Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cancer Immunol Immunother ; 71(11): 2691-2700, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35364740

RESUMO

Low efficacy of cancer immunotherapy encourages the search for possible resistance mechanisms and biomarkers that would predict the outcome of immunotherapy in oncology patients. Most cancer immunotherapies act on T lymphocytes, which can specifically recognize and kill tumor cells. However, for immunotherapy-activated T lymphocytes to be able to perform these functions, proper tumor Ag processing and surface presentation by MHC-I molecule is important. Knowing the significance of Ag processing and presentation mechanism (APM) in anti-tumor immune response, we sought to evaluate how the functionality of APM affects tumor immune microenvironment and response to dendritic cell vaccines (DCV) and anti-PD-1. By comparing murine Lewis lung carcinoma LLC1 and glioma GL261 models a decreased expression of APM-related genes, such as Psmb8, Psmb9, Psmb10, Tap1, Tap2, Erap1, B2m, and low expression of surface MHC-I molecule were found in LLC1 cells. Changes in APM-related gene expression affected the ability of T lymphocytes to recognize and kill LLC1 cells, resulting in the absence of cytotoxic immune response and resistance to DCV and anti-PD-1. An emerging cytotoxic immune reaction and sensitivity to DCV and anti-PD-1 were observed in GL261 tumors where APM remained functional. This study demonstrates that one of the possible mechanisms of tumor resistance to immunotherapy is a dysfunctional APM and reveals a predictive potential of APM-related gene set expression for the personalization of dendritic cell vaccine and anti-PD-1 therapies in murine pre-treated tumors.


Assuntos
Glioma , Vacinas , Aminopeptidases/metabolismo , Animais , Apresentação de Antígeno , Linhagem Celular Tumoral , Células Dendríticas , Glioma/metabolismo , Antígenos de Histocompatibilidade Classe I , Humanos , Imunoterapia , Camundongos , Antígenos de Histocompatibilidade Menor/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Microambiente Tumoral , Vacinas/metabolismo
3.
Medicina (Kaunas) ; 51(5): 302-6, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26674149

RESUMO

BACKGROUND AND OBJECTIVE: The aim of this study was to evaluate the efficiency of proton beam irradiation in pancreatic cancer cell line MIA PaCa-2 and its role in the cell cycle, apoptosis, and formation of histone γH2AX in different reparation times (72-h follow-up). MATERIAL AND METHODS: The MIA PaCa-2 pancreatic carcinoma cell line was irradiated with 1.6-Gy proton beam. After irradiation, cell viability was measured colorimetrically, and the cell cycle, apoptosis, and γH2AX expression were evaluated on a FACScan cytometer. RESULTS: Low-dose proton beam irradiation had an effect on the MIA PaCa-2 tumor cell line already 1h after exposure, but maximal lethality was reached after 72h postirradiation with a cell viability rate of 24%. The cell cycle went into partial G1/0 arrest, and was released after 72h. The expression of γH2AX was strong and its levels were significantly elevated as late as 48h post radiation. The apoptosis levels increased with post radiation incubation time to reach 79% after 72h. CONCLUSIONS: Our data demonstrate that low-doses proton beam irradiation had an effect on MIA PaCa-2 pancreatic carcinoma cell line. Full extent of irradiation had an impact only 24h postirradiation, triggering DNA arrested cell cycle in G1/0 phase. Formed DNA DSBs were found to be repaired via the NHEJ pathway mechanism within 72h. Unsuccessful repaired DSBs induced apoptotic cell death. After 72h reparation processes were completed, and cell cycle was released from arrest in G1/0 phase.


Assuntos
Histonas/metabolismo , Neoplasias Pancreáticas/radioterapia , Terapia com Prótons/métodos , Apoptose/efeitos da radiação , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Neoplasias Pancreáticas/metabolismo
4.
Medicina (Kaunas) ; 50(4): 237-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25458961

RESUMO

BACKGROUND AND OBJECTIVE: Superparamagnetic iron oxide nanoparticles (SPIONs) emerge as a promising tool for early cancer diagnostics and targeted therapy. However, both toxicity and biological activity of SPIONs should be evaluated in detail. The aim of this study was to synthesize superparamagnetic cobalt ferrite nanoparticles (Co-SPIONs), and to investigate their uptake, toxicity and effects on cancer stem-like properties in human pancreatic cancer cell line MiaPaCa2 and human ovarian cancer cell line A2780. MATERIALS AND METHODS: Co-SPIONs were produced by Massart's co-precipitation method. The cells were treated with Co-SPIONs at three different concentrations (0.095, 0.48, and 0.95µg/mL) for 24 and 48h. Cell viability and proliferation were analyzed after treatment. The stem-like properties of cells were assessed by investigating the cell clonogenicity and expression of cancer stem cell-associated markers, including CD24/ESA in A2780 cell line and CD44/ALDH1 in MiaPaCa2 cell line. Magnetically activated cell sorting was used for the separation of magnetically labeled and unlabeled cells. RESULTS: Both cancer cell lines accumulated Co-SPIONs, however differences in response to nanoparticles were observed between MiaPaCa2 and A2780 cell. In particular, A2780 cells were more sensitive to exposition to Co-SPIONs than MiaPaCa2 cells, indicating that a safe concentration of nanoparticles must be estimated individually for a particular cell type. Higher doses of Co-SPIONs decreased both the clonogenicity and ESA marker expression in A2780 cells. CONCLUSIONS: Co-SPIONs are not cytotoxic to cancer cells, at least when used at a concentration of up to 0.95µg/mL. Co-SPIONs have a dose-dependent effect on the clonogenic potential and ESA marker expression in A2780 cells. Magnetic detection of low concentrations of Co-SPIONS in cancer cells is a promising tool for further applications of these nanoparticles in cancer diagnosis and treatment; however, extensive research in this field is needed.


Assuntos
Cobalto/metabolismo , Compostos Férricos/metabolismo , Nanopartículas/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Pancreáticas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Cobalto/farmacologia , Feminino , Compostos Férricos/farmacologia , Citometria de Fluxo , Humanos , Tamanho da Partícula
5.
Front Immunol ; 15: 1401097, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39055716

RESUMO

Purpose: The aim of this study was to assess the role of sPD-L1 and sPD-1 as potential biomarkers in prostate cancer (PCa). The association of the values of these soluble proteins were correlated to the clinical data: stage of disease, Gleason score, biochemical recurrence etc. For a comprehensive study, the relationship between sPD-L1 and sPD-1 and circulating immune cells was further investigated. Methods: A total of 88 patients with pT2 and pT3 PCa diagnosis and 41 heathy men were enrolled. Soluble sPD-L1 and sPD-1 levels were measured in plasma by ELISA method. Immunophenotyping was performed by flow cytometry analysis. Results: Our study's findings demonstrate that PCa patients had higher levels of circulating sPD-L1 and sPD-1 comparing to healthy controls (p < 0.001). We found a statistically significant (p < 0.05) relationship between improved progression free survival and lower initial sPD-L1 values. Furthermore, patients with a lower sPD-1/sPD-L1 ratio were associated with a higher probability of disease progression (p < 0.05). Additionally, a significant (p < 0.05) association was discovered between higher Gleason scores and elevated preoperative sPD-L1 levels and between sPD-1 and advanced stage of disease (p < 0.05). A strong correlation (p < 0.05), between immunosuppressive CD4+CD25+FoxP3+ regulatory T cells and baseline sPD-L1 was observed in patients with unfavorable postoperative course of the disease, supporting the idea that these elements influence each other in cancer progression. In addition to the postoperative drop in circulating PD-L1, the inverse relationship (p < 0.05), between the percentage of M-MDSC and sPD-L1 in patients with BCR suggests that M-MDSC is not a source of sPD-L1 in PCa patients. Conclusion: Our findings suggest the potential of sPD-L1 as a promising prognostic marker in prostate cancer.


Assuntos
Antígeno B7-H1 , Biomarcadores Tumorais , Neoplasias da Próstata , Humanos , Masculino , Neoplasias da Próstata/sangue , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Antígeno B7-H1/sangue , Biomarcadores Tumorais/sangue , Pessoa de Meia-Idade , Idoso , Prognóstico , Gradação de Tumores , Estadiamento de Neoplasias
6.
Vaccine ; 42(3): 512-521, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38184395

RESUMO

Dendritic cell (DC) based immunotherapy is one of the strategies to combat cancer invoking a patient's immune system. This form of anticancer immunotherapy employs adjuvants to enhance the immune response, triggering mechanisms of innate immunity and thus increase immunotherapeutic efficiency. A conventional adjuvant for DCs maturation during production of anticancer vaccines is bacterial LPS. Nevertheless, synthetic dsRNAs were also shown to stimulate different receptors on innate immune cells and to activate immune responses through induction of cytokines via toll-like receptors. In our study we investigated the potential of Larifan as dsRNA of natural origin to stimulate maturation of DCs with proinflammatory (possible antitumoral) activity and to compare these immunostimulatory properties between Larifan's fractions with different molecular lengths. To explore the suitability of this product for therapy, it is necessary to study the properties of its different fractions and compare them to standard adjuvants. We investigated the effect of Larifan's fractions on immune system stimulation in vivo by monitoring the survival time of tumor-bearing mice. Murine DCs produced in vitro using Larifan and its fractions together with tumor antigens during production were also characterized. All Larifan fractions resulted in inducing high expression of immunogenic markers CD40, CD80, CD86, CCR7, MHC II and lower secretion of the immunosuppressive cytokine IL-10, compared to the maturation with LPS in mDCs. The lowest expression of tolerogenic gene Ido1 and highest expression of the immunogenic genes Clec7a, Tnf, Icosl, Il12rb2, Cd209a were characteristic to the unfractionated dsRNA and short fraction FR15. In the mouse model the best overall survival rate was observed in mice treated with medium-length FR9 and FR15. We can state that both Larifan and its fractions were superior to LPS as vaccine adjuvants in stimulating phenotype and functional activity of mature DCs. DCs maturation using these factors induces a valuable anticancer immune response.


Assuntos
Bacteriófagos , Neoplasias , Humanos , Camundongos , Animais , Adjuvantes de Vacinas , Lipopolissacarídeos , Células Dendríticas , Citocinas/metabolismo , Adjuvantes Imunológicos/metabolismo , Imunidade , Receptores de Interleucina-12 , Compostos Orgânicos
7.
Biomolecules ; 14(2)2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38397409

RESUMO

The spatial distribution of tumor infiltrating lymphocytes (TILs) defines several histologically and clinically distinct immune subtypes-desert (no TILs), excluded (TILs in stroma), and inflamed (TILs in tumor parenchyma). To date, robust classification of immune subtypes still requires deeper experimental evidence across various cancer types. Here, we aimed to investigate, define, and validate the immune subtypes in melanoma by coupling transcriptional and histological assessments of the lymphocyte distribution in tumor parenchyma and stroma. We used the transcriptomic data from The Cancer Genome Atlas melanoma dataset to screen for the desert, excluded, and inflamed immune subtypes. We defined subtype-specific genes and used them to construct a subtype assignment algorithm. We validated the two-step algorithm in the qPCR data of real-world melanoma tumors with histologically defined immune subtypes. The accuracy of a classifier encompassing expression data of seven genes (immune response-related: CD2, CD53, IRF1, and CD8B; and stroma-related: COL5A2, TNFAIP6, and INHBA) in a validation cohort reached 79%. Our findings suggest that melanoma tumors can be classified into transcriptionally and histologically distinct desert, excluded, and inflamed subtypes. Gene expression-based algorithms can assist physicians and pathologists as biomarkers in the rapid assessment of a tumor immune microenvironment while serving as a tool for clinical decision making.


Assuntos
Melanoma , Humanos , Melanoma/patologia , Biomarcadores/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Imunidade , Transcriptoma , Microambiente Tumoral/genética , Biomarcadores Tumorais/metabolismo
8.
J Clin Med ; 10(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34442004

RESUMO

BACKGROUND: Prostate cancer (PCa) is known to exhibit a wide spectrum of aggressiveness and relatively high immunogenicity. The aim of this study was to examine the effect of tumor excision on immunophenotype rearrangements in peripheral blood and to elucidate if it is associated with biochemical recurrence (BCR) in high risk (HR) and low risk (LR) patients. METHODS: Radical prostatectomy (RP) was performed on 108 PCa stage pT2-pT3 patients. Preoperative vs. postoperative (one and three months) immunophenotype profile (T- and B-cell subsets, MDSC, NK, and T reg populations) was compared in peripheral blood of LR and HR groups. RESULTS: The BCR-free survival difference was significant between the HR and LR groups. Postoperative PSA decay rate, defined as ePSA, was significantly slower in the HR group and predicted BCR at cut-off level ePSA = -2.0% d-1 (AUC = 0.85 (95% CI, 0.78-0.90). Three months following tumor excision, the LR group exhibited a recovery of natural killer CD3 - CD16+ CD56+ cells, from 232 cells/µL to 317 cells/µL (p < 0.05), which was not detectable in the HR group. Prostatectomy also resulted in an increased CD8+ population in the LR group, mostly due to CD8+ CD69+ compartment (from 186 cells/µL before surgery to 196 cells/µL three months after, p < 001). The CD8+ CD69+ subset increase without total T cell increase was present in the HR group (p < 0.001). Tumor excision resulted in a myeloid-derived suppressor cell (MDSC) number increase from 12.4 cells/µL to 16.2 cells/µL in the HR group, and no change was detectable in LR patients (p = 0.12). An immune signature of postoperative recovery was more likely to occur in patients undergoing laparoscopic radical prostatectomy (LRP). Open RP (ORP) was associated with increased MDSC numbers (p = 0.002), whereas LRP was characterized by an immunity sparing profile, with no change in MDSC subset (p = 0.16). CONCLUSION: Tumor excision in prostate cancer patients results in two distinct patterns of immunophenotype rearrangement. The low-risk group is highly responsive, revealing postoperative restoration of T cells, NK cells, and CD8+ CD69+ numbers and the absence of suppressor MDSC increase. The high-risk group presented a limited response, accompanied by a suppressor MDSC increase and CD8+ CD69+ increase. The laparoscopic approach, unlike ORP, did not result in an MDSC increase in the postoperative period.

9.
Am J Reprod Immunol ; 84(1): e13244, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32294293

RESUMO

PROBLEM: The current tumor immunology paradigm emphasizes the role of the immune tumor microenvironment and distinguishes several histologically and transcriptionally different immune tumor subtypes. However, the experimental validation of such classification is so far limited to selected cancer types. Here, we aimed to explore the existence of inflamed, excluded, and desert immune subtypes in ovarian cancer, as well as investigate their association with the disease outcome. METHOD OF STUDY: We used the publicly available ovarian cancer dataset from The Cancer Genome Atlas for developing subtype assignment algorithm, which was next verified in a cohort of 32 real-world patients of a known tumor subtype. RESULTS: Using clinical and gene expression data of 489 ovarian cancer patients in the publicly available dataset, we identified three transcriptionally distinct clusters, representing inflamed, excluded, and desert subtypes. We developed a two-step subtyping algorithm with COL5A2 serving as a marker for separating excluded tumors, and CD2, TAP1, and ICOS for distinguishing between inflamed and desert tumors. The accuracy of gene expression-based subtyping algorithm in a real-world cohort was 75%. Additionally, we confirmed that patients bearing inflamed tumors are more likely to survive longer. CONCLUSION: Our results highlight the presence of transcriptionally and histologically distinct immune subtypes among ovarian tumors and emphasize the potential benefit of immune subtyping as a clinical tool for treatment tailoring.


Assuntos
Inflamação/genética , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Ovarianas/genética , Algoritmos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Estudos de Coortes , Colágeno Tipo V/genética , Colágeno Tipo V/metabolismo , Biologia Computacional , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imunofenotipagem , Inflamação/diagnóstico , Inflamação/patologia , Pessoa de Meia-Idade , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/patologia , Prognóstico , Transcriptoma , Microambiente Tumoral
10.
Oncol Rep ; 41(2): 1238-1252, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30483809

RESUMO

The management of advanced ovarian cancer is challenging due to the high frequency of recurrence, often associated with the development of resistance to platinum­based chemotherapy. Molecular analyses revealed the complexity of ovarian cancer with particular emphasis on the immune system, which may contribute to disease progression and response to treatment. Cytokines and chemokines mediate the cross­talk between cancer and immune cells, and therefore, present as potential biomarkers, reflecting the tumor microenvironment. A panel of circulating C­C motif chemokine ligand (CCL) and C­X­C motif chemokine ligand (CXCL) chemokines were examined in the serum of 40 high­grade patients with ovarian cancer prior to primary surgery. The level of immune infiltration in tumors was also analyzed. The preoperative levels of chemokines differ between patients. Elevated levels of circulating CXCL4 + CCL20 + CXCL1 combination can discriminate patients with shorter recurrence­free survival and overall survival. The presence of tumor­infiltrating T lymphocytes was detected in half of the patients. The mRNA expression analysis suggests the presence of antitumoral and immunosuppressive elements in the tumor microenvironment. The combination of circulating CXCL9 + CXCL10 can distinguish immune­infiltrated tumors that will lead to shorter recurrence­free survival. The results suggest that preoperative profiling of circulating chemokines in patients with ovarian cancer may provide valuable information regarding tumor recurrence and immune infiltration. The findings demonstrate that combinations have better prognostic utility than single chemokines, and may serve as patient stratification tools.


Assuntos
Biomarcadores Tumorais/sangue , Quimiocina CCL20/sangue , Quimiocina CXCL1/sangue , Quimiocina CXCL9/sangue , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/imunologia , Microambiente Tumoral/imunologia , Adulto , Idoso , Progressão da Doença , Feminino , Humanos , Linfócitos do Interstício Tumoral/imunologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/imunologia , Neoplasias Ovarianas/sangue , Linfócitos T/imunologia
11.
Am J Reprod Immunol ; 80(3): e12996, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29904979

RESUMO

PROBLEM: Development of platinum resistance in ovarian cancer is mediated by both cancer cells and tumor microenvironment. Activation of epithelial-mesenchymal transition program in cancer cells may lead to enrichment for resistant clones. These processes can be affected by tumor-associated macrophages, a highly plastic population of cells that participate in tumor progression and response to treatment by shaping the microenvironment. We aimed to study how platinum resistance influences the crosstalk between macrophages and ovarian cancer cells. METHOD OF STUDY: Using cisplatin-sensitive ovarian cancer cell line A2780, we developed and characterized cisplatin-resistant A2780Cis and cisplatin and doxorubicin co-resistant A2780Dox cell lines. Next, we set up an indirect coculture system with THP-1 cell line-derived M0-type-, M1-type- and M2-type-like polarized macrophages. We monitored the expression of genes associated with cellular stemness, multidrug resistance, and epithelial-mesenchymal transition in cancer cells, and expression profile of M1/M2 markers in macrophages. RESULTS: Development of drug resistance in ovarian cancer cell lines was accompanied by increased migration, clonogenicity, and upregulated expression of transcription factors, associated with cellular stemness and epithelial-mesenchymal transition. Upon coculture, we noted that the most relevant changes in gene expression profile occurred in A2780 cells. Moreover, M0- and M1-type macrophages, but not M2-type macrophages, showed significant transcriptional alterations. CONCLUSION: Our results provide the evidence for bidirectional interplay between cancer cells and macrophages. Independent of platinum resistance status, ovarian cancer cells polarize macrophages toward M2-like type, whereas macrophages induce epithelial-mesenchymal transition and stemness-related gene expression profile in cisplatin-sensitive, but not cisplatin-resistant cancer cells.


Assuntos
Adenocarcinoma/imunologia , Cisplatino/uso terapêutico , Macrófagos/imunologia , Neoplasias Ovarianas/imunologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Cocultura , Citocinas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunomodulação , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Células Th2/imunologia , Evasão Tumoral , Microambiente Tumoral
12.
Oncol Lett ; 15(4): 5098-5104, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29552144

RESUMO

Immunotherapy in the form of anticancer vaccination relies on the mobilization of the patient's immune system against specific cancer antigens. Instead of focusing on an autologous cell lysate, which is not always available in clinical practice, the present study investigates vaccines utilizing xenogeneic foetal tissue that are rich in oncofoetal antigens. Lewis lung carcinoma (LLC)-challenged C57BL/6 mice were treated with either a xenogeneic vaccine made from chicken whole embryo, or a xenogeneic vaccine made from rat embryonic brain tissue, supplemented with a Bacillus subtilis protein fraction as an adjuvant. Median and overall survival, size of metastatic foci in lung tissue and levels of circulating CD8a+ T cells were evaluated and compared with untreated control mice. Following primary tumour removal, a course of three subcutaneous vaccinations with xenogeneic chicken embryo vaccine led to significant increase in overall survival rate (100% after 70 days of follow-up vs. 40% in untreated control mice), significant increase in circulating CD8a+ T cells (18.18 vs. 12.6% in untreated control mice), and a significant decrease in the area and incidence of metastasis foci. The xenogeneic rat brain tissue-based vaccine did not improve any of the investigated parameters, despite promising reports in other models. We hypothesize that the proper selection of antigen source (tissue) can constitute an effective immunotherapeutic product.

13.
Vaccine ; 32(32): 4015-24, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24837511

RESUMO

Accumulation of firm evidence that clinically apparent cancer develops only when malignant cells manage to escape immunosurveillance led to the introduction of tumor immunotherapy strategies aiming to reprogramm the cancer-dysbalanced antitumor immunity and restore its capacity to control tumor growth. There are several immunotherapeutical strategies, among which specific active immunotherapy or therapeutic cancer vaccination is one of the most promising. It targets dendritic cells (DCs) which have a unique ability of inducing naive and central memory T cell-mediated immune response in the most efficient manner. DCs can be therapeutically targeted either in vivo/in situ or by ex vivo manipulations followed by their re-injection back into the same patient. The majority of current DC targeting strategies are based on autologous or allogeneic tumor-associated antigens (TAAs) which possess various degrees of inherent tolerogenic potential. Therefore still limited efficacy of various tumor immunotherapy approaches may be attributed, among various other mechanisms, to the insufficient immunogenicity of self-protein-derived TAAs. Based on such an idea, the use of homologous xenogeneic antigens, derived from different species was suggested to overcome the natural immune tolerance to self TAAs. Xenoantigens are supposed to differ sufficiently from self antigens to a degree that renders them immunogenic, but at the same time preserves an optimal homology range with self proteins still allowing xenoantigens to induce cross-reactive T cells. Here we discuss the concept of xenogeneic vaccination, describe the cons and pros of autologous/allogeneic versus xenogeneic therapeutic cancer vaccines, present the results of various pre-clinical and several clinical studies and highlight the future perspectives of integrating xenovaccination into rapidly developing tumor immunotherapy regimens.


Assuntos
Antígenos Heterófilos/administração & dosagem , Vacinas Anticâncer/imunologia , Tolerância Imunológica , Imunoterapia Ativa/métodos , Neoplasias/terapia , Animais , Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Humanos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA