Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 329
Filtrar
1.
Cancer Res Commun ; 4(2): 505-515, 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38319147

RESUMO

In normal cells, binding of the transmembrane protein CD47 to signal regulatory protein-α (SIRPα) on macrophages induces an antiphagocytic signal. Tumor cells hijack this pathway and overexpress CD47 to evade immune destruction. Macrophage antitumor activity can be restored by simultaneously blocking the CD47-SIRPα signaling axis and inducing a prophagocytic signal via tumor-opsonizing antibodies. We identified a novel, fully human mAb (BMS-986351) that binds SIRPα with high affinity. BMS-986351 demonstrated broad binding coverage across SIRPα polymorphisms and potently blocked CD47-SIRPα binding at the CD47 binding site in a dose-dependent manner. In vitro, BMS-986351 increased phagocytic activity against cell lines from solid tumors and hematologic malignancies, and this effect was markedly enhanced when BMS-986351 was combined with the opsonizing antibodies cetuximab and rituximab. A phase I dose-escalation/-expansion study of BMS-986351 for the treatment of advanced solid and hematologic malignancies is underway (NCT03783403). SIGNIFICANCE: Increasing the phagocytotic capabilities of tumor-associated macrophages by modulating macrophage-tumor cell surface signaling via the CD47-SIRPα axis is a novel strategy. Molecules targeting CD47 have potential but its ubiquitous expression necessitates higher therapeutic doses to overcome potential antigen sink effects. The restricted expression pattern of SIRPα may limit toxicities and lower doses of the SIRPα antibody BMS-986351 may overcome target mediated drug disposition while maintaining the desired pharmacology.


Assuntos
Neoplasias Hematológicas , Neoplasias , Humanos , Antígeno CD47/genética , Receptores Imunológicos/genética , Fagocitose , Macrófagos , Neoplasias/tratamento farmacológico , Anticorpos Antineoplásicos/metabolismo , Proteínas Opsonizantes/metabolismo , Neoplasias Hematológicas/metabolismo
2.
Clin Exp Immunol ; 207(1): 84-94, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35020866

RESUMO

Human B cells and their expressed antibodies are crucial in conferring immune protection. Identifying pathogen-specific antibodies following infection is possible due to enhanced humoral immunity against well-described molecules on the pathogen surface. However, screening for cancer-reactive antibodies remains challenging since target antigens are often not identified a priori and the frequency of circulating B cells recognizing cancer cells is likely very low. We investigated whether combined ex vivo culture of human B cells with three innate stimuli, interleukin-17 (IL-17), B-cell activation factor (BAFF), and the toll-like receptor 9 (TLR-9) agonist DNA motif CpG ODN 2006 (CpG), each known to activate B cells through different signalling pathways, promote cell activation, proliferation, and antibody production. Combined IL-17+BAFF+CpG prolonged B-cell survival and increased proliferation compared with single stimuli. IL-17+BAFF+CpG triggered higher IgG secretion, likely by activating differentiated, memory and class-switched CD19+CD20+CD27+IgD- B cells. Regardless of anti-FOLR antibody seropositive status, IL-17+BAFF+CpG combined with a monovalent tumour-associated antigen (folate receptor alpha [FOLR]) led to secreted antibodies recognizing the antigen and the antigen-expressing IGROV1 cancer cells. In a seropositive individual, FOLR stimulation favoured class-switched memory B-cell precursors (CD27-CD38-IgD-), class-switched memory B cells and anti-FOLR antibody production, while IL-17+BAFF+CpG combined with FOLR, promoted class-switched memory B-cell precursors and antibody-secreting (CD138+IgD-) plasma cells. Furthermore, IL-17+BAFF+CpG stimulation of peripheral blood B cells from patients with melanoma revealed tumour cell-reactive antibodies in culture supernatants. These findings suggest that innate signals stimulate B-cell survival and antibody production and may help identify low-frequency antigen-reactive humoral responses.


Assuntos
Anticorpos Antineoplásicos , Neoplasias , Anticorpos Antineoplásicos/metabolismo , Formação de Anticorpos , Linfócitos B , Humanos , Ativação Linfocitária , Neoplasias/metabolismo
3.
Cells ; 10(11)2021 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-34831389

RESUMO

Immunotherapy targeting the PD-1-PD-L1 axis yielded good results in treating different immunologically ''hot'' tumors. A phase II study revealed good therapeutic activity of pembrolizumab in selected prostatic carcinoma (PC)-patients. We performed a systematic literature review (PRISMA guidelines), which analyzes the immunohistochemical expression of PD-L1 in human PC samples and highlights the pre-analytical and interpretation variables. Interestingly, 29% acinar PCs, 7% ductal PCs, and 46% neuroendocrine carcinomas/tumors were PD-L1+ on immunohistochemistry. Different scoring methods or cut-off criteria were applied on variable specimen-types, evaluating tumors showing different clinic-pathologic features. The positivity rate of different PD-L1 antibody clones in tumor cells ranged from 3% (SP142) to 50% (ABM4E54), excluding the single case tested for RM-320. The most tested clone was E1L3N, followed by 22C3 (most used for pembrolizumab eligibility), SP263, SP142, and 28-8, which gave the positivity rates of 35%, 11-41% (depending on different scoring systems), 6%, 3%, and 15%, respectively. Other clones were tested in <200 cases. The PD-L1 positivity rate was usually higher in tumors than benign tissues. It was higher in non-tissue microarray specimens (41-50% vs. 15%), as PC cells frequently showed heterogenous or focal PD-L1-staining. PD-L1 was expressed by immune or stromal cells in 12% and 69% cases, respectively. Tumor heterogeneity, inter-institutional preanalytics, and inter-observer interpretation variability may account for result biases.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Anticorpos Antineoplásicos/metabolismo , Humanos , Imuno-Histoquímica , Masculino
4.
Sci Rep ; 11(1): 12798, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34140620

RESUMO

The status of humoral immunity of cancer patients is not clear compared to cellular immunity because the ability of specific antibody production is difficult to analyze in vitro. We previously developed a humanized mouse model to evaluate antigen-specific antibody production by transplanting human peripheral blood mononuclear cells (PBMCs) into NOG-hIL-4-Tg mice (hu-PBL hIL-4 NOG). In this study, these mice were transplanted with PBMCs derived from breast cancer patients (BC) and immunized with a human epidermal growth factor receptor 2 (HER2) peptide, CH401MAP, to analyze humoral immunity of BCs. The hu-PBL hIL-4 NOG mice recapitulated immune environment of BCs as the ratio of CD8+/CD4+T cells was lower and that of PD-1 + T cells was higher compared to healthy donors (HDs). Diverse clusters were detected in BC-mouse (BC-M) plasma components involving immunoglobulins and complements unlike HD-M, and there was a significant diversity in CH401MAP-specific IgG titers in BC-M. The number of B cell clones producing high CH401MAP-specific IgG was not increased by immunization in BC-M unlike HD-M. These results demonstrated that the humoral immunity of BCs appeared as diverse phenotypes different from HDs in hu-PBL hIL-4 NOG mice, which may provide important information for the study of personalized medicine.


Assuntos
Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/imunologia , Imunidade Humoral , Linfócitos/imunologia , Receptor ErbB-2/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Antineoplásicos/metabolismo , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Proteínas Sanguíneas/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Imunidade Humoral/efeitos dos fármacos , Interleucina-4/metabolismo , Linfócitos/efeitos dos fármacos , Camundongos , Pessoa de Meia-Idade , Nivolumabe/farmacologia , Receptor de Morte Celular Programada 1/metabolismo , Baço/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Doadores de Tecidos
5.
Sci Rep ; 11(1): 13021, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158541

RESUMO

A syngeneic mouse model bearing a transplanted tumor is indispensable for the evaluation of the efficacy of immune checkpoint inhibitors (ICIs). However, few syngeneic mouse models of liver cancer are available. We established liver tumor cell lines (MHCF1 and MHCF5) from hepatitis C virus transgenic mice fed an atherogenic high-fat diet. MHCF1 and MHCF5 were successfully transplanted into the subcutaneous space of syngeneic C57BL/6 mice, in addition, they efficiently developed orthotopic tumors in the liver of syngeneic C57BL/6 mice. MHCF5 grew rapidly and showed a more malignant phenotype compared with MHCF1. Histologically, MHCF1-derived tumors were a combined type of hepatocellular carcinoma and MHCF5-derived tumors showed a sarcomatous morphology. Interestingly, MHCF1 and MHCF5 showed different sensitivity against an anti-PD1 antibody and MHCF5-derived tumors were resistant to this antibody. CD8 T cells infiltrated the MHCF1-derived tumors, but no CD8 T cells were found within the MHCF5-derived tumors. Gene expression profiling and whole-exon sequencing revealed that MHCF5 displayed the features of an activated cancer stem cell-like signature of sonic hedgehog and Wnt signaling. Therefore, these cell lines could be useful for the identification of new biomarkers and molecular mechanisms of ICI resistance and the development of new drugs against liver cancer.


Assuntos
Aterosclerose/patologia , Dieta Hiperlipídica , Hepacivirus/fisiologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Aloenxertos/patologia , Animais , Anticorpos Antineoplásicos/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Receptor de Morte Celular Programada 1/metabolismo , Transdução de Sinais/genética , Baço/patologia , Sequenciamento do Exoma
6.
Cells ; 10(4)2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33921301

RESUMO

Antibodies against inhibitory immune checkpoint molecules (ICPMs), referred to as immune checkpoint inhibitors (ICIs), have gained a prominent place in cancer therapy. Several ICIs in clinical use have been engineered to be devoid of effector functions because of the fear that ICIs with preserved effector functions could deplete immune cells, thereby curtailing antitumor immune responses. ICPM ligands (ICPMLs), however, are often overexpressed on a sizeable fraction of tumor cells of many tumor types and these tumor cells display an aggressive phenotype with changes typical of tumor cells undergoing an epithelial-mesenchymal transition. Moreover, immune cells expressing ICPMLs are often endowed with immunosuppressive or immune-deviated functionalities. Taken together, these observations suggest that compounds with the potential of depleting cells expressing ICPMLs may become useful tools for tumor therapy. In this article, we summarize the current state of the art of these compounds, including avelumab, which is the only ICI targeting an ICPML with preserved effector functions that has gained approval so far. We also discuss approaches allowing to obtain compounds with enhanced tumor cell-depleting potential compared to native antibodies. Eventually, we propose treatment protocols that may be applied in order to optimize the therapeutic efficacy of compounds that deplete cells expressing ICPMLs.


Assuntos
Proteínas de Checkpoint Imunológico/metabolismo , Neoplasias/patologia , Anticorpos Antineoplásicos/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Ligantes , Resultado do Tratamento
7.
J Vis Exp ; (168)2021 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-33682851

RESUMO

Within the same patient, absence of NKG2D ligands (NKG2DL) surface expression was shown to distinguish leukemic subpopulations with stem cell properties (so called leukemic stem cells, LSCs) from more differentiated counterpart leukemic cells that lack disease initiation potential although they carry similar leukemia specific genetic mutations. NKG2DL are biochemically highly diverse MHC class I-like self-molecules. Healthy cells in homeostatic conditions generally do not express NKG2DL on the cell surface. Instead, expression of these ligands is induced upon exposure to cellular stress (e.g., oncogenic transformation or infectious stimuli) to trigger elimination of damaged cells via lysis through NKG2D-receptor-expressing immune cells such as natural killer (NK) cells. Interestingly, NKG2DL surface expression is selectively suppressed in LSC subpopulations, allowing these cells to evade NKG2D-mediated immune surveillance. Here, we present a side-by-side analysis of two different flow cytometry methods that allow the investigation of NKG2DL surface expression on cancer cells i.e., a method involving pan-ligand recognition and a method involving staining with multiple antibodies against single ligands. These methods can be used to separate viable NKG2DL negative cellular subpopulations with putative cancer stem cell properties from NKG2DL positive non-LSC.


Assuntos
Citometria de Fluxo/métodos , Leucemia Mieloide Aguda/patologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Anticorpos Antineoplásicos/metabolismo , Biotinilação , Contagem de Células , Humanos , Ligantes , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Coloração e Rotulagem , Células Tumorais Cultivadas
8.
Sci Rep ; 10(1): 20777, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33247180

RESUMO

Radionuclide molecular imaging of cancer-specific targets is a promising method to identify patients for targeted antibody therapy. Radiolabeled full-length antibodies however suffer from slow clearance, resulting in high background radiation. To overcome this problem, a pretargeting system based on complementary peptide nucleic acid (PNA) probes has been investigated. The pretargeting relies on sequential injections of primary, PNA-tagged antibody and secondary, radiolabeled PNA probe, which are separated in time, to allow for clearance of non-bound primary agent. We now suggest to include a clearing agent (CA), designed for removal of primary tumor-targeting agent from the blood. The CA is based on the antibody cetuximab, which was conjugated to PNA and lactosaminated by reductive amination to improve hepatic clearance. The CA was evaluated in combination with PNA-labelled trastuzumab, T-ZHP1, for radionuclide HER2 pretargeting. Biodistribution studies in normal mice demonstrated that the CA cleared ca. 7 times more rapidly from blood than unmodified cetuximab. Injection of the CA 6 h post injection of the radiolabeled primary agent [131I]I-T-ZHP1 gave a moderate reduction of the radioactivity concentration in the blood after 1 h from 8.5 ± 1.8 to 6.0 ± 0.4%ID/g. These proof-of-principle results could guide future development of a more efficient CA.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/química , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Ácidos Nucleicos Peptídicos/administração & dosagem , Ácidos Nucleicos Peptídicos/química , Radioimunoterapia/métodos , Animais , Anticorpos Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cetuximab/administração & dosagem , Cetuximab/sangue , Cetuximab/química , Feminino , Humanos , Imunoconjugados/farmacocinética , Camundongos , Sondas Moleculares/administração & dosagem , Sondas Moleculares/química , Sondas Moleculares/farmacocinética , Terapia de Alvo Molecular/métodos , Ácidos Nucleicos Peptídicos/farmacocinética , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/imunologia , Distribuição Tecidual , Trastuzumab/administração & dosagem , Trastuzumab/sangue , Trastuzumab/química
9.
Int Immunopharmacol ; 88: 106850, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32777675

RESUMO

Targeted clearance of colorectal cancer stem cells (CCSCs) has become a novel strategy for tumor immunotherapy. Molecule mucin1 (MUC1) is one of targetable cell surface antigens in CCSCs. However, the critical role of MUC1 in anti-tumor effects of CCSC vaccine remains unclear. In the present study, we showed that MUC1 may be required for CCSC vaccine to exert tumor immunity. CD133+CCSCs were isolated from CT26 cell line using a magnetic-activated cell sorting system, and MUC1 shRNA or recombinant plasmid was further used to decrease or increase the expression of MUC1 in CD133+CCSCs. Mice were subcutaneously immunized with the CCSC lysates, MUC1 knockin CCSCs, and MUC1 knockdown CCSCs respectively, followed by a challenge with CT26 cells. We found that CCSC vaccine significantly reduced the tumor growth via a target killing of CCSCs as evidenced by a decrease of CD133+ cells and ALDH+ cells in tumors. Moreover, CCSC vaccine markedly increased the cytotoxicity of NK cells and the splenocytes, and promoted the release of IFN-γ, Perforin, and Granzyme B, and also reduced the TGF-ß1 expression. Additionally, CCSC vaccination enhanced the antibody production and decreased the myeloid derived suppressor cells and Treg subsets. More importantly, MUC1 knockdown partly impaired the anti-tumor efficacy of CCSC vaccine, whereas MUC1 overexpression dramatically enhanced the CCSC vaccine immunity. Overall, these results reveal a novel role and molecular mechanisms of MUC1 in CCSC vaccine against colorectal cancer.


Assuntos
Vacinas Anticâncer/imunologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/prevenção & controle , Mucina-1/biossíntese , Mucina-1/genética , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133/metabolismo , Aldeído Desidrogenase/metabolismo , Animais , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Feminino , Granzimas/metabolismo , Imunoterapia/métodos , Interferon gama/sangue , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Baço/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/sangue , Carga Tumoral/efeitos dos fármacos
10.
Molecules ; 25(10)2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32429033

RESUMO

High expression levels of the tumor-associated antigen MUC1 have been correlated with tumor aggressiveness, poor response to therapy, and poor survival in several tumor types, including breast, pancreatic, and epithelial ovarian cancer. Herein, we report the synthesis, characterization, and in vivo evaluation of a novel radioimmunoconjugate for the immuno-positron emission tomography (immunoPET) imaging of MUC1 expression based on the AR20.5 antibody. To this end, we modified AR20.5 with the chelator desferrioxamine (DFO) and labeled it with the positron-emitting radiometal zirconium-89 (t1/2 ~3.3 d) to produce [89Zr]Zr-DFO-AR20.5. In subsequent in vivo experiments in athymic nude mice bearing subcutaneous MUC1-expressing ovarian cancer xenografts, [89Zr]Zr-DFO-AR20.5 clearly delineated tumor tissue, producing a tumoral activity concentration of 19.1 ± 6.4 percent injected dose per gram (%ID/g) at 120 h post-injection and a tumor-to-muscle activity concentration ratio of 42.4 ± 10.6 at the same time point. Additional PET imaging experiments in mice bearing orthotopic MUC1-expressing ovarian cancer xenografts likewise demonstrated that [89Zr]Zr-DFO-AR20.5 enables the visualization of tumor tissue-including metastatic lesions-with promising tumor-to-background contrast.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Antineoplásicos/química , Imunoconjugados/química , Metástase Linfática/diagnóstico por imagem , Mucina-1/metabolismo , Neoplasias/diagnóstico por imagem , Radioisótopos/química , Zircônio/química , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacocinética , Anticorpos Antineoplásicos/metabolismo , Disponibilidade Biológica , Desferroxamina/química , Feminino , Expressão Gênica , Xenoenxertos , Humanos , Imunoconjugados/metabolismo , Imunoconjugados/farmacocinética , Metástase Linfática/patologia , Camundongos , Camundongos Nus , Mucina-1/genética , Neoplasias/metabolismo , Neoplasias/patologia , Tomografia por Emissão de Pósitrons/métodos , Ligação Proteica , Razão Sinal-Ruído , Distribuição Tecidual
11.
Cancer Med ; 9(13): 4756-4767, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32368864

RESUMO

INTRODUCTION: Ovarian cancer has only a 17% 5-year survival rate in patients diagnosed with late stage disease. Tumor-associated glycoprotein-72 (TAG72), expressed in 88% of all stages of ovarian cancer, is an excellent candidate for antibody-targeted therapy, as it is not expressed in normal human adult tissues, except in the secretory endometrium. METHODS: Using the clinically relevant anti-TAG72 murine monoclonal antibody CC49, we evaluated antibody drug conjugates (ADCs) incorporating the highly potent, synthetic antimitotic agent monomethylauristatin E (MMAE). MMAE was conjugated to CC49 via reduced disulfides in the hinge region, using three different types of linker chemistry, vinylsulfone (VS-MMAE), bromoacetamido (Br-MMAE), and maleimido (mal-MMAE). RESULTS: The drug antibody ratios (DARs) of the three ADCs were 2.3 for VS-MMAE, 10 for Br-MMAE, and 9.5 for mal-MMAE. All three ADCs exhibited excellent tumor to blood ratios on PET imaging, but the absolute uptake of CC49-mal-MMAE (3.3%ID/g) was low compared to CC49-Br-MMAE (6.43%ID/g), at 142 hours. Blood clearance at 43 hours was 38% for intact CC49, about 24% for both CC49-VS-MMAE and CC49-Br-MMAE, and 7% for CC49-mal-MMAE. CC49-VS-MMAE was not further studied due to its low DAR, while CC49-mal-MMAE was ineffective in the OVCAR3 xenograft likely due to its rapid blood clearance. In contrast, CC49-Br-MMAE treated mice exhibited an average of a 15.6 day tumor growth delay and a 40% increase in survival vs controls with four doses of 7.5 or 15 mg/kg of CC49-Br-MMAE. CONCLUSION: We conclude that CC49-Br-MMAE with a high DAR and stable linker performs well in a difficult to treat solid tumor model.


Assuntos
Anticorpos Antineoplásicos/uso terapêutico , Antígenos de Neoplasias/imunologia , Imunoconjugados/uso terapêutico , Fatores Imunológicos/uso terapêutico , Oligopeptídeos/uso terapêutico , Neoplasias Ovarianas/terapia , Acetatos/química , Animais , Anticorpos Antineoplásicos/sangue , Anticorpos Antineoplásicos/metabolismo , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Feminino , Células HT29 , Humanos , Fatores Imunológicos/farmacocinética , Camundongos , Oligopeptídeos/sangue , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/metabolismo , Tomografia por Emissão de Pósitrons , Distribuição Aleatória , Sulfonas/química
12.
Curr Protoc Cytom ; 93(1): e73, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32311834

RESUMO

The identification of residual leukemia following therapy, termed minimal or measurable residual disease (MRD), has emerged as one of the most important prognostic factors for patients with acute leukemia, including acute myeloid leukemia (AML). Flow cytometry is a preferred method for MRD detection due to its general applicability and the rapid results that it makes available. In this article, the basic protocol outlines a simple and efficient method for the labeling of hematopoietic cells from bone marrow or peripheral blood with a panel of monoclonal antibodies designed both to highlight patterns of normal maturation and allow identification of neoplastic hematopoietic progenitor populations with a high degree of sensitivity and specificity. The method was developed in a clinical laboratory setting for the diagnosis of myeloid stem cell disorders and neoplasms, and has been extensively validated both technically and clinically for the detection of MRD in AML. © 2020 The Authors. Basic Protocol: Staining and flow cytometry for AML minimal residual disease detection Support Protocol: Analysis and interpretation of data for AML minimal residual disease detection.


Assuntos
Citometria de Fluxo/métodos , Leucemia Mieloide Aguda/diagnóstico , Neoplasia Residual/diagnóstico , Anticorpos Antineoplásicos/metabolismo , Sobrevivência Celular , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/patologia , Antígenos Comuns de Leucócito/metabolismo , Mutação/genética , Neoplasia Residual/patologia , Proteínas Nucleares/genética , Nucleofosmina , Coloração e Rotulagem
13.
Gynecol Oncol ; 155(2): 324-330, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31477280

RESUMO

OBJECTIVE: To date, The Cancer Genome Atlas (TCGA) has provided the most extensive molecular characterization of invasive cervical cancer (ICC). Analysis of reverse phase protein array (RPPA) data from TCGA samples showed that cervical cancers could be stratified into 3 clusters exhibiting significant differences in survival outcome: hormone, EMT, and PI3K/AKT. The goals of the current study were to: 1) validate the TCGA RPPA results in an independent cohort of ICC patients and 2) to develop and validate an algorithm encompassing a small antibody set for clinical utility. METHODS: Subjects consisted of 2 ICC patient cohorts with accompanying RPPA and clinical-pathologic data: 155 samples from TCGA (TCGA-155) and 61 additional, unique samples (MCW-61). Using data from 173 common RPPA antibodies, we replicated Silhouette clustering analysis in both ICC cohorts. Further, an index score for each patient was calculated from the survival-associated antibodies (SAAs) identified using Random survival forests (RSF) and the Cox proportional hazard regression model. Kaplan-Meier survival analysis and the log-rank test were performed to assess and compare cluster or risk group survival outcome. RESULTS: In addition to validating the prognostic ability of the proteomic clusters reported by TCGA, we developed an algorithm based on 22 unique antibodies (SAAs) that stratified women with ICC into low-, medium-, or high-risk survival groups. CONCLUSIONS: We provide a signature of 22 antibodies which accurately predicted survival outcome in 2 separate groups of ICC patients. Future studies examining these candidate biomarkers in additional ICC cohorts is warranted to fully determine their clinical potential.


Assuntos
Proteômica , Neoplasias do Colo do Útero/mortalidade , Adulto , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/metabolismo , Biomarcadores Tumorais/metabolismo , Feminino , Humanos , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fatores de Risco , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/imunologia
14.
Curr Opin Rheumatol ; 31(6): 643-649, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31369431

RESUMO

PURPOSE OF REVIEW: The aim of this study is to provide a comprehensive overview of the current insight about the clinical utility of cancer-associated autoantibodies (CAAs) as biomarkers in paraneoplastic myositis syndrome (PMS). In addition, the possible mechanisms of the relationship between malignancy and myositis onset are discussed. RECENT FINDINGS: It has become increasingly clear that a subgroup of the myositis-specific autoantibodies could be considered as CAAs because they are closely related to the PMS. Increased risk of cancer was found in patients with antitranscriptional intermediary factor 1-γ (TIF1-γ), antinuclear matrix protein-2 (NXP-2), anti3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) or antismall ubiquitin-like modifier 1-activating enzyme (SAE) antibodies. However, the diagnosing sensitivity and specificity of these CAAs for PMS are different among different cohort studies. Abnormally expressed or mutated autoantigen genes in tumor could possibly induce cross immunity against self-proteins and subsequently lead to the development of PMS. SUMMARY: Anti-TIF1-γ, anti-NXP-2, anti-HMGCR and anti-SAE antibodies may act as CAAs in PMS. It is necessary to closely screen and monitor for cancer in patients with CAAs. The recent studies of the relationship between CAAs and PMS provided important new insights into the disease mechanisms.


Assuntos
Anticorpos Antineoplásicos/imunologia , Autoanticorpos/imunologia , Miosite/imunologia , Síndromes Paraneoplásicas/imunologia , Anticorpos Antineoplásicos/metabolismo , Autoanticorpos/metabolismo , Biomarcadores Tumorais/metabolismo , Humanos , Miosite/metabolismo , Síndromes Paraneoplásicas/metabolismo
15.
Adv Healthc Mater ; 8(13): e1900136, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30985092

RESUMO

Multidrug resistance is one of the leading causes of chemotherapy failure in cancer patients. Early detection and capture of drug-resistant tumor cells can facilitate the monitoring of the therapy process and improve the prognosis of patients. In this study, novel P-glycoprotein (P-gp) antibody modified porous hydrogel particles are proposed for drug-resistant tumor cells capture. The hydrogel particles employ a highly biocompatible hydrogel, methacrylate gelatin (GelMA), as the carrier and replicate from the silica colloidal crystal beads. By the modification of P-gp antibody probes on their surfaces, the hydrogel particles are endowed with the ability to capture drug-resistant tumor cells, which overexpress specific components of P-gp on their membranes. Additionally, the acquired ordered porous nanostructure of the particles can provide not only more surface area for antibody immobilization but also a nanopatterned platform for highly efficient target cell capture. The above advantages make the porous hydrogel particles ideal for efficient capture and detection of the drug-resistant tumor cells, which can be expected to facilitate the point-of-care pharmacotherapy and promisingly improve the patient outcomes.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/imunologia , Anticorpos Antineoplásicos/química , Resistencia a Medicamentos Antineoplásicos , Hidrogéis/química , Neoplasias/patologia , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/metabolismo , Anticorpos Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Gelatina/química , Humanos , Células K562 , Metacrilatos/química , Nanoestruturas/química , Neoplasias/metabolismo , Porosidade , Dióxido de Silício/química
16.
Front Immunol ; 9: 2233, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319664

RESUMO

Lymph nodes (LNs) are central in the generation of adaptive immune responses. Follicular helper CD4 T (Tfh) cells, a highly differentiated CD4 population, provide critical help for the development of antigen-specific B cell responses within the germinal center. Throughout the past decade, numerous studies have revealed the important role of Tfh cells in Human Immunodeficiency Virus (HIV) pathogenesis as well as in the development of neutralizing antibodies post-infection and post-vaccination. It has also been established that tumors influence various immune cell subsets not only in their proximity, but also in draining lymph nodes. The role of local or tumor associated lymph node Tfh cells in disease progression is emerging. Comparative studies of Tfh cells in chronic infections and cancer could therefore provide novel information with regards to their differentiation plasticity and to the mechanisms regulating their development.


Assuntos
Centro Germinativo/imunologia , Infecções por HIV/imunologia , Neoplasias/imunologia , Linfonodo Sentinela/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Plasticidade Celular/imunologia , Modelos Animais de Doenças , Progressão da Doença , Centro Germinativo/citologia , HIV/imunologia , HIV/patogenicidade , Infecções por HIV/virologia , Humanos , Imunidade Humoral , Neoplasias/patologia , Linfonodo Sentinela/citologia
17.
Glycoconj J ; 35(3): 333-342, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29926273

RESUMO

A promising approach capitalizing on the specific and highly sensitive characteristics of the body's own immune system is demonstrated in the context of revealing pancreatic ductal adenocarcinoma cancer (PDAC). IgA from a local biofluid called gastrointestinal lavage fluid (GLF) is used to investigate glycan reactivity to show the potential of this approach. IgA antibody responses, just as with IgG, result in amplification of a small signal which aids in detecting changes from a healthy state. IgA from GLF was screened against glycan arrays containing 609 glycan structures to investigate differential binding patterns associated with the disease. Samples included PDAC (n = 14) and non-PDAC (n = 6). Non-PDAC conditions included samples from healthy patients and the potentially confounding conditions of colon cancer and its precancerous lesion, colon adenoma. Results demonstrated characteristic reactivity in the PDAC sample group to a glycan structure. Also, IgA non-reactive motifs arose showing remarkable consistency within and between sample groups. While sample sizes are too small to identify putative biomarkers, these data show the use of IgA from GLF to be a promising avenue of research for local disease biomarker discovery.


Assuntos
Adenoma/metabolismo , Anticorpos Antineoplásicos/metabolismo , Especificidade de Anticorpos , Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/metabolismo , Imunoglobulina A/metabolismo , Intestinos , Lesões Pré-Cancerosas/metabolismo , Adenoma/patologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Neoplasias do Colo/patologia , Feminino , Humanos , Masculino , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/patologia
18.
Pigment Cell Melanoma Res ; 31(6): 708-719, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29778085

RESUMO

Little is known about the in vivo impacts of targeted therapy on melanoma cell abundance and protein expression. Here, 21 antibodies were added to an established melanoma mass cytometry panel to measure 32 cellular features, distinguish malignant cells, and characterize dabrafenib and trametinib responses in BRAFV600mut melanoma. Tumor cells were biopsied before neoadjuvant therapy and compared to cells surgically resected from the same site after 4 weeks of therapy. Approximately 50,000 cells per tumor were characterized by mass cytometry and computational tools t-SNE/viSNE, FlowSOM, and MEM. The resulting single-cell view of melanoma treatment response revealed initially heterogeneous melanoma tumors were consistently cleared of Nestin-expressing melanoma cells. Melanoma cell subsets that persisted to week 4 were heterogeneous but expressed SOX2 or SOX10 proteins and specifically lacked surface expression of MHC I proteins by MEM analysis. Traditional histology imaging of tissue microarrays from the same tumors confirmed mass cytometry results, including persistence of NES- SOX10+ S100ß+ melanoma cells. This quantitative single-cell view of melanoma treatment response revealed protein features of malignant cells that are not eliminated by targeted therapy.


Assuntos
Melanoma/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Nestina/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Anticorpos Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Melanoma/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oximas/farmacologia , Oximas/uso terapêutico , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Piridonas/farmacologia , Piridonas/uso terapêutico , Pirimidinonas/farmacologia , Pirimidinonas/uso terapêutico
19.
Biointerphases ; 13(2): 021001, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29571263

RESUMO

Methods for isolation and quantification of circulating tumor cells (CTCs) are attracting more attention every day, as the data for their unprecedented clinical utility continue to grow. However, the challenge is that CTCs are extremely rare (as low as 1 in a billion of blood cells) and a highly sensitive and specific technology is required to isolate CTCs from blood cells. Methods utilizing microfluidic systems for immunoaffinity-based CTC capture are preferred, especially when purity is the prime requirement. However, antibody immobilization strategy significantly affects the efficiency of such systems. In this study, two covalent and two bioaffinity antibody immobilization methods were assessed with respect to their CTC capture efficiency and selectivity, using an anti-epithelial cell adhesion molecule (EpCAM) as the capture antibody. Surface functionalization was realized on plain SiO2 surfaces, as well as in microfluidic channels. Surfaces functionalized with different antibody immobilization methods are physically and chemically characterized at each step of functionalization. MCF-7 breast cancer and CCRF-CEM acute lymphoblastic leukemia cell lines were used as EpCAM positive and negative cell models, respectively, to assess CTC capture efficiency and selectivity. Comparisons reveal that bioaffinity based antibody immobilization involving streptavidin attachment with glutaraldehyde linker gave the highest cell capture efficiency. On the other hand, a covalent antibody immobilization method involving direct antibody binding by N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (EDC)-N-hydroxysuccinimide (NHS) reaction was found to be more time and cost efficient with a similar cell capture efficiency. All methods provided very high selectivity for CTCs with EpCAM expression. It was also demonstrated that antibody immobilization via EDC-NHS reaction in a microfluidic channel leads to high capture efficiency and selectivity.


Assuntos
Anticorpos Antineoplásicos/imunologia , Células Sanguíneas , Separação Celular/métodos , Molécula de Adesão da Célula Epitelial/imunologia , Proteínas Imobilizadas/metabolismo , Células Neoplásicas Circulantes/imunologia , Anticorpos Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Molécula de Adesão da Célula Epitelial/metabolismo , Humanos , Microfluídica/métodos , Ligação Proteica
20.
Cell Rep ; 22(7): 1875-1888, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29444438

RESUMO

We have performed an in-depth single-cell phenotypic characterization of high-grade serous ovarian cancer (HGSOC) by multiparametric mass cytometry (CyTOF). Using a CyTOF antibody panel to interrogate features of HGSOC biology, combined with unsupervised computational analysis, we identified noteworthy cell types co-occurring across the tumors. In addition to a dominant cell subset, each tumor harbored rarer cell phenotypes. One such group co-expressed E-cadherin and vimentin (EV), suggesting their potential role in epithelial mesenchymal transition, which was substantiated by pairwise correlation analyses. Furthermore, tumors from patients with poorer outcome had an increased frequency of another rare cell type that co-expressed vimentin, HE4, and cMyc. These poorer-outcome tumors also populated more cell phenotypes, as quantified by Simpson's diversity index. Thus, despite the recognized genomic complexity of the disease, the specific cell phenotypes uncovered here offer a focus for therapeutic intervention and disease monitoring.


Assuntos
Citometria de Fluxo/métodos , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antineoplásicos/metabolismo , Carboplatina/farmacologia , Linhagem Celular Tumoral , Análise por Conglomerados , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Recidiva Local de Neoplasia/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fenótipo , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA