Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 818
Filtrar
1.
STAR Protoc ; 5(2): 103047, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38691463

RESUMEN

The tumor-associated mucin MUC1 is overexpressed in almost all types of epithelial tumor tissues, making it an attractive target antigen for cancer immunotherapy. Here we present a protocol to prepare MUC1 glycopeptide vaccines and to evaluate immunization effects in mice. We describe steps for synthesizing glycopeptide antigen and conjugating it with carrier protein to make vaccine candidates. We then detail procedures for mice immunization, antibody response evaluation, and cellular immune response. For complete details on the use and execution of this protocol, please refer to Cai et al.1,2.


Asunto(s)
Vacunas contra el Cáncer , Glicopéptidos , Mucina-1 , Animales , Mucina-1/inmunología , Ratones , Glicopéptidos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunización/métodos , Femenino
2.
J Med Chem ; 67(8): 6822-6838, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38588468

RESUMEN

Weak antigens represented by MUC1 are poorly immunogenic, which greatly constrains the development of relevant vaccines. Herein, we developed a multifunctional lipidated protein as a carrier, in which the TLR1/2 agonist Pam3CSK4 was conjugated to the N-terminus of MUC1-loaded carrier protein BSA through pyridoxal 5'-phosphate-mediated transamination reaction. The resulting Pam3CSK4-BSA-MUC1 conjugate was subsequently incorporated into liposomes, which biomimics the membrane structure of tumor cells. The results indicated that this lipidated protein carrier significantly enhanced antigen uptake by APCs and obviously augmented the retention of the vaccine at the injection site. Compared with the BSA-MUC1 and BSA-MUC1 + Pam3CSK4 groups, Pam3CSK4-BSA-MUC1 evoked 22- and 11-fold increases in MUC1-specific IgG titers. Importantly, Pam3CSK4-BSA-MUC1 elicited robust cellular immunity and significantly inhibited tumor growth. This is the first time that lipidated protein was constructed to enhance antigen immunogenicity, and this universal carrier platform exhibits promise for utilization in various vaccines, holding the potential for further clinical application.


Asunto(s)
Liposomas , Mucina-1 , Animales , Mucina-1/inmunología , Mucina-1/química , Ratones , Humanos , Lipopéptidos/química , Lipopéptidos/inmunología , Lipopéptidos/farmacología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/química , Albúmina Sérica Bovina/química , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Femenino , Ratones Endogámicos BALB C , Antígenos/inmunología , Línea Celular Tumoral
3.
Int J Mol Sci ; 24(17)2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37685842

RESUMEN

Specific changes in mucin-type O-glycosylation are common for many cancers, including gastric ones. The most typical alterations include incomplete synthesis of glycan structures, enhanced expression of truncated O-glycans (Tn, T antigens and their sialylated forms), and overexpression of fucosylation. Such altered glycans influence many cellular activities promoting cancer development. Tiliroside is a glycosidic dietary flavonoid with pharmacological properties, including anti-cancer. In this study, we aim to assess the effect of the combined action of anti-MUC1 and tiliroside on some cancer-related factors in AGS gastric cancer cells. Cancer cells were treated with 40, 80, and 160 µM tiliroside, 5 µg/mL anti-MUC1, and flavonoid together with mAb. Real-Time PCR, ELISA, and Western blotting were applied to examine MUC1 expression, specific, tumor-associated antigens, enzymes taking part in their formation, Gal-3, Akt, and NF-κB. MUC1 expression was significantly reduced by mAb action. The combined action of anti-MUC1 and tiliroside was more effective in comparison with monotherapy in the case of C1GalT1, ST3GalT1, FUT4, Gal-3, NF-κB, Akt mRNAs, and Tn antigen, as well as sialyl T antigen expression. The results of our study indicate that applied combined therapy may be a promising anti-gastric cancer strategy.


Asunto(s)
FN-kappa B , Neoplasias Gástricas , Humanos , Anticuerpos Monoclonales/farmacología , Flavonoides , Fucosiltransferasas , Proteínas Proto-Oncogénicas c-akt , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/terapia , Mucina-1/inmunología
4.
Int J Immunopathol Pharmacol ; 36: 3946320221112358, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35839304

RESUMEN

INTRODUCTION: Naked DNA is one of the attractive tools for vaccination studies. We studied naked DNA vaccination against the human tumor antigen, mucin, which is encoded by the MUC1 gene. METHODS: We constructed the pcDNA3.0-MUC1 (pcDNA-MUC1) plasmid expressing an underglycosylated MUC1 protein. BALB/c mice were immunized intradermally thrice at 2-weeks intervals with pcDNA-MUC1. Two weeks after the last immunization, tumor challenge experiments were performed using either the CT26 or TA3HA tumor cell lines, both of which transduce human MUC1. RESULTS: Immune cell population monitoring from pcDNA-MUC1-immunized animals indicated that immune cell activation was induced by MUC1-specific immunization. Using intracellular fluorescence activated cell sorting and enzyme-linked immunosorbent spot assay, we reported that interferon-γ secreting CD8+ T cells were mainly involved in MUC1-specific immunization. In all mice immunized with MUC1 DNA, tumor growth inhibition was observed, whereas control mice developed tumors (p < 0.001). CONCLUSION: Our results suggest that intradermal immunization with MUC1 DNA induces MUC1-specific CD8+ T cell infiltration into tumors, elicits tumor-specific Th1-type immune response, and inhibits tumor growth.


Asunto(s)
Mucina-1 , Neoplasias , Animales , Linfocitos T CD8-positivos , ADN/genética , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Mucina-1/genética , Mucina-1/inmunología , Mucina-1/uso terapéutico , Neoplasias/metabolismo , Plásmidos/genética , Plásmidos/uso terapéutico , Vacunación/métodos
5.
Cell Mol Gastroenterol Hepatol ; 14(4): 789-811, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35809803

RESUMEN

BACKGROUND & AIMS: MUC1 is abnormally expressed in colorectal cancer, including colitis-associated colorectal cancer (CAC), but its role in tumorigenesis is unclear. This study investigated MUC1's effects in murine models of colitis and CAC and elucidated mechanisms of action. METHODS: Colitis and CAC were induced in mice by exposure to dextran sodium sulfate or azoxymethane plus dextran sodium sulphate. Clinical parameters, immune cell infiltration, and tumor development were monitored throughout disease progression. Experiments in knockout mice and bone marrow chimeras were combined with an exploration of immune cell abundance and function. RESULTS: Deficiency of Muc1 suppressed inflammation, inhibited tumor progression, increased abundance of CD8+ T lymphocytes, and reduced abundance of macrophages in colon tumors. Bone marrow chimeras showed promotion of CAC was primarily mediated by Muc1-expressing hematopoietic cells, and that MUC1 promoted a pro-tumoral immunosuppressive macrophage phenotype within tumors. Mechanistic studies revealed that Muc1 deficiency remarkably reduced interleukin-6 levels in the colonic tissues and tumors that was mainly produced by infiltrating macrophages at day 21, 42, and 85. In bone marrow-derived macrophages, MUC1 promoted responsiveness to chemoattractant and promoted activation into a phenotype with high Il6 and Ido1 expression, secreting factors which inhibited CD8+ T cell proliferation. MUC1 potently drives macrophages to produce interleukin-6, which in turn drives a pro-tumorigenic activation of signal transducer and activator of transcription 3 in colon epithelial tumor and stromal cells, ultimately increasing the occurrence and development of CAC. CONCLUSIONS: Our findings provide cellular and molecular mechanisms for the pro-tumorigenic functions of MUC1 in the inflamed colon. Therapeutic strategies to inhibit MUC1 signal transduction warrant consideration for the prevention or therapy of CAC.


Asunto(s)
Neoplasias Asociadas a Colitis , Interleucina-6 , Activación de Macrófagos , Mucina-1 , Factor de Transcripción STAT3 , Animales , Azoximetano/toxicidad , Carcinogénesis , Factores Quimiotácticos , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/inmunología , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Sulfato de Dextran/toxicidad , Interleucina-6/genética , Interleucina-6/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Ratones , Ratones Noqueados , Mucina-1/genética , Mucina-1/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología
6.
Oncoimmunology ; 11(1): 2029298, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35127252

RESUMEN

The oncogenic MUC1-C protein drives dedifferentiation of castrate resistant prostate cancer (CRPC) cells in association with chromatin remodeling. The present work demonstrates that MUC1-C is necessary for expression of IFNGR1 and activation of the type II interferon-gamma (IFN-γ) pathway. We show that MUC1-C→ARID1A/BAF signaling induces IFNGR1 transcription and that MUC1-C-induced activation of the NuRD complex suppresses FBXW7 in stabilizing the IFNGR1 protein. MUC1-C and NuRD were also necessary for expression of the downstream STAT1 and IRF1 transcription factors. We further demonstrate that MUC1-C and PBRM1/PBAF are necessary for IRF1-induced expression of (i) IDO1, WARS and PTGES, which metabolically suppress the immune tumor microenvironment (TME), and (ii) the ISG15 and SERPINB9 inhibitors of T cell function. Of translational relevance, we show that MUC1 associates with expression of IFNGR1, STAT1 and IRF1, as well as the downstream IDO1, WARS, PTGES, ISG15 and SERPINB9 immunosuppressive effectors in CRPC tumors. Analyses of scRNA-seq data further demonstrate that MUC1 correlates with cancer stem cell (CSC) and IFN gene signatures across CRPC cells. Consistent with these results, MUC1 associates with immune cell-depleted "cold" CRPC TMEs. These findings demonstrate that MUC1-C integrates chronic activation of the type II IFN-γ pathway and induction of chromatin remodeling complexes in linking the CSC state with immune evasion.


Asunto(s)
Ensamble y Desensamble de Cromatina , Interferón gamma , Mucina-1 , Neoplasias de la Próstata Resistentes a la Castración , Ensamble y Desensamble de Cromatina/inmunología , Humanos , Terapia de Inmunosupresión , Masculino , Mucina-1/inmunología , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores de Interferón/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral , Receptor de Interferón gamma
7.
Mol Oncol ; 16(2): 485-507, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34694686

RESUMEN

Alteration in glycosylation pattern of MUC1 mucin tandem repeats during carcinomas has been shown to negatively affect adhesive properties of malignant cells and enhance tumor invasiveness and metastasis. In addition, MUC1 overexpression is closely interrelated with angiogenesis, making it a great target for immunotherapy. Alongside, easier interaction of nanobodies (single-domain antibodies) with their antigens, compared to conventional antibodies, is usually associated with superior desirable results. Herein, we evaluated the preclinical efficacy of a recombinant nanobody against MUC1 tandem repeats in suppressing tumor growth, angiogenesis, invasion, and metastasis. Expressed nanobody demonstrated specificity only toward MUC1-overexpressing cancer cells and could internalize in cancer cell lines. The IC50 values (the concentration at which the nanobody exerted half of its maximal inhibitory effect) of the anti-MUC1 nanobody against MUC1-positive human cancer cell lines ranged from 1.2 to 14.3 nm. Similar concentrations could also effectively induce apoptosis in MUC1-positive cancer cells but not in normal cells or MUC1-negative human cancer cells. Immunohistochemical staining of spontaneously developed mouse breast tumors prior to in vivo studies confirmed cross-reactivity of nanobody with mouse MUC1 despite large structural dissimilarities between mouse and human MUC1 tandem repeats. In vivo, a dose of 3 µg nanobody per gram of body weight in tumor-bearing mice could attenuate tumor progression and suppress excessive circulating levels of IL-1a, IL-2, IL-10, IL-12, and IL-17A pro-inflammatory cytokines. Also, a significant decline in expression of Ki-67, MMP9, and VEGFR2 biomarkers, as well as vasculogenesis, was evident in immunohistochemically stained tumor sections of anti-MUC1 nanobody-treated mice. In conclusion, the anti-MUC1 tandem repeat nanobody of the present study could effectively overcome tumor growth, invasion, and metastasis.


Asunto(s)
Proliferación Celular/genética , Neoplasias Mamarias Animales/patología , Mucina-1/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Neovascularización Patológica/genética , Anticuerpos de Dominio Único/genética , Secuencias Repetidas en Tándem , Animales , Apoptosis/genética , Línea Celular Tumoral , Quimiocinas/metabolismo , Reacciones Cruzadas , Citocinas/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Mucina-1/inmunología , Unión Proteica , Anticuerpos de Dominio Único/inmunología
8.
ESMO Open ; 7(1): 100311, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34920291

RESUMEN

BACKGROUND: Gatipotuzumab is a humanized monoclonal antibody recognizing the carbohydrate-induced epitope of the tumor-associated mucin-1 (TA-MUC1). This study aimed to evaluate the efficacy and safety of switch maintenance therapy with gatipotuzumab in patients with TA-MUC1-positive recurrent ovarian, fallopian tube, or primary high-grade serous peritoneal cancer. PATIENTS AND METHODS: In this double-blind, randomized, placebo-controlled, phase II trial, patients with at least stable disease (SD) following chemotherapy were randomized 2:1 to receive intravenous gatipotuzumab (500 mg followed by 1700 mg 1 week later) or placebo every 3 weeks until tumor progression or unacceptable toxicity occurred. Stratification factors were the number of prior chemotherapy lines (2 versus 3-5), response versus SD after the most recent chemotherapy, and progression-free survival (PFS) <6 versus 6-12 months following the prior therapy. Primary endpoint was PFS according to modified immune-related RECIST 1.1 response criteria. Secondary endpoints were PFS at 6 months, safety, overall response rate, CA-125 progression, overall survival, quality of life, and pharmacokinetics. RESULTS: Overall, 216 patients were randomized to gatipotuzumab (n  = 151) or placebo (n  = 65). Median PFS with gatipotuzumab was 3.5 months as compared with 3.5 months with placebo (hazard ratio 0.96, 95% confidence interval 0.69-1.33, P  = 0.80). No advantage for gatipotuzumab over placebo was seen in the secondary efficacy endpoints or in any stratified subgroups. Gatipotuzumab was well tolerated, with mild to moderate infusion-related reactions being the most common adverse events. CONCLUSIONS: Gatipotuzumab switch maintenance therapy does not improve outcome in TA-MUC1-positive ovarian cancer patients. TRIAL REGISTRATION: ClinicalTrials.govNCT01899599; https://clinicaltrials.gov/ct2/show/NCT01899599.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Antineoplásicos Inmunológicos , Mucina-1 , Neoplasias Ováricas , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Método Doble Ciego , Femenino , Humanos , Quimioterapia de Mantención , Mucina-1/inmunología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Neoplasias Ováricas/tratamiento farmacológico , Calidad de Vida
9.
Mikrochim Acta ; 189(1): 38, 2021 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-34958417

RESUMEN

A sensitive detection of carbohydrate antigen 15-3 (CA15-3) levels may allow for early diagnosis and monitoring the treatment of breast cancer, but this can only be made in routine clinical practice if low-cost immunosensors are available. In this work, we developed a sandwich-type electrochemical immunosensor capable of rapid detection of CA15-3 with an ultra-low limit of detection (LOD) of 0.08 fg mL-1 within a wide linear concentration range from 0.1 fg mL-1 to 1 µg mL-1. The immunosensor had a matrix of a layer-by-layer film of Au nanoparticles and reduced graphene oxide (Au-rGO) co-electrodeposited on screen-printed carbon electrodes (SPCE). The high sensitivity was achieved by using secondary antibodies (Ab2) labeled with horseradish peroxidase (HRP) in the presence of hydrogen peroxide (H2O2) as signal amplifiers, and hydroquinone (HQ) was used as an electron mediator. The immunosensor was selective for CA15-3 in human serum and artificial saliva samples, robust, and stable to permit storage at 4 °C for more than 30 days. With its high performance, the immunosensor may be incorporated into future point-of-care (POC) devices to determine CA15-3 in distinct biological fluids, including in blood and saliva samples.


Asunto(s)
Biomarcadores de Tumor/sangre , Técnicas Electroquímicas/métodos , Grafito/química , Inmunoensayo/métodos , Nanopartículas del Metal/química , Mucina-1/sangre , Anticuerpos Inmovilizados/inmunología , Armoracia/enzimología , Biomarcadores de Tumor/inmunología , Oro/química , Peroxidasa de Rábano Silvestre/química , Humanos , Peróxido de Hidrógeno/química , Hidroquinonas/química , Límite de Detección , Mucina-1/inmunología , Reproducibilidad de los Resultados , Saliva/química
10.
Molecules ; 26(19)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34641504

RESUMEN

The role of sialic acids on MUC1 in peritoneal dissemination of ovarian cancer cells was investigated. A human ovarian carcinoma cell line, ES-2, was transfected with full-length MUC1 containing 22 or 42 tandem repeats. These transfectants were less adherent to monolayers of patient-derived mesothelial cells than ES-2/mock transfectants. When these cells were inoculated into the abdominal cavity of female nude mice, mice that had received the transfectants showed better survival. When the transfectants were mixed with sialidase and injected, the survival was poorer, whereas when they were mixed with N-acetyl-2,3-dehydro-2-deoxyneuraminic acid, a sialidase inhibitor, the survival was significantly prolonged. These behaviors, concerned with peritoneal implantation and dissemination observed in vitro and in vivo, were dependent on the expression of MUC1. Therefore, sialic acid linked to MUC1 in the form, at least in part, of sialyl-T, as shown to be recognized by monoclonal antibody MY.1E12, is responsible for the suppression of adhesion of these cells to mesothelial cells and the suppression of peritoneal implantation and dissemination.


Asunto(s)
Mucina-1/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neoplasias Ováricas/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Epítopos/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Mucina-1/genética , Mucina-1/inmunología , Neuraminidasa/metabolismo , Neuraminidasa/farmacología , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/secundario , Peritoneo/citología , Polisacáridos/química , Polisacáridos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int Immunopharmacol ; 101(Pt A): 108173, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34607233

RESUMEN

In previous studies, we have obtained a notable anti-tumor efficacy of the recombinant MUC1-MBP vaccine in the process of mouse B16-MUC1 melanoma treatment. However, the tumor cannot be eliminated completely. We found that the tumor inhibition rate decreased from 81.67% (five immunizations) to 43.67% (eight immunizations) after more than five immunizations, indicating persistent vaccine stimulation may activate immunosuppressive factors. In the present study, we revealed that programmed cell death 1 (PD1), an inhibitory molecule suppressing T cell function, expressed on splenic and tumor-infiltrating T cells were up-regulated by the vaccine. Therefore, to optimize the anti-tumor efficacy of the vaccine, we employed combination immunotherapy with MUC1-MBP vaccine and αPD1 (anti-PD1 antibody). Results showed that combination immunotherapy induced a more remarkable anti-tumor efficacy, the tumor clearance being increased to 80% from 20% which obtain by MUC1-MBP vaccine immunizations. To investigate the possible underlying mechanism, IFN-γ secretion and cytotoxic T lymphocyte (CTL) cytotoxicity were measured by enzyme-linked immunosorbent assay (ELISA) and xCELLigence real-time cell analyzer (RTCA) respectively. T cell subsets and immunosuppressive cells in the mouse spleen and tumor microenvironment were analyzed by FACS. Results showed that the proportion of splenic CD8+T cells and tumor infiltration was increased and the activity of CTL killing, T helper 1 (Th1), Type 1 CD8+T (Tc1) was enhanced, indicating that the anti-tumor efficacy enhanced by combination immunotherapy was mainly through boosting CD8+T cells mediated anti-tumor cellular immunity. Additionally, combination immunotherapy significantly decreased the splenic and tumor-infiltrating myeloid derived suppressor cells (MDSCs). These results demonstrated that combination immunotherapy with MUC1-MBP vaccine and αPD1 was capable to invoke a more potent anti-tumor immune response and provide a foundation for further research.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/farmacología , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral/trasplante , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Mucina-1/administración & dosificación , Mucina-1/genética , Mucina-1/inmunología , Proteína Básica de Mielina/administración & dosificación , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/inmunología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
12.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1625-1639, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34586349

RESUMEN

Mucin 1 (MUC1) has been regarded as an ideal target for cancer treatment, since it is overexpressed in a variety of different cancers including the majority of breast cancer. However, there are still no approved monoclonal antibody drugs targeting MUC1. In this study, we generated a humanized MUC1 (HzMUC1) antibody from our previously developed MUC1 mouse monoclonal antibody that only recognizes MUC1 on the surface of tumor cells. Furthermore, an antibody-drug conjugate (ADC) was generated by conjugating HzMUC1 with monomethyl auristatin (MMAE), and the efficacy of HzMUC1-MMAE on the MUC1-positive HER2+ breast cancer in vitro and in 'Xenograft' model was tested. Results from western blot analysis and immunoprecipitation revealed that the HzMUC1 antibody did not recognize cell-free MUC1-N in sera from breast cancer patients. Confocal microscopy analysis showed that HzMUC1 antibody bound to MUC1 on the surface of breast cancer cells. Results from mapping experiments suggested that HzMUC1 may recognize an epitope present in the interaction region between MUC1-N and MUC1-C. Results from colony formation assay and flow cytometry demonstrated that HzMUC1-MMAE significantly inhibited cell growth by inducing G2/M cell cycle arrest and apoptosis in trastuzumab-resistant HER2-positive breast cancer cells. Meanwhile, HzMUC1-MMAE significantly reduced the growth of HCC1954 xenograft tumors by inhibiting cell proliferation and enhancing cell death. In conclusion, our results indicate that HzMUC1-ADC is a novel therapeutic drug that can overcome trastuzumab resistance of breast cancer. HzMUC1-ADC should also be an effective therapeutic drug for the treatment of different MUC1-positive cancers in clinic.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Inmunoconjugados/farmacología , Mucina-1/metabolismo , Trastuzumab/farmacología , Animales , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Epítopos , Humanos , Inmunoconjugados/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mucina-1/sangre , Mucina-1/química , Mucina-1/inmunología , Oligopéptidos/química , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Med Sci ; 18(15): 3380-3388, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34522164

RESUMEN

Lung cancer remains a global challenge due to high morbidity and mortality rates and poor response to treatment, and there are still no effective strategies to solve it. The bispecific antibody (BsAb) is a novel antibody, which can target two different antigens and mediate specific killing effects by selectively redirecting effector cells to the target cells. In this study, we combined two BsAbs to achieve a dual-target therapy strategy of EpCAM+ and MUC-1+ with high affinity and specificity. The results showed that the combination of two BsAbs against EpCAM and MUC-1 could inhibit the growth of lung cancer more effectively in cell lines and primary tumors. The superior antitumor effect of two BsAbs could be attributable to enhanced CTL and increased production of type I IFNs. At the same time, the combination of EpCAM/CD3 BsAb and MUC-1/CD3 BsAb significantly regulated T population in the TDLNs. Therefore, we have found a potential immunotherapeutic strategy, which was the combination therapy with EpCAM/CD3 BsAb and MUC-1/CD3 BsAb for the treatment of non-small cell lung cancer.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Complejo CD3/inmunología , Molécula de Adhesión Celular Epitelial/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Mucina-1/inmunología , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/inmunología , Linfocitos T/inmunología
14.
Sci Rep ; 11(1): 13065, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158526

RESUMEN

Recently, the siderophores have opened new horizons in nanomedicine. The current study aimed to design a theranostic platform based on superparamagnetic iron oxide nanoparticles-pyoverdine (SPION/PVD) conjugates bound to MUC1 aptamer (MUC1Apt) and loaded with doxorubicin (DOX) as an anti-cancer agent. The SPION/PVD complex was covalently conjugated to MUC1Apt and loaded with DOX to prepare a targeted drug delivery system (SPION/PVD/MUC1Apt/DOX). The investigation of cellular cytotoxicity and uptake of formulations by MTT and flow cytometry in both MUC1 positive (C26) and MUC1 negative (CHO) cell lines revealed that MUC1Apt could improve both cellular uptake and toxicity in the C26 cell line. The evaluation of tumor-targeting activity by in vivo bio-distribution showed that the targeted formulation could enhance tumor inhibitory growth effect and survival rate in C26 tumor-bearing mice. Furthermore, the potential of synthesized SPION/PVD/MUC1Apt/DOX complex as diagnostic agents was investigated by magnetic resonance imaging (MRI) which improved the contrast of tumor site in MRI. Our findings confirm that aptamer-targeted PVD chelated the SPION as a diagnostic agent and loaded with DOX as a chemotherapeutic drug, would be beneficial as a novel theranostic platform.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Nanopartículas de Magnetita/uso terapéutico , Sideróforos/uso terapéutico , Animales , Aptámeros de Nucleótidos/uso terapéutico , Carcinoma/diagnóstico por imagen , Carcinoma/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias del Colon/diagnóstico por imagen , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mucina-1/inmunología , Sideróforos/química , Nanomedicina Teranóstica/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Eur J Pharmacol ; 902: 174119, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-33930385

RESUMEN

MUC1 seems to be promising target in cancer cells due to its abundant and specifically altered expression as well as differential distribution pattern relative to normal tissues. Rosmarinic acid (RA) is a natural, polyphenolic compound with pharmacological activities, including anti-cancer. Herein, we aim to explore the effect of combined action of anti-MUC1 and RA on selected cancer-related factors in AGS gastric cancer cells. Cancer cells were treated with 100, 200 µM rosmarinic acid, 5 µg/ml anti-MUC1 and acid together with antibody. Western blotting, ELISA and RT-PCR were used to assess the expression of MUC1, selected sugar antigens, enzymes participating in protein glycosylation, Gal-3, p53, pro- and anti-apoptotic factors, and caspases-3,-8,-9 in cancer cells. MUC1 mRNA was significantly suppressed by combined action of anti-MUC1 and RA. Such treatment markedly inhibited expression of cancer-related Tn, T, sialyl Tn, sialyl T, and fucosylated sugar antigens as well as mRNA expression of enzymes participating in their formation: ppGalNAcT2, C1GalT1, ST6GalNAcT2, ST3GalT1 and FUT4. C1GalT1 was suppressed also on protein level. Gal-3, factor likely participating in metastasis, was significantly suppressed on mRNA level by RA administrated with anti-MUC1. Pro-apoptotic Bax protein and Bad mRNA were significantly induced, and anti-apoptotic Bcl-2 mRNA expression was inhibited by such treatment. Combined action of mAb and RA markedly increased caspase-9 mRNA expression. Results of the study indicate that combined action of anti-MUC1 and RA is more effective than monotherapy in relation to examined cancer related factors. Such treatment can be considered as new, promising strategy in gastric cancer therapy.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Antineoplásicos/farmacología , Cinamatos/farmacología , Depsidos/farmacología , Mucina-1/inmunología , Mucina-1/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada , Galectinas/genética , Galectinas/metabolismo , Glicosilación/efectos de los fármacos , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Humanos , Mucina-1/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Ácido Rosmarínico
16.
Ther Apher Dial ; 25(4): 415-424, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33886156

RESUMEN

The prognosis of patients with advanced pancreatic cancer is poor. In the present phase I/II study, we have evaluated the safety and the feasibility of Wilms' tumor 1 (WT1) and/or mucin1 (MUC1) peptide-pulsed dendritic cell (DC) vaccination in combination with chemotherapy in patients with advanced or relapsed pancreatic ductal adenocarcinoma (PDAC). Sixty-five eligible patients were enrolled. No severe adverse events related to the vaccinations were observed. Objective response rate and disease control rate was 12.3% and 50.8%, respectively. Median progression-free survival and overall survival were 4.9 and 9.6 months, respectively. DC vaccinations augmented WT1- and MUC1-specific immunity which might be related to clinical outcome. These results indicate that DC-based immunotherapy combined with a conventional chemotherapy is safe and feasible for patients in advanced stage of PDAC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/terapia , Células Dendríticas/inmunología , Inmunoterapia/métodos , Mucina-1/inmunología , Neoplasias Pancreáticas/terapia , Vacunación/métodos , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/inmunología , Terapia Combinada , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Pronóstico , Tasa de Supervivencia
17.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33762322

RESUMEN

BACKGROUND: Antitumor vaccines targeting tumor-associated antigens (TAAs) can generate antitumor immune response. A novel vaccine platform using adenovirus 5 (Ad5) vectors [E1-, E2b-] targeting three TAAs-prostate-specific antigen (PSA), brachyury, and MUC-1-has been developed. Both brachyury and the C-terminus of MUC-1 are overexpressed in metastatic castration-resistant prostate cancer (mCRPC) and have been shown to play an important role in resistance to chemotherapy, epithelial-mesenchymal transition, and metastasis. The transgenes for PSA, brachyury, and MUC-1 all contain epitope modifications for the expression of CD8+ T-cell enhancer agonist epitopes. We report here the first-in-human trial of this vaccine platform. METHODS: Patients with mCRPC were given concurrently three vaccines targeting PSA, brachyury, and MUC-1 at 5×1011 viral particles (VP) each, subcutaneously every 3 weeks for a maximum of three doses (dose de-escalation cohort), followed by a booster vaccine every 8 weeks for 1 year (dose-expansion cohort only). The primary objective was to determine the safety and the recommended phase II dose. Immune assays and clinical responses were evaluated. RESULTS: Eighteen patients with mCRPC were enrolled between July 2018 and September 2019 and received at least one vaccination. Median PSA was 25.58 ng/mL (range, 0.65-1006 ng/mL). The vaccine was tolerable and safe, and no grade >3 treatment-related adverse events or dose-limiting toxicities (DLTs) were observed. One patient had a partial response, while five patients had confirmed PSA decline and five had stable disease for >6 months. Median progression-free survival was 22 weeks (95% CI: 19.1 to 34). Seventeen (100%) of 17 patients mounted T-cell responses to at least one TAA, whereras 8 (47%) of 17 patients mounted immune responses to all three TAAs. Multifunctional T-cell responses to PSA, MUC-1, and brachyury were also detected after vaccination in the majority of the patients. CONCLUSIONS: Ad5 PSA/MUC-1/brachyury vaccine is well tolerated. The primary end points were met and there were no DLTs. The recommended phase II dose is 5×1011 VP. The vaccine demonstrated clinical activity, including one partial response and confirmed PSA responses in five patients. Three patients with prolonged PSA responses received palliative radiation therapy. Further research is needed to evaluate the clinical benefit and immunogenicity of this vaccine in combination with other immuno-oncology agents and/or palliative radiation therapy. TRIAL REGISTRATION NUMBER: NCT03481816.


Asunto(s)
Adenoviridae/inmunología , Vacunas contra el Cáncer/uso terapéutico , Proteínas Fetales/inmunología , Calicreínas/inmunología , Mucina-1/inmunología , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Proteínas de Dominio T Box/inmunología , Vacunas Combinadas/uso terapéutico , Adenoviridae/genética , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Proteínas Fetales/genética , Vectores Genéticos , Humanos , Calicreínas/genética , Masculino , Persona de Mediana Edad , Mucina-1/genética , Supervivencia sin Progresión , Antígeno Prostático Específico/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Proteínas de Dominio T Box/genética , Factores de Tiempo , Vacunación , Eficacia de las Vacunas , Vacunas Combinadas/efectos adversos , Vacunas Combinadas/genética , Vacunas Combinadas/inmunología , Vacunas Virales
18.
Org Biomol Chem ; 19(11): 2448-2455, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33645601

RESUMEN

MUC1 glycopeptides are attractive antigens for anti-cancer vaccine development. One potential drawback in using the native MUC1 glycopeptide for vaccine design is the instability of the O-glycosyl linkage between the glycan and the peptide backbone to glycosidase. To overcome this challenge, a MUC1 glycopeptide mimic has been synthesized with the galactose-galactosamine disaccharide linked with threonine (Thomsen-Friedenreich or Tf antigen) through an unnatural ß-glycosyl bond. The resulting MUC1-ß-Tf had a much-enhanced stability toward a glycosidase capable of cleaving the glycan from the corresponding MUC1 glycopeptide with the natural α-Tf linkage. The MUC1-ß-Tf was subsequently conjugated with a powerful carrier bacteriophage Qß. The conjugate induced high levels of IgG antibodies in clinically relevant human MUC1 transgenic mice, which cross-recognized not only the natural MUC1-α-Tf glycopeptide but also MUC1 expressing tumor cells, supporting the notion that a simple switch of the stereochemistry of the glycan/peptide linkage can be a strategy for anti-cancer vaccine epitope design for glycopeptides.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/química , Vacunas contra el Cáncer/química , Glicopéptidos/química , Mucina-1/química , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Disacáridos/química , Diseño de Fármacos , Galactosamina/química , Galactosa/química , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/farmacología , Ratones , Ratones Transgénicos , Mucina-1/inmunología
19.
Sci Rep ; 11(1): 6276, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737613

RESUMEN

Current treatments for cholangiocarcinoma (CCA) are largely unsuccessful due to late diagnosis at advanced stage, leading to high mortality rate. Consequently, improved therapeutic approaches are urgently needed. Chimeric antigen receptor (CAR) T cell therapy is a newly potential therapy that can recognize specific surface antigen without major histocompatibility complex (MHC) restriction. Mucin 1 (MUC1) is an attractive candidate antigen as it is highly expressed and associated with poor prognosis and survival in CCA. We, therefore, set forth to create the fourth-generation CAR (CAR4) construct containing anti-MUC1-single-chain variable fragment (scFv) and three co-stimulatory domains (CD28, CD137, and CD27) linked to CD3ζ and evaluate anti-MUC1-CAR4 T cells in CCA models. Compared to untransduced T cells, anti-MUC1-CAR4 T cells produced increased levels of TNF-α, IFN-γ and granzyme B when exposed to MUC1-expressing KKU-100 and KKU-213A CCA cells (all p < 0.05). Anti-MUC1-CAR4 T cells demonstrated specific killing activity against KKU-100 (45.88 ± 7.45%, p < 0.05) and KKU-213A cells (66.03 ± 3.14%, p < 0.001) at an effector to target ratio of 5:1, but demonstrated negligible cytolytic activity against immortal cholangiocytes. Furthermore, the anti-MUC1-CAR4 T cells could effectively disrupt KKU-213A spheroids. These activities of anti-MUC1-CAR4 T cells supports the development of this approach as an adoptive T cell therapeutic strategy for CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/terapia , Trasplante de Células/métodos , Colangiocarcinoma/inmunología , Colangiocarcinoma/terapia , Inmunoterapia Adoptiva/métodos , Mucina-1/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Técnicas de Cocultivo , Citocinas/biosíntesis , Células HEK293 , Humanos , Células MCF-7 , Mucina-1/metabolismo , Receptores Quiméricos de Antígenos/genética , Anticuerpos de Cadena Única/inmunología , Esferoides Celulares/inmunología , Transfección , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
20.
Int J Nanomedicine ; 16: 403-420, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33469292

RESUMEN

BACKGROUND: Therapeutic tumor vaccines are one of the most promising strategies and have attracted great attention in cancer treatment. However, most of them have shown unsatisfactory immunogenicity, there are still few available vaccines for clinical use. Therefore, there is an urgent demand to develop novel strategies to improve the immune efficacy of antitumor vaccines. PURPOSE: This study aimed to develop novel adjuvants and carriers to enhance the immune effect of MUC1 glycopeptide antigen-based antitumor vaccines. METHODS: An antitumor vaccine was developed, in which MUC1 glycopeptide was used as tumor-associated antigen, α-GalCer served as an immune adjuvant and AuNPs was a multivalent carrier. RESULTS: Immunological evaluation results indicated that the constructed vaccines enabled a significant antibody response. FACS analysis and immunofluorescence assay showed that the induced antisera exhibited a specific binding with MUC1 positive MCF-7 cells. Moreover, the induced antibody can mediate CDC to kill MCF-7 cells. Besides stimulating B cells to produce MUC1-specific antibodies, the prepared vaccines also induced MUC1-specific CTLs in vitro. Furthermore, the vaccines significantly delayed tumor development in tumor-bearing mice model. CONCLUSION: These results showed that the construction of vaccines by presenting α-GalCer adjuvant and an antigen on gold nanoparticles offers a potential strategy to improve the antitumor response in cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Galactosilceramidas/farmacología , Oro/farmacología , Nanopartículas del Metal/química , Mucina-1/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antineoplásicos/inmunología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Galactosilceramidas/síntesis química , Galactosilceramidas/química , Humanos , Sueros Inmunes/metabolismo , Melanoma/inmunología , Melanoma/patología , Nanopartículas del Metal/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/patología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA