Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Biol Ther ; 24(1): 2269637, 2023 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-37878417

RESUMO

Targeted monoclonal antibody therapy has emerged as a powerful therapeutic strategy for cancer. However, only a minority of patients have durable responses and the development of resistance remains a major clinical obstacle. Antibody-dependent cell-mediated cytotoxicity (ADCC) represents a crucial therapeutic mechanism of action; however, few studies have explored ADCC resistance. Using multiple in vitro models of ADCC selection pressure, we have uncovered both shared and distinct resistance mechanisms. Persistent ADCC selection pressure yielded ADCC-resistant cells that are characterized by a loss of NK cell conjugation and this shared resistance phenotype is associated with cell-line dependent modulation of cell surface proteins that contribute to immune synapse formation and NK cell function. We employed single-cell RNA sequencing and proteomic screens to interrogate molecular mechanisms of resistance. We demonstrate that ADCC resistance involves upregulation of interferon/STAT1 and DNA damage response signaling as well as activation of the immunoproteasome. Here, we identify pathways that modulate ADCC sensitivity and report strategies to enhance ADCC-mediated elimination of cancer cells. ADCC resistance could not be reversed with combinatorial treatment approaches. Hence, our findings indicate that tumor cells utilize multiple strategies to inhibit NK cell mediated-ADCC. Future research and development of NK cell-based immunotherapies must incorporate plans to address or potentially prevent the induction of resistance.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Proteômica , Humanos , Linhagem Celular Tumoral , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Células Matadoras Naturais
2.
Int J Mol Sci ; 24(10)2023 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-37240219

RESUMO

The tumor microenvironment regulates many aspects of cancer progression and anti-tumor immunity. Cancer cells employ a variety of immunosuppressive mechanisms to dampen immune cell function in the tumor microenvironment. While immunotherapies that target these mechanisms, such as immune checkpoint blockade, have had notable clinical success, resistance is common, and there is an urgent need to identify additional targets. Extracellular adenosine, a metabolite of ATP, is found at high levels in the tumor microenvironment and has potent immunosuppressive properties. Targeting members of the adenosine signaling pathway represents a promising immunotherapeutic modality that can potentially synergize with conventional anti-cancer treatment strategies. In this review, we discuss the role of adenosine in cancer, present preclinical and clinical data on the efficacy adenosine pathway inhibition, and discuss possible combinatorial approaches.


Assuntos
Adenosina , Neoplasias , Humanos , Adenosina/metabolismo , Imunoterapia , Neoplasias/metabolismo , Imunossupressores/uso terapêutico , Transdução de Sinais , Microambiente Tumoral
3.
Transplant Cell Ther ; 28(10): 650-656, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35788086

RESUMO

The advent of chimeric antigen receptor (CAR) engineering has led to the development of powerful cellular therapies for cancer. CAR T cell-based treatments have had notable clinical success, but logistical issues and associated toxicities are recognized limitations. There is emerging interest in using other immune effector cell types for CAR therapy. Natural killer (NK) cells are part of the innate immune system, and these lymphocytes play major roles in immunosurveillance and antitumor immune responses. Incorporating CARs into NK cells provides the opportunity to harness and enhance their innate cytotoxic potential toward malignancies. In this review, we discuss the production of CAR-engineered NK cells, highlight data on their preclinical and clinical efficacy, and examine the obstacles and strategies to overcome them.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Imunoterapia , Imunoterapia Adotiva , Células Matadoras Naturais , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T
4.
Antibodies (Basel) ; 9(3)2020 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-32698317

RESUMO

Monoclonal antibody-based immunotherapy is now considered to be a main component of cancer therapy, alongside surgery, radiation, and chemotherapy. Monoclonal antibodies possess a diverse set of clinically relevant mechanisms of action. In addition, antibodies can directly target tumor cells while simultaneously promoting the induction of long-lasting anti-tumor immune responses. The multifaceted properties of antibodies as a therapeutic platform have led to the development of new cancer treatment strategies that will have major impacts on cancer care. This review focuses on the known mechanisms of action, current clinical applications for the treatment of cancer, and mechanisms of resistance of monoclonal antibody therapy. We further discuss how monoclonal antibody-based strategies have moved towards enhancing anti-tumor immune responses by targeting immune cells instead of tumor antigens as well as some of the current combination therapies.

5.
Semin Cancer Biol ; 65: 28-37, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31866479

RESUMO

Traditional cancer treatment approaches have focused on surgery, radiation therapy, and cytotoxic chemotherapy. However, with rare exceptions, metastatic cancers were considered to be incurable by traditional therapy. Over the past 20 years a fourth modality - immunotherapy - has emerged as a potentially curative approach for patients with advanced metastatic cancer. However, in many patients cancer "finds a way" to evade the anti-tumor effects of immunotherapy. Immunotherapy resistance mechanisms can be employed by both cancer cells and the non-cancer elements of tumor microenvironment. This review focuses on the resistance mechanisms that are specifically mediated by cancer cells. In order to extend the impact of immunotherapy to more patients and across all cancer types, and to inhibit the development of acquired resistance, the underlying biology driving immune escape needs to be better understood. Elucidating mechanisms of immune escape may shed light on new therapeutic targets, and lead to successful combination therapeutic strategies.


Assuntos
Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/efeitos adversos , Neoplasias/tratamento farmacológico , Microambiente Tumoral/imunologia , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Microambiente Tumoral/efeitos dos fármacos
6.
Prog Mol Biol Transl Sci ; 164: 61-100, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31383409

RESUMO

The immune system plays a key role in the interactions between host and tumor. Immune selection pressure is a driving force behind the sculpting and evolution of malignant cancer cells to escape this immune attack. Several common tumor cell-based mechanisms of resistance to immune attack have been identified and can be broadly categorized into three main classes: loss of antigenicity, loss of immunogenicity, and creation of an immunosuppressive microenvironment. In this review, we will discuss in detail the relevant literature associated with each class of resistance and will describe the relevance of these mechanisms to human cancer patients. To conclude, we will outline the implications these mechanisms have for the treatment of cancer using currently available therapeutic approaches. Immunotherapy has been a successful addition to current treatment approaches, but many patients either do not respond or quickly become resistant. This reflects the ability of tumors to continue to adapt to immune selection pressure at all stages of development. Additional study of immune escape mechanisms and immunotherapy resistance mechanisms will be needed to inform future treatment approaches.


Assuntos
Neoplasias/imunologia , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Regulação para Baixo , Humanos , Terapia de Imunossupressão , Microambiente Tumoral/imunologia
7.
Int J Mol Sci ; 20(1)2019 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-30621125

RESUMO

Immune checkpoint blockade therapy is a powerful treatment strategy for many cancer types. Many patients will have limited responses to monotherapy targeted to a single immune checkpoint. Both inhibitory and stimulatory immune checkpoints continue to be discovered. Additionally, many receptors previously identified to play a role in tumor formation and progression are being found to have immunomodulatory components. The success of immunotherapy depends on maximizing pro-anti-tumor immunity while minimizing immunosuppressive signaling. Combining immune checkpoint targeted approaches with each other or with other receptor targets is a promising schema for future therapeutic regimen designs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Imunomodulação , Ipilimumab/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Nivolumabe/uso terapêutico , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Quimioterapia Combinada , Humanos , Ipilimumab/imunologia , Nivolumabe/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Resultado do Tratamento
8.
Cancer Immunol Res ; 7(2): 230-243, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30563830

RESUMO

Targeted monoclonal antibody therapy is a promising therapeutic strategy for cancer, and antibody-dependent cell-mediated cytotoxicity (ADCC) represents a crucial mechanism underlying these approaches. The majority of patients have limited responses to monoclonal antibody therapy due to the development of resistance. Models of ADCC provide a system for uncovering immune-resistance mechanisms. We continuously exposed epidermal growth factor receptor (EGFR+) A431 cells to KIR-deficient NK92-CD16V effector cells and the anti-EGFR cetuximab. Persistent ADCC exposure yielded ADCC-resistant cells (ADCCR1) that, compared with control ADCC-sensitive cells (ADCCS1), exhibited reduced EGFR expression, overexpression of histone- and interferon-related genes, and a failure to activate NK cells, without evidence of epithelial-to-mesenchymal transition. These properties gradually reversed following withdrawal of ADCC selection pressure. The development of resistance was associated with lower expression of multiple cell-surface molecules that contribute to cell-cell interactions and immune synapse formation. Classic immune checkpoints did not modulate ADCC in this unique model system of immune resistance. We showed that the induction of ADCC resistance involves genetic and epigenetic changes that lead to a general loss of target cell adhesion properties that are required for the establishment of an immune synapse, killer cell activation, and target cell cytotoxicity.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Modelos Biológicos , Animais , Citotoxicidade Celular Dependente de Anticorpos/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Xenoenxertos , Histonas/metabolismo , Humanos , Interferons/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Proteoma , Proteômica/métodos
9.
Antib Ther ; 1(1): 7-12, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33928217

RESUMO

The targeting of surface antigens expressed on tumor cells by monoclonal antibodies (mAbs) has revolutionized cancer therapeutics. One mechanism of action of antibody-based immunotherapy is the activation of immune effector cells to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). This review will summarize the process of ADCC, its important role in the efficacy of mAb therapy, how to measure it, and finally future strategies for antibody design that can take advantage of it to improve clinical performance.

10.
Pediatr Radiol ; 47(2): 221-226, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27812743

RESUMO

BACKGROUND: Glomerular hyperfiltration has recently been reported in children with malignancies and has been attributed to increased solute from breakdown of tumor tissues. OBJECTIVE: To evaluate the prevalence of hyperfiltration in the pediatric oncology population and explore its pathophysiological mechanism. MATERIALS AND METHODS: Tc-99 m diethylenetriaminepentaacetic acid (DTPA) glomerular filtration rate (GFR) examinations (437 studies) and medical records of 177 patients <21 years of age diagnosed with a malignancy between January 2005 and October 2013 were retrospectively reviewed. Hyperfiltration was defined as a GFR ≥ 160 ml/min/1.73 m2. RESULTS: Seventy-seven (43.5%) patients had hyperfiltration in at least one GFR exam. A significantly higher percentage of patients with central nervous system (CNS) tumors (63.6%) had hyperfiltration when compared to other tumor types (27.3%, P < 0.001). No association was found between hyperfiltration and age, gender, race or bone marrow involvement. There was a significant trend toward decreasing hyperfiltration after the second cycle of chemotherapy (P = 0.006) and a significant increase in subjects with low GFR (<100 ml/min/1.73 m2) with increasing number of cycles of chemotherapy (P = 0.005). CONCLUSION: Glomerular hyperfiltration is common in children with malignancies at diagnosis and during initial cycles of chemotherapy. It is particularly prevalent in patients with central nervous tumors, which are frequently smaller in volume. Therefore, the pathophysiological mechanism of hyperfiltration cannot be explained solely on the basis of large tumor volume and subsequent cell breakdown. We hypothesize that host hypermetabolic state plays an important role in pathophysiology of hyperfiltration.


Assuntos
Taxa de Filtração Glomerular , Nefropatias/fisiopatologia , Neoplasias/fisiopatologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neoplasias/tratamento farmacológico , Prevalência , Estudos Retrospectivos , Fatores de Risco , Pentetato de Tecnécio Tc 99m/análogos & derivados
11.
Virchows Arch ; 469(3): 345-50, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27271275

RESUMO

Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm. Approximately 50 % of IMTs show an anaplastic lymphoma kinase (ALK) gene fusion resulting in ALK overexpression on immunohistochemistry (IHC). A novel anti-ALK monoclonal antibody (D5F3) has been suggested to be of superior sensitivity to the ALK1 antibody which is currently used. We compared the performance of D5F3 in detecting ALK protein expression in IMTs from various anatomic sites compared to the currently utilized ALK1. We selected 25 IMTs from our surgical pathology files (2005-2015). The novel rabbit monoclonal anti-human CD246 (clone D5F3) and the currently used mouse monoclonal anti-human CD246 (clone ALK1) were used for immunohistochemical staining (IHC) in an automated slide stainer. The percentage of immunoreactive tumor cells (0, <5 %, 5-50 %, >50 %) and cytoplasmic staining intensity (graded 0-3) were assessed and compared between the two antibodies. Fluorescence in situ hybridization (FISH) studies for ALK gene rearrangement were performed on 11 tumors. D5F3 antibody stained 76 % and ALK1 antibody stained 72 % of IMTs (p = 0.747). Compared to staining with ALK1, D5F3 stained a higher proportion of cases extensively (>50 % cells) (76 vs. 28 %, p < 0.001) and with high intensity (grade 3 76 % vs 0; p < 0.001). FISH and IHC findings (for both antibodies) were concordant in 9/10 (90 %) IMTs, in which results were informative. The novel anti-ALK rabbit monoclonal antibody (D5F3 clone) demonstrates superior overall performance in term of intensity and extent of staining of ALK protein in IMT. We found IHC staining with both antibody clones to correlate equally well with FISH results for detection of ALK rearrangement.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/metabolismo , Receptores Proteína Tirosina Quinases/imunologia , Adulto , Quinase do Linfoma Anaplásico , Anticorpos/imunologia , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Feminino , Fusão Gênica/genética , Rearranjo Gênico/genética , Humanos , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente/métodos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Receptores Proteína Tirosina Quinases/metabolismo
12.
Hum Pathol ; 54: 184-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27068523

RESUMO

Solitary fibrous tumor (SFT) diagnosis in prostate can be challenging on small biopsies. Prostatic stromal tumors of unknown malignant potential (STUMP) and SFT have overlapping features. NAB2-STAT6 gene fusions that were recently identified in various SFTs lead to nuclear translocalization of STAT6. Nuclear STAT6 immunostaining is now considered an adjunct for SFT diagnosis. We evaluated STAT6 and an emerging stemness marker, ALDH1, in the differential diagnosis of SFT versus prostatic stromal lesions. Sixteen STUMPs, 12 SFTs, and 4 prostatic stromal sarcomas (12 needle biopsies, 13 radical prostatectomies, 7 transurethral resections) were retrieved (1995-2015). Sections were stained with polyclonal STAT6 antibody (Santa Cruz Biotechnology, Santa Cruz, CA; S20, 1:100) and monoclonal ALDH1 antibody (BD Biosciences, San Jose, CA; clone 44, 1:250). In STAT6 cases, only unequivocal nuclear staining (with/without cytoplasmic staining) was considered positive. Cytoplasmic ALDH1 staining was counted positive. Ten of 11 evaluable SFTs demonstrated strong and diffuse nuclear STAT6 positivity; 4 of 16 STUMPs had nuclear staining that was weak (1/4) or focal (1/4). ALDH1 positivity was seen in 10 of 12 evaluable SFTs and 3 of 15 STUMPs. Prostatic stromal sarcomas were STAT6 negative (4/4); 2 of 4 were ALDH1 positive. The sensitivity and specificity for STAT6 for the diagnosis of SFT were 91% and 75%, respectively. Coexpression of STAT6 and ALDH1 yielded the same sensitivity but improved the specificity (100%) for the diagnosis of SFT. STAT6 is a useful marker in the differential diagnosis of SFT versus STUMP. Using STAT6 and ALDH1 together increases specificity. STUMPs can show STAT6 positivity, and when they do, it is likely to be weak or focal.


Assuntos
Biomarcadores Tumorais/análise , Imuno-Histoquímica , Isoenzimas/análise , Neoplasias da Próstata/enzimologia , Retinal Desidrogenase/análise , Fator de Transcrição STAT6/análise , Sarcoma/enzimologia , Tumores Fibrosos Solitários/enzimologia , Células Estromais/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Família Aldeído Desidrogenase 1 , Diagnóstico Diferencial , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Neoplasias da Próstata/patologia , Reprodutibilidade dos Testes , Sarcoma/patologia , Tumores Fibrosos Solitários/patologia , Células Estromais/patologia
13.
Exp Cell Res ; 315(11): 1850-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19362550

RESUMO

The majority of human tumors are angiogenesis dependent. Understanding the specific mechanisms that contribute to angiogenesis may offer the best approach to develop therapies to inhibit angiogenesis in cancer. Endothelial monocyte activating polypeptide-II (EMAP-II) is an anti-angiogenic cytokine with potent effects on endothelial cells (ECs). It inhibits EC proliferation and cord formation, and it suppresses primary and metastatic tumor growth in-vivo. However, very little is known about the molecular mechanisms behind the anti-angiogenic activity of EMAP-II. In the present study, we explored the molecular mechanism behind the anti-angiogenic activity exerted by this protein on ECs. Our results demonstrate that EMAP-II binds to the cell surface alpha5beta1 integrin receptor. The cell surface binding of EMAP-II results in its internalization into the cytoplasmic compartment where it interacts with its cytoplasmic partner PSMA7, a component of the proteasome degradation pathway. This interaction increases hypoxia-inducible factor 1-alpha (HIF-1alpha) degradation under hypoxic conditions. The degradation results in the inhibition of HIF-1alpha mediated transcriptional activity as well as HIF-1alpha mediated angiogenic sprouting of ECs. HIF-1alpha plays a critical role in angiogenesis by activating a variety of angiogenic growth factors. Our results suggest that one of the major anti-angiogenic functions of EMAP-II is exerted through its inhibition of the HIF-1alpha activities.


Assuntos
Citocinas/metabolismo , Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Bases , Sítios de Ligação , Hipóxia Celular , Linhagem Celular , Células Cultivadas , Citocinas/genética , Citoplasma/metabolismo , Células Endoteliais/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Integrina alfa5beta1/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/genética , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica , Complexo de Endopeptidases do Proteassoma/genética , Inibidores de Proteassoma , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA