Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biomater Sci ; 12(9): 2292-2301, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38498328

RESUMEN

Colorectal cancer (CRC) ranks among the most prevalent cancers globally, demanding innovative therapeutic strategies. Immunotherapy, a promising avenue, employs cancer vaccines to activate the immune system against tumors. However, conventional approaches fall short of eliciting robust responses within the gastrointestinal (GI) tract, where CRC originates. Harnessing the potential of all-trans retinoic acid (ATRA) and cytosine-phosphorothioate-guanine (CpG), we developed layered nanoparticles using a layer-by-layer assembly method to co-deliver these agents. ATRA, crucial for gut immunity, was efficiently encapsulated alongside CpG within these nanoparticles. Administering these ATRA@CpG-NPs, combined with ovalbumin peptide (OVA), effectively inhibited orthotopic CRC growth in mice. Our approach leveraged the inherent benefits of ATRA and CpG, demonstrating superior efficacy in activating dendritic cells, imprinting T cells with gut-homing receptors, and inhibiting tumor growth. This mucosal adjuvant presents a promising strategy for CRC immunotherapy, showcasing the potential for targeting gut-associated immune responses in combating colorectal malignancies.


Asunto(s)
Neoplasias Colorrectales , Fosfatos de Dinucleósidos , Nanopartículas , Tretinoina , Tretinoina/química , Tretinoina/administración & dosificación , Tretinoina/farmacología , Animales , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/inmunología , Nanopartículas/química , Nanopartículas/administración & dosificación , Ratones , Humanos , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Ratones Endogámicos C57BL , Femenino , Inmunoterapia/métodos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/química , Línea Celular Tumoral , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Nanopartículas Capa por Capa
2.
In Vivo ; 37(1): 286-293, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36593062

RESUMEN

BACKGROUND/AIM: Vitamin D3 (VD3) affects the regulation of the immune system, including the differentiation and function of regulatory T-cells (Tregs). Tregs play an important role in maintaining immune homeostasis in patients with colorectal cancer (CRC). The effects of VD3 on Treg-associated immune function were investigated in Thai patients in the early stages of CRC. MATERIALS AND METHODS: Twenty-eight patients were randomized to one of two groups: Untreated or treatment with VD3 for 3 months. Whole blood samples were collected at baseline, and at 1 and 3 months. Peripheral blood mononuclear cells were isolated and the populations of forkhead box P3-positive Treg cells was analyzed by flow cytometry. The levels of Treg-associated cytokines, interleukin 10 (IL-10) and transforming growth factor beta 1 (TGF-ß1), were measured by enzyme-linked immunosorbent assays. RESULTS: Serum VD3 levels of the VD3-treated group were significantly increased at 1 (p=0.017) and 3 months (p<0.001) compared to the untreated control group. The mean percentage of Tregs was maintained between 1 and 3 months in the VD3-treated group. At 3 months, the untreated group had significantly lower Treg levels than the VD3-treated group (p=0.043). Serum IL-10 levels of the VD3-treated group were statistically increased at 1 month compared to the control group (p=0.032). No significant difference in serum TGF-ß1 levels was observed between the two groups. However, the TGF-ß1 level in the VD3-treated group at 1 month was lower than that of the control. CONCLUSION: Our findings suggest that VD3 supplementation can maintain immune responses in the early stages of CRC, helping to control Treg function. Therefore, VD3 should be supplemented to maintain immune homeostasis, especially in patients with vitamin D deficiency.


Asunto(s)
Colecalciferol , Neoplasias Colorrectales , Linfocitos T Reguladores , Humanos , Colecalciferol/administración & dosificación , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/cirugía , Suplementos Dietéticos , Homeostasis , Interleucina-10/inmunología , Leucocitos Mononucleares/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología
3.
Pharmacol Res ; 176: 106082, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032662

RESUMEN

Patchouli Essential Oil (PEO) has been used as a scent for various healing purposes since the ancient Egyptian period. The primary source of the oil is Pogostemon cablin (PC), a medicinal plant for treating gastrointestinal symptoms. However, the pharmacological function has not been addressed. Here, we report the cancer prevention and gut microbiota (GM) modulating property of PEO and its derivatives patchouli alcohol (PA) and pogostone (PO) in the ApcMin /+ colorectal cancer mice model. We found that PEO, PA, and PO significantly reduced the tumor burden. At the same time, it strengthened the epithelial barrier, evidenced by substantially increasing the number of the goblet and Paneth cells and upregulation of tight junction and adhesion molecules. In addition, PEO, PA, and PO shifted M1 to M2 macrophage phenotypes and remodeled the inflammatory milieu of ApcMin /+ mice. We also found suppression of CD4+CD25+ and stimulation CD4+ CD8+ cells in the spleen, blood, mesenteric lymph nodes (MLNs), and Peyer's patches (PPs) of the treated mice. The composition of the gut microbiome of the drug-treated mice was distinct from the control mice. The drugs stimulated the short-chain fatty acids (SCFAs)-producers and the key SCFA-sensing receptors (GPR41, GPR43, and GPR109a). The activation of SCFAs/GPSs also triggered the alterations of PPAR-γ, PYY, and HSDCs signaling mediators in the treated mice. Our work showed that PEO and its derivatives exert potent anti-cancer effects by modulating gut microbiota and improving the intestinal microenvironment of the ApcMmin /+ mice.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Aceites Volátiles/uso terapéutico , Pogostemon , Animales , Antineoplásicos Fitogénicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Aceites Volátiles/farmacología , Ganglios Linfáticos Agregados/efectos de los fármacos , Bazo/efectos de los fármacos
4.
Comput Math Methods Med ; 2021: 1619809, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34950221

RESUMEN

OBJECTIVE: To explore the effects of Xiaoyutang combined with intraperitoneal heat perfusion chemotherapy on immune function, circulating Mir, and prognosis and survival of postoperative patients with colorectal cancer. METHODS: A total of 96 patients with colorectal cancer who were treated in our hospital from May 2018 to August 2019 and followed up to August 2021 were selected as the study subjects. The patients were randomly divided into a control group and study group by a 1 : 1 random number table method, 48 cases in each group. Patients in the control group were given intraperitoneal thermal perfusion chemotherapy after surgery, and patients in the research group were treated with Xiaoyutang on this basis. The treatment cycle was 21 days, and all patients were treated for 3 consecutive cycles. The therapeutic efficacy, immune function (CD3+, CD4+, and CD4+/CD8+), circulating mir (mir-29a, mir-145, and mir-92a), prognosis, and survival of the two groups were compared. RESULTS: After 3 cycles of treatment, ORR and DCR in the study group were higher than those in the control group (60.42% vs. 37.50%) and 85.42% vs. 66.67%, respectively, with statistical significance (P < 0.05). There were statistically significant differences in CD3+, CD4+, CD4+/CD8+, mir-29a, mir-145, and mir-92a time points and intergroup and intergroup interactions between the two groups (P < 0.05); the levels of CD3+, CD4+, and CD4+/CD8+ in the study group were higher than those in the control group after 1, 2, and 3 cycles of treatment (P < 0.05); the expressions of mir-29a, mir-145, and mir-92a were significantly lower than those in the control group (P < 0.05). By the end of follow-up, 3 cases were lost to follow-up in the study group and 5 cases in the control group. The recurrence rate and mortality of the study group were lower than those of the control group at 1- and 2-year follow-up (P > 0.05), and the mean survival time of patients in the study group was higher than that in the control group; the differences were statistically significant (χ 2 = 5.151, P = 0.023). CONCLUSION: Xiaoyutang combined with peritoneal heat perfusion chemotherapy has a good postoperative effect on patients with colorectal cancer, which can effectively improve the immune function and circulating Mir of patients with colorectal cancer, reduce tumor recurrence, and improve the prognosis of patients.


Asunto(s)
Neoplasias Colorrectales/terapia , Medicamentos Herbarios Chinos/administración & dosificación , Quimioterapia Intraperitoneal Hipertérmica , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , China/epidemiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Terapia Combinada , Biología Computacional , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/sangre , MicroARNs/genética , Persona de Mediana Edad , Periodo Posoperatorio , Pronóstico
5.
Bioengineered ; 12(2): 12461-12469, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34931923

RESUMEN

Severe mortality due to the COVID-19 pandemic resulted from the lack of effective treatment. Although COVID-19 vaccines are available, their side effects have become a challenge for clinical use in patients with chronic diseases, especially cancer patients. In the current report, we applied network pharmacology and systematic bioinformatics to explore the use of biochanin A in patients with colorectal cancer (CRC) and COVID-19 infection. Using the network pharmacology approach, we identified two clusters of genes involved in immune response (IL1A, IL2, and IL6R) and cell proliferation (CCND1, PPARG, and EGFR) mediated by biochanin A in CRC/COVID-19 condition. The functional analysis of these two gene clusters further illustrated the effects of biochanin A on interleukin-6 production and cytokine-cytokine receptor interaction in CRC/COVID-19 pathology. In addition, pathway analysis demonstrated the control of PI3K-Akt and JAK-STAT signaling pathways by biochanin A in the treatment of CRC/COVID-19. The findings of this study provide a therapeutic option for combination therapy against COVID-19 infection in CRC patients.


Asunto(s)
Anticarcinógenos/uso terapéutico , Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Neoplasias Colorrectales/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genisteína/uso terapéutico , Fitoestrógenos/uso terapéutico , Atlas como Asunto , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/virología , Ciclina D1/genética , Ciclina D1/inmunología , Receptores ErbB/genética , Receptores ErbB/inmunología , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Quinasas Janus/genética , Quinasas Janus/inmunología , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Terapia Molecular Dirigida/métodos , Familia de Multigenes , Farmacología en Red/métodos , PPAR gamma/genética , PPAR gamma/inmunología , Farmacogenética/métodos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Transducción de Señal
6.
Front Immunol ; 12: 737076, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659228

RESUMEN

Immune checkpoints such as programmed death-1 (PD-1) have been proven as antitumor targets by enhancing cytotoxic T cell activity. All immune checkpoint blockades are antibody therapeutics that have large size and high affinity, as well as known immune-related side effects and low responses. To overcome the limitation of antibody therapeutics, we have explored PD-1/PD-L1 (programmed death-ligand 1) blockades in traditional oriental medicine, which has a long history but has not yet studied PD-1/PD-L1 blockades. Sanguisorbae Radix extract (SRE) blocked PD-1 and PD-L1 binding in competitive ELISA. SRE effectively inhibited the PD-1/PD-L1 interaction, thereby improving T cell receptor (TCR) signaling and the NFAT-mediated luciferase activity of T cells. SRE treatment reduced tumor growth in the humanized PD-L1 MC38 cell allograft humanized PD-1 mouse model. Additionally, the combination of SRE and pembrolizumab (anti-PD-1 antibody) suppressed tumor growth and increased infiltrated cytotoxic T cells to a greater extent did either agent alone. This study showed that SRE alone has anticancer effects via PD-1/PD-L1 blockade and that the combination therapy of SRE and pembrolizumab has enhanced immuno-oncologic effects.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Extractos Vegetales/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Sanguisorba , Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células CHO , Técnicas de Cocultivo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cricetulus , Humanos , Células Jurkat , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Extractos Vegetales/aislamiento & purificación , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Sanguisorba/química , Transducción de Señal , Carga Tumoral
7.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360782

RESUMEN

Colorectal carcinogenesis is the second most common cause of mortality across all types of malignancies, followed by hepatic and stomach cancers. Chemotherapy and radiotherapy are key approaches to treating cancer patients, but these carry major concerns, such as a high risk of side effects, poor accessibility, and the non-selective nature of chemotherapeutics. A number of natural products have been identified as countering various forms of cancer with fewer side effects. The potential impact of vitamins and minerals on long-term health, cognition, healthy development, bone formation, and aging has been supported by experimental and epidemiological studies. Successful treatment may thus be highly influenced by the nutritional status of patients. An insufficient diet could lead to detrimental effects on immune status and tolerance to treatment, affecting the ability of chemotherapy to destroy cancerous cells. In recent decades, most cancer patients have been taking vitamins and minerals to improve standard therapy and/or to decrease the undesirable side effects of the treatment together with the underlying disease. On the other hand, taking dietary supplements during cancer therapy may affect the effectiveness of chemotherapy. Thus, micronutrients in complementary oncology must be selected appropriately and should be taken at the right time. Here, the potential impact of micronutrients on gastro-intestinal and hepatic cancers is explored and their molecular targets are laid down.


Asunto(s)
Neoplasias Colorrectales , Suplementos Dietéticos , Neoplasias Hepáticas , Micronutrientes , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/inmunología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Micronutrientes/inmunología , Micronutrientes/uso terapéutico
8.
Cell Host Microbe ; 29(10): 1573-1588.e7, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34453895

RESUMEN

Despite overall success, T cell checkpoint inhibitors for cancer treatment are still only efficient in a minority of patients. Recently, intestinal microbiota was found to critically modulate anti-cancer immunity and therapy response. Here, we identify Clostridiales members of the gut microbiota associated with a lower tumor burden in mouse models of colorectal cancer (CRC). Interestingly, these commensal species are also significantly reduced in CRC patients compared with healthy controls. Oral application of a mix of four Clostridiales strains (CC4) in mice prevented and even successfully treated CRC as stand-alone therapy. This effect depended on intratumoral infiltration and activation of CD8+ T cells. Single application of Roseburia intestinalis or Anaerostipes caccae was even more effective than CC4. In a direct comparison, the CC4 mix supplementation outperformed anti-PD-1 therapy in mouse models of CRC and melanoma. Our findings provide a strong preclinical foundation for exploring gut bacteria as novel stand-alone therapy against solid tumors.


Asunto(s)
Terapia Biológica , Clostridiales/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Microbioma Gastrointestinal , Animales , Linfocitos T CD8-positivos/inmunología , Clostridiales/fisiología , Neoplasias Colorrectales/microbiología , Humanos , Inmunidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Simbiosis
9.
Front Immunol ; 12: 635899, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777027

RESUMEN

Iron deficiency, with or without anemia, is the most frequent hematological manifestation in individuals with cancer, and is especially common in patients with colorectal cancer. Iron is a vital micronutrient that plays an essential role in many biological functions, in the context of which it has been found to be intimately linked to cancer biology. To date, however, whereas a large number of studies have comprehensively investigated and reviewed the effects of excess iron on cancer initiation and progression, potential interrelations of iron deficiency with cancer have been largely neglected and are not well-defined. Emerging evidence indicates that reduced iron intake and low systemic iron levels are associated with the pathogenesis of colorectal cancer, suggesting that optimal iron intake must be carefully balanced to avoid both iron deficiency and iron excess. Since iron is vital in the maintenance of immunological functions, insufficient iron availability may enhance oncogenicity by impairing immunosurveillance for neoplastic changes and potentially altering the tumor immune microenvironment. Data from clinical studies support these concepts, showing that iron deficiency is associated with inferior outcomes and reduced response to therapy in patients with colorectal cancer. Here, we elucidate cancer-related effects of iron deficiency, examine preclinical and clinical evidence of its role in tumorigenesis, cancer progression and treatment response. and highlight the importance of adequate iron supplementation to limit these outcomes.


Asunto(s)
Anemia Ferropénica/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Hierro/metabolismo , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/inmunología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/prevención & control , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Hierro/inmunología , Compuestos de Hierro/uso terapéutico , Estrés Oxidativo , Escape del Tumor , Microambiente Tumoral
10.
Cancer Treat Rev ; 95: 102174, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33721596

RESUMEN

In 5% of metastatic colorectal cancer (mCRC) patients, tumours display a deficient mismatch repair (dMMR) system. Immunotherapy is beneficial in dMMR mCRC patients and has recently been approved by the Food and Drug Administration for patients with unresectable or metastatic dMMR CRC. Although dMMR and proficient MMR (pMMR) CRC tumours are biologically distinct, they are commonly treated with the same chemotherapy and monoclonal antibodies. This includes dMMR mCRC patients who did not respond to immunotherapy (20-30%). However, it is unclear if these treatments are equally beneficial in dMMR mCRC. Of note, dMMR mCRC patients have a worse prognosis compared to pMMR, which may in part be caused by a lower response to treatment. To avoid unnecessary exposure to ineffective treatments and their associated toxicity, it is important to identify which systemic treatments are most beneficial in dMMR mCRC patients, thus improving their outcome. Indeed, future treatment strategies are likely to involve combinations of immunotherapy, chemotherapy and monoclonal antibodies. In this evidence-based review, we summarize clinical trials reporting treatment efficacy of different types of chemotherapy and monoclonal antibodies in dMMR mCRC patients. We also review the biological rationale behind a potential differential benefit of chemotherapy with or without monoclonal antibodies in dMMR mCRC patients. A barrier in the interpretation of preclinical results is the choice of model systems. They largely comprise traditional models, including cell lines and xenografts, rather than more representative models, such as patient-derived organoids. We provide concrete recommendations for clinical investigators and fundamental researchers to accelerate research regarding which systemic therapy is most effective in dMMR mCRC patients.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Reparación de la Incompatibilidad de ADN/genética , Animales , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/secundario , Evaluación Preclínica de Medicamentos , Humanos , Inmunoterapia
11.
Cancer Res ; 81(9): 2495-2509, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33509940

RESUMEN

Tyrosine phosphorylation (pTyr) plays a pivotal role in signal transduction and is commonly dysregulated in cancer. As a result, profiling tumor pTyr levels may reveal therapeutic insights critical to combating disease. Existing discovery and targeted mass spectrometry-based methods used to monitor pTyr networks involve a tradeoff between broad coverage of the pTyr network, reproducibility in target identification across analyses, and accurate quantification. To address these limitations, we developed a targeted approach, termed "SureQuant pTyr," coupling low input pTyr enrichment with a panel of isotopically labeled internal standard peptides to guide data acquisition of low-abundance tyrosine phosphopeptides. SureQuant pTyr allowed for reliable quantification of several hundred commonly dysregulated pTyr targets with high quantitative accuracy, improving the robustness and usability of targeted mass spectrometry assays. We established the clinical applicability of SureQuant pTyr by profiling pTyr signaling levels in human colorectal tumors using minimal sample input, characterizing patient-specific oncogenic-driving mechanisms. While in some cases pTyr profiles aligned with previously reported proteomic, genomic, and transcriptomic molecular characterizations, we highlighted instances of new insights gained using pTyr characterization and emphasized the complementary nature of pTyr measurements with traditional biomarkers for improving patient stratification and identifying therapeutic targets. The turn-key nature of this approach opens the door to rapid and reproducible pTyr profiling in research and clinical settings alike and enables pTyr-based measurements for applications in precision medicine. SIGNIFICANCE: SureQuant pTyr is a mass spectrometry-based targeted method that enables sensitive and selective targeted quantitation of several hundred low-abundance tyrosine phosphorylated peptides commonly dysregulated in cancer, including oncogenic signaling networks.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/análisis , Transducción de Señal , Tirosina/metabolismo , Células A549 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Cromatografía Liquida/métodos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Receptores ErbB/metabolismo , Humanos , Espectrometría de Masas/métodos , Fosfopéptidos/análisis , Fosfopéptidos/metabolismo , Fosforilación , Mapas de Interacción de Proteínas , Proteoma/metabolismo , Proteómica/métodos
12.
Front Immunol ; 12: 791565, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069573

RESUMEN

Intestinal diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancer (CRC) generally characterized by clinical symptoms, including malabsorption, intestinal dysfunction, injury, and microbiome imbalance, as well as certain secondary intestinal disease complications, continue to be serious public health problems worldwide. The role of vitamin K (VK) on intestinal health has drawn growing interest in recent years. In addition to its role in blood coagulation and bone health, several investigations continue to explore the role of VK as an emerging novel biological compound with the potential function of improving intestinal health. This study aims to present a thorough review on the bacterial sources, intestinal absorption, uptake of VK, and VK deficiency in patients with intestinal diseases, with emphasis on the effect of VK supplementation on immunity, anti-inflammation, intestinal microbes and its metabolites, antioxidation, and coagulation, and promoting epithelial development. Besides, VK-dependent proteins (VKDPs) are another crucial mechanism for VK to exert a gastroprotection role for their functions of anti-inflammation, immunomodulation, and anti-tumorigenesis. In summary, published studies preliminarily show that VK presents a beneficial effect on intestinal health and may be used as a therapeutic drug to prevent/treat intestinal diseases, but the specific mechanism of VK in intestinal health has yet to be elucidated.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino , Mucosa Intestinal , Deficiencia de Vitamina K , Vitamina K , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/terapia , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/terapia , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Vitamina K/inmunología , Vitamina K/uso terapéutico , Deficiencia de Vitamina K/inmunología , Deficiencia de Vitamina K/microbiología
13.
Cancer Prev Res (Phila) ; 14(1): 65-76, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32917645

RESUMEN

Increased COX-2 and decreased 15-hydroxyprostaglandin dehydrogenase (15-HPGD) expression promote prostaglandin-mediated inflammation and colorectal carcinogenesis. Experimental studies suggest that vitamin D and calcium may inhibit these pathways, but their effects on colorectal tissue COX-2 and 15-HPGD expression in humans are unknown. We tested the effects of supplemental vitamin D (1,000 IU/day) and/or calcium (1,200 mg/day) on COX-2 and 15-HPGD expression in the morphologically normal rectal mucosa from 62 paients with colorectal adenoma in a placebo-controlled chemoprevention trial. We measured biomarker expression using automated IHC and quantitative image analysis at baseline and 1-year follow-up, and assessed treatment effects using mixed linear models. The primary outcome was the COX-2/15-HPGD expression ratio, because these enzymes function as physiologic antagonists. After 1 year of treatment, the mean COX-2/15-HPGD expression ratio in full-length crypts proportionately decreased 47% in the vitamin D group (P = 0.001), 46% in the calcium group (P = 0.002), and 34% in the calcium + vitamin D group (P = 0.03), relative to the placebo group. Among individuals with the functional vitamin D-binding protein isoform DBP2 (GC rs4588*A), the COX-2/15-HPDG ratio decreased 70% (P = 0.0006), 75% (P = 0.0002), and 60% (P = 0.006) in the vitamin D, calcium, and combined supplementation groups, respectively, relative to placebo. These results show that vitamin D and calcium favorably modulate the balance of expression of COX-2 and 15-HPGD-biomarkers of inflammation that are strongly linked to colorectal carcinogenesis-in the normal-appearing colorectal mucosa of patients with colorectal adenoma (perhaps especially those with the DBP2 isoform). PREVENTION RELEVANCE: Supplemental calcium and vitamin D reduce indicators of cancer-promoting inflammation in normal colorectal tissue in humans, thus furthering our understanding of how they may help prevent colorectal cancer.


Asunto(s)
Adenoma/prevención & control , Carbonato de Calcio/administración & dosificación , Neoplasias Colorrectales/prevención & control , Mucosa Intestinal/inmunología , Vitamina D/administración & dosificación , Adenoma/inmunología , Adenoma/patología , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Colon/efectos de los fármacos , Colon/enzimología , Colon/inmunología , Colon/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/análisis , Ciclooxigenasa 2/metabolismo , Suplementos Dietéticos , Femenino , Estudios de Seguimiento , Humanos , Hidroxiprostaglandina Deshidrogenasas/análisis , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Inflamación/diagnóstico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Recto/efectos de los fármacos , Recto/enzimología , Recto/inmunología , Recto/patología , Resultado del Tratamiento
14.
Clin Cancer Res ; 27(1): 288-300, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33028592

RESUMEN

PURPOSE: The DNA damage immune response (DDIR) assay was developed in breast cancer based on biology associated with deficiencies in homologous recombination and Fanconi anemia pathways. A positive DDIR call identifies patients likely to respond to platinum-based chemotherapies in breast and esophageal cancers. In colorectal cancer, there is currently no biomarker to predict response to oxaliplatin. We tested the ability of the DDIR assay to predict response to oxaliplatin-based chemotherapy in colorectal cancer and characterized the biology in DDIR-positive colorectal cancer. EXPERIMENTAL DESIGN: Samples and clinical data were assessed according to DDIR status from patients who received either 5-fluorouracil (5-FU) or 5FUFA (bolus and infusion 5-FU with folinic acid) plus oxaliplatin (FOLFOX) within the FOCUS trial (n = 361, stage IV), or neoadjuvant FOLFOX in the FOxTROT trial (n = 97, stage II/III). Whole transcriptome, mutation, and IHC data of these samples were used to interrogate the biology of DDIR in colorectal cancer. RESULTS: Contrary to our hypothesis, DDIR-negative patients displayed a trend toward improved outcome for oxaliplatin-based chemotherapy compared with DDIR-positive patients. DDIR positivity was associated with microsatellite instability (MSI) and colorectal molecular subtype 1. Refinement of the DDIR signature, based on overlapping IFN-related chemokine signaling associated with DDIR positivity across colorectal cancer and breast cancer cohorts, further confirmed that the DDIR assay did not have predictive value for oxaliplatin-based chemotherapy in colorectal cancer. CONCLUSIONS: DDIR positivity does not predict improved response following oxaliplatin treatment in colorectal cancer. However, data presented here suggest the potential of the DDIR assay in identifying immune-rich tumors that may benefit from immune checkpoint blockade, beyond current use of MSI status.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bioensayo/métodos , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/terapia , Daño del ADN/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Daño del ADN/efectos de los fármacos , Análisis Mutacional de ADN , Femenino , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Leucovorina/farmacología , Leucovorina/uso terapéutico , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Mutación , Terapia Neoadyuvante/métodos , Compuestos Organoplatinos/farmacología , Compuestos Organoplatinos/uso terapéutico , Supervivencia sin Progresión , Ensayos Clínicos Controlados Aleatorios como Asunto
15.
Theranostics ; 10(26): 11998-12010, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33204325

RESUMEN

Rationale: Vascular abnormality stemming from the hypoxia-driven elevation of proangiogenic factors is a hallmark for many solid malignant tumors, including colorectal cancer (CRC) and its liver metastasis. We report a hypoxia-triggered liposome-supported metal-polyphenol-gene bio-nanoreactor to tune the proangiogenic factor-mediated immunotolerance and synergize the elicited tumoricidal immunity for CRC treatment. Methods: With the aid of polyphenol gallic acid, Cu2+ ion-based intracellular bio-nanoreactor was synthesized for the delivery of small interfering RNA targeting vascular endothelial growth factor and then cloaked with a hybrid liposomal membrane that harbored a hypoxia-responsive azobenzene derivative. In hypoxic tumor, the liposomal shell disintegrated, and a shrunk-size bio-nanoreactor was burst released. Intracellularly, Cu2+ from the bio-nanoreactor catalyzed a Fenton-like reaction with glutathione, which efficiently converted H2O2 to •OH and enabled a chemodynamic therapy (CDT) in tumor sites. With the alleviation of proangiogenic factor-mediated immunotolerance and high production of CDT-induced tumor-associated antigens, robust tumoricidal immunity was co-stimulated. Results: With colorectal tumor and its liver metastasis models, we determined the underlying mechanism of proangiogenic factor-mediated immunotolerance and highlighted that the liposomal bio-nanoreactor could create positive feedback among the critical players in the vascular endothelium and synergize the elicited tumoricidal immunity. Conclusion: Our work provides an alternative strategy for exerting efficient tumoricidal immunity in the proangiogenic factor-upregulated subpopulation of CRC patients and may have a wide-ranging impact on cancer immune-anti-angiogenic complementary therapy in clinics.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas del Metal/administración & dosificación , Hipoxia Tumoral/efectos de los fármacos , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Cobre/química , Sinergismo Farmacológico , Femenino , Ácido Gálico , Humanos , Liposomas , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Nanopartículas del Metal/química , Ratones , Polifenoles/química , ARN Interferente Pequeño/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Nanomedicina Teranóstica/métodos , Escape del Tumor/efectos de los fármacos , Hipoxia Tumoral/genética , Hipoxia Tumoral/inmunología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Int J Mol Sci ; 21(15)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32751239

RESUMEN

Colorectal cancer (CRC) is a multifaceted disease influenced by both environmental and genetic factors. A large body of literature has demonstrated the role of gut microbes in promoting inflammatory responses, creating a suitable microenvironment for the development of skewed interactions between the host and the gut microbiota and cancer initiation. Even if surgery is the primary therapeutic strategy, patients with advanced disease or cancer recurrence after surgery remain difficult to cure. Therefore, the gut microbiota has been proposed as a novel therapeutic target in light of recent promising data in which it seems to modulate the response to cancer immunotherapy. The use of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics, and fecal microbiota transplantation, is therefore considered to support current therapies in CRC management. In this review, we will discuss the importance of host-microbe interactions in CRC and how promoting homeostatic immune responses through microbe-targeted therapies may be useful in preventing/treating CRC development.


Asunto(s)
Neoplasias Colorrectales/terapia , Disbiosis/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Recurrencia Local de Neoplasia/terapia , Probióticos/uso terapéutico , Antibacterianos/uso terapéutico , Productos Biológicos/uso terapéutico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/patología , Interacciones Microbiota-Huesped/inmunología , Humanos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/microbiología , Recurrencia Local de Neoplasia/patología , Prebióticos/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
17.
Cell Commun Signal ; 18(1): 113, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677955

RESUMEN

BACKGROUND: Progression of Colorectal cancer (CRC) is influenced by single or compounded environmental factors. Accumulating evidence shows that microbiota can influence the outcome of cancer immunotherapy. T cell, one of the main populations of effector immune cells in antitumor immunity, has been considered as a double-edged sword during the progression of CRC. Our previous studies indicate that traditional Chinese herbs (TCM) have potential anticancer effects in improving quality of life and therapeutic effect. However, little is known about the mechanism of TCM formula in cancer prevention. METHODS: Here, we used C57BL/6 J ApcMin/+ mice, an animal model of human intestinal tumorigenesis, to investigate the gut bacterial diversity and their mechanisms of action in gastrointestinal adenomas, and to evaluate the effects of Yi-Yi-Fu-Zi-Bai-Jiang-San (YYFZBJS) on of colon carcinogenesis in vivo and in vitro. Through human-into-mice fecal microbiota transplantation (FMT) experiments from YYFZBJS volunteers or control donors, we were able to differentially modulate the tumor microbiome and affect tumor growth as well as tumor immune infiltration. RESULTS: We report herein, YYFZBJS treatment blocked tumor initiation and progression in ApcMin/+ mice with less change of body weight and increased immune function. Moreover, diversity analysis of fecal samples demonstrated that YYFZBJS regulated animal's natural gut flora, including Bacteroides fragilis, Lachnospiraceae and so on. Intestinal tumors from conventional and germ-free mice fed with stool from YYFZBJS volunteers had been decreased. Some inflammation' expression also have been regulated by the gut microbiota mediated immune cells. Intestinal lymphatic, and mesenteric lymph nodes (MLN), accumulated CD4+ CD25+ Foxp3 positive Treg cells were reduced by YYFZBJS treatment in ApcMin/+ mice. Although YYFZBJS had no inhibition on CRC cell proliferation by itself, the altered Tregs mediated by YYFZBJS repressed CRC cancer cell growth, along with reduction of the phosphorylation of ß-catenin. CONCLUSIONS: In conclusion, we demonstrated that gut microbiota and Treg were involved in CRC development and progression, and we propose YYFZBJS as a new potential drug option for the treatment of CRC. Video abstract.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/microbiología , Progresión de la Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Microbioma Gastrointestinal , Linfocitos T Reguladores/inmunología , Proteína de la Poliposis Adenomatosa del Colon , Animales , Bacteroides fragilis , Bromodesoxiuridina/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Células HCT116 , Humanos , Inmunidad/efectos de los fármacos , Antígeno Ki-67/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bazo/efectos de los fármacos , Bazo/patología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/efectos de los fármacos
18.
Expert Rev Clin Immunol ; 16(7): 689-709, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32536227

RESUMEN

INTRODUCTION: Treatment of colorectal cancer as one of the most commonly diagnosed and a frequent cause of cancer-related deaths is of great challenges in health-related issues. AREAS COVERED: Immunotherapy is the fourth pillar of cancer treatment which provides more novel therapeutic options with expanding investigational potentials. One of the modalities in immunotherapy is the use of bispecific antibodies. Despite demonstrating many promising roles, it still needs more advanced studies to identify the actual pros and cons. In this review, the application of bispecific antibody in the treatment of colorectal cancer has been explained, based on preclinical and clinical studies. The literature search was conducted mainly through PubMed in June and September 2019. EXPERT OPINION: Bispecific antibody is in its early stages in colorectal cancer treatment, requiring modern technologies in manufacturing, better biomarkers and more specific target antigens, more studies on individual genetic variations, and conducting later phase clinical trials and systematic reviews to achieve better survival benefits.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/terapia , Inmunoterapia/tendencias , Linfocitos T/inmunología , Animales , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/inmunología , Evaluación Preclínica de Medicamentos , Humanos
19.
J Sci Food Agric ; 100(15): 5442-5449, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32567144

RESUMEN

BACKGROUND: Yacón (Smallanthus sonchifolius) roots store carbohydrate in the form of prebiotic fructooligosaccharides (FOS), which improve intestinal health. Yacon has the potential to prevent the intestinal barrier alterations associated with colorectal cancer (CRC). This study aimed to investigate the preventive effects of yacón flour (YF) on alterations promoted by CRC induced by 1,2-dimethylhydrazine in rats. RESULTS: CRC increased tumor necrosis factor alpha levels (group CY = 10.2 ± 0.72; group C = 9.6 ± 1.0; group Y = 5.8 ± 0.54; group S = 5.95 ± 0.6 pg mL-1 ) and short-chain fatty acid production, and decreased total antioxidant capacity (group CY = 4.7 ± 0.72; group C = 3.3 ± 0.3; group Y = 4.1 ± 0.47; group S = 6.7 ± 0.78 U mL-1 ). Furthermore, YF treatment reduced intraluminal pH (group CY = 6.45 ± 0.47; group C = 7.65 ± 0.44; group Y = 6.75 ± 0.46; group S = 8.13 ± 0.2), lactulose/mannitol ratio, tumor necrosis factor-alpha (TNF-α)/interleukin (IL)-10 ratio, and increased secretory immunoglobulin A (group CY = 9.48 ± 1.46; group C = 10.95 ± 3.87; group Y = 15.95 ± 7.36; group S = 9.19 ± 1.52), but did not affect IL-10, IL-12, and TNF-α levels nor the IL-12/IL-10 ratio. CONCLUSION: YF as a source of fructooligosaccharides may help to maintain the integrity of intestinal health, which is altered in induced CRC in rats. © 2020 Society of Chemical Industry.


Asunto(s)
Asteraceae/química , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/prevención & control , Oligosacáridos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/metabolismo , Animales , Carcinogénesis , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Modelos Animales de Enfermedad , Ácidos Grasos Volátiles/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Intestinos/efectos de los fármacos , Intestinos/inmunología , Masculino , Raíces de Plantas/química , Prebióticos/análisis , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
Front Immunol ; 11: 684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425932

RESUMEN

Background: Colorectal cancer (CRC) is frequently associated with dysbiosis of the gut microbiome which, together with a compromised gut barrier, can result in perioperative endotoxin leakage into the circulation. Constant local and systemic inflammatory activity is suggested to facilitate metastases formation. Previous studies have pointed to the capacity of a colostrum preparation to neutralize endotoxins within the gastrointestinal tract which could ameliorate associated inflammatory responses and tumor recurrence in affected patients. This study aimed to examine the effects of the colostrum preparation, KMP01D, on the inflammatory activity of patient-derived immune cells. Methods: The effects of KMP01D on pro-/anti-inflammatory cytokine responses and apoptosis were examined ex vivo using immune cells from CRC patients (stages I-IV, n = 48). The expression of CD14, CD68, Toll-like receptor (TLR)4, and insulin-like growth factor (IGF)-1 was also analyzed. Results: KMP01D increased interleukin (IL)-10 and IL-13 anti-inflammatory cytokine expression in patient-derived peripheral blood mononuclear cells (PBMCs). Interestingly, KMP01D also decreased the secretion of IL-1ß, IL-6, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-12 inflammatory cytokines, and IGF-1 in these cells. Moreover, CD14 and TLR4 expression involved in endotoxin signaling was downregulated in PBMCs and tumor-derived cells. Apoptosis of immune cells and tumor-derived cells was likewise enhanced with KMP01D. Addition of vitamin D3 as a cofactor demonstrated enhanced anti-inflammatory effects. Conclusions: KMP01D demonstrated beneficial ex vivo effects on inflammatory cytokine responses in PBMCs and enhanced apoptosis of immune cells from CRC patients. In line with previous clinical trials, we present new evidence endorsing KMP01D as a treatment strategy to regulate stage-dependent local and systemic inflammation in CRC patients.


Asunto(s)
Antiinflamatorios/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Calostro , Monocitos/efectos de los fármacos , Anciano , Apoptosis , Colecalciferol/farmacología , Neoplasias Colorrectales/inmunología , Citocinas/biosíntesis , Microbioma Gastrointestinal/fisiología , Humanos , Inmunoglobulinas Intravenosas/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Persona de Mediana Edad , Monocitos/inmunología , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA