Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Chinese Journal of Biologicals ; (12): 11-15+20, 2023.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-965559

RESUMO

Objective@#To evaluate the inhibitory effect of tumor vaccines in colon carcinoma model mice.@*Methods@# Mouse bone marrow⁃derived dendritic cells(BMDCs)were stimulated by using CpG β⁃glucan nanoparticles(CNP)in vitro. The BMDCs were divided into PBS group,NP group(without CpG nanoparticles),Lysate group(MC38 cell lysate)and CpG group(CpG1826),which were determined for the expression of marker molecules on the surface by flow cytometry and for the contents of interleukin⁃6(IL⁃6)and IL⁃12p40 in the culture supernatant by ELISA. The tumor lysate nano⁃vaccine was pre⁃ pared by mixing 50 mg/mL tumor lysate(MC38 cell lysate)with 200 mg/mL CNP in a volume ratio of 1∶1,with which mice were subcutaneously immunized as Vaccine group. Vaccine group,PBS group,CNP group and Lysate group were im⁃ munized once a week,for three times in total. Mice were subcutaneously inoculated with MC38 cells,2 × 105 cells for each, in the right lower limb 1 h after the last immunization,and measured for tumor volume once every three days to plot the tumor growth curve. The ratios of CD3+ CD4+ T and CD3+ CD8+ T cells in the blood were analyzed by flow cytometry and the levels of tumor necrosis factor⁃α(TNF⁃α)and interferon γ(IFNγ)in the blood and spleen of mice were determined by ELISA.@*Results@# CNP effectively increased the expression of CD11c+ CD80+,CD11c+ CD86+,CD11c+ MHC⁃Ⅱ+ and the secretion of IL⁃6 and IL⁃12p40 in BMDCs in vitro,which were significantly higher than those in other 4 groups(t = 4. 3 ~ 46. 2,each P < 0. 05). Compared with that of the other three groups,the tumor volume of mice in Vaccine group decreased significantly(t =2.6~3.4,eachP <0. 05);TherewasnosignificantdifferenceinCD3+ CD8+ TandCD3+ CD8+ Tcellratios(t = 0.5~ 1. 9,each P > 0. 05);The content of IFNγ in blood increased significantly(t = 3. 8 ~ 4. 6,P < 0. 05),while thatof TNF⁃α showed no significant difference(t = 0. 4 ~ 2. 0,each P > 0. 05);However,the contents of IFN γ and TNF⁃α in spleen increased significantly(t = 6. 3 ~ 13. 0,each P < 0. 001).@*Conclusion@#The prepared nano⁃vaccine of tumor lysate improvedtheimmune level in mice and effectively inhibited the growth of colon carcinoma.

2.
Int Immunopharmacol ; 107: 108684, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35272171

RESUMO

Cancer stem cells (CSCs), which represent the root cause of resistance to conventional treatments, recurrence, and metastasis, constitute the critical point of failure in cancer treatments. Targeting CSCs with dendritic cell (DC)-based vaccines have been an effective strategy, but sialic acids on the surface of DCs limit the interaction with loaded antigens. We hypothesized that removal of sialic acid moieties on immature DCs (iDCs) could significantly affect DC-CSC-antigen loading, thereby leading to DC maturation and improving immune recognition and activity. The lysate of CD44+/CD24-/low breast CSCs (BCSCs) was pulsed with sialidase-treated DCs to obtain mature dendritic cells (mDCs). The roles of cytoskeletal elements in antigen uptake and dendritic cell maturation were determined by immunofluorescence staining, flow cytometry, and cytokine measurement, respectively. To test the efficacy of the vaccine in vivo, CSCs tumor-bearing mice were immunized with iDC or mDC. Pulsing DCs with antigen increased the expression levels of actin, gelsolin, talin, WASp, and Arp2, especially in podosome-like regions. Compared with iDCs, mDCs expressed high levels of CD40, CD80, CD86 costimulatory molecules and increased IL-12 production. Vaccination with mDC: i) increased CD8+ and CD4 + T-cell numbers, ii) prevented tumor growth with anti-mitotic activity and apoptotic induction, iii) suppressed metastasis by decreasing Snail, Slug, and Twist expressions. This study reveals for the first time that sialic acid removal and loading with CSC antigens induces significant molecular, morphological, and functional changes in DCs and that this new DC identity may be considered for future combined immunotherapy strategies against breast tumors.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Vacinas Anticâncer/uso terapêutico , Células Dendríticas , Camundongos , Ácido N-Acetilneuramínico , Células-Tronco Neoplásicas
3.
Macromol Biosci ; 22(2): e2100356, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34822219

RESUMO

Cancer vaccination is a powerful strategy to combat cancer. A very attractive approach to prime the immune system against cancer cells involves the use of tumor lysate as antigen source. The immunogenicity of tumor lysate can be further enhanced by treatment with hypochlorous acid. This study explores poly(lactic-co-glycolic acid) (PLGA) nanoparticles to enhance the delivery of oxidized tumor lysate to dendritic cells. Using human donor-derived dendritic cells, it is found that the use of PLGA nanoparticles enhances antigen uptake and dendritic cell maturation, as compared to the use of the free tumor lysate. The ability of the activated dendritic cells to stimulate autologous peripheral blood mononuclear cells (PBMCs) is assessed in vitro by coculturing PBMCs with A375 melanoma cells. Live cell imaging analysis of this experiment highlights the potential of nanoparticle-mediated dendritic-cell-based vaccination approaches. Finally, the efficacy of the PLGA nanoparticle formulation is evaluated in vivo in a therapeutic vaccination study using B16F10 tumor-bearing C57BL/6J mice. Animals that are challenged with the polymer nanoparticle-based oxidized tumor lysate formulation survive for up to 50 days, in contrast to a maximum of 41 days for the group that receives the corresponding free oxidized tumor lysate-based vaccine.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Animais , Células Dendríticas , Leucócitos Mononucleares , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
4.
Vaccines (Basel) ; 9(5)2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34069348

RESUMO

BACKGROUND: Malignant pleural mesothelioma (MPM) is a fatal neoplasm with, if untreated, poor survival of approximately nine months from diagnosis. Until recently, phase II-III immunotherapy trials did not show any significant benefit. The lack of immunotherapy efficacy can be explained by the fact that mesothelioma is a tumor with an "immune desert" phenotype, meaning a non-inflamed tumor characterized by low T-cell infiltration. By administration of DCs, which were ex-vivo cultured, exposed to (tumor-associated) antigens, and subsequently activated, this "immune desert" phenotype might be turned into an "inflamed" phenotype. Three phase I/II studies have been performed and published using activated DCs, which support this concept. We here report on the long-term survival of patients treated with DCs in three phase I/II studies. METHODS: Survival data of the phase I/II trials using DC therapy in MPM patients were obtained and subsequently analyzed. In the first two trials, DCs were loaded with autologous tumor lysate. In the third trial, DCs were loaded with allogeneic mesothelioma tumor cell line lysate. RESULTS: In the three studies combined, 29 patients with MPM were treated with DC vaccination between 2006 and 2015. At data cut-off, the median OS was 27 months (95% CI: 21-47 months). OS at 2 years was 55.2% (95% CI: 39.7-76.6%), and OS at 5 years was 20.7% (95% CI: 10.1-42.2%). CONCLUSIONS: The long-term survival of DC therapy in MPM in these three trials is promising, which is the basis for the randomized phase II/III DENIM study. This DENIM study is currently enrolling, and the results of which have to be awaited for definite conclusions.

5.
ACS Appl Bio Mater ; 4(12): 8291-8300, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-35005925

RESUMO

Peptides and proteins represent an emerging class of powerful therapeutics. Peptide and protein nanogels are attractive carriers for the transport and delivery of biologically active peptides and proteins because they allow essentially quantitative encapsulation of these biologics. One interesting field of use of peptide and protein nanogels is the transport of antigens and adjuvants in cancer immunotherapy. This study demonstrates the use of reduction-sensitive protein nanogels for the delivery of ovalbumin and oxidized tumor lysate-based antigens to mouse and human-donor-derived dendritic cells. Challenging mouse-derived and human dendritic cells with reduction-sensitive ovalbumin nanogels was found to significantly enhance antigen uptake as compared to the use of the corresponding free protein antigen. The experiments with mouse-derived dendritic cells further showed that the administration of ovalbumin in the form of reduction-sensitive nanogels enhanced dendritic cell maturation as well as the presentation of the SIINFEKL epitope as compared to experiments that use free ovalbumin. In addition to ovalbumin as a model antigen, the feasibility of reduction-sensitive nanogels was also demonstrated for the delivery of oxidized, whole tumor lysate-based cancer antigens. In experiments with dendritic cells harvested from human donors, dendritic cell uptake of the oxidized tumor lysate antigen was significantly enhanced in experiments that used oxidized tumor lysate nanogels as compared to the free antigen.


Assuntos
Antígenos de Neoplasias , Neoplasias , Animais , Células Dendríticas , Humanos , Camundongos , Nanogéis , Ovalbumina , Peptídeos
6.
J Exp Clin Cancer Res ; 39(1): 257, 2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33228738

RESUMO

Gliomas, especially glioblastomas, represent one of the most aggressive and difficult-to-treat human brain tumors. In the last few decades, clinical immunotherapy has been developed and has provided exceptional achievements in checkpoint inhibitors and vaccines for cancer treatment. Immunization with cellular vaccines has the advantage of containing specific antigens and acceptable safety to potentially improve cancer therapy. Based on T cells, dendritic cells (DC), tumor cells and natural killer cells, the safety and feasibility of cellular vaccines have been validated in clinical trials for glioma treatment. For TAA engineered T cells, therapy mainly uses chimeric antigen receptors (IL13Rα2, EGFRvIII and HER2) and DNA methylation-induced technology (CT antigen) to activate the immune response. Autologous dendritic cells/tumor antigen vaccine (ADCTA) pulsed with tumor lysate and peptides elicit antigen-specific and cytotoxic T cell responses in patients with malignant gliomas, while its pro-survival effect is biased. Vaccinations using autologous tumor cells modified with TAAs or fusion with fibroblast cells are characterized by both effective humoral and cell-mediated immunity. Even though few therapeutic effects have been observed, most of this therapy showed safety and feasibility, asking for larger cohort studies and better guidelines to optimize cellular vaccine efficiency in anti-glioma therapy.


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/imunologia , Glioma/terapia , Imunoterapia/métodos , Neoplasias Encefálicas/patologia , Feminino , Glioma/patologia , Humanos , Masculino
7.
J Transl Med ; 18(1): 402, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33087163

RESUMO

BACKGROUND: Cancer vaccines provide a complex source of neoantigens. Still, increasing evidence reveals that the neoantigen quality rather than the quantity is predictive for treatment outcome. METHODS: Using the preclinical Mlh1-/- tumor model, we performed a side-by side comparison of two autologous cell-line derived tumor lysates (namely 328 and A7450 T1 M1) harboring different tumor mutational burden (TMB; i.e. ultra-high: 328; moderate-high: A7450 T1 M1). Mice received repetitive prophylactic or therapeutic applications of the vaccine. Tumor incidence, immune responses and tumor microenvironment was examined. RESULTS: Both tumor cell lysates delayed tumor formation in the prophylactic setting, with the A7450 T1 M1 lysate being more effective in decelerating tumor growth than the 328 lysate (median overall survival: 37 vs. 25 weeks). Comparable results were achieved in therapeutic setting and could be traced back to antigen-driven immune stimulation. Reactive T cells isolated from A7450 T1 M1-treated mice recognized autologous Mlh1-/- tumor cells in IFNγ ELISpot, but likewise YAC-1 cells, indicative for stimulation of both arms of the immune system. By deciphering local effects, vaccines shaped the tumor microenvironment differently. While A7450 T1 M1 prophylactically vaccinated tumors harbored low numbers of myeloid-derived suppressor cells (MDSC) and elevated CD8-T cell infiltrates, vaccination with the 328 lysate evoked MDSC infiltration. Similar effects were seen in the therapeutic setting with stable disease induction only upon A7450 T1 M1 vaccination. Untangling individual response profiles revealed strong infiltration with LAG3+ and PD-L1+ immune cells when treatments failed, but almost complete exclusion of checkpoint-expressing lymphocytes in long-term survivors. CONCLUSIONS: By applying two tumor cell lysates we demonstrate that neoantigen quality outranks quantity. This should be considered prior to designing cancer vaccine-based combination approaches.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Linfócitos T CD8-Positivos , Linhagem Celular , Camundongos , Microambiente Tumoral , Vacinação
8.
Oncoimmunology ; 9(1): 1792058, 2020 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-32923156

RESUMO

Development of T cell-directed immune checkpoint inhibitors (ICI) has revolutionized metastatic melanoma (MM) therapy, but <50% of treated patients experience durable responses. This phase I trial (NCT01946373) investigates the safety/feasibility of tumor-infiltrating lymphocyte (TIL) adoptive cell therapy (ACT) combined with dendritic cell (DC) vaccination in MM patients progressing on ICI. An initial cohort (5 patients) received TIL therapy alone to evaluate safety and allow for optimization of TIL expansion protocols. A second cohort (first-in-man, 5 patients) received TIL combined with autologous tumor lysate-loaded DC vaccination. All patients received cyclophosphamide/fludarabine preconditioning prior to, and intravenous (i.v.) IL-2 after, TIL transfer. The DC vaccine was given as five intradermal injections after TIL and IL-2 administration. [18F]-FDG PET/CT radiology was performed to evaluate clinical response, according to RECIST 1.1 (on the CT part). Immunological monitoring was performed by flow cytometry and T-cell receptor (TCR) sequencing. In the safety/optimization cohort, all patients had a mixed response or stable disease, but none durable. In the combination cohort, two patients experienced complete responses (CR) that are still ongoing (>36 and >18 months, respectively). In addition, two patients had partial responses (PR), one still ongoing (>42 months) with only a small bone-lesion remaining, and one of short duration (<4 months). One patient died early during treatment and did not receive DC. Long-lasting persistency of the injected TILs was demonstrated in blood. In summary, we report clinical responses by TIL therapy combined with DC vaccination in 4 out of 4 treated MM patients who previously failed ICI.


Assuntos
Inibidores de Checkpoint Imunológico , Melanoma , Humanos , Imunoterapia Adotiva , Melanoma/terapia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Vacinação
9.
Vaccines (Basel) ; 8(2)2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570793

RESUMO

Background: Targeting inhibitory immune checkpoint molecules has highlighted the need to find approaches enabling the activation of immune responses against cancer. Therapeutic vaccination, which induces specific immune responses against tumor antigens (Ags), is an attractive option. Methods: Utilizing a K-RasG12Dp53null murine lung cancer model we determined tumor burden, tumor-infiltrating T cell (TIL) cytolysis, immunohistochemistry, flow cytometry, and CD4 and CD8 depletion to evaluate the efficacy of PD-1 blockade combined with CCL21-DC tumor lysate vaccine. Results: Anti-PD-1 plus CCL21-DC tumor lysate vaccine administered to mice bearing established tumors (150 mm3) increased expression of perforin and granzyme B in the tumor microenvironment (TME), increased tumor-infiltrating T cell (TIL) activity, and caused 80% tumor eradication. Mice with treatment-induced tumor eradication developed immunological memory, enabling tumor rejection upon challenge and cancer-recurrence-free survival. The depletion of CD4 or CD8 abrogated the antitumor activity of combined therapy. PD-1 blockade or CCL21-DC tumor lysate vaccine monotherapy reduced tumor burden without tumor eradication. Conclusion: Immune checkpoint blockade promotes the activity of the therapeutic cancer vaccine. PD-1 blockade plus CCL21-DC tumor lysate vaccine therapy could benefit lung cancer patients.

10.
Vaccines (Basel) ; 8(1)2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31947581

RESUMO

With the emergence of immune checkpoint inhibitors and adoptive T-cell therapies, there is a considerable interest in using personalized autologous dendritic cell (DC) vaccines in combination with T cell-targeting immunotherapies to potentially maximize the therapeutic impact of DC vaccines. Here, we describe the development and optimization of a Good Manufacturing Practice (GMP)-compliant manufacturing process based on tumor lysate as a tumor antigen source for the production of an oxidized tumor cell lysate loaded DC (OC-DC) vaccine. The manufacturing process required one day for lysate preparation and six days for OC-DC vaccine production. Tumor lysate production was standardized based on an optimal tumor digestion protocol and the immunogenicity was improved through oxidation using hypochloric acid prior to freeze-thaw cycles resulting in the oxidized tumor cell lysate (OC-L). Next, monocytes were selected using the CliniMACS prodigy closed system and were placed in culture in cell factories in the presence of IL-4 and GM-CSF. Immature DCs were loaded with OC-L and matured using MPLA-IFNγ. After assessing the functionality of the OC-DC cells (IL12p70 secretion and COSTIM assay), the OC-DC vaccine was cryopreserved in multiple doses for single use. Finally, the stability of the formulated doses was tested and validated. We believe this GMP-compliant DC vaccine manufacturing process will facilitate access of patients to personalized DC vaccines, and allow for multi-center clinical trials.

11.
Sovrem Tekhnologii Med ; 12(2): 34-41, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34513051

RESUMO

Although major progress has been made in the standard treatment for glioblastomas, encompassing the maximal surgical resection, chemotherapy and radiation therapy, it is possible to increase survival rates significantly only in a few patients. Therefore, it is necessary to explore new therapeutic modalities, one of which is immunotherapy. The aim of the study was to evaluate the efficacy of the combined use of autologous and pooled tumor lysates in comprehensive treatment of patients with glioblastoma. MATERIALS AND METHODS: All patients (n=58, including 30 males and 28 females aged 18-70 years) were randomized into three groups, two of which received immunotherapy based on injection of autologous dendritic cells pulsed with autologous tumor lysates (first protocol) or pooled lysates (second protocol) from more than one tumor, in addition to the planned standard treatment. The patients of group 3 (control) received the standard comprehensive treatment encompassing the maximum safe tumor resection followed by radiation therapy and chemotherapy. RESULTS: The tolerability of both applied immunotherapy protocols was good: there were no anaphylactic reactions observed or patients who prematurely discontinued participation in the study. The final analysis of the data revealed no significant differences in median survival values of patients in each of the three groups. However, when analyzing the Karnofsky Performance Status in patients of group 2, it was found that it tended to improve. CONCLUSION: The study shows that the proposed immunotherapy protocols are safe for clinical use and have the potential to improve the patient's life quality. However, these findings should be considered intermediate until the findings of multicenter randomized clinical trials with a larger number of patients are obtained.

12.
Immunol Invest ; 48(8): 794-808, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31094258

RESUMO

Stimulating antitumor T cells using dendritic cells (DCs) is a novel and promising method in cancer therapy. Poly lactic-co-glycolic acid is one of the best-known polymers used for encapsulating antigen to protect them against proteolytic enzymes. In this study, poly lactic-co-glycolic acid nanoparticles (NPs) were used as DC antigen delivery vehicles in a preclinical model of immunotherapy of gastric cancer. The DCs were generated from peripheral blood monocytes by conventional in vitro differentiation and loaded with either soluble tumor lysate or lysate encapsulated in NPs using a double emulsion/solvent evaporation technique. Morphology of NPs was determined by scanning electron microscopy. Tumor lysate, either in the soluble form or encapsulated in NPs, was loaded into DC and stimulatory capacity was compared using patient-derived autologous CD3+ T cells as responders. The amount of relevant cytokines produced by Ag-loaded DC and in DC/T cell cocultures was evaluated as a measure of initial DC stimulation and T-cell responses, respectively. Significance increases in expression of DC surface molecules (i.e., CD80, CD83, CD86, and Human Leukocyte Antigen-DR (HLA-DR)) and cytokine production by both DC and DC/T cell cocultures (i.e., interleukin (IL)-12:IL-10 and interferon [IFN]-γ:IL-4 ratios) was observed following loading with lysate NP versus controls. The results suggest that NP-encapsulated antigen can shift antitumor T-cell responses toward a Th1 bias, which potentially increases DC vaccine potency in clinical settings.


Assuntos
Antígenos de Neoplasias/imunologia , Células Dendríticas/imunologia , Imunoterapia/métodos , Nanopartículas/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Neoplasias Gástricas/terapia , Antígenos de Neoplasias/química , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Técnicas de Cocultura , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Humanos , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Microscopia Eletrônica de Varredura , Nanopartículas/ultraestrutura , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
13.
Cancers (Basel) ; 11(2)2019 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-30682795

RESUMO

Recent advances in immunotherapy have included inhibition of immune checkpoint proteins in the tumor microenvironment and tumor lysate-based vaccination strategies. We combined these approaches in pet dogs with high-grade glioma. Administration of a synthetic peptide targeting the immune checkpoint protein, CD200, enhanced the capacity of antigen-presenting cells to prime T-cells to mediate an anti-glioma response. We found that in canine spontaneous gliomas, local injection of a canine-specific, CD200-directed peptide before subcutaneous delivery of an autologous tumor lysate vaccine prolonged survival relative to a historical control treated with autologous tumor lysate alone (median survivals of 12.7 months and 6.36 months, respectively). Antigen-presenting cells and T-lymphocytes primed with this peptide suppressed their expression of the inhibitory CD200 receptor, thereby enhancing their ability to initiate immune reactions in a glioblastoma microenvironment replete with the immunosuppressive CD200 protein. These results support consideration of a CD200 ligand as a novel glioblastoma immunotherapeutic agent.

14.
J Immunother Cancer ; 7(1): 8, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30630527

RESUMO

BACKGROUND: Mismatch Repair Deficiency (MMR-D)-related tumors are highly immunogenic and constitute ideal vaccination targets. In a proof-of-concept study delayed tumorigenesis and prolonged survival has been shown in a clinically-relevant mouse model for MMR-D-related diseases (=MLH1 knock out mice). To refine this approach, vaccination was combined with immune modulatory low-dose chemotherapy to polarize immune regulatory subtypes. METHODS: Mice (prophylactic: 8-10 weeks; therapeutic: > 36 weeks) received a single injection of cyclophosphamide (CPX, 120 mg/kg bw, i.p.) or gemcitabine (GEM, 100 mg/kg bw, i.p.) prior to vaccination (lysate of a gastrointestinal tumor allograft, 10 mg/kg bw, n = 9 mice/group). The vaccine was given repetitively (10 mg/kg bw, s.c., 4 x / once a week, followed by monthly boosts) until tumor formation or progression. Tumor growth ([18F] FDG PET/CT imaging) and immune responses were monitored (flow cytometry, IFNγ ELISpot). The microenvironment was analyzed by immunofluorescence. RESULTS: Prophylactic application of GEM + lysate delayed tumorigenesis compared to lysate monotherapy and CPX-pre-treatment (median time of onset: 53 vs. 47 vs. 48 weeks). 33% of mice even remained tumor-free until the experimental endpoint (= 65 weeks). This was accompanied by long-term effect on cytokine plasma levels; splenic myeloid derived suppressor cells (MDSC) as well as regulatory T cell numbers. Assessment of tumor microenvironment from GEM + lysate treated mice revealed low numbers of MDSCs, but enhanced T cell infiltration, in some cases co-expressing PD-L1. Therapeutic chemo-immunotherapy (GEM + lysate) had minor impact on overall survival (median time: 12 (GEM + lysate) vs. 11.5 (lysate) vs. 3 weeks (control)), but induced complete remission in one case. Dendritic and T cell infiltrates increased in both treatment groups. Reactive T cells specifically recognized MLH1-/- tumor cells in IFNγ ELISpot, but lacked response towards NK cell targets YAC-1. CONCLUSIONS: Combined chemo-immunotherapy impairs tumor onset and growth likely attributable to modulation of immune responses. Depleting or 're-educating' immunosuppressive cell types, such as MDSC, may help moving a step closer to combat cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Vacinas Anticâncer/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Ciclofosfamida/uso terapêutico , Desoxicitidina/análogos & derivados , Neoplasias Gastrointestinais/tratamento farmacológico , Imunoterapia , Síndromes Neoplásicas Hereditárias/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Desoxicitidina/uso terapêutico , Camundongos , Células Supressoras Mieloides/imunologia , Baço/citologia , Linfócitos T Reguladores/imunologia , Gencitabina
15.
BMC Complement Altern Med ; 18(1): 232, 2018 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-30081891

RESUMO

BACKGROUND: Dendritic cells (DCs) are unique antigen presenting cells (APC) which play a pivotal role in immunotherapy and induction of an effective immune response against tumors. In the present study, 80% ethanol extract of Phyllanthus amarus was used to generate tumor lysate (TLY) derived from HCT 116 and MCF-7 cancer cell lines via induction of apoptosis. Monocyte-derived DCs were generated ex vivo from the adherent population of peripheral blood mononuclear cells (PBMCs). The generated TLY were used to impulse DCs to investigate its effect on their cellular immune functions including antigen presentation capacity, phagocytic activity, chemotaxis capacity, T-cell proliferation and cytokines release. METHODS: The effect of P. amarus-generated TLY on DCs maturation was evaluated by determination of MHC class I, II and CD 11c expression as well as the co-stimulatory molecules CD 83 and 86 by using flow cytometry. The phagocytic capacity of TLY-pulsed DCs was investigated through FITC-dextran uptake by using flow cytometry. The effect on the cytokines release including IL-12, IL-6 and IL-10 was elucidated by using ELISA. The migration capacity and T cell proliferation activity of pulsed DCs were measured. The relative gene expression levels of cytokines were determined by using qRT-PCR. The major constituents of P. amarus extract were qualitatively and quantitatively analyzed by using validated reversed-phase high performance liquid chromatography (HPLC) methods. RESULTS: P. amarus-generated TLY significantly up-regulated the expression levels of MHC class I, CD 11 c, CD 83 and 86 in pulsed DCs. The release of interleukin IL-12 and IL-6 was enhanced by TLY-DCs at a ratio of 1 DC: 3 tumor apoptotic bodies (APO), however, the release of IL-10 was suppressed. The migration ability as well as allogeneic T-cell proliferation activities of loaded DCs were significantly enhanced, but their phagocytic capacity was highly attenuated. The gene expression profiles for IL-12 and IL-6 of DCs showed increase in their mRNA gene expression in TLY pulsed DCs versus unloaded and LPS-treated only DCs. CONCLUSION: The effect of P. amarus-generated TLY on the immune effector mechanisms of DCs verified its potential to induce an in vitro anti-tumor immune response against the recognized tumor antigen.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fatores Imunológicos/farmacologia , Phyllanthus/química , Extratos Vegetais/farmacologia , Apresentação de Antígeno/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/imunologia , Fagocitose/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
16.
Cancer ; 123(9): 1576-1584, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28241093

RESUMO

BACKGROUND: There are limited options for the curative treatment of refractory bone and soft tissue sarcomas. The purpose of this phase 1/2 study was to assess the immunological and clinical effects of dendritic cells (DCs) pulsed with autologous tumor lysate (TL) in patients with advanced bone and soft tissue sarcomas. METHODS: Thirty-seven patients with metastatic or recurrent sarcomas were enrolled in this study. Peripheral blood mononuclear cells obtained from the patients were suspended in media containing interleukin 4 (IL-4) and granulocyte-macrophage colony-stimulating factor. Subsequently, these cells were treated with TL, tumor necrosis factor α, and OK-432. The DCs were injected into the inguinal or axillary region. One treatment course comprised 6 weekly DC injections. The toxicity, clinical response (tumor volume, serum interferon-γ [IFN-γ], and serum IL-12), and oncological outcomes were observed. RESULTS: In total, 47 courses of DC therapy were performed in 37 patients. No severe adverse events or deaths associated with the DC injections were observed in the study patients. Increased serum IFN-γ and IL-12 levels were observed 1 month after the DC injection. Among the 37 patients, 35 patients were assessed for clinical responses: 28 patients showed tumor progression, 6 patients had stable disease, and 1 patient showed a partial response 8 weeks after the DC injection. The 3-year overall and progression-free survival rates of the patients were 42.3% and 2.9%, respectively. CONCLUSIONS: Although DC therapy appears safe and resulted in an immunological response in patients with refractory sarcoma, it resulted in an improvement of the clinical outcome in only a small number of patients. Cancer 2017;123:1576-1584. © 2017 American Cancer Society.


Assuntos
Neoplasias Ósseas/terapia , Células Dendríticas , Imunoterapia/métodos , Leucócitos Mononucleares , Sarcoma/terapia , Neoplasias de Tecidos Moles/terapia , Adolescente , Adulto , Idoso , Antineoplásicos , Neoplasias Ósseas/sangue , Criança , Condrossarcoma/sangue , Condrossarcoma/terapia , Intervalo Livre de Doença , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Histiocitoma Fibroso Maligno/sangue , Histiocitoma Fibroso Maligno/terapia , Humanos , Interferon gama/sangue , Interleucina-12/sangue , Interleucina-4 , Leiomiossarcoma/sangue , Leiomiossarcoma/terapia , Masculino , Pessoa de Meia-Idade , Osteossarcoma/sangue , Osteossarcoma/terapia , Picibanil , Sarcoma/sangue , Sarcoma de Células Claras/sangue , Sarcoma de Células Claras/terapia , Sarcoma Sinovial/sangue , Sarcoma Sinovial/terapia , Neoplasias de Tecidos Moles/sangue , Resultado do Tratamento , Fator de Necrose Tumoral alfa , Adulto Jovem
17.
Tumour Biol ; 2016 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-27726097

RESUMO

Dendritic cells (DC) have been exploited for vaccination against cancer for years. DC loading autologous tumor lysate (ATL-DC) have been assessed in ongoing clinical trials, but frequently do not meet expectation. In this study, we found that mice immunized with ATL-DC induced less protective anti-tumor effect than immunized with DC alone. The percentage of CD8+ T cells and the lysis efficiency of CTLs to auto tumor cells in ATL-DC vaccination group was less than that of DC group. Moreover, vaccination of mice with ATL-DC also promoted tumor angiogenesis by analyzing the CD31 positive microvessel density and hemoglobin content of tumor specimens. Human umbilical vein endothelial cells (HUVEC) have been proved effective in the anti-angiogenesis immunity against cancer. However, in the following research we found that the anti-tumor effect was attenuated while immunized mice with HUVEC combined with ATL-DC (HUVEC + ATL-DC). Furthermore, immunized mice with HUVEC + ATL-DC profoundly increased the tumor angiogenesis by analyzing the microvessel density and hemoglobin content of tumor specimens. These data suggest that vaccination using ATL-DC antagonized HUVEC induced anti-angiogenesis effect. Our research for the first time indicated that ATL-DC have the potential to promote the process of tumor angiogenesis in vivo. As vaccines based on DC loading autologous tumor lysate have been used in clinical, this find warned that the safety of this kind of vaccine should be taken into consideration seriously.

18.
Expert Opin Emerg Drugs ; 21(2): 133-45, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27223671

RESUMO

INTRODUCTION: Immunotherapy for brain cancer has evolved dramatically over the past decade, owed in part to our improved understanding of how the immune system interacts with tumors residing within the central nervous system (CNS). Glioblastoma (GBM), the most common primary malignant brain tumor in adults, carries a poor prognosis (<15 months) and only few advances have been made since the FDA's approval of temozolomide (TMZ) in 2005. Importantly, several immunotherapies have now entered patient trials based on promising preclinical data, and recent studies have shed light on how GBM employs a slew of immunosuppressive mechanisms that may be targeted for therapeutic gain. Altogether, accumulating evidence suggests immunotherapy may soon earn its keep as a mainstay of clinical management for GBM. AREAS COVERED: Here, we review cancer vaccines, checkpoint inhibitors, adoptive T-cell immunotherapy, and oncolytic virotherapy. EXPERT OPINION: Checkpoint blockade induces antitumor activity by preventing negative regulation of T-cell activation. This platform, however, depends on an existing frequency of tumor-reactive T cells. GBM tumors are exceptionally equipped to prevent this, occupying low levels of antigen expression and elaborate mechanisms of immunosuppression. Therefore, checkpoint blockade may be most effective when used in combination with a DC vaccine or adoptively transferred tumor-specific T cells generated ex vivo. Both approaches have been shown to induce endogenous immune responses against tumor antigens, providing a rationale for use with checkpoint blockade where both primary and secondary responses may be potentiated.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Imunoterapia/métodos , Adulto , Animais , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Vacinas Anticâncer/administração & dosagem , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Terapia Viral Oncolítica/métodos , Prognóstico , Linfócitos T/imunologia
19.
Oncoimmunology ; 5(2): e1083669, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27057467

RESUMO

Dendritic cell (DC)-based immunotherapy has yielded promising results against high-grade glioma (HGG). However, the efficacy of DC vaccines is abated by HGG-induced immunosuppression and lack of attention toward the immunogenicity of the tumor lysate/cells used for pulsing DCs. A literature analysis of DC vaccination clinical trials in HGG patients delineated the following two most predominantly applied methods for tumor lysate preparation: freeze-thaw (FT)-induced necrosis or FT-necrosis followed by X-ray irradiation. However, from the available clinical evidence, it is unclear which of both methodologies has superior immunogenic potential. Using an orthotopic HGG murine model (GL261-C57BL/6), we observed that prophylactic vaccination with DCs pulsed with irradiated FT-necrotic cells (compared to FT-necrotic cells only) prolonged overall survival by increasing tumor rejection in glioma-challenged mice. This was associated, both in prophylactic and curative vaccination setups, with an increase in brain-infiltrating Th1 cells and cytotoxic T lymphocytes (CTL), paralleled by a reduced accumulation of regulatory T cells, tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). Further analysis showed that irradiation treatment of FT-necrotic cells considerably increased the levels of carbonylated proteins - a surrogate-marker of oxidation-associated molecular patterns (OAMPs). Through further application of antioxidants and hydrogen peroxide, we found a striking correlation between the amount of lysate-associated protein carbonylation/OAMPs and DC vaccine-mediated tumor rejection capacity thereby suggesting for the first time a role for protein carbonylation/OAMPs in at least partially mediating antitumor immunity. Together, these data strongly advocate the use of protein oxidation-inducing modalities like irradiation for increasing the immunogenicity of tumor lysate/cells used for pulsing DC vaccines.

20.
Springerplus ; 4: 780, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26702369

RESUMO

For the production of tumor-specific vaccines, including dendritic cell (DC) vaccines, the tumor cells themselves are an ideal source. Floating tumor cells in the ascites fluid from patients with malignant ascites are a good candidate source, but it is not easy to obtain pure tumor cells from ascites because of various types of cell contamination as well as protein aggregates. We here report an effective method to recover pure tumor cells from malignant ascites. We used lavage fluid from 13 patients with malignant ascites who were treated with modified cell-free and concentrated ascites reinfusion therapy (KM-CART). Cellular components were separated from the lavage fluid by centrifugation, enzymatic digestion and hemolysis. Tumor cells were purified by depleting CD45(+) leukocytes with antibody-conjugated magnetic beads. The tumor cell lysate was extracted by freeze-and-thaw cycles. The mean obtained total cell number was 7.50 × 10(7) cells (range 4.40 × 10(6)-2.48 × 10(8) cells). From this fraction, 6.39 × 10(6) (range 3.23 × 10(5)-2.53 × 10(7)) CD45(-) cells were collected, and the tumor cell purity was over 80 % defined as CD45(-)CD326(+). A sufficient amount of tumor lysate, average  = 2416 µg (range 25-8743 µg), was extracted from CD45(-)CD326(+) tumor cells. We here established an effective method to produce highly purified tumor cells from KM-CART lavage fluid. The clinical feasibility of this simple preparation method for generating tumor lysate should be examined in clinical studies of DC vaccines.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...