Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cancer Res Clin Oncol ; 148(5): 1045-1055, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35072775

RESUMEN

PURPOSE: Multiple myeloma (MM) remains an incurable hematologic malignancy which ultimately develops drug resistance and evades treatment. Despite substantial therapeutic advances over the past years, the clinical failure rate of preclinically promising anti-MM drugs remains substantial. More realistic in vitro models are thus required to better predict clinical efficacy of a preclinically active compound. METHODS: Here, we report on the establishment of a conical agarose 3D co-culture platform for the preclinical propagation of primary MM cells ex vivo. Cell growth was compared to yet established 2D and liquid overlay systems. MM cell lines (MMCL: RPMI-8226, U266, OPM-2) and primary patient specimens were tested. Drug sensitivity was examined by exploring the cytotoxic effect of bortezomib and the deubiquitinase inhibitor auranofin under various conditions. RESULTS: In contrast to 2D and liquid overlay, cell proliferation in the 3D array followed a sigmoidal curve characterized by an initial growth delay but more durable proliferation of MMCL over 12 days of culture. Primary MM specimens did not expand in ex vivo monoculture, but required co-culture support by a human stromal cell line (HS-5, MSP-1). HS-5 induced a > fivefold increase in cluster volume and maintained long-term viability of primary MM cells for up to 21 days. Bortezomib and auranofin induced less cytotoxicity under 3D vs. 2D condition and in co- vs. monoculture, respectively. CONCLUSIONS: This study introduces a novel model that is capable of long-term propagation and drug testing of primary MM specimens ex vivo overcoming some of the pitfalls of currently available in vitro models.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Auranofina/farmacología , Bortezomib/farmacología , Bortezomib/uso terapéutico , Línea Celular Tumoral , Técnicas de Cocultivo , Humanos , Mieloma Múltiple/patología
2.
Front Oncol ; 11: 708231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34485145

RESUMEN

OBJECTIVE: In clinical trials (CTs), the assessment of minimal residual disease (MRD) has proven to have prognostic value for multiple myeloma (MM) patients. Multiparameter flow cytometry (MFC) and next-generation sequencing are currently used in CTs as effective tools for outcome prediction. We have previously described 6- and 8-color MFC panels with and without kappa/lambda, which were equally reliable in detecting aberrant plasma cells (aPC) in myeloma bone marrow (BM) specimens. This follow-up study a) established a highly sensitive single-tube 10-color MFC panel for MRD detection in myeloma samples carrying different disease burden (monoclonal gammopathy of unknown significance (MGUS), smoldering multiple myeloma (SMM), MM), b) evaluated additional, rarely used markers included in this panel, and c) assessed MRD levels and the predictive value in apheresis vs. BM samples of MM patients undergoing autologous stem cell transplantation (ASCT). METHODS + RESULTS: The 10-color MFC was performed in BM and apheresis samples of 128 MM and pre-MM (MGUS/SMM) patients. The markers CD28, CD200, CD19, and CD117 underwent closer examination. The analysis revealed distinct differences in these antigens between MM, MGUS/SMM, and patients under treatment. In apheresis samples, the 10-color panel determined MRD negativity in 44% of patients. Absence of aPC in apheresis corresponded with disease burden, cytogenetics, and response to induction. It also determined MRD negativity in BM samples after ASCT and was associated with improved progression-free survival. CONCLUSION: These results highlight the significance of the evaluation of both BM and apheresis samples with a novel highly sensitive 10-color MFC panel.

4.
Cell Mol Life Sci ; 76(2): 369-380, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30357422

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) represent the lifelong source of all blood cells and continuously regenerate the hematopoietic system through differentiation and self-renewal. The process of differentiation is initiated in the G1 phase of the cell cycle, when stem cells leave their quiescent state. During G1, the anaphase-promoting complex or cyclosome associated with the coactivator Cdh1 is highly active and marks proteins for proteasomal degradation to regulate cell proliferation. Following Cdh1 knockdown in HSPCs, we analyzed human and mouse hematopoiesis in vitro and in vivo in competitive transplantation assays. We found that Cdh1 is highly expressed in human CD34+ HSPCs and downregulated in differentiated subsets; whereas, loss of Cdh1 restricts myeloid differentiation, supports B cell development and preserves immature short-term HSPCs without affecting proliferation or viability. Our data highlight a role of Cdh1 as a regulator of balancing the maintenance of HSPCs and differentiation into mature blood cells.


Asunto(s)
Proteínas Cdh1/metabolismo , Diferenciación Celular/genética , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteínas Cdh1/antagonistas & inhibidores , Proteínas Cdh1/genética , Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/deficiencia , Proteínas Proto-Oncogénicas c-kit/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Células Madre/citología , Células Madre/metabolismo
6.
Br J Haematol ; 179(1): 36-49, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28670693

RESUMEN

Cell adhesion-mediated drug resistance (CAM-DR) by the bone marrow (BM) is fundamental to multiple myeloma (MM) propagation and survival. Targeting BM protection to increase the efficacy of current anti-myeloma treatment has not been extensively pursued. To extend the understanding of CAM-DR, we hypothesized that the cytotoxic effects of novel anti-myeloma agents may be abrogated by the presence of BM stroma cells (BMSCs) and restored by addition of the CXCL12 antagonist NOX-A12 or the CXCR4 inhibitor plerixafor. Following this hypothesis, we evaluated different anti-myeloma agents alone, with BMSCs and when combined with plerixafor or NOX-A12. We verified CXCR4, CD49d (also termed ITGA4) and CD44 as essential mediators of BM adhesion on MM cells. Additionally, we show that CXCR7, the second receptor of stromal-derived-factor-1 (CXCL12), is highly expressed in active MM. Co-culture proved that co-treatment with plerixafor or NOX-A12, the latter inhibiting CXCR4 and CXCR7, functionally interfered with MM chemotaxis to the BM. This led to the resensitization of MM cells to the anti-myeloma agents vorinostat and pomalidomide and both proteasome inhibitors bortezomib and carfilzomib. Within a multicentre phase I/II study, NOX-A12 was tested in combination with bortezomib-dexamethasone, underlining the feasibility of NOX-A12 as an active add-on agent to antagonize myeloma CAM-DR.


Asunto(s)
Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Resistencia a Antineoplásicos , Mieloma Múltiple/metabolismo , Receptores CXCR/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Aptámeros de Nucleótidos/farmacología , Bencilaminas , Biomarcadores , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12/genética , Aberraciones Cromosómicas , Técnicas de Cocultivo , Ciclamas , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Compuestos Heterocíclicos/uso terapéutico , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Estadificación de Neoplasias , Oligopéptidos/farmacología , Fosforilación , Multimerización de Proteína , Transporte de Proteínas , Receptores CXCR/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
7.
Oncotarget ; 8(13): 21153-21166, 2017 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-28416751

RESUMEN

Elderly and frail patients, diagnosed with acute myeloid leukemia (AML) and ineligible to undergo intensive treatment, have a dismal prognosis. The small molecule inhibitor volasertib induces a mitotic block via inhibition of polo-like kinase 1 and has shown remarkable anti-leukemic activity when combined with low-dose cytarabine. We have demonstrated that AML cells are highly vulnerable to cell death in mitosis yet manage to escape a mitotic block through mitotic slippage by sustained proteasome-dependent slow degradation of cyclin B. Therefore, we tested whether interfering with mitotic slippage through proteasome inhibition arrests and kills AML cells more efficiently during mitosis. We show that therapeutic doses of bortezomib block the slow degradation of cyclin B during a volasertib-induced mitotic arrest in AML cell lines and patient-derived primary AML cells. In a xenotransplant mouse model of human AML, mice receiving volasertib in combination with bortezomib showed superior disease control compared to mice receiving volasertib alone, highlighting the potential therapeutic impact of this drug combination.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bortezomib/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Mitosis/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Pteridinas/farmacología , Anciano , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bortezomib/uso terapéutico , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Ciclina B/genética , Ciclina B/metabolismo , Citarabina/farmacología , Citarabina/uso terapéutico , Anciano Frágil , Humanos , Ratones , Ratones Endogámicos NOD , Inhibidores de Proteasoma/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pteridinas/uso terapéutico , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
9.
Cancer Res ; 75(11): 2166-76, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840983

RESUMEN

Cancer stem cells (CSC) drive tumorigenesis and contribute to genotoxic therapy resistance, diffuse infiltrative invasion, and immunosuppression, which are key factors for the incurability of glioblastoma multiforme (GBM). The AC133 epitope of CD133 is an important CSC marker for GBM and other tumor entities. Here, we report the development and preclinical evaluation of a recombinant AC133×CD3 bispecific antibody (bsAb) that redirects human polyclonal T cells to AC133(+) GBM stem cells (GBM-SC), inducing their strong targeted lysis. This novel bsAb prevented the outgrowth of AC133-positive subcutaneous GBM xenografts. Moreover, upon intracerebral infusion along with the local application of human CD8(+) T cells, it exhibited potent activity in prophylactic and treatment models of orthotopic GBM-SC-derived invasive brain tumors. In contrast, normal hematopoietic stem cells, some of which are AC133-positive, were virtually unaffected at bsAb concentrations effective against GBM-SCs and retained their colony-forming abilities. In conclusion, our data demonstrate the high activity of this new bsAb against patient-derived AC133-positive GBM-SCs in models of local therapy of highly invasive GBM.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD/inmunología , Glioblastoma/terapia , Glicoproteínas/inmunología , Células Madre Neoplásicas/inmunología , Péptidos/inmunología , Antígeno AC133 , Anticuerpos Biespecíficos/inmunología , Antígenos CD/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Carcinogénesis/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epítopos/inmunología , Glioblastoma/inmunología , Glioblastoma/patología , Glicoproteínas/uso terapéutico , Humanos , Inmunoterapia/métodos , Células Madre Neoplásicas/patología , Péptidos/uso terapéutico
10.
PLoS One ; 8(11): e79939, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223204

RESUMEN

BACKGROUND: We systematically analyzed multiple myeloma (MM) cell lines and patient bone marrow cells for their engraftment capacity in immunodeficient mice and validated the response of the resulting xenografts to antimyeloma agents. DESIGN AND METHODS: Using flow cytometry and near infrared fluorescence in-vivo-imaging, growth kinetics of MM cell lines L363 and RPMI8226 and patient bone marrow cells were investigated with use of a murine subcutaneous bone implant, intratibial and intravenous approach in NOD/SCID, NOD/SCID treated with CD122 antibody and NOD/SCID IL-2Rγ(null) mice (NSG). RESULTS: Myeloma growth was significantly increased in the absence of natural killer cell activity (NSG or αCD122-treated NOD/SCID). Comparison of NSG and αCD122-treated NOD/SCID revealed enhanced growth kinetics in the former, especially with respect to metastatic tumor sites which were exclusively observed therein. In NSG, MM cells were more tumorigenic when injected intratibially than intravenously. In NOD/SCID in contrast, the use of juvenile long bone implants was superior to intratibial or intravenous cancer cell injection. Using the intratibial NSG model, mice developed typical disease symptoms exclusively when implanted with human MM cell lines or patient-derived bone marrow cells, but not with healthy bone marrow cells nor in mock-injected animals. Bortezomib and dexamethasone delayed myeloma progression in L363- as well as patient-derived MM cell bearing NSG. Antitumor activity could be quantified via flow cytometry and in vivo imaging analyses. CONCLUSIONS: Our results suggest that the intratibial NSG MM model mimics the clinical situation of the disseminated disease and serves as a valuable tool in the development of novel anticancer strategies.


Asunto(s)
Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Receptores de Interleucina-2/deficiencia , Animales , Ácidos Borónicos/uso terapéutico , Bortezomib , Línea Celular Tumoral , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/uso terapéutico , Receptores de Interleucina-2/genética
11.
Br J Haematol ; 161(1): 104-16, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23384035

RESUMEN

Despite considerable advances, multiple myeloma (MM) remains incurable and the development of novel therapies targeting the interplay between plasma cells (PCs) and their bone marrow (BM) microenvironment remains essential. We investigated the effect of various agents in vitro on the proliferation, phenotype, morphology, actin polymerization and migration of MM cells and, in vivo, the tumour growth of L363-bearing non-obese diabetic severe combined immunodeficient mice with a deficient interleukin-2 receptor gamma chain (NSG). In vitro, we observed a dose-dependent cytotoxicity with bortezomib and sorafenib. Using RPMI8226 cells co-expressing histone 2B-mCherry and cytochrome c-GFP, bortezomib- and sorafenib-induced apoptosis was confirmed, and both agents combined showed synergism. Sorafenib induced CD138-downregulation and abolished CXCL12-induced actin polymerization. L363 cells expressed CCR4 and CCR5 and migrated to their common ligand CCL5. Chemotaxis to BM stroma cells was notable and significantly reduced by sorafenib. Downregulation of phospho-ERK appeared relevant for the inhibition of actin polymerization and chemotaxis. Sorafenib alone, and combined with bortezomib, showed substantial antitumour activity in L363-bearing NSG. Correspondingly, sorafenib induced clinical responses in MM-/AL-amyloidosis patients. We conclude that, in addition to the cytotoxic and anti-angiogenic effects of sorafenib, blocking of MM cell migration and homing represent promising mechanisms to interrupt the interplay between PCs and their supportive microenvironment.


Asunto(s)
Actinas/metabolismo , Antineoplásicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Sindecano-1/biosíntesis , Anciano , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Ácidos Borónicos/uso terapéutico , Bortezomib , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/biosíntesis , Niacinamida/administración & dosificación , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Fosforilación/efectos de los fármacos , Polimerizacion/efectos de los fármacos , Pirazinas/uso terapéutico , Sorafenib , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Leuk Res ; 35(9): 1265-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21684596

RESUMEN

We analyzed (1) early endothelial progenitors (EPCs; CD34(+)/AC133(+)/VEGFR2(+)), mature EPCs (CD34(+)/VEGFR2(+)) and VEGFR2(+)-cells in bone marrow (BM)-specimens of multiple myeloma (MM)- vs. monoclonal gammopathy (MGUS)-patients and healthy controls; (2) differences of BM-, peripheral blood (PB)- and leukapheresis (LP)-samples; and (3) the association of EPCs and VEGFR2(+)-cells with MM-parameters. MM patients demonstrated highest early and mature EPCs and VEGFR2(+)-cells in the BM, particularly with advanced and active disease. Endothelial cells differed in BM-, PB- and LP-specimens, albeit seemed less associated with unfavorable prognostic MM-parameters. Our data suggest that especially VEGFR2(+)-cells and mature EPCs in MM are of value to explore further.


Asunto(s)
Compartimento Celular , Células Endoteliales/fisiología , Mieloma Múltiple/patología , Células Madre/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Compartimento Celular/fisiología , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Células Madre/metabolismo , Células Madre/patología
13.
Leuk Lymphoma ; 51(9): 1632-42, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20578819

RESUMEN

Cyclins D1, D2, and D3 (CCND1, 2, 3) are regulated by proteasomal degradation. Their overexpression in multiple myeloma (MM) has prognostic value. We performed this pilot study to analyze a possible association between CCND1-3 overexpression and response to treatment with the proteasome inhibitor bortezomib, since a specific prognostic marker for bortezomib response has not been reported, but would be ideal to predict who benefits most from bortezomib in times of several potentially efficient therapeutic options. Bone marrow (BM) specimens of 20/47 consecutive patients were available for reliable CCND1-3 analyses by real-time PCR. With CCND1 overexpression in 9/20 patients, the risk for progression after bortezomib treatment was significantly decreased (HR 0.102, 95% CI 0.021-0.498, p = 0.0048) and progression-free survival substantially prolonged (p = 0.0011). Our study is the first to suggest that overexpressed CCND1 in MM is an independent prognostic marker associated with a more durable response to bortezomib. These preliminary results warrant a larger study.


Asunto(s)
Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Ciclina D1/genética , Ciclina D2/genética , Ciclina D3/genética , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Médula Ósea , Bortezomib , Ciclina D1/metabolismo , Ciclina D2/metabolismo , Ciclina D3/metabolismo , ADN/análisis , ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Proyectos Piloto , Reacción en Cadena de la Polimerasa , Terapia Recuperativa , Tasa de Supervivencia , Resultado del Tratamiento
14.
Exp Hematol ; 35(10): 1550-61, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17889722

RESUMEN

OBJECTIVE: To develop an in vitro culture system for rapid assessment of multiple myeloma (MM) cell growth. METHODS: MM cells lines (MMCLs) L363, U266, and RPMI-8226, and bone marrow (BM)-derived plasma cells (PCs) from MM patients were evaluated for their in vitro growth using various stroma (BMSC, M210-B4, and osteoclasts [OCs]) and cytokine support combinations (combination A: interleukin [IL]-6, vascular endothelial growth factor, insulin-like growth factor-1 vs combination B: A plus hepatocyte growth factor, IL-13 vs combination C: IL-6, insulin-like growth factor -1, stromal-derived growth factor-1, Galectin-1, IL-1alpha). RESULTS: We found a significant effect of stroma, notably affecting growth of L363 cells. Cytokine combination A had the highest growth impact, whereas B and C were of lesser benefit. The contribution of combined cytokines and stroma for MMCL growth was moderate and the viability of MMCLs was best preserved with OCs. One of the most commonly used PC-marker CD138, expressed on all MMCLs on day 0, showed a gradual downmodulation upon all culture conditions, possibly induced as a stroma-triggered phenotypic change, and leading to the ability of MM cells to dedifferentiate into immature, resilient phenotypes. PC-enriched BM samples from 7 of 10 MM patients could be maintained in culture, again profiting from stroma more than cytokines alone. CONCLUSIONS: Our data demonstrate a consistent growth advantage provided by BMSCs on MMCLs and primary MM cells, preserved viability with OCs, and phenotypic and morphologic heterogeneity of primary MM cells during culture. Further identification of key components involved in MM cell growth, coupled with our understanding of drug sensitivity offers the potential to better define the disease pathogenesis and to identify novel therapeutic targets.


Asunto(s)
Técnicas de Cultivo de Célula , Mieloma Múltiple/patología , Células Plasmáticas/patología , Anciano , Diferenciación Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Fenotipo , Células Plasmáticas/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Sindecano-1/biosíntesis , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA