Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 161(2): 201-4, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25860604

RESUMO

Although cancer immunotherapy can lead to durable outcomes, the percentage of patients who respond to this disruptive approach remains modest to date. Encouragingly, nanotechnology can enhance the efficacy of immunostimulatory small molecules and biologics by altering their co-localization, biodistribution, and release kinetics.


Assuntos
Vacinas Anticâncer/uso terapêutico , Imunoterapia , Nanotecnologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Células Dendríticas/imunologia , Humanos , Linfócitos T/imunologia
2.
Cancer Immunol Immunother ; 65(7): 787-96, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26646852

RESUMO

Immunotherapy confers durable clinical benefit to melanoma, lung, and kidney cancer patients. Challengingly, most other solid tumors, including ovarian carcinoma, are not particularly responsive to immunotherapy, so combination with a complementary therapy may be beneficial. Recent findings suggest that epigenetic modifying drugs can prime antitumor immunity by increasing expression of tumor-associated antigens, chemokines, and activating ligands by cancer cells as well as cytokines by immune cells. This review, drawing from both preclinical and clinical data, describes some of the mechanisms of action that enable DNA methyltransferase inhibitors to facilitate the establishment of antitumor immunity.


Assuntos
Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Animais , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/imunologia , Metilases de Modificação do DNA/imunologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Neoplasias/enzimologia , Neoplasias/genética
3.
Biochem Biophys Res Commun ; 463(4): 551-6, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26047697

RESUMO

Familial breast and ovarian cancer are often caused by inherited mutations of BRCA1. While current prognoses for such patients are rather poor, inhibition of poly-ADP ribose polymerase 1 (PARP1) induces synthetic lethality in cells that are defective in homologous recombination. BMN 673 is a potent PARP1 inhibitor that is being clinically evaluated for treatment of BRCA-mutant cancers. Using the Brca1-deficient murine epithelial ovarian cancer cell line BR5FVB1-Akt, we investigated whether the antitumor effects of BMN 673 extend beyond its known pro-apoptotic function. Administration of modest amounts of BMN 673 greatly improved the survival of mice bearing subcutaneous or intraperitoneal tumors. We thus hypothesized that BMN 673 may influence the composition and function of immune cells in the tumor microenvironment. Indeed, BMN 673 significantly increases the number of peritoneal CD8(+) T cells and NK cells as well as their production of IFN-γ and TNF-α. These data suggest that the cell stress caused by BMN 673 induces not only cancer cell-intrinsic apoptosis but also cancer cell-extrinsic antitumor immune effects in a syngeneic murine model of ovarian cancer. BMN 673 may therefore serve as a promising adjuvant therapy to immunotherapy to achieve durable responses among patients whose tumors harbor defects in homologous recombination.


Assuntos
Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Genes BRCA1 , Neoplasias Ovarianas/imunologia , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Feminino , Citometria de Fluxo , Camundongos , Camundongos Knockout , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Poli(ADP-Ribose) Polimerase-1 , Reação em Cadeia da Polimerase em Tempo Real
4.
Biomacromolecules ; 15(11): 4187-94, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25251833

RESUMO

Development of drug resistance is a central challenge to the treatment of ovarian cancer. Metronomic chemotherapy decreases the extent of drug-free periods, thereby hindering development of drug resistance. Intraperitoneal chemotherapy allows for treatment of tumors confined within the peritoneum, but achieving sustained tumor-localized chemotherapy remains difficult. We hypothesized that modulating the surface properties of poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles could enhance their drug retention ability and extend their release profile, thereby enabling metronomic, localized chemotherapy in vivo. Paclitaxel was encapsulated in particles coated with a layer of polydopamine and a subsequent layer of poly(ethylene glycol) (PEG). These particles achieved a 3.8-fold higher loading content compared to that of nanoparticles formulated from linear PLGA-PEG copolymers. In vitro release kinetic studies and in vivo drug distribution profiles demonstrate sustained release of paclitaxel. Although free drug conferred no survival advantage, low-dose intraperitoneal administration of paclitaxel-laden surface-coated nanoparticles to drug-resistant ovarian tumor-bearing mice resulted in significant survival benefits in the absence of any apparent systemic toxicity.


Assuntos
Administração Metronômica , Modelos Animais de Doenças , Ácido Láctico/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/administração & dosagem , Ácido Poliglicólico/administração & dosagem , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Humanos , Ácido Láctico/química , Camundongos , Nanopartículas/química , Neoplasias Ovarianas/patologia , Paclitaxel/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Propriedades de Superfície
5.
Methods ; 63(2): 95-100, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23403216

RESUMO

Short interfering RNAs (siRNAs) mediate the catalytic sequence-specific cleavage of target messenger RNA (mRNA) molecules, resulting in the silencing of gene products in an efficient and precise manner. One apparent application of this technology is the knockdown of genes responsible for cancer progression, including pro-proliferative oncogenes, inhibitors of apoptosis, and mediators of angiogenesis. Delivery of siRNAs into particular cells has remained the principal obstacle to the realization of the potential of RNA interference (RNAi) in the clinic. Several groups have worked to develop carriers that facilitate siRNA delivery into ovarian cancer cells in mouse models of ovarian cancer. The results have been promising, often leading to significant survival extension. Such benefit is critical for a disease that is characterized by very poor outcomes and demands novel treatment options. This review describes advancements in siRNA delivery for the treatment of ovarian cancer.


Assuntos
Terapia Genética , Neoplasias Ovarianas/terapia , RNA Interferente Pequeno/genética , Animais , Feminino , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Humanos , Injeções Intraperitoneais , Lipossomos , Neoplasias Ovarianas/genética , Interferência de RNA
6.
Proc Natl Acad Sci U S A ; 108(2): 745-50, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187397

RESUMO

Inhibition of the DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP1) with small molecules has been shown to be an effective treatment for ovarian cancer with BRCA mutations. Here, we report the in vivo administration of siRNA to Parp1 in mouse models of ovarian cancer. A unique member of the lipid-like materials known as lipidoids is shown to deliver siRNA to disseminated murine ovarian carcinoma allograft tumors following intraperitoneal (i.p.) injection. siParp1 inhibits cell growth, primarily by induction of apoptosis, in Brca1-deficient cells both in vitro and in vivo. Additionally, the treatment extends the survival of mice bearing tumors derived from Brca1-deficient ovarian cancer cells but not from Brca1 wild-type cells, confirming the proposed mechanism of synthetic lethality. Because there are 17 members of the Parp family, the inherent complementarity of RNA affords a high level of specificity for therapeutically addressing Parp1 in the context of impaired homologous recombination.


Assuntos
Proteína BRCA1/genética , Nanopartículas/química , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Poli(ADP-Ribose) Polimerases/genética , RNA Interferente Pequeno/metabolismo , Animais , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Nanotecnologia/métodos , Transplante de Neoplasias , Interferência de RNA , Resultado do Tratamento
7.
Nano Lett ; 12(6): 2685-9, 2012 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-22432731

RESUMO

The development of responsive nanomaterials, nanoscale systems that actively respond to stimuli, is one general goal of nanotechnology. Here we develop nanoparticles that can be controllably triggered to synthesize proteins. The nanoparticles consist of lipid vesicles filled with the cellular machinery responsible for transcription and translation, including amino acids, ribosomes, and DNA caged with a photolabile protecting group. These particles served as nanofactories capable of producing proteins including green fluorescent protein (GFP) and enzymatically active luciferase. In vitro and in vivo, protein synthesis was spatially and temporally controllable, and could be initiated by irradiating micrometer-scale regions on the time scale of milliseconds. The ability to control protein synthesis inside nanomaterials may enable new strategies to facilitate the study of orthogonal proteins in a confined environment and for remotely activated drug delivery.


Assuntos
Cristalização/métodos , Nanoestruturas/química , Nanoestruturas/ultraestrutura , Engenharia de Proteínas/métodos , Proteínas/síntese química , Robótica/métodos , Teste de Materiais , Tamanho da Partícula , Conformação Proteica , Propriedades de Superfície
8.
Nat Rev Cancer ; 19(10): 587-602, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31492927

RESUMO

The 2018 Nobel Prize in Physiology or Medicine was awarded to pioneers in the field of cancer immunotherapy, as the utility of leveraging a patient's coordinated and adaptive immune system to fight the patient's unique tumour has now been validated robustly in the clinic. Still, the proportion of patients who respond to immunotherapy remains modest (~15% objective response rate across indications), as tumours have multiple means of immune evasion. The immune system is spatiotemporally controlled, so therapies that influence the immune system should be spatiotemporally controlled as well, in order to maximize the therapeutic index. Nanoparticles and biomaterials enable one to program the location, pharmacokinetics and co-delivery of immunomodulatory compounds, eliciting responses that cannot be achieved upon administration of such compounds in solution. The convergence of cancer immunotherapy, nanotechnology, bioengineering and drug delivery is opportune, as each of these fields has matured independently to the point that it can now be used to complement the others substantively and rationally, rather than modestly and empirically. As a result, unmet needs increasingly can be addressed with deductive intention. This Review explores how nanotechnology and related approaches are being applied to augmenting both endogenous leukocytes and adoptively transferred ones by informing specificity, influencing localization and improving function.


Assuntos
Imunoterapia/métodos , Nanomedicina/métodos , Neoplasias/imunologia , Neoplasias/terapia , Transferência Adotiva , Animais , Materiais Biocompatíveis/química , Sistemas de Liberação de Medicamentos , Humanos , Sistema Imunitário , Fatores Imunológicos/metabolismo , Leucócitos/citologia , Nanopartículas/química , Ácidos Nucleicos/metabolismo , Linfócitos T/citologia
9.
iScience ; 12: 369-378, 2019 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-30769282

RESUMO

Vaccination approaches have generally focused on the antigen rather than the resultant antibodies generated, which differ greatly in quality and function between individuals. The ability to replace the variable regions of the native B cell receptor (BCR) heavy and light chain loci with defined recombined sequences of a preferred monoclonal antibody could enable curative adoptive cell transfer. We report CRISPR-mediated homologous recombination (HR) into the BCR of primary human B cells. Ribonucleoprotein delivery enabled editing at the model CXCR4 locus, as demonstrated by T7E1 assay, flow cytometry, and TIDE analysis. Insertion via HR was confirmed by sequencing, cross-boundary PCR, and restriction digest. Optimized conditions were used to achieve HR at the BCR variable heavy and light chains. Insertion was confirmed at the DNA level, and transgene expression from the native BCR promoters was observed. Reprogramming the specificity of antibodies in the genomes of B cells could have clinical importance.

10.
J Immunother Cancer ; 7(1): 199, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31362778

RESUMO

BACKGROUND: Despite major advancements in immunotherapy among a number of solid tumors, response rates among ovarian cancer patients remain modest. Standard treatment for ovarian cancer is still surgery followed by taxane- and platinum-based chemotherapy. Thus, there is an urgent need to develop novel treatment options for clinical translation. METHODS: Our approach was to analyze the effects of standard chemotherapy in the tumor microenvironment of mice harboring orthotopic, syngeneic ID8-Vegf-Defb29 ovarian tumors in order to mechanistically determine a complementary immunotherapy combination. Specifically, we interrogated the molecular and cellular consequences of chemotherapy by analyzing gene expression and flow cytometry data. RESULTS: These data show that there is an immunosuppressive shift in the myeloid compartment, with increased expression of IL-10 and ARG1, but no activation of CD3+ T cells shortly after chemotherapy treatment. We therefore selected immunotherapies that target both the innate and adaptive arms of the immune system. Survival studies revealed that standard chemotherapy was complemented most effectively by a combination of anti-IL-10, 2'3'-cGAMP, and anti-PD-L1. Immunotherapy dramatically decreased the immunosuppressive myeloid population while chemotherapy effectively activated dendritic cells. Together, combination treatment increased the number of activated T and dendritic cells as well as expression of cytotoxic factors. It was also determined that the immunotherapy had to be administered concurrently with the chemotherapy to reverse the acute immunosuppression caused by chemotherapy. Mechanistic studies revealed that antitumor immunity in this context was driven by CD4+ T cells, which acquired a highly activated phenotype. Our data suggest that these CD4+ T cells can kill cancer cells directly via granzyme B-mediated cytotoxicity. Finally, we showed that this combination therapy is also effective at delaying tumor growth substantially in an aggressive model of lung cancer, which is also treated clinically with taxane- and platinum-based chemotherapy. CONCLUSIONS: This work highlights the importance of CD4+ T cells in tumor immunology. Furthermore, the data support the initiation of clinical trials in ovarian cancer that target both innate and adaptive immunity, with a focus on optimizing dosing schedules.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Antineoplásicos Imunológicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Perfilação da Expressão Gênica/métodos , Imunidade Inata/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos Imunológicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Carboplatina/administração & dosagem , Carboplatina/farmacologia , Terapia Combinada , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-10/antagonistas & inibidores , Camundongos , Terapia de Alvo Molecular , Nucleotídeos Cíclicos/administração & dosagem , Nucleotídeos Cíclicos/farmacologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/imunologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Análise de Sobrevida , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Sci Transl Med ; 10(433)2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29563317

RESUMO

Cancer immunotherapy can confer durable benefit, but the percentage of patients who respond to this approach remains modest. The ability to concentrate immunostimulatory compounds at the site of disease can overcome local immune tolerance and reduce systemic toxicity. Surgical resection of tumors may improve the efficacy of immunotherapy by removing the concentrated immunosuppressive microenvironment; however, it also removes tumor-specific leukocytes as well as tumor antigens that may be important to establishing antitumor immunity. Moreover, surgery produces a transient immunosuppressive state associated with wound healing that has been correlated with increased metastasis. Using multiple models of spontaneous metastasis, we show that extended release of agonists of innate immunity-including agonists of Toll-like receptor 7/8 (TLR7/8) or stimulator of interferon genes (STING)-from a biodegradable hydrogel placed in the tumor resection site cured a much higher percentage of animals than systemic or local administration of the same therapy in solution. Depletion and neutralization experiments confirmed that the observed prevention of local tumor recurrence and eradication of existing metastases require both the innate and adaptive arms of the immune system. The localized therapy increased the numbers of activated natural killer (NK) cells, dendritic cells, and T cells and induced production of large amounts of type I interferons, thereby converting an immunosuppressive post-resection microenvironment into an immunostimulatory one. The results suggest that the perioperative setting may prove to be a useful context for immunotherapy, particularly when the release of the therapy is extended locally.


Assuntos
Imunidade Inata/imunologia , Imunoterapia/métodos , Metástase Neoplásica/prevenção & controle , Animais , Células Dendríticas/imunologia , Feminino , Hidrogel de Polietilenoglicol-Dimetacrilato , Células Matadoras Naturais/imunologia , Camundongos , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo
12.
Science ; 361(6409)2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30262472

RESUMO

Cancer cells from a primary tumor can disseminate to other tissues, remaining dormant and clinically undetectable for many years. Little is known about the cues that cause these dormant cells to awaken, resume proliferating, and develop into metastases. Studying mouse models, we found that sustained lung inflammation caused by tobacco smoke exposure or nasal instillation of lipopolysaccharide converted disseminated, dormant cancer cells to aggressively growing metastases. Sustained inflammation induced the formation of neutrophil extracellular traps (NETs), and these were required for awakening dormant cancer. Mechanistic analysis revealed that two NET-associated proteases, neutrophil elastase and matrix metalloproteinase 9, sequentially cleaved laminin. The proteolytically remodeled laminin induced proliferation of dormant cancer cells by activating integrin α3ß1 signaling. Antibodies against NET-remodeled laminin prevented awakening of dormant cells. Therapies aimed at preventing dormant cell awakening could potentially prolong the survival of cancer patients.


Assuntos
Carcinogênese/metabolismo , Armadilhas Extracelulares/enzimologia , Laminas/metabolismo , Neoplasias Pulmonares/patologia , Neutrófilos/enzimologia , Pneumonia/patologia , Animais , DNA/metabolismo , Humanos , Inflamação/induzido quimicamente , Inflamação/microbiologia , Integrina alfa3beta1/metabolismo , Elastase de Leucócito/metabolismo , Lipopolissacarídeos , Pulmão/patologia , Células MCF-7 , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/patologia , Pneumonia/induzido quimicamente , Pneumonia/microbiologia , Pneumonia Bacteriana/etiologia , Pneumonia Bacteriana/patologia , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas/antagonistas & inibidores , Desiminases de Arginina em Proteínas/metabolismo , Proteólise , Ratos , Transdução de Sinais , Fumar , Nicotiana
13.
Nanomedicine (Lond) ; 12(7): 729-743, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28322107

RESUMO

AIM: We aimed to enhance the stability and therapeutic efficiency of protein-based therapeutic formulations. MATERIALS & METHODS: Proteins were immobilized on the surface of nanoparticles (NPs) to improve both protein stability and protein function, especially enzymatic activity. The modularity of the platform was demonstrated by coating proteins of varied molecular weights and functionalities on the surface of poly(lactic-co-glycolic acid)-based NPs. RESULTS: Coating proteins to the particle surface greatly enhanced the stability of the NPs, preventing particle aggregation and improving enzymatic potency, including in vivo. Specifically, coating of collagenase I to the particle surface greatly improved the ability of the enzyme to degrade tumor collagen relative to free enzyme, thereby increasing the penetration of adjuvant chemotherapy (doxorubicin). Additionally, the protein coating reduced the rate of doxorubicin release, enabling sustained release of the small-molecule payload. CONCLUSION: The straightforward procedure described herein permits the formulation of modular NPs that can combine and sustain the benefits of small molecules and biologics.


Assuntos
Antineoplásicos/administração & dosagem , Colagenases/química , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Ácido Láctico/química , Nanopartículas/química , Ácido Poliglicólico/química , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/química , Complexo CD3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Quimioterapia Adjuvante , Colágeno/metabolismo , Doxorrubicina/química , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Indóis/química , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Polietilenoglicóis/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química , Propriedades de Superfície , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral
14.
Cancer Discov ; 7(7): 675-693, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28630051

RESUMO

DNA-damaging agents are widely used in clinical oncology and exploit deficiencies in tumor DNA repair. Given the expanding role of immune checkpoint blockade as a therapeutic strategy, the interaction of tumor DNA damage with the immune system has recently come into focus, and it is now clear that the tumor DNA repair landscape has an important role in driving response to immune checkpoint blockade. Here, we summarize the mechanisms by which DNA damage and genomic instability have been found to shape the antitumor immune response and describe clinical efforts to use DNA repair biomarkers to guide use of immune-directed therapies.Significance: Only a subset of patients respond to immune checkpoint blockade, and reliable predictive biomarkers of response are needed to guide therapy decisions. DNA repair deficiency is common among tumors, and emerging experimental and clinical evidence suggests that features of genomic instability are associated with response to immune-directed therapies. Cancer Discov; 7(7); 675-93. ©2017 AACR.


Assuntos
Biomarcadores Tumorais/genética , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Neoplasias/imunologia , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/imunologia , Dano ao DNA/imunologia , Reparo do DNA/imunologia , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/imunologia , Humanos , Imunoterapia/efeitos adversos , Neoplasias/genética , Neoplasias/terapia
15.
Nat Commun ; 8(1): 1747, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29170511

RESUMO

Targeted delivery of compounds to particular cell subsets can enhance therapeutic index by concentrating their action on the cells of interest. Because attempts to target tumors directly have yielded limited benefit, we instead target endogenous immune cell subsets in the circulation that can migrate actively into tumors. We describe antibody-targeted nanoparticles that bind to CD8+ T cells in the blood, lymphoid tissues, and tumors of mice. PD-1+ T cells are successfully targeted in the circulation and tumor. The delivery of an inhibitor of TGFß signaling to PD-1-expressing cells extends the survival of tumor-bearing mice, whereas free drugs have no effect at such doses. This modular platform also enables PD-1-targeted delivery of a TLR7/8 agonist to the tumor microenvironment, increasing the proportion of tumor-infiltrating CD8+ T cells and sensitizing tumors to subsequent anti-PD-1. Targeted delivery of immunotherapy to defined subsets of endogenous leukocytes may be superior to administration of free drugs.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoterapia/métodos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Imidazóis/administração & dosagem , Linfócitos do Interstício Tumoral/imunologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral/imunologia
16.
J Clin Invest ; 127(12): 4554-4568, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29130934

RESUMO

Transcriptional repression of ubiquitin B (UBB) is a cancer-subtype-specific alteration that occurs in a substantial population of patients with cancers of the female reproductive tract. UBB is 1 of 2 genes encoding for ubiquitin as a polyprotein consisting of multiple copies of ubiquitin monomers. Silencing of UBB reduces cellular UBB levels and results in an exquisite dependence on ubiquitin C (UBC), the second polyubiquitin gene. UBB is repressed in approximately 30% of high-grade serous ovarian cancer (HGSOC) patients and is a recurrent lesion in uterine carcinosarcoma and endometrial carcinoma. We identified ovarian tumor cell lines that retain UBB in a repressed state, used these cell lines to establish orthotopic ovarian tumors, and found that inducible expression of a UBC-targeting shRNA led to tumor regression, and substantial long-term survival benefit. Thus, we describe a recurrent cancer-specific lesion at the level of ubiquitin production. Moreover, these observations reveal the prognostic value of UBB repression and establish UBC as a promising therapeutic target for ovarian cancer patients with recurrent UBB silencing.


Assuntos
Inativação Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias Ovarianas/metabolismo , Ubiquitina C/biossíntese , Ubiquitina/biossíntese , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Ubiquitina/genética , Ubiquitina C/genética
17.
Sci Transl Med ; 8(361): 361ra138, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27798263

RESUMO

Neutrophils, the most abundant type of leukocytes in blood, can form neutrophil extracellular traps (NETs). These are pathogen-trapping structures generated by expulsion of the neutrophil's DNA with associated proteolytic enzymes. NETs produced by infection can promote cancer metastasis. We show that metastatic breast cancer cells can induce neutrophils to form metastasis-supporting NETs in the absence of infection. Using intravital imaging, we observed NET-like structures around metastatic 4T1 cancer cells that had reached the lungs of mice. We also found NETs in clinical samples of triple-negative human breast cancer. The formation of NETs stimulated the invasion and migration of breast cancer cells in vitro. Inhibiting NET formation or digesting NETs with deoxyribonuclease I (DNase I) blocked these processes. Treatment with NET-digesting, DNase I-coated nanoparticles markedly reduced lung metastases in mice. Our data suggest that induction of NETs by cancer cells is a previously unidentified metastasis-promoting tumor-host interaction and a potential therapeutic target.


Assuntos
Armadilhas Extracelulares , Metástase Neoplásica , Neutrófilos/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Desoxirribonuclease I/química , Humanos , Pulmão/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Neutrófilos/citologia
18.
Adv Drug Deliv Rev ; 91: 38-51, 2015 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-25280471

RESUMO

While nanoparticles have traditionally been used to deliver cytotoxic drugs directly to tumors to induce cancer cell death, emerging data suggest that nanoparticles are likely to generate a larger impact on oncology through the delivery of agents that can stimulate antitumor immunity. Tumor-targeted nanocarriers have generally been used to localize chemotherapeutics to tumors and thus decrease off-target toxicity while enhancing efficacy. Challengingly, tumor heterogeneity and evolution render tumor-intrinsic approaches likely to succumb to relapse. The immune system offers exquisite specificity, cytocidal potency, and long-term activity that leverage an adaptive memory response. For this reason, the ability to manipulate immune cell specificity and function would be desirable, and nanoparticles represent an exciting means by which to perform such manipulation. Dendritic cells and tumor-associated macrophages are cells of the myeloid lineage that function as natural phagocytes, so they naturally take up nanoparticles. Dendritic cells direct the specificity and potency of cellular immune responses that can be targeted for cancer vaccines. Herein, we discuss the specific criteria needed for efficient vaccine design, including but not limited to the route of administration, size, morphology, surface charge, targeting ligands, and nanoparticle composition. In contrast, tumor-associated macrophages are critical mediators of immunosuppression whose trans-migratory abilities can be exploited to localize therapeutics to the tumor core and which can be directly targeted for elimination or for repolarization to a tumor suppressive phenotype. It is likely that a combination of targeting dendritic cells to stimulate antitumor immunity and tumor-associated macrophages to reduce immune suppression will impart significant benefits and result in durable antitumor responses.


Assuntos
Imunoterapia/métodos , Células Mieloides/imunologia , Neoplasias/terapia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Sistemas de Liberação de Medicamentos , Humanos , Nanopartículas , Neoplasias/imunologia
19.
Mol Cancer Ther ; 14(7): 1521-31, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25964202

RESUMO

Tumor multidrug resistance (MDR) is a serious clinical challenge that significantly limits the effectiveness of cytotoxic chemotherapy. As such, complementary therapeutic strategies are being explored to prevent relapse. The altered metabolic state of cancer cells, which perform aerobic glycolysis, represents an interesting target that can enable discrimination between healthy cells and cancer cells. We hypothesized that cosilencing of genes responsible for aerobic glycolysis and for MDR would have synergistic antitumor effect. In this study, siRNA duplexes against pyruvate kinase M2 and multidrug resistance gene-1 were encapsulated in hyaluronic acid-based self-assembling nanoparticles. The particles were characterized for morphology, size, charge, encapsulation efficiency, and transfection efficiency. In vivo studies included biodistribution assessment, gene knockdown confirmation, therapeutic efficacy, and safety analysis. The benefit of active targeting of cancer cells was confirmed by modifying the particles' surface with a peptide targeted to epidermal growth factor receptor, which is overexpressed on the membranes of the SKOV-3 cancer cells. To augment the studies involving transplantation of a paclitaxel-resistant cell line, an in vivo paclitaxel resistance model was developed by injecting repeated doses of paclitaxel following tumor inoculation. The nanoparticles accumulated significantly in the tumors, hindering tumor volume doubling time (P < 0.05) upon combination therapy in both the wild-type (2-fold) and resistant (8-fold) xenograft models. Although previous studies indicated that silencing of MDR-1 alone sensitized MDR ovarian cancer to paclitaxel only modestly, these data suggest that concurrent silencing of PKM-2 improves the efficacy of paclitaxel against MDR ovarian cancer.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Neoplasias Ovarianas/terapia , Piruvato Quinase/genética , Terapêutica com RNAi/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Ácido Hialurônico/química , Camundongos Nus , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Paclitaxel/farmacologia , Piruvato Quinase/metabolismo , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacocinética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
20.
Cancer Immunol Res ; 3(9): 1030-41, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26056145

RESUMO

The lack of second-line treatment for relapsed ovarian cancer necessitates the development of improved combination therapies. Targeted therapy and immunotherapy each confer clinical benefit, albeit limited as monotherapies. Ovarian cancer is not particularly responsive to immune checkpoint blockade, so combination with a complementary therapy may be beneficial. Recent studies have revealed that a DNA methyl transferase inhibitor, azacytidine, alters expression of immunoregulatory genes in ovarian cancer. In this study, the antitumor effects of a related DNA methyl transferase inhibitor, decitabine (DAC), were demonstrated in a syngeneic murine ovarian cancer model. Low-dose DAC treatment increases the expression of chemokines that recruit NK cells and CD8(+) T cells, promotes their production of IFNγ and TNFα, and extends the survival of mice bearing subcutaneous or orthotopic tumors. While neither DAC nor immune checkpoint blockade confers durable responses as a monotherapy in this model, the efficacy of anti-CTLA-4 was potentiated by combination with DAC. This combination promotes differentiation of naïve T cells into effector T cells and prolongs cytotoxic lymphocyte responses as well as mouse survival. These results suggest that this combination therapy may be worthy of further consideration for improved treatment of drug-resistant ovarian cancer.


Assuntos
Antineoplásicos/uso terapêutico , Azacitidina/análogos & derivados , Antígeno CTLA-4/antagonistas & inibidores , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Líquido Ascítico/imunologia , Azacitidina/administração & dosagem , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Movimento Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/imunologia , Citocinas/biossíntese , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunoterapia/métodos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Neoplasias Ovarianas/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA