Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 813
Filtrar
1.
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
2.
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
3.
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
4.
Drug Deliv ; 25(1): 1826-1839, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30458644

RESUMO

Nanoparticles (NPs) have proven to be effective drug carriers in diagnosis and therapy of cancer. But, they faced a contradictory issue that NPs with large size appear weak tumor penetration, meanwhile small size resulted in poor tumor retention. Herein, we fabricated doxorubicin conjugated carbon dots (CDs-DOX) and indocyanine green (ICG)-loaded liposomes (ICG-LPs) named CDs-ICG-LPs using a modified reverse phase evaporation process, and with high incorporation in the aqueous core. The CDs-ICG-LPs exhibited good monodispersity, excellent fluorescence/size stability, and consistent spectra characteristics compared with free ICG or DOX. Moreover, the CDs-ICG-LPs showed higher temperature response, faster DOX release under laser irradiation. In the meantime, the fluorescence of DOX and ICG in CDs-ICG-LPs was also visualized for the process of subcellular location in vitro. In comparison with chemo or photothermal treatment alone, the combined treatment of CDs-ICG-LPs with laser irradiation synergistically induced the apoptosis and death of DOX-sensitive HepG2 cells. In vivo antitumor activities demonstrated CDs-ICG-LPs could reach higher antitumor activity compared with CDs-DOX and ICG-LPs for H22 tumor cells, and suppressed H22 tumor growth in vivo. Notably, no systemic toxicity occurrence was observed after repeated dose of CDs-ICG-LPs with laser irradiation. Hence, the well-defined CDs-ICG-LPs exhibited great potential in targeting cancer imaging and chemo-photothermal therapy.


Assuntos
Antineoplásicos/administração & dosagem , Carbono/administração & dosagem , Doxorrubicina/administração & dosagem , Verde de Indocianina/administração & dosagem , Fármacos Fotossensibilizantes/administração & dosagem , Pontos Quânticos/administração & dosagem , Sarcoma Experimental/terapia , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carbono/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Desenho de Fármacos , Liberação Controlada de Fármacos , Quimioterapia Combinada , Células Hep G2 , Humanos , Hipertermia Induzida/métodos , Verde de Indocianina/uso terapêutico , Lipossomos , Masculino , Camundongos , Nanopartículas , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia
5.
Nanomedicine ; 14(2): 289-301, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28993266

RESUMO

In order to prevent the aggregation of ICG and enhance its stability, a novel nanoplatform (TiO2:Yb,Ho,F-ß-CD@ICG/HA) was designed for NIR-induced phototherapy along with multi-mode imaging(UCL/MRI/Flu). In this nanosysytem: TiO2:Yb,Ho,F was used as upconversion materials and applied in vivo for the first time; ß-CD acted as a "protective umbrella" to load separated ICG and avoid the low phototherapy efficiency because of its aggregation; HA was the capping agent of ß-CD to prevent ICG unexpected leaking and a target to recognize CD44 receptor. The nanosystem exhibited excellent size (~200 nm) and photo- and thermal-stability, preferable reactive oxygen yield and temperature response (50.4 °C) under 808 nm laser. It could efficiently target and suppress tumor growth. The imaging ability (UCL/MRI) of TiO2:Yb,Ho,F could facilitate diagnosis of the tumor, especially for deep tissues. Altogether, our work successfully improved the phototherapy efficacy through incorporating the ICG into the cavity of ß-CD and applied TiO2:Yb,Ho,F for upconversion imaging in vivo.


Assuntos
Verde de Indocianina/metabolismo , Neoplasias Mamárias Experimentais/terapia , Imagem Multimodal/métodos , Nanopartículas/administração & dosagem , Fototerapia , Sarcoma Experimental/terapia , Animais , Apoptose , Ciclo Celular , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Nanopartículas/química , Espécies Reativas de Oxigênio/metabolismo , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Células Tumorais Cultivadas
6.
J Immunother Cancer ; 5(1): 98, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29254507

RESUMO

BACKGROUND: We have previously shown that radiotherapy (RT) augments natural killer (NK) functions in pre-clinical models of human and mouse cancers, including sarcomas. Since dogs are an excellent outbred model for immunotherapy studies, we sought to assess RT plus local autologous NK transfer in canine sarcomas. METHODS: Dog NK cells (CD5dim, NKp46+) were isolated from PBMCs and expanded with irradiated K562-C9-mIL21 feeder cells and 100 IU/mL recombinant human IL-2. NK homing and cytotoxicity ± RT were evaluated using canine osteosarcoma tumor lines and dog patient-derived xenografts (PDX). In a first-in-dog clinical trial for spontaneous osteosarcoma, we evaluated RT and intra-tumoral autologous NK transfer. RESULTS: After 14 days, mean NK expansion and yield were 19.0-fold (±8.6) and 258.9(±76.1) ×106 cells, respectively. Post-RT, NK cytotoxicity increased in a dose-dependent fashion in vitro reaching ~ 80% at effector:target ratios of ≥10:1 (P < 0.001). In dog PDX models, allogeneic NK cells were cytotoxic in ex vivo killing assays and produced significant PDX tumor growth delay (P < 0.01) in vivo. After focal RT and intravenous NK transfer, we also observed significantly increased NK homing to tumors in vivo. Of 10 dogs with spontaneous osteosarcoma treated with focal RT and autologous NK transfer, 5 remain metastasis-free at the 6-month primary endpoint with resolution of suspicious pulmonary nodules in one patient. We also observed increased activation of circulating NK cells after treatment and persistence of labelled NK cells in vivo. CONCLUSIONS: NK cell homing and cytotoxicity are increased following RT in canine models of sarcoma. Results from a first-in-dog clinical trial are promising, including possible abscopal effects.


Assuntos
Células Matadoras Naturais/citologia , Osteossarcoma/terapia , Radioterapia/métodos , Sarcoma Experimental/terapia , Animais , Técnicas de Cocultura , Terapia Combinada , Citotoxicidade Imunológica , Doenças do Cão/terapia , Cães , Humanos , Células K562 , Células Matadoras Naturais/transplante , Osteossarcoma/veterinária , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Integr Biol (Camb) ; 9(6): 555-565, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28513646

RESUMO

Magnetic liposome-mediated combined chemotherapy and hyperthermia is gaining importance as an effective therapeutic modality for cancer. However, control and maintenance of optimum hyperthermia are major challenges in clinical settings due to the overheating of tissues. To overcome this problem, we developed a novel magnetic liposomes formulation co-entrapping a dextran coated biphasic suspension of La0.75Sr0.25MnO3 (LSMO) and iron oxide (Fe3O4) nanoparticles for self-controlled hyperthermia and chemotherapy. However, the general apprehension about biocompatibility and safety of the newly developed formulation needs to be addressed. In this work, in vitro and in vivo biocompatibility and therapeutic evaluation studies of the novel magnetic liposomes are reported. Biocompatibility study of the magnetic liposomes formulation was carried out to evaluate the signs of preliminary systemic toxicity, if any, following intravenous administration of the magnetic liposomes in Swiss mice. Therapeutic efficacy of the magnetic liposomes formulation was evaluated in the fibrosarcoma tumour bearing mouse model. Fibrosarcoma tumour-bearing mice were subjected to hyperthermia following intratumoral injection of single or double doses of the magnetic liposomes with or without chemotherapeutic drug paclitaxel. Hyperthermia (three spurts, each at 3 days interval) with drug loaded magnetic liposomes following single dose administration reduced the growth of tumours by 2.5 fold (mean tumour volume 2356 ± 550 mm3) whereas the double dose treatment reduced the tumour growth by 3.6 fold (mean tumour volume 1045 ± 440 mm3) compared to their corresponding control (mean tumour volume 3782 ± 515 mm3). At the end of the tumour efficacy studies, the presence of MNPs was studied in the remnant tumour tissues and vital organs of the mice. No significant leaching or drainage of the magnetic liposomes during the study was observed from the tumour site to the other vital organs of the body, suggesting again the potential of the novel magnetic liposomes formulation for possibility of developing as an effective modality for treatment of drug resistant or physiologically vulnerable cancer.


Assuntos
Hipertermia Induzida/métodos , Lipossomos/uso terapêutico , Magnetismo , Neoplasias/terapia , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Materiais Biocompatíveis/uso terapêutico , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Humanos , Lipossomos/administração & dosagem , Lipossomos/toxicidade , Nanopartículas de Magnetita/administração & dosagem , Nanopartículas de Magnetita/uso terapêutico , Nanopartículas de Magnetita/toxicidade , Teste de Materiais , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Paclitaxel/administração & dosagem , Sarcoma Experimental/tratamento farmacológico , Sarcoma Experimental/metabolismo , Sarcoma Experimental/terapia , Distribuição Tecidual
8.
PLoS One ; 11(4): e0153633, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27088717

RESUMO

PURPOSE: Prenyl ammonium iodides (Amino-Prenols, APs), semi-synthetic polyprenol derivatives were studied as prospective novel gene transfer agents. METHODS: AP-7, -8, -11 and -15 (aminoprenols composed of 7, 8, 11 or 15 isoprene units, respectively) were examined for their capacity to form complexes with pDNA, for cytotoxicity and ability to transfect genes to cells. RESULTS: All the carriers were able to complex DNA. The highest, comparable to commercial reagents, transfection efficiency was observed for AP-15. Simultaneously, AP-15 exhibited the lowest negative impact on cell viability and proliferation--considerably lower than that of commercial agents. AP-15/DOPE complexes were also efficient to introduce pDNA to cells, without much effect on cell viability. Transfection with AP-15/DOPE complexes influenced the expression of a very few among 44 tested genes involved in cellular lipid metabolism. Furthermore, complexes containing AP-15 and therapeutic plasmid, encoding the TIMP metallopeptidase inhibitor 2 (TIMP2), introduced the TIMP2 gene with high efficiency to B16-F10 melanoma cells but not to B16-F10 melanoma tumors in C57BL/6 mice, as confirmed by TIMP2 protein level determination. CONCLUSION: Obtained results indicate that APs have a potential as non-viral vectors for cell transfection.


Assuntos
Compostos de Amônio/farmacologia , DNA/administração & dosagem , Sistemas de Liberação de Medicamentos , Terapia Genética , Melanoma Experimental/terapia , Neopreno/química , Sarcoma Experimental/terapia , Animais , Western Blotting , Sobrevivência Celular , Portadores de Fármacos , Feminino , Técnicas de Transferência de Genes , Técnicas Imunoenzimáticas , Lipossomos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcoma Experimental/genética , Sarcoma Experimental/patologia , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Bone Joint Surg Am ; 98(1): 48-55, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26738903

RESUMO

BACKGROUND: Radiation-induced fibrosis (RIF) of musculoskeletal tissue is a common complication of radiation therapy for extremity soft-tissue sarcoma, with no standardized strategy for prevention and treatment. Angiotensin-(1-7) (Ang-[1-7]), a well-tolerated endogenous heptapeptide hormone with antitumor and antifibrotic properties, was tested as a radioprotectant for RIF and stiffening of irradiated muscles. METHODS: Male CD-1 mice were randomized to one of three treatment groups: control, simulated sarcoma radiation therapy to the gastrocnemius and soleus muscles, or radiation therapy along with continuous Ang-(1-7) delivery initiated three days before radiation therapy. The biologically equivalent dose of radiation (∼100.3 Gy) absorbed by normal musculature during the course of radiation therapy for extremity sarcoma was delivered by means of four dose fractions of 7.3 Gy over two weeks. Fibrosis (n = 5 per group) and mechanical properties (n = 4 to 6 per group) of the muscles were measured at six weeks and four months after radiation therapy, and the intramuscular concentration of the profibrotic cytokines transforming growth factor-beta (TGF-ß) and connective tissue growth factor (CTGF) (n = 8 to 10 per group) were measured at six weeks. RESULTS: Interstitial (p < 0.01) and perivascular (p < 0.05) fibrosis increased significantly in the muscles treated with radiation therapy alone versus the nonirradiated controls at both six weeks (interstitial, +89%; perivascular, +112%) and four months (interstitial, +154%; perivascular, +88%). The muscles treated with radiation alone also exhibited increased tension (p < 0.01) versus nonirradiated controls at both six weeks (+779%) and four months (+1761%) when placed under 5% strain, and at four months (+1390%; p < 0.001) under 10% strain. At four months, muscle stiffness had increased in the mice treated with radiation therapy alone (+90%; p = 0.002) compared with nonirradiated controls. TGF-ß production was also greater in this group at six weeks (+37%; p = 0.06) versus control. Ang-(1-7) administration prevented RIF and stiffening, with no differences observed for any other outcome between those receiving radiation therapy with Ang-(1-7) and the nonirradiated controls. Likewise, Ang-(1-7) mitigated the increase in TGF-ß and CTGF concentration from radiation therapy. CONCLUSIONS: Ang-(1-7) attenuated RIF, stiffening, and production of profibrotic cytokines that were elevated in mouse skeletal muscles after simulated radiation therapy for extremity sarcoma. CLINICAL RELEVANCE: Ang-(1-7) may serve as a potential therapy for the prevention of RIF in patients who require radiation therapy as adjuvant treatment for soft-tissue sarcoma.


Assuntos
Angiotensina I/administração & dosagem , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/efeitos da radiação , Fragmentos de Peptídeos/administração & dosagem , Sarcoma Experimental/terapia , Espasmo/prevenção & controle , Análise de Variância , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Fibrose/etiologia , Fibrose/prevenção & controle , Membro Posterior , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Neoplasias Musculares/patologia , Neoplasias Musculares/terapia , Músculo Esquelético/patologia , Distribuição Aleatória , Valores de Referência , Sarcoma Experimental/patologia , Sensibilidade e Especificidade , Espasmo/patologia
10.
J Clin Invest ; 125(4): 1603-19, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25751063

RESUMO

Here, we describe the multiple lentiviral expression (MuLE) system that allows multiple genetic alterations to be introduced simultaneously into mammalian cells. We created a toolbox of MuLE vectors that constitute a flexible, modular system for the rapid engineering of complex polycistronic lentiviruses, allowing combinatorial gene overexpression, gene knockdown, Cre-mediated gene deletion, or CRISPR/Cas9-mediated (where CRISPR indicates clustered regularly interspaced short palindromic repeats) gene mutation, together with expression of fluorescent or enzymatic reporters for cellular assays and animal imaging. Examples of tumor engineering were used to illustrate the speed and versatility of performing combinatorial genetics using the MuLE system. By transducing cultured primary mouse cells with single MuLE lentiviruses, we engineered tumors containing up to 5 different genetic alterations, identified genetic dependencies of molecularly defined tumors, conducted genetic interaction screens, and induced the simultaneous CRISPR/Cas9-mediated knockout of 3 tumor-suppressor genes. Intramuscular injection of MuLE viruses expressing oncogenic H-RasG12V together with combinations of knockdowns of the tumor suppressors cyclin-dependent kinase inhibitor 2A (Cdkn2a), transformation-related protein 53 (Trp53), and phosphatase and tensin homolog (Pten) allowed the generation of 3 murine sarcoma models, demonstrating that genetically defined autochthonous tumors can be rapidly generated and quantitatively monitored via direct injection of polycistronic MuLE lentiviruses into mouse tissues. Together, our results demonstrate that the MuLE system provides genetic power for the systematic investigation of the molecular mechanisms that underlie human diseases.


Assuntos
Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas , Clonagem Molecular/métodos , Vetores Genéticos , Lentivirus/genética , Animais , Apoptose , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Doxiciclina/farmacologia , Resistência a Medicamentos/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Camundongos SCID , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/genética , RNA Interferente Pequeno/genética , Recombinação Genética , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/genética , Sarcoma Experimental/genética , Sarcoma Experimental/terapia , Transdução Genética , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Radiat Oncol Biol Phys ; 91(3): 621-30, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25544668

RESUMO

PURPOSE: To examine the addition of genetic or pharmacologic inhibition of hypoxia-inducible factor 1α (HIF-1α) to radiation therapy (RT) and vascular endothelial growth factor A (VEGF-A) inhibition (ie trimodality therapy) for soft-tissue sarcoma. METHODS AND MATERIALS: Hypoxia-inducible factor 1α was inhibited using short hairpin RNA or low metronomic doses of doxorubicin, which blocks HIF-1α binding to DNA. Trimodality therapy was examined in a mouse xenograft model and a genetically engineered mouse model of sarcoma, as well as in vitro in tumor endothelial cells (ECs) and 4 sarcoma cell lines. RESULTS: In both mouse models, any monotherapy or bimodality therapy resulted in tumor growth beyond 250 mm(3) within the 12-day treatment period, but trimodality therapy with RT, VEGF-A inhibition, and HIF-1α inhibition kept tumors at <250 mm(3) for up to 30 days. Trimodality therapy on tumors reduced HIF-1α activity as measured by expression of nuclear HIF-1α by 87% to 95% compared with RT alone, and cytoplasmic carbonic anhydrase 9 by 79% to 82%. Trimodality therapy also increased EC-specific apoptosis 2- to 4-fold more than RT alone and reduced microvessel density by 75% to 82%. When tumor ECs were treated in vitro with trimodality therapy under hypoxia, there were significant decreases in proliferation and colony formation and increases in DNA damage (as measured by Comet assay and γH2AX expression) and apoptosis (as measured by cleaved caspase 3 expression). Trimodality therapy had much less pronounced effects when 4 sarcoma cell lines were examined in these same assays. CONCLUSIONS: Inhibition of HIF-1α is highly effective when combined with RT and VEGF-A inhibition in blocking sarcoma growth by maximizing DNA damage and apoptosis in tumor ECs, leading to loss of tumor vasculature.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Neovascularização Patológica/terapia , RNA Interferente Pequeno/uso terapêutico , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/terapia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Terapia Combinada/métodos , Dano ao DNA , Doxorrubicina/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Transgênicos/genética , Tolerância a Radiação , Radioterapia , Sarcoma Experimental/genética , Sarcoma Experimental/metabolismo , Sarcoma Experimental/patologia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Int J Cancer ; 136(4): 965-76, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24978211

RESUMO

Isolated limb perfusion (ILP) is a treatment for advanced extremity sarcoma and in-transit melanoma. Advancing this procedure by investigating the addition of novel agents, such as cancer-selective oncolytic viruses, may improve both the therapeutic efficacy of ILP and the tumour-targeted delivery of oncolytic virotherapy. Standard in vitro assays were used to characterise single agent and combinatorial activities of melphalan, tumour necrosis factor-alpha (TNF-α) and Lister strain vaccinia virus (GLV-1h68) against BN175 rat sarcoma cells. An orthotopic model of advanced extremity sarcoma was used to evaluate survival of animals after ILP with combinations of TNF-α, melphalan and GLV-1h68. We investigated the efficiency of viral tumour delivery by ILP compared to intravenous therapy, the locoregional and systemic biodistribution of virus after ILP, and the effect of mode of administration on antibody response. The combination of melphalan and GLV-1h68 was synergistic in vitro. The addition of virus to standard ILP regimens was well tolerated and demonstrated superior tumour targeting compared to intravenous administration. Triple therapy (melphalan/TNF-α/GLV-1h68) resulted in increased tumour growth delay and enhanced survival compared to other treatment regimens. Live virus was recovered in large amounts from perfused regions, but in smaller amounts from systemic organs. The addition of oncolytic vaccinia virus to existing TNF-α/melphalan-based ILP strategies results in survival advantage in an immunocompetent rat model of advanced extremity sarcoma. Virus administered by ILP has superior tumour targeting compared to intravenous delivery. Further evaluation and clinical translation of this approach is warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Membro Posterior/patologia , Vírus Oncolíticos/fisiologia , Sarcoma Experimental/terapia , Vaccinia virus/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Quimioterapia do Câncer por Perfusão Regional , Terapia Combinada , Membro Posterior/efeitos dos fármacos , Humanos , Masculino , Melfalan/administração & dosagem , Transplante de Neoplasias , Ratos Endogâmicos , Sarcoma Experimental/irrigação sanguínea , Sarcoma Experimental/patologia , Fator de Necrose Tumoral alfa/administração & dosagem
13.
Cancer Immunol Res ; 1(1): 64-76, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24777249

RESUMO

Collaborative action between tumor cells and host-derived suppressor cells leads to peripheral tolerance of T cells to tumor antigens. Here, we showed that in tumor-bearing mice, generation of tumor antigen-specific effector T-helper cells (TH1) was significantly attenuated, and impaired TH1 differentiation was restored by the temporal blockade of interleukin (IL)-6 activity at the T-cell priming phase. Furthermore, we found that Gr-1(+) myeloid-derived suppressor cells (MDSC) served as a source of IL-6 in tumor-bearing mice. Adoptive transfer of effector CD4(+) T cells revealed that MDSC-sensitized effector CD4(+) T cells were less potent in mounting antitumor immune responses, although effector T cells generated together with Gr-1(+) cells from tumor-free mice eradicated established tumors. CD8(+) T cells, IFN-γ, and MHC-class II expression in host mice were indispensable for the antitumor activity initiated by effector CD4(+) T cells. Despite comparable suppressive activity of IL-6(+/+) and IL-6(-/-) MDSC on primary T-cell activation, transfer of IL-6(+/+) MDSC, but not IL-6(-/-) MDSC, dampened the efficient induction of effector TH1 cells and counteracted CD4(+) T cell-mediated antitumor immunity including cognate help for CD8(+) T cells in vivo. These findings suggest that, apart from the inhibitory effects on primary T-cell activation, MDSC promote tumor progression by attenuating functional differentiation of tumor-specific CD4(+) T cells into effector TH1 cells through IL-6 production to promote tumor progression. This novel mode of MDSC-induced tolerance of effector CD4(+) T cells should be considered as the basis for the rational design of effective T cell-mediated antitumor therapies.


Assuntos
Interleucina-6/imunologia , Células Mieloides/imunologia , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Progressão da Doença , Interleucina-6/biossíntese , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia
14.
Immunotherapy ; 4(10): 1023-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23148754

RESUMO

Soft tissue sarcomas represent a rare but diverse family of tumors affecting patients of all ages. Conventional chemotherapy rarely eradicates metastatic disease and newer targeted agents are successful in only very specific histologic subgroups. Therefore, scientists and clinicians are investigating immunotherapy techniques, primarily involving cellular immunity focused on the T-cell response to tumor antigens. In both animal models and human sarcoma trials, dendritic cells have been shown to induce an effective antitumor immune response. Radiotherapy, particularly when delivered in a hypofractionated manner prior to sarcoma excision, may potentiate the function of the dendritic cells through the induction of apoptosis. The synergistic effect may carry over to other cancer types and warrants further multidisciplinary investigation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Imunoterapia/métodos , Sarcoma/imunologia , Sarcoma/terapia , Animais , Antígenos de Neoplasias/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunidade Celular , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Linfócitos T/imunologia
15.
PLoS One ; 7(4): e34467, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496812

RESUMO

The provision of T cell co-stimulation via members of the TNFR super-family, including OX40 (CD134) and 4-1BB (CD137), provides critical signals that promote T cell survival and differentiation. Recent studies have demonstrated that ligation of OX40 can augment T cell-mediated anti-tumor immunity in pre-clinical models and more importantly, OX40 agonists are under clinical development for cancer immunotherapy. OX40 is of particular interest as a therapeutic target as it is not expressed on naïve T cells but rather, is transiently up-regulated following TCR stimulation. Although TCR engagement is necessary for inducing OX40 expression, the downstream signals that regulate OX40 itself remain unclear. In this study, we demonstrate that OX40 expression is regulated through a TCR and common gamma chain cytokine-dependent signaling cascade that requires JAK3-mediated activation of the downstream transcription factors STAT3 and STAT5. Furthermore, combined treatment with an agonist anti-OX40 mAb and IL-2 augmented tumor immunotherapy against multiple tumor types. Dual therapy was also able to restore the function of anergic tumor-reactive CD8 T cells in mice with long-term well-established (>5 wks) tumors, leading to increased survival of the tumor-bearing hosts. Together, these data reveal the ability of TCR/common gamma chain cytokine signaling to regulate OX40 expression and demonstrate a novel means of augmenting cancer immunotherapy by providing dual anti-OX40/common gamma chain cytokine-directed therapy.


Assuntos
Imunoterapia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Interleucina-2/uso terapêutico , Receptores OX40/fisiologia , Sarcoma Experimental/imunologia , Sarcoma Experimental/terapia , Animais , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Citocinas/metabolismo , Quimioterapia Combinada , Feminino , Citometria de Fluxo , Janus Quinase 3/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Transcrição STAT3/fisiologia , Fator de Transcrição STAT5/fisiologia , Sarcoma Experimental/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Tumorais Cultivadas
17.
Hum Gene Ther ; 23(2): 173-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21958207

RESUMO

Sarcoma at advanced stages remains a clinically challenging disease. Interferons (IFNs) can target cancer cells by multiple antitumor activities, including the induction of cancer cell death and enhancement of immune response. However, the development of an effective cancer immunotherapy is often difficult, because cancer generates an immunotolerant microenvironment against the host immune system. An autologous hematopoietic stem cell transplantation (HSCT) is expected to reconstitute a fresh immune system, and expand tumor-specific T cells through the process of homeostatic proliferation. Here we examined whether a combination of autologous HSCT and IFNs could induce an effective tumor-specific immune response against sarcoma. First, we found that a type I IFN gene transfer significantly suppressed the cell growth of various sarcoma cell lines, and that IFN-ß gene transfer was more effective in inducing cell death than was IFN-α in sarcoma cells. Then, to examine the antitumor effect in vivo, human sarcoma cells were inoculated in immune-deficient mice, and a lipofection of an IFN-ß-expressing plasmid was found to suppress the growth of subcutaneous tumors significantly. Finally, the IFN gene transfer was combined with syngeneic HSCT in murine osteosarcoma models. Intratumoral IFN-ß gene transfer markedly suppressed the growth of vector-injected tumors and inhibited formation of spontaneous lung and liver metastases in syngeneic HSCT mice, and an infiltration of many immune cells was recognized in metastatic tumors of the treated mice. The treated mice showed no significant adverse events. A combination of intratumoral IFN gene transfer with autologous HSCT could be a promising therapeutic strategy for patients with sarcoma.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Interferon beta/genética , Sarcoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Imunoterapia , Interferon beta/imunologia , Lipídeos , Camundongos , Camundongos Nus , Metástase Neoplásica , Transplante de Neoplasias , Plasmídeos , Sarcoma Experimental/imunologia , Sarcoma Experimental/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Immunol ; 187(10): 4935-46, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22013118

RESUMO

Optimum efficacy of therapeutic cancer vaccines may require combinations that generate effective antitumor immune responses, as well as overcome immune evasion and tolerance mechanisms mediated by progressing tumor. Previous studies showed that IL-13Rα2, a unique tumor-associated Ag, is a promising target for cancer immunotherapy. A targeted cytotoxin composed of IL-13 and mutated Pseudomonas exotoxin induced specific killing of IL-13Rα2(+) tumor cells. When combined with IL-13Rα2 DNA cancer vaccine, surprisingly, it mediated synergistic antitumor effects on tumor growth and metastasis in established murine breast carcinoma and sarcoma tumor models. The mechanism of synergistic activity involved direct killing of tumor cells and cell-mediated immune responses, as well as elimination of myeloid-derived suppressor cells and, consequently, regulatory T cells. These novel results provide a strong rationale for combining immunotoxins with cancer vaccines for the treatment of patients with advanced cancer.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Exotoxinas/uso terapêutico , Marcação de Genes/métodos , Imunoterapia/métodos , Subunidade alfa2 de Receptor de Interleucina-13/uso terapêutico , Neoplasias Mamárias Experimentais/terapia , Sarcoma Experimental/terapia , Vacinas de DNA/uso terapêutico , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/genética , ADP Ribose Transferases/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Exotoxinas/genética , Exotoxinas/imunologia , Feminino , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Subunidade alfa2 de Receptor de Interleucina-13/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Vacinas de DNA/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
19.
J Radiat Res ; 52(5): 568-74, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799293

RESUMO

The effect of hyperoxygenation with carbogen (95% O(2) + 5% CO(2)) inhalation on RIF-1 tumor pO(2 )and its consequence on growth inhibition with fractionated radiotherapy is reported. The temporal changes in the tumor pO(2) were assessed by in vivo Electron Paramagnetic Resonance (EPR) oximetry in mice breathing 30% O(2) or carbogen and the tumors were irradiated with 4 Gy/day for 5 consecutive days; a protocol that emulates the clinical application of carbogen. The RIF-1 tumors were hypoxic with a tissue pO(2) of 5-9 mmHg. Carbogen (CB) breathing significantly increased tumor pO(2), with a maximum increase at 22.9-31.2 min on days 1-5, however, the magnitude of increase in pO(2) declined on day 5. Radiotherapy during carbogen inhalation (CB/RT) resulted in a significant tumor growth inhibition from day 3 to day 6 as compared to 30%O(2)/RT and carbogen (CB/Sham RT) groups. The results provide unambiguous quantitative information on the effect of carbogen inhalation on tumor pO(2) over the course of 5 days. Tumor growth inhibition in the CB/RT group confirms that the tumor oxygenation with carbogen was radiobiologically significant. Repeated tumor pO(2) measurements by EPR oximetry can provide temporal information that could be used to improve therapeutic outcomes by scheduling doses at times of improved tumor oxygenation.


Assuntos
Sarcoma Experimental/terapia , Animais , Dióxido de Carbono/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Hipóxia/metabolismo , Hipóxia/radioterapia , Hipóxia/terapia , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Induzidas por Radiação/metabolismo , Neoplasias Induzidas por Radiação/radioterapia , Neoplasias Induzidas por Radiação/terapia , Oximetria , Oxigênio/administração & dosagem , Oxigênio/metabolismo , Tolerância a Radiação , Radiossensibilizantes/administração & dosagem , Sarcoma Experimental/metabolismo , Sarcoma Experimental/radioterapia
20.
J Exp Clin Cancer Res ; 30: 24, 2011 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-21352555

RESUMO

BACKGROUND: The immune factors heat shock protein (HSP)/peptides (HSP/Ps) can induce both adaptive and innate immune responses. Treatment with HSP/Ps in cancer cell-bearing mice and cancer patients revealed antitumor immune activity. We aimed to develop immunotherapy strategies by vaccination with a mixture of HSP/Ps (mHSP/Ps, HSP60, HSP70, Gp96 and HSP110) enhanced with cyclophosphamide (CY) and interleukin-12 (IL-12). METHODS: We extracted mHSP/Ps from the mouse sarcoma cell line S180 using chromatography. The identity of proteins in this mHSP/Ps was assayed using SDS-PAGE and Western blot analysis with antibodies specific to various HSPs. BALB/C mice bearing S180 cells were vaccinated with mHSP/Ps ×3, then were injected intraperitoneally with low-dose CY and subcutaneously with IL-12, 100 µg/day, ×5. After vaccination, T lymphocytes in the peripheral blood were analyzed using FACScan and Cytotoxicity (CTL) was analyzed using lactate dehydrogenase assay. ELISPOT assay was used to evaluate interferon γ (IFN-γ), and immune cell infiltration in tumors was examined in the sections of tumor specimen. RESULTS: In mice vaccinated with enhanced vaccine (mHSP/Ps and CY plus IL-12), 80% showed tumor regression and long-term survival, and tumor growth inhibition rate was 82.3% (30 days), all controls died within 40 days. After vaccination, lymphocytes and polymorphonuclear leukocytes infiltrated into the tumors of treated animals, but no leukocytes infiltrated into the tumors of control mice. The proportions of natural killer cells, CD8+, and interferon-γ-secreting cells were all increased in the immune group, and tumor-specific cytotoxic T lymphocyte activity was increased. CONCLUSIONS: In this mice tumor model, vaccination with mHSP/Ps combined with low-dose CY plus IL-12 induced an immunologic response and a marked antitumor response to autologous tumors. The regimen may be a promising therapeutic agent against tumors.


Assuntos
Protocolos Antineoplásicos , Vacinas Anticâncer/uso terapêutico , Ciclofosfamida/uso terapêutico , Proteínas de Choque Térmico/uso terapêutico , Imunoterapia/métodos , Interleucina-12/uso terapêutico , Sarcoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Feminino , Proteínas de Choque Térmico/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Sarcoma Experimental/imunologia , Linfócitos T/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA