Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.956
Filter
1.
Signal Transduct Target Ther ; 9(1): 118, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702343

ABSTRACT

Antitumor therapies based on adoptively transferred T cells or oncolytic viruses have made significant progress in recent years, but the limited efficiency of their infiltration into solid tumors makes it difficult to achieve desired antitumor effects when used alone. In this study, an oncolytic virus (rVSV-LCMVG) that is not prone to induce virus-neutralizing antibodies was designed and combined with adoptively transferred T cells. By transforming the immunosuppressive tumor microenvironment into an immunosensitive one, in B16 tumor-bearing mice, combination therapy showed superior antitumor effects than monotherapy. This occurred whether the OV was administered intratumorally or intravenously. Combination therapy significantly increased cytokine and chemokine levels within tumors and recruited CD8+ T cells to the TME to trigger antitumor immune responses. Pretreatment with adoptively transferred T cells and subsequent oncolytic virotherapy sensitizes refractory tumors by boosting T-cell recruitment, down-regulating the expression of PD-1, and restoring effector T-cell function. To offer a combination therapy with greater translational value, mRNA vaccines were introduced to induce tumor-specific T cells instead of adoptively transferred T cells. The combination of OVs and mRNA vaccine also displays a significant reduction in tumor burden and prolonged survival. This study proposed a rational combination therapy of OVs with adoptive T-cell transfer or mRNA vaccines encoding tumor-associated antigens, in terms of synergistic efficacy and mechanism.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Animals , Mice , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Virotherapy/methods , Combined Modality Therapy , mRNA Vaccines/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Tumor Microenvironment/immunology , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/genetics , Cancer Vaccines/administration & dosage
2.
J Nanobiotechnology ; 22(1): 230, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720322

ABSTRACT

Tumor vaccines, a crucial immunotherapy, have gained growing interest because of their unique capability to initiate precise anti-tumor immune responses and establish enduring immune memory. Injected tumor vaccines passively diffuse to the adjacent draining lymph nodes, where the residing antigen-presenting cells capture and present tumor antigens to T cells. This process represents the initial phase of the immune response to the tumor vaccines and constitutes a pivotal determinant of their effectiveness. Nevertheless, the granularity paradox, arising from the different requirements between the passive targeting delivery of tumor vaccines to lymph nodes and the uptake by antigen-presenting cells, diminishes the efficacy of lymph node-targeting tumor vaccines. This study addressed this challenge by employing a vaccine formulation with a tunable, controlled particle size. Manganese dioxide (MnO2) nanoparticles were synthesized, loaded with ovalbumin (OVA), and modified with A50 or T20 DNA single strands to obtain MnO2/OVA/A50 and MnO2/OVA/T20, respectively. Administering the vaccines sequentially, upon reaching the lymph nodes, the two vaccines converge and simultaneously aggregate into MnO2/OVA/A50-T20 particles through base pairing. This process enhances both vaccine uptake and antigen delivery. In vitro and in vivo studies demonstrated that, the combined vaccine, comprising MnO2/OVA/A50 and MnO2/OVA/T20, exhibited robust immunization effects and remarkable anti-tumor efficacy in the melanoma animal models. The strategy of controlling tumor vaccine size and consequently improving tumor antigen presentation efficiency and vaccine efficacy via the DNA base-pairing principle, provides novel concepts for the development of efficient tumor vaccines.


Subject(s)
Cancer Vaccines , Lymph Nodes , Manganese Compounds , Mice, Inbred C57BL , Nanoparticles , Ovalbumin , Oxides , Animals , Cancer Vaccines/immunology , Lymph Nodes/immunology , Mice , Ovalbumin/immunology , Ovalbumin/chemistry , Oxides/chemistry , Nanoparticles/chemistry , Manganese Compounds/chemistry , Immunity, Cellular , Female , Cell Line, Tumor , DNA/chemistry , DNA/immunology , Immunotherapy/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Particle Size , Antigens, Neoplasm/immunology
3.
Elife ; 122024 May 28.
Article in English | MEDLINE | ID: mdl-38805560

ABSTRACT

Solid tumors generally exhibit chromosome copy number variation, which is typically caused by chromosomal instability (CIN) in mitosis. The resulting aneuploidy can drive evolution and associates with poor prognosis in various cancer types as well as poor response to T-cell checkpoint blockade in melanoma. Macrophages and the SIRPα-CD47 checkpoint are understudied in such contexts. Here, CIN is induced in poorly immunogenic B16F10 mouse melanoma cells using spindle assembly checkpoint MPS1 inhibitors that generate persistent micronuclei and diverse aneuploidy while skewing macrophages toward a tumoricidal 'M1-like' phenotype based on markers and short-term anti-tumor studies. Mice bearing CIN-afflicted tumors with wild-type CD47 levels succumb similar to controls, but long-term survival is maximized by SIRPα blockade on adoptively transferred myeloid cells plus anti-tumor monoclonal IgG. Such cells are the initiating effector cells, and survivors make de novo anti-cancer IgG that not only promote phagocytosis of CD47-null cells but also suppress tumor growth. CIN does not affect the IgG response, but pairing CIN with maximal macrophage anti-cancer activity increases durable cures that possess a vaccination-like response against recurrence.


Subject(s)
Chromosomal Instability , Immunoglobulin G , Macrophages , Animals , Mice , Macrophages/immunology , CD47 Antigen/metabolism , CD47 Antigen/genetics , CD47 Antigen/immunology , Mice, Inbred C57BL , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/genetics , Cell Line, Tumor , Female
4.
Biochem Biophys Res Commun ; 718: 150058, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38729076

ABSTRACT

The therapeutic efficacy of radiotherapy (RT) is primarily driven by two factors: biophysical DNA damage in cancer cells and radiation-induced anti-tumor immunity. However, Anti-tumor immune responses between X-ray RT (XRT) and carbon-ion RT (CIRT) remain unclear. In this study, we, employed mouse models to assess the immunological contribution, especially cytotoxic T-lymphocyte (CTL)-mediated immunity, to the therapeutic effectiveness of XRT and CIRT in shrinking tumors. We irradiated mouse intradermal tumors of B16F10-ovalbumin (OVA) mouse melanoma cells and 3LL-OVA mouse lung cancer cells with carbon-ion beams or X-rays in the presence or absence of CTLs. CTL removal was performed by administration of anti-CD8 monoclonal antibody (mAb) in mice. Based on tumor growth delay, we determined the tumor growth and regression curves. The enhancement ratio (ER) of the slope of regression lines in the presence of CTLs, relative to the absence of CTLs, indicates the dependency of RT on CTLs for shrinking mouse tumors, and the biological effectiveness (RBE) of CIRT relative to XRT were calculated. Tumor growth curves revealed that the elimination of CD8+ CTLs by administrating anti-CD8 mAb accelerated tumor growth compared to the presence of CTLs in both RTs. The ERs were larger in CIRT compared to XRT in the B16F10-OVA tumor models, but not in the 3LL-OVA models, suggesting a greater contribution of CTL-mediated anti-tumor immunity to tumor reduction in CIRT compared to XRT in the B16F10-OVA tumor model. In addition, the RBE values for both models were larger in the presence of CTLs compared to models without CTLs, suggesting that CIRT may utilize CTL-mediated anti-tumor immunity more than X-ray. The findings from this study suggest that although immunological contribution to therapeutic efficacy may vary depending on the type of tumor cell, CIRT utilizes CTL-mediated immunity to a greater extent compared to XRT.


Subject(s)
Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic , Animals , T-Lymphocytes, Cytotoxic/immunology , Mice , Cell Line, Tumor , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Heavy Ion Radiotherapy/methods , X-Ray Therapy , Female , Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Lung Neoplasms/pathology
5.
Nat Commun ; 15(1): 4444, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789421

ABSTRACT

Mitochondrial respiration is essential for the survival and function of T cells used in adoptive cellular therapies. However, strategies that specifically enhance mitochondrial respiration to promote T cell function remain limited. Here, we investigate methylation-controlled J protein (MCJ), an endogenous negative regulator of mitochondrial complex I expressed in CD8 cells, as a target for improving the efficacy of adoptive T cell therapies. We demonstrate that MCJ inhibits mitochondrial respiration in murine CD8+ CAR-T cells and that deletion of MCJ increases their in vitro and in vivo efficacy against murine B cell leukaemia. Similarly, MCJ deletion in ovalbumin (OVA)-specific CD8+ T cells also increases their efficacy against established OVA-expressing melanoma tumors in vivo. Furthermore, we show for the first time that MCJ is expressed in human CD8 cells and that the level of MCJ expression correlates with the functional activity of CD8+ CAR-T cells. Silencing MCJ expression in human CD8 CAR-T cells increases their mitochondrial metabolism and enhances their anti-tumor activity. Thus, targeting MCJ may represent a potential therapeutic strategy to increase mitochondrial metabolism and improve the efficacy of adoptive T cell therapies.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy, Adoptive , Mitochondria , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Humans , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Cell Respiration , Cell Line, Tumor , Female , Ovalbumin/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy
6.
Cancer Lett ; 592: 216934, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38710299

ABSTRACT

The Staphylococcal nuclease and Tudor domain containing 1 (SND1) has been identified as an oncoprotein. Our previous study demonstrated that SND1 impedes the major histocompatibility complex class I (MHC-I) assembly by hijacking the nascent heavy chain of MHC-I to endoplasmic reticulum-associated degradation. Herein, we aimed to identify inhibitors to block SND1-MHC-I binding, to facilitate the MHC-I presentation and tumor immunotherapy. Our findings validated the importance of the K490-containing sites in SND1-MHC-I complex. Through structure-based virtual screening and docking analysis, (-)-Epigallocatechin (EGC) exhibited the highest docking score to prevent the binding of MHC-I to SND1 by altering the spatial conformation of SND1. Additionally, EGC treatment resulted in increased expression levels of membrane-presented MHC-I in tumor cells. The C57BL/6J murine orthotopic melanoma model validated that EGC increases infiltration and activity of CD8+ T cells in both the tumor and spleen. Furthermore, the combination of EGC with programmed death-1 (PD-1) antibody demonstrated a superior antitumor effect. In summary, we identified EGC as a novel inhibitor of SND1-MHC-I interaction, prompting MHC-I presentation to improve CD8+ T cell response within the tumor microenvironment. This discovery presents a promising immunotherapeutic candidate for tumors.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Catechin , Endonucleases , Mice, Inbred C57BL , Animals , CD8-Positive T-Lymphocytes/immunology , Mice , Humans , Antigen Presentation/immunology , Endonucleases/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Molecular Docking Simulation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism
7.
Nanotechnology ; 35(29)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38593752

ABSTRACT

Melanoma is one of the most aggressive and lethal types of cancer owing to its metastatic propensity and chemoresistance property. An alternative therapeutic option is photodynamic and photothermal therapies (PDT/PTT), which employ near-infrared (NIR) light to generate heat and reactive oxygen species (ROS). As per previous reports, Melanin (Mel), and its synthetic analogs (i.e. polydopamine nanoparticles) can induce NIR light-mediated heat energy, thereby selectively targeting and ameliorating cancer cells. Similarly, chlorin e6 (Ce6) also has high ROS generation ability and antitumor activity against various types of cancer. Based on this tenet, In the current study, we have encapsulated Mel-Ce6 in a polydopamine (PDA) nanocarrier (MCP NPs) synthesized by the oxidation polymerization method. The hydrodynamic diameter of the synthesized spherical MCP NPs was 139 ± 10 nm. The MCP NPs, upon irradiation with NIR 690 nm laser for 6 min, showed photothermal efficacy of more than 50 °C. Moreover, the red fluorescence in the MCP NPs due to Ce6 can be leveraged for diagnostic purposes. Further, the MCP NPs exhibited considerable biocompatibility with the L929 cell line and exerted nearly 70% ROS-mediated cytotoxicity on the B16 melanoma cell line after the laser irradiation. Thus, the prepared MCP NPs could be a promising theranostic agent for treating the B16 melanoma cancer.


Subject(s)
Chlorophyllides , Indoles , Melanins , Melanoma, Experimental , Nanoparticles , Polymers , Porphyrins , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Polymers/pharmacology , Nanoparticles/chemistry , Animals , Mice , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Cell Line, Tumor , Porphyrins/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Phototherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photochemotherapy/methods , Photothermal Therapy
8.
Pharmacol Res ; 203: 107174, 2024 May.
Article in English | MEDLINE | ID: mdl-38580185

ABSTRACT

The emergence of immune checkpoint inhibitors (ICIs) has revolutionized the clinical treatment for tumor. However, the low response rate of ICIs remains the major obstacle for curing patients and effective approaches for patients with primary or secondary resistance to ICIs remain lacking. In this study, immune stimulating agent unmethylated CG-enriched (CpG) oligodeoxynucleotide (ODN) was locally injected into the tumor to trigger a robust immune response to eradicate cancer cells, while anti-CD25 antibody was applied to remove immunosuppressive regulatory T cells, which further enhanced the host immune activity to attack tumor systematically. The combination of CpG and anti-CD25 antibody obtained notable regression in mouse melanoma model. Furthermore, rechallenge of tumor cells in the xenograft model has resulted in smaller tumor volume, which demonstrated that the combinational treatment enhanced the activity of memory T cells. Remarkably, this combinational therapy presented significant efficacy on multiple types of tumors as well and was able to prevent relapse of tumor partially. Taken together, our combinational immunotherapy provides a new avenue to enhance the clinical outcomes of patients who are insensitive or resistant to ICIs treatments.


Subject(s)
Oligodeoxyribonucleotides , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Oligodeoxyribonucleotides/therapeutic use , Oligodeoxyribonucleotides/pharmacology , Mice , Mice, Inbred C57BL , Female , Humans , Cell Line, Tumor , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Interleukin-2 Receptor alpha Subunit/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/therapy , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Vaccination , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
9.
ACS Nano ; 18(19): 12261-12275, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38683132

ABSTRACT

Cancer immunotherapy holds significant promise for addressing diverse malignancies. Nevertheless, its efficacy remains constrained by the intricate tumor immunosuppressive microenvironment. Herein, a light-triggered nanozyme Fe-TCPP-R848-PEG (Fe-MOF-RP) was designed for remodeling the immunosuppressive microenvironment. The Fe-TCPP-MOFs were utilized not only as a core catalysis component against tumor destruction but also as a biocompatible delivery vector of an immunologic agonist, improving its long circulation and tumor enrichment. Concurrently, it catalyzes the decomposition of H2O2 within the tumor, yielding oxygen to augment photodynamic therapy. The induced ferroptosis, in synergy with photodynamic therapy, prompts the liberation of tumor-associated antigens from tumor cells inducing immunogenic cell death. Phototriggered on-demand release of R848 agonists stimulated the maturation of dendritic cells and reverted the tumor-promoting M2 phenotypes into adoptive M1 macrophages, which further reshaped the tumor immunosuppressive microenvironment. Notably, the nanozyme effectively restrains well-established tumors, such as B16F10 melanoma. Moreover, it demonstrates a distal tumor-inhibiting effect upon in situ light treatment. What is more, in a lung metastasis model, it elicits robust immune memory, conferring enduring protection against tumor rechallenge. Our study presents a straightforward and broadly applicable strategy for crafting nanozymes with the potential to effectively thwart cancer recurrence and metastasis.


Subject(s)
Ferroptosis , Light , Tumor Microenvironment , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Ferroptosis/drug effects , Mice , Mice, Inbred C57BL , Photochemotherapy , Tumor Hypoxia/drug effects , Nanoparticles/chemistry , Immunotherapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Cell Line, Tumor
10.
J Immunother Cancer ; 12(4)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658032

ABSTRACT

BACKGROUND: While immunotherapy has been highly successful for the treatment of some cancers, for others, the immune response to tumor antigens is weak leading to treatment failure. The resistance of tumors to checkpoint inhibitor therapy may be caused by T cell exhaustion resulting from checkpoint activation. METHODS: In this study, lentiviral vectors that expressed T cell epitopes of an experimentally introduced tumor antigen, ovalbumin, or the endogenous tumor antigen, Trp1 were developed. The vectors coexpressed CD40 ligand (CD40L), which served to mature the dendritic cells (DCs), and a soluble programmed cell death protein 1 (PD-1) microbody to prevent checkpoint activation. Vaccination of mice bearing B16.OVA melanomas with vector-transduced DCs induced the proliferation and activation of functional, antigen-specific, cytolytic CD8 T cells. RESULTS: Vaccination induced the expansion of CD8 T cells that infiltrated the tumors to suppress tumor growth. Vector-encoded CD40L and PD-1 microbody increased the extent of tumor growth suppression. Adoptive transfer demonstrated that the effect was mediated by CD8 T cells. Direct injection of the vector, without the need for ex vivo transduction of DCs, was also effective. CONCLUSIONS: This study suggests that therapeutic vaccination that induces tumor antigen-specific CD8 T cells coupled with a vector-expressed checkpoint inhibitor can be an effective means to suppress the growth of tumors that are resistant to conventional immunotherapy.


Subject(s)
Cancer Vaccines , Immune Checkpoint Inhibitors , Lentivirus , Animals , Mice , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Lentivirus/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Humans , Dendritic Cells/immunology , Disease Models, Animal , CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Cell Line, Tumor , Mice, Inbred C57BL , Female
11.
Theranostics ; 14(6): 2573-2588, 2024.
Article in English | MEDLINE | ID: mdl-38646638

ABSTRACT

Background: Hypofractionated radiotherapy (hRT) can induce a T cell-mediated abscopal effect on non-irradiated tumor lesions, especially in combination with immune checkpoint blockade (ICB). However, clinically, this effect is still rare, and ICB-mediated adverse events are common. Lenalidomide (lena) is an anti-angiogenic and immunomodulatory drug used in the treatment of hematologic malignancies. We here investigated in solid tumor models whether lena can enhance the abscopal effect in double combination with hRT. Methods: In two syngeneic bilateral tumor models (B16-CD133 melanoma and MC38 colon carcinoma), the primary tumor was treated with hRT. Lena was given daily for 3 weeks. Besides tumor size and survival, the dependence of the antitumor effects on CD8+ cells, type-I IFN signaling, and T cell costimulation was determined with depleting or blocking antibodies. Tumor-specific CD8+ T cells were quantified, and their differentiation and effector status were characterized by multicolor flow cytometry using MHC-I tetramers and various antibodies. In addition, dendritic cell (DC)-mediated tumor antigen cross-presentation in vitro and directly ex vivo and the composition of tumor-associated vascular endothelial cells were investigated. Results: In both tumor models, the hRT/lena double combination induced a significant abscopal effect. Control of the non-irradiated secondary tumor and survival were considerably better than with the respective monotherapies. The abscopal effect was strongly dependent on CD8+ cells and associated with an increase in tumor-specific CD8+ T cells in the non-irradiated tumor and its draining lymph nodes. Additionally, we found more tumor-specific T cells with a stem-like (TCF1+ TIM3- PD1+) and a transitory (TCF1- TIM3+ CD101- PD1+) exhausted phenotype and more expressing effector molecules such as GzmB, IFNγ, and TNFα. Moreover, in the non-irradiated tumor, hRT/lena treatment also increased DCs cross-presenting a tumor model antigen. Blocking type-I IFN signaling, which is essential for cross-presentation, completely abrogated the abscopal effect. A gene expression analysis of bone marrow-derived DCs revealed that lena augmented the expression of IFN response genes and genes associated with differentiation, maturation (including CD70, CD83, and CD86), migration to lymph nodes, and T cell activation. Flow cytometry confirmed an increase in CD70+ CD83+ CD86+ DCs in both irradiated and abscopal tumors. Moreover, the hRT/lena-induced abscopal effect was diminished when these costimulatory molecules were blocked simultaneously using antibodies. In line with the enhanced infiltration by DCs and tumor-specific CD8+ T cells, including more stem-like cells, hRT/lena also increased tumor-associated high endothelial cells (TA-HECs) in the non-irradiated tumor. Conclusions: We demonstrate that lena can augment the hRT-induced abscopal effect in mouse solid tumor models in a CD8 T cell- and IFN-I-dependent manner, correlating with enhanced anti-tumor CD8 T cell immunity, DC cross-presentation, and TA-HEC numbers. Our findings may be helpful for the planning of clinical trials in (oligo)metastatic patients.


Subject(s)
CD8-Positive T-Lymphocytes , Disease Models, Animal , Lenalidomide , Radiation Dose Hypofractionation , Animals , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Mice, Inbred C57BL , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor , Combined Modality Therapy/methods , Female , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/therapy
12.
J Control Release ; 369: 251-265, 2024 May.
Article in English | MEDLINE | ID: mdl-38493950

ABSTRACT

Immunotherapy is currently a standard of care in the treatment of many malignancies. However, predictable side effects caused by systemic administration of highly immunostimulatory molecules have been a serious concern within this field. Intratumoural expression or silencing of immunogenic and immunoinhibitory molecules using nucleic acid-based approaches such as plasmid DNA (pDNA) and small interfering RNA (siRNA), respectively, could represent a next generation of cancer immunotherapy. Here, we employed lipid nanoparticles (LNPs) to deliver either non-specific pDNA and siRNA, or constructs targeting two prominent immunotherapeutic targets OX40L and indoleamine 2,3-dioxygenase-1 (IDO), to tumours in vivo. In the B16F10 mouse model, intratumoural delivery of LNP-formulated non-specific pDNA and siRNA led to strong local immune activation and tumour growth inhibition even at low doses due to the pDNA immunogenic nature. Replacement of these non-specific constructs by pOX40L and siIDO resulted in more prominent immune activation as evidenced by increased immune cell infiltration in tumours and tumour-draining lymph nodes. Consistently, pOX40L alone or in combination with siIDO could prolong overall survival, resulting in complete tumour regression and the formation of immunological memory in tumour rechallenge models. Our results suggest that intratumoural administration of LNP-formulated pDNA and siRNA offers a promising approach for cancer immunotherapy.


Subject(s)
DNA , Immunotherapy , Mice, Inbred C57BL , Nanoparticles , Plasmids , RNA, Small Interfering , Animals , Immunotherapy/methods , RNA, Small Interfering/administration & dosage , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Plasmids/administration & dosage , DNA/administration & dosage , DNA/immunology , Mice , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Female , Cell Line, Tumor , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Lipids/chemistry , Lipids/administration & dosage , Drug Carriers/chemistry
13.
Am J Pathol ; 194(6): 1007-1019, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38442804

ABSTRACT

Cytotoxic CD8+ T cells are central to the antitumor immune response by releasing cytotoxic granules that kill tumor cells. They are activated by antigen-presenting cells, which become activated by DAMPs (damage associated molecular patterns) through MyD88. However, the suppressive tumor microenvironment promotes T-cell tolerance to tumor antigens, in part by enhancing the activity of immune checkpoint molecules that prevent CD8+ T-cell activation and cytotoxicity. MyD88 limits CD4+ T-cell activation during cardiac adaptation to stress. A similar mechanism is hypothesized to exist in CD8+ T cells that could be modulated to improve antitumor immunity. Herein, adoptive transfer of MyD88-/- CD8+ T cells in melanoma-bearing T-cell-deficient mice resulted in slower tumor growth, greater intratumoral T-cell accumulation, and higher melanoma cell death compared with transfer of wild-type CD8+ T cells. These findings were also observed in T-cell-specific MyD88-/- mice compared with wild-type littermates implanted with melanoma. Mechanistically, deletion of MyD88 enhanced CD8+ T-cell activation and survival, and T-cell receptor induced degranulation of cytotoxic molecules, overall improving the killing of melanoma cells. This enhanced cytotoxicity was retained in mice bearing tumors expressing the specific antigen for which cytotoxic T-cells were restricted. This study's results demonstrate a conserved mechanism for MyD88 in modulating CD8+ T-cell activation and represent a novel target in improving cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Myeloid Differentiation Factor 88 , Animals , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Mice , CD8-Positive T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Melanoma/immunology , Melanoma/pathology , Melanoma/genetics , Melanoma/therapy , Mice, Knockout , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Melanoma, Experimental/genetics
14.
Methods Mol Biol ; 2773: 157-163, 2024.
Article in English | MEDLINE | ID: mdl-38236544

ABSTRACT

Efficacy of novel cancer immunization protocols could be tested in cell line-derived xenograft tumor models (CDX), which are based on the implantation of human tumor cell lines into mice for the development of different tumors by numerous means, such as subcutaneous implantation and orthotopic, venial, or peritoneal injections. However, the disadvantages of this model are the biological alteration of the derived cells or the inability of the cell lines to accurately reflect the complexity of tumor heterogeneity. Therefore, syngeneic mouse models, which offer a relatively simple grafting technique, preservation of lineage hierarchy, and the ability to generate tumors in as little as 2-8 weeks, are being used to study potential future applications in medical treatment, particularly immunotherapies. Here, we describe a B16.F10 C57Bl/6 mouse melanoma model we selected for therapeutic studies employing IL-2 and IL-12 immunization protocols. Procedure of tumor cells inoculation and melanoma development in mice is described in detail, as first and necessary set-up for successful immunization experiments.


Subject(s)
Cancer Vaccines , Melanoma, Experimental , Humans , Animals , Mice , Immunotherapy , Melanoma, Experimental/therapy , Cell Line, Tumor , Disease Models, Animal , Mice, Inbred C57BL
15.
Mil Med Res ; 11(1): 3, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38173045

ABSTRACT

BACKGROUND: Targeted T-cell therapy has emerged as a promising strategy for the treatment of hematological malignancies. However, its application to solid tumors presents significant challenges due to the limited accessibility and heterogeneity. Localized delivery of tumor-specific T-cells using biomaterials has shown promise, however, procedures required for genetic modification and generation of a sufficient number of tumor-specific T-cells ex vivo remain major obstacles due to cost and time constraints. METHODS: Polyethylene glycol (PEG)-based three-dimensional (3D) scaffolds were developed and conjugated with positively charged poly-L-lysine (PLL) using carbamide chemistry for efficient loading of lentiviruses (LVs) carrying tumor antigen-specific T-cell receptors (TCRs). The physical and biological properties of the scaffold were extensively characterized. Further, the scaffold loaded with OVA-TCR LVs was implanted in B16F10 cells expressing ovalbumin (B16-OVA) tumor model to evaluate the anti-tumor response and the presence of transduced T-cells. RESULTS: Our findings demonstrate that the scaffolds do not induce any systemic inflammation upon subcutaneous implantation and effectively recruit T-cells to the site. In B16-OVA melanoma tumor-bearing mice, the scaffolds efficiently transduce host T-cells with OVA-specific TCRs. These genetically modified T-cells exhibit homing capability towards the tumor and secondary lymphoid organs, resulting in a significant reduction of tumor size and systemic increase in anti-tumor cytokines. Immune cell profiling revealed a significantly high percentage of transduced T-cells and a notable reduction in suppressor immune cells within the tumors of mice implanted with these scaffolds. CONCLUSION: Our scaffold-based T-cell therapy presents an innovative in situ localized approach for programming T-cells to target solid tumors. This approach offers a viable alternative to in vitro manipulation of T-cells, circumventing the need for large-scale in vitro generation and culture of tumor-specific T-cells. It offers an off-the-shelf alternative that facilitates the use of host cells instead of allogeneic cells, thereby, overcoming a major hurdle.


Subject(s)
Melanoma, Experimental , T-Lymphocytes , Mice , Animals , T-Lymphocytes/pathology , Cell Line, Tumor , Immunotherapy , Genetic Engineering , Receptors, Antigen, T-Cell/genetics , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology
16.
J Mater Chem B ; 11(45): 10923-10928, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37934507

ABSTRACT

The strategy of using tumor cells to construct whole-cell cancer vaccines has received widespread attention. However, the limited immunogenicity of inactivated tumor cells and the challenge of overcoming immune suppression in solid tumors have hindered the application of whole-cell-based cancer immune therapy. Inspired by the regulatory effects of MnO2 and spatiotemporal control capability of material layers in cell surface engineering, we developed a manganese (Mn)-mineralized tumor cell, B16F10@MnO2, by inactivating B16F10 melanoma cells with KMnO4 to generate manganese-mineralized tumor cells. The cell-based composite was formed by combining amorphous MnO2 with the membrane structure of cells based on the redox reaction between KMnO4 and tumor cells. The MnO2 layer induced a stronger phagocytosis of ovalbumin (OVA)-expressing tumor cells by antigen presenting cells than formaldehyde-fixed cells did, resulting in specific antigen-presentation in vitro and in vivo and subsequent immune responses. Intratumoral therapy with B16F10@MnO2 inhibited B16F10 tumor growth. Moreover, the infiltration of CD8+ T cells within B16F10 solid tumors and the proportion of central memory T cells both increased in B16F10@MnO2 treated tumor-bearing mice, indicating enhanced adaptive immunity. This study provides a convenient and effective method to improve whole-cell-based anti-tumor therapy.


Subject(s)
Cancer Vaccines , Melanoma, Experimental , Mice , Animals , CD8-Positive T-Lymphocytes , Manganese , Manganese Compounds/pharmacology , Melanoma, Experimental/therapy , Oxides/pharmacology , Immunotherapy/methods
17.
J Control Release ; 363: 484-495, 2023 11.
Article in English | MEDLINE | ID: mdl-37778468

ABSTRACT

Blocking programmed cell death protein 1 (PD-1) is an effective therapeutic strategy for melanoma. However, patients often develop tumor recurrence postoperatively due to the low response rate to the anti-PD-1 antibody (aPD-1). In this study, we developed an in situ sprayable fibrin gel that contains cytosine-guanine oligodeoxynucleotides (CpG ODNs)-modified ovalbumin (OVA) antigen-expressing bone marrow dendritic cell (DC)-derived small extracellular vesicles (DC-sEVs) and aPD-1. CpG ODNs can activate DCs, which have potent immunostimulatory effects, by stimulating both the maturation and activation of tumor-infiltrating dendritic cells (TIDCs) and DCs in tumor-draining lymph nodes (TDLNs). In addition, DC-sEVs can deliver OVA to the same DCs, leading to the specific expression of tumor antigens by antigen-presenting cells (APCs). In brief, the unique synergistic combination of aPD-1 and colocalized delivery of immune adjuvants and tumor antigens enhances antitumor T-cell immunity, not only in the tumor microenvironment (TME) but also in TDLNs. This effectively attenuates local tumor recurrence and metastasis. Our results suggest that dual activation by CpG ODNs prolongs the survival of mice and decreases the recurrence rate in an incomplete tumor resection model, providing a promising approach to prevent B16-F10-OVA melanoma tumor recurrence and metastasis.


Subject(s)
Melanoma, Experimental , Neoplasm Recurrence, Local , Humans , Animals , Mice , Immunotherapy/methods , Melanoma, Experimental/therapy , Antigens, Neoplasm , Oligodeoxyribonucleotides/therapeutic use , Dendritic Cells , Mice, Inbred C57BL , Tumor Microenvironment
18.
Nutrients ; 15(16)2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37630828

ABSTRACT

(1) Background: Although the important role of dietary energy intake in regulating both cancer progression and host immunity has been widely recognized, it remains unclear whether dietary calorie restriction (CR) has any impact on anti-tumor immune responses. (2) Methods: Using an immunogenic B16 melanoma cell expressing ovalbumin (B16-OVA), we examined the effect of the CR diet on B16-OVA tumor growth and host immune responses. To further test whether the CR diet affects the efficacy of cancer immunotherapy, we examined the effect of CR against anti-PD-1 monoclonal antibody (anti-PD-1 Ab) treatment. (3) Results: The CR diet significantly slowed down the tumor growth of B16-OVA without affecting both CD4+ and CD8+ T cell infiltration into the tumor. Although in vivo depletion of CD8+ T cells facilitated B16-OVA tumor growth in the control diet group, there was no significant change in the tumor growth in the CR diet group with or without CD8+ T cell-depletion. Anti-PD-1 Ab treatment lost its efficacy to suppress tumor growth along with the activation and metabolic shift of CD8+ T cells under CR condition. (4) Conclusions: Our present results suggest that a physical condition restricted in energy intake in cancer patients may impair CD8+ T cell immune surveillance and the efficacy of immunotherapy.


Subject(s)
Caloric Restriction , Melanoma, Experimental , Humans , Animals , CD8-Positive T-Lymphocytes , Energy Intake , Melanoma, Experimental/therapy , Immunity
19.
Eur J Immunol ; 53(12): e2250182, 2023 12.
Article in English | MEDLINE | ID: mdl-37615189

ABSTRACT

Hypoxia-inducible factor 1 alpha (HIF1α), under hypoxic conditions, is known to play an oxygen sensor stabilizing role by exerting context- and cell-dependent stimulatory and inhibitory functions in immune cells. Nevertheless, how HIF1α regulates T cell differentiation and functions in tumor settings has not been elucidated. Herein, we demonstrated that T-cell-specific deletion of HIF1α improves the inflammatory potential and memory phenotype of CD8+ T cells. We validated that T cell-specific HIF1α ablation reduced the B16 melanomas development with the indication of ameliorated antitumor immune response with enhanced IFN-γ+ CD8+ T cells despite the increase in the Foxp3+ regulatory T-cell population. This was further verified by treating tumor-bearing mice with a HIF1α inhibitor. Results indicated that HIF1α inhibitor also recapitulates HIF1α ablation effects by declining tumor growth and enhancing the memory and inflammatory potential of CD8+ T cells. Furthermore, a combination of Treg inhibitor with HIF1α inhibitor can substantially reduce tumor size. Collectively, these findings highlight the notable roles of HIF1α in distinct CD8+ T-cell subsets. This study suggests the significant implications for enhancing the potential of T cell-based antitumor immunity by combining HIF1α and Tregs inhibitors.


Subject(s)
Melanoma, Experimental , T-Lymphocytes, Regulatory , Mice , Animals , CD8-Positive T-Lymphocytes , T-Lymphocyte Subsets , Melanoma, Experimental/therapy , Immunity
20.
Biomaterials ; 301: 122279, 2023 10.
Article in English | MEDLINE | ID: mdl-37591187

ABSTRACT

Efficient and safe delivery of vulnerable mRNA is a long-standing challenge for the broad application of the emerging mRNA-based therapeutics. Herein, a combinatorial library containing 119 novel lipids was constructed via sequential aza-Michael addition reactions of arylates and varying amines to tackle the ongoing challenge in mRNA delivery. Through in vitro screening of the lipid library on IGROV 1 cells, we identified several synthetic lipids with superior mRNA delivery efficacy. The delivery capability of these lipids was verified by the potent expression of luciferase in BALB/c mice upon intravenous administration of luciferase-encoding mRNA lipid nanoparticles (LNPs). Further investigations on the structure-activity relationship revealed that lipids with branched hydrophobic tails were better at delivering mRNA than those containing linear tails at the similar total number of carbons. In comparison to linear tails, the branched tails endowed LNPs with less inner hydrophobicity, fewer surface charges, and proper stability, which benefits the cellular uptake of LNPs and the intracellular trafficking of mRNA, thus improves the delivery efficacy of mRNA. The therapeutical potential of the lead LNPs was evaluated by delivering ovalbumin (OVA)-encoding mRNA to mice bearing B16-OVA melanoma tumors. The results demonstrated that the administration of OVA mRNA LNPs significantly activated CD8+ T cells in tumor microenvironment and substantially prohibited the growth of the aggressive B16-OVA tumors. The robust antitumor efficacy highlights the great potential of these LNPs in cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma, Experimental , Animals , Mice , Immunotherapy , Liposomes , Ovalbumin , Melanoma, Experimental/therapy , Lipids , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...