Published in last 50 years
Articles published on Dual Targeting
- New
- Research Article
- 10.1002/advs.202515664
- Nov 6, 2025
- Advanced science (Weinheim, Baden-Wurttemberg, Germany)
- Shichao Zhang + 9 more
Acute kidney injury (AKI) represents a critical clinical condition marked by abrupt deterioration of renal function, primarily driven by oxidative stress, inflammation, and apoptosis. However, effective targeted therapies remain limited. Here, a smart, biomimetic nanoplatform (CeAst@MK) that synergistically addresses oxidative and inflammatory injury in AKI is reported. CeAst nanoparticles are formed via coordination between Ce3⁺ ions and astragalin (Ast), a natural flavonoid with intrinsic ROS-scavenging and anti-inflammatory properties. To enhance immune evasion and renal targeting specificity, CeAst is cloaked with macrophage membranes (MCM) and modified with a kidney-targeting peptide (KTP), yielding the final CeAst@MK system. The platform exhibits pH-responsive release in the acidic microenvironment of injured renal tissues, enabling precise and rapid therapeutic delivery. In both LPS- and ischemia reperfusion-induced AKI models, CeAst@MK significantly improves renal function, suppresses proinflammatory cytokines, and promotes M2 macrophage polarization. Mechanistically, it modulates PI3K/Akt and NF-κB pathways, achieving dual antioxidative and anti-inflammatory effects. This study presents a translationally promising nanotherapeutic system integrating natural antioxidants, biomimetic camouflage, and tissue-specific delivery, offering an effective and precise strategy for AKI intervention.
- New
- Research Article
- 10.1007/s00262-025-04215-9
- Nov 6, 2025
- Cancer immunology, immunotherapy : CII
- Mengqing Chen + 8 more
Cancer stem cells (CSCs) are considered the 'seeds' of recurrence after chemotherapy, but eliminating CSCs remains notoriously challenging. This study aims to examine whether cell cycle checkpoint kinase 1 (CHK1) blockade can abrogate the stemness of ovarian cancer (OC) cells, making them easier targets of anti-tumor immunity. Prexasertib was used to block CHK1 in OC cell lines and xenografts, and its cytotoxicity was assessed in vitro and in vivo. In vitro tumor-sphere formation assays and stemness markers were used to evaluate cell stemness. PD-L1 expressions were examined via qRT-PCR, Western blot, flow cytometry, and immunohistochemistry. Prexasertib in combination with anti-PD-L1 antibody Atezolizumab was tested in immune-proficient mice bearing OC xenografts in terms of effects on tumor growth, tumor cell stemness, and tumor infiltrating lymphocytes via tumor volume monitoring, immunohistochemistry, and flow cytometry. Prexasertib effectively inhibited CHK1 phosphorylation, exhibited significant anti-tumor effects in vitro and in vivo, accompanied by decreased OC cell stemness. CHK1 was highly expressed in tumor spheres versus tumor cells cultured in 2D system, and Prexasertib treatment suppressed sphere formation and reduced the ALDH+ cell fraction. Unexpectedly, Prexasertib upregulated PD-L1 expression in tumor cells. In vivo, combining Prexasertib with Atezolizumab led to more remarkable remission of tumors, when compared with Prexasertib or Atezolizumab alone. Meanwhile, the tumor-infiltrating CD8+ T cells significantly increased in the combination group, while exhausted T cells decreased; the treatments did not affect CD4+ cell infiltration. Dual targeting of CHK1 and PD-L1 may improve OC treatment by simultaneously suppressing stemness and enhancing anti-tumor immunity.
- New
- Research Article
- 10.1038/s41598-025-22669-w
- Nov 5, 2025
- Scientific reports
- Doretta Cuffaro + 11 more
Antibody-drug conjugates (ADCs) represent a promising strategy in cancer therapy, enabling the targeted delivery of cytotoxic agents to tumor cells. In this study, we developed and characterized novel ADCs combining the anti-EGFR monoclonal therapeutic antibody Cetuximab (Cet) with two aminobisphosphonates (N-BPs) analogues of zoledronic acid (ZA): DC310 and the aminothiazole DC315. These conjugates aim to enhance antitumor efficacy of Cet in colorectal cancer (CRC) by both directly inhibiting tumor cell growth and activating Vδ2 T lymphocytes. We optimized the drug-antibody ratio (DAR), achieving significantly higher DARs compared to previously reported Cet-ZA conjugate, particularly with Cet-DC315 (DAR ≈ 23). Both ADCs retained selective EGFR binding in CRC cell lines and patient-derived organoids (PDO). Functionally, Cet-DC315 markedly inhibited proliferation of EGFR⁺ CRC cell lines in conventional cultures and 3D spheroids. Furthermore, Cet-DC-315 uniquely induced expansion and cytotoxic activation of Vδ2 T cells in co-cultures with CRC cell lines, PDO, and primary tumor samples. These findings suggest that ADCs incorporating novel N-BPs such as DC315 represent a potent approach for dual antitumor targeting through direct cytostatic effects and immune activation, offering a potential therapeutic advantage in the treatment of EGFR+ colorectal cancer.
- New
- Research Article
- 10.1007/s00210-025-04687-7
- Nov 5, 2025
- Naunyn-Schmiedeberg's archives of pharmacology
- Lei Wang + 6 more
This study aims to evaluate the efficacy ofTP (paclitaxel plus carboplatin)chemotherapy combined with trastuzumab and pertuzumab dual-target therapy for HER2-positive breast cancer using serum tumor markers. A total of96patients with HER2-positive breast cancer treated at our hospital from January 2021 to January 2023 were selected and randomly divided into a control group and a study group, with48cases in each group. The efficacy and safety of the treatments were compared between the two groups. Serum levels of CEA, CA125, and CA153 were measured before and after treatment. After treatment, the overall response rate and disease control rate in the study group were significantly higher than those in the control group (P < 0.05). There were no significant differences in serum levels of CEA, CA125, and CA153 between the two groups before treatment (P > 0.05). However, after treatment, the serum levels of CEA, CA125, and CA153 in the study group were significantlylowerthan those in the control group (P < 0.05). ROC curve analysis showed that the combined prediction of CEA, CA125, and CA153 had higher AUC, sensitivity, and specificity compared to individual markers. There were no significant differences between the two groups in terms of liver function abnormalities, hemoglobin reduction, platelet reduction, cardiotoxicity, gastrointestinal symptoms, and joint muscle pain (P > 0.05). Evaluating the efficacy ofTP (paclitaxel plus carboplatin)chemotherapy combined with trastuzumab and pertuzumab dual-target therapy for HER2-positive breast cancer using serum tumor markers has significant clinical value.
- New
- Research Article
- 10.1002/1878-0261.70155
- Nov 5, 2025
- Molecular oncology
- Juhyeon Son + 13 more
Rearranged during transfection (RET) fusions drive subsets of non-small cell lung cancer (NSCLC) and papillary thyroid carcinoma (PTC). Despite new selective RET tyrosine kinase inhibitors (TKIs), resistance usually occurs and is often driven by RET-independent bypass mechanisms. Previous studies have implied crosstalk between RET and proto-oncogene tyrosine-protein kinase SRC, but the anticancer effects of targeting SRC combined with selective RET TKIs, and the underlying molecular mechanisms involved, are not fully understood. Our results show that the multitargeted SRC TKI dasatinib significantly enhanced the efficacy of RET TKIs in RET fusion-positive (RET+) NSCLC and PTC cells. Genetic rescue experiments validated that the combination effects between RET TKIs and dasatinib were indeed SRC-dependent. Phosphoproteomics analysis and validation using selective inhibitors and small interfering RNAs (siRNAs) determined that synergy was primarily mediated by suppression of downstream serine/threonine-protein kinase PAK signaling, with contributions from AKT and ribosomal protein S6. Importantly, synergy was also observed with eCF506 (NXP900), a next-generation clinical SRC inhibitor. Finally, both SRC TKIs restored sensitivity in selpercatinib-resistant RET+ PTC cells. These results elucidate RET and SRC signaling crosstalk in RET+ NSCLC and PTC, suggesting that co-inhibiting SRC has clinical potential in TKI-naïve and -resistant RET+ cancers.
- New
- Research Article
- 10.1007/s12672-025-03844-x
- Nov 4, 2025
- Discover Oncology
- Gamze Guney Eskiler + 3 more
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) have attracted significant attention in triple negative breast cancer (TNBC) treatment. However, the acquired or de novo PARP inhibitor resistance limits treatment success. Ataxia telangiectasia and Rad3-related (ATR) regulates genome integrity, and thus, the aberrant activation of ATR could play a significant role in the pathogenesis of TNBC and be associated with PARPi resistance in especially homologous recombinant deficiency tumors. We aimed to assess the efficacy of ATR and PARP inhibitors combination in TNBC cells and the reversal of PARPi resistance in resistant cells. HCC1937 and HCC1937-R Talazoparib (TAL) resistant cells were treated with Elimusertib (ELI) alone as ATR inhibitor (ATRi) and ELI and TAL combination. Then, the cytotoxic, apoptotic and ATR based DNA damage response (DDR) were evaluated by WST-1, Annexin V, AO/PI, cell cycle and western blot analysis. Our results showed that the ELI and TAL combination could overcome TAL resistance by downregulating cell cycle checkpoint proteins and ATR-based DDR pathways through synergistic effects (ZIP score > 10). The overexpression of ATR and associated cell cycle proteins could play a role in PARPi resistance. However, this combination did not exert synergism in TNBC cells despite a higher apoptotic rate and increased DNA damage compared with the drug alone. Therefore, the dual targeting of ATR and PARP is a promising modality to reverse PARPi resistance with the downregulation of ATR-Chk1 based DNA damage response. However, further preclinical and clinical investigations should be required to elucidate the underlying molecular mechanisms behind ATRi and PARPi interactions in TNBC cells.
- New
- Research Article
- 10.1038/s44318-025-00603-0
- Nov 3, 2025
- The EMBO journal
- Anna Schönbichler + 16 more
Two transmissible cancers, Devil Facial Tumour 1 (DFT1) and Devil Facial Tumour 2 (DFT2), have caused a significant decline in the Tasmanian devil population. DFT1 is driven by ERBB, while DFT2 is driven by PDGFRA. We show that DFT cancer cells exhibit distinct kinase phosphorylation profiles that dictate their responses to tyrosine kinase inhibitors. Upon long-term treatment, both DFT cell lines develop resistance, with DFT1 cells rapidly evading ERBB inhibition without major copy number alterations or significant changes in phosphorylation, suggesting signalling plasticity and engagement of alternative oncogenic drivers. In contrast, DFT2 cells exhibit a slowed development of resistance to imatinib, a selective kinase inhibitor with known activity against PDGFRs. Moreover, DFT2 cell resistance is accompanied by copy number alterations and an activation of ERBB and JAK/STAT signalling with MHCI downregulation, resembling DFT1 signalling. Dual targeting of ERBB and PDGFR shows synergistic effects in DFT1 and may prevent resistance emergence. These findings provide critical insight into the adaptive capacity of transmissible cancers and inform conservation strategies. Moreover, they highlight broader principles of kinase-driven resistance relevant to human cancers with high pathway plasticity.
- New
- Research Article
- 10.1002/jmv.70677
- Nov 1, 2025
- Journal of medical virology
- Huu Y Le + 10 more
While enterovirus D68 (EV-D68) has emerged as a significant pediatric pathogen causing severe respiratory illness and acute flaccid myelitis, no effective therapeutics exist. Through systematic screening of natural products, we identified geranyl-p-trans-coumaric acid (GCA) as a potent EV-D68 inhibitor with EC50 values of 20‒40 μM against multiple clinical strains. Time-of-addition analysis revealed that GCA primarily targets early viral entry processes, with strongest inhibition during the adsorption phase. Mechanistic investigations demonstrated that GCA directly binds to EV-D68 particles and prevents viral uncoating without interfering with initial cellular attachment. Thermal protection and PaSTRy assays confirmed GCA-mediated capsid stabilization, increasing RNA release temperature from 51°C to 53°C. Receptor binding studies showed that GCA selectively inhibits virus interaction with heparan sulfate proteoglycans but not ICAM-5, indicating specific interference with secondary receptor engagement. Resistance selection identified critical mutations in VP1 (T92N) and nonstructural protein 2 C (K6R), with resistance requiring both mutations simultaneously, suggesting cooperative dual-targeting mechanisms. Molecular docking revealed GCA binding to the VP1 canyon region, and combination studies with pleconaril demonstrated synergistic effects (combination index = 0.83). Single mutations alone maintained drug sensitivity, but double mutants exhibited marked resistance (EC50 > 200 μM), confirming dual targeting of viral capsid and cellular host factors. In vivo efficacy studies demonstrated dose-dependent protection, with 71% survival at 12 mg/kg/day compared to 18% in controls (p < 0.01) and significant viral burden reduction in brain and muscle tissues. These findings identify GCA as a promising therapeutic candidate against EV-D68 operating through novel dual mechanisms.
- New
- Research Article
- 10.1016/j.bioorg.2025.109195
- Nov 1, 2025
- Bioorganic chemistry
- Hongyan Lin + 10 more
Click-chemistry mediated synthesis of podophyllotoxin triazole ether derivatives: Inhibiting human CRC tumor growth via dual targeting of AKT1 and tubulin.
- New
- Research Article
- 10.1016/j.ejmech.2025.118025
- Nov 1, 2025
- European journal of medicinal chemistry
- Jiaqi Li + 11 more
Oligopyridinium peptidomimetics with dual bacterial membrane and DNA targeting as resistance-resistant antibacterials.
- New
- Research Article
- 10.1016/j.ejmech.2025.117899
- Nov 1, 2025
- European journal of medicinal chemistry
- R M C Di Martino + 21 more
Rational optimization of D3R/GSK-3β dual target-directed ligands as potential treatment for bipolar disorder: Design, synthesis, X-ray crystallography, molecular dynamics simulations, in vitro ADME, and in vivo pharmacokinetic studies.
- New
- Research Article
- 10.1016/j.cej.2025.167858
- Nov 1, 2025
- Chemical Engineering Journal
- Ibrahim Shaw + 6 more
Microfluidics engineered methotrexate-loaded cRGD lipid-based nanoparticles for the selective targeting and inhibition of αvβ3 integrin to provide dual mechanistic targeting of psoriasis
- New
- Research Article
- 10.4196/kjpp.25.056
- Nov 1, 2025
- The Korean Journal of Physiology & Pharmacology : Official Journal of the Korean Physiological Society and the Korean Society of Pharmacology
- Weifeng Wan + 6 more
Glioblastoma, an aggressive brain tumor that largely depends on angiogenesis, has limited treatment options and poor prognosis. This study explores the therapeutic potential of fimepinostat, a dual HDAC/PI3K inhibitor, as a single agent alone and in combination of temozolomide in glioblastoma using preclinical tumor and angiogenesis models. We show that fimepinostat at nanomolar concentrations inhibited proliferation and induced apoptosis in a panel of glioblastoma cell lines. In addition, fimepinostat inhibited capillary network formation of microvascular endothelial cells derived from patients, indicating that fimepinostat inhibits glioblastoma angiogenesis. Combination index analysis indicates that fimepinostat and temozolomide is synergistic in inhibiting glioblastoma. Consistent with the in vitro findings, fimepinostat significantly inhibited glioblastoma growth in mice without causing any toxicity. The combination of fimepinostat and temozolomide significantly inhibited tumor growth and prolonged survival compared to monotherapy or control. Mechanism studies confirmed that fimepinostat acts on glioblastoma cells through suppressing Akt/MYC. Our findings suggest that dual targeting of tumor and angiogenesis by fimepinostat may provide an alternative approach for anti-glioblastoma therapy.
- New
- Research Article
- 10.2174/0109298673361923250101072831
- Nov 1, 2025
- Current medicinal chemistry
- Saurav Kumar Mishra + 8 more
This study aimed to screen the potential phytochemicals derived from Asparagus racemosus (Shatavari) against Thymidylate Kinase (TMPK) and D9 decapping enzyme, which is the vital target of the monkeypox virus and helps in the host-- pathogen interaction mechanism, using integrated docking, QSAR analysis, and a molecular dynamics approach. The Monkeypox Virus (MPXV) is a recently emerging outbreak with ongoing infection cases. Drugs and vaccines for smallpox are being used to contain it. However, no specific drugs or vaccines are available to combat this infection. The TMPK and D9 decapping enzymes were retrieved from the MPXV virus UK strain in FASTA format. Due to the unavailability of an experimentally determined structure, the 3D structure was modelled via SWISS-MODEL and further enhanced and validated. The structure was subjected to docking analysis with the derived phytochemicals from Asparagus racemosus using a maestro module. The potential inhibitors were examined via QSAR analysis. Additionally, through MD simulation 250ns, the stability was analyzed, and the MM-GBSA was employed to calculate the binding affinities. The molecular investigation revealed asparoside-C (PubChem ID: 158598) and asparoside-D (PubChem ID: 158597) to be potential hits among others for both targets (TMPK and D9 decapping enzyme) compared to the reference drugs, i.e., tecovirimat, brincidofovir, and cidofovir, possessing antiviral and required bioactivity analyzed via the ADME and QSAR analyses. Moreover, the simulation study of over 250ns revealed strong stability, followed by RMSD, RMSF, etc. The free energy calculation via MMGBSA exhibited strong affinities of asparoside-C and asparoside-D towards the TMPK and the D9 decapping enzyme according to their respective scores. The docking, QSAR, and simulation investigation revealed dual-target inhibitors activity of phytochemicals from Asparagus racemosus towards the MPXV via targeting TMPK and D9 decapping enzyme. It has been observed that asparoside-D and asparoside-C can potentially combat MPXV.
- New
- Research Article
- 10.1016/j.phymed.2025.157323
- Nov 1, 2025
- Phytomedicine : international journal of phytotherapy and phytopharmacology
- Chun-Lan Dai + 11 more
Dual targeting of lipogenesis and PUFAs homeostasis by compound kushen injection suppresses breast cancer bone metastasis.
- New
- Research Article
- 10.1016/j.electacta.2025.147055
- Nov 1, 2025
- Electrochimica Acta
- Barış Dost + 4 more
A dual target voltammetric approach: Simultaneous electrochemical sensing of ammunition stabilizer resorcinol and of dihydroxybenzene isomers by semi-derivative voltammetry
- New
- Research Article
- 10.1016/j.bioorg.2025.109181
- Nov 1, 2025
- Bioorganic chemistry
- Zhichang Xu + 5 more
Exploration of piperazine-azole hybrids as antifungal agents against drug-resistant Candida albicans.
- New
- Research Article
- 10.1002/advs.202513986
- Oct 31, 2025
- Advanced science (Weinheim, Baden-Wurttemberg, Germany)
- Bingjie Zhang + 13 more
The immunosuppressive tumor microenvironment imposes significant metabolic constraints that impair dendritic cell (DC) maturation and antigen presentation, ultimately undermining antitumor immunity. In colorectal cancer (CRC), elevated glutamine uptake by tumor cells depletes extracellular glutamine, thereby limiting DC functionality and disrupting T cell priming. While glutamine antagonists such as JHU083 inhibit tumor metabolism, they are insufficient to fully restore DC activity. Here, the development of T26, a bifunctional immunometabolic prodrug that links JHU083 with the STING agonist MSA-2 via a cleavable amide bond, is reported, enabling synchronized intratumoral release and dual targeting of glutamine metabolism and innate immune activation. In murine CRC models, T26 restores DC maturation, promotes CD8⁺ T cell activation, and reprograms tumor cell-derived extracellular vesicles to enhance antigen presentation and immune stimulation. Importantly, T26 significantly inhibits the growth and proliferation of CRC patient-derived organoids, underscoring its translational potential in human CRC. Notably, T26 also demonstrates strong synergy with chemotherapy, immune checkpoint blockade, and anti-angiogenic therapy, significantly improving tumor control without inducing systemic toxicity. These findings position T26 as a mechanistically integrated and translationally promising strategy to overcome glutamine-driven immune suppression and enhance immunotherapy efficacy in CRC and other metabolically dysregulated malignancies.
- New
- Research Article
- 10.1016/j.intimp.2025.115388
- Oct 30, 2025
- International immunopharmacology
- Jiaqi Ban + 9 more
Oridonin alleviates SiNPs-induced pulmonary fibrosis by inhibiting pyroptosis via IRE1α-XBP1s-NLRP3 pathway.
- New
- Research Article
- 10.1016/j.actbio.2025.10.054
- Oct 29, 2025
- Acta biomaterialia
- Zhouzhou Liao + 9 more
Endometriosis is an estrogen-dependent disease that severely affects the physical and mental health of women of childbearing age. Due to the significant side effects of traditional hormone therapies, non-hormonal treatment strategies are urgently needed. In this study, a targeted liposomal nanosystem (cRGD/iRGD-LP@DP) was developed, co-loaded with the pyroptosis inhibitor disulfiram and the estrogen receptor β (ERβ) antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol (PHTPP), to synergistically block the positive feedback loop between inflammation and estrogen. By modifying the dual-targeting peptides cRGD and iRGD, the nanocarrier can specifically bind to the integrin αvβ3 receptor, which is highly expressed in the lesions and promote the delivery of drugs to deep tissues with the help of the penetration function of iRGD. In vitro experiments showed that cRGD/iRGD-LP@DP significantly inhibited the proliferation and migration of ectopic endometrial stromal cells and the expression of pyroptosis-related proteins (NLRP3/Caspase-1/GSDMD), and reduced the release of inflammatory factors IL-1β and IL-18. In the mouse model, intraperitoneal injection of this nanomedicine could efficiently accumulate in the ectopic lesions, reducing the volume and weight of the lesions without significantly affecting the normal endometrium and ovarian function. Mechanistically, disulfiram blocks pyroptosis by inhibiting the formation of Gasdermin D pores, while PHTPP inhibits the nuclear translocation of ERβ. The two work synergistically to break the vicious cycle between inflammation and estrogen. In addition, this nanosystem has good biosafety and no hepatotoxicity or nephrotoxicity. This study provides an efficient and targeted non-hormonal treatment platform for endometriosis with potential for clinical translation. STATEMENT OF SIGNIFICANCE: Endometriosis is an estrogen-dependent chronic inflammatory disease, and dual targeting of estrogen and inflammation can effectively inhibit the progression of this disease. However, fibrosis of ectopic lesions impedes drug penetration. Additionally, both the estrogen receptor β antagonist PHTPP and the inflammatory inhibitor disulfiram are lipophilic drugs with limited bioavailability. To address this challenge, researchers have developed a nanomedicine modified with cRGD and iRGD dual-targeting peptides. This nanomedicine can specifically bind to integrin αvβ3 receptors, which are highly expressed in ectopic lesions, allowing for deep penetration into the interior of ectopic lesions, and achieving efficient drug delivery. Results demonstrate that this nanomedicine exerts a favorable therapeutic effect on endometriosis, providing an innovative strategy for the non-hormonal treatment of endometriosis.