Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Case Rep Hematol ; 2021: 8891376, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777461

RESUMO

Inspite of medication compliance, some chronic myeloid leukemia (CML) patients will relapse/progress into an accelerated phase or blast crisis. Central nervous system (CNS) involvement is a rare manifestation of such a relapse. Here, we report a case of 23-year-old female who was diagnosed with CML in the accelerated phase and subsequently treated with imatinib. She developed early relapse in her CNS, and her treatment was switched to dasatinib and intrathecal chemotherapy with cytarabine and methotrexate. Her CNS disease went into remission, and she underwent matched unrelated donor (MUD) hematopoietic stem cell transplant (HSCT). We discuss various mechanisms of treatment failure, importance of vigilance for symptoms and signs of treatment failure/relapse, indications for use of different tyrosine kinase inhibitors (TKIs), and management of blast crises in CML.

2.
Cancer Chemother Pharmacol ; 76(5): 949-55, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26395450

RESUMO

PURPOSE: The sequence bendamustine (B) + Irinotecan (I) followed by etoposide (E) + carboplatin (C) was hypothesized to increase progression-free survival (PFS) and overall survival (OS) in untreated extensive-disease small cell lung cancer (EDSCLC) patients compared to historical controls by exploiting mitotic catastrophe. Absent expression of ERCC-1 and expression of topoisomerases were hypothesized to be predictive for PFS and OS. METHODS: This was a phase I/IIa trial in 30 patients to determine the maximum tolerated dose (MTD) of B + I and the PFS of B + I E + C with secondary end points including overall response rate (ORR) and OS. Biomarkers measured by immunohistochemistry (IHC) obtained from diagnostic specimens were correlated with outcome. RESULTS: The MTD of B + I was not reached. During treatment with B + I, there were two grade 5 toxicities from neutropenic sepsis and metabolic encephalopathy. Other toxicities included fatigue, nausea/vomiting, diarrhea, and weight loss. For the sequence, the PFS and OS were 6.0 months and 10 months, respectively. The ORR for B + I and the sequence were 82% and 83%, respectively. Topoisomerase-2 expression was predictive for TTP and OS, but absent ERCC-1 expression was not, contrary to our hypothesis. CONCLUSIONS: B + I is an active regimen in EDSCLC. Toxicities included two grade 5 events but were otherwise manageable. The novel sequence B + I E + C increased PFS and OS compared to historical controls. Correlative studies are conflicting regarding the mechanism of action of this novel sequence.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cloridrato de Bendamustina/administração & dosagem , Cloridrato de Bendamustina/efeitos adversos , Cloridrato de Bendamustina/farmacologia , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Encefalopatias Metabólicas/etiologia , Camptotecina/administração & dosagem , Camptotecina/efeitos adversos , Camptotecina/análogos & derivados , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Carcinoma de Células Pequenas/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Esquema de Medicação , Endonucleases/deficiência , Endonucleases/genética , Etoposídeo/administração & dosagem , Etoposídeo/efeitos adversos , Fadiga/induzido quimicamente , Feminino , Gastroenteropatias/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Humanos , Irinotecano , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Mitose/efeitos dos fármacos , Insuficiência de Múltiplos Órgãos/etiologia , Pneumonia/induzido quimicamente , Resultado do Tratamento , Redução de Peso/efeitos dos fármacos
4.
Adv Exp Med Biol ; 632: 159-74, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19025121

RESUMO

Anti-tumor monoclonal antibody therapy represents one of the earliest targeted therapies in clinical cancer care and has achieved great clinical promise. Complement activation mediated by anti-tumor mAbs can result in direct tumor lysis or enhancement of antibody-dependent cellular cytotoxicy. Chemotaxis of phagocytic cells by complement activation products C5a is also required for certain cancer immunotherapy such as combined beta-glucan with anti-tumor mAb therapy. However, high expression levels of membrane-bound complement regulatory proteins (mCRPs) such as CD46, CD55 and CD59 on tumors significantly limit the anti-tumor mAb therapeutic efficacy. In addition, mCRPs have been shown to directly or indirectly down-regulate adaptive T cell responses. Therefore, it is desirable to combine anti-tumor mAb therapy or tumor vaccines with the blockade of mCRPs. Such strategies so far include the utilization of neutralizing mAbs for mCRPs, small interfering RNAs or anti-sense oligos for mCRPs, and chemotherapeutic drugs or cytokines. In vitro studies have demonstrated the feasibility and efficacy of such methods, although concerns have been raised about the utilization of neutralizing mAbs in vivo due to widespread expression of mCRPs on normal cells and tissues. Strategies have been developed to address these issues and more in vivo studies are needed to further validate these combination approaches.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antígenos CD/imunologia , Antígenos CD55/imunologia , Antígenos CD59/imunologia , Vacinas Anticâncer/uso terapêutico , Proteínas do Sistema Complemento/imunologia , Estudos de Viabilidade , Humanos , Oligonucleotídeos Antissenso/uso terapêutico , RNA Interferente Pequeno/uso terapêutico
5.
Cancer Res ; 67(15): 7421-30, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671212

RESUMO

Administration of a combination of yeast-derived beta-glucan with antitumor monoclonal antibodies (mAb) has significant therapeutic efficacy in a variety of syngeneic murine tumor models. We have now tested this strategy using human carcinomas implanted in immunocompromised severe combined immunodeficient mice. Combined immunotherapy was therapeutically effective in vivo against NCI-H23 human non-small-cell lung carcinomas, but this modality was surprisingly ineffective against SKOV-3 human ovarian carcinomas. Whereas NCI-H23 tumors responded to this combination therapy with increased intratumoral neutrophil infiltration and C5a production, these responses were lacking in treated SKOV-3 tumors. Further results suggested that SKOV-3 tumors were protected by up-regulation of the membrane complement regulatory protein CD55 (decay-accelerating factor). Blockade of CD55 in vitro led to enhanced deposition of C activation product C3b and increased cytotoxicity mediated by beta-glucan-primed neutrophils. In vivo, administration of anti-CD55 mAb along with beta-glucan and anti-Her-2/neu mAb caused tumor regression and greatly improved long-term survival in animals bearing the previously resistant SKOV-3 tumors. This was accompanied by increased intratumoral neutrophil accumulation and C5a production. We conclude that CD55 suppresses tumor killing by antitumor mAb plus beta-glucan therapy (and, perhaps, in other circumstances). These results suggest a critical role for CD55 to regulate iC3b and C5a release and in turn to influence the recruitment of beta-glucan-primed neutrophils eliciting killing activity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD55/metabolismo , Carcinoma Pulmonar de Células não Pequenas/terapia , Complemento C5a/fisiologia , Infiltração de Neutrófilos/imunologia , Neoplasias Ovarianas/terapia , beta-Glucanas/uso terapêutico , Animais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Complemento C3a/fisiologia , Quimioterapia Combinada , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Infiltração de Neutrófilos/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Saccharomyces cerevisiae/metabolismo , Células Tumorais Cultivadas
6.
Folia Histochem Cytobiol ; 45(2): 107-14, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17597024

RESUMO

Beta (1-3)-D-glucans were identified almost 40 years ago as biological response modifiers that stimulated tumor rejection. In vitro studies have shown that beta-glucans bind to a lectin domain within complement receptor type 3 (CR3), or to, more recently described dectin-1 a beta-glucan specific receptor, acting mainly on phagocytic cells. In this study, we assessed the intracellular cytokine profiles of peripheral blood lymphocytes from mice bearing mammary tumors receiving i.v. anti-tumor mAbs combined or not with whole glucan particle suspension given orally (WGP, 400 microg every 24 hours). The proportions of T cells producing IL-4 and IFNgamma were determined by flow cytometry. The proportion of T cells producing IL-4 was significantly higher in tumor-bearing mice not receiving beta-glucan-enhanced therapy. Conversely, T cells from mice undergoing beta-glucan-enhanced therapy showed increased production of the Th1 cytokine IFNgamma. The switch from a Th2 to a Th1 response after WGP therapy was possibly mediated by intestinal mucosal macrophages releasing IL-12.


Assuntos
Imunidade Celular/imunologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Células Th1/imunologia , Células Th2/imunologia , beta-Glucanas/administração & dosagem , beta-Glucanas/uso terapêutico , Administração Oral , Animais , Anticorpos Monoclonais/imunologia , Quimioterapia Adjuvante , Feminino , Imunoterapia , Interferon gama/biossíntese , Interleucina-12/biossíntese , Interleucina-4/biossíntese , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
7.
J Immunol ; 177(3): 1661-9, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16849475

RESUMO

Anti-tumor mAbs hold promise for cancer therapy, but are relatively inefficient. Therefore, there is a need for agents that might amplify the effectiveness of these mAbs. One such agent is beta-glucan, a polysaccharide produced by fungi, yeast, and grains, but not mammalian cells. Beta-glucans are bound by C receptor 3 (CR3) and, in concert with target-associated complement fragment iC3b, elicit phagocytosis and killing of yeast. Beta-glucans may also promote killing of iC3b-opsonized tumor cells engendered by administration of anti-tumor mAbs. In this study, we report that tumor-bearing mice treated with a combination of beta-glucan and an anti-tumor mAb show almost complete cessation of tumor growth. This activity evidently derives from a 25-kDa fragment of beta-glucan released by macrophage processing of the parent polysaccharide. This fragment, but not parent beta-glucan, binds to neutrophil CR3, induces CBRM 1/5 neoepitope expression, and elicits CR3-dependent cytotoxicity. These events require phosphorylation of the tyrosine kinase, Syk, and consequent PI3K activation because beta-glucan-mediated CR3-dependent cytotoxicity is greatly decreased by inhibition of these signaling molecules. Thus, beta-glucan enhances tumor killing through a cascade of events, including in vivo macrophage cleavage of the polysaccharide, dual CR3 ligation, and CR3-Syk-PI3K signaling. These results are important inasmuch as beta-glucan, an agent without evident toxicity, may be used to amplify tumor cell killing and may open new opportunities in the immunotherapy of cancer.


Assuntos
Complemento C3b/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Linfoma/imunologia , Antígeno de Macrófago 1/fisiologia , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/imunologia , beta-Glucanas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica , Linhagem Celular Tumoral , Complemento C3b/metabolismo , Humanos , Injeções Intravenosas , Linfoma/enzimologia , Linfoma/mortalidade , Linfoma/terapia , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Opsonizantes/metabolismo , Análise de Sobrevida , Quinase Syk , Leveduras/imunologia , beta-Glucanas/metabolismo , beta-Glucanas/uso terapêutico
8.
Blood ; 107(2): 835-40, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16179370

RESUMO

Myelotoxic injury in the bone marrow (BM) as a consequence of total body irradiation (TBI) or granulocyte colony-stimulating factor (G-CSF) mobilization results in the deposition of iC3b on BM stroma (stroma-iC3b). In the present study, we have examined how stroma-iC3b interacts with hematopoietic progenitor cells (HPCs) and the role of complement (C) and complement receptor 3 (CR3) in BM injury/repair. We demonstrate here that stroma-iC3b tethers HPCs via the inserted (I) domain of HPC complement receptor 3 (CR3, CD11b/CD18, Mac-1). Following irradiation, stroma-iC3b was observed in the presence of purified IgM and normal mouse serum (NMS), but not serum from Rag-2(-/-) mice, implicating a role for antibody (Ab) and the classic pathway of C activation. Furthermore, a novel role for soluble yeast beta-glucan, a ligand for the CR3 lectin-like domain (LLD), in the priming of CR3(+) HPC is suggested. Soluble yeast beta-glucan could enhance the proliferation of tethered HPCs, promote leukocyte recovery following sublethal irradiation, and increase the survival of lethally irradiated animals following allogeneic HPC transplantation in a CR3-dependent manner. Taken together, these observations suggest a novel role for C, CR3, and beta-glucan in the restoration of hematopoiesis following injury.


Assuntos
Medula Óssea/imunologia , Medula Óssea/lesões , Antígenos CD18/metabolismo , Complemento C3b/imunologia , Complemento C3b/metabolismo , Antígeno de Macrófago 1/imunologia , beta-Glucanas/metabolismo , Animais , Medula Óssea/metabolismo , Antígenos CD18/imunologia , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/fisiologia , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Imunoglobulina M/imunologia , Lectinas , Leucócitos/citologia , Leucócitos/metabolismo , Antígeno de Macrófago 1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Saccharomyces cerevisiae , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , beta-Glucanas/imunologia
9.
Expert Opin Biol Ther ; 5(5): 691-702, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15934844

RESUMO

Beta-glucans, biological response modifiers (BRMs) derived from the cell walls of yeast and other sources, have been demonstrated to prime leukocyte complement receptor 3 (CR3), thus enabling these cells to kill tumours opsonised with complement fragment iC3b. Many tumours activate complement via the classical pathway mediated by antitumour monoclonal antibodies (mAbs) or natural antibodies. Studies into the cellular and molecular mechanisms of action have demonstrated that orally administrated yeast beta-glucans are ingested and processed by macrophages. These macrophages secrete the active moiety that primes neutrophil CR3 to kill iC3b-opsonised tumour cells. Extensive studies in preclinical animal tumour models have demonstrated the efficacy of combined oral particulate yeast beta-glucan with antitumour mAb therapy in terms of tumour regression and long-term survival. It is proposed that the addition of beta-glucan will further improve the clinical therapeutic efficacy of antitumour mAbs in cancer patients.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Neoplasias/imunologia , Leveduras/imunologia , beta-Glucanas/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , beta-Glucanas/uso terapêutico
10.
J Immunol ; 174(11): 7050-6, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905548

RESUMO

Intravenous and orally administered beta-glucans promote tumor regression and survival by priming granulocyte and macrophage C receptor 3 (CR3, iC3bR and CD11b/CD18) to trigger the cytotoxicity of tumor cells opsonized with iC3b via anti-tumor Abs. Despite evidence for priming of macrophage CR3 by oral beta-glucan in vivo, the current study in C57BL/6 and BALB/c mice showed that granulocytes were the essential killer cells in mAb- and oral beta-glucan-mediated tumor regression, because responses were absent in granulocyte-depleted mice. Among granulocytes, neutrophils were the major effector cells, because tumor regression did not occur when C5a-dependent chemotaxis was blocked with a C5aR antagonist, whereas tumor regression was normal in C3aR(-/-) mice. Neutrophil recruitment by C5a in vivo required amplification via leukotriene B(4), because both C5a-mediated leukocyte recruitment into the peritoneal cavity and tumor regression were suppressed in leukotriene B(4)R-deficient (BLT-1(-/-)) mice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Quimiotaxia de Leucócito/imunologia , Complemento C5a/fisiologia , Leucotrieno B4/fisiologia , Linfoma/imunologia , Linfoma/terapia , Infiltração de Neutrófilos/imunologia , beta-Glucanas/uso terapêutico , Administração Oral , Animais , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/genética , Complemento C3a/fisiologia , Granulócitos/citologia , Granulócitos/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/genética , Receptores do Leucotrieno B4/deficiência , Receptores do Leucotrieno B4/genética , Receptores do Leucotrieno B4/fisiologia , beta-Glucanas/administração & dosagem
11.
J Mol Histol ; 35(3): 233-45, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15339043

RESUMO

Chemokines, small pro-inflammatory chemoattractant cytokines, that bind to specific G-protein-coupled seven-span transmembrane receptors present on plasma membranes of target cells are the major regulators of cell trafficking. In addition some chemokines have been reported to modulate cell survival and growth. Moreover, compelling evidence is accumulating that cancer cells may employ several mechanisms involving chemokine-chemokine receptor axes during their metastasis that also regulate the trafficking of normal cells. Of all the chemokines, stromal-derived factor-1 (SDF-1), an alpha-chemokine that binds to G-protein-coupled CXCR4, plays an important and unique role in the regulation of stem/progenitor cell trafficking. First, SDF-1 regulates the trafficking of CXCR4+ haemato/lymphopoietic cells, their homing/retention in major haemato/lymphopoietic organs and accumulation of CXCR4+ immune cells in tissues affected by inflammation. Second, CXCR4 plays an essential role in the trafficking of other tissue/organ specific stem/progenitor cells expressing CXCR4 on their surface, e.g., during embryo/organogenesis and tissue/organ regeneration. Third, since CXCR4 is expressed on several tumour cells, these CXCR4 positive tumour cells may metastasize to the organs that secrete/express SDF-1 (e.g., bones, lymph nodes, lung and liver). SDF-1 exerts pleiotropic effects regulating processes essential to tumour metastasis such as locomotion of malignant cells, their chemoattraction and adhesion, as well as plays an important role in tumour vascularization. This implies that new therapeutic strategies aimed at blocking the SDF-1-CXCR4 axis could have important applications in the clinic by modulating the trafficking of haemato/lymphopoietic cells and inhibiting the metastatic behaviour of tumour cells as well. In this review, we focus on a role of the SDF-1-CXCR4 axis in regulating the metastatic behaviour of tumour cells and discuss the molecular mechanisms that are essential to this process.


Assuntos
Movimento Celular/fisiologia , Quimiocinas CXC/metabolismo , Quimiotaxia/fisiologia , Neoplasias/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais , Animais , Adesão Celular/fisiologia , Quimiocina CXCL12 , Humanos , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Neoplasias/patologia
12.
J Immunol ; 173(2): 797-806, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15240666

RESUMO

Antitumor mAb bind to tumors and activate complement, coating tumors with iC3b. Intravenously administered yeast beta-1,3;1,6-glucan functions as an adjuvant for antitumor mAb by priming the inactivated C3b (iC3b) receptors (CR3; CD11b/CD18) of circulating granulocytes, enabling CR3 to trigger cytotoxicity of iC3b-coated tumors. Recent data indicated that barley beta-1,3;1,4-glucan given orally similarly potentiated the activity of antitumor mAb, leading to enhanced tumor regression and survival. This investigation showed that orally administered yeast beta-1,3;1,6-glucan functioned similarly to barley beta-1,3;1,4-glucan with antitumor mAb. With both oral beta-1,3-glucans, a requirement for iC3b on tumors and CR3 on granulocytes was confirmed by demonstrating therapeutic failures in mice deficient in C3 or CR3. Barley and yeast beta-1,3-glucan were labeled with fluorescein to track their oral uptake and processing in vivo. Orally administered beta-1,3-glucans were taken up by macrophages that transported them to spleen, lymph nodes, and bone marrow. Within the bone marrow, the macrophages degraded the large beta-1,3-glucans into smaller soluble beta-1,3-glucan fragments that were taken up by the CR3 of marginated granulocytes. These granulocytes with CR3-bound beta-1,3-glucan-fluorescein were shown to kill iC3b-opsonized tumor cells following their recruitment to a site of complement activation resembling a tumor coated with mAb.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Monoclonais/efeitos dos fármacos , Glucanos/farmacologia , Neoplasias/tratamento farmacológico , beta-Glucanas , Animais , Anticorpos Monoclonais/imunologia , Complemento C3/efeitos dos fármacos , Tecido Linfoide/efeitos dos fármacos , Antígeno de Macrófago 1/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Neoplasias/imunologia
13.
Blood ; 103(6): 2071-8, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14604969

RESUMO

The mechanisms regulating the homing/mobilization of hematopoietic stem/progenitor cells (HSPCs) are not fully understood. In our previous studies we showed that the complement C3 activation peptide, C3a, sensitizes responses of HSPCs to stromal-derived factor 1 (SDF-1). In this study, mobilization was induced with granulocyte colony-stimulating factor (G-CSF) in both C3-deficient (C3-/-) and C3a receptor-deficient (C3aR-/-) mice as well as in wild-type (wt) mice in the presence or absence of a C3aR antagonist, SB 290157. The data indicated (1) significantly increased G-CSF-induced mobilization in C3-/- and C3aR-/- mice compared with wt mice, (2) significantly accelerated and enhanced G-CSF-induced mobilization in wt, but not in C3-/- or C3aR-/-, mice treated with SB 290157, and (3) deposition of C3b/iC3b fragments onto the viable bone marrow (BM) cells of G-CSF-treated animals. Furthermore, mobilization studies performed in chimeric mice revealed that wt mice reconstituted with C3aR-/- BM cells, but not C3aR-/- mice reconstituted with wt BM cells, are more sensitive to G-CSF-induced mobilization, suggesting that C3aR deficiency on graft-derived cells is responsible for this increased mobilization. Hence we suggest that C3 is activated in mobilized BM into C3a and C3b, and that the C3a-C3aR axis plays an important and novel role in retention of HSPCs (by counteracting mobilization) by increasing their responsiveness to SDF-1, the concentration of which is reduced in BM during mobilization. The C3a-C3aR axis may prevent an uncontrolled release of HSPCs into peripheral blood. These data further suggest that the C3aR antagonist SB 290157 could be developed as a drug to mobilize HSPCs for transplantation.


Assuntos
Arginina/análogos & derivados , Células da Medula Óssea/citologia , Complemento C3/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Membrana/genética , Receptores de Complemento/genética , Animais , Arginina/farmacologia , Compostos Benzidrílicos/farmacologia , Transplante de Medula Óssea , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Ativação do Complemento/efeitos dos fármacos , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Quimera por Radiação , Receptores de Complemento/antagonistas & inibidores
14.
Cancer Res ; 63(24): 9023-31, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14695221

RESUMO

The tumor-killing mechanisms available to monoclonal antibodies (mAbs; e.g., antagonism of growth factor receptors, antibody-dependent cell-mediated cytotoxicity) limit efficacy. Previous studies suggested that i.v. beta-glucan might function as an adjuvant for antitumor mAbs. beta- Glucan had been shown to function via the iC3b-receptor complement receptor 3 (CR3; CD11b/CD18) thereby enhancing leukocyte killing of tumor cells coated with iC3b via naturally occurring antitumor antibodies. Therapy with beta-glucans was limited by levels of natural antibodies and by tumor escape through elimination of antigen-positive cells. Accordingly, it was hypothesized that beta-glucan responses could be improved by combined administration with antitumor mAbs. Five tumor models were explored in BALB/c or C57Bl/6 mice using tumors that expressed either high levels of naturally occurring antigens (e.g., G(D2) ganglioside) or recombinant human MUC1. In comparison with antitumor mAb or beta-glucan alone, combined treatment with mAb plus beta-glucan produced significantly greater tumor regression in all models that included mammary, s.c., and hepatic tumors. Tumor-free survival only occurred in models that incorporated stable expression of the target antigen. beta-Glucan enhancement of the mAb tumoricidal response did not occur in mice deficient in either leukocyte CR3 (CD11b(-/-)) or serum C3, confirming the requirement for CR3 on leukocytes and iC3b on tumors. Granulocytes appeared to be primarily responsible for tumoricidal activity, because beta-glucan therapeutic responses did not occur in granulocyte-depleted mice. These data suggest that the therapeutic efficacy of mAbs known to activate complement (e.g., Herceptin, Rituxan, and Erbitux) could be significantly enhanced if they were combined with beta-glucan.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Monoclonais/farmacologia , Glucanos/farmacologia , Granulócitos/imunologia , Imunoterapia/métodos , beta-Glucanas , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Animais , Anticorpos Monoclonais/imunologia , Complemento C3/deficiência , Complemento C3/imunologia , Complemento C3/metabolismo , Complemento C3b/imunologia , Glucanos/imunologia , Antígeno de Macrófago 1/imunologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA