Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Ann Hematol ; 97(5): 839-849, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29359239

RESUMEN

A promising approach to the treatment of multiple myeloma (MM) involves agents that target not only the myeloma cells directly, but also the tumor microenvironment which promotes tumor cell growth, angiogenesis, and MM bone disease. Here we investigate the orally available multikinase inhibitor, regorafenib (BAY 73-4506), for its therapeutic efficacy in MM. Regorafenib is a potent inhibitor of angiogenic (VEGFR 1-3, PDGFR-b) as well as oncogenic (c-KIT, RET, FGFR, Raf) kinases. We show that regorafenib induces apoptosis in all MM cell lines at below clinically achievable concentrations. Regorafenib overcomes the growth advantage conferred by a stroma cell MM and an endothelial cell MM, co-culture systems, and abrogates growth factor-stimulated MEK, ERK, and AKT phosphorylation at nanomolar to micromolar concentrations. Moreover, it inhibits endothelial cell growth and tubule formation, abrogates both VEGF secretion and VEGF-induced MM cell migration, inhibits osteoclastogenesis, and shows synergistic cytotoxicity with dexamethasone, the immunomodulatory drug pomalidomide, and the p110δ inhibitor idelalisib. Most importantly, regorafenib significantly delays tumor growth in a xenograft mouse model of human MM. These results provide the rationale for further clinical evaluation of regorafenib, alone and in combination, in the treatment of MM.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Compuestos de Fenilurea/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas/administración & dosificación , Administración Oral , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Desnudos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Compuestos de Fenilurea/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Piridinas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
Hematol Oncol ; 34(4): 200-207, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25784529

RESUMEN

Therapeutic options in heavily pretreated relapsed/refractory multiple myeloma patients are often very limited because of impaired bone marrow function. Bendamustine is effective in multiple myeloma and has a favourable toxicity profile. We hypothesized that dose-intensified bendamustine (180 mg/m2 , day 1 and 2) followed by autologous blood stem cell support (ASCS) would improve bone marrow function with low post-transplant toxicity in patients with severely impaired haematopoiesis. We analyzed 28 consecutive myeloma patients, with a median of three prior lines of therapy (range 2-7), who had relapsed from the last treatment with very limited bone marrow function and were therefore ineligible for conventional chemotherapy, novel agents or trial enrolment. Dose-intensified bendamustine with ASCS improved haematopoiesis as reflected by increased platelet counts (median 40/nl vs 94/nl, p = 0.0004) and white blood cell counts (3.0/nl vs 4.8/nl, p = 0.02) at day +100. The median time until engraftment of platelets (>50/nl) was 11 days (0-24 days) and of white cell counts (>1.0/nl) 0 days (0-24 days). At least, a minimal response was achieved in 36% of patients. The disease stabilization rate was 50% while the median progression-free survival rate was limited to 2.14 months. Most importantly, patients were once again eligible for alternative treatments including enrolment into clinical trials. We conclude that dose-intensified bendamustine followed by ASCS is safe and feasible for multiple myeloma patients with very limited bone marrow reserve. Copyright © 2015 John Wiley & Sons, Ltd.


Asunto(s)
Clorhidrato de Bendamustina/administración & dosificación , Médula Ósea/fisiopatología , Hematopoyesis , Mieloma Múltiple , Trasplante de Células Madre de Sangre Periférica , Acondicionamiento Pretrasplante , Adulto , Anciano , Autoinjertos , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Mieloma Múltiple/fisiopatología , Mieloma Múltiple/terapia , Tasa de Supervivencia
3.
Blood Cancer J ; 14(1): 148, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191769

RESUMEN

Risk of progression of monoclonal gammopathy of undetermined significance (MGUS) into multiple myeloma and related plasma cell disorders can be determined by three major risk stratification models, namely Mayo2005, Sweden2014, and NCI2019. This retrospective study of 427 patients with MGUS diagnosed according to the 2014 International Myeloma Working Group criteria aimed to describe and analyze the longitudinal applicability of these risk models. In all three models, the majority of patients remained at their baseline risk group, whereas small numbers of patients migrated to a different risk group. Proportions of patients among risk groups remained stable over time (e.g. Mayo2005 model, low-risk group, at baseline: 43%, after 1, 2, 3, 4, 5, and 8 years: 40%, 37%, 37%, 43%, 44%, and 43%). All three risk models reliably distinguished risk of progression at baseline, upon yearly reassessment (e.g. 1 year from diagnosis) and in time-dependent analyses. Upstaging to a high-risk category was associated with an increased risk of progression in all three models (Mayo2005: hazard ratio [HR] = 5.43, 95% confidence interval [95% CI] 1.21-24.39, p = 0.027; Sweden2014: HR = 13.02, 95% CI 5.25-32.28, p < 0.001; NCI2019: HR = 5.85, 95% CI 2.49-13.74, p < 0.001). Our study shows that MGUS risk stratification models can be applied longitudinally to repeatedly determine and improve individual risk of progression. Patient migration to higher risk categories during follow up should prompt more frequent monitoring in clinical routine.


Asunto(s)
Progresión de la Enfermedad , Gammopatía Monoclonal de Relevancia Indeterminada , Humanos , Gammopatía Monoclonal de Relevancia Indeterminada/epidemiología , Gammopatía Monoclonal de Relevancia Indeterminada/diagnóstico , Masculino , Femenino , Anciano , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Anciano de 80 o más Años , Estudios Longitudinales , Adulto , Mieloma Múltiple/epidemiología , Mieloma Múltiple/diagnóstico , Factores de Riesgo
4.
EJHaem ; 3(4): 1377-1380, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36467828

RESUMEN

Due to differences in the protein folding mechanisms, it is exceedingly rare for amyloid light chain (AL) amyloidosis and monoclonal gammopathy of renal significance (MGRS) to coexist. We herein report the first case of concurrent AL amyloidosis and a subclass of MGRS, light chain proximal tubulopathy (LCPT). The 53-year-old female was diagnosed with smoldering myeloma immunoglobulin G kappa and AL amyloidosis with deposits in fat and gastrointestinal tissue. The kidney biopsy did not show amyloid deposits but electron microscopy revealed the presence of LCPT with crystal formation in proximal tubular epithelial cells. This case illustrates the complex pathophysiology of protein deposition in monoclonal gammopathies.

5.
Blood ; 113(7): 1513-21, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19018094

RESUMEN

Targeting protein kinase C (PKC) isoforms by the small molecule inhibitor enzastaurin has shown promising preclinical activity in a wide range of tumor cells. We further delineated its mechanism of action in multiple myeloma (MM) cells and found a novel role of beta-catenin in regulating growth and survival of tumor cells. Specifically, inhibition of PKC leads to rapid accumulation of beta-catenin by preventing the phosphorylation required for its proteasomal degradation. Microarray analysis and small-interfering RNA (siRNA)-mediated gene silencing in MM cells revealed that accumulated beta-catenin activates early endoplasmic reticulum stress signaling via eIF2alpha, C/EBP-homologous protein (CHOP), and p21, leading to immediate growth inhibition. Furthermore, accumulated beta-catenin contributes to enzastaurin-induced cell death. Sequential knockdown of beta-catenin, c-Jun, and p73, as well as overexpression of beta-catenin or p73 confirmed that accumulated beta-catenin triggers c-Jun-dependent induction of p73, thereby conferring MM cell apoptosis. Our data reveal a novel role of beta-catenin in endoplasmic reticulum (ER) stress-mediated growth inhibition and a new proapoptotic mechanism triggered by beta-catenin on inhibition of PKC isoforms. Moreover, we identify p73 as a potential novel therapeutic target in MM. Based on these and previous data, enzastaurin is currently under clinical investigation in a variety of hematologic malignancies, including MM.


Asunto(s)
Apoptosis/fisiología , Retículo Endoplásmico/metabolismo , Mieloma Múltiple/metabolismo , Proteína Quinasa C/metabolismo , beta Catenina/metabolismo , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/metabolismo , Indoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Proteínas Nucleares/genética , Proteína Quinasa C/antagonistas & inhibidores , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/fisiología , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética
6.
Cancers (Basel) ; 13(18)2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34572934

RESUMEN

Salvage high-dose chemotherapy and autologous stem cell transplantation (HDCT/ASCT) is a treatment option for relapsed and/or refractory multiple myeloma (RRMM). No data are available on salvage HDCT/ASCT following re-induction treatment with state-of-the-art triplet regimens. We retrospectively report on 44 patients receiving salvage HDCT/ASCT following re-induction with carfilzomib/lenalidomide/dexamethasone (KRd). All patients received frontline HDCT/ASCT with median time to progression (TTP1) of 2.9 (1.2-13.5) years, enabling paired comparison of frontline and salvage HDCT/ASCT. After re-induction and before salvage transplant, 25/44 patients (57%) attained ≥ very good partial response (VGPR), which increased to 34/44 (77%) at best response after salvage HDCT/ASCT. Median progression-free survival (PFS) was 23.3 months from salvage HDCT/ASCT. Patients with ≥ VGPR at the time of salvage HDCT/ASCT and those receiving maintenance treatment post salvage HDCT/ASCT had significantly superior PFS (hazard ratio (HR) 0.19, p = 0.001 and HR 0.20, p = 0.009). In patients achieving at least an equal depth of response before salvage HDCT/ASCT as before frontline HDCT/ASCT, PFS after salvage HDCT/ASCT was comparable to the frontline situation (p = 0.3). This is the first report of state-of-the-art triplet re-induction and salvage HDCT/ASCT for RRMM after frontline transplantation. Deep remissions achieved with KRd translate into prolonged PFS following salvage HDCT/ASCT and are enhanced by maintenance treatment.

7.
Lancet ; 374(9686): 324-39, 2009 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-19541364

RESUMEN

Multiple myeloma is characterised by clonal proliferation of malignant plasma cells, and mounting evidence indicates that the bone marrow microenvironment of tumour cells has a pivotal role in myeloma pathogenesis. This knowledge has already expanded treatment options for patients with multiple myeloma. Prototypic drugs thalidomide, bortezomib, and lenalidomide have each been approved for the treatment of this disease by targeting both multiple myeloma cells and the bone marrow microenvironment. Although benefit was first shown in relapsed and refractory disease, improved overall response, duration of response, and progression-free and overall survival can be achieved when these drugs are part of first-line regimens. This treatment framework promises to improve outcome not only for patients with multiple myeloma, but also with other haematological malignancies and solid tumours.


Asunto(s)
Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/uso terapéutico , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Bortezomib , Ensayos Clínicos como Asunto , Terapia Combinada , Predisposición Genética a la Enfermedad/genética , Humanos , Lenalidomida , Melfalán/uso terapéutico , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/epidemiología , Mieloma Múltiple/etiología , Estadificación de Neoplasias , Inhibidores de Proteasas/uso terapéutico , Inducción de Remisión , Trasplante de Células Madre , Tasa de Supervivencia , Resultado del Tratamiento
8.
Br J Haematol ; 141(5): 659-71, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18410446

RESUMEN

This study investigated the biological significance of the inhibition of fatty acid synthase (FAS) in multiple myeloma (MM) using the small molecule inhibitor Cerulenin. Cerulenin triggered growth inhibition in both MM cell lines and MM patient cells, and overcame the survival and growth advantages conferred by interleukin-6, insulin-like growth factor-1, and bone marrow stromal cells. It induced apoptosis in MM cell lines with only modest activation of caspase -8, -9, -3 and PARP; moreover, the pan-caspase inhibitor Z-VAD-FMK did not inhibit Cerulenin-induced apoptosis and cell death. In addition, treatment of MM cells with Cerulenin primarily up-regulated apoptosis-inducing factor/endonuclease G, mediators of caspase-independent apoptosis. Importantly, Cerulenin induced endoplasmic reticulum stress response via up-regulation of the Grp78/IRE1alpha/JNK pathway. Although the C-Jun-NH(2)-terminal kinase (JNK) inhibitor SP600215 blocked Cerulenin-induced cytotoxicity, it did not inhibit apoptosis and caspase cleavage. Furthermore, Cerulenin showed synergistic cytotoxic effects with various agents including Bortezomib, Melphalan and Doxorubicin. Our results therefore indicate that inhibition of FAS by Cerulenin primarily triggered caspase-independent apoptosis and JNK-dependent cytotoxicity in MM cells. This report demonstrated that inhibition of FAS has anti-tumour activity against MM cells, suggesting that it represents a novel therapeutic target in MM.


Asunto(s)
Cerulenina/uso terapéutico , Ácido Graso Sintasas/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 8/biosíntesis , Caspasa 9/biosíntesis , Línea Celular Tumoral , Cerulenina/farmacología , Sistemas de Liberación de Medicamentos , Evaluación de Medicamentos , Chaperón BiP del Retículo Endoplásmico , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , MAP Quinasa Quinasa 4/metabolismo , Mieloma Múltiple/enzimología , Transducción de Señal , Células Tumorales Cultivadas , Receptor fas/metabolismo
9.
Haematologica ; 93(1): 124-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18166796

RESUMEN

In the prospective phase 3 HOVON-50/GMMG-HD3 trial, patients randomized to TAD (thalidomide, doxorubicin, dexamethasone) had a significantly higher response rate (at least PR) after induction compared with patients randomized to VAD (vincristine, adriamycin, dexamethasone, 72% vs. 54%, p<0.001). Complete remission (CR) and very good partial remission (VGPR) were also higher after TAD. After High Dose melphalan 200mg/m(2) response was comparable in both arms, 76% and 79% respectively. However, CR plus VGPR were significantly higher in the patients randomized to TAD (49% vs. 32%, p<0.001). CTC grade 3-4 adverse events were similar in both arms.


Asunto(s)
Oncología Médica/métodos , Mieloma Múltiple/tratamiento farmacológico , Talidomida/uso terapéutico , Adulto , Anciano , Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citarabina/uso terapéutico , Daunorrubicina/uso terapéutico , Dexametasona/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inducción de Remisión , Tioguanina/uso terapéutico , Resultado del Tratamiento , Vincristina/uso terapéutico
10.
Expert Opin Drug Metab Toxicol ; 4(7): 973-85, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18624684

RESUMEN

BACKGROUND: After the tragic events in the early 1960s, thalidomide has re-emerged as therapeutic for multiple myeloma (MM). It was first approved for the treatment of erythema nodosum leprosum, and is now under evaluation for hematologic and non-hematologic disorders. Its complex mechanism of action is not fully understood; however extensive preclinical studies in MM have revealed its antiangiogenic and immunomodulatory properties. OBJECTIVE: In this review, we focus on the importance and toxicity of thalidomide in today's clinical use. METHODS: Key preclinical and clinical trials available as well as data on the pharmacokinetics and pharmacodynamics of thalidomide in humans are summarized. CONCLUSIONS: Thalidomide is widely used as first-line treatment and in relapsed/refractory MM. The most common side effects are fatigue, constipation and peripheral neuropathy, and careful monitoring is required to avoid fetal exposure.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Inmunosupresores/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Talidomida/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Interacciones Farmacológicas , Femenino , Humanos , Inmunosupresores/efectos adversos , Inmunosupresores/farmacocinética , Inmunosupresores/farmacología , Masculino , Embarazo , Talidomida/efectos adversos , Talidomida/farmacocinética , Talidomida/farmacología
11.
Cancer Res ; 66(13): 6675-82, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818641

RESUMEN

Recent studies have underscored the role of B-cell-activating factor (BAFF), a member of the tumor necrosis factor superfamily, in promoting the survival of malignant B cells, including human multiple myeloma. We here characterized the functional significance of BAFF in the interaction between multiple myeloma and bone marrow stromal cells (BMSC) and further defined the molecular mechanisms regulating these processes. BAFF is detected on BMSCs derived from multiple myeloma patients as evidenced by flow cytometry. BAFF secretion is 3- to 10-fold higher in BMSCs than in multiple myeloma cells, and tumor cell adhesion to BMSCs augments BAFF secretion by 2- to 5-fold, confirmed by both ELISA and immunoblotting. Adhesion of MM1S and MCCAR multiple myeloma cell lines to KM104 BMSC line transfected with a luciferase reporter vector carrying the BAFF gene promoter (BAFF-LUC) significantly enhanced luciferase activity, suggesting that nuclear factor-kappaB (NF-kappaB) activation induced by multiple myeloma adhesion to BMSCs mediates BAFF up-regulation. Moreover, BAFF (0-100 ng/mL) increases adhesion of multiple myeloma lines to BMSCs in a dose-dependent manner; conversely, transmembrane activator and calcium modulator and cyclophylin ligand interactor-Ig or B-cell maturation antigen/Fc blocked BAFF stimulation. Using adenoviruses expressing dominant-negative and constitutively expressed AKT as well as NF-kappaB inhibitors, we further showed that BAFF-induced multiple myeloma cell adhesion is primarily mediated via activation of AKT and NF-kappaB signaling. Importantly, BAFF similarly increased adhesion of CD138-expressing patient multiple myeloma cells to BMSCs. These studies establish a role for BAFF in localization and survival of multiple myeloma cells in the bone marrow microenvironment and strongly support novel therapeutics, targeting the interaction between BAFF and its receptors in human multiple myeloma.


Asunto(s)
Neoplasias de la Médula Ósea/patología , Proteínas de la Membrana/fisiología , Mieloma Múltiple/patología , Factor de Necrosis Tumoral alfa/fisiología , Factor Activador de Células B , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Neoplasias de la Médula Ósea/metabolismo , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/farmacología , Mieloma Múltiple/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Interleucina-4/biosíntesis , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/patología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
12.
Cancer Res ; 65(24): 11712-20, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16357183

RESUMEN

SGN-40, a humanized immoglobulin G1 (IgG1) anti-CD40 monoclonal antibody, mediates cytotoxicity against human multiple myeloma (MM) cells via suppression of interleukin (IL)-6-induced proliferative and antiapoptotic effects as well as antibody-dependent cell-mediated cytotoxicity (ADCC). Here, we studied the clinical significance of an immunomodulatory drug lenalidomide on SGN-40-induced cytotoxicity against CD138(+)CD40(+) MM lines and patient MM cells. Pretreatment with lenalidomide sensitized MM cells to SGN-40-induced cell death. Combined lenalidomide and SGN-40 significantly induced MM apoptosis, evidenced by enhanced cleavage of caspase-3/8/poly(ADP-ribose)polymerase and increased sub-G(0) cells, compared with either single agent at the same doses. Pretreatment of effector cells with lenalidomide augmented SGN-40-induced MM cell lysis, associated with an increased number of CD56(+)CD3(-) natural killer (NK) cells expressing CD16 and LFA-1. Importantly, pretreatment with lenalidomide or lenalidomide and SGN-40 markedly enhanced NK-cell-mediated lysis of autologous patient MM cells triggered by SGN-40. Lenalidomide also up-regulated CD40L on CD56(+)CD3(-) NK cells, facilitating IL-2-mediated activation of NK cells. In addition, lenalidomide induced the CD56(dim) NK subset, which are more potent mediators of ADCC against target MM cells than the CD56(bright) NK subset. Finally, pretreatment of both effector and target MM cells with lenalidomide markedly enhanced SGN-40-mediated ADCC against CD40-expressing MM cells. These studies, therefore, show that the addition of lenalidomide to SGN-40 enhances cytotoxicity against MM cells, providing the framework for combined lenalidomide and SGN-40 in a new treatment paradigm to both target MM cells directly and induce immune effectors against MM.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Apoptosis/efectos de los fármacos , Antígenos CD40/inmunología , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Talidomida/análogos & derivados , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD40/metabolismo , Ligando de CD40/inmunología , Antígeno CD56/inmunología , Antígeno CD56/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Humanos , Inmunización Pasiva , Interleucina-6/farmacología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Lenalidomida , Mieloma Múltiple/metabolismo , Talidomida/uso terapéutico
13.
Haematologica ; 91(11): 1555-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17082013

RESUMEN

Patients with multiple myeloma and end-stage renal failure on dialysis are frequently not considered eligible for high-dose therapy (HDT) due to higher transplant-related mortality (TRM). Our aim was to evaluate the toxicity and survival of dialysis-dependent patients after HDT with melphalan (100 mg/m(2)) compared to those of patients without renal insufficiency (melphalan 200 mg/m(2)) in a matched pairs analysis of 34 patients. No significant differences were observed between hematologic toxicity, TRM or disease response. Dialysis patients showed comparable event-free and overall survival. They required significantly extended intravenous antibiotic treatment and longer hospitalization. Thus, melphalan 100 mg/m2 is less toxic, yet equally efficient and improves the prognosis of this group of patients.


Asunto(s)
Fallo Renal Crónico/terapia , Mieloma Múltiple/terapia , Diálisis Renal , Trasplante de Células Madre , Adulto , Anciano , Femenino , Humanos , Fallo Renal Crónico/mortalidad , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Diálisis Renal/mortalidad , Trasplante de Células Madre/mortalidad , Trasplante Autólogo/mortalidad , Resultado del Tratamiento
14.
Oncotarget ; 5(21): 10237-50, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25296978

RESUMEN

The inhibitor-of-apoptosis family member survivin has been reported to inhibit apoptosis and regulate mitosis and cytokinesis. In multiple myeloma, survivin has been described to be involved in downstream sequelae of various therapeutic agents. We assessed 1093 samples from previously untreated patients, including two independent cohorts of 392 and 701 patients, respectively. Survivin expression was associated with cell proliferation, adverse prognostic markers, and inferior event-free and overall survival, supporting the evaluation of survivin as a therapeutic target in myeloma. The small molecule suppressant of survivin--YM155--is in clinical development for the treatment of solid tumors. YM155 potently inhibited proliferation and induced apoptosis in primary myeloma cells and cell lines. Gene expression and protein profiling revealed the critical roles of IL6/STAT3-signaling and the unfolded protein response in the efficacy of YM155. Both pathways converged to down regulate anti-apoptotic Mcl-1 in myeloma cells. Conversely, growth inhibition and apoptotic cell death by YM155 was rescued by ectopic expression of Mcl-1 but not survivin, identifying Mcl-1 as the pivotal downstream target of YM155 in multiple myeloma. Mcl-1 expression was likewise associated with adverse prognostic markers, and inferior survival. Our results strongly support the clinical evaluation of YM155 in patients with multiple myeloma.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Naftoquinonas/farmacología , Respuesta de Proteína Desplegada , Apoptosis/efectos de los fármacos , Apoptosis/genética , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Estudios de Cohortes , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Interleucina-6/metabolismo , Mieloma Múltiple/mortalidad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Pronóstico , Factor de Transcripción STAT3/metabolismo , Análisis de Supervivencia , Survivin , Transcriptoma , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/genética
15.
Cancer Res ; 72(20): 5374-85, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22942257

RESUMEN

In contrast to normal cells, malignant cells are frequently aneuploid and contain multiple centrosomes. To allow for bipolar mitotic division, supernumerary centrosomes are clustered into two functional spindle poles in many cancer cells. Recently, we have shown that griseofulvin forces tumor cells with supernumerary centrosomes to undergo multipolar mitoses resulting in apoptotic cell death. Here, we describe the characterization of the novel small molecule GF-15, a derivative of griseofulvin, as a potent inhibitor of centrosomal clustering in malignant cells. At concentrations where GF-15 had no significant impact on tubulin polymerization, spindle tension was markedly reduced in mitotic cells upon exposure to GF-15. Moreover, isogenic cells with conditional centrosome amplification were more sensitive to GF-15 than parental controls. In a wide array of tumor cell lines, mean inhibitory concentrations (IC(50)) for proliferation and survival were in the range of 1 to 5 µmol/L and were associated with apoptotic cell death. Importantly, treatment of mouse xenograft models of human colon cancer and multiple myeloma resulted in tumor growth inhibition and significantly prolonged survival. These results show the in vitro and in vivo antitumor efficacy of a prototype small molecule inhibitor of centrosomal clustering and strongly support the further evaluation of this new class of molecules.


Asunto(s)
Centrosoma/efectos de los fármacos , Griseofulvina/análogos & derivados , Línea Celular Tumoral , Griseofulvina/farmacología , Humanos , Técnicas In Vitro , Distribución Tisular
16.
Br J Haematol ; 139(1): 55-63, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17854307

RESUMEN

Osteolytic bone disease in multiple myeloma (MM) is associated with upregulation of osteoclast (OCL) activity and constitutive inhibition of osteoblast function. The extracellular signal-regulated kinase 1/2 (ERK1/2) pathway mediates OCL differentiation and maturation. We hypothesized that inhibition of ERK1/2 could prevent OCL differentiation and downregulate OCL function. It was found that AZD6244, a mitogen-activated or extracellular signal-regulated protein kinase (MEK) inhibitor, blocked OCL differentiation and formation in a dose-dependent manner, evidenced by decreased alphaVbeta3-integrin expression and tartrate-resistant acid phosphatase positive (TRAP+) cells. Functional dentine disc cultures showed inhibition of OCL-induced bone resorption by AZD6244. Major MM growth and survival factors produced by OCLs including B-cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL), as well as macrophage inflammatory protein (MIP-1alpha), which mediates OCL differentiation and MM, were also significantly inhibited by AZD6244. In addition to ERK inhibition, NFATc1 (nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1) and c-fos were both downregulated, suggesting that AZD6244 targets a later stage of OCL differentiation. These results indicate that AZD6244 inhibits OCL differentiation, formation and bone resorption, thereby abrogating paracrine MM cell survival in the bone marrow microenvironment. The present study therefore provides a preclinical rationale for the evaluation of AZD6244 as a potential new therapy for patients with MM.


Asunto(s)
Bencimidazoles/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Osteoclastos/patología , Fosfatasa Ácida/metabolismo , Biomarcadores/análisis , Western Blotting/métodos , Resorción Ósea/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL3 , Quimiocina CCL4 , Citocinas/metabolismo , Depresión Química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Integrina alfaVbeta3/metabolismo , Isoenzimas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Proteínas Inflamatorias de Macrófagos/metabolismo , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Ligando RANK/farmacología , Fosfatasa Ácida Tartratorresistente , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
17.
Eur J Haematol ; 78(1): 21-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17042762

RESUMEN

BACKGROUND: We analyzed the hematopoietic reconstitution and outcome of 508 patients with multiple myeloma (MM) with respect to the number of CD34+ cells reinfused at our center. PATIENTS AND METHODS: Each cohort of 390 patients (unselected CD34+ cell transplant) and 118 patients (CD34+ selected transplant) was divided into four subgroups. Among the 390 transplantations, 86 patients received a high dose (HD-) of > or =6.50 x 10(6) unselected CD34+ cells/kg, 116 patients a low dose (LD-) of <3.00 x 10(6) CD34+ cells/kg. Among the patients treated with CD34+ selected PBSC, 34 received > or =6.50 x 10(6) CD34+ cells/kg (HD+) and 16 <3.00 x 10(6) CD34+ cells/kg (LD+). RESULTS: HD- patients experienced a reduced median time to leukocyte (13 d vs. 14 d) (P < 0.001) and platelet reconstitution >20 x 10(9)/L (10 d vs. 12 d) (P < 0.001). Similarly, HD+ showed a reduced median time to leukocyte (12 d vs. 15 d) (P < 0.001) and platelet recovery >20 x 10(9)/L (10 d vs. 11 d) (P = 0.058). CD34+ cell-dose was significant for long-term platelet recovery at day 360 (unselected transplant P = 0.015, selected transplant P = 0.023). Number of transplanted CD34+ cells had no significant impact on transplant related mortality, overall survival or CR/PR rates within 100 d. In terms of supportive care the differences of high-/low-dose grafts were minimal. CONCLUSIONS: These results confirm that high doses of CD34+ PBSC shorten hematopoietic reconstitution and reduce hospitalization. Nevertheless secure engraftment results from transplantation of 2.00-3.00 x 10(6) CD34+ cells/kg. As 60% of our pretreated patients are able to collect > or =5.00 x 10(6) CD34+ cells/kg within a single leukapheresis, division into two or more freezing bags allows safe tandem transplantation in the majority of MM patients.


Asunto(s)
Antígenos CD34/biosíntesis , Hematopoyesis , Células Madre Hematopoyéticas/inmunología , Mieloma Múltiple/terapia , Trasplante de Células Madre de Sangre Periférica , Adulto , Anciano , Estudios de Cohortes , Relación Dosis-Respuesta Inmunológica , Femenino , Supervivencia de Injerto , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Recuento de Plaquetas , Pronóstico , Tasa de Supervivencia , Acondicionamiento Pretrasplante , Trasplante Autólogo , Resultado del Tratamiento
18.
Blood ; 109(4): 1669-77, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17023575

RESUMEN

In multiple myeloma (MM) protein kinase C (PKC) signaling pathways have been implicated in cell proliferation, survival, and migration. Here we investigated the novel, orally available PKC-inhibitor enzastaurin for its anti-MM activity. Enzastaurin specifically inhibits phorbol ester-induced activation of PKC isoforms, as well as phosphorylation of downstream signaling molecules MARCKS and PKCmu. Importantly, it also inhibits PKC activation triggered by growth factors and cytokines secreted by bone marrow stromal cells (BMSCs), costimulation with fibronectin, vascular endothelial growth factor (VEGF), or interleukin-6 (IL-6), as well as MM patient serum. Consequently, enzastaurin inhibits proliferation, survival, and migration of MM cell lines and MM cells isolated from multidrug-resistant patients and overcomes MM-cell growth triggered by binding to BMSCs and endothelial cells. Importantly, strong synergistic cytotoxicity is observed when enzastaurin is combined with bortezomib and moderate synergistic or additive effects when combined with melphalan or lenalidomide. Finally, tumor growth, survival, and angiogenesis are abrogated by enzastaurin in an in vivo xenograft model of human MM. Our results therefore demonstrate in vitro and in vivo efficacy of the orally available PKC inhibitor enzastaurin in MM and strongly support its clinical evaluation, alone or in combination therapies, to improve outcome in patients with MM.


Asunto(s)
Indoles/farmacología , Mieloma Múltiple/tratamiento farmacológico , Ésteres del Forbol/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Administración Oral , Animales , Comunicación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Citocinas/farmacología , Sistemas de Liberación de Medicamentos , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/patología , Neovascularización Patológica/tratamiento farmacológico , Fosforilación , Isoformas de Proteínas , Células del Estroma/citología , Trasplante Heterólogo , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas
19.
Blood ; 110(10): 3744-52, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17715391

RESUMEN

The interaction between osteoclasts (OCs) and multiple myeloma (MM) cells plays a key role in the pathogenesis of MM-related osteolytic bone disease (OBD). MM cells promote OC formation and, in turn, OCs enhance MM cell proliferation. Chemokines are mediators of MM effects on bone and vice versa; in particular, CCL3 enhances OC formation and promotes MM cell migration and survival. Here, we characterize the effects of MLN3897, a novel specific antagonist of the chemokine receptor CCR1, on both OC formation and OC-MM cell interactions. MLN3897 demonstrates significant impairment of OC formation (by 40%) and function (by 70%), associated with decreased precursor cell multinucleation and down-regulation of c-fos signaling. OCs secrete high levels of CCL3, which triggers MM cell migration; conversely, MLN3897 abrogates its effects by inhibiting Akt signaling. Moreover, MM cell-to-OC adhesion was abrogated by MLN3897, thereby inhibiting MM cell survival and proliferation. Our results therefore show novel biologic sequelae of CCL3 and its inhibition in both osteoclastogenesis and MM cell growth, providing the preclinical rationale for clinical trials of MLN3897 to treat OBD in MM.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Receptores CCR1/antagonistas & inhibidores , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Fusión Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL3/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Genes fos , Humanos , Mieloma Múltiple/metabolismo , Osteoclastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA