Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Cancer ; 12(1): 143, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24257318

RESUMO

BACKGROUND: PAM4, an antibody that has high specificity for pancreatic ductal adenocarcinoma (PDAC), compared to normal pancreas, benign lesions of the pancreas, and cancers originating from other tissues, is being investigated as a biomarker for early detection, as well as antibody-targeted imaging and therapy. Therefore, the identity of the antigen bound by this monoclonal antibody (MAb) can provide information leading to improved use of the antibody. Prior results suggested the antigen is a mucin-type glycoprotein rich in cysteine disulfide bridges that provide stable conformation for the PAM4-epitope. METHODS: Indirect and sandwich enzyme immunoassays (EIA) were performed to compare and contrast the reactivity of PAM4 with several anti-mucin antibodies having known reactivity to specific mucin species (e.g., MUC1, MUC4, MUC5AC, etc.). Studies designed to block reactivity of PAM4 with its specific antigen also were performed. RESULTS: We demonstrate that MAbs 2-11 M1 and 45 M1, each reactive with MUC5AC, are able to provide signal in a heterologous sandwich immunoassay where PAM4 is the capture antibody. Further, we identify MAbs 21 M1, 62 M1, and 463 M1, each reactive with MUC5AC, as inhibiting the reaction of PAM4 with its specific epitope. MAbs directed to MUC1, MUC3, MUC4, MUC16 and CEACAM6 are not reactive with PAM4-captured antigen, nor are they able to block the reaction of PAM4 with its antigen. CONCLUSIONS: These data implicate MUC5AC as a specific mucin species to which PAM4 is reactive. Furthermore, this realization may allow for the improvement of the current PAM4 serum-based immunoassay for detection of early-stage PDAC by the application of anti-MUC5AC MAbs as probes in this sandwich EIA.


Assuntos
Anticorpos Imobilizados/química , Anticorpos Monoclonais/química , Carcinoma Ductal Pancreático/diagnóstico , Mucina-5AC/metabolismo , Neoplasias Pancreáticas/diagnóstico , Animais , Carcinoma Ductal Pancreático/tratamento farmacológico , Ensaios Clínicos como Assunto , Detecção Precoce de Câncer , Mapeamento de Epitopos , Humanos , Imunoensaio , Camundongos , Camundongos Nus , Mucina-5AC/imunologia , Transplante de Neoplasias , Neoplasias Pancreáticas/tratamento farmacológico
2.
Biol Blood Marrow Transplant ; 19(1): 28-39, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23025988

RESUMO

Prevention and treatment of graft-versus-host disease (GVHD) remains a major challenge, given that current T-cell depletion and mainstay immunosuppressive therapies compromise preexisting T-cell immunity, often leading to severe infections and disease relapse. Thus, there is a critical need for novel anti-GVHD agents that can spare protective T-cell memory. Here we show that milatuzumab (hLL1), a humanized anti-CD74 antagonist monoclonal antibody, can moderately reduce the numbers of CD74-expressing B cells and myeloid dendritic cells, but has no effect on the survival of T cells that are CD74(-). Consequently, milatuzumab inhibits allogeneic T-cell proliferation in mixed leukocyte reactions. In a human/mouse xenogeneic SCID mouse model in which GVHD is induced and mediated by engrafted human CD4(+) T cells and dendritic cells, milatuzumab effectively prevents the onset and manifestations of acute GVHD, suppresses serum levels of human IFN-γ and IL-5, eliminates the infiltration of human lymphocytes into GVHD target organs (ie, lung, liver, and spleen), and significantly promotes survival (90% versus 20% for controls; P = .0012). Importantly, exposure to milatuzumab does not affect the number of cytomegalovirus-specific, IFN-γ-producing human CD8(+) T cells in allogeneic mixed leukocyte reactions. These encouraging results warrant further exploration of milatuzumab as a possible new therapeutic agent for GVHD.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antígenos de Diferenciação de Linfócitos B , Doença Enxerto-Hospedeiro/prevenção & controle , Antígenos de Histocompatibilidade Classe II , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Células Dendríticas/transplante , Modelos Animais de Doenças , Feminino , Doença Enxerto-Hospedeiro/sangue , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Humanos , Interferon gama/sangue , Interferon gama/imunologia , Interleucina-5/sangue , Interleucina-5/imunologia , Masculino , Camundongos , Camundongos SCID , Transplante Heterólogo
3.
Cancer ; 119(3): 522-8, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22898932

RESUMO

BACKGROUND: The monoclonal antibody PAM4 has high specificity for pancreatic ductal adenocarcinoma (PDAC), as well as its precursor lesions, but has not been found to be reactive with normal and benign pancreatic tissues. The objective of the current study was to evaluate a PAM4-based serum enzyme immunoassay alone and in combination with the carbohydrate antigen (CA) 19-9 assay for the detection of PDAC, with particular attention to early stage disease. METHODS: Sera from patients with confirmed PDAC (N = 298), other cancers (N = 99), benign disease of the pancreas (N = 120), and healthy adults (N = 79) were evaluated by a specific enzyme immunoassay for the concentration of PAM4 and CA 19-9 antigen levels by blinded analyses. All tests for statistical significance were 2-sided. RESULTS: The overall sensitivity for PAM4 detection of PDAC was 76%, with 64% of patients with stage I disease also identified. The detection rate was considerably higher (85%) for patients with advanced disease. The assay demonstrated high specificity compared with benign pancreatic disease (85%), with a positive likelihood ratio of 4.93. CA 19-9 provided an overall sensitivity of 77%, and was positive in 58% of patients with stage I disease; however, the specificity was significantly lower for CA 19-9 (68%), with a positive likelihood ratio of 2.85 (P = .026 compared with PAM4). It is important to note that a combined PAM4 and CA 19-9 biomarker serum assay demonstrated an improved sensitivity (84%) for the overall detection of PDAC without a significant loss of specificity (82%) compared with either arm alone. CONCLUSIONS: The PAM4 enzyme immunoassay identified approximately two-thirds of patients with stage I PDAC with high discriminatory power with respect to benign, nonneoplastic pancreatic disease. These results provide a rationale for testing patient groups considered to be at high risk for PDAC with a combined PAM4 and CA 19-9 biomarker serum assay for the detection of early stage PDAC.


Assuntos
Adenocarcinoma/diagnóstico , Antígenos de Neoplasias/análise , Antígeno CA-19-9/análise , Detecção Precoce de Câncer/métodos , Neoplasias Pancreáticas/diagnóstico , Adenocarcinoma/sangue , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Antígenos de Neoplasias/metabolismo , Análise Química do Sangue/métodos , Antígeno CA-19-9/metabolismo , Carcinoma Ductal Pancreático/sangue , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Estudos de Casos e Controles , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Concentração Osmolar , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Sensibilidade e Especificidade
4.
Int J Cancer ; 131(1): 49-58, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21796629

RESUMO

Cell fusion in vitro has been used to study cancer, gene mapping and regulation, and the production of antibodies via hybridomas. However, in-vivo heterosynkaryon formation by cell-cell fusion has received less attention. This investigation describes the spontaneous fusion of a human glioblastoma with normal hamster cells after xenogeneic transplantation, resulting in malignant cells that express both human and hamster genes and gene products, and retention of glioblastoma traits with an enhanced ability to metastasize. Three of 7 human genes found showed translation of their proteins during serial propagation in vivo or in vitro for years; namely, CD74, CXCR4 and PLAGL2, each implicated with malignancy or glioblastoma. This supports the thesis that genetic hybridization of cancer and normal cells can transmit malignancy and also, as first described herein, regulatory genes involved in the tumor's organotypic morphology. Evidence also is increasing that even cell-free human cancer DNA can induce malignancy and transfer genetic information to normal cells. Hence, we posit that the transfer of genetic information between tumor and stromal cells, whether by cell-cell fusion or other mechanisms, is implicated in the progression of malignancy, and may further define the crosstalk between cancer cells and their stromal neighbors.


Assuntos
Fusão Celular , Glioblastoma/genética , Glioblastoma/patologia , Células Híbridas/patologia , Animais , Antígenos de Diferenciação de Linfócitos B/biossíntese , Antígenos de Diferenciação de Linfócitos B/genética , Linhagem Celular Tumoral , Quimera , Cricetinae , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Progressão da Doença , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Metástase Neoplásica , Transplante de Neoplasias , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Células Estromais/patologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transplante Heterólogo
5.
Cancer ; 118(22): 5497-506, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22569804

RESUMO

BACKGROUND: It has been demonstrated that the humanized clivatuzumab tetraxetan (hPAM4) antibody targets pancreatic ductal carcinoma selectively. After a trial of radioimmunotherapy that determined the maximum tolerated dose of single-dose yttrium-90-labeled hPAM4 ((90) Y-hPAM4) and produced objective responses in patients with advanced pancreatic ductal carcinoma, the authors studied fractionated radioimmunotherapy combined with low-dose gemcitabine in this disease. METHODS: Thirty-eight previously untreated patients (33 patients with stage IV disease and 5 patients with stage III disease) received gemcitabine 200 mg/m(2) weekly for 4 weeks with (90) Y-hPAM4 given weekly in Weeks 2, 3, and 4 (cycle 1), and the same cycle was repeated in 13 patients (cycles 2-4). In the first part of the study, 19 patients received escalating weekly (90) Y doses of 6.5 mCi/m(2) , 9.0 mCi/m(2) , 12.0 mCi/m(2) , and 15.0 mCi/m(2) . In the second portion, 19 additional patients received weekly doses of 9.0 mCi/m(2) or 12.0 mCi/m(2) . RESULTS: Grade 3/4 thrombocytopenia or neutropenia (according to version 3.0 of the National Cancer Institute's Common Terminology Criteria for Adverse Events) developed in 28 of 38 patients after cycle 1 and in all retreated patients; no grade >3 nonhematologic toxicities occurred. Fractionated dosing of cycle 1 allowed almost twice the radiation dose compared with single-dose radioimmunotherapy. The maximum tolerated dose of (90) Y-hPAM4 was 12.0 mCi/m(2) weekly for 3 weeks for cycle 1, with ≤9.0 mCi/m(2) weekly for 3 weeks for subsequent cycles, and that dose will be used in future trials. Six patients (16%) had partial responses according to computed tomography-based Response Evaluation Criteria in Solid Tumors, and 16 patients (42%) had stabilization as their best response (58% disease control). The median overall survival was 7.7 months for all 38 patients, including 11.8 months for those who received repeated cycles (46% [6 of 13 patients] ≥1 year), with improved efficacy at the higher radioimmunotherapy doses. CONCLUSIONS: Fractionated radioimmunotherapy with (90) Y-hPAM4 and low-dose gemcitabine demonstrated promising therapeutic activity and manageable myelosuppression in patients with advanced pancreatic ductal carcinoma.


Assuntos
Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/radioterapia , Radioimunoterapia , Radioisótopos de Ítrio/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/uso terapêutico , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/radioterapia , Terapia Combinada , Desoxicitidina/uso terapêutico , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neutropenia/etiologia , Doses de Radiação , Radiossensibilizantes/uso terapêutico , Trombocitopenia/etiologia , Radioisótopos de Ítrio/efeitos adversos , Gencitabina
6.
Clin Cancer Res ; 13(24): 7380-7, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18094420

RESUMO

PURPOSE: The anti-MUC1 monoclonal antibody (MAb), PAM4, has a high specificity for pancreatic adenocarcinoma compared with other cancers, normal tissues, or pancreatitis. In order to assess its role in early pancreatic cancer development, we examined the expression of the PAM4-reactive MUC1 in the noninvasive precursor lesions, pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasia (IPMN). EXPERIMENTAL DESIGN: Tissue microarrays prepared from formalin-fixed, paraffin-embedded specimens were assessed by immunohistology for expression of the PAM4-reactive, non-variable number of tandem repeats (VNTR), MUC1 epitope, and the VNTR epitope bound by the MA5 MAb. RESULTS: The PAM4-reactive MUC1 epitope was not detected in normal pancreas but was expressed in 87% (48 of 55) of invasive pancreatic adenocarcinomas, including early stage 1 disease: PAM4 labeled 94% (44 of 47) of the earliest PanIN lesions, PanIN-1A and 1B, along with 91% (10 of 11) of PanIN-2, 40% (2 of 5) of PanIN-3, and 86% (31 of 36) of intraductal papillary mucinous neoplasia lesions. A mostly diffuse pattern of labeling was observed. A second, unrelated, anti-MUC1 MAb, MA5, showed considerably less sensitivity with early PanIN-1 lesions; only 61% (25 of 41) were positive and the labeling did not differentiate normal pancreas from PanINs. CONCLUSIONS: The results suggest that expression of the PAM4-reactive antigen may represent an early event in the development of invasive pancreatic adenocarcinoma, and is unrelated to the VNTR peptide core epitopes of MUC1. Detection of this biomarker using immunohistology, in vitro immunoassays, and in vivo antibody-based imaging may provide new opportunities for the early detection and improved diagnosis of pancreatic cancer.


Assuntos
Anticorpos Monoclonais , Biomarcadores Tumorais/análise , Carcinoma in Situ/diagnóstico , Carcinoma Ductal Pancreático/diagnóstico , Mucina-1/metabolismo , Neoplasias Pancreáticas/diagnóstico , Anticorpos Monoclonais/imunologia , Área Sob a Curva , Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Epitopos/imunologia , Humanos , Imuno-Histoquímica , Repetições Minissatélites , Mucina-1/imunologia , Neoplasias Pancreáticas/metabolismo , Curva ROC , Sensibilidade e Especificidade , Análise Serial de Tecidos
7.
Mol Cancer Ther ; 5(2): 200-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16505092

RESUMO

Considerable progress has been made recently in our understanding of the role of ceramide in the induction of apoptotic cell death. Ceramide is produced by cancer cells in response to exposure to radiation and most chemotherapeutics and is an intracellular second messenger that activates enzymes, leading to apoptosis. Because of its central role in apoptosis, pharmacologic manipulation of intracellular ceramide levels should result in attenuation or enhancement of drug resistance. This may be achieved through direct application of sphingolipids or by the inhibition/activation of the enzymes that either produce or use ceramide. In addition, attention should be given to the subcellular location of ceramide generation, because this has been shown to affect the biological activity of sphingolipids. This review summarizes the sphingolipid biosynthetic pathway, as it relates to the identification of important targets for drug discovery, and the development of novel agents capable of enhancing chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Ceramidas/metabolismo , Neoplasias/tratamento farmacológico , Esfingolipídeos/metabolismo , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Morte Celular , Ceramidas/análise , Ceramidas/biossíntese , Humanos , Neoplasias/metabolismo , Esfingolipídeos/análise , Esfingolipídeos/biossíntese
8.
Cancer Res ; 64(22): 8405-10, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15548711

RESUMO

We have examined the mechanism by which sphingomyelin (SM) enhances chemotherapy in human pancreatic cancer cells, focusing on the correlation between ceramide metabolism and apoptosis. Dose response curves for gemcitabine in the absence or presence of 0.2 mg/mL SM provided IC(50) values of 78.3 +/- 13.7 and 13.0 +/- 3.0 nmol/L, respectively. The cytotoxic effect of the combined treatment was synergistic (combination index = 0.36). Using annexin-V staining, the percentage of apoptotic cells was 3.6 +/- 2.6% for the untreated cells, 6.5 +/- 3.8% for the 0.2 mg/mL SM-treated cells, and 19.9 +/- 12.9% for the 100 nmol/L gemcitabine-treated cells, but increased significantly to 42.1 +/- 12.7% with the combined treatment (P < 0.001, compared with gemcitabine-treated group). The percentage of cells losing mitochondrial membrane potential followed a similar trend. The ceramide content of untreated and gemcitabine-treated cells was not significantly different (0.46 +/- 0.29 and 0.59 +/- 0.34 pmol ceramide/nmole PO(4)). However, when 0.2 mg/mL SM was added, ceramide levels were 1.09 +/- 0.42 and 1.58 +/- 0.55 pmol ceramide/nmol PO(4), for the SM alone and SM with gemcitabine-treated cells, respectively (P = 0.038). Acidic SMase was activated by exposure to gemcitabine but not SM, whereas the activities of neutral SMase and glycosylceramide synthase did not change with either gemcitabine or SM. The data are consistent with gemcitabine-induced activation of acidic SMase and indicate that the addition of SM can yield increased production of ceramide, mitochondrial depolarization, apoptosis, and cell death. Because SM by itself is relatively nontoxic, addition of this lipid to agents that induce apoptosis may prove useful to enhance apoptosis and increase cytotoxicity in cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Ceramidas/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Neoplasias Pancreáticas/patologia , Esfingomielinas/farmacologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Ativação Enzimática , Glucosiltransferases/metabolismo , Humanos , Neoplasias Pancreáticas/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Gencitabina
9.
Int J Oncol ; 26(6): 1591-6, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15870874

RESUMO

Mu-9, an anti-colon specific antigen-p (CSAp) monoclonal antibody (MAb), has shown excellent gastrointestinal cancer targeting in both pre-clinical and clinical trials. With the recent development of the humanized version of this MAb, Mu-9 will receive further attention as a potential therapeutic agent for colorectal cancer. Hence, we have undertaken studies to examine the nature of the CSAp antigen and the structure of the Mu-9 epitope. M13 phage displaying random 12-mer peptides were used to isolate a peptide that binds both murine and humanized Mu-9 MAbs. The peptide, Mu-9-p16, was synthesized and found to inhibit binding of Mu-9 to CSAp with a Ka for the humanized antibody of 4.28 x 10(-7) +/- 0.91 x 10(-7) M. Control peptides did not bind Mu-9, nor did they inhibit the Mu-9-CSAp interaction. Three overlapping peptides were synthesized and used to demonstrate that the last six residues were sufficient to inhibit the Mu-9-CSAp interaction. A search of GenBank revealed that the peptide sequence IHPRP, was also present within CA125, a very high molecular weight ovarian cancer-associated antigen. The sequence is present outside of the known antigenic regions identified by the Oc125, M-11 and Ov197 anti-CA125 antibodies. To demonstrate that CSAp and CA125 may be the same protein, a heterologous sandwich enzyme immunoassay was developed with anti-CA125 MAbs used as capture reagents and Mu-9 as probe. By use of this ELISA system, we were able to specifically identify CSAp. In conclusion, our results indicate that the Mu-9-p16 peptide isolated through our screen identifies a peptide epitope shared by CSAp and CA125 and suggest that these proteins are related.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/análise , Antígeno Ca-125/análise , Sequência de Aminoácidos , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Ligação Competitiva , Antígeno Ca-125/química , Antígeno Ca-125/imunologia , Humanos , Dados de Sequência Molecular , Biblioteca de Peptídeos
10.
Clin Cancer Res ; 9(10 Pt 2): 3929S-37S, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-14506191

RESUMO

PURPOSE: Monoclonal antibody PAM4 is reactive with the MUC1 mucin as expressed by >85% of human pancreatic cancers. Significant antitumor effects have been demonstrated using radiolabeled PAM4 for radioimmunotherapy (RAIT) of experimental pancreatic cancer. The goal of the present study was to determine whether the addition of low-dose (90)Y-PAM4 RAIT to a clinically relevant regimen of gemcitabine chemotherapy would provide enhanced antitumor efficacy over that observed by chemotherapy alone without the addition of significant toxicity to normal tissues. EXPERIMENTAL DESIGN: Mice bearing human pancreatic tumor xenografts (CaPan1) were administered three cycles of gemcitabine chemotherapy (1000 mg/m(2)/week for 3 weeks with 1 week off) concomitant with (90)Y-labeled PAM4 RAIT (25 micro Ci; 10% of the single agent MTD) given at weeks 0, 4, and 7. Control groups of mice received chemotherapy alone, (90)Y-PAM4 RAIT alone, or an equidose of (90)Y-labeled nontargeting control antibody with and without gemcitabine. RESULTS: Mice that received (90)Y-PAM4 RAIT with gemcitabine had tumors that were significantly smaller in size than all of the other treatment groups (P < 0.005). A median survival of 24 weeks was achieved in mice that received the combined treatment versus 10 weeks for mice that received only gemcitabine (P < 0.001) and 16 weeks for mice that received only (90)Y-PAM4 RAIT (P < 0.040). The combined treatment regimen was well tolerated. CONCLUSIONS: A combined chemoimmunotherapy and RAIT approach using gemcitabine and low-dose (90)Y-PAM4 provided significantly increased antitumor efficacy than was observed for each treatment arm given alone. Importantly, the enhanced antitumor efficacy was achieved with minimal toxicity to normal tissues. These studies provide justification for clinical trials using the combined modality treatment for patients with pancreatic cancer.


Assuntos
Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Neoplasias Pancreáticas/terapia , Radioimunoterapia/métodos , Radioisótopos de Ítrio/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Antimetabólitos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Ensaios Clínicos como Assunto , Terapia Combinada , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais , Fatores de Tempo , Distribuição Tecidual , Gencitabina
11.
Clin Cancer Res ; 10(10): 3552-61, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15161715

RESUMO

PURPOSE: The early detection and diagnosis of pancreatic cancer remains a major clinical challenge in which imaging procedures have a central role. The purpose of this study was to evaluate a pretargeting method with a bispecific PAM4 (bsPAM4; anti-MUC1) antibody for radioimmunoscintigraphy of experimental human pancreatic cancer. EXPERIMENTAL DESIGN: A bispecific F(ab')(2) antibody was generated from chimeric PAM4 Fab' and murine 734 (anti-indium-diethylenetriaminepentaacetic acid) Fab' fragments and then used in conjunction with 2 peptide haptens ((111)In-IMP-156 and (99m)Tc-IMP-192). Biodistribution studies and radioimmunoscintigraphic imaging properties of the radiolabeled bsPAM4, and pretargeted, radiolabeled peptides were examined in the CaPan1 human pancreatic tumor grown as s.c. xenografts in athymic nude mice. Tumor uptake and tumor:nontumor ratios were compared with a nontargeting irrelevant anti-CD20, bispecific rituximab, radiolabeled peptides alone, and with directly labeled PAM4. RESULTS: Biodistribution results indicated significantly greater tumor uptake of radiolabeled peptides at 3 h after injection when pretargeting was performed with bsPAM4 as compared with the bispecific rituximab [20.2 +/- 5.5 percentage of injected dose per gram of tissue (%ID/g) versus 0.9 +/- 0.1%ID/g, respectively, for (111)In-IMP-156, and 16.8 +/- 4.8%ID/g versus 1.1 +/- 0.2%ID/g, respectively, for (99m)Tc-IMP-192]. Similar results were obtained at the 24-h time point. Tumor:nontumor ratios were >30 for all of the tissues except the kidneys, where a ratio of 7.8 +/- 2.8 was observed. By immunoscintigraphy, tumors could be visualized as early as 30 min after injection of the radiolabeled peptide. CONCLUSIONS: These studies demonstrate the feasibility of using the pretargeted, bispecific antibody technology for nuclear imaging of pancreatic cancer. The advantage of pretargeted bsPAM4 antibody as an imaging platform is the high specificity for pancreatic cancer as compared with the physicochemical parameters identified by current imaging technologies.


Assuntos
Anticorpos Biespecíficos/química , Neoplasias Pancreáticas/imunologia , Radioimunodetecção/métodos , Animais , Anticorpos/química , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Antígenos CD20/biossíntese , Linhagem Celular Tumoral , Haptenos/química , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/química , Peptídeos/química , Rituximab , Fatores de Tempo
12.
Clin Cancer Res ; 10(19): 6606-11, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15475450

RESUMO

PURPOSE: CD74 (HLA-DR-associated invariant chain) plays a role in antigen presentation. In addition to its expression on antigen-presenting cells, it is expressed by carcinomas of renal, lung, gastric, and thymic origin and by certain sarcomas. The restricted expression of CD74 by normal tissues and its very rapid internalization make CD74 an attractive therapeutic target for both cancer and immunologic diseases. Preclinical efficacy of anti-CD74 monoclonal antibody (mAb) therapy has been demonstrated in B-lymphoma models. Because there are few validated antigenic targets in multiple myeloma, CD74 expression was examined. EXPERIMENTAL DESIGN: CD74 expression was assessed by immunohistochemistry in bone marrow biopsies of known multiple myeloma cases. Its expression was measured by flow cytometry in multiple myeloma lines, and CD74 mRNA expression was determined by reverse transcription-PCR. In addition, the in vitro antiproliferative effect of LL1 mAb was evaluated on a CD74+ multiple myeloma cell line using a [3H]thymidine incorporation assay. RESULTS: CD74 expression was observed in 19 of 22 cases of multiple myeloma, with most expressing moderate to high levels in the majority of malignant plasma cells. CD74 was expressed by most multiple myeloma cell lines, as was CD74 mRNA, at levels mirroring CD74 protein. Also, unlabeled LL1 mAb mediated in vitro growth inhibition of a CD74+ multiple myeloma cell line. CONCLUSIONS: CD74 expression is frequent in multiple myeloma, with predominant expression by the malignant plasma cells. Because anti-CD74 mAbs internalize very rapidly and LL1 mAb has shown efficacy in B-lymphoma models, CD74 represents a novel and promising target for treatment of multiple myeloma. Therefore, LL1 mAb is well suited as a carrier of radionuclides, drugs, or toxins, and also has activity as an unlabeled mAb, thereby supporting its development for this unmet need in cancer therapy.


Assuntos
Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Mieloma Múltiplo/patologia , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Diferenciação de Linfócitos B/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imuno-Histoquímica , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Plasmócitos/química , Plasmócitos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina/farmacocinética , Trítio
13.
Oncotarget ; 6(6): 4274-85, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25595893

RESUMO

PAM4 is a monoclonal antibody showing high specificity for pancreatic ductal adenocarcinoma (PDAC). Humanized PAM4 labeled with 90Y in combination with low-dose gemcitabine has shown promising therapeutic activity, and is being evaluated in a phase III clinical trial. Prior efforts have suggested that PAM4 potentially reacts with MUC5AC, a secretory mucin expressed de novo in early pancreatic neoplasia and retained throughout disease progression. In present study, we provide further evidence validating MUC5AC as the PAM4 antigen, and locate PAM4-reactive epitope within the N-terminal cysteine-rich subdomain 2 (Cys2), thus differentiating PAM4 from most anti-MUC5AC antibodies known to-date. Specifically, we show (i) PAM4-antigen and MUC5AC were co-localized in multiple human cancer cell lines, including Capan-1, BxPC-3, and CFPAC-1; (ii) MUC5AC-specific siRNA prominently reduced the expression of both MUC5AC and PAM4-antigen in CFPAC-1 cells; (iii) PAM4 preferentially binds to the void-volume fractions from Sepharose-CL2B chromatography of Capan-1 culture supernatants, which were revealed by Western blot to display the ladder pattern characteristic of oligomeric MUC5AC; and (iv) the N-terminal Cys2 within several recombinant MUC5AC fragments is essential for binding to PAM4. These findings shed light on the mechanism of PAM4-based diagnosis and treatment for pancreatic cancer, and guide further exploration of its clinical utility.


Assuntos
Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/metabolismo , Mucina-5AC/metabolismo , Neoplasias Pancreáticas/metabolismo , Antígenos de Neoplasias/análise , Antígenos de Neoplasias/química , Western Blotting , Linhagem Celular Tumoral , Epitopos/análise , Epitopos/química , Humanos , Microscopia de Fluorescência , Mucina-5AC/química
14.
Arch Pathol Lab Med ; 138(2): 220-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24476519

RESUMO

CONTEXT: PAM4 is a monoclonal antibody that shows high specificity for pancreatic ductal adenocarcinoma (PDAC) and its neoplastic precursor lesions. A PAM4-based serum immunoassay is able to detect 71% of early-stage patients and 91% with advanced disease. However, approximately 20% of patients diagnosed with chronic pancreatitis (CP) are also positive for circulating PAM4 antigen. The specificity of the PAM4 antibody is critical to the interpretation of the serum-based and immunohistochemical assays for detection of PDAC. OBJECTIVE: To determine whether PAM4 can differentiate PDAC from nonneoplastic lesions of the pancreas. DESIGN: Tissue microarrays of PDAC (N = 43) and surgical specimens from CP (N = 32) and benign cystic lesions (N = 19) were evaluated for expression of the PAM4 biomarker, MUC1, MUC4, CEACAM5/6, and CA19-9. RESULTS: PAM4 and monoclonal antibodies (MAbs) to MUC1, MUC4, CEACAM5/6, and CA19-9 were each reactive with the majority of PDAC cases; however, PAM4 was the only monoclonal antibody not to react with adjacent, nonneoplastic parenchyma. Although PAM4 labeled 19% (6 of 32) of CP specimens, reactivity was restricted to pancreatic intraepithelial neoplasia associated with CP; inflamed tissues were negative in all cases. In contrast, MUC1, MUC4, CEACAM5/6, and CA19-9 were detected in 90%, 78%, 97%, and 100% of CP, respectively, with reactivity also present in nonneoplastic inflamed tissue. CONCLUSIONS: PAM4 was the only monoclonal antibody able to differentiate PDAC (and pancreatic intraepithelial neoplasia precursor lesions) from benign, nonneoplastic tissues of the pancreas. These results suggest the use of PAM4 for evaluation of tissue specimens, and support its role as an immunoassay for detection of PDAC.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/diagnóstico , Mucina-1/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/diagnóstico , Pancreatite Crônica/diagnóstico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/cirurgia , Diagnóstico Diferencial , Humanos , Imuno-Histoquímica , Gradação de Tumores , Estadiamento de Neoplasias , Pâncreas/imunologia , Pâncreas/patologia , Pâncreas/cirurgia , Pancreatectomia , Cisto Pancreático/diagnóstico , Cisto Pancreático/metabolismo , Cisto Pancreático/patologia , Cisto Pancreático/cirurgia , Ductos Pancreáticos/imunologia , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/patologia , Ductos Pancreáticos/cirurgia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Pancreatite Crônica/metabolismo , Pancreatite Crônica/patologia , Pancreatite Crônica/cirurgia , Lesões Pré-Cancerosas/diagnóstico , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Lesões Pré-Cancerosas/cirurgia , Análise Serial de Tecidos
15.
Mol Cancer Ther ; 12(6): 968-78, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23427296

RESUMO

CD74 is an attractive target for antibody-drug conjugates (ADC), because it internalizes and recycles after antibody binding. CD74 mostly is associated with hematologic tumors but is expressed also in solid cancers. Therefore, ADCs of the humanized anti-CD74 antibody, milatuzumab, were examined for the therapy of CD74-expressing solid tumors. Milatuzumab-doxorubicin and two milatuzumab-SN-38 conjugates with cleavable linkers, differing in their stability in serum and how they release SN-38 in the lysosome, were prepared. CD74 expression was determined by flow cytometry and immunohistology. In vitro cytotoxicity and in vivo therapeutic studies were conducted in the human cancer cell lines A-375 (melanoma), HuH-7 and Hep-G2 (hepatoma), Capan-1 (pancreatic), NCI-N87 (gastric), and Raji Burkitt lymphoma. The milatuzumab-SN-38 ADC was compared with SN-38 ADCs prepared with anti-Trop-2 and anti-CEACAM6 antibodies in xenografts expressing their target antigens. Milatuzumab-doxorubicin was most effective in the lymphoma model, whereas in A-375 and Capan-1 solid tumors, only milatuzumab-SN-38 showed a therapeutic benefit. Despite much lower surface expression of CD74 than Trop-2 or CEACAM6, milatuzumab-SN-38 had similar efficacy in Capan-1 as anti-Trop-2-SN-38, but in NCI-N87, anti-CEACAM6 and anti-Trop-2 conjugates were superior. Studies in two hepatoma lines at a single dose level showed significant benefit over saline controls but not against an irrelevant immunoglobulin G conjugate. CD74 is a suitable target for ADCs in some solid tumor xenografts, with efficacy largely influenced by uniformity of CD74 expression and with SN-38 conjugates providing the best therapeutic responses; SN-38 conjugates were preferable in solid cancers, whereas doxorubicin ADC was better in lymphoma tested.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Histocompatibilidade Classe II/genética , Neoplasias/tratamento farmacológico , Anticorpos Monoclonais , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Neoplasias/metabolismo , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Moléculas de Adesão Celular/metabolismo , Doxorrubicina/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Células Hep G2 , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Irinotecano , Neoplasias/imunologia , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 8(2): e55324, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23405135

RESUMO

We report the in-vivo fusion of two Hodgkin lymphomas with golden hamster cheek pouch cells, resulting in serially-transplanted (over 5-6 years) GW-532 and GW-584 heterosynkaryon tumor cells displaying both human and hamster DNA (by FISH), lymphoma-like morphology, aggressive metastasis, and retention of 7 human genes (CD74, CXCR4, CD19, CD20, CD71, CD79b, and VIM) out of 24 tested by PCR. The prevalence of B-cell restricted genes (CD19, CD20, and CD79b) suggests that this uniform population may be the clonal initiating (malignant) cells of Hodgkin lymphoma, despite their not showing translation to their respective proteins by immunohistochemical analysis. This is believed to be the first report of in-vivo cell-cell fusion of human lymphoma and rodent host cells, and may be a method to disclose genes regulating both organoid and metastasis signatures, suggesting that the horizontal transfer of tumor DNA to adjacent stromal cells may be implicated in tumor heterogeneity and progression. The B-cell gene signature of the hybrid xenografts suggests that Hodgkin lymphoma, or its initiating cells, is a B-cell malignancy.


Assuntos
Transferência Genética Horizontal , Doença de Hodgkin/genética , Animais , Linfócitos B/metabolismo , Fusão Celular/métodos , Linhagem Celular Tumoral , Cricetinae , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Transferência de Genes , Doença de Hodgkin/metabolismo , Humanos , Mesocricetus , Células Estromais/metabolismo
17.
Int J Clin Exp Pathol ; 6(11): 2476-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24228110

RESUMO

BACKGROUND: We investigated a series of pancreaticoduodenectomy and duodenal biopsies with a panel of immunohistochemical markers to identify duodenal mucosal invasion by pancreatic ductal adenocarcinoma (PDAC), including markers of poor prognosis and targets of promising novel immunotherapies. MATERIALS AND METHODS: Eighteen consecutive pancreaticoduodenectomy specimens with duodenal mucosal invasion by PDAC were examined for expression of MUC1, MUC4, MUC5AC, MUC6, mesothelin, MUC2, CDX2, and DPC4 on formalin-fixed, paraffin-embedded sections of duodenal-ampullary-pancreatic junctions. Expression of all but MUC6 was also assessed in duodenal biopsies from 12 patients with duodenal mucosal invasion by PDAC. RESULTS: The duodenal mucosa expressed MUC1 (crypts), MUC2 (goblet cells), MUC6 (Brunner glands), CDX2, and DPC4. PDACs in the duodenal mucosa from the resection (n=16-18) and biopsy (n=12) specimens were marked as follows: MUC1 100% (30/30), MUC4 83% (24/29), MUC5AC 83% (25/30), mesothelin 82% (23/28), MUC2 7% (2/30), and CDX2 36% (10/28). Loss of DPC4 expression was seen in 16 of 29 (55%) cases. Reactive mucosa adjacent to PDAC expressed MUC4, MUC5AC and mesothelin in 65% (17/26), 19% (5/27), and 19% (5/26) of cases, respectively. While MUC5AC and mesothelin had high diagnostic accuracy for detection of PDAC, MUC2, CDX2 and DPC4 expression demonstrated negative correlation with PDAC, with absent expression being highly specific for PDAC. CONCLUSION: Immunohistochemical labeling for PDAC biomarkers may aid the diagnosis of PDAC in duodenal biopsy, especially in situations where diagnosis of a pancreatic mass is challenging.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Ductal Pancreático/química , Duodeno/química , Imuno-Histoquímica , Mucosa Intestinal/química , Neoplasias Pancreáticas/química , Biópsia , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/cirurgia , Duodeno/patologia , Duodeno/cirurgia , Humanos , Mucosa Intestinal/patologia , Mucosa Intestinal/cirurgia , Invasividade Neoplásica , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Pancreaticoduodenectomia , Valor Preditivo dos Testes , Prognóstico
18.
Hum Immunol ; 73(6): 612-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22504409

RESUMO

The current poxvirus vaccine is associated with rare, but serious adverse events. Therefore, we investigated a non-replicating approach to vaccine design. Peptides encoding potential HLA-binding motifs were derived from the orthopoxvirus genes, D8L, A27L, and C12L (the IL-18-binding protein [vIL18BP105]), all of which are preserved among poxviruses that infect humans, and which may be a target of host immunity. The peptides were tested with poxvirus-vaccinated human PBMC and serum for eliciting memory responses, as well as with splenocytes and serum from peptide-immunized, human HLA-DR04 transgenic (HLA tg) mice. vIL18BP105 induced 5-fold proliferation of vaccinated-donor PBMC over non-vaccinated (P<0.001), including IL-2-producing CD8+ cells. Serum IgG recognizing vIL18BP105 was detected (P<0.002 vs non-vaccinated) by ELISA. Viral peptides were conjugated to the HLA-targeting mAb, L243, for immunization of HLA tg mice. Splenocytes from vIL18BP105-L243-immunized mice proliferated upon exposure to vIL18BP105 (P<0.001). Proliferating splenocytes were interferon-γ-producing CD4(+)CD45RA(neg). vIL18BP105-L243-immunized mice generated IgG more rapidly than free-peptide-immunized mice. Peptide-specific antibody was also detected when different L243-peptide conjugates were combined. vIL18BP, by eliciting human memory responses, is a viable antigen for inclusion in a virus-free vaccine. The immunogenicity of peptides was boosted by conjugation to L243, whether administered alone or combined.


Assuntos
Genes Virais/imunologia , Antígenos HLA/imunologia , Imunoconjugados/imunologia , Interleucina-18/imunologia , Orthopoxvirus/imunologia , Varíola/prevenção & controle , Vacinação , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Sítios de Ligação , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Antígenos HLA/química , Antígenos HLA/genética , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-18/química , Interleucina-2/biossíntese , Interleucina-2/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Transgênicos , Orthopoxvirus/efeitos dos fármacos , Orthopoxvirus/genética , Peptídeos/química , Peptídeos/imunologia , Peptídeos/farmacologia , Varíola/imunologia , Varíola/virologia , Baço/citologia , Baço/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
19.
Clin Cancer Res ; 17(12): 4091-100, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21527562

RESUMO

PURPOSE: Humanized antibody hPAM4 specifically binds a mucin glycoprotein expressed in pancreatic adenocarcinomas. This phase I study evaluated a single dose of (90)Y-clivatuzumab tetraxetan ((90)Y-labeled hPAM4) in patients with advanced pancreatic cancer. EXPERIMENTAL DESIGN: Twenty-one patients (4 stage III; 17 stage IV) received (111)In-hPAM4 for imaging and serum sampling before (90)Y-hPAM4. Study procedures evaluated adverse events, safety laboratories, computed tomography (CT) scans, biomarkers, pharmacokinetics, radiation dosimetry, and immunogenicity (HAHA). RESULTS: (111)In-hPAM4 showed normal biodistribution with radiation dose estimates to red marrow and solid organs acceptable for radioimmunotherapy and with tumor targeting in 12 patients. One patient withdrew before (90)Y-hPAM4; otherwise, 20 patients received (90)Y doses of 15 (n = 7), 20 (n = 9), and 25 mCi/m(2) (n = 4). Treatment was well tolerated; the only significant drug-related toxicities were (NCI CTC v.3) grade 3 to 4 neutropenia and thrombocytopenia increasing with (90)Y dose. There were no bleeding events or serious infections, and most cytopenias recovered to grade 1 within 12 weeks. Three patients at 25 mCi/m(2) encountered dose-limiting toxicity with grade 4 cytopenias more than 7 days, establishing 20 mCi/m(2) as the maximal tolerated (90)Y dose. Two patients developed HAHA of uncertain clinical significance. Most patients progressed rapidly and with CA19-9 levels increasing within 1 month of therapy, but 7 remained progression-free by CT for 1.5 to 5.6 months, including 3 achieving transient partial responses (32%-52% tumor diameter shrinkage). CONCLUSION: (90)Y-Clivatuzumab tetraxetan was well tolerated with manageable hematologic toxicity at the maximal tolerated (90)Y dose, and is a potential new therapeutic for advanced pancreatic cancer.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Antineoplásicos/efeitos adversos , Antineoplásicos/imunologia , Antineoplásicos/farmacocinética , Biomarcadores Tumorais/sangue , Carcinoma/sangue , Carcinoma/diagnóstico por imagem , Carcinoma/imunologia , Carcinoma/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Radiografia , Radiometria , Distribuição Tecidual , Resultado do Tratamento
20.
Int J Clin Exp Pathol ; 4(1): 1-12, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21228923

RESUMO

CD74, a transmembrane glycoprotein that associates with MHC II, is an important chaperone that regulates antigen presentation for immune response. In addition, CD74 is the receptor for macrophage migration-inhibitory factor which, when bound to CD74, initiates survival pathways and cell proliferation. Formalin fixed, paraffin embedded clinical specimens were evaluated by immunohistochemical procedures for expression of CD74. Overall, expression of CD74 within gastrointestinal carcinomas showed a statistically greater expression than in the normal tissue counterparts (P<0.001 or better). CD74 expression was observed in 95% of pancreatic carcinomas with the majority of cases presenting a mostly intense, diffuse labeling pattern. The results suggested a trend towards greater expression within the higher grade carcinomas (P=0.06). Colorectal and gastric carcinomas gave similar results with 60% and 86%, respectively, positive for CD74 with an intense, diffuse staining pattern. We hypothesized that precursor lesions would express levels of CD74 as high, or higher, than their respective carcinomas, since activation of survival pathways would be of particular importance at the early stages of neoplastic development. For PanIN lesions there was greater expression of CD74 within higher grade, PanIN-3 lesions, whereas the colonic adenomas showed no such trend, but overall, a higher frequency and intensity of CD74 labeling than was observed within the colon carcinomas. These findings are supportive of a role for CD74 in the development and maintenance of gastrointestinal neo-plasia, and provide a rationale for development of therapeutic agents that are able to block CD74 function, specifically within the tumor cell.


Assuntos
Adenocarcinoma/metabolismo , Adenoma/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Neoplasias do Sistema Digestório/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Adenocarcinoma/patologia , Adenoma/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias do Sistema Digestório/patologia , Humanos , Imuno-Histoquímica , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Análise Serial de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA