Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 11(14): e2308280, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38298111

ABSTRACT

Despite strides in immunotherapy, glioblastoma multiforme (GBM) remains challenging due to low inherent immunogenicity and suppressive tumor microenvironment. Converting "cold" GBMs to "hot" is crucial for immune activation and improved outcomes. This study comprehensively characterized a therapeutic vaccination strategy for preclinical GBM models. The vaccine consists of Mannan-BAM-anchored irradiated whole tumor cells, Toll-like receptor ligands [lipoteichoic acid (LTA), polyinosinic-polycytidylic acid (Poly (I:C)), and resiquimod (R-848)], and anti-CD40 agonistic antibody (rWTC-MBTA). Intracranial GBM models (GL261, SB28 cells) are used to evaluate the vaccine efficacy. A substantial number of vaccinated mice exhibited complete regression of GBM tumors in a T-cell-dependent manner, with no significant toxicity. Long-term tumor-specific immune memory is confirmed upon tumor rechallenge. In the vaccine-draining lymph nodes of the SB28 model, rWTC-MBTA vaccination triggered a major rise in conventional dendritic cell type 1 (cDC1) 12 h post-treatment, followed by an increase in conventional dendritic cell type 2 (cDC2), monocyte-derived dendritic cell (moDC), and plasmacytoid dendritic cell (pDC) on Day 5 and Day 13. Enhanced cytotoxicity of CD4+ and CD8+ T cells in vaccinated mice is verified in co-culture with tumor cells. Analyses of immunosuppressive signals (T-cell exhaustion, myeloid-derived suppressor cells (MDSC), M2 macrophages) in the GBM microenvironment suggest potential combinations with other immunotherapies for enhanced efficacy. In conclusion, the authors findings demonstrate that rWTC-MBTA induces potent and long-term adaptive immune responses against GBM.


Subject(s)
Glioblastoma , Vaccines , Mice , Animals , Glioblastoma/metabolism , CD8-Positive T-Lymphocytes , Vaccines/metabolism , Dendritic Cells , Immunity , Tumor Microenvironment
2.
J Exp Clin Cancer Res ; 42(1): 163, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37434263

ABSTRACT

BACKGROUND: Autologous tumor cell-based vaccines (ATVs) aim to prevent and treat tumor metastasis by activating patient-specific tumor antigens to induce immune memory. However, their clinical efficacy is limited. Mannan-BAM (MB), a pathogen-associated molecular pattern (PAMP), can coordinate an innate immune response that recognizes and eliminates mannan-BAM-labeled tumor cells. TLR agonists and anti-CD40 antibodies (TA) can enhance the immune response by activating antigen-presenting cells (APCs) to present tumor antigens to the adaptive immune system. In this study, we investigated the efficacy and mechanism of action of rWTC-MBTA, an autologous whole tumor cell vaccine consisting of irradiated tumor cells (rWTC) pulsed with mannan-BAM, TLR agonists, and anti-CD40 antibody (MBTA), in preventing tumor metastasis in multiple animal models. METHODS: The efficacy of the rWTC-MBTA vaccine was evaluated in mice using breast (4T1) and melanoma (B16-F10) tumor models via subcutaneous and intravenous injection of tumor cells to induce metastasis. The vaccine's effect was also assessed in a postoperative breast tumor model (4T1) and tested in autologous and allogeneic syngeneic breast tumor models (4T1 and EMT6). Mechanistic investigations included immunohistochemistry, immunophenotyping analysis, ELISA, tumor-specific cytotoxicity testing, and T-cell depletion experiments. Biochemistry testing and histopathology of major tissues in vaccinated mice were also evaluated for potential systemic toxicity of the vaccine. RESULTS: The rWTC-MBTA vaccine effectively prevented metastasis and inhibited tumor growth in breast tumor and melanoma metastatic animal models. It also prevented tumor metastasis and prolonged survival in the postoperative breast tumor animal model. Cross-vaccination experiments revealed that the rWTC-MBTA vaccine prevented autologous tumor growth, but not allogeneic tumor growth. Mechanistic data demonstrated that the vaccine increased the percentage of antigen-presenting cells, induced effector and central memory cells, and enhanced CD4+ and CD8+ T-cell responses. T-cells obtained from mice that were vaccinated displayed tumor-specific cytotoxicity, as shown by enhanced tumor cell killing in co-culture experiments, accompanied by increased levels of Granzyme B, TNF-α, IFN-γ, and CD107a in T-cells. T-cell depletion experiments showed that the vaccine's antitumor efficacy depended on T-cells, especially CD4+ T-cells. Biochemistry testing and histopathology of major tissues in vaccinated mice revealed negligible systemic toxicity of the vaccine. CONCLUSION: The rWTC-MBTA vaccine demonstrated efficacy in multiple animal models through T-cell mediated cytotoxicity and has potential as a therapeutic option for preventing and treating tumor metastasis with minimal systemic toxicity.


Subject(s)
Breast Neoplasms , Cancer Vaccines , Melanoma , Animals , Mice , Humans , Female , Mannans , Immunologic Memory , Cancer Vaccines/therapeutic use , CD40 Antigens , Antigens, Neoplasm , Breast Neoplasms/therapy
3.
Front Endocrinol (Lausanne) ; 14: 1030412, 2023.
Article in English | MEDLINE | ID: mdl-37342258

ABSTRACT

Cancer immunotherapy has shown remarkable clinical progress in recent years. Although age is one of the biggest leading risk factors for cancer development and older adults represent a majority of cancer patients, only a few new cancer immunotherapeutic interventions have been preclinically tested in aged animals. Thus, the lack of preclinical studies focused on age-dependent effect during cancer immunotherapy could lead to different therapeutic outcomes in young and aged animals and future modifications of human clinical trials. Here, we compare the efficacy of previously developed and tested intratumoral immunotherapy, based on the combination of polysaccharide mannan, toll-like receptor ligands, and anti-CD40 antibody (MBTA immunotherapy), in young (6 weeks) and aged (71 weeks) mice bearing experimental pheochromocytoma (PHEO). The presented results point out that despite faster growth of PHEO in aged mice MBTA intratumoral immunotherapy is effective approach without age dependence and could be one of the possible therapeutic interventions to enhance immune response to pheochromocytoma and perhaps other tumor types in aged and young hosts.


Subject(s)
Adrenal Gland Neoplasms , Pheochromocytoma , Humans , Animals , Mice , Aged , Pheochromocytoma/therapy , Immunotherapy/methods , Toll-Like Receptors , CD40 Antigens , Adrenal Gland Neoplasms/therapy
4.
Int Immunopharmacol ; 118: 110150, 2023 May.
Article in English | MEDLINE | ID: mdl-37030115

ABSTRACT

Despite constant advances in cancer research, the treatment of pancreatic adenocarcinoma remains extremely challenging. The intratumoral immunotherapy approach that was developed by our research group and was based on a combination of mannan-BAM, TLR ligands, and anti-CD40 antibody (MBTA) showed promising therapeutic effects in various murine tumor models, including a pancreatic adenocarcinoma model (Panc02). However, the efficacy of MBTA therapy in the Panc02 model was negatively correlated with tumor size at the time of therapy initiation. Here, we aimed to further improve the outcome of MBTA therapy in the Panc02 model using the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON). The combination of intratumoral MBTA therapy and intraperitoneal administration of DON resulted in the complete elimination of advanced Panc02 subcutaneous tumors (140.8 ± 46.8 mm3) in 50% of treated animals and was followed by development of long-term immune memory. In the bilateral Panc02 subcutaneous tumor model, we observed a significant reduction in tumor growth in both tumors as well as prolonged survival of treated animals. The appropriate timing and method of administration of DON were also addressed to maximize its therapeutic effects and minimize its side effects. In summary, our findings demonstrate that the intraperitoneal application of DON significantly improves the efficacy of intratumoral MBTA therapy in both advanced and bilateral Panc02 subcutaneous tumor murine models.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Animals , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Glutamine/therapeutic use , Adenocarcinoma/drug therapy , Immunotherapy/methods , Cell Line, Tumor , Pancreatic Neoplasms
5.
Cancers (Basel) ; 13(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34439097

ABSTRACT

Immunotherapy has become an essential component in cancer treatment. However, the majority of solid metastatic cancers, such as pheochromocytoma, are resistant to this approach. Therefore, understanding immune cell composition in primary and distant metastatic tumors is important for therapeutic intervention and diagnostics. Combined mannan-BAM, TLR ligand, and anti-CD40 antibody-based intratumoral immunotherapy (MBTA therapy) previously resulted in the complete eradication of murine subcutaneous pheochromocytoma and demonstrated a systemic antitumor immune response in a metastatic model. Here, we further evaluated this systemic effect using a bilateral pheochromocytoma model, performing MBTA therapy through injection into the primary tumor and using distant (non-injected) tumors to monitor size changes and detailed immune cell infiltration. MBTA therapy suppressed the growth of not only injected but also distal tumors and prolonged MBTA-treated mice survival. Our flow cytometry analysis showed that MBTA therapy led to increased recruitment of innate and adaptive immune cells in both tumors and the spleen. Moreover, adoptive CD4+ T cell transfer from successfully MBTA-treated mice (i.e., subcutaneous pheochromocytoma) demonstrates the importance of these cells in long-term immunological memory. In summary, this study unravels further details on the systemic effect of MBTA therapy and its use for tumor and metastasis reduction or even elimination.

6.
Cancer Immunol Immunother ; 70(11): 3303-3312, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33855601

ABSTRACT

Pancreatic adenocarcinoma is one of the leading causes of cancer-related deaths, and its therapy remains a challenge. Our proposed therapeutic approach is based on the intratumoral injections of mannan-BAM, toll-like receptor ligands, and anti-CD40 antibody (thus termed MBTA therapy), and has shown promising results in the elimination of subcutaneous murine melanoma, pheochromocytoma, colon carcinoma, and smaller pancreatic adenocarcinoma (Panc02). Here, we tested the short- and long-term effects of MBTA therapy in established subcutaneous Panc02 tumors two times larger than in previous study and bilateral Panc02 models as well as the roles of CD4+ and CD8+ T lymphocytes in this therapy. The MBTA therapy resulted in eradication of 67% of Panc02 tumors with the development of long-term memory as evidenced by the rejection of Panc02 cells after subcutaneous and intracranial transplantations. The initial Panc02 tumor elimination is not dependent on the presence of CD4+ T lymphocytes, although these cells seem to be important in long-term survival and resistance against tumor retransplantation. The resistance was revealed to be antigen-specific due to its inability to reject B16-F10 melanoma cells. In the bilateral Panc02 model, MBTA therapy manifested a lower therapeutic response. Despite numerous combinations of MBTA therapy with other therapeutic approaches, our results show that only simultaneous application of MBTA therapy into both tumors has potential for the treatment of the bilateral Panc02 model.


Subject(s)
Adenocarcinoma/pathology , CD40 Antigens/antagonists & inhibitors , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Mannans/pharmacology , Pancreatic Neoplasms/pathology , Poly I-C/pharmacology , Teichoic Acids/pharmacology , Adenocarcinoma/immunology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immunotherapy , Ligands , Mice , Pancreatic Neoplasms/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors , Pancreatic Neoplasms
7.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810617

ABSTRACT

The foundation of precision immunotherapy in oncology is rooted in computational biology and patient-derived sample sequencing to enrich for and target immunogenic epitopes. Discovery of these tumor-specific epitopes through tumor sequencing has revolutionized patient outcomes in many types of cancers that were previously untreatable. However, these therapeutic successes are far from universal, especially with cancers that carry high intratumoral heterogeneity such as glioblastoma (GBM). Herein, we present the technical aspects of Mannan-BAM, TLR Ligands, Anti-CD40 Antibody (MBTA) vaccine immunotherapy, an investigational therapeutic that potentially circumvents the need for in silico tumor-neoantigen enrichment. We then review the most promising GBM vaccination strategies to contextualize the MBTA vaccine. By reviewing current evidence using translational tumor models supporting MBTA vaccination, we evaluate the underlying principles that validate its clinical applicability. Finally, we showcase the translational potential of MBTA vaccination as a potential immunotherapy in GBM, along with established surgical and immunologic cancer treatment paradigms.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , CD40 Antigens/immunology , Glioblastoma/immunology , Glioblastoma/therapy , Immunotherapy/methods , Animals , Antigen-Presenting Cells/chemistry , Cancer Vaccines , Computational Biology , Epitopes/chemistry , Humans , Immunophenotyping , Ligands , Medical Oncology/trends , Mice , Neoplasm Metastasis , Peptides/chemistry , Treatment Outcome
8.
Article in English | MEDLINE | ID: mdl-33499322

ABSTRACT

In this paper, we map navigational needs and preferences of patients and visitors to evaluate the appropriateness of a smartphone navigation application in the hospital in contrast to other, more traditional navigational cues. We test the effects of sociodemographic variables (age, gender, education) on wayfinding strategies and preferences of respondents (using chi2 tests). Empirical research is based on the survey among 928 patients/visitors of the Vítkovice Hospital in Ostrava, Czechia. We found a relatively weak association between gender and wayfinding-no major differences between men and women in navigational preferences were found. Age was the most important predictor of wayfinding. Respondents in the over-60-year age group were characteristic of a lower interest in changes of the navigational system and low willingness to use mobile applications for navigation-people between 41 years and 60 years were the biggest supporters of changes. Correspondingly, demand for improvement of navigation (including a mobile application) was positively correlated with educational level.


Subject(s)
Spatial Navigation , Adult , Cues , Female , Hospitals , Humans , Male , Surveys and Questionnaires , Technology
9.
Adv Ther (Weinh) ; 3(9)2020 Sep.
Article in English | MEDLINE | ID: mdl-33709018

ABSTRACT

Emerging evidence is demonstrating the extent of T-cell infiltration within the tumor microenvironment has favorable prognostic and therapeutic implications. Hence, immunotherapeutic strategies that augment the T-cell signature of tumors hold promising therapeutic potential. Recently, immunotherapy based on intratumoral injection of mannan-BAM, toll-like receptor ligands and anti-CD40 antibody (MBTA) demonstrated promising potential to modulate the immune phenotype of injected tumors. The strategy promotes the phagocytosis of tumor cells to facilitate the recognition of tumor antigens and induce a tumor-specific adaptive immune response. Using a syngeneic colon carcinoma model, we demonstrate MBTA's potential to augment CD8+ T-cell tumor infiltrate when administered intratumorally or subcutaneously as part of a whole tumor cell vaccine. Both immunotherapeutic strategies proved effective at controlling tumor growth, prolonged survival and induced immunological memory against the parental cell line. Collectively, our investigation demonstrates MBTA's potential to trigger a potent anti-tumor immune response.

10.
Semin Oncol ; 46(4-5): 385-392, 2019.
Article in English | MEDLINE | ID: mdl-31739997

ABSTRACT

There is no doubt that immunotherapy lies in the spotlight of current cancer research and clinical trials. However, there are still limitations in the treatment response in certain types of tumors largely due to the presence of the complex network of immunomodulatory and immunosuppressive pathways. These limitations are not likely to be overcome by current immunotherapeutic options, which often target isolated steps in immune pathways preferentially involved in adaptive immunity. Recently, we have developed an innovative anti-cancer immunotherapeutic strategy that initially elicits a strong innate immune response with subsequent activation of adaptive immunity in mouse models. Robust primary innate immune response against tumor cells is induced by toll-like receptor ligands and anti-CD40 agonistic antibodies combined with the phagocytosis-stimulating ligand mannan, anchored to a tumor cell membrane by biocompatible anchor for membrane. This immunotherapeutic approach results in a dramatic therapeutic response in large established murine subcutaneous tumors including melanoma, sarcoma, pancreatic adenocarcinoma, and pheochromocytoma. Additionally, eradication of metastases and/or long-lasting resistance to subsequent re-challenge with tumor cells was also accomplished. Current and future advantages of this immunotherapeutic approach and its possible combinations with other available therapies are discussed in this review.


Subject(s)
Immunotherapy , Neoplasms/therapy , Adaptive Immunity , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Combined Modality Therapy , Humans , Immune System/immunology , Immune System/metabolism , Immunity, Innate , Immunomodulation , Immunotherapy/methods , Ligands , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Phagocytosis/drug effects , Phagocytosis/immunology , Toll-Like Receptors/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
Cancers (Basel) ; 11(5)2019 May 11.
Article in English | MEDLINE | ID: mdl-31083581

ABSTRACT

Therapeutic options for metastatic pheochromocytoma/paraganglioma (PHEO/PGL) are limited. Here, we tested an immunotherapeutic approach based on intratumoral injections of mannan-BAM with toll-like receptor ligands into subcutaneous PHEO in a mouse model. This therapy elicited a strong innate immunity-mediated antitumor response and resulted in a significantly lower PHEO volume compared to the phosphate buffered saline (PBS)-treated group and in a significant improvement in mice survival. The cytotoxic effect of neutrophils, as innate immune cells predominantly infiltrating treated tumors, was verified in vitro. Moreover, the combination of mannan-BAM and toll-like receptor ligands with agonistic anti-CD40 was associated with increased mice survival. Subsequent tumor re-challenge also supported adaptive immunity activation, reflected primarily by long-term tumor-specific memory. These results were further verified in metastatic PHEO, where the intratumoral injections of mannan-BAM, toll-like receptor ligands, and anti-CD40 into subcutaneous tumors resulted in significantly less intense bioluminescence signals of liver metastatic lesions induced by tail vein injection compared to the PBS-treated group. Subsequent experiments focusing on the depletion of T cell subpopulations confirmed the crucial role of CD8+ T cells in inhibition of bioluminescence signal intensity of liver metastatic lesions. These data call for a new therapeutic approach in patients with metastatic PHEO/PGL using immunotherapy that initially activates innate immunity followed by an adaptive immune response.

12.
Int Immunopharmacol ; 59: 86-96, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29635103

ABSTRACT

Immunotherapy emerges as a fundamental approach in cancer treatment. Up to date, the efficacy of numerous different immunotherapies has been evaluated. The use of microorganisms or their parts for immune cell activation, referred to as Pathogen-Associated Molecular Patterns (PAMPs), represents highly promising concept. The therapeutic effect of PAMPs can be further amplified by suitable combination of different types of PAMPs such as Toll like receptor (TLR) agonists and phagocytosis activating ligands. Previously, we used the combination of phagocytosis activating ligand (mannan) and mixture of TLR agonists (resiquimod (R-848), poly(I:C), inactivated Listeria monocytogenes) for successful treatment of melanoma in murine B16-F10 model. In the present study, we optimized the composition and timing of previously used mixture. Therapeutic mixture based on well-defined chemical compounds consisted of mannan anchoring to tumor cell surface by biocompatible anchor for membranes (BAM) and TLR agonists resiquimod, poly(I:C), and lipoteichoic acid (LTA). The optimization resulted in (1) eradication of advanced stage progressive melanoma in 83% of mice, (2) acquisition of resistance to tumor re-transplantation, and (3) potential anti-metastatic effect. After further investigation of mechanisms, underlying anti-tumor responses, we concluded that both innate and adaptive immunity are activated and involved in these processes. We tested the efficacy of our treatment in Panc02 murine model of aggressive pancreatic tumor as well. Simultaneous application of agonistic anti-CD40 antibody was necessary to achieve effective therapeutic response (80% recovery) in this model. Our results suggest that herein presented immunotherapeutic approach is a promising cancer treatment strategy with the ability to eradicate not only primary tumors but also metastases.


Subject(s)
Adenocarcinoma/therapy , Melanoma, Experimental/therapy , Pancreatic Neoplasms/therapy , Phagocytosis , Toll-Like Receptors/agonists , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Female , Imidazoles/therapeutic use , Immunotherapy , Lipopolysaccharides/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Mannans/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neutrophils/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Poly I-C/therapeutic use , Teichoic Acids/therapeutic use , Tumor Burden/drug effects
13.
BMC Cancer ; 16(1): 940, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927165

ABSTRACT

BACKGROUND: Using killed microorganisms or their parts to stimulate immunity for cancer treatment dates back to the end of 19th century. Since then, it undergone considerable development. Our novel approach binds ligands to the tumor cell surface, which stimulates tumor phagocytosis. The therapeutic effect is further amplified by simultaneous application of agonists of Toll-like receptors. We searched for ligands that induce both a strong therapeutic effect and are safe for humans. METHODS: B16-F10 murine melanoma model was used. For the stimulation of phagocytosis, mannan or N-formyl-methionyl-leucyl-phenylalanine, was covalently bound to tumor cells or attached using hydrophobic anchor. The following agonists of Toll-like receptors were studied: monophosphoryl lipid A (MPLA), imiquimod (R-837), resiquimod (R-848), poly(I:C), and heat killed Listeria monocytogenes. RESULTS: R-848 proved to be the most suitable Toll-like receptor agonist for our novel immunotherapeutic approach. In combination with covalently bound mannan, R-848 significantly reduced tumor growth. Adding poly(I:C) and L. monocytogenes resulted in complete recovery in 83% of mice and in their protection from the re-transplantation of melanoma cells. CONCLUSION: An efficient cancer treatment results from the combination of Toll-like receptor agonists and phagocytosis stimulating ligands bound to the tumor cells.


Subject(s)
Immunity, Innate , Immunotherapy , Neoplasms/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Imidazoles/pharmacology , Immunotherapy/methods , Ligands , Mannans/immunology , Melanoma, Experimental , Mice , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis , Poly I-C/immunology , Respiratory Burst/immunology , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
14.
Int Immunopharmacol ; 39: 295-306, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27505858

ABSTRACT

The idea of using killed microorganisms or their parts for a stimulation of immunity in the cancer immunotherapy is very old, but the question of interactions and binding of these preparations to tumor cells has not been addressed so far. The attachment of Zymosan A and both Gram-positive and Gram-negative bacteria to tumor cells was tested in in vivo experiments. This binding was accomplished by charge interactions, anchoring based on hydrophobic chains and covalent bonds and proved to be crucial for a strong immunotherapeutic effect. The establishment of conditions for simultaneous stimulation of both Toll-like and phagocytic receptors led to very strong synergy. It resulted in tumor shrinkage and its temporary or permanent elimination. The role of neutrophils in cancer immunotherapy was demonstrated and the mechanism of their action (frustrated phagocytosis) was proposed. Finally, therapeutic approaches applicable for safe human cancer immunotherapy are discussed. Heat killed Mycobacterium tuberculosis covalently attached to tumor cells seems to be promising tool for this therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Listeria monocytogenes/immunology , Melanoma/therapy , Mycobacterium tuberculosis/immunology , Skin Neoplasms/therapy , Zymosan/therapeutic use , Animals , Hot Temperature , Humans , Immunity , Immunomodulation , Killer Cells, Natural/immunology , Listeria monocytogenes/chemistry , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/chemistry , Neoplasm Transplantation , Neutrophils/immunology , Phagocytosis , Skin Neoplasms/immunology , Tumor Burden , Zymosan/chemistry
15.
PLoS One ; 9(1): e85222, 2014.
Article in English | MEDLINE | ID: mdl-24454822

ABSTRACT

The application of the phagocytic receptor agonists in cancer immunotherapy was studied. Agonists (laminarin, molecules with terminal mannose, N-Formyl-methioninyl-leucyl-phenylalanine) were firmly anchored to the tumor cell surface. When particular agonists of phagocytic receptors were used together with LPS (Toll-like receptor agonist), high synergy causing tumour shrinkage and a temporary or permanent disappearance was observed. Methods of anchoring phagocytic receptor agonists (charge interactions, anchoring based on hydrophobic chains, covalent bonds) and various regimes of phagocytic agonist/LPS mixture applications were tested to achieve maximum therapeutic effect. Combinations of mannan/LPS and f-MLF/LPS (hydrophobic anchors) in appropriate (pulse) regimes resulted in an 80% and 60% recovery for mice, respectively. We propose that substantial synergy between agonists of phagocytic and Toll-like receptors (TLR) is based on two events. The TLR ligand induces early and massive inflammatory infiltration of tumors. The effect of this cell infiltrate is directed towards tumor cells, bearing agonists of phagocytic receptors on their surface. The result of these processes was effective killing of tumor cells. This novel approach represents exploitation of innate immunity mechanisms for treating cancer.


Subject(s)
Immunotherapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Receptors, Immunologic/agonists , Animals , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Glucans , Ligands , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Mannose/chemistry , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Polysaccharides/pharmacology , Polysaccharides/therapeutic use , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Survival Analysis , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...