Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Breast Cancer Res ; 24(1): 35, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35606863

RESUMO

BACKGROUND: CCR5 is a motility chemokine receptor implicated in tumor progression, whose activation and subsequent endocytosis may identify highly aggressive breast cancer cell subtypes likely to spread into the circulatory system. METHODS: The MDA-MB-231 cell line was used to model and visualize CCR5 activation by stimulation with RANTES, in an effort to quantify CCR5 endocytosis from the cell surface to the perinuclear space. CCR5 expression was then examined in tumor-associated cells (TACs), consisting of circulating tumor cells and circulating stromal cells, isolated from the peripheral blood of 54 metastatic breast cancer (mBC) patients to evaluate these CCR5 pooling patterns as they relate to progression and survival over 2 years. RESULTS: In MB231 experiments, it was observed that CCR5 formed ~ 1 micron clusters identified as "CCR5 pools" on the surface of the cell, which in the presence of RANTES were endocytosed and translocated to the cell cytoplasm. When TACs from patients were analyzed, CCR5 pools were observed on the cell surface and translocating to the nuclear area, with CCR5 also having a positive statistical correlation between increased numbers of TACs and increased CCR5 pools on the cells. Further, it was determined that patients with very high numbers of CCR5 (> 10 CCR5 pools), specifically in the circulating stromal cells, were associated with worse progression-free survival (hazard ratio = 4.5, p = 0.002) and worse overall survival (hazard ratio = 3.7, p = 0.014). CONCLUSIONS: Using a liquid biopsy approach, we evaluated two populations of tumor-associated cells emanating from primary tumors, with data suggesting that upregulation of the motility chemokine CCR5 in TACs provides clinically relevant opportunities for treating and tracking drug targetable receptors in mBC.


Assuntos
Neoplasias da Mama , Quimiocina CCL5 , Células Neoplásicas Circulantes , Receptores CCR5 , Neoplasias da Mama/metabolismo , Quimiocina CCL5/genética , Endocitose , Feminino , Humanos , Receptores CCR5/genética , Receptores CCR5/metabolismo
2.
Invest New Drugs ; 32(3): 526-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24464266

RESUMO

INTRODUCTION: Vorinostat is a small molecule inhibitor of class I and II histone deacetylases with preclinical activity in melanoma. METHODS: We evaluated 32 patients with advanced primary cutaneous or ocular melanoma in a multi-institutional setting (PMH Phase II Consortium) with continuous daily oral vorinostat 400 mg. The primary endpoint was response rate by RECIST, with time to progression as a secondary endpoint. The study was designed to distinguish a response rate of 20 % from a RR of 5 % and to distinguish a 2 month median progression-free survival (PFS), from one of 3.1 months. The study proceeded to stage 2 following 2 of 16 responses.. We also assessed VEGF, FGF levels, P52 polymorphisms and chromatin-associated proteins as potential biomarkers. RESULTS: Therapy was associated with significant side effects, including fatigue, nausea, lymphopenia, and hyperglycemia. Eleven patients experienced at least one grade 3 or higher adverse event. There were two confirmed PRs in patients with cutaneous melanoma. Sixteen patients had stable disease and 14 patients had progressive disease for best response. In addition, two patients with cutaneous melanoma scored as stable disease had early unconfirmed partial responses with subsequent progression. Patients with stable disease or partial response (n = 18) had a median progression free survival of 5 months. (range 2-12 months). CONCLUSIONS: Vorinostat demonstrated some early responses and a high proportion of patients with stable disease, but did not meet its primary endpoint of response. Different schedules of this agent with BRAF mutation status and markers of histone acetylation could be explored in melanoma.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Melanoma/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Biomarcadores/sangue , Intervalo Livre de Doença , Feminino , Fatores de Crescimento de Fibroblastos/sangue , Inibidores de Histona Desacetilases/efeitos adversos , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/efeitos adversos , Ácidos Hidroxâmicos/farmacologia , Masculino , Melanoma/genética , Melanoma/metabolismo , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Neoplasias Cutâneas , Proteína Supressora de Tumor p53/genética , Fator A de Crescimento do Endotélio Vascular/sangue , Vorinostat , Melanoma Maligno Cutâneo
3.
Breast Cancer Res Treat ; 140(1): 23-33, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23784380

RESUMO

Inflammatory breast cancer (IBC) is the most aggressive type of advanced breast cancer characterized by rapid proliferation, early metastatic development and poor prognosis. Since there are few preclinical models of IBC, there is a general lack of understanding of the complexity of the disease. Recently, we have developed a new model of IBC derived from the pleural effusion of a woman with metastatic secondary IBC. FC-IBC02 cells are triple negative and form clusters (mammospheres) in suspension that are strongly positive for E-cadherin, ß-catenin and TSPAN24, all adhesion molecules that play an important role in cell migration and invasion. FC-IBC02 cells expressed stem cell markers and some, but not all of the characteristics of cells undergoing epithelial mesenchymal transition (EMT). Breast tumor FC-IBC02 xenografts developed quickly in SCID mice with the presence of tumor emboli and the development of lymph node and lung metastases. Remarkably, FC-IBC02 cells were able to produce brain metastasis in mice on intracardiac or intraperitoneal injections. Genomic studies of FC-IBC02 and other IBC cell lines showed that IBC cells had important amplification of 8q24 where MYC, ATAD2 and the focal adhesion kinase FAK1 are located. MYC and ATAD2 showed between 2.5 and 7 copies in IBC cells. FAK1, which plays important roles in anoikis resistance and tumor metastasis, showed 6-4 copies in IBC cells. Also, CD44 was amplified in triple-negative IBC cells (10-3 copies). Additionally, FC-IBC02 showed amplification of ALK and NOTCH3. These results indicate that MYC, ATAD2, CD44, NOTCH3, ALK and/or FAK1 may be used as potential targeted therapies against IBC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/patologia , ATPases Associadas a Diversas Atividades Celulares , Adenosina Trifosfatases/genética , Quinase do Linfoma Anaplásico , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Antígenos CD4/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Cromossomos Humanos Par 8 , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal , Feminino , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Genes myc , Humanos , Neoplasias Inflamatórias Mamárias/metabolismo , Perda de Heterozigosidade , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores Proteína Tirosina Quinases/genética , Receptor Notch3 , Receptores Notch/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Gynecol Oncol ; 129(1): 86-91, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23321064

RESUMO

OBJECTIVES: Vascular endothelial growth factors (VEGF) and their receptors have a critical role in stimulating the growth of ovarian cancer cells. Motesanib is a small molecule inhibitor of multiple receptor tyrosine kinases including VEGF receptors 1-3, as well as c-KIT and platelet-derived growth factor which are related to the VEGF family. PATIENTS AND METHODS: Twenty-two eligible patients with recurrent ovarian, fallopian tube or primary peritoneal carcinoma were treated with an oral daily dose of 125 mg of motesanib. Peripheral blood was analyzed for circulating tumor cells (CTC) and circulating endothelial cells/circulating endothelial progenitors (CEC/CEP), VEGF levels and cell-free circulating DNA (cfDNA). RESULTS: The study was abruptly halted after four patients developed posterior reversible encephalopathy syndrome. One patient had a partial response and seven patients had stable disease at the time they were removed from study treatment. Twelve of the 22 patients (50%) had indeterminate responses at trial closure. Early closure without clinical efficacy data precludes meaningful correlative studies. CONCLUSIONS: The serious central nervous system toxicity observed in patients with recurrent ovarian cancer precluded full examination of this agent in this population. There were no clear cut explanations for the high incidence of this known class effect in the study population compared with patients with other cancers.


Assuntos
Neoplasias das Tubas Uterinas/tratamento farmacológico , Indóis/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Niacinamida/análogos & derivados , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Peritoneais/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/uso terapêutico , DNA de Neoplasias/análise , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Humanos , Indóis/efeitos adversos , Pessoa de Meia-Idade , Niacinamida/efeitos adversos , Niacinamida/uso terapêutico , Oligonucleotídeos , Fator A de Crescimento do Endotélio Vascular/sangue
5.
Cancer Res ; 54(15): 4084-90, 1994 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-8033141

RESUMO

Macrophage colony-stimulating factor (M-CSF) is a known inducer of proliferation and differentiation of cells of the mononuclear phagocyte lineage, and gamma-interferon (gamma-IFN) is a known activator of mononuclear phagocytes. In this Phase I clinical trial of combined therapy with M-CSF and gamma-IFN, 36 patients were treated with 14-day continuous infusions of M-CSF at doses ranging from 10 to 140 micrograms/kg/day. In all but five patients, gamma-IFN was administered by daily s.c. injection on days 8-14 of the M-CSF infusion at doses of 0.05 or 0.1 mg/m2/day. A total of 73 courses of M-CSF and 66 courses of gamma-IFN were administered. The maximally tolerated dose combination was 120 micrograms/kg/day M-CSF, 0.1 mg/m2/day gamma-IFN. The addition of gamma-IFN did not alter the maximally tolerated dose of M-CSF therapy, although some additional toxicities were noted with combined therapy. At the 140-micrograms/kg/day M-CSF dose level, grade 4 thrombocytopenia occurred in 2 of 3 patients, with a median platelet count nadir of 26,000/mm3 after 7-10 days of M-CSF infusion. At this dose level, there was one reversible grade 3 hepatic toxicity, and one grade 3 exacerbation of underlying chronic obstructive lung disease. Peripheral blood monocytosis was observed at all M-CSF dose levels exceeding 40 micrograms/kg/day, approaching 3-fold elevations at the 100-micrograms/kg/day M-CSF dose level. The induction of monocytosis was correlated with the development of thrombocytopenia. At the conclusion of therapy with 100 micrograms/kg/day M-CSF, 0.1 mg/m2/day gamma-IFN, 78% of peripheral blood monocytes expressed the low affinity Fc gamma receptor for aggregated immunoglobulin, Fc gamma RIII (CD16), and CD14 was expressed by only 36% of the cells. This phenotype has been shown previously to be associated with cellular activation. In contrast, 35% of monocytes from patients treated with M-CSF therapy alone at the same dose expressed CD16 and 88% expressed CD14. A partial clinical response was noted in a patient with metastatic renal cell carcinoma, and minor clinical responses were observed in patients with a diffuse/follicular lymphoma, metastatic renal cell carcinoma, and metastatic thymoma. At M-CSF doses exceeding 20 micrograms/kg/day within the maximally tolerated dose range, gamma-IFN did not modulate the ability of M-CSF to reliably induce peripheral blood monocytosis. This study shows that M-CSF and gamma-IFN therapy induces the proliferation and differentiation of circulating mononuclear phagocytes.


Assuntos
Interferon gama/uso terapêutico , Fator Estimulador de Colônias de Macrófagos/uso terapêutico , Neoplasias/terapia , Adulto , Idoso , Esquema de Medicação , Quimioterapia Combinada , Feminino , Humanos , Interferon gama/efeitos adversos , Leucocitose/etiologia , Fator Estimulador de Colônias de Macrófagos/efeitos adversos , Masculino , Pessoa de Meia-Idade , Monócitos , Neoplasias/sangue , Fenótipo , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/uso terapêutico , Transtornos Respiratórios/etiologia , Trombocitopenia/etiologia
6.
Cancer Res ; 55(20): 4586-93, 1995 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-7553634

RESUMO

2B1 is a bispecific murine monoclonal antibody (BsMAb) with specificity for the c-erbB-2 and Fc gamma RIII extracellular domains. This BsMAb promotes the targeted lysis of malignant cells overexpressing the c-erbB-2 gene product of the HER2/neu proto-oncogene by human natural killer cells and mononuclear phagocytes expressing the Fc gamma RIII A isoform. In a Phase I clinical trial of 2B1, 15 patients with c-erbB-2-overexpressing tumors were treated with 1 h i.v. infusions of 2B1 on days 1, 4, 5, 6, 7, and 8 of a single course of treatment. Three patients were treated with daily doses of 1.0 mg/m2, while six patients each were treated with 2.5 mg/m2 and 5.0 mg/m2, respectively. The principal non-dose-limiting transient toxicities were fevers, rigors, nausea, vomiting, and leukopenia. Thrombocytopenia was dose limiting at the 5.0 mg/m2 dose level in two patients who had received extensive prior myelosuppressive chemotherapy. Murine antibody was detectable in serum following 2B1 administration, and its bispecific binding properties were retained. The pharmacokinetics of this murine antibody were variable and best described by nonlinear kinetics with an average t 1/2 of 20 h. Murine antibody bound extensively to all neutrophils and to a proportion of monocytes and lymphocytes. The initial 2B1 treatment induced more than 100-fold increases in circulating levels of tumor necrosis factor-alpha, interleukin 6, and interleukin 8 and lesser rises in granulocyte-monocyte colony-stimulating factor and IFN-gamma. Brisk human anti-mouse antibody responses were induced in 14 of 15 patients. Several minor clinical responses were observed, with reductions in the thickness of chest wall disease in one patient with disseminated breast cancer. Resolution of pleural effusions and ascites, respectively, were noted in two patients with metastatic colon cancer, and one of two liver metastases resolved in a patient with metastatic colon cancer. Treatment with 2B1 BsMAb has potent immunological consequences. The maximum tolerated dose and Phase II daily dose for patients with extensive prior myelosuppressive chemotherapy was 2.5 mg/m2. Continued dose escalation is required to identify the maximally tolerated dose for patients who have been less heavily pretreated.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Neoplasias/terapia , Receptor ErbB-2/imunologia , Receptores de IgG/imunologia , Adulto , Idoso , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/farmacocinética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Feminino , Humanos , Imunoterapia , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas de Neoplasias/imunologia , Neutrófilos/imunologia , Proto-Oncogene Mas , Fatores de Tempo , Distribuição Tecidual , Fator de Necrose Tumoral alfa/metabolismo
7.
Cancer Res ; 61(12): 4750-5, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11406547

RESUMO

Antitumor monoclonal antibodies must bind to tumor antigens with high affinity to achieve durable tumor retention. This has spurred efforts to generate high affinity antibodies for use in cancer therapy. However, it has been hypothesized that very high affinity interactions between antibodies and tumor antigens may impair efficient tumor penetration of the monoclonal antibodies and thus diminish effective in vivo targeting (K. Fujimori et al., J. Nucl. Med., 31: 1191-1198, 1990). Here we show that intrinsic affinity properties regulate the quantitative delivery of antitumor single-chain Fv (scFv) molecules to solid tumors and the penetration of scFv from the vasculature into tumor masses. In biodistribution studies examining a series of radioiodinated scFv mutants with affinities ranging from 10(-7)-10(-11) M, quantitative tumor retention did not significantly increase with enhancements in affinity beyond 10(-9) M. Similar distribution patterns were observed when the scFv were evaluated in the absence of renal clearance in anephric mice, indicating that the rapid renal clearance of the scFv was not responsible for these observations. IHC and IF evaluations of tumor sections after the i.v. administration of scFv affinity mutants revealed that the lowest affinity molecule exhibited diffuse tumor staining whereas the highest affinity scFv was primarily retained in the perivascular regions of the tumor. These results indicate that antibody-based molecules with extremely high affinity have impaired tumor penetration properties that must be considered in the design of antibody-based cancer therapies.


Assuntos
Afinidade de Anticorpos , Antígenos de Neoplasias/imunologia , Fragmentos de Imunoglobulinas/imunologia , Fragmentos de Imunoglobulinas/metabolismo , Neoplasias Ovarianas/metabolismo , Receptor ErbB-2/imunologia , Animais , Antígenos de Neoplasias/metabolismo , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Humanos , Rim/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos , Camundongos SCID , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/imunologia , Coelhos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Res ; 56(1): 113-20, 1996 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8548750

RESUMO

The bispecific murine monoclonal antibody (MAb) 1A10 has specificity for the human transferrin receptor (TfR) and the human tumor-associated antigen gp40. This antibody, therefore, functions as an "antigen fork" by binding to two distinct antigens on the same malignant cell. Highly purified 1A10 inhibits the growth of cells coexpressing high levels of human TfR and the tumor-associated antigen gp40 by binding to both target antigens. In SW948 cells, the majority of 1A10 binding is via its gp40 specificity, and half-maximal inhibition of cell growth by 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay requires 20-30-micrograms/ml concentrations of 1A10. The binding of 1A10 correlates with growth inhibition in the cell lines HT-29, SK-OV-3, OVCAR-2, and OVCAR-3. The growth of OVCAR-10 cells, which express little gp40 and TfR, is not inhibited by 1A10. However, SK-BR-3 cells, which express abundant gp40 and extremely high levels of TfR, are insensitive to the effects of 1A10. In some cell lines, combined exposure to 1A10 and the iron chelator deferoxamine mesylate has synergistic antiproliferative effects. A single i.p. dose of 600 micrograms 1A10 is sufficient to achieve an estimated tumor concentration of at least 30 micrograms/ml for 7 days in C.B17/Icr-scid mice bearing SW948 human tumor xenografts. Treatment of scid mice bearing day 2 or day 4 SW948 xenografts with single or multiple 1A10 doses inhibits tumor growth in a dose-related fashion. Antitumor effects are not seen with therapy using either parental antibody of 1A10. The antiproliferative properties of 1A10 in tumor cells overexpressing gp40 and TfR suggest avenues for the development of new bispecific antibody-promoted treatment strategies.


Assuntos
Anticorpos Biespecíficos/imunologia , Antígenos de Neoplasias/imunologia , Glicoproteínas de Membrana/imunologia , Receptores da Transferrina/imunologia , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ligação Competitiva , Divisão Celular/efeitos dos fármacos , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias Experimentais/terapia , Ensaio Radioligante , Células Tumorais Cultivadas
9.
J Clin Oncol ; 15(5): 1994-2007, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9164211

RESUMO

PURPOSE: To establish the maximum-tolerated dose (MTD) and define the toxicities of a single-dose infusion of PNU-214565, a recombinant Escherichia coli-derived fusion protein of Staphylococcal enterotoxin A (SEA) and the Fab-fragment of the C242 monoclonal antibody in patients with advanced colorectal and pancreatic carcinomas. To investigate the capability of PNU-214565 to induce a superantigen (SAg) response resulting in cytokine production and tumor regression. PATIENTS AND METHODS: Twenty-one patients (age range, 39 to 76 years; median, 64; 12 men, nine women; 18 colorectal, three pancreatic cancers) were treated with a single 3-hour infusion of PNU-214565, with doses ranging from 0.01 to 1.5 ng/kg. All patients had prior chemotherapy and a good performance status Eastern Cooperative Oncology Group [ECOG] performance status [PS] = 0 [n = 10]; PS = 1 [n = 11]), 10 had prior radiation, and 18 had prior surgery. RESULTS: Fever and hypotension were the most common toxicities. Fever of any grade occurred in 16 of 21 patients (76%): four of 21 (19%) with grade 2 and two of 21 (9.5%) with grade 3. Hypotension of any grade occurred in 13 of 21 (62%): four of 21 with grade 2 and one of 21 (5%) with grade 3. Interleukin-2 (IL-2) and tumor necrosis factor alpha (TNF alpha) induction correlated with toxicity. In the two patients with grade 3 fever, peak IL-2 and TNF alpha levels were 2.9 IU/mL and 165 pg/mL, and 8.3 IU/mL and 245 pg/mL, respectively. Transient, > or = 50% decreases in circulating monocytes were observed in 17 of 21 patients as early as 0.5 hours (median time, 2 hours) from the start of infusion. Decreases (mean 33%) in circulating lymphocytes were observed in seven of 21 patients. All three patients with grade 3 toxicity were treated at the 0.5-ng/kg dose. The significance of baseline anti-SEA, human antimouse antibody (HAMA), CA242-soluble antigen levels, and T-cell receptor variable beta region (TCR V beta) subsets and histocompatibility leukocyte antigen-DR (HLA-DR) genotypes was assessed as possible predictors of toxicity. All toxicities were transient and easily managed. No grade 3 toxicity occurred at the higher dose levels. CONCLUSION: PNU-214565, a SAg-based tumor targeted therapy, is safe when given as a single 3-hour infusion at doses up to 1.5 ng/kg. The MTD for a single dose was not determined. The safety of a repeated dose schedule is currently under investigation, beginning with doses determined to be safe in this trial.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/terapia , Enterotoxinas/uso terapêutico , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Imunoterapia , Imunotoxinas/uso terapêutico , Neoplasias Pancreáticas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Neoplasias Retais/terapia , Superantígenos/imunologia , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/sangue , Antígenos de Neoplasias/sangue , Neoplasias do Colo/imunologia , Enterotoxinas/efeitos adversos , Enterotoxinas/sangue , Feminino , Genótipo , Antígenos HLA-DR/genética , Humanos , Fragmentos Fab das Imunoglobulinas/efeitos adversos , Fragmentos Fab das Imunoglobulinas/sangue , Imunoterapia/efeitos adversos , Interleucina-2/sangue , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/sangue , Neoplasias Retais/imunologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Clin Cancer Res ; 4(8): 1903-14, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9717818

RESUMO

Antibody-directed, superantigen-induced cytotoxicity has been shown to have potent in vitro and in vivo antitumor effects in preclinical models. In the present study, PNU-214565, a recombinant fusion protein consisting of the Fab of the monoclonal antibody C242 and staphylococcal enterotoxin A (SEA), was used in an escalating repeat dose Phase I clinical trial in patients with advanced gastrointestinal malignancies. A prior single-dose Phase I clinical trial had demonstrated safety at doses of 1.5 ng/kg with toxicities of fever and hypotension that were not dose related. Twenty-seven patients (age range, 36-75 years; median, 62; 14 males and 13 females; 23 colorectal and 4 pancreatic) were treated in the present study with one cycle of four consecutive daily 3-h infusions of PNU-214565 at doses of 0.15 ng/kg (n = 3); 0.5 ng/kg (n = 3), 1.5 ng/kg (n = 4), 2.75 ng/kg (n = 12), and 3.5 ng/kg (n = 5). All patients had a good performance status [Eastern Cooperative Oncology Group: PS = 0 (n = 15), PS = 1 (n = 12)]. As in the single-dose trial, fever and hypotension were the most common toxicities. Dose-limiting toxicity (DLT), consisting of transient hypotension responsive to dopamine, was experienced by one patient treated at the 2.75 ng/kg dose level. One patient with pancreatic cancer metastatic to the liver experienced a partial response of hepatic metastases with stable pancreatic head abnormalities by computed tomography scan. Further dose escalation was suspended when two patients treated in a companion repeat dose Phase I study experienced DLT at the 4 ng/kg dose level. Multiparameter analyses on all patients treated in the two companion single-dose and two-repeated-dose Phase I trials revealed that the levels of patients' pretreatment anti-SEA antibodies protected against toxicity at a given drug dose. By jointly considering weight and the baseline anti-SEA concentration in a patient, it is possible to assign a PNU-214565 dose that will induce systemic cytokine release (a surrogate test to assess for the presence of uncomplexed drug and its ability to induce systemic cellular activation) without DLT. This pharmacodynamically based dosing scheme will be tested in future repeated-dose clinical trials and will define maximally tolerated doses of this powerful new immunotherapy approach.


Assuntos
Enterotoxinas/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Fragmentos de Imunoglobulinas/uso terapêutico , Imunotoxinas/uso terapêutico , Indutores de Interferon/uso terapêutico , Superantígenos/imunologia , Adulto , Idoso , Relação Dose-Resposta a Droga , Esquema de Medicação , Enterotoxinas/efeitos adversos , Feminino , Neoplasias Gastrointestinais/imunologia , Humanos , Fragmentos de Imunoglobulinas/efeitos adversos , Imunotoxinas/efeitos adversos , Infusões Intravenosas , Indutores de Interferon/efeitos adversos , Masculino , Pessoa de Meia-Idade
11.
Clin Cancer Res ; 7(5): 1181-91, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11350882

RESUMO

Granulocyte macrophage colony-stimulating factor (GM-CSF) has been shown to be an effective vaccine adjuvant because it enhances antigen processing and presentation by dendritic cells. ALVAC-CEA B7.1 is a canarypox virus encoding the gene for the tumor-associated antigen carcinoembryonic antigen (CEA) and for a T-cell costimulatory molecule, B7.1. After an initial dose escalation phase, this study evaluated vaccination with 4.5 x 10(8) plaque-forming units ALVAC-CEA B7.1 alone (n = 30) or with GM-CSF (n = 30) in patients with advanced CEA-expressing tumors to determine whether the addition of the adjuvant GM-CSF enhances induction of CEA-specific T-cells. Patients were vaccinated with vaccine intradermally every other week for 8 weeks. GM-CSF was given s.c. for 5 days beginning 2 days before vaccination. Patients with stable or responding disease after four immunizations received monthly boost injections alone or with GM-CSF. Biopsies of vaccine sites were obtained 48 h after vaccination to evaluate leukocytic infiltration and CEA expression. Induction of peripheral blood CEA-specific T-cell precursors was assessed in HLA-A2 positive patients by an ELISPOT assay looking for the production of IFN-gamma. Therapy was well tolerated. All of the patients had evidence of leukocytic infiltration and CEA expression in vaccine biopsy sites. In the patients receiving GM-CSF, leukocytic infiltrates were greater in cell number but were less likely to have a predominant lymphocytic infiltrate compared with patients receiving vaccine in the absence of the cytokine adjuvant. After four vaccinations, CEA-specific T-cell precursors were statistically increased in HLA-A2 positive patients who received vaccine alone. However, the GM-CSF plus vaccine cohort of HLA-A2 positive did not demonstrate a statistically significant increase in their CEA-specific T-cell precursor frequencies compared with baseline results. The number of prior chemotherapy regimens was negatively correlated with the generation of a T-cell response, whereas there was a positive correlation between the number of months from the last chemotherapy regimen and the T-cell response. ALVAC-CEA B7.1 is safe in patients with advanced, recurrent adenocarcinomas that express CEA, is associated with the induction of a CEA-specific T-cell response in patients treated with vaccine alone but not with vaccine and GM-CSF, and can lead to disease stabilization for up to 13 months.


Assuntos
Vacinas Anticâncer/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias/tratamento farmacológico , Vacinas Sintéticas/uso terapêutico , Adulto , Idoso , Biópsia , Vacinas Anticâncer/efeitos adversos , Quimioterapia Adjuvante , Estudos de Coortes , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Humanos , Imunidade/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Resultado do Tratamento , Vacinas Sintéticas/efeitos adversos
12.
Med Oncol ; 15(3): 191-8, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9819796

RESUMO

The inflammatory tumor lymphocytic infiltrates and spontaneous tumor regressions seen in patients with metastatic malignant melanomas suggest a cellular immune involvement. Enhancement of such responses has been the goal of R24 (GD3 ganglioside-specific) monoclonal antibody trials, alone and in combination with other agents. This study reports the results of 21 patients treated in a phase IB trial employing R24 (0, 5, 25, 50 mg/m2) administered by continuous i.v. infusion on days 1-5 followed by 3 MU each of interleukin-2 (IL-2) and alpha interferon (alpha-IFN) given subcutaneously on days 8-12, 15-19 and 22-26. R24-related toxicities occurred pre-dominantly at the 25 and 50 mg/m2 doses. One patient (50 mg/m2 R24) exhibited a dose-limiting Grade 4 anaphylaxis. Cytokine-related toxicities required IL-2/alpha-IFN dose reduction in two patients and early termination of treatment in five additional patients. Nine of 20 baseline biopsies showed chronic inflammation; six with lymphocytic tumor infiltration and three where inflammation was confined to the perivascular/peritumoral spaces. No day 8 or 29 biopsies in the R24-treated groups demonstrated treatment-induced tumor lymphocytic infiltrates. However, one patient randomized to no R24 treatment, showed a significant inflammatory tumor lymphocytic infiltration at days 8 and 29. Eighteen of 21 treated patients were evaluable for response. One (5%) patient receiving IL-2/alpha-IFN alone had stable disease lasting 1.5 years. Five (28%) R24, IL-2/alpha-IFN-treated patients had stable disease ranging from 6 to 32 weeks, with one patient remaining alive 2.5 years post-treatment. Although this combined treatment program was generally well tolerated, no objective responses were seen and significant R24-induced tumor lymphocytic infiltrates were not demonstrated.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Gangliosídeos/imunologia , Interferon-alfa/uso terapêutico , Interleucina-2/uso terapêutico , Melanoma/tratamento farmacológico , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Interferon-alfa/administração & dosagem , Interleucina-2/administração & dosagem , Masculino , Melanoma/imunologia , Melanoma/patologia , Pessoa de Meia-Idade , Linfócitos T
13.
Cancer Immunol Immunother ; 45(3-4): 190-2, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9435871

RESUMO

Commencing with the discovery and characterization of bispecific antibodies, numerous investigations have shown that such antibodies are capable of redirecting cellular cytotoxicity. Clinical trials testing diverse strategies, including those targeting CD16-expressing effector cells, have been conducted or are in progress. This manuscript reviews our clinical trials efforts with bispecific antibodies and describes our experience employing multi-functional binding proteins containing tumor-targeting antibody Fab fragments linked to bacterial superantigens, such as staphylococcal enterotoxin A.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Receptor ErbB-2/imunologia , Receptores de IgG/imunologia , Superantígenos/uso terapêutico , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/imunologia , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/uso terapêutico , Superantígenos/imunologia
14.
J Hematother ; 4(5): 453-6, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8581384

RESUMO

Bispecific monoclonal antibodies (BsmAb) can be used to specifically target tumor cells for cytotoxicity mediated by defined effector cells. One such BsmAb, 2B1, targets the extracellular domains of both the c-erbB-2 protein product of the HER-2/neu oncogene and Fc gamma RIII (CD16), the Fc gamma receptor expressed by human natural killer cells, neutrophils, and differentiated mononuclear phagocytes. 2B1 promotes the conjugation of cells expressing these target antigens. It efficiently promotes the specific lysis of tumor cells expressing c-erbB-2 by human NK cells and macrophages over a broad concentration range. 2B1 selectively targets c-erbB-2-positive human tumor xenografts growing in immunodeficient SCID mice. Treatment of such mice with 2B1 plus interleukin 2 (IL-2) inhibits the growth of early, established human tumor xenografts overexpressing c-erbB-2. A phase I clinical trial of 2B1 has been initiated to determine the toxicity profile and maximum tolerated dose (MTD) of this BsmAb and to examine the biodistribution of the antibody and the biologic effects of treatment. Preliminary results of this trial indicate that the dose-limiting toxicity for patients with extensive prior bone marrow-toxic therapy is thrombocytopenia for as yet undetermined reasons. Toxicities of fevers, rigors, and associated constitutional symptoms are explained, in part, by treatment-induced systemic expression of cytokines, such as tumor necrosis factor-alpha. Circulating, functional BsmAb is easily detectible in treatment patients' sera and exhibits complex elimination patterns. HAMA and anti-idiotypic treatment-induced antibodies are induced by 2B1 treatment. Some preliminary indications of clinical activity have been observed. BsmAb therapy targeting tumor antigens and Fc gamma RIII has potent immunologic effects. Future studies will include the development of more relevant animal models for BsmAb therapy targeting human Fc gamma RIII. The ongoing phase I trial will be completed to identify the MTD for patients without extensive prior bone marrow-toxic chemotherapy and radiation. A phase II clinical trial of 2B1 therapy in women with metastatic breast cancer is planned, as is a phase I trial incorporating treatment with both 2B1 and IL-2.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/terapia , Fagócitos/imunologia , Receptor ErbB-2/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Citocinas/metabolismo , Feminino , Febre/induzido quimicamente , Humanos , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias/imunologia , Trombocitopenia/induzido quimicamente
15.
Cancer Immunol Immunother ; 42(3): 141-50, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8640842

RESUMO

2B1 is a bispecific murine monoclonal antibody (bsmAb) targeting the c-erbB-2 and CD16 (Fc gamma RIII) antigens. c-erbB-2 is over-expressed by a variety of adenocarcinomas, and CD16, the low-affinity Fc gamma receptor for aggregated immunoglobulins, is expressed by polymorphonuclear leukocytes (PMN), natural killer (NK) cells and differentiated mononuclear phagocytes. 2B1 potentiates the in vitro lysis of c-erb-2 over-expressing tumors by NK cells and macrophages. In this report, the interactions between 2B1 and PMN were investigated to assess the impact of these associations on in vitro 2B1-promoted tumor cytotoxicity by human NK cells. The peak binding of 2B1 to PMN was observed at a concentration of 10 microgram/ml 2B1. However, 2B1 rapidly dissociated from PMN in vitro at 37 degrees C in non-equilibrium conditions. This dissociation was not caused by CD16 shedding. When PMN were labeled witn 125I-2B1 and incubated at 37 degrees C and the supernatants examined by HPLC analysis, the Fab regions of dissociated 2B1 were not complexed with shed CD16 extracellular domain. While most of the binding of 2B1 PMN was solely attributable to Fab-directed binding to Fc gamma RIII, PMN-associated 2B1 also bound through Fc gamma-domain/Fc gamma RII interactions. 2B1 did not promote in vitro PMN cytotoxicity against c-erbB-2-expressing SK-OV-3 tumor cells. When PMN were coincubated with peripheral blood lymphocytes, SK-OV-3 tumor and 2B1, the concentration of 2B1 required for maximal tumor lysis was lowered. Although PMN may serve as a significant competitive binding pool of systemically administered 2B1 in vivo, the therapeutic potential of the targeted cytotoxicity properties of this bsmAb should not be compromised.


Assuntos
Anticorpos Biespecíficos/metabolismo , Anticorpos Monoclonais/metabolismo , Neutrófilos/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/terapia , Receptores de IgG/metabolismo , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Citotoxicidade Imunológica , Feminino , Humanos , Imunoterapia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Neutrófilos/ultraestrutura , Neoplasias Ovarianas/imunologia , Células Tumorais Cultivadas
16.
Cancer Immunol Immunother ; 48(8): 443-55, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10550549

RESUMO

Fcgamma receptor (FcgammaR) engagement is pivotal for many effector functions of macrophages, polymorphonuclear neutrophils (PMN), and natural killer (NK) cells. Mice transgenic for the A and B isoforms of human (h) FcgammaRIII on macrophages, PMN, and NK cells were constructed to permit the study of mechanisms and potential in vivo strategies to utilize the cytotoxic effector and antigen-presenting functions of cells expressing the hFcgammaR. The present report characterizes the phenotypic and functional expression of hFcgammaRIII in transgenic mice derived by crossing hFcgammaRIIIA and hFcgammaRIIIB transgenic mice. Interleukin-2 (IL-2) induces hFcgammaRIII expression by myeloid cells and their precursors, and these transgenic receptors promote in vitro cytotoxicity and anti-hFcgammaRIII antibody internalization. Splenocytes from untreated and IL-2-treated hFcgammaRIIIA, hFcgammaRIIIB, and hFcgammaRIIIA/B mice exhibited enhanced in vitro cytotoxicity toward HER-2/neu-overexpressing SK-OV-3 human ovarian carcinoma cells when incubated with the murine bispecific mAb 2B1, which has specificity for HER-2/neu and hFcgammaRIII. These results indicate that hFcgammaRIII transgenes are expressed on relevant murine cellular subsets, exhibit inducible up-regulation patterns similar to those seen in humans, and code for functional proteins. hFcgammaRIII transgenic mice exhibiting specific cellular subset expression will permit the examination of strategies designed to enhance hFcgammaRIII-dependent immunological effector functions and will provide a model system in which to evaluate preclinically potential candidate molecules that recognize hFcgammaRIII for the immunotherapy of cancer.


Assuntos
Receptores de IgG/genética , Animais , Citotoxicidade Celular Dependente de Anticorpos , Células Apresentadoras de Antígenos/imunologia , Medula Óssea/metabolismo , Cruzamento , Endocitose , Feminino , Humanos , Interleucina-2/farmacologia , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos SCID , Camundongos Transgênicos , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de IgG/biossíntese , Receptores de IgG/imunologia , Baço/citologia , Baço/metabolismo , Células Tumorais Cultivadas
17.
J Immunol ; 151(5): 2877-86, 1993 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8103070

RESUMO

Bispecific monoclonal antibodies (BsmAb) with specificity for tumor Ag and effector cell trigger molecules have been shown to redirect the cytotoxicity of several peripheral blood mononuclear cell populations against relevant tumor. The BsmAb, 2B1, binds to the extracellular domain of the c-erbB-2 gene product of the HER2/neu proto-oncogene and to CD16. In this report, the binding and cytotoxic characteristics of 2B1 are presented. Maximal saturation binding of 2B1 to PBL and c-erbB-2 expressing SK-OV-3 cells occurred in the 1 microgram/ml concentration range. However, substantial lysis potentiation was observed at 1000-fold lower BsmAb concentrations. Optimal tumor lysis was obtained when the BsmAb, PBL, and target cells were continuously coincubated. When PBL were franked with 2B1, washed, and added to labeled targets, substantially less lysis was observed. These results suggest that the best way to therapeutically exploit the cytotoxic attributes of 2B1 may be to obtain continuous BsmAb exposure to tumor. Approaches based on franking of this BsmAb to PBL may not be warranted.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Citotoxicidade Imunológica , Animais , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Humanos , Imunoglobulina G/imunologia , Células Matadoras Ativadas por Linfocina/imunologia , Camundongos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Receptor ErbB-2 , Receptores de IgG/análise , Células Tumorais Cultivadas
18.
Cancer Immunol Immunother ; 44(5): 265-72, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9247561

RESUMO

The bispecific monoclonal antibody (bsmAb) 2B1, targeting the extracellular domain of c-erbB-2, the protein product of the HER-2/neu proto-ocogene, and Fc gamma RIII (CD16), expressed by human natural killer cells, neutrophils and differentiated monocytes, mediates the specific cytotoxic activity of these effector cells to tumor cells. A group of 24 patients with c-erbB-2-overexpressing tumors were treated with intravenously administered 2B1 in a phase I clinical trial and followed after treatment to evaluate the diversity and extent of the 2B1-induced humoral immune responses. As expected, 17 of 24 patients developed human anti-(murine Ig) antibodies (HAMA) to whole 2B1 IgG in a range from 100 ng/ml to more than 50000 ng/ml; 10 of these patients (42%) had strong (at least 1000 ng/ml) HAMA responses, some of which were still detectable at day 191. These responses were usually associated with similar reactivity to the F(ab')2 fragments of the parental antibodies 520C9 (anti-c-erbB-2) and 3G8 (anti-CD16). We sought evidence of an idiotypic cascade induction, indicating a prolonged specific treatment-induced effect on at least one selected target of 2B1. Using competition-based enzyme-linked immunosorbent assays, specific anti-idiotypic antibodies (Ab2) were detectable against 520C9 in 11 patients and against 3G8 in 13 patients. Peak anti-idiotypic antibodies generally occurred 3-5 weeks from treatment initiation, with a downward trend thereafter. There was a statistically significant correlation among the induction of significant HAMA responses, anti-idiotypic antibody production and the development of antibodies to c-erbB-2. The anti-c-erbB-2 responses, which were distinct from anti-anti-idiotypic (Ab3) antibodies, were detected in the post-treatment sera of 6/16 patients examined. No obvious correlation could be made between the development of humoral immune responses, the dose received, and the clinical response. Future investigation involving 2B1 therapy will concentrate on investigating an association of these humoral responses to any c-erbB-2-specific cellular responses. Manipulations of 2B1 therapy effects that augment immunity to c-erbB-2 could provide additional avenues for immunotherapy with this and other bispecific antibodies.


Assuntos
Adenocarcinoma/prevenção & controle , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos , Receptor ErbB-2/imunologia , Vacinação , Adenocarcinoma/secundário , Citotoxicidade Imunológica , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Receptores de IgG/imunologia
19.
Blood ; 85(10): 2910-7, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7742551

RESUMO

The small subset of circulating monocytes that express the maturation-associated CD16 antigen has recently been reported to be elevated in patients with bacterial sepsis. We now show that this novel CD16+ monocyte population is also spontaneously expanded in patients with cancer. We studied 14 patients with metastatic gastrointestinal carcinoma enrolled ina clinical trial of recombinant human macrophage colony-stimulating factor (rhMCSF) plus monoclonal antibody D612. We found that before any cytokine treatment, 12 of 14 patients constitutively displayed significant elevations in both the percentage and the absolute number of CD16+ monocytes, as compared with both normal subjects and ill patients with elevated monocyte counts but without malignancy. CD16+ monocytes accounted for 46% +/- 22% of total monocytes in the patients with cancer versus 5% +/- 3% for controls (P < .01). The increase was not attributable to infection or intercurrent illness and appeared to be associated with the underlying malignancy itself. A similar spontaneous elevation of CD16+ monocytes was observed in 35 of 44 additional patients diagnosed with a variety of other solid tumors. When patients with gastrointestinal carcinoma were treated with rhMCSF, there was a marked further increase in the percentage of CD16+ monocytes (to 83% +/- 11%), as well as in the absolute number of CD16+ cells and the level of CD16 antigen expression. In every case, the patients with cancer showed a greater CD16+ monocyte response than the maximal response obtained in normal volunteer subjects treated witha similar regimen of rhMCSF (n = 5, P < .001), suggesting that the presence of malignancy primed patients for enhanced responsiveness to rhMCSF. We hypothesize that spontaneous expansion of the CD16+ monocyte population may represent a novel biologic marker for a widespread and previously unsuspected host immune response to malignancy.


Assuntos
Fator Estimulador de Colônias de Macrófagos/farmacologia , Monócitos/citologia , Neoplasias/imunologia , Receptores de IgG/metabolismo , Adulto , Citometria de Fluxo , Humanos , Imunofenotipagem , Fator Estimulador de Colônias de Macrófagos/uso terapêutico , Metástase Neoplásica , Neoplasias/patologia
20.
Blood ; 88(4): 1215-24, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8695839

RESUMO

The CD16 receptor (Fc gamma R-III) is found on many tissue macrophages (M phi s), but its expression on circulating monocytes is restricted to a small, phenotypically distinct subset. The number of these CD16+ monocytes may be markedly increased in response to sepsis, human immunodeficiency virus infection, or metastatic malignancy. We have recently shown that the CD16+ monocyte population is selectively expanded by administration of recombinant human macrophage colony-stimulating factor (rhM-CSF). In the current study, we used the highly rhM-CSF-responsive cynomolgus primate model to further characterize this novel monocyte population. Animals treated with rhM-CSF underwent a progressive and essentially complete conversion to the CD16+ monocyte phenotype, with up to a 50-fold increase in the number of CD16+ cells. This increase was paralleled by the emergence of a population of circulating cells that morphologically resembled large granular lymphocytes (LGLs). However, quantitatively, this population corresponded closely to the number of CD16+ monocytes, and fluorescence-activated cell sorting (FACS) confirmed that they were the same. In addition to their LGL-like morphology, many rhM-CSF-induced CD16+ monocytes showed a pattern of size, granularity, and quantitative cell surface marker expression that closely resembled the pretreatment LGL/natural killer (NK) cell population but that did not resemble the pretreatment monocyte population. However, rhM-CSF-induced CD16+ monocytes could be distinguished from LGL/ NK cells by fact that they all expressed cell surface receptors for rhM-CSF, and many of them showed reduced but detectable phagocytic and respiratory burst activity. Studies of human subjects treated with rhM-CSF also showed an analogous population of "LGL-appearing" CD16+ mononuclear cells. Thus, our studies reveal a previously unsuspected ability of cells in the monocyte lineage to adopt a phenotype similar to that of LGL/NK cells. The extent of this phenotypic convergence suggests that the two lineages retain access to elements of a similar developmental pathway.


Assuntos
Antígenos de Diferenciação Mielomonocítica/análise , Células Matadoras Naturais/citologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Monócitos/citologia , Animais , Antígeno CD56/análise , Citometria de Fluxo , Humanos , Imunofenotipagem , Receptores de Lipopolissacarídeos/análise , Macaca fascicularis , Masculino , Monócitos/imunologia , Fagocitose , Receptores de IgG/análise , Proteínas Recombinantes , Explosão Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA