • Most cited Mathematics Research papers

Latest papers from journals

  • Scientific Reports latest papers
  • PLOS ONE latest papers
  • Journal of Clinical Oncology latest papers
  • Nature Communications latest papers
  • BMC Geriatrics latest papers
  • Science of The Total Environment latest papers
  • Medical Physics latest papers
  • Cureus latest papers
  • Cancer Research latest papers
  • Chemosphere latest papers
  • International Journal of Advanced Research in Science latest papers
  • Communication and Technology latest papers

Latest papers from institutions

  • Latest research from French National Centre for Scientific Research
  • Latest research from Chinese Academy of Sciences
  • Latest research from Harvard University
  • Latest research from University of Toronto
  • Latest research from University of Michigan
  • Latest research from University College London
  • Latest research from Stanford University
  • Latest research from The University of Tokyo
  • Latest research from Johns Hopkins University
  • Latest research from University of Washington
  • Latest research from University of Oxford
  • Latest research from University of Cambridge

Popular Collections

  • Research on Reduced Inequalities
  • Research on No Poverty
  • Research on Gender Equality
  • Research on Peace Justice & Strong Institutions
  • Research on Affordable & Clean Energy
  • Research on Quality Education
  • Research on Clean Water & Sanitation
  • Research on COVID-19
  • Research on Monkeypox
  • Research on Medical Specialties
  • Research on Climate Justice
Discovery logo
FacebookTwitterLinkedinInstagram

Download the FREE App

  • Play store Link
  • App store Link
  • Scan QR code to download FREE App

    Scan to download FREE App

  • Google PlayApp Store
FacebookTwitterTwitterInstagram
  • Universities & Institutions
  • Publishers
  • R Discovery PrimeNew
  • Ask R Discovery
  • Blog
  • Accessibility
  • Topics
  • Journals
  • Open Access Papers
  • Year-wise Publications
  • Recently published papers
  • Pre prints
  • Questions
  • FAQs
  • Contact us
Lead the way for us

Your insights are needed to transform us into a better research content provider for researchers.

Share your feedback here.

FacebookTwitterLinkedinInstagram
Cactus Communications logo

Copyright 2025 Cactus Communications. All rights reserved.

Privacy PolicyCookies PolicyTerms of UseCareers