• 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
Sign In
Paper
Search Paper
Cancel
Pricing Sign In
  • My Feed iconMy Feed
  • Search Papers iconSearch Papers
  • Library iconLibrary
  • Explore iconExplore
  • Ask R Discovery iconAsk R Discovery Star Left icon
  • Chat PDF iconChat PDF Star Left icon
  • Chrome Extension iconChrome Extension
    External link
  • Use on ChatGPT iconUse on ChatGPT
    External link
  • iOS App iconiOS App
    External link
  • Android App iconAndroid App
    External link
  • Contact Us iconContact Us
    External link
Discovery Logo menuClose menu
  • My Feed iconMy Feed
  • Search Papers iconSearch Papers
  • Library iconLibrary
  • Explore iconExplore
  • Ask R Discovery iconAsk R Discovery Star Left icon
  • Chat PDF iconChat PDF Star Left icon
  • Chrome Extension iconChrome Extension
    External link
  • Use on ChatGPT iconUse on ChatGPT
    External link
  • iOS App iconiOS App
    External link
  • Android App iconAndroid App
    External link
  • Contact Us iconContact Us
    External link

Tumor Cell Membrane 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
1548 Articles

Published in last 50 years

Related Topics

  • Expression In Tumor Cells
  • Expression In Tumor Cells
  • Cancer Cell Membrane
  • Cancer Cell Membrane
  • Target Tumor Cells
  • Target Tumor Cells

Articles published on Tumor Cell Membrane

Authors
Select Authors
Journals
Select Journals
Duration
Select Duration
1497 Search results
Sort by
Recency
  • New
  • Research Article
  • 10.3760/cma.j.cn112151-20250508-00331
Consistency of claudin 18.2 expression with antibodies of different clones in gastric adenocarcinoma
  • Nov 8, 2025
  • Zhonghua bing li xue za zhi = Chinese journal of pathology
  • C S Wang + 8 more

Objective: To explore the consistency of claudin 18.2 immunohistochemistry (IHC) using 4 different clone antibodies in gastric adenocarcinoma. Methods: A total of 226 gastric adenocarcinomas diagnosed and treated at the Drum Tower Hospital Affiliated to Nanjing University Medical College between January 2022 to March 2023 were included in this study. The cohort consisted of 165 males and 61 females, with a mean age of (61.3±12.1) years. Tumor tissues from radical resection specimens were collected for tissue microarrays. IHC detection of claudin 18.2 was performed using the EnVision method, utilizing 4 clones of antibody: OTIR157B5, 43-14A, EPR19202 and D313D22. The results were interpreted based on both the intensity of staining on tumor cell membranes and the percentage of positive tumor cells relative to the total tumor cells. Results: The positive cutoff value was set as moderately to strongly linear membrane staining in ≥75% of all viable invasive tumor cells, and clone OTIR157B5 demonstrated the highest positive expression rate at 52.2% (118/226). Additionally, the clones OTIR157B5, 43-14A, and EPR19202 were consistently and strongly positive, with all agreement rates of Cohen κ exceeding 0.8. In gastric adenocarcinoma and its three Lauren subtypes, OTIR157B5 exhibited clear membranous localization. Conclusions: Clone OTIR157B5 of claudin 18.2 antibody shows the highest rate of moderately to strongly linear membrane-positive staining, accounting for ≥75% of all viable invasive tumor cells, and clones 43-14A and EPR19202 show strong consistency and high sensitivity.

  • New
  • Research Article
  • 10.1002/advs.202517228
Magnetoelectric Nanotherapy Achieves Complete Tumor Ablation and Prolonged Survival in Pancreatic Cancer Murine Models.
  • Nov 3, 2025
  • Advanced science (Weinheim, Baden-Wurttemberg, Germany)
  • John Michael Bryant + 28 more

Magnetoelectric nanoparticles (MENPs), when activated by a magnetic field, are shown to provide a minimally invasive, drug-free, theranostic approach to pancreatic ductal adenocarcinoma (PDAC) treatment. The magnetoelectric effect allows intravenously administered MENPs to be magnetically guided to PDAC xenograft tumors and remotely activated with a 7T-MRI field to induce targeted, electrode-free tumor ablation with real-time imaging feedback. A single MENP treatment achieved a threefold median reduction in tumor volume and complete tumor responses in 33.3% of mice at 300 and 600µg doses (N = 17) and significantly longer mean overall survival as compared to the control cohorts (54.1 vs 28.8 days, χ2 = 40.14, p = 0.045), without evident toxicity in any imaged organ. In contrast, mice receiving subtherapeutic doses, non-activated MENPs, or saline controls showed no significant response. MRI T2* relaxation time decreases closely correlated with tumor reduction (ρ = -0.73, p < 0.001), supporting MENPs as both a therapeutic and imaging biomarker. Mechanistically, MENPs preferentially target cancer cells via magnetic-field-driven electrostatic interactions specific to tumor cell membranes, in agreement with multiphysics numerical simulations. Flow cytometry confirmed that MENP activation primarily induces apoptosis, with minimal necrosis, and time-course studies showed a progressive apoptotic response over 3-hour post-treatment. The findings establish MENPs as a versatile, image-guided, theranostic platform with translational promise for minimally invasive oncology.

  • New
  • Research Article
  • 10.1016/j.bioadv.2025.214339
Mechanism of selenium-doped black phosphorus nanosheets wrapped with biomimetic tumor cell membrane for prostate cancer immunotherapy.
  • Nov 1, 2025
  • Biomaterials advances
  • Xingjian Yan + 4 more

Mechanism of selenium-doped black phosphorus nanosheets wrapped with biomimetic tumor cell membrane for prostate cancer immunotherapy.

  • New
  • Research Article
  • 10.1021/jacs.5c13182
Cell Membrane-Anchored Click Reaction Enhances Porphyrin Uptake for Highly Efficient Photodynamic Therapy of Breast Tumors.
  • Oct 31, 2025
  • Journal of the American Chemical Society
  • Yu Ma + 7 more

Enhancing cell uptake of drugs for better therapy is a fundamental scientific problem in pharmaceutics. The "hydrophobic-hydrophilic-hydrophobic" structure has shown the potential of enhancing cell uptake of drugs. Thus, cell membrane-anchored formation of this structure should additionally enhance cell uptake of drugs but has not been reported. In this work, we rationally designed acid-deshielding cysteine-PEG-DSPE (DA-Cys-PD) and PE-porphyrin-PEG-CBT (Por-CBT). After DA-Cys-PD anchors on the tumor cell membrane, the weak acidic environment of the cell deshields the anchor to yield Cys-PD, which subsequently click-reacts with Por-CBT to yield Por-Luc-PD with a "hydrophobic-hydrophilic-hydrophobic" structure. This in situ formed structure significantly enhances the cellular uptake of porphyrin and its consequent photodynamic therapeutic effect on breast tumors. Particularly, with the assistance of the cell membrane-anchored click reaction, porphyrin uptake in cancer cells or breast tumors is increased roughly to be 7.8-fold or 3.9-fold of that of the negative control group whose Cys is acid-inactive, respectively. The enhanced porphyrin uptake leads to highly efficient photodynamic therapy of breast tumors with a remarkable tumor growth inhibition rate of 64.1% compared with that of 5.5% of the negative control group. This approach of cell membrane-anchored click reaction provides people with a simple and feasible avenue for enhancing cell uptake of drugs/probes, as well as their therapeutic/diagnostic effects.

  • New
  • Research Article
  • 10.1038/s41419-025-08125-7
Development of antibody drug conjugates targeting epithelial membrane protein 2-highly expressed lung cancer
  • Oct 31, 2025
  • Cell Death & Disease
  • Mengge Zheng + 14 more

Lung cancer is one of the most devastating types of cancer, and the treatment of lung cancer has been facing great challenges. Antibody-drug conjugates (ADCs), a new type of targeted therapy, have been widely used in cancer therapy and have opened up a new perspective for the treatment of lung cancer. Here, using a tissue-microarray-based antibody library screening, we identified the FK002 antibody that specifically binds to the tumor cell membrane across various tumor types. We determined that FK002 targets epithelial membrane protein 2 (EMP2), a member of the tetraspanin superfamily of proteins. Subsequently, we developed the EMP2-directed ADC, FK002-exatecan, with a potent DNA topoisomerase I inhibitor (exatecan). In-depth in vitro and in vivo experiments have shown the efficacy and specificity of the EMP2-directed ADC. We validated that the FK002-exatecan ADC effectively eradicated tumors in various lung cancer cell lines and xenograft mouse models, including patient-derived xenograft (PDX) and patient-derived tumor-like cell cluster (PTC) models, as well as xenograft tumors. Mechanistically, FK002-exatecan specifically bound to EMP2 and was internalized into tumor cells, followed by intracellular trafficking to the lysosome and exatecan release, which induced cell cycle arrest and apoptosis. This study identifies EMP2 as a novel molecular target for lung cancer therapy and establishes a foundation for developing ADCs that selectively eradicate lung cancer cells.

  • New
  • Research Article
  • 10.1002/anie.202516998
A Smart Self-Immobilization Magnetic Resonance Contrast Agent for Delayed Tumor Imaging In Vivo.
  • Oct 29, 2025
  • Angewandte Chemie (International ed. in English)
  • Zheng Huang + 5 more

Achieving tumor-specific accumulation and high relaxivity remains critical yet challenging for improving magnetic resonance imaging (MRI) probe sensitivity. Here, we present P-QM-Gd, an enzyme-responsive, self-immobilizing small-molecule MRI probe designed for high-sensitivity tumor imaging. Upon activation by membrane-bound alkaline phosphatase (ALP), P-QM-Gd is enzymatically converted to generate reactive quinone methide intermediates. These intermediates readily undergo nucleophilic addition with proximal protein residues, enabling the covalent conjugation of paramagnetic gadolinium complexes directly onto tumor cell membranes. This enzymatic self-immobilization markedly increased the longitudinal relaxivity (r1) from 7.35 to 13.15 mM-1 s-1 (0.5 T, 21.3MHz, 32°C) and effectively "traps" the probe at the tumor site. P-QM-Gd showed rapid tumor uptake and efficient covalent labeling, achieving distinct MR contrast enhancement (> 60%). The imaging window was extended for up to 24h in subcutaneous HeLa and orthotopic K7M2 tumor models. Notably, delayed MRI with P-QM-Gd enabled precision visualization of ∼1.3mm orthotopic K7M2 tumors in mice. By overcoming the rapid washout and low sensitivity inherent to traditional small-molecule MRI probes through this enzymatic self-immobilization strategy, P-QM-Gd offers a promising approach for high-sensitivity, high-spatial resolution delayed tumor MRI, making it particularly effective for early-stage lesion detection.

  • New
  • Research Article
  • 10.1002/ange.202516998
A Smart Self‐Immobilization Magnetic Resonance Contrast Agent for Delayed Tumor Imaging In Vivo
  • Oct 29, 2025
  • Angewandte Chemie
  • Zheng Huang + 5 more

Abstract Achieving tumor‐specific accumulation and high relaxivity remains critical yet challenging for improving magnetic resonance imaging (MRI) probe sensitivity. Here, we present P‐QM‐Gd , an enzyme‐responsive, self‐immobilizing small‐molecule MRI probe designed for high‐sensitivity tumor imaging. Upon activation by membrane‐bound alkaline phosphatase (ALP), P‐QM‐Gd is enzymatically converted to generate reactive quinone methide intermediates. These intermediates readily undergo nucleophilic addition with proximal protein residues, enabling the covalent conjugation of paramagnetic gadolinium complexes directly onto tumor cell membranes. This enzymatic self‐immobilization markedly increased the longitudinal relaxivity ( r 1 ) from 7.35 to 13.15 mM −1 s −1 (0.5 T, 21.3 MHz, 32 °C) and effectively “traps” the probe at the tumor site. P‐QM‐Gd showed rapid tumor uptake and efficient covalent labeling, achieving distinct MR contrast enhancement (&gt; 60%). The imaging window was extended for up to 24 h in subcutaneous HeLa and orthotopic K7M2 tumor models. Notably, delayed MRI with P‐QM‐Gd enabled precision visualization of ∼1.3 mm orthotopic K7M2 tumors in mice. By overcoming the rapid washout and low sensitivity inherent to traditional small‐molecule MRI probes through this enzymatic self‐immobilization strategy, P‐QM‐Gd offers a promising approach for high‐sensitivity, high‐spatial resolution delayed tumor MRI, making it particularly effective for early‐stage lesion detection.

  • New
  • Research Article
  • 10.1002/advs.202512135
Sequential Tumor Microenvironment Reprogramming by Nanoplatform Potentiates Sonodynamic-Chemodynamic Therapy and Immune Checkpoint Blockade in Breast Cancer.
  • Oct 24, 2025
  • Advanced science (Weinheim, Baden-Wurttemberg, Germany)
  • Yang Yu + 5 more

The complex tumor microenvironment (TME) remains a major barrier to effective breast cancer therapy. A modular nanoplatform capable of sequentially reprogramming the TME through cascade actions and responsive therapeutic functions is developed to enhance breast cancer immunotherapy. A hybrid nanoparticle (MCC) containing manganese dioxide (MnO2), calcium peroxide (CaO2), and chlorin e6 (Ce6) is synthesized and subsequently camouflaged with a tumor cell membrane. Surface conjugation of a PD-L1 antibody (αP) is then achieved via a glutathione (GSH)-responsive fragment, resulting in the formation of an integrated nanoplatform MCC@TM-αP. Through dual-targeting mechanisms involving the tumor cell membrane and the PD-L1 antibody, MCC@TM-αP achieves efficient enrichment at tumor sites. MCC@TM-αP alleviates hypoxia by generating O2 from CaO2 in the acidic TME and scavenges GSH via the MnO2-mediated Fenton-like reaction, thereby markedly amplifying the sonodynamic efficacy of Ce6. The combined effects of sonodynamic therapy and chemodynamic therapy ablate tumors and reprogram the immunosuppressive TME. Upon cleavage of the GSH-responsive fragment by intratumoral GSH, MCC@TM-αP releases the PD-L1 antibody, eliciting a robust immune response that eradicates metastatic tumors. In murine breast cancer models, this therapeutic strategy enhances tumor infiltration by effector T cells and suppresses metastatic progression. By sequentially decoupling the immunosuppressive mechanisms, this study provides a programmable approach to potentiate immunotherapy and overcome TME-driven resistance.

  • New
  • Research Article
  • 10.1021/acsbiomaterials.5c01048
Activating Immunogenic Cell Death and Reprogramming the Tumor Microenvironment to Enhance Radiotherapy for Breast Cancer via Low-Frequency Ultrasound-Driven "Artificial T Cells".
  • Oct 23, 2025
  • ACS biomaterials science & engineering
  • Lei Dong + 6 more

The immunosuppressive tumor microenvironment (TME) is a key factor that reduces the effectiveness of radiotherapy in breast cancer, as it limits the ability of cytotoxic T lymphocytes (CTLs) to effectively target and eliminate tumor cells. In this study, T cell membrane components were incorporated and granzyme B (GrB) was loaded to construct biomimetic microbubbles with a core-shell structure (GrB@TMBs), named "artificial T cells". Low-frequency ultrasound (LFUS) mediates the "cavitation effect" of GrB@TMBs, causing the formation of micropores on the tumor cell membrane and releasing GrB cross these pores, thereby mimicking the process of CTLs killing tumor cells. Consequently, up to 1.99-fold of dendritic cells and 2.87-fold of tumor-specific T cells in the TME were observed with the US + GrB@TMBs group compared to the free GrB group. Furthermore, the use of LFUS-driven "artificial T cells" to enhance breast cancer radiotherapy led to the elimination of 40% of tumors, prolonged survival, and the promotion of long-term immune memory formation. In summary, LFUS-driven "artificial T cells" provide a promising strategy for improving the efficacy of breast cancer radiotherapy.

  • New
  • Research Article
  • 10.1002/smll.202509285
Three-in-One: Biomimetic Trimetallic Ionic-Site Nanozyme Composite as a Redox Homeostasis Disruptor for Ferroptosis/Cuproptosis/Pyroptosis Co-Activated Nanocatalytic Therapy.
  • Oct 22, 2025
  • Small (Weinheim an der Bergstrasse, Germany)
  • Lin-Jiao Yang + 11 more

Given the critical role of tumor redox homeostasis in sustaining malignant growth, simultaneously targeting multiple aspects of intracellular balance may offer a more efficient therapeutic strategy. Herein, a trimetallic ionic-site nanozyme is engineered by integrating Au3⁺, Ru3⁺, and Cu2⁺ ions into a nanoscale metal-organic framework (tis-ARC). The nanozyme is further loaded with gambogic acid (GA) and buthionine sulfoximine (BSO) and cloaked in tumor cell membranes (tis-ARC-GB@M) to enhance targeting and homologous recognition. The resulting tis-ARC-GB@M exhibited multi-enzyme mimetic catalytic activities that disrupted tumor redox balance by simultaneously amplifying reactive oxygen species (ROS) production and depleting glutathione (GSH), thereby dismantling the tumor's intrinsic antioxidant defenses. This cascade of events triggered several cell death pathways-including ferroptosis, cuproptosis, and pyroptosis, and released damage-associated biomarker molecules that reprogrammed the tumor microenvironment (TME). Mechanistically, oxidative stress-enhanced ferroptosis, cuproptosis, and pyroptosis collectively disrupted mitochondrial metabolism, which in turn exacerbated intracellular oxidative stress, resulting in a mutually reinforcing therapeutic effect. In vitro and in vivo studies demonstrated that tis-ARC-GB@M significantly suppressed tumor growth in tumor-bearing models. Overall, this approach establishes a novel paradigm for antitumor nanocatalytic therapy through the targeted disruption of intracellular homeostasis.

  • New
  • Research Article
  • 10.1039/d5cc04269k
A membrane camouflaged copper single-atom nanozyme for combined mild photothermal and chemodynamic therapy.
  • Oct 21, 2025
  • Chemical communications (Cambridge, England)
  • Chunmei Guo + 9 more

A copper (Cu) single-atom nanozyme coated with 4T1 tumor cell membrane was constructed to enhance tumor targeting and biocompatibility. The platform combines chemodynamic therapy (CDT) and mild photothermal therapy (PTT) for improved antitumor efficacy and enables precise tumor detection via photoacoustic imaging (PAI).

  • New
  • Research Article
  • 10.1007/s13346-025-01994-7
Preliminary study on targeted therapy of breast cancer using tumor cell membrane-coated dual-loaded liposomes based on chemo-photothermal synergistic effects.
  • Oct 20, 2025
  • Drug delivery and translational research
  • Yuwei Wang + 6 more

Addressing the clinical challenges of lacking effective therapeutic targets and the high recurrence rate in triple-negative breast cancer (TNBC), this study innovatively constructed a targeted nanoplatform (CM@DOX-GO NPs) based on the synergistic combination of photothermal therapy (PTT) and chemotherapy. This platform achieves a breakthrough integration of spatiotemporally coordinated PTT-chemotherapy and precise targeting by co-loading the highly efficient photothermal agent, monolayer graphene oxide (GO), and the chemotherapeutic drug doxorubicin hydrochloride (DOX) into core-shell structured liposomes, followed by biomimetic modification with tumor cell membranes (derived from MDA-MB-231 cells). Serving as a novel near-infrared (NIR) photosensitizer, GO exhibits a unique photothermal effect that not only directly induces tumor cell death but also enhances cellular membrane permeability through hyperthermia, thereby promoting the intertumoral penetration and targeted release of DOX. Experimental results confirmed that the tumor cell membrane-camouflaged nanoparticles exhibit significantly enhanced homologous targeting capability compared to conventional formulations. Their optimized systemic circulation characteristics and specific fluorescence enrichment within the tumor region collectively validate the effectiveness of the biomimetic strategy. Under NIR irradiation, this system leverages the spatiotemporally coordinated mechanism of PTT-chemotherapy, substantially improving tumor cell eradication efficiency while simultaneously reducing systemic toxicity through precise energy control. This innovative design successfully overcomes the limitations inherent in conventional PTT, such as uneven energy distribution and low bioavailability of photosensitizers, offering a promising new strategy for highly efficient and low-toxicity TNBC treatment.

  • Research Article
  • 10.1016/j.ijpharm.2025.126091
Self-amplifying ROS-responsive trinity nanodelivery system for enhanced cancer therapy.
  • Oct 15, 2025
  • International journal of pharmaceutics
  • Jia Deng + 8 more

Self-amplifying ROS-responsive trinity nanodelivery system for enhanced cancer therapy.

  • Research Article
  • 10.1002/advs.202515006
Engineering Metal-Organic-Framework-Based STING Nanoagonists for PROTAC-Enhanced Cancer Chemo-Metalloimmunotherapy.
  • Oct 13, 2025
  • Advanced science (Weinheim, Baden-Wurttemberg, Germany)
  • Zhenzhen Chen + 3 more

Chemo-metalloimmunotherapy is emerging as a promising strategy for cancer treatment by integrating chemotherapy-induced immunogenicity with metal ion-mediated immune activation. However, its efficacy is hampered by chemoresistance and immune escape driven by PD-L1 upregulation. Here, a multifunctional manganese-based metal-organic framework nanoplatform (Mn-CDDP-dBET6@CM) is reported that integrates metalloimmunotherapy, chemotherapy, and Proteolysis-targeting chimera (PROTAC) -mediated epigenetic modulation for enhanced cancer treatment. This system co-delivers Mn2+ to activate the stimulator of interferon genes (STING) pathway, cisplatin (CDDP) to induce nucleus DNA damage, and the bromodomain-containing protein 4 (BRD4) -targeting PROTAC dBET6 to promote mitochondrial DNA release and suppress PD-L1-mediated immune evasion. Coated with tumor cell membranes for homologous targeting and immune evasion, Mn-CDDP-dBET6@CM effectively induces cellular senescence, robust innate and adaptive immune activation, and tumor microenvironment remodeling. In vitro and in vivo studies demonstrate potent tumor growth inhibition, enhance dendritic cell maturation, and increase cytotoxic T cell infiltration. This nanoplatform offers a promising strategy to overcome chemoresistance and immunosuppression, providing a versatile approach for next-generation chemo-metalloimmunotherapy.

  • Research Article
  • 10.3760/cma.j.cn112151-20250402-00230
Clinicopathological and genetic features of hyalinizing trabecular tumor of the thyroid
  • Oct 8, 2025
  • Zhonghua bing li xue za zhi = Chinese journal of pathology
  • D J Hu + 6 more

Objective: To analyze the cytological, histological, immunohistochemical, and molecular pathological features of hyalinizing trabecular tumor (HTT). Methods: Clinical and pathological data of the HTT cases diagnosed at Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine between 2020 and 2024 were collected and analyzed. HE staining, special staining, immunohistochemical staining, and next-generation sequencing were performed on all cases. Results: Among the 10 HTT patients, 4 were male and 6 were female. The age at onset ranged from 29 to 85 years, with a median age of 49 (35,61) years. The maximum tumor diameter ranged from 0.3 to 5.3 cm. Cytologically, the smears were hypercellular and showed tumor cells arranged in nested clusters with visible basement membrane-like material. The nuclei were oval with finely granular chromatin, and nuclear pseudoinclusions were readily identifiable. Histologically, the tumors were well demarcated. The tumor cells were arranged in a paraganglioma-like pattern, exhibiting typical nuclear features of papillary thyroid carcinoma and psammoma bodies. Yellow bodies were observed in the cytoplasm. The stroma was rich in hyalinized material, which was periodic acid-Schiff stain (PAS)-positive. Immunohistochemically, the tumor cells showed diffuse expression of TTF-1 and focal expression of thyroglobulin. Aberrant immunoreaction with Ki-67 was present in the cytoplasm and membrane of the tumor cells. Molecular testing was performed on 8 cases. The PAX8-GLIS3 gene fusion was detected in 7 cases. Among these fusion-positive cases, 4 exhibited additional genetic abnormalities: one concurrent TSHR point mutation (p.D617H); one concurrent HRAS point mutation (p.Q61R); one concurrent LRP1B point mutation (p.S1752L), SUGCT point mutation (p.K137), and TERT point mutation (p.P785L); one concurrent MTOR mutation (7528+27A>T) and FLT3 mutation (p.E77K). The key initiating factors for thyroid carcinoma, including the BRAF V600E mutation and RET rearrangements, were absent in all cases tested. Conclusions: Cellular pleomorphism, yellow bodies and basement membrane-like material constitute important cytological and histological features for the differential diagnosis of HTT. Immunophenotypically, thyroglobulin may show focal expression, while Ki-67 is typically localized in the tumor cell membrane and cytoplasm. This study also demonstrates that PAX8-GLIS3 fusion is a characteristic molecular abnormality in HTT, although cases with wild type of GLIS gene may also present. Although rare, HTT may harbor point mutations in HRAS and TSHR, and other uncommon genetic alterations.

  • Research Article
  • 10.1093/neuonc/noaf193.109
P01.29.A GLIOBLASTOMA CELL MOTILITY AND INVASION IS REGULATED BY MEMBRANE-ASSOCIATED HEAT SHOCK PROTEIN HSP70
  • Oct 3, 2025
  • Neuro-Oncology
  • E Oganesyan + 13 more

Abstract BACKGROUND Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor which characterized by a high ability to migrate and invade into surrounding brain tissue. The main problem in therapy is the tumor’s high invasiveness. The heat shock protein 70 (Hsp70) has a cytoplasmic localization in normal cells but specifically localized on the plasma membrane (mHsp70) of tumor cells which makes it a promising therapeutic target. We investigated the role of mHsp70 in motility of GBM cells. MATERIAL AND METHODS The study was performed using primary human glioblastoma cell cultures (n=4) isolated from biopsy material. The expression of mHsp70 was detected with confocal microscopy, flow cytometry, Western blot, and proteome analysis of lipid rafts. Сells were sorted into two subpopulations - high (mHsp70High) and low expressed mHsp70 (mHsp70Low). The motility characteristics were studied using automatic single-cell tracking and transwell analysis with Hsp70 inhibitors (PES, JG-98) blocking different domains. RESULTS The expression of mHsp70 was observed in 100% of cell populations. The highest content of mHsp70 revealed in lamellipodia and filopodia. Proteomic analysis of lipid rafts identified the interaction of mHsp70 with proteins involved in cytoskeletal remodeling and migration (tubulin-β, tubulin-α4a, myosin IX, filamin A, integrin β-1, α-enolase and the small GTPase RhoA) that may indicate the chaperone participation in the regulation of migration. The mHsp70High subpopulations had a mean speed of 1.5-2 times higher than mHsp70Low. The inhibitors of Hsp70 effectively decrease GBM cells speed. CONCLUSION This study expands our understanding of mHsp70 influence to the migration of GBM cells. The protein expression level correlates with motility. The Hsp70 inhibitors reduces the invasive potential of cells, which allows to use them as an adjuvant agents in the development of novel approaches to the treatment of malignant tumors. Research was funded by the Ministry of Science and Higher Education of Russia (agreement № 075-15-2022-301).

  • Research Article
  • 10.1093/neuonc/noaf193.102
P01.22.B MEMBRANE-BOUND HEAT SHOCK PROTEINS: MHSP70 AS A TARGET FOR THERANOSTICS IN NEURO-ONCOLOGY
  • Oct 3, 2025
  • Neuro-Oncology
  • M Shevtsov + 10 more

Abstract BACKGROUND Heat shock protein mHsp70 can be exposed on the surface of the plasma membrane of tumor cells. We demonstrate that mHsp70 of malignant brain tumors is required for high migratory and invasive activity of cancer cells and can serve as a target for RAS70 peptide for tumor diagnostics. MATERIAL AND METHODS Protein expression on the cell membrane surface of tumor cell lines and primary brain tumors of adult and pediatric GBM patients was studied using FACS, live-cell confocal microscopy, and isolated lipid raft mass-spectrometry. Protein involvement in cell invasion and migration was assessed using low-molecular inhibitors (PES, JG-98) in vitro and in orthotopic glioma animal models. The target properties of the fluorescently-labeled peptide RAS70, which recognizes membrane-bound mHsp70, were assessed in primary patient tumors and in animal models. RESULTS Live-cell inverted confocal microscopy of brain tumor samples (n=32) showed mHsp70 overexpression on the membrane. Mass-spectrometry analysis of lipid rafts isolated from brain tumor cells confirmed the presence of the protein in HSP cluster, which in turn, during interactome analysis, was associated with proteins involved in cell migration and invasion (i.e., Rac1, RhoC, myosin-9). Application of small-molecule inhibitors of HSP70 (JG-98, PES) led to a significant decrease in the invasive potential of cells isolated from a tumor sample of patients, which indicates the role of the chaperone in invasion. Moreover, the use of chaperone inhibitors in orthotopic brain tumors model in rodents significantly decreased tumor progression, which was accompanied by an increased overall survival. Data demonstrate that chaperone inhibitors, particularly JG-98, disrupt the function of mHsp70, thereby providing an opportunity to better understand the diverse functions of this protein and offer aid in the development of novel cancer therapies. Apart from chaperone inhibitors we developed mHsp70-targeted peptide that could efficiently cross the blood-brain barrier accumulating in the glioblastoma cells in vivo. Subsequently, in the orthotopic GL261 mice and C6 rat glioma, intravenous administration of the fluorescently labeled peptide resulted in the tumor retention of agent (further confirmed by histological studies). Topical application of the mHsp70-targeted peptide (sprayed over the freshly dissected brain tissues) helped to delineate the tumors in neuro-oncological adult patients employing intraoperative fluorescent imaging system. CONCLUSION Membrane-bound mHsp70 could be used as a target for tumor theranostics. RAS70 peptide recognizing mHsp70 demonstrated high targeting properties which could be employed for the intraoperative visualization as well as for developing of a potential carrier for drug delivery.

  • Research Article
  • 10.1093/neuonc/noaf193.214
P05.09.A RAS70 PEPTIDE-MEDIATED FLUORESCENCE IMAGING OF HSP70-EXPRESSING HIGH-GRADE GLIOMAS AND BRAIN METASTASES
  • Oct 3, 2025
  • Neuro-Oncology
  • A Nechaeva + 6 more

Abstract BACKGROUND The 70kDa heat shock protein (Hsp70) exhibits a unique expression pattern, being prominently displayed on the plasma membrane of tumor cells while remaining absent from normal cells. This differential expression makes mHsp70 an attractive target for selective tumor imaging. In this study, a 17-mer RAS70 peptide, designed to recognize the membrane-bound mHsp70 on cancer cells, was linked to Cy7.5 fluorophore. This conjugate was then employed for epifluorescent detection of mHsp70-positive brain tumors and metastases. METHODS Adult patients with glioblastoma (n=6) and brain metastases (n=6) were included in the pilot study between November 2024 and April 2025. Histologically confirmed tumor specimens were obtained from three tumor regions based on preoperative MRI scanning: necrotic, the contrast-enhanced, and peritumoral zones. Specimens were ex vivo sprayed with Cy7.5-conjugated RAS70 peptide (exposure time of 5 min), followed by an intensive rinsing of the sample with PBS. Cy7.5-conjugated scramble peptide was used as a control agent. RAS70 was excited by an 800 nm light source on an operating microscope (Leica M720 OH5, Germany). Images were analyzed using ImageJ software with IBRs estimated as the Ratio of fluorescence signal intensity of Interest regions (ROIs, n=102) and the mean signal intensity of black Background. RESULTS Сomparison of IBRs in samples from different tumor areas revealed significantly higher IBRs for RAS70 in samples compared to control scramble-peptide (p&amp;lt;0.0001), constituting: necrotic zone (18.7 ± 6.1 a.u), contrast-enhanced zone (9.7 ± 5.1 a.u.), peritumoral zone (9.3 ± 5.0 a.u), control (1.9 ± 1.1 a.u). The mean fluorescence intensities did not significantly differ between glioblastoma and metastasis specimens. According to ROC analysis, the sensitivity and specificity of the RAS70 peptide for diagnosing the contrast-enhanced zone were 95.56% and 88.89% (threshold &amp;gt;3.05 a.u., AUC 0.9790, p&amp;lt;0.0001), for the peritumoral zone 95.0% and 88.89% (threshold &amp;gt;3.05 a.u., AUC 0.9971, p&amp;lt;0.0001), for the necrotic zone 94.74% and 94.44% (threshold &amp;gt;5.60 a.u., AUC 0.9971, p&amp;lt;0.0001). CONCLUSION The RAS70 peptide, exhibiting targeting properties for brain tumors and metastases, holds promise not only for fluorescence-guided surgery but also as a potential drug delivery vehicle.

  • Research Article
  • 10.1002/anie.202507700
Persistent Membrane-Anchored Oligomeric Peptides with Nanopore Formation for Targeted Immune Modulation.
  • Oct 1, 2025
  • Angewandte Chemie (International ed. in English)
  • Jingtian Cao + 12 more

In the field of targeted diagnosis and therapy, one of the key challenges is the off-target effect of these recognition molecules. Herein, a new class of oligomeric helical peptides is introduced through rational design and high-throughput screening to address this challenge. Following amino acid mutations and structural optimizations, lead candidates TA03 and TA10 were identified. Thereinto, TA03 was shown to specifically recognize the PD-L1 target and employ a unique aromatic side-chain anchoring strategy to form stable nanopores on the tumor cell membrane, ensuring prolonged residence time. This structure from the precise match between helix length and lipid bilayer thickness, alongside the positioning of two tryptophan (Trp) residues near the peptide terminus. Both Trp side chains interact with the phospholipid membrane via aromatic-hydrophobic forces, allowing TA03 to form an "hourglass-like" pore structure. This design ensures proper membrane localization and extends residence time on tumor cells of TA03, increasing the persistent interactions between TA03 and PD-L1, effectively avoiding off-target effects. Furthermore, the unique structure and PD-L1 targeting of TA03 cause physical disruption to tumor cells boosting immunotherapy effects and inhibiting tumor growth. It provides an avenue for new molecular structure to overcome off-target effect towards the membrane associate targets.

  • Research Article
  • 10.1016/j.mtbio.2025.102153
A polymeric nanovesicle delivers sulfopin and gemcitabine to remodel tumor microenvironment for enhanced chemoimmunotherapy against orthotopic pancreatic cancer.
  • Oct 1, 2025
  • Materials today. Bio
  • Weiting Jiang + 6 more

A polymeric nanovesicle delivers sulfopin and gemcitabine to remodel tumor microenvironment for enhanced chemoimmunotherapy against orthotopic pancreatic cancer.

  • 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