Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurooncol Adv ; 3(1): vdab142, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34729484

RESUMEN

BACKGROUND: This phase I/II trial in patients with recurrent glioblastoma (GBM) evaluates the safety and preliminary efficacy of marizomib, an irreversible pan-proteasome inhibitor that crosses the blood-brain barrier. METHODS: Part A assessed the safety and efficacy of marizomib monotherapy. In Part B, escalating doses of marizomib (0.5-0.8 mg/m2) in combination with bevacizumab were evaluated. Part C explored intra-patient dose escalation of marizomib (0.8-1.0 mg/m2) for the combination. RESULTS: In Part A, 30 patients received marizomib monotherapy. The most common AEs were fatigue (66.7%), headache (46.7%), hallucination (43.3%), and insomnia (43.3%). One patient (3.3%) achieved a partial response. In Part B, the recommended phase II dose of marizomib was 0.8 mg/m2 when combined with bevacizumab 10 mg/kg. In Part C, dose escalation to 1.0 mg/m2 was not tolerated. Pooled analysis of 67 patients treated with marizomib ≤0.8 mg/m2 and bevacizumab showed a nonoverlapping safety profile consistent with the known safety profile of each agent: the most common grade ≥3 AEs were hypertension (16.4%), confusion (13.4%), headache (10.4%), and fatigue (10.4%). The overall response rate was 34.3%, including 2 patients with complete response. Six-month progression-free survival was 29.8%; median overall survival was 9.1 months. CONCLUSIONS: The safety profile of marizomib as monotherapy and in combination with bevacizumab was consistent with previous observations that marizomib crosses the blood-brain barrier. Preliminary efficacy did not demonstrate a meaningful benefit of the addition of marizomib to bevacizumab for the treatment of recurrent GBM.

2.
Bioconjug Chem ; 29(6): 2009-2020, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29734804

RESUMEN

Nanoparticles provide a unique opportunity to explore the benefits of selective distribution and release of cancer therapeutics at sites of disease through varying particle sizes and compositions that exploit the enhanced permeability of tumor-associated blood vessels. Though delivery of larger as opposed to smaller and/or actively transported molecules to the brain is prima facie a challenging endeavor, we wondered whether nanoparticles could improve the therapeutic index of existing drugs for use in treating brain tumors via these vascular effects. We therefore selected a family of nanoparticles composed of cabazitaxel-carboxymethyl cellulose amphiphilic polymers to investigate the potential for delivering a brain-penetrant taxane to intracranial brain tumors in mice. Among a small set of nanoparticle formulations, we found evidence for nanoparticle accumulation in the brain, and one such formulation demonstrated activity in an orthotopic model of glioma, suggesting that such nanoparticles could be useful for the treatment of glioma and brain metastases of other tumor types.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Carboximetilcelulosa de Sodio/química , Nanopartículas/química , Polietilenglicoles/química , Taxoides/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/ultraestructura , Taxoides/química , Taxoides/farmacocinética , Taxoides/uso terapéutico
3.
Br J Haematol ; 180(1): 41-51, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29076150

RESUMEN

Marizomib (MRZ) is an irreversible, pan-subunit proteasome inhibitor (PI) in clinical development for relapsed/refractory multiple myeloma (RRMM) and glioma. This study analysed MRZ, pomalidomide (POM) and low-dose dexamethasone (Lo-DEX) [PMD] in RRMM to evaluate safety and determine the maximum tolerated dose (MTD) and/or recommended Phase 2 dose (RP2D). Intravenous MRZ (0·3-0·5 mg/m2 ) was administered over 2 h on days 1, 4, 8, 11; POM (3-4 mg) on days 1-21; and Lo-DEX (5 or 10 mg) on days 1, 2, 4, 5, 8, 9, 11, 12, 15, 16, 22 and 23 of every 28-day cycle. Thirty-eight patients were enrolled that had received a median of 4 (range 1-10) prior lines of therapy; all patients received prior lenalidomide and bortezomib. No dose-limiting toxicities (DLTs) were observed and 0·5 mg/m2 MRZ was determined to be the RP2D. The most common treatment-related ≥Grade 3 adverse events were: neutropenia (11/38 patients: 29%), pneumonia (4/38 patients 11%), anaemia (4/38 patients; 11%) and thrombocytopenia (4/38 patients; 11%). The overall response rate and clinical benefit rate was 53% (19/36) and 64% (23/36), respectively. In conclusion, PMD was well tolerated and demonstrated promising activity in heavily pre-treated, high-risk RRMM patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Resistencia a Antineoplásicos , Femenino , Humanos , Lactonas/administración & dosificación , Lactonas/farmacocinética , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Pirroles/administración & dosificación , Pirroles/farmacocinética , Recurrencia , Retratamiento , Análisis de Supervivencia , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Talidomida/farmacocinética , Resultado del Tratamiento
4.
Br J Haematol ; 177(2): 221-225, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28387460

RESUMEN

Marizomib, a natural marine product, is an irreversible proteasome inhibitor currently under investigation in relapsed-refractory multiple myeloma (RRMM) and malignant glioma. Central nervous system-multiple myeloma (CNS-MM) is a rare manifestation of extra-medullary disease with few therapeutic options, highlighting the unmet clinical need in these patients. Marizomib demonstrated encouraging activity in RRMM and has emerging clinical activity in glioma, making it a potential CNS-MM therapeutic intervention. Herein, we present two patients with RRMM and CNS involvement who benefited from marizomib-based therapy. These cases provide the first proof of principle for further exploring marizomib in CNS-MM patients.


Asunto(s)
Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Lactonas/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirroles/uso terapéutico , Adulto , Neoplasias del Sistema Nervioso Central/patología , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/patología
5.
Br J Haematol ; 174(5): 711-20, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27161872

RESUMEN

Proteasome inhibitors (PIs) are highly active in multiple myeloma (MM) but resistance is commonly observed. All clinical stage PIs effectively inhibit chymotrypsin-like (CT-L) activity; one possible mechanism of resistance is compensatory hyperactivation of caspase-like (C-L) and trypsin-like (T-L) subunits, in response to CT-L blockade. Marizomib (MRZ), an irreversible PI that potently inhibits all three 20S proteasome subunits with a specificity distinct from other PIs, is currently in development for treatment of MM and malignant glioma. The pan-proteasome pharmacodynamic activity in packed whole blood and peripheral blood mononuclear cells was measured in two studies in patients with advanced solid tumours and haematological malignancies. Functional inhibition of all proteasome subunits was achieved with once- or twice-weekly MRZ dosing; 100% inhibition of CT-L was frequently achieved within one cycle at therapeutic doses. Concomitantly, C-L and T-L activities were either unaffected or increased, suggesting compensatory hyperactivation of these subunits. Importantly, this response was overcome by continued administration of MRZ, with robust inhibition of T-L and C-L (up to 80% and 50%, respectively) by the end of Cycle 2 and maintained thereafter. This enhanced proteasome inhibition was independent of tumour type and may underlie the clinical activity of MRZ in patients resistant to other PIs.


Asunto(s)
Lactonas/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/uso terapéutico , Pirroles/administración & dosificación , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Quimotripsina/efectos de los fármacos , Quimotripsina/metabolismo , Activación Enzimática/efectos de los fármacos , Glioma/tratamiento farmacológico , Humanos , Lactonas/farmacocinética , Lactonas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacocinética , Inhibidores de Proteasoma/farmacología , Pirroles/farmacocinética , Pirroles/farmacología , Tripsina/efectos de los fármacos , Tripsina/metabolismo
6.
Clin Cancer Res ; 22(18): 4559-66, 2016 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-27117181

RESUMEN

PURPOSE: Marizomib (NPI-0052) is an irreversible proteasome inhibitor, derived from a marine actinomycete, with activity and specificity that is distinct from other proteasome inhibitors. EXPERIMENTAL DESIGN: Phase I study (NPI-0052-102) evaluated the MTD, pharmacokinetics, and pharmacodynamics of marizomib intravenously on two dosing schedules. RESULTS: Forty-two patients with advanced malignancies received Schedule A (0.1-0.9 mg/m(2) over 1-10 minutes on days 1, 8, 15 in 4-week cycles); 44 patients with relapsed and/or refractory multiple myeloma (RRMM) and other hematologic malignancies received Schedule B (0.075-0.6 mg/m(2) over 1 minute to 2 hours on days 1, 4, 8, 11, in 3-week cycles). The Schedule A recommended phase II dose was 0.7 mg/m(2) over 10 minutes; Schedule B was 0.5 mg/m(2) over 2 hours. The most common (>25% of patients) related adverse events were fatigue, nausea, diarrhea, and infusion site pain (Schedule A); and fatigue (Schedule B). Overall response rate of 11% was seen in 27 efficacy-evaluable RRMM Schedule B patients (1 very good partial response, 3 partial responses, 4 minimal responses, and 12 stable disease). One Schedule A patient with transformed marginal zone lymphoma had complete response. Marizomib has a short half-life (<30 minutes), with high volume of distribution (∼15-416 L) and clearance (∼0.9-22 L/minutes). CONCLUSIONS: Marizomib does not exhibit the severe peripheral neuropathy or hematologic toxicity observed with other proteasome inhibitors. Marizomib was generally well tolerated with low-dose dexamethasone, demonstrated activity in heavily pretreated RRMM patients, and warrants further evaluation. Clin Cancer Res; 22(18); 4559-66. ©2016 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Lactonas/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Proteasoma/uso terapéutico , Pirroles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Esquema de Medicación , Femenino , Humanos , Lactonas/administración & dosificación , Lactonas/efectos adversos , Lactonas/farmacocinética , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Inhibidores de Proteasoma/administración & dosificación , Inhibidores de Proteasoma/efectos adversos , Inhibidores de Proteasoma/farmacocinética , Pirroles/administración & dosificación , Pirroles/efectos adversos , Pirroles/farmacocinética , Recurrencia , Retratamiento , Resultado del Tratamiento , Adulto Joven
7.
Blood ; 127(22): 2693-700, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27009059

RESUMEN

Marizomib (MRZ) is a novel, irreversible proteasome inhibitor in clinical development for the treatment of relapsed or relapsed and refractory multiple myeloma (RRMM). MRZ inhibits the 3 proteolytic activities of the 20S proteasome with specificity distinct from bortezomib and carfilzomib. Study NPI-0052-101 Part 1 enrolled relapsed or RRMM patients into an open-label, dose-escalation design to determine the maximum tolerated dose and recommended phase 2 dose (RP2D) of MRZ administered intravenously on 2 different schedules: schedule A (0.025-0.7 mg/m(2) once weekly on days 1, 8, and 15 of 4-week cycles) and schedule B (0.15-0.6 mg/m(2) twice weekly on days 1, 4, 8, and 11 of 3-week cycles; concomitant dexamethasone was allowed with schedule B). Patients had received an average of 4.9 and 7.3 prior treatment regimens (schedules A and B, respectively). MRZ schedule A was administered to 32 patients, and the RP2D was established as 0.7 mg/m(2) infused over 10 minutes. Schedule B was administered to 36 patients, and the RP2D was determined to be 0.5 mg/m(2) infused over 2 hours. The most common (>20% of patients) related adverse events were fatigue, headache, nausea, diarrhea, dizziness, and vomiting. Six patients achieved clinical benefit responses (defined as minimal response or better), including 5 partial responses (1 patient on schedule A and 4 on schedule B; 3 of these 4 patients received concomitant dexamethasone). MRZ was generally well tolerated, and results suggest activity in previously treated RRMM patients. Combination studies using pomalidomide and dexamethasone are now underway. The trial was registered at www.clinicaltrials.gov as #NCT00461045.


Asunto(s)
Lactonas/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Pirroles/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Femenino , Humanos , Lactonas/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Pirroles/efectos adversos , Recurrencia , Talidomida/administración & dosificación , Talidomida/efectos adversos , Talidomida/análogos & derivados
8.
Neuro Oncol ; 18(6): 840-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26681765

RESUMEN

BACKGROUND: The proteasome plays a vital role in the physiology of glioblastoma (GBM), and proteasome inhibition can be used as a strategy for treating GBM. Marizomib is a second-generation, irreversible proteasome inhibitor with a more lipophilic structure that suggests the potential for penetrating the blood-brain barrier. While bortezomib and carfilzomib, the 2 proteasome inhibitors approved for treatment of multiple myeloma, have little activity against malignant gliomas in vivo, marizomib could be a novel therapeutic strategy for primary brain tumors. METHODS: The in-vitro antitumor activity of marizomib was studied in glioma cell lines U-251 and D-54. The ability of marizomib to cross the blood-brain barrier and regulate proteasome activities was evaluated in cynomolgus monkeys and rats. The antitumor effect of marizomib in vivo was tested in an orthotopic xenograft model of human GBM. RESULTS: Marizomib inhibited the proteasome activity, proliferation, and invasion of glioma cells. Meanwhile, free radical production and apoptosis induced by marizomib could be blocked by antioxidant N-acetyl cysteine. In animal studies, marizomib distributed into the brain at 30% of blood levels in rats and significantly inhibited (>30%) baseline chymotrypsin-like proteasome activity in brain tissue of monkeys. Encouragingly, the immunocompromised mice, intracranially implanted with glioma xenografts, survived significantly longer than the control animals (P < .05) when treated with marizomib. CONCLUSIONS: These preclinical studies demonstrated that marizomib can cross the blood-brain barrier and inhibit proteasome activity in rodent and nonhuman primate brain and elicit a significant antitumor effect in a rodent intracranial model of malignant glioma.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Glioma/tratamiento farmacológico , Lactonas/farmacología , Inhibidores de Proteasoma/farmacología , Pirroles/farmacología , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Ratones Desnudos
9.
Br J Haematol ; 171(5): 798-812, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26456076

RESUMEN

The proteasome inhibitor bortezomib is an effective therapy for the treatment of relapsed and refractory multiple myeloma (RRMM); however, prolonged treatment can be associated with toxicity, peripheral neuropathy and drug resistance. Our earlier studies showed that the novel proteasome inhibitor marizomib is distinct from bortezomib in its chemical structure, mechanisms of action and effects on proteasomal activities, and that it can overcome bortezomib resistance. Pomalidomide, like lenalidomide, has potent immunomodulatory activity and has been approved by the US Food and Drug Administration for the treatment of RRMM. Here, we demonstrate that combining low concentrations of marizomib with pomalidomide induces synergistic anti-MM activity. Marizomib plus pomalidomide-induced apoptosis is associated with: (i) activation of caspase-8, caspase-9, caspase-3 and PARP cleavage, (ii) downregulation of cereblon (CRBN), IRF4, MYC and MCL1, and (iii) suppression of chymotrypsin-like, caspase-like, and trypsin-like proteasome activities. CRBN-siRNA attenuates marizomib plus pomalidomide-induced MM cells death. Furthermore, marizomib plus pomalidomide inhibits the migration of MM cells and tumour-associated angiogenesis, as well as overcomes cytoprotective effects of bone marrow microenvironment. In human MM xenograft model studies, the combination of marizomib and pomalidomide is well tolerated, inhibits tumour growth and prolongs survival. These preclinical studies provide the rationale for on-going clinical trials of combined marizomib and pomalidomide to improve outcome in patients with RRMM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Mieloma Múltiple/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Lactonas/administración & dosificación , Lactonas/farmacología , Ratones SCID , Péptido Hidrolasas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteasoma/farmacología , Pirroles/administración & dosificación , Pirroles/farmacología , ARN Interferente Pequeño/metabolismo , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Talidomida/farmacología , Trasplante Heterólogo , Ubiquitina-Proteína Ligasas
10.
Anticancer Res ; 29(1): 131-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19331142

RESUMEN

BACKGROUND: The alpha(v)beta3 integrin in the endothelial cell membrane is important for the growth and migration of capillaries into tumour tissue and is also a survival factor for these cells. The alphaII(b)beta3 (GPIIb/IIIa) integrin is responsible for platelet activation and, with concomitant release of different stored proangiogenic factors, and tumour cell-platelet interactions. MATERIALS AND METHODS: An immunodeficient nude rat model was used to study tumour growth in tibial bone, with tumour cells negative for the target alpha(v)beta3 and alphaII(b)beta3 integrins. RESULTS: Daily intraperitoneal injections of m7E3 F(ab')2 antibody fragment, blocking human and rat alpha(v)beta3 and alphaII(b)beta3 integrins, reduced the measured size of the tumours growing in the tibial bone by 35% (p = 0.012), and also the microvessel density in these tumours. The concentration of the important proangiogenic factor bFGF was significantly reduced by 41% in the treated tumours. The treatment slightly increased the time to the appearance of the tumour from 22.2 to 24.9 days, indicating a small but significant effect on the early stages of tumour growth and invasion through the bone tissue. CONCLUSION: Integrin-targeted treatment reduced tumour growth, solely targeting the host angiogenesis. This treatment strategy should be further exploited for use in combination with conventional treatment strategies, or the combined targeting of alternative antiangiogenic pathways.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Femenino , Citometría de Flujo , Células HeLa , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Fragmentos de Inmunoglobulinas/farmacología , Integrina alfaVbeta3/biosíntesis , Integrina alfaVbeta3/inmunología , Masculino , Metástasis de la Neoplasia , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/biosíntesis , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Biol Ther ; 6(10): 1559-68, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17912026

RESUMEN

Alternatively spliced integrins may play an important role in integrin mediated tumor cell adhesion, spreading, and migration. Here we report in human tumor cells a naturally occurring alternatively spliced variant of the beta3 integrin [i.e., truncated (tr) beta3] that lacked a cytoplasmic and a transmembrane domain. The presence of trbeta3 was demonstrated at the mRNA level by RT-PCR, cloning, and sequencing; at the protein level by immunohistochemistry and Western Blotting. The alternately spliced beta3 integrin was detected in human prostate carcinomas, breast carcinomas, and melanoma cells. Expression in vivo was confirmed by immunohistochemistry with an antibody to trbeta3 that does not recognize wild type beta3. Tumor cells secreted this protein and deposited it on the extracellular matrix. Secreted trbeta3 inhibited adhesion of melanoma and prostate cancer cells to fibronectin and vitronectin, which was partially reversed by adsorption of trbeta3 from the media. Confocal microscopy and time lapse live cell microscopy demonstrated that trbeta3 distributed to the trailing edge of migrating cells, which may represent an alternative cell detachment mechanism in these cells. Results suggest that trbeta3 may act as an anti-integrin and play a crucial role in cell migration, which is an important process in tumor invasion and metastasis.


Asunto(s)
Movimiento Celular , Integrina beta3/metabolismo , Neoplasias/patología , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Fibronectinas/metabolismo , Humanos , Inmunohistoquímica , Integrina beta3/genética , Masculino , Datos de Secuencia Molecular , Neoplasias/química , Neoplasias/metabolismo , Estructura Terciaria de Proteína/genética , Transcripción Genética , Vitronectina/metabolismo
12.
Clin Cancer Res ; 13(12): 3689-95, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575234

RESUMEN

PURPOSE: Targeted delivery of cytotoxic agents to solid tumors through cell surface antigens can potentially reduce systemic toxicity and increase the efficacy of the targeted compounds. The purpose of this study was to show the feasibility of treating solid tumors by targeting alpha(v) integrins with antibody-maytansinoid conjugates and to test the relative in vivo activities of several linker-maytansinoid chemistries. EXPERIMENTAL DESIGN: CNTO 364, CNTO 365, and CNTO 366 are targeted cytotoxic agents created by conjugating the CNTO 95 anti-alpha(v) integrin antibody with three distinct maytansinoid-linker structures. These structures were designed to have varying degrees of chemical substitution surrounding the disulfide bond linking the cytotoxic agent to the antibody. A model conjugate was shown to be specifically cytotoxic in vitro and highly active against established human tumor xenografts in immunocompromised rats. The in vivo antitumor activities of CNTO 364, CNTO 365, and CNTO 366 were compared in rat xenograft models. RESULTS: CNTO 365, with a linker chemistry of expected intermediate stability, was shown to be substantially more active than the other two conjugates with lesser or greater substitution around the disulfide linkage. CONCLUSION: CNTO 95-maytansinoid immunoconjugates are potent antitumor agents against alpha(v) integrin-expressing human carcinomas. These studies show for the first time the feasibility of targeting alpha(v) integrins on solid tumors with tumor-activated prodrugs. The DM4 linker-maytansinoid configuration of CNTO 365 was substantially more active in the models tested here when compared with alternative configurations with greater or lesser chemical substitution surrounding the linker.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Inmunoconjugados/administración & dosificación , Integrina alfa5/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Especificidad de Anticuerpos , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Femenino , Citometría de Flujo , Humanos , Inmunoconjugados/química , Inmunoterapia , Ratones , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Res ; 66(6): 3087-95, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540658

RESUMEN

Initially, prostate cancer is androgen dependent. However, most cases progress to an androgen-independent state through unknown mechanisms. Interleukin-6 (IL-6) has been associated with prostate cancer progression including activation of the androgen receptor (AR). To determine if IL-6 plays a role in the conversion of prostate cancer from androgen dependent to androgen independent, we established androgen-dependent LuCaP 35 human prostate cancer xenografts in nude mice, castrated the mice, and blocked IL-6 activity using a neutralizing antibody (CNT0328) for a period of 18 weeks. IL-6 inhibition increased survival of mice and inhibited tumor growth, as reflected by decreased tumor volume and prostate-specific antigen levels, compared with that in mice receiving isotype control antibody. To test the effect of IL-6 inhibition on the conversion from androgen dependent to androgen independent, tumor cells from the treated mice were assessed for their androgen dependence both in vitro and by implanting them into sham-operated or orchiectomized mice. Tumor cells derived from the isotype-treated animals converted to androgen-independent state, whereas tumor cells from the anti-IL-6 antibody-treated mice were still androgen dependent in vitro and in vivo. Although there was no difference in AR levels between the androgen-independent and androgen-dependent tumors, IL-6 inhibition promoted both apoptosis and inhibited cell proliferation in tumors and blocked the orchiectomy-induced expression of histone acetylases, p300 and CBP, which are AR cofactors. These data show that IL-6 contributes to the development of androgen independence in prostate cancer and suggest that it mediates this effect, in part, through modulation of p300 and CBP.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Interleucina-6/antagonistas & inhibidores , Neoplasias Hormono-Dependientes/patología , Neoplasias Hormono-Dependientes/terapia , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Interleucina-6/inmunología , Interleucina-6/fisiología , Masculino , Ratones , Ratones Desnudos , Orquiectomía , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Int J Cancer ; 116(1): 27-35, 2005 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15761867

RESUMEN

Previous studies indicated that transfection of the platelet integrin alphaIIbbeta3 into human melanoma cells expressing integrin alphavbeta3 promoted their in vivo (but not in vitro) growth and cell survival. To reveal the underlying pathomechanism, we have analyzed the angiogenic phenotype of alphaIIbbeta3 integrin-transduced human melanoma cells expressing integrin alphavbeta3. Upon heterotopic or orthotopic (intracutaneous) injections into SCID mice, the alphaIIbbeta3 integrin-overexpressing clones, ESL, ESH, 19L and 19H, grew more rapidly than the mock transfectant (alphavbeta3 expressing) clone, 3.1P. Morphometry demonstrated an increased intratumoral microvessel density in 19L and 19H tumors compared to 3.1P. Immunocytochemistry and flow cytometry indicated that vascular endothelial growth factor (VEGF) is constitutively expressed in the majority of the cells of both the mock and the alphaIIbbeta3 integrin-transfected clones. However, the mock transfectant clone, 3.1P, did not express basic fibroblast growth factor (bFGF) at protein level (<1%), unlike the alphaIIbbeta3 integrin-transfected clones, 19L and 19H, (33.9 and 84.1%, respectively). Quantitative PCR analysis of 6 related human melanoma clones with various levels of alphaIIbbeta3 integrin expressions revealed a correlation between the alphaIIb protein and bFGF mRNA expressions. Furthermore, cDNA microarray analysis of the 19H cells revealed 12 downregulated and 36 upregulated genes [among them 3 upregulated vasculogenic mimicry-genes (CD34, endothelin receptor B, Prostaglandin I-2 synthase)] when compared to 3.1P cells. The altered bFGF expression may be influenced by integrin-linked signaling, since bbeta3-endonexin is upregulated in alphaIIbbeta3-transfected cells and tyrosine kinase inhibitors downregulate bFGF both at mRNA and protein levels. We propose here that the illegitimate expression of alphaIIbbeta3 integrin in human melanoma cells already expressing alphavbeta3 integrin may alter their in vivo growth properties due to the modulation of their angiogenic phenotype.


Asunto(s)
Integrina alfaVbeta3/metabolismo , Melanoma/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Animales , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Integrina alfaVbeta3/genética , Ratones , Ratones SCID , Neovascularización Patológica , Fenotipo , Transducción de Señal , Transfección
15.
J Support Oncol ; 3(1): 37-50, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15724944

RESUMEN

The prevalence of depression among patients diagnosed with cancer is higher than among the general medical population and is associated with faster tumor progression and shortened survival time. Cancer-related depression often occurs in association with anorexia and cachexia, although until recently the relationship between these conditions has not been well understood. Cachexia is associated with poorer quality of life and survival outcomes and is theeventual cause of death in approximately 30% of all patients with cancer. Recent evidence has linked elevated levels of inflammatory cytokines with both depression and cachexia, and experiments have shown that introducing cytokines induces depression and cachectic symptoms in both humans and rodents, suggesting that there may be a common etiology at the molecular level. Therapeutic agents targeting specific cytokine molecules, such as interleukin-6 or tumor necrosis factor-alpha, are currently being evaluated for their potential to simultaneously treat both depression and cachexia pharmacologically. This review summarizes the available data suggesting a dual role for cytokines in the development of cancer-related depression and cachexia and describes how biologic therapies targeting specific cytokines may improve outcomes beyond depression and cachexia, such as survival and quality of life.


Asunto(s)
Caquexia/diagnóstico , Citocinas/metabolismo , Trastorno Depresivo/diagnóstico , Trastorno Depresivo/tratamiento farmacológico , Neoplasias/diagnóstico , Calidad de Vida , Animales , Antidepresivos/uso terapéutico , Biomarcadores de Tumor/análisis , Caquexia/epidemiología , Caquexia/terapia , Ensayos Clínicos como Asunto , Terapia Combinada , Citocinas/análisis , Trastorno Depresivo/epidemiología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Neoplasias/epidemiología , Pronóstico , Medición de Riesgo , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
16.
Int J Cancer ; 111(4): 592-5, 2004 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-15239138

RESUMEN

IL-6 is a multifunctional cytokine implicated in several cancers. IL-6 is a growth factor for certain tumors and contributes to drug resistance, cachexia and bone resorption. Cachexia is characterized by progressive weight loss and depletion of host reserves of adipose tissue and skeletal muscle. We have developed CNTO 328 (cCLB8), a human-mouse chimeric MAb to IL-6 (K(d) approx. 10(-12) M) that inhibits IL-6 function. A phase I study with CNTO 328 in multiple myeloma patients demonstrated that the antibody was safe and had a circulating half-life of approximately 17 days. Since IL-6 is implicated in cachexia, we hypothesized that CNTO 328 could inhibit tumor-induced cachexia. We used 2 human tumor-induced cachexia models in nude mice. In the first model, human melanoma cells were inoculated in female nude mice. Control treated animals lost 19% (+/-7.7%) body weight from day 0 to day 31, whereas CNTO 328 (10 mg/kg)-treated animals lost only 1.5% (+/-1.3%) body weight from day 0 to day 31 (p = 0.023). In the second cachexia model, human prostate tumor cells were injected into male nude mice. By day 29, control treated animals lost 6% (+/-3.5%) body weight, whereas CNTO 328 (10 mg/kg)-treated animals gained 7% (+/-4%) body weight (p = 0.01). Since CNTO 328 blocks human IL-6 but not mouse IL-6, the data indicate that tumor cell-secreted IL-6 directly contributes to body weight loss, highlighting the potential role for CNTO 328 as an anticachectic agent.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Caquexia/inmunología , Caquexia/terapia , Interleucina-6/inmunología , Interleucina-6/farmacología , Melanoma/complicaciones , Neoplasias de la Próstata/complicaciones , Neoplasias Cutáneas/complicaciones , Animales , Peso Corporal , Modelos Animales de Enfermedad , Humanos , Masculino , Melanoma/veterinaria , Ratones , Ratones Desnudos , Neoplasias de la Próstata/veterinaria , Neoplasias Cutáneas/veterinaria , Trasplante Heterólogo
17.
Int J Cancer ; 110(3): 326-35, 2004 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15095296

RESUMEN

Integrins of the alphav family, such as alphavbeta3 and alphavbeta5, are implicated in tumor-induced angiogenesis; but their role in tumor growth has not been fully explored. CNTO 95 is a fully human antibody that recognizes the alphav family of integrins and is likely to be less immunogenic in humans compared to chimeric or humanized antibodies. CNTO 95 bound to purified alphavbeta3 and alphavbeta5 with a Kd of approximately 200 pM and to alphav integrin-expressing human cells with a Kd of 1-24 nM. In vitro, CNTO 95 inhibited human melanoma cell adhesion, migration and invasion at doses ranging 7-20 nM. In a rat aortic ring sprouting assay, CNTO 95 (approx. 70 nM) completely inhibited sprouting. Using a human melanoma xenograft model in nude mice wherein CNTO 95 recognized alphavbeta3 and alphavbeta5 on human tumor cells but not mouse angiogenic integrins, CNTO 95 (10 mg/kg, 3 times/week) inhibited growth of human melanoma tumors in nude mice by approximately 80% (p = 0.0005), suggesting that CNTO 95 inhibited human tumor growth independently of its antiangiogenic activity. In a nude rat human xenograft model where CNTO 95 binds and blocks both tumor and host integrins, this antibody (10 mg/kg once/week) reduced final tumor weight by >99% (p < 0.0001). Based on these preclinical data, a dose-escalating phase I clinical trial in cancer patients has been initiated. To our knowledge, CNTO 95 is the first fully human MAb to alphav integrins that has potent antitumor and antiangiogenic properties in in vivo preclinical models.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Integrina alfaV/química , Melanoma/terapia , Animales , Anticuerpos Monoclonales Humanizados , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Aorta/metabolismo , Aorta/patología , Western Blotting , Bovinos , Adhesión Celular , División Celular , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Colágeno/farmacología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Endotelio Vascular/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Haplorrinos , Humanos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Cinética , Laminina/farmacología , Macaca fascicularis , Melanoma/inmunología , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica , Placenta/metabolismo , Placenta/patología , Unión Proteica , Proteoglicanos/farmacología , Ratas , Ratas Desnudas , Receptores de Vitronectina/metabolismo , Factores de Tiempo
18.
Clin Cancer Res ; 9(13): 4653-65, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14581334

RESUMEN

Interleukin (IL)-6, a pleiotropic cytokine with varied systemic functions, plays a major role in inflammatory processes. It modulates the transcription of several liver-specific genes during acute inflammatory states, particularly C-reactive protein, and controls the survival of normal plasmablastic cells. In addition, IL-6 has been implicated in hematopoiesis as a cofactor in stem cell amplification and differentiation. This article is the first review of clinical studies in the 1990s with anti-IL-6 monoclonal antibodies (mAbs) in the treatment of patients with cancer and related lymphoproliferative disorders. In six clinical studies of mAbs to IL-6 with BE-8 or CNTO 328 in patients with multiple myeloma, renal cell carcinoma, and B-lymphoproliferative disorders, anti-IL-6 mAb treatment decreased C-reactive protein levels in all patients. In most patients, levels decreased below detectable limits. The antibodies were well tolerated, and no serious adverse effects were observed in the vast majority of studies. The fact that anti-IL-6 mAb therapy decreased the incidence of cancer-related anorexia and cachexia may also be useful in the treatment of cancer patients.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoterapia/métodos , Interleucina-6/química , Interleucina-6/fisiología , Neoplasias/terapia , Proteína C-Reactiva/biosíntesis , Carcinoma de Células Renales/terapia , Humanos , Neoplasias Renales/terapia , Linfoma de Células B/terapia , Mieloma Múltiple/terapia
19.
Clin Exp Metastasis ; 20(5): 413-20, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14524530

RESUMEN

The growth of metastatic prostate cancer cells in the bone involves an intimate interaction between the tumor cells and various elements of the bone microenvironment, resulting in increased rate of bone turnover and rapid tumor growth. The alpha(v)beta3 integrin has been shown to play an important role in tumor growth and angiogenesis, and is known to be critical to osteoclast formation and activity. This study was designed to examine the role of alpha(v)beta3 expressed by cells native to the bone in the growth and pathogenesis of prostate cancer bone metastases. Human prostate cancer cells which do not express alpha(v)beta3 or alpha(IIb)beta3 integrins were injected directly into human bone fragments previously implanted subcutaneously in SCID mice (SCID-human-bone model). At the same time treatment with anti-beta3 antibody fragment (m7E3 F(ab')2) i.p. at 300 microg/dose 3 x per week was initiated and continued for 2 weeks. In this system, m7E3 F(ab')2 only recognizes human bone-derived alpha(v)beta3. Antibody inhibition of alpha(v)beta3 integrin in vivo resulted in a specific reduction in the proportion of antigenically-human blood vessels within tumor-bearing bone implants (from 73.5% +/- 3.93 in controls to 17.74% +/- 5.64 in treated animals). Proliferation of the alpha(v)beta3-negative tumor cells was also reduced, although the overall vessel density was maintained by compensating mouse vasculature. Blockage of human bone-derived alpha(v)beta3 also significantly reduced the recruitment of osteoclasts in response to tumor cells, as well as degradation of calcified bone tissue. Together these observations confirm the importance of alpha(v)beta3 in bone metabolism and angiogenesis, and point to the role of these processes in controlling growth of metastatic prostate cancer cells in the bone.


Asunto(s)
Neoplasias Óseas/patología , Huesos/metabolismo , Integrina alfaVbeta3/antagonistas & inhibidores , Neovascularización Patológica , Animales , Apoptosis , División Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Microscopía Fluorescente , Metástasis de la Neoplasia , Osteoclastos/metabolismo , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas
20.
Curr Opin Biotechnol ; 13(6): 609-14, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12482522

RESUMEN

The specificity of antibodies has been harnessed to target cancer cells and the first therapeutic antibodies for use in oncology are now finding application in the clinic. Studies are currently under way to develop new and improved antibodies. Recent developments have been made in the identification of novel targets, including the use of genomic and proteomic technologies. Several methods are also being developed to enhance antibody efficacy.


Asunto(s)
Anticuerpos Monoclonales/clasificación , Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Ensayos Clínicos Fase III como Asunto , Terapia Combinada/métodos , Aprobación de Drogas , Humanos , Ratones , Neoplasias/inmunología , Control de Calidad , Radioinmunoterapia/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA