Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.076
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-34502247

RESUMO

Previously, we showed that a nitric oxide synthase (NOS) inhibitor, compound T1023, induces transient hypoxia and prevents acute radiation syndrome (ARS) in mice. Significant efficacy (according to various tests, dose modifying factor (DMF)-1.6-1.9 against H-ARS/G-ARS) and safety in radioprotective doses (1/5-1/4 LD10) became the reason for testing its ability to prevent complications of tumor radiation therapy (RT). Research methods included studying T1023 effects on skin acute radiation reactions (RSR) in rats and mice without tumors and in tumor-bearing animals. The effects were evaluated using clinical, morphological and histological techniques as well as RTOG classification. T1023 administration prior to irradiation significantly limited the severity of acute RSR. This was due to a decrease in radiation alteration of the skin and underlying tissues, and the preservation of the functional activity of cell populations that are critical in the pathogenesis of radiation burn. The DMF values for T1023 for skin protection were 1.4-1.7. Moreover, its radioprotective effect was fully selective to normal tissues in RT models of solid tumors-T1023 reduced the severity of acute RSR and did not modify the antitumor effects of γ-radiation. The results indicate that T1023 can selectively protect the non-malignant tissues against γ-radiation due to hypoxic mechanism of action and potentiate opportunities of NOS inhibitors in RT complications prevention.


Assuntos
Carcinoma de Ehrlich/radioterapia , Raios gama/efeitos adversos , Óxido Nítrico Sintase/antagonistas & inibidores , Protetores contra Radiação/farmacologia , Radiodermite/tratamento farmacológico , Sarcoma Experimental/radioterapia , Tioureia/análogos & derivados , Animais , Carcinoma de Ehrlich/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteção Radiológica/métodos , Radiodermite/etiologia , Radiodermite/patologia , Ratos , Ratos Sprague-Dawley , Sarcoma Experimental/patologia , Tioureia/farmacologia
2.
PLoS One ; 16(7): e0254426, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34292968

RESUMO

Aberrant NF-κB signaling fuels tumor growth in multiple human cancer types including both hematologic and solid malignancies. Chronic elevated alternative NF-κB signaling can be modeled in transgenic mice upon activation of a conditional NF-κB-inducing kinase (NIK) allele lacking the regulatory TRAF3 binding domain (NT3). Here, we report that expression of NT3 in the mesenchymal lineage with Osterix (Osx/Sp7)-Cre or Fibroblast-Specific Protein 1 (FSP1)-Cre caused subcutaneous, soft tissue tumors. These tumors displayed significantly shorter latency and a greater multiple incidence rate in Fsp1-Cre;NT3 compared to Osx-Cre;NT3 mice, regardless of sex. Histological assessment revealed poorly differentiated solid tumors with some spindled patterns, as well as robust RelB immunostaining, confirming activation of alternative NF-κB. Even though NT3 expression also occurs in the osteolineage in Osx-Cre;NT3 mice, we observed no bony lesions. The staining profiles and pattern of Cre expression in the two lines pointed to a mesenchymal tumor origin. Immunohistochemistry revealed that these tumors stain strongly for alpha-smooth muscle actin (αSMA), although vimentin staining was uniform only in Osx-Cre;NT3 tumors. Negative CD45 and S100 immunostains precluded hematopoietic and melanocytic origins, respectively, while positive staining for cytokeratin 19 (CK19), typically associated with epithelia, was found in subpopulations of both tumors. Principal component, differential expression, and gene ontology analyses revealed that NT3 tumors are distinct from normal mesenchymal tissues and are enriched for NF-κB related biological processes. We conclude that constitutive activation of the alternative NF-κB pathway in the mesenchymal lineage drives spontaneous sarcoma and provides a novel mouse model for NF-κB related sarcomas.


Assuntos
Regulação Neoplásica da Expressão Gênica , Integrases , Proteínas de Neoplasias , Proteínas Serina-Treonina Quinases , Proteína A4 de Ligação a Cálcio da Família S100 , Sarcoma Experimental , Fator de Transcrição Sp7 , Animais , Indução Enzimática , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Fator de Transcrição Sp7/genética , Fator de Transcrição Sp7/metabolismo , Quinase Induzida por NF-kappaB
3.
Cell Mol Immunol ; 18(3): 711-722, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32728200

RESUMO

In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.


Assuntos
Fibrossarcoma/patologia , Receptores de Hialuronatos/metabolismo , Interleucina-33/metabolismo , Neoplasias Pulmonares/patologia , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Apoptose , Proliferação de Células , Fibrossarcoma/etiologia , Fibrossarcoma/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Receptores de Hialuronatos/genética , Interleucina-33/genética , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos DBA , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Sarcoma Experimental/etiologia , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Phys Med Biol ; 65(20): 205012, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-32702686

RESUMO

Preclinical micro-CT provides a hotbed in which to develop new imaging technologies, including spectral CT using photon counting detector (PCD) technology. Spectral imaging using PCDs promises to expand x-ray CT as a functional imaging modality, capable of molecular imaging, while maintaining CT's role as a powerful anatomical imaging modality. However, the utility of PCDs suffers due to distorted spectral measurements, affecting the accuracy of material decomposition. We attempt to improve material decomposition accuracy using our novel hybrid dual-source micro-CT system which combines a PCD and an energy integrating detector. Comparisons are made between PCD-only and hybrid CT results, both reconstructed with our iterative, multi-channel algorithm based on the split Bregman method and regularized with rank-sparse kernel regression. Multi-material decomposition is performed post-reconstruction for separation of iodine (I), gold (Au), gadolinium (Gd), and calcium (Ca). System performance is evaluated first in simulations, then in micro-CT phantoms, and finally in an in vivo experiment with a genetically modified p53fl/fl mouse cancer model with Au, Gd, and I nanoparticle (NP)-based contrasts agents. Our results show that the PCD-only and hybrid CT reconstructions offered very similar spatial resolution at 10% MTF (PCD: 3.50 lp mm-1; hybrid: 3.47 lp mm-1) and noise characteristics given by the noise power spectrum. For material decomposition we note successful separation of the four basis materials. We found that hybrid reconstruction reduces RMSE by an average of 37% across all material maps when compared to PCD-only of similar dose but does not provide much difference in terms of concentration accuracy. The in vivo results show separation of targeted Au and accumulated Gd NPs in the tumor from intravascular iodine NPs and bone. Hybrid spectral micro-CT can benefit nanotechnology and cancer research by providing quantitative imaging to test and optimize various NPs for diagnostic and therapeutic applications.


Assuntos
Algoritmos , Meios de Contraste , Imagens de Fantasmas , Fótons , Sarcoma Experimental/diagnóstico por imagem , Sarcoma/diagnóstico por imagem , Microtomografia por Raio-X/instrumentação , Animais , Gadolínio , Humanos , Processamento de Imagem Assistida por Computador , Iodo , Camundongos , Sarcoma/induzido quimicamente , Sarcoma/patologia , Sarcoma Experimental/induzido quimicamente , Sarcoma Experimental/patologia
5.
J Clin Invest ; 130(9): 4921-4934, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32544087

RESUMO

Cachexia, a devastating wasting syndrome characterized by severe weight loss with specific losses of muscle and adipose tissue, is driven by reduced food intake, increased energy expenditure, excess catabolism, and inflammation. Cachexia is associated with poor prognosis and high mortality and frequently occurs in patients with cancer, chronic kidney disease, infection, and many other illnesses. There is no effective treatment for this condition. Hypothalamic melanocortins have a potent and long-lasting inhibitory effect on feeding and anabolism, and pathophysiological processes increase melanocortin signaling tone, leading to anorexia, metabolic changes, and eventual cachexia. We used 3 rat models of anorexia and cachexia (LPS, methylcholanthrene sarcoma, and 5/6 subtotal nephrectomy) to evaluate efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor. Our data show that peripheral treatment using TCMCB07 with intraperitoneal, subcutaneous, and oral administration increased food intake and body weight and preserved fat mass and lean mass during cachexia and LPS-induced anorexia. Furthermore, administration of TCMCB07 diminished hypothalamic inflammatory gene expression in cancer cachexia. These results suggest that peripheral TCMCB07 treatment effectively inhibits central melanocortin signaling and therefore stimulates appetite and enhances anabolism, indicating that TCMCB07 is a promising drug candidate for treating cachexia.


Assuntos
Caquexia/tratamento farmacológico , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores , Insuficiência Renal Crônica/tratamento farmacológico , Sarcoma Experimental/tratamento farmacológico , Animais , Apetite/efeitos dos fármacos , Caquexia/etiologia , Caquexia/metabolismo , Caquexia/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Melanocortina/metabolismo , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Sarcoma Experimental/complicações , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia
6.
J Immunother Cancer ; 8(1)2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32457127

RESUMO

BACKGROUND: High-dose radiotherapy (RT) is known to be immunogenic, but is rarely capable of driving clinically relevant abscopal antitumor immunity as monotherapy. RT is known to increase antigen presentation, type I/II interferon responses, and immune cell trafficking to irradiated tumors. Bempegaldesleukin (NKTR-214) is a CD122-preferential interleukin 2 (IL-2) pathway agonist that has been shown to increase tumor-infiltrating lymphocytes, T cell clonality, and increase PD-1 expression. NKTR-214 has increased drug half-life, decreased toxicity, and increased CD8+ T cell and natural killer cell stimulation compared with IL-2. METHODS: Animals bearing bilateral subcutaneous MCA-205 fibrosarcoma or CT26 colorectal tumors were treated with NKTR-214, RT, or combination therapy, and tumor growth of irradiated and abscopal lesions was assessed. Focal RT was delivered using a small animal radiation research platform. Peripheral and tumor-infiltrating immune phenotype and functional analyses were performed by flow cytometry. RNA expression profiling from both irradiated and abscopal lesions was performed using microarray. RESULTS: We demonstrate synergy between RT of a single tumor and NKTR-214 systemic therapy resulting in dramatically increased cure rates of mice bearing bilateral tumors compared with RT or NKTR-214 therapy alone. Combination therapy resulted in increased magnitude and effector function of tumor-specific CD8+ T cell responses and increased trafficking of these T cells to both irradiated and distant, unirradiated, tumors. CONCLUSIONS: Given the increasing role of hypofractionated and stereotactic body RT as standard of care treatments in the management of locally advanced and metastatic cancer, these data have important implications for future clinical trial development. The combination of RT and NKTR-214 therapy potently stimulates systemic antitumor immunity and should be evaluated for the treatment of patients with locally advanced and metastatic solid tumors.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias Colorretais/terapia , Fibrossarcoma/terapia , Interleucina-2/análogos & derivados , Linfócitos do Interstício Tumoral/imunologia , Polietilenoglicóis/uso terapêutico , Radioterapia/métodos , Sarcoma Experimental/terapia , Animais , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada , Feminino , Fibrossarcoma/imunologia , Fibrossarcoma/patologia , Imunoterapia/métodos , Interleucina-2/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Linfócitos T Reguladores/imunologia
7.
J Immunother Cancer ; 8(1)2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32273347

RESUMO

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) possess efficient antigen presentation and cross-presentation activity, as well as potent T cell priming ability. Tissue-resident cDC1s (CD103+ cDC1s in mice, CD141+ cDC1s in humans) are linked with improved tumor control, yet the efficacy of immunotherapy using this population is understudied. METHODS: We generated murine CD103+ cDC1s in vitro and examined their expression of cDC1-related factors, antigen cross-presentation activity, and accumulation in tumor-draining lymph nodes (TdLNs). The antitumor efficacy of the in vitro-generated CD103+ cDC1s was studied in murine melanoma and osteosarcoma models. We evaluated tumor responses on vaccination with CD103+ cDC1s, compared these to vaccination with monocyte-derived DCs (MoDCs), tested CD103+ cDC1 vaccination with checkpoint blockade, and examined the antimetastatic activity of CD103+ cDC1s. RESULTS: In vitro-generated CD103+ cDC1s produced cDC1-associated factors such as interleukin-12p70 and CXCL10, and demonstrated antigen cross-presentation activity on stimulation with the toll-like receptor 3 agonist polyinosinic:polycytidylic acid (poly I:C). In vitro-generated CD103+ cDC1s also migrated to TdLNs following poly I:C treatment and intratumoral delivery. Vaccination with poly I:C-activated and tumor antigen-loaded CD103+ cDC1s enhanced tumor infiltration of tumor antigen-specific and interferon-γ+ CD8+ T cells, and suppressed melanoma and osteosarcoma growth. CD103+ cDC1s showed superior antitumor efficacy compared with MoDC vaccination, and led to complete regression of 100% of osteosarcoma tumors in combination with CTLA-4 antibody-mediated checkpoint blockade. In vitro-generated CD103+ cDC1s effectively protected mice from pulmonary melanoma and osteosarcoma metastases. CONCLUSIONS: Our data indicate an in vitro-generated CD103+ cDC1 vaccine elicits systemic and long-lasting tumor-specific T cell-mediated cytotoxicity, which restrains primary and metastatic tumor growth. The CD103+ cDC1 vaccine was superior to MoDCs and enhanced response to immune checkpoint blockade. These results indicate the potential for new immunotherapies based on use of cDC1s alone or in combination with checkpoint blockade.


Assuntos
Antígenos CD/imunologia , Células Dendríticas/imunologia , Cadeias alfa de Integrinas/imunologia , Neoplasias Pulmonares/imunologia , Melanoma Experimental/imunologia , Osteossarcoma/imunologia , Sarcoma Experimental/imunologia , Vacinas/administração & dosagem , Animais , Apresentação de Antígeno/imunologia , Antígenos CD/metabolismo , Antígenos de Neoplasias/imunologia , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Apresentação Cruzada , Células Dendríticas/transplante , Imunoterapia , Técnicas In Vitro , Cadeias alfa de Integrinas/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Osteossarcoma/patologia , Osteossarcoma/terapia , Sarcoma Experimental/patologia , Sarcoma Experimental/terapia , Células Tumorais Cultivadas
8.
J Immunother Cancer ; 8(1)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32217757

RESUMO

BACKGROUND: CD6 is a lymphocyte surface co-receptor physically associated with the T-cell receptor (TCR)/CD3 complex at the center of the immunological synapse. There, CD6 assists in cell-to-cell contact stabilization and modulation of activation/differentiation events through interaction with CD166/ALCAM (activated leukocyte cell adhesion molecule), its main reported ligand. While accumulating evidence is attracting new interest on targeting CD6 for therapeutic purposes in autoimmune disorders, little is known on its potential in cancer. In an attempt to elucidate the in vivo relevance of blocking CD6-mediated interactions in health and disease, we explored the consequences of expressing high circulating levels of a soluble form CD6 (sCD6) as a decoy receptor. METHODS: High sCD6 serum levels were achieved by using transgenic C57BL/6 mice expressing human sCD6 under the control of lymphoid-specific transcriptional elements (shCD6LckEµTg) or wild type either transduced with hepatotropic adeno-associated virus coding for mouse sCD6 or undergoing repeated infusions of recombinant human sCD6 protein. Characterization of sCD6-induced changes was performed by ex vivo flow cytometry and functional analyses of mouse lymphoid organ cells. The in vivo relevance of those changes was explored by challenging mice with subcutaneous or metastatic tumors induced by syngeneic cancer cells of different lineage origins. RESULTS: Through a combination of in vitro and in vivo studies, we show that circulating sCD6 expression induces defective regulatory T cell (Treg) generation and function, decreased CD166/ALCAM-mediated tumor cell proliferation/migration and impaired galectin-induced T-cell apoptosis, supporting the fact that sCD6 modulates antitumor lymphocyte effector function and tumorigenesis. Accordingly, sCD6 expression in vivo resulted in delayed subcutaneous tumor growth and/or reduced metastasis on challenge of mice with syngeneic cancer cells. CONCLUSIONS: Evidence is provided for the disruption of CD6 receptor-ligand interactions as a feasible immunomodulatory approach in cancer.


Assuntos
Antígenos CD/sangue , Antígenos de Diferenciação de Linfócitos T/sangue , Neoplasias Pulmonares/imunologia , Linfoma de Células T/imunologia , Melanoma Experimental/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Reguladores/imunologia , Molécula de Adesão de Leucócito Ativado/imunologia , Molécula de Adesão de Leucócito Ativado/metabolismo , Animais , Antígenos CD/administração & dosagem , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/genética , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Linfoma de Células T/metabolismo , Masculino , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Sarcoma Experimental/sangue , Sarcoma Experimental/patologia , Linfócitos T Reguladores/metabolismo
9.
Sci Rep ; 9(1): 17220, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31748650

RESUMO

Cooperating gene mutations are typically required to transform normal cells enabling growth in soft agar or in immunodeficient mice. For example, mutations in Kras and transformation-related protein 53 (Trp53) are known to transform a variety of mesenchymal and epithelial cells in vitro and in vivo. Identifying other genes that can cooperate with oncogenic Kras and substitute for Trp53 mutation has the potential to lead to new insights into mechanisms of carcinogenesis. Here, we applied a genome-wide CRISPR/Cas9 knockout screen in KrasG12D immortalized mouse embryonic fibroblasts (MEFs) to search for genes that when mutated cooperate with oncogenic Kras to induce transformation. We also tested if mutation of the identified candidate genes could cooperate with KrasG12D to generate primary sarcomas in mice. In addition to identifying the well-known tumor suppressor cyclin dependent kinase inhibitor 2A (Cdkn2a), whose alternative reading frame product p19 activates Trp53, we also identified other putative tumor suppressors, such as F-box/WD repeat-containing protein 7 (Fbxw7) and solute carrier family 9 member 3 (Slc9a3). Remarkably, the TCGA database indicates that both FBXW7 and SLC9A3 are commonly co-mutated with KRAS in human cancers. However, we found that only mutation of Trp53 or Cdkn2a, but not Fbxw7 or Slc9a3 can cooperate with KrasG12D to generate primary sarcomas in mice. These results show that mutations in oncogenic Kras and either Fbxw7 or Slc9a3 are sufficient for transformation in vitro, but not for in vivo sarcomagenesis.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/patologia , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Sarcoma Experimental/prevenção & controle , Animais , Sistemas CRISPR-Cas , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas de Neoplasias/genética , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Transdução de Sinais
10.
Mol Pharm ; 16(4): 1714-1722, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30860846

RESUMO

Tumor associated macrophages (TAMs) are important components residing in the tumor microenvironment. They are immunosuppressive and promote tumor progression. Targeting TAMs and reprogramming their phenotype may be a promising strategy that can restore antitumor immune responses. In this study, we developed a microRNA delivery system based on lipid-coated calcium phosphonate nanoparticles (CaP/miR@pMNPs) containing conjugated mannose and sterically shielded with a pH-responsive material. The nanocarrier could respond to the low pH in the tumor microenvironment and expose mannose to promote cellular internalization in TAMs. The carrier could reactivate TAMs and reprogram their functions, reverse the immunosuppressive tumor microenvironment, and inhibit tumor growth in a tumor-bearing mouse model. In summary, redirecting the polarization of TAMs is a potential therapeutic strategy for tumor immunotherapy.


Assuntos
Imunoterapia , Macrófagos/imunologia , MicroRNAs/administração & dosagem , Nanopartículas/administração & dosagem , Sarcoma Experimental/terapia , Microambiente Tumoral/imunologia , Animais , Apoptose , Fosfatos de Cálcio/química , Proliferação de Células , Humanos , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , MicroRNAs/química , MicroRNAs/genética , Nanopartículas/química , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cancer Res ; 79(8): 1938-1951, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30777853

RESUMO

Cancer induces alteration of hematopoiesis to fuel disease progression. We report that in tumor-bearing mice the macrophage colony-stimulating factor elevates the myeloid cell levels of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD salvage pathway, which acts as negative regulator of the CXCR4 retention axis of hematopoietic cells in the bone marrow. NAMPT inhibits CXCR4 through a NAD/Sirtuin 1-mediated inactivation of HIF1α-driven CXCR4 gene transcription, leading to mobilization of immature myeloid-derived suppressor cells (MDSC) and enhancing their production of suppressive nitric oxide. Pharmacologic inhibition or myeloid-specific ablation of NAMPT prevented MDSC mobilization, reactivated specific antitumor immunity, and enhanced the antitumor activity of immune checkpoint inhibitors. Our findings identify NAMPT as a metabolic gate of MDSC precursor function, providing new opportunities to reverse tumor immunosuppression and to restore clinical efficacy of immunotherapy in patients with cancer. SIGNIFICANCE: These findings identify NAMPT as a metabolic gate of MDSC precursor function, providing new opportunities to reverse tumor immunosuppression and to restore clinical efficacy of immunotherapy in cancer patients.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Mamárias Experimentais/patologia , Células Supressoras Mieloides/patologia , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Sarcoma Experimental/patologia , Animais , Apoptose , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Hematopoese , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Supressoras Mieloides/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Invest New Drugs ; 37(5): 1044-1051, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30680583

RESUMO

Obtustatin, isolated from the Levantine Viper snake venom (Macrovipera lebetina obtusa -MLO), is the shortest known monomeric disintegrin shown to specifically inhibit the binding of the α1ß1 integrin to collagen IV. Its oncostatic effect is due to the inhibition of angiogenesis, likely through α1ß1 integrin inhibition in endothelial cells. To explore the therapeutic potential of obtustatin, we studied its effect in S-180 sarcoma-bearing mice model in vivo as well as in human dermal microvascular endothelial cells (HMVEC-D) in vitro, and tested anti-angiogenic activity in vivo using the chick embryo chorioallantoic membrane assay (CAM assay). Our in vivo results show that obtustatin inhibits tumour growth by 33%. The expression of vascular endothelial growth factor (VEGF) increased after treatment with obtustatin, but the level of expression of caspase 8 did not change. In addition, our results demonstrate that obtustatin inhibits FGF2-induced angiogenesis in the CAM assay. Our in vitro results show that obtustatin does not exhibit cytotoxic activity in HMVEC-D cells in comparison to in vivo results. Thus, our findings disclose that obtustatin might be a potential candidate for the treatment of sarcoma in vivo with low toxicity.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Sarcoma Experimental/tratamento farmacológico , Venenos de Víboras/farmacologia , Animais , Apoptose , Proliferação de Células , Embrião de Galinha , Membrana Corioalantoide , Integrina alfa1beta1/antagonistas & inibidores , Camundongos , Neovascularização Patológica/patologia , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 83(2): 361-374, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30506269

RESUMO

Macrophage-mediated drug delivery system has emerged and gained wide interest as a novel strategy for cancer treatment. Among them, RAW264.7 cell was commonly used as the macrophage model for antitumor drug loading and delivery. However, this cell line was a macrophage-like cancerous cell with both immunogenicity and pro-tumorigenic properties, which may interfere with the positive response of the host immune system to developed tumor. Thus, the safety and efficacy of the RAW264.7 cell line as a drug carrier for cancer therapy remain questionable. Here, we constructed doxorubicin-loaded RAW264.7 cells and examined its antitumor efficacy in S180 tumor-bearing mice. The bio-distribution of RAW264.7 cells was determined by in vivo imaging technique, showing a high accumulation level of RAW264.7 cells in mice livers, spleens, and thymuses. A phenomenon of accelerated tumor growth was observed in mice treated with doxorubicin-loaded RAW264.7 cells. Thereafter, the effect of frequency, dose, and viability of injected RAW264.7 cells on S180 tumor growth was further investigated. The underlying mechanism was confirmed, attributing to the immune tolerance induced by excessive RAW264.7 cells. Our findings emphasized the latent limitation of RAW264.7 cells as drug carrier in current researches, and provided an experimental basis for the clinical safety of cell-mediated drug delivery system.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Materiais Biomiméticos/administração & dosagem , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Macrófagos/transplante , Sarcoma Experimental/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/farmacocinética , Apoptose , Proliferação de Células , Doxorrubicina/farmacocinética , Liberação Controlada de Fármacos , Humanos , Macrófagos/citologia , Masculino , Camundongos , Células RAW 264.7 , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
15.
Photochem Photobiol Sci ; 18(1): 129-139, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30357250

RESUMO

Although infrared radiation (IR) represents more than 50% of the solar radiation reaching the Earth's surface, this waveband has been hardly investigated in terms of tumourigenesis. The objective of the present study was to investigate the influence of IR on ultraviolet B (UVB)-induced carcinogenesis in male and female wild type mice. For this purpose, male and female C57BL/6N mice were subjected to a long-term irradiation protocol. Mice were irradiated once neonatally and from the age of eight weeks for 36 weeks with a cumulative dose of 576 kJ m-2 UVB and/or 78 895 kJ m-2 IR. In order to resemble natural sun irradiation, exposure to physiological doses of UVB and IR was performed simultaneously. Mice were screened for arising lesions twice a week. Lesions were excised and histologically diagnosed. Kaplan-Meier analyses were carried out and lesion counts and cumulated hazard rates for the development of lesions in the UVB and IR + UVB-exposed groups in male and female mice were compared. We found that IR-exposure did not change the number of epithelial malignant tumours in UVB-exposed wild type mice. In combination with IR there was a tendency of more tumours with increased malignancy: 23 vs. seven spindle cell shaped sarcomas and seven vs. two MelanA+/S100+ tumours in groups of 35 C57BL/6 mice. IR did not influence UVB-induced carcinogenesis differently in male and female mice. However, comparing UVB and sham irradiated animals irrespective of IR exposure, UVB-induced non-epithelial tumours arose significantly earlier in male mice than in female mice.


Assuntos
Carcinogênese/efeitos da radiação , Raios Infravermelhos/efeitos adversos , Neoplasias Induzidas por Radiação/etiologia , Sarcoma Experimental/etiologia , Neoplasias Cutâneas/etiologia , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Feminino , Humanos , Estimativa de Kaplan-Meier , Antígeno MART-1/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas S100/análise , Sarcoma Experimental/patologia , Fatores Sexuais , Pele/patologia , Neoplasias Cutâneas/patologia
16.
Anticancer Agents Med Chem ; 19(3): 365-374, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30523769

RESUMO

OBJECTIVE: The aim of this study is to investigate the inhibitory effect of camptothecin derivative 3j on Non-Small Cell Lung Cancer (NSCLCs) cells and the potential anti-tumor mechanisms. BACKGROUND: Camptothecin compounds are considered as the third largest natural drugs which are widely investigated in the world and they suffered restriction because of serious toxicity, such as hemorrhagic cystitis and bone marrow suppression. METHODS: Using cell proliferation assay and S180 tumor mice model, a series of 20(S)-O-substituted benzoyl 7- ethylcamptothecin compounds were screened and evaluated the antitumor activities in vitro and in vivo. Camptothecin derivative 3j was selected for further study using flow cytometry in NSCLCs cells. Cell cycle related protein cyclin A2, CDK2, cyclin D and cyclin E were detected by Western Blot. Then, computer molecular docking was used to confirm the interaction between 3j and Topo I. Also, DNA relaxation assay and alkaline comet assay were used to investigate the mechanism of 3j on DNA damage. RESULTS: Our results demonstrated that camptothecin derivative 3j showed a greater antitumor effect in eleven 20(S)-O-substituted benzoyl 7-ethylcamptothecin compounds in vitro and in vivo. The IC50 of 3j was 1.54± 0.41 µM lower than irinotecan with an IC50 of 13.86±0.80 µM in NCI-H460 cell, which was reduced by 8 fold. In NCI-H1975 cell, the IC50 of 3j was 1.87±0.23 µM lower than irinotecan (IC50±SD, 5.35±0.38 µM), dropped by 1.8 fold. Flow cytometry analysis revealed that 3j induced significant accumulation in a dose-dependent manner. After 24h of 3j (10 µM) treatment, the percentage of NCI-H460 cell in S-phase significantly increased (to 93.54 ± 4.4%) compared with control cells (31.67 ± 3.4%). Similarly, the percentage of NCI-H1975 cell in Sphase significantly increased (to 83.99 ± 2.4%) compared with control cells (34.45 ± 3.9%) after treatment with 10µM of 3j. Moreover, increased levels of cyclin A2, CDK2, and decreased levels of cyclin D, cyclin E further confirmed that cell cycle arrest was induced by 3j. Furthermore, molecular docking studies suggested that 3j interacted with Topo I-DNA and DNA-relaxation assay simultaneously confirmed that 3j suppressed the activity of Topo I. Research on the mechanism showed that 3j exhibited anti-tumour activity via activating the DNA damage response pathway and suppressing the repair pathway in NSCLC cells. CONCLUSION: Novel camptothecin derivative 3j has been demonstrated as a promising antitumor agent and remains to be assessed in further studies.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , Inibidores da Topoisomerase I/farmacologia , Animais , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/química , Camptotecina/síntese química , Camptotecina/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Estrutura Molecular , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Relação Estrutura-Atividade , Inibidores da Topoisomerase I/síntese química , Inibidores da Topoisomerase I/química , Células Tumorais Cultivadas
17.
PLoS One ; 13(8): e0201220, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30080874

RESUMO

In recent years, immunogenic cell death (ICD) has emerged as a revolutionary concept in the development of novel anticancer therapies. This particular form of cell death is able, through the spatiotemporally defined emission of danger signals by the dying cell, to induce an effective antitumor immune response, allowing the immune system to recognize and eradicate malignant cells. To date, only a restricted number of chemotherapeutics can trigger ICD of cancer cells. We previously reported that a peptide, called RT53, spanning the heptad leucine repeat region of the survival protein AAC-11 fused to a penetrating sequence, selectively induces cancer cell death in vitro and in vivo. Interestingly, B16F10 melanoma cells treated by RT53 were able to mediate anticancer effects in a tumor vaccination model. Stimulated by this observation, we investigated whether RT53 might mediate ICD of cancer cells. Here, we report that RT53 treatment induces all the hallmarks of immunogenic cell death, as defined by the plasma membrane exposure of calreticulin, release of ATP and the exodus of high-mobility group box 1 protein (HMGB1) from dying cancer cells, through a non-regulated, membranolytic mode of action. In a prophylactic mouse model, vaccination with RT53-treated fibrosarcomas prevented tumor growth at the challenge site. Finally, local intratumoral injection of RT53 into established cancers led to tumor regression together with T-cell infiltration and the mounting of an inflammatory response in the treated animals. Collectively, our results strongly suggest that RT53 can induce bona fide ICD of cancer cells and illustrate its potential use as a novel antitumor and immunotherapeutic strategy.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Peptídeos Penetradores de Células/farmacologia , Proteínas Nucleares/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Proteínas Reguladoras de Apoptose/química , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Células HL-60 , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Peptídeos/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Sci Rep ; 8(1): 2194, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391414

RESUMO

A newly designed organoselenium compound, methyl substituted umbelliferone selenocyanate (MUS), was synthesized as a primary hit against the myelotoxic activity of carboplatin. MUS was administered at 6 mg/kg b.wt, p.o. in concomitant and pretreatment schedules with carboplatin (12 mg/kg b.wt, i.p. for 10 days) in female Swiss albino mouse. MUS treatment reduced (P < 0.001) the percentage of chromosomal aberrations, micronuclei formation, DNA damage and apoptosis in murine bone marrow cells and also enhanced (P < 0.001) the bone marrow cell proliferation of the carboplatin-treated mice. These activities cumulatively restored the viable bone marrow cell count towards normalcy. Myeloprotection by MUS was achieved, in part, due to a significant reduction in the ROS/RNS formation and restoration of glutathione redox pool. Additionally, MUS synergistically enhanced the cytotoxicity of carboplatin against two human cancer cell lines (MCF-7 and Colo-205). Furthermore, MUS can effectively potentiate the antitumour activity of carboplatin against two murine cancers (Dalton's Lymphoma and Sarcoma-180) in vivo. These preclinical findings clearly indicate that MUS can improve the therapeutic index of carboplatin and ensures more effective therapeutic strategy against cancer for clinical development.


Assuntos
Antineoplásicos/farmacologia , Cumarínicos/química , Desenho de Fármacos , Linfoma/tratamento farmacológico , Mielopoese/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Sarcoma Experimental/tratamento farmacológico , Animais , Antineoplásicos/química , Antioxidantes/química , Antioxidantes/farmacologia , Apoptose , Proliferação de Células , Quimioterapia Adjuvante , Aberrações Cromossômicas , Dano ao DNA/efeitos dos fármacos , Feminino , Peroxidação de Lipídeos , Linfoma/patologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
19.
Mol Cancer Ther ; 17(4): 858-868, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29437879

RESUMO

Carbon ion therapy (CIT) offers several potential advantages for treating cancers compared with X-ray and proton radiotherapy, including increased biological efficacy and more conformal dosimetry. However, CIT potency has not been characterized in primary tumor animal models. Here, we calculate the relative biological effectiveness (RBE) of carbon ions compared with X-rays in an autochthonous mouse model of soft tissue sarcoma. We used Cre/loxP technology to generate primary sarcomas in KrasLSL-G12D/+; p53fl/fl mice. Primary tumors were irradiated with a single fraction of carbon ions (10 Gy), X-rays (20 Gy, 25 Gy, or 30 Gy), or observed as controls. The RBE was calculated by determining the dose of X-rays that resulted in similar time to posttreatment tumor volume quintupling and exponential growth rate as 10 Gy carbon ions. The median tumor volume quintupling time and exponential growth rate of sarcomas treated with 10 Gy carbon ions and 30 Gy X-rays were similar: 27.3 and 28.1 days and 0.060 and 0.059 mm3/day, respectively. Tumors treated with lower doses of X-rays had faster regrowth. Thus, the RBE of carbon ions in this primary tumor model is 3. When isoeffective treatments of carbon ions and X-rays were compared, we observed significant differences in tumor growth kinetics, proliferative indices, and immune infiltrates. We found that carbon ions were three times as potent as X-rays in this aggressive tumor model and identified unanticipated differences in radiation response that may have clinical implications. Mol Cancer Ther; 17(4); 858-68. ©2018 AACR.


Assuntos
Proliferação de Células , Radioterapia com Íons Pesados , Sarcoma Experimental/radioterapia , Sarcoma/radioterapia , Animais , Apoptose , Relação Dose-Resposta à Radiação , Feminino , Masculino , Camundongos , Radiometria , Eficiência Biológica Relativa , Sarcoma/patologia , Sarcoma Experimental/patologia
20.
J BUON ; 23(6): 1904-1911, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30610820

RESUMO

PURPOSE: To examine the antitumor effects of Mebendazole (MZ) in a model of experimental fibrosarcoma induced by inoculation of BHK-21/C13 cells in Syrian golden hamster. METHODS: Hamsters were inoculated with a suspension of BHK cells by subcutaneous injection and randomly divided into 5 experimental and 2 control groups. Treatment started on the 10th day after inoculation, when the tumor grew to a diameter of 5mm. The experimental design was based on distributing the total amount of drug MZ(z) in different protocols and approaches (oral/intraperitoneal) to the 5 experimental groups. The positive control group received doxorubicin intraperitoneally. Negative control group received olive oil orally. The total amount of MZ(z) was chosen to be the highest for the animal to survive during the experiment. For antitumor effect evaluation, the main parameters were tumor size, number of mitoses, cytochrome-C immunopositivity and tumor tissue morphology incuding cytoarchitecture and percentage of preserved tumor tissue in stereologically reconstructed tumor mass. RESULTS: The results of this study showed absence of objective MZ antitumor effect on experimental fibrosarcoma. MZ does not exhibit activity similar to DNA-damaging agents on the fibrosarcoma model. CONCLUSIONS: It might be postulated that soft tissue tumors on animal models could show high level of resistance to MZ effect.


Assuntos
Proliferação de Células/efeitos dos fármacos , Reposicionamento de Medicamentos/métodos , Fibrossarcoma/patologia , Mebendazol/uso terapêutico , Sarcoma Experimental/patologia , Moduladores de Tubulina/uso terapêutico , Animais , Fibrossarcoma/tratamento farmacológico , Mesocricetus , Sarcoma Experimental/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...