Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.898
Filter
1.
J Immunother Cancer ; 12(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39089738

ABSTRACT

BACKGROUND: Enhanced glucose metabolism has been reported in many cancers. Glucose-6-phosphate dehydrogenase (G6PD) is a rate-limiting enzyme involved in the pentose phosphate pathway, which maintains NADPH levels and protects cells from oxidative damage. We recently found that low G6PD expression correlates with active tumor immunity. However, the mechanism involving G6PD and tumor immunity remained unclear. METHODS: We conducted in vitro studies using G6PD-knocked down malignant melanoma cells, pathway analysis using the GEO dataset, in vivo studies in combination with immune checkpoint inhibitors (ICIs) using a mouse melanoma model, and prognostic analysis in 42 melanoma patients and 30 lung cancer patients who were treated with ICIs. RESULTS: Inhibition of G6PD, both chemically and genetically, has been shown to decrease the production of NADPH and reduce their oxidative stress tolerance. This leads to cell death, which is accompanied by the release of high mobility group box 1 and the translocation of calreticulin to the plasma membrane. These findings suggested that inhibiting G6PD can induce immunogenic cell death. In experiments with C57BL/6 mice transplanted with G6PD-knockdown B16 melanoma cells and treated with anti-PD-L1 antibody, a significant reduction in tumor size was observed. Interestingly, inhibiting G6PD in only a part of the lesions increased the sensitivity of other lesions to ICI. Additionally, out of 42 melanoma patients and 30 lung cancer patients treated with ICIs, those with low G6PD expression had a better prognosis than those with high G6PD expression (p=0.0473; melanoma, p=0.0287; lung cancer). CONCLUSION: G6PD inhibition is a potent therapeutic strategy that triggers immunogenic cell death in tumors, significantly augmenting the efficacy of immunotherapies.


Subject(s)
Glucosephosphate Dehydrogenase , Immunogenic Cell Death , Immunotherapy , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Animals , Humans , Mice , Immunotherapy/methods , Immunogenic Cell Death/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Female , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Cell Line, Tumor , Male , Melanoma/drug therapy , Melanoma/immunology , Melanoma/pathology
2.
J Exp Clin Cancer Res ; 43(1): 214, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39090759

ABSTRACT

BACKGROUND: Melanoma progression is based on a close interaction between cancer cells and immune cells in the tumor microenvironment (TME). Thus, a better understanding of the mechanisms controlling TME dynamics and composition will help improve the management of this dismal disease. Work from our and other groups has reported the requirement of an active Hedgehog-GLI (HH-GLI) signaling for melanoma growth and stemness. However, the role of the downstream GLI1 transcription factor in melanoma TME remains largely unexplored. METHODS: The immune-modulatory activity of GLI1 was evaluated in a syngeneic B16F10 melanoma mouse model assessing immune populations by flow cytometry. Murine polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were differentiated from bone marrow cells and their immunosuppressive ability was assessed by inhibition of T cells. Conditioned media (CM) from GLI1-overexpressing mouse melanoma cells was used to culture PMN-MDSCs, and the effects of CM were evaluated by Transwell invasion assay and T cell inhibition. Cytokine array analysis, qPCR and chromatin immunoprecipitation were performed to explore the regulation of CX3CL1 expression by GLI1. Human monocyte-derived dendritic cells (moDCs) were cultured in CM from GLI1-silenced patient-derived melanoma cells to assess their activation and recruitment. Blocking antibodies anti-CX3CL1, anti-CCL7 and anti-CXCL8 were used for in vitro functional assays. RESULTS: Melanoma cell-intrinsic activation of GLI1 promotes changes in the infiltration of immune cells, leading to accumulation of immunosuppressive PMN-MDSCs and regulatory T cells, and to decreased infiltration of dendric cells (DCs), CD8 + and CD4 + T cells in the TME. In addition, we show that ectopic expression of GLI1 in melanoma cells enables PMN-MDSC expansion and recruitment, and increases their ability to inhibit T cells. The chemokine CX3CL1, a direct transcriptional target of GLI1, contributes to PMN-MDSC expansion and recruitment. Finally, silencing of GLI1 in patient-derived melanoma cells promotes the activation of human monocyte-derived dendritic cells (moDCs), increasing cytoskeleton remodeling and invasion ability. This phenotype is partially prevented by blocking the chemokine CCL7, but not CXCL8. CONCLUSION: Our findings highlight the relevance of tumor-derived GLI1 in promoting an immune-suppressive TME, which allows melanoma cells to evade the immune system, and pave the way for the design of new combination treatments targeting GLI1.


Subject(s)
Melanoma , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , Zinc Finger Protein GLI1 , Animals , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Mice , Humans , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Melanoma/pathology , Melanoma/metabolism , Melanoma/immunology , Melanoma/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Inbred C57BL
3.
Mikrochim Acta ; 191(7): 435, 2024 06 29.
Article in English | MEDLINE | ID: mdl-38949689

ABSTRACT

A novel scaffold for in situ electrochemical detection of cell biomarkers was developed using electrospun nanofibers and commercial adhesive polymeric membranes. The electrochemical sensing of cell biomarkers requires the cultivation of the cells on/near the (bio)sensor surface in a manner to preserve an appropriate electroactive available surface and to avoid the surface passivation and sensor damage. This can be achieved by employing biocompatible nanofiber meshes that allow the cells to have a normal behavior and do not alter the electrochemical detection. For a better mechanical stability and ease of handling, nylon 6/6 nanofibers were collected on commercial polymeric membranes, at an optimal fiber density, obtaining a double-layered platform. To demonstrate the functionality of the fabricated scaffold, the screening of cellular stress has been achieved integrating melanoma B16-F10 cells and the (bio)sensor components on the transducer whereas the melanin exocytosis was successfully quantified using a commercial electrode. Either directly on the surface of the (bio)sensor or spatially detached from it, the integration of cell cultures in biosensing platforms based on electrospun nanofibers represents a powerful bioanalytical tool able to provide real-time information about the biomarker release, enzyme activity or inhibition, and monitoring of various cellular events.


Subject(s)
Biosensing Techniques , Electrochemical Techniques , Nanofibers , Nanofibers/chemistry , Animals , Mice , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Biosensing Techniques/methods , Cell Line, Tumor , Melanins , Biomarkers/analysis , Tissue Scaffolds/chemistry , Exocytosis , Melanoma, Experimental/pathology , Melanoma, Experimental/diagnosis
4.
Exp Biol Med (Maywood) ; 249: 10081, 2024.
Article in English | MEDLINE | ID: mdl-38974834

ABSTRACT

The lack of effective treatment options for an increasing number of cancer cases highlights the need for new anticancer therapeutic strategies. Immunotherapy mediated by Salmonella enterica Typhimurium is a promising anticancer treatment. Candidate strains for anticancer therapy must be attenuated while retaining their antitumor activity. Here, we investigated the attenuation and antitumor efficacy of two S. enterica Typhimurium mutants, ΔtolRA and ΔihfABpmi, in a murine melanoma model. Results showed high attenuation of ΔtolRA in the Galleria mellonella model, and invasion and survival in tumor cells. However, it showed weak antitumor effects in vitro and in vivo. Contrastingly, lower attenuation of the attenuated ΔihfABpmi strain resulted in regression of tumor mass in all mice, approximately 6 days after the first treatment. The therapeutic response induced by ΔihfABpmi was accompanied with macrophage accumulation of antitumor phenotype (M1) and significant increase in the mRNAs of proinflammatory mediators (TNF-α, IL-6, and iNOS) and an apoptosis inducer (Bax). Our findings indicate that the attenuated ΔihfABpmi exerts its antitumor activity by inducing macrophage infiltration or reprogramming the immunosuppressed tumor microenvironment to an activated state, suggesting that attenuated S. enterica Typhimurium strains based on nucleoid-associated protein genes deletion could be immunotherapeutic against cancer.


Subject(s)
Salmonella typhimurium , Animals , Salmonella typhimurium/immunology , Salmonella typhimurium/genetics , Mice , Mice, Inbred C57BL , Melanoma/immunology , Melanoma/genetics , Melanoma/pathology , Immunotherapy/methods , Macrophages/immunology , Macrophages/metabolism , Cell Line, Tumor , Mutation , Female , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Disease Models, Animal
5.
Theranostics ; 14(10): 3810-3826, 2024.
Article in English | MEDLINE | ID: mdl-38994034

ABSTRACT

Rationale: Surgical resection is a primary treatment for solid tumors, but high rates of tumor recurrence and metastasis post-surgery present significant challenges. Manganese (Mn2+), known to enhance dendritic cell-mediated cancer immunotherapy by activating the cGAS-STING pathway, has potential in post-operative cancer management. However, achieving prolonged and localized delivery of Mn2+ to stimulate immune responses without systemic toxicity remains a challenge. Methods: We developed a post-operative microenvironment-responsive dendrobium polysaccharide hydrogel embedded with Mn2+-pectin microspheres (MnP@DOP-Gel). This hydrogel system releases Mn2+-pectin microspheres (MnP) in response to ROS, and MnP shows a dual effect in vitro: promoting immunogenic cell death and activating immune cells (dendritic cells and macrophages). The efficacy of MnP@DOP-Gel as a post-surgical treatment and its potential for immune activation were assessed in both subcutaneous and metastatic melanoma models in mice, exploring its synergistic effect with anti-PD1 antibody. Result: MnP@DOP-Gel exhibited ROS-responsive release of MnP, which could exert dual effects by inducing immunogenic cell death of tumor cells and activating dendritic cells and macrophages to initiate a cascade of anti-tumor immune responses. In vivo experiments showed that the implanted MnP@DOP-Gel significantly inhibited residual tumor growth and metastasis. Moreover, the combination of MnP@DOP-Gel and anti-PD1 antibody displayed superior therapeutic potency in preventing either metastasis or abscopal brain tumor growth. Conclusions: MnP@DOP-Gel represents a promising drug-free strategy for cancer post-operative management. Utilizing this Mn2+-embedding and ROS-responsive delivery system, it regulates surgery-induced immune responses and promotes sustained anti-tumor responses, potentially increasing the effectiveness of surgical cancer treatments.


Subject(s)
Dendrobium , Hydrogels , Manganese , Mice, Inbred C57BL , Microspheres , Polysaccharides , Animals , Mice , Hydrogels/chemistry , Manganese/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Dendrobium/chemistry , Macrophages/immunology , Macrophages/drug effects , Melanoma/immunology , Melanoma/drug therapy , Melanoma/therapy , Immunotherapy/methods , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor , Female , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Reactive Oxygen Species/metabolism , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/drug therapy
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1088-1097, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977338

ABSTRACT

OBJECTIVE: To investigate the effect of the aqueous extract of Chuan Xiong Rhizoma (CR) on brain metastasis of melanoma B16F10 cells in mice. METHODS: C57BL/6J mouse models of brain metastasis of melanoma were established by ultrasound-guided intraventricular injection of Luc-labeled B16F10 cells, and brain tumor growth was monitored by in vivo imaging. The mouse models were then randomized for daily gavage of saline or aqueous extract of CR (equivalent crude drug concentration of 1 mg/g). Behavioral tests were used to evaluate the neuroprotective effects of CR in the tumor-bearing mice, and the changes in proteins associated with blood-brain barrier integrity, neuronal cell proliferation and apoptosis, and microglial cell apoptosis and activation were observed using immunofluorescence assay. The efficacy of CR combined with temozolomide (25 mg/kg) against brain metastases of B16F10 cells was observed by in vivo imaging. RESULTS: CR-treated mouse models did not show obvious progression of brain metastases and had a reduced rate of body weight loss and lowered protein expressions of ZO-1, claudin-5, occludin, P-gp, TNF-α, AQP4 and PDGFRß. In the behavioral tests, the CR-treated mice showed prolonged stay on the wooden stick with a shortened time of sticky stick removal. Immunofluorescence assay showed increased proliferation and decreased apoptosis of neuronal cells and microglia in CR-treated mice. CR treatment significantly increased the levels of CD86, CD206, IL-4 and IL-10 and decreased the levels of CD163 and IL-1ß in the microenvironment of brain metastases. The mice receiving combined treatments with CR and temozolomide showed significantly lower intensity of fluorescent signals in the brain than those treated with temozolomide alone. CONCLUSION: CR does not promote brain metastasis of melanoma while inducing opening of the blood-brain barrier, and its combined use with TMZ results in enhanced inhibition against brain metastasis of melanoma B16F10 cells in mice.


Subject(s)
Brain Neoplasms , Drugs, Chinese Herbal , Mice, Inbred C57BL , Temozolomide , Animals , Temozolomide/pharmacology , Mice , Brain Neoplasms/secondary , Brain Neoplasms/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Blood-Brain Barrier/drug effects , Apoptosis/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology
7.
Elife ; 122024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973593

ABSTRACT

Pyrimidine nucleotide biosynthesis is a druggable metabolic dependency of cancer cells, and chemotherapy agents targeting pyrimidine metabolism are the backbone of treatment for many cancers. Dihydroorotate dehydrogenase (DHODH) is an essential enzyme in the de novo pyrimidine biosynthesis pathway that can be targeted by clinically approved inhibitors. However, despite robust preclinical anticancer efficacy, DHODH inhibitors have shown limited single-agent activity in phase 1 and 2 clinical trials. Therefore, novel combination therapy strategies are necessary to realize the potential of these drugs. To search for therapeutic vulnerabilities induced by DHODH inhibition, we examined gene expression changes in cancer cells treated with the potent and selective DHODH inhibitor brequinar (BQ). This revealed that BQ treatment causes upregulation of antigen presentation pathway genes and cell surface MHC class I expression. Mechanistic studies showed that this effect is (1) strictly dependent on pyrimidine nucleotide depletion, (2) independent of canonical antigen presentation pathway transcriptional regulators, and (3) mediated by RNA polymerase II elongation control by positive transcription elongation factor B (P-TEFb). Furthermore, BQ showed impressive single-agent efficacy in the immunocompetent B16F10 melanoma model, and combination treatment with BQ and dual immune checkpoint blockade (anti-CTLA-4 plus anti-PD-1) significantly prolonged mouse survival compared to either therapy alone. Our results have important implications for the clinical development of DHODH inhibitors and provide a rationale for combination therapy with BQ and immune checkpoint blockade.


Subject(s)
Antigen Presentation , Dihydroorotate Dehydrogenase , Immune Checkpoint Inhibitors , Animals , Mice , Humans , Antigen Presentation/drug effects , Cell Line, Tumor , Immune Checkpoint Inhibitors/pharmacology , Quinoxalines/pharmacology , Enzyme Inhibitors/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Mice, Inbred C57BL , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Biphenyl Compounds , Quinaldines
8.
Front Immunol ; 15: 1396349, 2024.
Article in English | MEDLINE | ID: mdl-39011040

ABSTRACT

Introduction: Immunogenic cell death (ICD) has emerged as a novel option for cancer immunotherapy. The key determinants of ICD encompass antigenicity (the presence of antigens) and adjuvanticity, which involves the release of damage-associated molecular patterns (DAMPs) and various cytokines and chemokines. CX3CL1, also known as neurotactin or fractalkine, is a chemokine involved in cellular signalling and immune cell interactions. CX3CL1 has been denoted as a "find me" signal that stimulates chemotaxis of immune cells towards dying cells, facilitating efferocytosis and antigen presentation. However, in the context of ICD, it is uncertain whether CX3CL1 is an important mediator of the effects of ICD. Methods: In this study, we investigated the intricate role of CX3CL1 in immunogenic apoptosis induced by mitoxantrone (MTX) in cancer cells. The Luminex xMAP technology was used to quantify murine cytokines, chemokines and growth factors to identify pivotal regulatory cytokines released by murine fibrosarcoma MCA205 and melanoma B16-F10 cells undergoing ICD. Moreover, a murine tumour prophylactic vaccination model was employed to analyse the effect of CX3CL1 on the activation of an adaptive immune response against MCA205 cells undergoing ICD. Furthermore, thorough analysis of the TCGA-SKCM public dataset from 98 melanoma patients revealed the role of CX3CL1 and its receptor CX3CR1 in melanoma patients. Results: Our findings demonstrate enhanced CX3CL1 release from apoptotic MCA205 and B16-F10 cells (regardless of the cell type) but not if they are undergoing ferroptosis or accidental necrosis. Moreover, the addition of recombinant CX3CL1 to non-immunogenic doses of MTX-treated, apoptotically dying cancer cells in the murine prophylactic tumour vaccination model induced a robust immunogenic response, effectively increasing the survival of the mice. Furthermore, analysis of melanoma patient data revealed enhanced survival rates in individuals exhibiting elevated levels of CD8+ T cells expressing CX3CR1. Conclusion: These data collectively underscore the importance of the release of CX3CL1 in eliciting an immunogenic response against dying cancer cells and suggest that CX3CL1 may serve as a key switch in conferring immunogenicity to apoptosis.


Subject(s)
Apoptosis , Chemokine CX3CL1 , Animals , Chemokine CX3CL1/metabolism , Mice , Humans , Cell Line, Tumor , Mice, Inbred C57BL , Melanoma, Experimental/immunology , Female , Immunogenic Cell Death , Cytokines/metabolism
9.
Nanoscale ; 16(28): 13580-13596, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38953490

ABSTRACT

Melanoma is the most invasive and lethal form of skin cancer that arises from the malignant transformation of specialized pigment-producing cell melanocytes. Nanomedicine represents an important prospect to mitigate the difficulties and provide significant benefits to cure melanoma. In the present study, we investigated in vitro and in vivo therapeutic efficacies of copper nitroprusside analogue nanoparticles (abbreviated as CuNPANP) towards melanoma. Initially, in vitro anti-cancer activities of CuNPANP towards melanoma cells (B16F10) were evaluated by several experiments such as [methyl-3H]-thymidine incorporation assay, cell cycle and apoptosis assays using FACS analysis, ROS generation using DCFDA, DHE and DAF2A reagents, internalization of nanoparticles through ICP-OES analysis, co-localization of the nanoparticles using confocal microscopy, JC-1 staining to investigate the mitochondrial membrane potential (MMP) and immunofluorescence studies to analyze the expressions of cytochrome-c, Ki-67, E-cadherin as well as phalloidin staining to analyze the cytoskeletal integrity. Further, the in vivo therapeutic effectiveness of the nanoparticles was established towards malignant melanoma by inoculating B16F10 cells in the dorsal right abdomen of C57BL/6J mice. The intraperitoneal administration of CuNPANP inhibited tumor growth and increased the survivability of melanoma mice. The in vivo immunofluorescence studies (Ki-67, CD-31, and E-cadherin) and TUNEL assay further support the anti-cancer and apoptosis-inducing potential of CuNPANP, respectively. Finally, various signaling pathways and molecular mechanisms involved in anti-cancer activities were further evaluated by Western blot analysis. The results altogether indicated the potential use of copper-based nanomedicines for the treatment of malignant melanoma.


Subject(s)
Apoptosis , Copper , Melanoma, Experimental , Mice, Inbred C57BL , Nitroprusside , Animals , Mice , Cell Line, Tumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Apoptosis/drug effects , Copper/chemistry , Copper/pharmacology , Nitroprusside/pharmacology , Nitroprusside/chemistry , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Nanoparticles/chemistry , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Cell Proliferation/drug effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use
10.
Front Immunol ; 15: 1410564, 2024.
Article in English | MEDLINE | ID: mdl-39007148

ABSTRACT

Background: Immune checkpoint blockade (ICB) is rapidly becoming a standard of care in the treatment of many cancer types. However, the subset of patients who respond to this type of therapy is limited. Another way to promote antitumoral immunity is the use of immunostimulatory molecules, such as cytokines or T cell co-stimulators. The systemic administration of immunotherapeutics leads to significant immune-related adverse events (irAEs), therefore, the localized antitumoral action is needed. One way to achieve this is intratumoral non-viral gene-immune therapy, which allows for prolonged and localized gene expression, and multiple drug administration. In this study, we combined the previously described non-viral gene delivery system, PEG-PEI-TAT copolymer, PPT, with murine OX40L-encoding plasmid DNA. Methods: The resulting OX40L/PPT nanoparticles were characterized via gel mobility assay, dynamic light scattering analysis and in vitro transfection efficiency evaluation. The antitumoral efficacy of intratumorally (i.t.) administered nanoparticles was estimated using subcutaneously (s.c.) implanted CT26 (colon cancer), B16F0 (melanoma) and 4T1 (breast cancer) tumor models. The dynamics of stromal immune cell populations was analyzed using flow cytometry. Weight loss and cachexia were used as irAE indicators. The effect of combination of i.t. OX40L/PPT with intraperitoneal PD-1 ICB was estimated in s.c. CT26 tumor model. Results: The obtained OX40L/PPT nanoparticles had properties applicable for cell transfection and provided OX40L protein expression in vitro in all three investigated cancer models. We observed that OX40L/PPT treatment successfully inhibited tumor growth in B16F0 and CT26 tumor models and showed a tendency to inhibit 4T1 tumor growth. In B16F0 tumor model, OX40L/PPT treatment led to the increase in antitumoral effector NK and T killer cells and to the decrease in pro-tumoral myeloid cells populations within tumor stroma. No irAE signs were observed in all 3 tumor models, which indicates good treatment tolerability in mice. Combining OX40L/PPT with PD-1 ICB significantly improved treatment efficacy in the CT26 subcutaneous colon cancer model, providing protective immunity against CT26 colon cancer cells. Conclusion: Overall, the anti-tumor efficacy observed with OX40L non-viral gene therapy, whether administered alone or in combination with ICB, highlights its potential to revolutionize cancer gene therapy, thus paving the way for unprecedented advancements in the cancer therapy field.


Subject(s)
Immunotherapy , OX40 Ligand , Animals , OX40 Ligand/genetics , Mice , Immunotherapy/methods , Cell Line, Tumor , Female , Genetic Therapy/methods , Nanoparticles , Gene Transfer Techniques , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Microenvironment/immunology , Polyethyleneimine/chemistry , Humans , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Polyethylene Glycols/chemistry
11.
Chem Biol Drug Des ; 104(2): e14594, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39072923

ABSTRACT

Melanoma is one of the most aggressive and fatal skin cancers owing to its ability to metastasize and develop resistance to chemotherapy. Photodynamic therapy (PDT) is a minimally noninvasive treatment modality comprising photosensitizers (PSs), light sources, and endogenous molecular oxygen that exert a localized cytotoxic effect on cancer cells. The current study explores the therapeutic potential of sodium copper chlorophyllin-loaded chitosan nanoparticles (CH-SCC NPs) along with handheld laser-based PDT on B16 cancer cells. A modified chlorophyll derivative identified as sodium copper chlorophyllin (SCC) is a dietary supplement that has anticancer properties. Herein, we have synthesized CH-SCC NPs using the ionic gelation method to enhance the PS's bioavailability and efficiency. Chitosan nanoparticles exhibited high biocompatibility in a normal cell line L929, zebrafish, and chick embryos, and were successfully employed to deliver the SCC to cancer cells. CH-SCC NPs showed an enhanced PDT effect that killed approximately 80%-85% of B16 cells. CH-SCC NPs in combination with a handheld portable laser source showed significant therapeutic potential against the B16 skin cancer cell line. The experimental findings further strengthen our device-repurposing strategy, which suggests that SCC nanoformulations along with handheld laser can be a suitable treatment for skin cancer even in remote areas where power source and treatment cost can be a limitation.


Subject(s)
Chitosan , Chlorophyllides , Melanoma, Experimental , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Zebrafish , Animals , Nanoparticles/chemistry , Chitosan/chemistry , Mice , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Cell Line, Tumor , Chick Embryo , Copper/chemistry , Copper/pharmacology , Cell Survival/drug effects
12.
J Immunother Cancer ; 12(7)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043603

ABSTRACT

BACKGROUND: Lymphocyte activation gene 3 (LAG-3) is expressed on activated immune cells and has emerged as a promising target for immune checkpoints blockade. However, conflicting findings have been reported regarding the association between LAG-3 expression in tumors and patient prognosis, indicating the need for further investigation into the significance of LAG-3 expression levels in tumor therapies. In this study, 68Ga-NOTA-XH05, a novel peptide-based positron emission tomography (PET) tracer targeting LAG-3, was constructed to non-invasively detect LAG-3 expression in melanoma after CpG oligonucleotide (CpG) treatment and explore the relationship between LAG-3 expression and therapeutic effect. METHODS: The tracer 68Ga-NOTA-XH05 was identified by high-performance liquid chromatography after being prepared and purified. Cell uptake and blocking essays were performed to verify the specificity of the tracer in vitro. The expression of LAG-3 in B16-F10 subcutaneous tumors was monitored by flow cytometry, and its correlation with the tracer uptake was analyzed to evaluate the tracer specificity. PET imaging and biodistribution studies were conducted after CpG treatment of unilateral or bilateral B16-F10 subcutaneous tumor models to assess the ability of 68Ga-NOTA-XH05 in monitoring immunotherapy efficacy and the abscopal effect of CpG. RESULTS: Following purification, 68Ga-NOTA-XH05 exhibited high radiochemical purity and specificity. Flow cytometry analysis revealed a positive correlation between LAG-3 expression in tumors and the uptake of 68Ga-NOTA-XH05. In B16-F10 bearing mice treated with CpG, PET imaging using 68Ga-NOTA-XH05 demonstrated a higher tumor to blood ratio (TBR) compared with the control group. Furthermore, TBR values obtained from CpG-treated mice allowed for differentiation between responders and non-responders. In a bilateral subcutaneous tumor model where only right-sided tumors were treated with intratumoral injection of CpG, TBR values of left-sided tumors were significantly higher than those in the control group, indicating that 68Ga-NOTA-XH05 could effectively monitor the systemic effect of local CpG injection. CONCLUSION: Our findings highlight the detection capability of 68Ga-NOTA-XH05 in assessing LAG-3 expression levels within tumors and evaluating response to immunotherapy, thereby suggesting promising clinical translational prospects.


Subject(s)
Immunotherapy , Lymphocyte Activation Gene 3 Protein , Positron-Emission Tomography , Animals , Mice , Positron-Emission Tomography/methods , Immunotherapy/methods , Humans , Antigens, CD/metabolism , Cell Line, Tumor , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/metabolism , Peptides , Gallium Radioisotopes , Melanoma/diagnostic imaging , Melanoma/immunology , Melanoma/drug therapy , Melanoma/metabolism , Female , Radiopharmaceuticals
13.
Int J Pharm ; 661: 124380, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38950661

ABSTRACT

Melanoma is an aggressive form of skin cancer with elevated propensity to metastasize. One of the major critical issues in the treatment of oncological patients is represented by the development of toxicity and resistance to the available therapies. Great progress has been made in the field of nanotechnologies to limit the unwanted effects of anti-cancer treatments. We explored the potential of creating oil-in-water nanoemulsions composed of oleic acid, as a bioactive carrier for lipophilic drug delivery. This bioactive nanoemulsion was loaded with Curcumin, a natural fluorescent lipophilic compound, used as a model drug to evaluate nanoemulsion capability to: i) encapsulate the lipophilic moiety; ii) interact with the specific cells, and iii) improve the efficacy of the loaded model drug compared to the free one. Therefore, we evaluated the physical-chemical features of Curcumin-loaded nanoemulsions, confirming their pH sensibility and their stability over time. Moreover, the nanoemulsions were able to preserve the loaded Curcumin by degradation/destabilization phenomena. Finally, we verified some of the biological functions of Curcumin delivered by nanoemulsions in the B16F10 melanoma cell line. We obtained evidence of the biological action of Curcumin, suggesting oleic-based nanoemulsions as an efficient nanocarrier for lipophilic drug delivery.


Subject(s)
Curcumin , Emulsions , Melanoma, Experimental , Nanoparticles , Oleic Acid , Curcumin/administration & dosage , Curcumin/chemistry , Curcumin/pharmacology , Hydrogen-Ion Concentration , Cell Line, Tumor , Oleic Acid/chemistry , Animals , Mice , Melanoma, Experimental/drug therapy , Nanoparticles/chemistry , Drug Carriers/chemistry , Cell Survival/drug effects , Melanoma/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Delivery Systems
14.
Int J Pharm ; 661: 124422, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38977163

ABSTRACT

The challenges posed by intractable relapse and metastasis in cancer treatment have led to the development of various forms of photodynamic therapy (PDT). However, traditional drug delivery systems, such as virus vectors, liposomes, and polymers, often suffer from issues like desynchronized drug release, carrier instability, and drug leakage during circulation. To address these problems, we have developed a dual-prodrug nanogel (PVBN) consisting of Pyro (Pyropheophorbide a) and SAHA (Vorinostat) bound to BSA (Bovine Serum Albumin), which facilitates synchronous and spontaneous drug release in situ within the lysosome. Detailed results indicate that PVBN-treated tumor cells exhibit elevated levels of ROS and Acetyl-H3, leading to necrosis, apoptosis, and cell cycle arrest, with PDT playing a dominant role in the synergistic therapeutic effect. Furthermore, the anti-tumor efficacy of PVBN was validated in melanoma-bearing mice, where it significantly inhibited tumor growth and pulmonary metastasis. Overall, our dual-prodrug nanogel, formed by the binding of SAHA and Pyro to BSA and releasing drugs within the lysosome, represents a novel and promising strategy for enhancing the clinical efficacy of photochemotherapy.


Subject(s)
Chlorophyll , Nanogels , Photochemotherapy , Prodrugs , Serum Albumin, Bovine , Vorinostat , Animals , Vorinostat/administration & dosage , Vorinostat/pharmacology , Vorinostat/chemistry , Photochemotherapy/methods , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , Chlorophyll/administration & dosage , Chlorophyll/pharmacology , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/administration & dosage , Cell Line, Tumor , Nanogels/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Mice , Apoptosis/drug effects , Drug Liberation , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Reactive Oxygen Species/metabolism , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Mice, Inbred C57BL , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma, Experimental/drug therapy , Polyethyleneimine/chemistry
15.
Biomed Pharmacother ; 177: 117083, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38968793

ABSTRACT

Cancer stem cells (CSCs) can self-renew and differentiate, contributing to tumor heterogeneity, metastasis, and recurrence. Their resistance to therapies, including immunotherapy, underscores the importance of targeting them for complete remission and relapse prevention. Olfactomedin 4 (OLFM4), a marker associated with various cancers such as colorectal cancer, is expressed on CSCs promoting immune evasion and tumorigenesis. However, its potential as a target for CSC-specific immunotherapy remains underexplored. The primary aim of this study is to evaluate the effectiveness of targeting OLFM4 with dendritic cell (DC)-based vaccines in inhibiting tumor growth and metastasis. To improve antigen delivery and immune response, OLFM4 was conjugated with a protein-transduction domain (PTD) from the antennapedia of Drosophila called penetratin, creating a fusion protein (P-OLFM4). The efficacy of DCs pulsed with P-OLFM4 (DCs [P-OLFM4]) was compared to DCs pulsed with OLFM4 (DCs [OLFM4]) and PBS (DCs [PBS]). DCs [P-OLFM4] inhibited tumor growth by 91.2 % and significantly reduced lung metastasis of OLFM4+ melanoma cells by 97 %, compared to the DCs [PBS]. DCs [OLFM4] also demonstrated a reduction in lung metastasis by 59.7 % compared to DCs [PBS]. Immunization with DCs [P-OLFM4] enhanced OLFM4-specific T-cell proliferation, interferon-γ production, and cytotoxic T cell activity in mice. The results indicate that OLFM4 is a viable target for CSC-focused immunotherapy. DC [P-OLFM4] vaccines can elicit robust immune responses, significantly inhibiting tumor growth and metastasis. This strategy holds promise for developing more effective cancer treatments that specifically target CSCs, potentially leading to better patient outcomes by reducing the likelihood of tumor relapse and metastasis.


Subject(s)
Dendritic Cells , Mice, Inbred C57BL , Animals , Dendritic Cells/immunology , Mice , Cell-Penetrating Peptides , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Female , Cell Line, Tumor , Cell Proliferation , Immunotherapy/methods , Lung Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Neoplasm Metastasis
16.
Viruses ; 16(7)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39066315

ABSTRACT

To explore whether the p17 protein of oncolytic avian reovirus (ARV) mediates cell migration and invadopodia formation, we applied several molecular biological approaches for studying the involved cellular factors and signal pathways. We found that ARV p17 activates the p53/phosphatase and tensin homolog (PTEN) pathway to suppress the focal adhesion kinase (FAK)/Src signaling and downstream signal molecules, thus inhibiting cell migration and the formation of invadopodia in murine melanoma cancer cell line (B16-F10). Importantly, p17-induced formation of invadopodia could be reversed in cells transfected with the mutant PTENC124A. p17 protein was found to significantly reduce the expression levels of tyrosine kinase substrate 5 (TKs5), Rab40b, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), and matrix metalloproteinases (MMP9), suggesting that TKs5 and Rab40b were transcriptionally downregulated by p17. Furthermore, we found that p17 suppresses the formation of the TKs5/NCK1 complex. Coexpression of TKs5 and Rab40b in B16-F10 cancer cells reversed p17-modulated suppression of the formation of invadopodia. This work provides new insights into p17-modulated suppression of invadopodia formation by activating the p53/PTEN pathway, suppressing the FAK/Src pathway, and inhibiting the formation of the TKs5/NCK1 complex.


Subject(s)
Cell Movement , Focal Adhesion Kinase 1 , Orthoreovirus, Avian , Podosomes , Signal Transduction , Animals , Mice , Orthoreovirus, Avian/physiology , Orthoreovirus, Avian/genetics , Cell Line, Tumor , Podosomes/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Oncolytic Viruses/physiology , Oncolytic Viruses/genetics , src-Family Kinases/metabolism , src-Family Kinases/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics
17.
Anticancer Res ; 44(8): 3295-3306, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060064

ABSTRACT

BACKGROUND/AIM: Despite the established antitumor effectiveness and synergistic interactions of melatonin with photon irradiation, its role in carbon-ion radiotherapy remains uncertain. This study aimed to elucidate the mechanisms and potential clinical advantages of combining exogenous melatonin therapy with carbon-ion radiotherapy. MATERIALS AND METHODS: The investigation assessed the impact of combining exogenous melatonin with photon or carbon-ion irradiation on cell-cycle modulation and DNA-repair capability using the melanoma cell line B16F10. RNA sequencing and bioinformatics analysis were conducted to explore mechanisms and evaluate potential clinical benefits, with validation performed on the osteosarcoma cell line LM8. RESULTS: Pre-treatment with melatonin reduced the survival fraction of B16F10 and LM8 cells upon exposure to photon and carbon-ion radiation. Mechanistically, melatonin was found to inhibit G2/M arrest, preserve DNA damage, and suppress key genes involved in DNA double-strand break repair after 8 Gy carbon-ion radiation. Furthermore, RNA sequencing and bioinformatics analysis revealed favorable changes in genes associated with survival and metastasis, highlighting potential clinical significance. LM8 cells treated with melatonin exhibited increased radiosensitivity and suppression of DNA-repair proteins. CONCLUSION: The combination of exogenous melatonin not only heightened radiosensitivity and modulated hallmark tumor gene sets in vitro but also markedly suppressed the efficiency of DNA double-strand break-repair pathway, thus enhancing the cytotoxicity of carbon-ion radiotherapy.


Subject(s)
DNA Repair , Heavy Ion Radiotherapy , Melatonin , Radiation Tolerance , Radiation-Sensitizing Agents , Melatonin/pharmacology , Cell Line, Tumor , Radiation Tolerance/drug effects , Mice , Animals , Humans , DNA Repair/drug effects , DNA Repair/radiation effects , Radiation-Sensitizing Agents/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects
18.
Biomed Pharmacother ; 177: 117063, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38968800

ABSTRACT

Off-targeting toxicity and immunosuppressive tumor microenvironment still restrict the therapeutic requirement of photodynamic therapy (PDT). The development of metal ion-coordination-based nanoparticles (NPs) for cancer therapy has advantages, such as precious nanostructure and potent therapeutic effect as well as great safety. In this study, we prepared calcium ions (Ca2+)-coordination photosensitizer NPs, based on Ca2+-pyrochloric acid (PPA)-coordination as the new photosensitive nanoamplifiers, and microneedles (MNs) as the personalized apparatus, and investigated the nanoamplifiers for treating the melanoma via transdermal administration. This nanoamplifiers was synthesized via a simple coordination of Ca2+ and PPA with the addition of bovine serum albumin (BSA), and further fabricated into MNs (nanoamplifiers@MNs). Following inserted into the tumor, the released nanoamplifiers from the tips and back layer exhibited great photodynamic activity under irradiation, inducing cancer cell death. Meanwhile, Ca2+ acted as the second messenger, promoting M1 polarization of macrophages and maturation of dendritic cells (DCs), thereby enhancing the immune activation effect in the tumor microenvironment. As a result, such nanoamplifiers effectively achieved significant efficacy against malignant melanoma tumors by synergistically tumor killing and potent anti-tumor immune activation without obviously side effect. This work demonstrated the potential of MNs-mediated metal ion-coordination-based nanoamplifier as a novel photodynamic therapeutic platform for the efficient and safe treatment of cancer.


Subject(s)
Calcium , Needles , Photochemotherapy , Photosensitizing Agents , Tumor Microenvironment , Photochemotherapy/methods , Tumor Microenvironment/drug effects , Animals , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Calcium/metabolism , Mice , Cell Line, Tumor , Nanoparticles/chemistry , Mice, Inbred C57BL , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Melanoma/drug therapy , Melanoma/pathology , Dendritic Cells/drug effects , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology
19.
Biosci Biotechnol Biochem ; 88(8): 900-907, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38835135

ABSTRACT

Melanoma, a cancer arising from melanocytes, requires a novel treatment strategy because of the ineffectiveness of conventional therapies in certain patients. Fustin is a flavanonol found in young fustic (Cotinus coggygria). However, little is known about its antimelanoma effects. Our study demonstrates that fustin suppresses the growth of B16 melanoma cells. Phalloidin staining of cytoskeletal actin revealed that fustin induced a conformational change in the actin structure of melanoma cells, accompanied by suppressed phosphorylation of myosin regulatory light chain 2 (MLC2), a regulator of actin structure. Furthermore, the protein kinase A (cAMP-dependent protein kinase) inhibitor H89 completely attenuated fustin-induced downregulation of phosphorylated myosin phosphatase targeting subunit 1, which is involved in dephosphorylation of MLC2. In a mouse model, administration of fustin suppressed tumor growth in B16 melanoma cells without adverse effects. In conclusion, our findings suggest that fustin effectively suppresses melanoma cell growth both in vitro and in vivo.


Subject(s)
Cell Proliferation , Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Melanoma, Experimental , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/drug therapy , Cell Line, Tumor , Mice , Phosphorylation/drug effects , Myosin Light Chains/metabolism , Cardiac Myosins/metabolism , Flavonoids/pharmacology , Isoquinolines/pharmacology , Actins/metabolism , Sulfonamides/pharmacology , Humans , Melanoma/pathology , Melanoma/metabolism , Melanoma/drug therapy , Mice, Inbred C57BL
20.
Front Immunol ; 15: 1345046, 2024.
Article in English | MEDLINE | ID: mdl-38827732

ABSTRACT

Introduction: Recently, more and more research illustrated the importance of inducing CD4+ T helper type (Th)-1 dominant immunity for the success of tumor immunotherapy. Our prior studies revealed the crucial role of CD4+ Th1 cells in orchestrating systemic and durable antitumor immunity, which contributes to the satisfactory outcomes of the novel cryo-thermal therapy in the B16F10 tumor model. However, the mechanism for maintaining the cryo-thermal therapy-mediated durable CD4+ Th1-dominant response remains uncovered. Additionally, cryo-thermal-induced early-stage CD4+ Th1-dominant T cell response showed a correlation with the favorable prognosis in patients with colorectal cancer liver metastasis (CRCLM). We hypothesized that CD4+ Th1-dominant differentiation induced during the early stage post cryo-thermal therapy would affect the balance of CD4+ subsets at the late phase. Methods: To understand the role of interferon (IFN)-γ, the major effector of Th1 subsets, in maintaining long-term CD4+ Th1-prone polarization, B16F10 melanoma model was established in this study and a monoclonal antibody was used at the early stage post cryo-thermal therapy for interferon (IFN)-γ signaling blockade, and the influence on the phenotypic and functional change of immune cells was evaluated. Results: IFNγ at the early stage after cryo-thermal therapy maintained long-lasting CD4+ Th1-prone immunity by directly controlling Th17, Tfh, and Tregs polarization, leading to the hyperactivation of Myeloid-derived suppressor cells (MDSCs) represented by abundant interleukin (IL)-1ß generation, and thereby further amplifying Th1 response. Discussion: Our finding emphasized the key role of early-phase IFNγ abundance post cryo-thermal therapy, which could be a biomarker for better prognosis after cryo-thermal therapy.


Subject(s)
Cell Differentiation , Interferon-gamma , Melanoma, Experimental , Mice, Inbred C57BL , Th1 Cells , Animals , Th1 Cells/immunology , Mice , Interferon-gamma/metabolism , Cell Differentiation/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Cryotherapy/methods , Cell Line, Tumor , Female
SELECTION OF CITATIONS
SEARCH DETAIL