Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Int J Clin Oncol ; 21(1): 1-12, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26081252

RÉSUMÉ

The purpose of this article is to disseminate the standard of antiemetic therapy for Japanese clinical oncologists. On the basis of the Appraisal of Guidelines for Research and Evaluation II instrument, which reflects evidence-based clinical practice guidelines, a working group of the Japanese Society of Clinical Oncology (JSCO) reviewed clinical practice guidelines for antiemesis and performed a systematic review of evidence-based domestic practice guidelines for antiemetic therapy in Japan. In addition, because health-insurance systems in Japan are different from those in other countries, a consensus was reached regarding standard treatments for chemotherapy that induce nausea and vomiting. Current evidence was collected by use of MEDLINE, from materials from meetings of the American Society of Clinical Oncology National Comprehensive Cancer Network, and from European Society of Medical Oncology/Multinational Association of Supportive Care in Cancer guidelines for antiemesis. Initially, 21 clinical questions (CQ) were selected on the basis of CQs from other guidelines. Patients treated with highly emetic agents should receive a serotonin (5-hydroxytryptamine; 5HT3) receptor antagonist, dexamethasone, and a neurokinin 1 receptor antagonist. For patients with moderate emetic risk, 5HT3 receptor antagonists and dexamethasone were recommended, whereas for those receiving chemotherapy with low emetic risk dexamethasone only is recommended. Patients receiving high-emetic-risk radiation therapy should also receive a 5HT3 receptor antagonist. In this paper the 2010 JSCO clinical practice guidelines for antiemesis are presented in English; they reveal high concordance of Japanese medical circumstances with other antiemetic guidelines that are similarly based on evidence.


Sujet(s)
Antiémétiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Oncologie médicale , Nausée/induit chimiquement , Guides de bonnes pratiques cliniques comme sujet , Vomissement/induit chimiquement , Dexaméthasone/usage thérapeutique , Humains , Japon , Nausée/traitement médicamenteux , Antagonistes des récepteurs 5-HT3 de la sérotonine/usage thérapeutique , Sociétés médicales , Facteurs temps , Vomissement/traitement médicamenteux
2.
Oncol Rep ; 30(3): 1053-8, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23836376

RÉSUMÉ

Various anticancer drugs, including camptothecins and indolocarbazoles, target DNA topoisomerase I (Top1). We previously described the camptothecin-resistant colon cancer cell line DLDSNR6, which has a Gly365Ser missense mutation in Top1. In the present study, we established highly camptothecin-resistant sublines from DLDSNR6 cells by continuous exposure to higher camptothecin concentrations. The established sublines grew in the presence of 30 µM of camptothecin, but exhibited markedly retarded growth. In addition to Gly365Ser, these sublines harbored a Top1 Gly717Arg mutation and some had also a Top1 Gln421Arg mutation. Top1 activity was reduced to approximately one-eighth in highly resistant cell lines compared with that in parental DLD-1 cells. Resistant clones with 3 Top1 mutations including Gln421RArg exhibited the highest resistance to the indolocarbazole J-107088 in terms of the effect on the cell cycle distribution. The Gln421 mutation was equivalent to a mutation recently found in camptothecin biosynthesizing plants, but it has not previously been found in mammalian cells.


Sujet(s)
Camptothécine/pharmacologie , Tumeurs du côlon/génétique , ADN topoisomérases de type I/génétique , Résistance aux médicaments antinéoplasiques/génétique , Mutation faux-sens/génétique , Inhibiteurs de la topoisomérase-I/pharmacologie , Transporteurs ABC/génétique , Transporteurs ABC/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Carbazoles/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/métabolisme , ADN topoisomérases de type I/composition chimique , ADN topoisomérases de type I/métabolisme , ADN tumoral/génétique , Cytométrie en flux , Humains , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , RT-PCR , Cellules cancéreuses en culture
3.
Blood ; 116(9): 1460-8, 2010 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-20505158

RÉSUMÉ

In this study, we demonstrate expression and examined the biologic sequelae of PI3K/p110delta signaling in multiple myeloma (MM). Knockdown of p110delta by small interfering RNA caused significant inhibition of MM cell growth. Similarly, p110delta specific small molecule inhibitor CAL-101 triggered cytotoxicity against LB and INA-6 MM cell lines and patient MM cells, associated with inhibition of Akt phosphorylation. In contrast, CAL-101 did not inhibit survival of normal peripheral blood mononuclear cells. CAL-101 overcame MM cell growth conferred by interleukin-6, insulin-like growth factor-1, and bone marrow stromal cell coculture. Interestingly, inhibition of p110delta potently induced autophagy. The in vivo inhibition of p110delta with IC488743 was evaluated in 2 murine xenograft models of human MM: SCID mice bearing human MM cells subcutaneously and the SCID-hu model, in which human MM cells are injected within a human bone chip implanted subcutaneously in SCID mice. IC488743 significantly inhibited tumor growth and prolonged host survival in both models. Finally, combined CAL-101 with bortezomib induced synergistic cytotoxicity against MM cells. Our studies therefore show that PI3K/p110delta is a novel therapeutic target in MM and provide the basis for clinical evaluation of CAL-101 to improve patient outcome in MM.


Sujet(s)
Mouvement cellulaire , Myélome multiple/thérapie , Phosphatidylinositol 3-kinases/métabolisme , Purines/pharmacologie , Quinazolinones/pharmacologie , Animaux , Marqueurs biologiques/métabolisme , Technique de Western , Moelle osseuse/métabolisme , Adhérence cellulaire , Prolifération cellulaire , Cellules cultivées , Phosphatidylinositol 3-kinases de classe I , Endothélium vasculaire/cytologie , Endothélium vasculaire/métabolisme , Antienzymes/pharmacologie , Test ELISA , Technique d'immunofluorescence , Analyse de profil d'expression de gènes , Humains , Souris , Souris SCID , Myélome multiple/génétique , Myélome multiple/métabolisme , Séquençage par oligonucléotides en batterie , Phosphatidylinositol 3-kinases/génétique , Inhibiteurs des phosphoinositide-3 kinases , Phosphorylation , Petit ARN interférent/pharmacologie , Cellules souches/métabolisme , Veines ombilicales/cytologie , Veines ombilicales/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Eur J Haematol ; 85(1): 68-75, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20192985

RÉSUMÉ

OBJECTIVES: Bortezomib (PS-341; Velcade), a proteasome inhibitor, is used as a therapeutic agent for multiple myeloma. Bortezomib has been shown to strongly induce osteoblast differentiation and elevate the levels of osteoblast-related differentiation markers in the serum of patients with myeloma. Bortezomib also reportedly increases the activity of the transcription factor, Runx2. However, the mechanism of action by which bortezomib-elevated Runx2 activity mediates osteoblast differentiation remains unclear. On the other hand, fibroblast growth factor 2 (FGF-2) is found at high levels in patients with multiple myeloma. We previously reported that FGF-2 reduces the levels of the transcriptional coactivator with PDZ-binding motif (TAZ). We therefore investigated the effects of bortezomib on TAZ protein levels in the presence of FGF-2. METHODS: Osteoblastic MC3T3-E1 cells were treated with different concentrations of bortezomib in the presence or absence of FGF-2 and various biologic responses were investigated by immunoblotting, RT-PCR, quantitative PCR, and alizarin red staining. RESULTS: We found that bortezomib inhibited FGF-2-induced reduction of TAZ levels through a pathway other than that used for proteasome inhibition, while maintaining TAZ function, which in turn, enhanced the expression of Runx2-transcribed osteogenic differentiation markers. Bortezomib also suppressed the antimineralization effect of FGF-2. CONCLUSIONS: These findings suggest that bortezomib inhibited FGF-2-induced reduction of TAZ and consequently stimulated osteogenic differentiation independently of proteasome inhibition. These findings may contribute to elucidate the osteolytic mechanism in multiple myeloma, and to the development of new drugs for multiple myeloma and other osteolytic diseases.


Sujet(s)
Acides boroniques/pharmacologie , Facteur de croissance fibroblastique de type 2/pharmacologie , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Inhibiteurs du protéasome , Pyrazines/pharmacologie , Facteurs de transcription/métabolisme , Cellules 3T3 , Acyltransferases , Animaux , Séquence nucléotidique , Acides boroniques/administration et posologie , Bortézomib , Différenciation cellulaire/effets des médicaments et des substances chimiques , Sous-unité alpha 1 du facteur CBF/métabolisme , Amorces ADN/génétique , Humains , Souris , Modèles biologiques , Myélome multiple/complications , Myélome multiple/traitement médicamenteux , Ostéoblastes/cytologie , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéolyse/traitement médicamenteux , Ostéolyse/étiologie , Inhibiteurs de protéases/administration et posologie , Inhibiteurs de protéases/pharmacologie , Pyrazines/administration et posologie , ARN messager/génétique , ARN messager/métabolisme , Protéines recombinantes/pharmacologie , Facteurs de transcription/génétique
6.
Clin Cancer Res ; 15(18): 5829-39, 2009 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-19737962

RÉSUMÉ

PURPOSE: The increasing incidence of osteonecrosis of the jaw and its possible association with high cumulative doses of bisphosphonate led us to study the effects of high doses of zoledronic acid (ZA) on bone remodeling. EXPERIMENTAL DESIGN: Five-week-old C57BL6 mice were treated with saline or ZA weekly for 3 weeks at increasing doses (0.05-1 mg/Kg). Effects of ZA on bone remodeling were studied using standard assays. RESULTS: We observed an increase in bone mineral density and content in treated animals at doses of 0.05 mg/Kg, which was not further enhanced at higher doses of ZA. Trabecular bone volume at the proximal tibia and the distal femur assessed by histomorphometry and microCT, respectively, increased significantly in ZA-treated groups. There was however no difference between 0.5 and 1 mg/kg, suggesting a ceiling effect for ZA. ZA led to decreased numbers of osteoclasts and osteoblasts per bone perimeter that paralleled a significant reduction of serum levels of TRAC5b and osteocalcin in vivo. Effects on osteoblasts were confirmed in in vitro assays. Mechanical testing of the femur showed increased brittleness in ZA-treated mice. CONCLUSIONS: High doses of ZA inhibit both osteoclast and osteoblasts function and bone remodeling in vivo interfering with bone mechanical properties. No dose response was noted beyond 0.5 mg/kg suggesting that lower doses of ZA may be adequate in inhibiting bone resorption. Our data may help inform future studies of ZA use with respect to alternate and lower doses in the treatment of patients with cancer bone disease.


Sujet(s)
Remodelage osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/effets des médicaments et des substances chimiques , Os et tissu osseux/physiologie , Lignage cellulaire/effets des médicaments et des substances chimiques , Diphosphonates/administration et posologie , Diphosphonates/pharmacologie , Imidazoles/administration et posologie , Imidazoles/pharmacologie , Ostéoblastes/effets des médicaments et des substances chimiques , Adulte , Animaux , Densité osseuse/effets des médicaments et des substances chimiques , Agents de maintien de la densité osseuse/administration et posologie , Agents de maintien de la densité osseuse/pharmacologie , Relation dose-effet des médicaments , Femelle , Humains , Souris , Souris de lignée C57BL , Ostéoblastes/cytologie , Ostéoblastes/physiologie , Cellules cancéreuses en culture , Acide zolédronique
7.
Clin Cancer Res ; 15(12): 4028-37, 2009 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-19509164

RÉSUMÉ

PURPOSE: We investigated the antitumor effect of murine/human chimeric CD138-specific monoclonal antibody nBT062 conjugated with highly cytotoxic maytansinoid derivatives against multiple myeloma (MM) cells in vitro and in vivo. EXPERIMENTAL DESIGN: We examined the growth inhibitory effect of BT062-SPDB-DM4, BT062-SMCC-DM1, and BT062-SPP-DM1 against MM cell lines and primary tumor cells from MM patients. We also examined in vivo activity of these agents in murine MM cell xenograft model of human and severe combined immunodeficient (SCID) mice bearing implant bone chips injected with human MM cells (SCID-hu model). RESULTS: Anti-CD138 immunoconjugates significantly inhibited growth of MM cell lines and primary tumor cells from MM patients without cytotoxicity against peripheral blood mononuclear cells from healthy volunteers. In MM cells, they induced G(2)-M cell cycle arrest, followed by apoptosis associated with cleavage of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase. Nonconjugated nBT062 completely blocked cytotoxicity induced by nBT062-maytansinoid conjugate, confirming that specific binding is required for inducing cytotoxicity. Moreover, nBT062-maytansinoid conjugates blocked adhesion of MM cells to bone marrow stromal cells. The coculture of MM cells with bone marrow stromal cells protects against dexamethasone-induced death but had no effect on the cytotoxicity of immunoconjugates. Importantly, nBT062-SPDB-DM4 and nBT062-SPP-DM1 significantly inhibited MM tumor growth in vivo and prolonged host survival in both the xenograft mouse models of human MM and SCID-hu mouse model. CONCLUSION: These results provide the preclinical framework supporting evaluation of nBT062-maytansinoid derivatives in clinical trials to improve patient outcome in MM.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques d'origine végétale/usage thérapeutique , Immunoconjugués/usage thérapeutique , Maitansine/usage thérapeutique , Myélome multiple/traitement médicamenteux , Syndécane-1/immunologie , Animaux , Cellules de la moelle osseuse/métabolisme , Lignée cellulaire tumorale , Humains , Facteur de croissance IGF-I/métabolisme , Interleukine-6/métabolisme , Souris , Souris SCID , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Cellules stromales/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Blood ; 114(5): 1046-52, 2009 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-19436050

RÉSUMÉ

Bortezomib is a proteasome inhibitor with remarkable preclinical and clinical antitumor activity in multiple myeloma (MM) patients. The initial rationale for its use in MM was inhibition of nuclear factor (NF)-kappaB activity by blocking proteasomal degradation of inhibitor of kappaBalpha (IkappaBalpha). Bortezomib inhibits inducible NF-kappaB activity; however, its impact on constitutive NF-kappaB activity in MM cells has not yet been defined. In this study, we demonstrate that bortezomib significantly down-regulated IkappaBalpha expression and triggered NF-kappaB activation in MM cell lines and primary tumor cells from MM patients. Importantly, no inhibition of p65 (RelA) nuclear translocation was recognized after bortezomib treatment in a murine xenograft model bearing human MM cells. Bortezomib-induced NF-kappaB activation was mediated via the canonical pathway. Moreover, other classes of proteasome inhibitors also induced IkappaBalpha down-regulation associated with NF-kappaB activation. Molecular mechanisms whereby bortezomib induced IkappaBalpha down-regulation were further examined. Bortezomib triggered phosphorylation of IkappaB kinase (IKKbeta) and its upstream receptor-interacting protein 2, whereas IKKbeta inhibitor MLN120B blocked bortezomib-induced IkappaBalpha down-regulation and NF-kappaB activation, indicating receptor-interacting protein 2/IKKbeta signaling plays crucial role in bortezomib-induced NF-kappaB activation. Moreover, IKKbeta inhibitors enhanced bortezomib-induced cytotoxicity. Our studies therefore suggest that bortezomib-induced cytotoxicity cannot be fully attributed to inhibition of canonical NF-kappaB activity in MM cells.


Sujet(s)
Antinéoplasiques/pharmacologie , Acides boroniques/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Myélome multiple/génétique , Facteur de transcription NF-kappa B/effets des médicaments et des substances chimiques , Protéines tumorales/effets des médicaments et des substances chimiques , Inhibiteurs de protéases/pharmacologie , Pyrazines/pharmacologie , Animaux , Antinéoplasiques/usage thérapeutique , Acides boroniques/usage thérapeutique , Bortézomib , Régulation négative/effets des médicaments et des substances chimiques , Activation enzymatique/effets des médicaments et des substances chimiques , Humains , I-kappa B Kinase/antagonistes et inhibiteurs , I-kappa B Kinase/métabolisme , Protéines I-kappa B/métabolisme , Souris , Souris SCID , Myélome multiple/traitement médicamenteux , Inhibiteur alpha de NF-KappaB , Facteur de transcription NF-kappa B/physiologie , Protéines tumorales/physiologie , Phosphorylation , Inhibiteurs de protéases/usage thérapeutique , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , Pyrazines/usage thérapeutique , Receptor-Interacting Protein Serine-Threonine Kinase 2/métabolisme , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Blood ; 113(21): 5228-36, 2009 May 21.
Article de Anglais | MEDLINE | ID: mdl-19270264

RÉSUMÉ

Nuclear factor-kappaB (NF-kappaB) has an important role in multiple myeloma (MM) cell pathogenesis in the context of the bone marrow (BM) microenvironment. In NF-kappaB signaling cascades, IkappaB kinase alpha (IKKalpha) and IKKbeta are key molecules that predominantly mediate noncanonical and canonical pathways, respectively. In this study, we examined the biologic sequelae of the inhibition of IKKalpha versus IKKbeta in MM cell lines. All MM cell lines have constitutive canonical NF-kappaB activity, and a subset of MM cell lines shows noncanonical NF-kappaB activity. Adhesion to BM stromal cells further activates both canonical and noncanonical NF-kappaB activity. IKKbeta inhibitor MLN120B blocks canonical pathway and growth of MM cell lines but does not inhibit the noncanonical NF-kappaB pathway. Although IKKalpha knockdown induces significant growth inhibition in the cell lines with both canonical and noncanonical pathways, it does not inhibit NF-kappaB activation. Importantly, IKKalpha down-regulation decreases expression of beta-catenin and aurora-A, which are known to mediate MM cell growth and survival. Finally, IKKbeta inhibitor enhances the growth inhibition triggered by IKKalpha down-regulation in MM cells with both canonical and noncanonical NF-kappaB activity. Combination therapy targeting these kinases therefore represents a promising treatment strategy in MM.


Sujet(s)
I-kappa B Kinase/antagonistes et inhibiteurs , Myélome multiple/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Cellules de la moelle osseuse , Adhérence cellulaire , Lignée cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Myélome multiple/métabolisme , Sous-unités de protéines , Transduction du signal , Cellules stromales/cytologie
10.
Blood ; 113(4): 846-55, 2009 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-18948577

RÉSUMÉ

Heat-shock protein 90 (Hsp90) acts as a molecular chaperone required for maintaining the conformational stability of client proteins regulating cell proliferation, survival, and apoptosis. Here we investigate the biologic significance of Hsp90 inhibition in multiple myeloma (MM) and other hematologic tumors using an orally available novel small molecule inhibitor SNX-2112, which exhibits unique activities relative to 17-allyamino-17-demethoxy-geldanamycin (17-AAG). SNX-2112 triggers growth inhibition and is more potent than 17-AAG against MM and other malignancies. It induces apoptosis via caspase-8, -9, -3, and poly (ADP-ribose) polymerase cleavage. SNX-2112 inhibits cytokine-induced Akt and extracellular signal-related kinase (ERK) activation and also overcomes the growth advantages conferred by interleukin-6, insulin-like growth factor-1, and bone marrow stromal cells. Importantly, SNX-2112 inhibits tube formation by human umbilical vein endothelial cells via abrogation of eNOS/Akt pathway and markedly inhibits osteoclast formation via down-regulation of ERK/c-fos and PU.1. Finally, SNX-2112, delivered by its prodrug SNX-5422, inhibits MM cell growth and prolongs survival in a xenograft murine model. Our results indicate that blockade of Hsp90 by SNX-2112 not only inhibits MM cell growth but also acts in the bone marrow microenvironment to block angiogenesis and osteoclastogenesis. Taken together, our data provide the framework for clinical studies of SNX-2112 to improve patient outcome in MM and other hematologic malignancies.


Sujet(s)
Extracellular Signal-Regulated MAP Kinases/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Leucémies/métabolisme , Myélome multiple/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Protéines du choc thermique HSP90/métabolisme , Composés hétérocycliques avec 4 noyaux ou plus/composition chimique , Composés hétérocycliques avec 4 noyaux ou plus/usage thérapeutique , Humains , Facteur de croissance IGF-I/pharmacologie , Interleukine-6/pharmacologie , Leucémies/traitement médicamenteux , Leucémies/anatomopathologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Souris SCID , Structure moléculaire , Myélome multiple/vascularisation , Myélome multiple/traitement médicamenteux , Myélome multiple/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Ostéoclastes/cytologie , Ostéoclastes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Br J Haematol ; 141(5): 659-71, 2008 May.
Article de Anglais | MEDLINE | ID: mdl-18410446

RÉSUMÉ

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.


Sujet(s)
Cérulénine/usage thérapeutique , Fatty acid synthases/antagonistes et inhibiteurs , Myélome multiple/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Caspase-3/biosynthèse , Caspase 8/biosynthèse , Caspase-9/biosynthèse , Lignée cellulaire tumorale , Cérulénine/pharmacologie , Systèmes de délivrance de médicaments , Évaluation de médicament , Chaperonne BiP du réticulum endoplasmique , Activation enzymatique , Humains , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 4/métabolisme , Myélome multiple/enzymologie , Transduction du signal , Cellules cancéreuses en culture , Antigènes CD95/métabolisme
12.
Br J Haematol ; 141(5): 598-606, 2008 May.
Article de Anglais | MEDLINE | ID: mdl-18397345

RÉSUMÉ

The interaction between multiple myeloma (MM) cells and the bone marrow (BM) microenvironment induces proliferation and survival of MM cells, as well as osteoclastogenesis. This study investigated the therapeutic potential of novel p38 mitogen-activated protein kinase (p38MAPK) inhibitor LY2228820 (LY) in MM. Although cytotoxicity against MM cell lines was modest, LY significantly enhanced the toxicity of bortezomib by down-regulating bortezomib-induced heat shock protein 27 phosphorylation. LY inhibited interleukin-6 secretion from long term cultured-BM stromal cells and BM mononuclear cells (BMMNCs) derived from MM patients in remission. LY also inhibited macrophage inflammatory protein-1alpha secretion from patient MM cells and BMMNCs as well as normal CD14 positive osteoclast precursor cells. Moreover, LY significantly inhibited in vitro osteoclastogenesis from CD14 positive cells induced by macrophage-colony stimulating factor and soluble receptor activator of nuclear factor-kappaB ligand. Finally, LY also inhibited in vivo osteoclatogenesis in a severe combined immunodeficiency mouse model of human MM. These results suggest that LY represents a promising novel targeted approach to improve MM patient outcome both by enhancing the effect of bortezomib and by reducing osteoskeletal events.


Sujet(s)
Acides boroniques/pharmacologie , Imidazoles/pharmacologie , Myélome multiple/anatomopathologie , Ostéoclastes/effets des médicaments et des substances chimiques , Pyrazines/pharmacologie , Pyridines/pharmacologie , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Animaux , Antinéoplasiques , Bortézomib , Mort cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Synergie des médicaments , Protéines du choc thermique/métabolisme , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Souris SCID , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Transplantation tumorale , Phosphorylation/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , p38 Mitogen-Activated Protein Kinases/physiologie
13.
Br J Haematol ; 138(6): 783-91, 2007 Sep.
Article de Anglais | MEDLINE | ID: mdl-17760810

RÉSUMÉ

Akt mediates growth and drug resistance in multiple myeloma (MM) cells in the bone marrow (BM) microenvironment. We have shown that a novel Akt inhibitor Perifosine induces significant cytotoxicity in MM cells in the BM milieu. This study further delineated molecular mechanisms whereby Perifosine triggered cytotoxicity in MM cells. Neither the intensity of Jun NH(2)-terminal kinase phosphorylation nor caspase/poly (ADP-ribose) polymerase cleavage correlated with Perifosine-induced cytotoxicity in MM.1S, INA6, OPM1 and OPM2 MM cells. However, survivin, which regulates caspase-3 activity, was markedly downregulated by Perifosine treatment, without changes in other anti-apoptotic proteins. Downregulation of survivin by siRNA significantly inhibited OPM1 MM cell growth, confirming that survivin mediates MM cell survival. Perifosine significantly downregulated both function and protein expression of beta-catenin. Co-culture with BM stromal cells (BMSCs) upregulated both beta-catenin and survivin expression in MM cells, which was blocked by Perifosine. Importantly, Perifosine treatment also downregulated survivin expression in human MM cells grown in vivo in a severe combined immunodeficient mouse xenograft model. Finally, Perifosine inhibited bortezomib-induced upregulation of survivin, associated with enhanced cytotoxicity of combined bortezomib and Perifosine treatment. These preclinical studies provide the framework for clinical trials of bortezomib with Perifosine to improve patient outcome in MM.


Sujet(s)
Régulation négative , Protéines associées aux microtubules/métabolisme , Myélome multiple/traitement médicamenteux , Protéines tumorales/métabolisme , Phosphoryl-choline/analogues et dérivés , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Acides boroniques/usage thérapeutique , Bortézomib , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Protéines IAP , Souris , Protéines associées aux microtubules/génétique , Modèles animaux , Myélome multiple/métabolisme , Protéines tumorales/génétique , Transplantation tumorale , Phosphoryl-choline/pharmacologie , Phosphoryl-choline/usage thérapeutique , Inhibiteurs de protéases/usage thérapeutique , Pyrazines/usage thérapeutique , Interférence par ARN , Petit ARN interférent/pharmacologie , Survivine , Transplantation hétérologue
14.
Blood ; 110(10): 3744-52, 2007 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-17715391

RÉSUMÉ

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.


Sujet(s)
Communication cellulaire/effets des médicaments et des substances chimiques , Myélome multiple/anatomopathologie , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/physiologie , Récepteurs CCR1/antagonistes et inhibiteurs , Adhérence cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Fusion cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Chimiokine CCL3/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Évaluation préclinique de médicament , Gènes fos , Humains , Myélome multiple/métabolisme , Ostéoclastes/métabolisme
15.
Mol Cancer Ther ; 6(6): 1718-27, 2007 Jun.
Article de Anglais | MEDLINE | ID: mdl-17575103

RÉSUMÉ

In this study, we investigated the cytotoxicity of 5-azacytidine, a DNA methyltransferase inhibitor, against multiple myeloma (MM) cells, and characterized DNA damage-related mechanisms of cell death. 5-Azacytidine showed significant cytotoxicity against both conventional therapy-sensitive and therapy-resistant MM cell lines, as well as multidrug-resistant patient-derived MM cells, with IC(50) of approximately 0.8-3 micromol/L. Conversely, 5-azacytidine was not cytotoxic to peripheral blood mononuclear cells or patient-derived bone marrow stromal cells (BMSC) at these doses. Importantly, 5-azacytidine overcame the survival and growth advantages conferred by exogenous interleukin-6 (IL-6), insulin-like growth factor-I (IGF-I), or by adherence of MM cells to BMSCs. 5-Azacytidine treatment induced DNA double-strand break (DSB) responses, as evidenced by H2AX, Chk2, and p53 phosphorylations, and apoptosis of MM cells. 5-Azacytidine-induced apoptosis was both caspase dependent and independent, with caspase 8 and caspase 9 cleavage; Mcl-1 cleavage; Bax, Puma, and Noxa up-regulation; as well as release of AIF and EndoG from the mitochondria. Finally, we show that 5-azacytidine-induced DNA DSB responses were mediated predominantly by ATR, and that doxorubicin, as well as bortezomib, synergistically enhanced 5-azacytidine-induced MM cell death. Taken together, these data provide the preclinical rationale for the clinical evaluation of 5-azacytidine, alone and in combination with doxorubicin and bortezomib, to improve patient outcome in MM.


Sujet(s)
Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Azacitidine/pharmacologie , Acides boroniques/pharmacologie , Altération de l'ADN , ADN tumoral/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Antienzymes/pharmacologie , Myélome multiple/anatomopathologie , Pyrazines/pharmacologie , Bortézomib , Division cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Humains
16.
Int J Biochem Cell Biol ; 39(1): 171-80, 2007.
Article de Anglais | MEDLINE | ID: mdl-16979371

RÉSUMÉ

Histones are key components of chromatin. We investigated histone H2A-immunoreactive proteins in acute monocytic leukemia THP-1 cells using three polyclonal antibodies raised against peptides corresponding to distinct regions of H2A. Two unknown immunoreactive proteins (9- and 12-kDa proteins), H2A (14kDa) and ubiquitinated H2A (23kDa) were found in the cell lysates prepared by immediate direct addition of SDS-PAGE sample buffer to the cells as well as in the nuclear and chromatin fractions. However, they were not found in the cytoplasmic fraction. The unknown proteins were successfully purified by immunoaffinity chromatography from the cell nucleus extract and identified as 9-kDa H2A(1-87) and 12-kDa H2A(1-114), suggesting that both were produced by limited proteolysis of intact H2A(1-129). The truncated forms of H2A probably persisted as chromatin constituents, since the stability of H2A(1-87) in the chromatin fraction was sensitive to treatment with micrococcal nuclease, and H2A(1-114) was solubilized with lower ionic strength from the chromatin fraction obtained by micrococcal nuclease treatment. Truncated H2A proteins in THP-1 cells were transiently increased in amount by short-term treatment with phorbol 12-myristate 13-acetate or all-trans-retinoic acid, both of which induce macrophage-like differentiation. Furthermore, these increases were suppressed by preceding treatment with carbobenzoxy-l-leucyl-l-leucyl-l-leucinal (MG132) but not with carbobenzoxy-l-isoleucyl-gamma-t-butyl-l-glutamyl-l-alanyl-l-leucinal (PSI), both of which are generally known as proteasome inhibitors. Our results suggest that histone H2A is cleaved at least at two sites by protease(s) that remain obscure, and might affect chromatins in the early stage of THP-1 cell differentiation.


Sujet(s)
Anticorps/composition chimique , Chromatine/composition chimique , Histone/composition chimique , Histone/isolement et purification , Monocytes/composition chimique , Ubiquitines/composition chimique , Ubiquitines/isolement et purification , Anticorps/immunologie , Antinéoplasiques/pharmacologie , Cancérogènes/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Chromatine/métabolisme , Histone/biosynthèse , Histone/immunologie , Humains , Leupeptines , Monocytes/métabolisme , Maturation post-traductionnelle des protéines/effets des médicaments et des substances chimiques , 12-Myristate-13-acétate de phorbol/pharmacologie , Trétinoïne/pharmacologie , Ubiquitines/biosynthèse , Ubiquitines/immunologie
17.
Br J Haematol ; 136(3): 414-23, 2007 Feb.
Article de Anglais | MEDLINE | ID: mdl-17173546

RÉSUMÉ

We have previously shown that heat shock protein (Hsp) 27 or its upstream activator p38 mitogen-activated protein kinase (MAPK) confers resistance to bortezomib and dexamethasone (Dex) in multiple myeloma (MM) cells. This study examined anti-MM activity of a novel p38 MAPK inhibitor, BIRB 796, alone and in combination with conventional and novel therapeutic agents. BIRB 796 blocked baseline and bortezomib-triggered upregulation of p38 MAPK and Hsp27 phosphorylation, thereby enhancing cytotoxicity and caspase activation. The Hsp90 inhibitor 17-allylamino-17-demethoxy-geldanamycin (17-AAG) upregulated protein expression and phosphorylation of Hsp27; conversely, BIRB 796 inhibited this phosphorylation and enhanced 17-AAG-induced cytotoxicity. Importantly, BIRB 796 inhibited Hsp27 phosphorylation induced by 17-AAG plus bortezomib, thereby enhancing cytotoxicity. In bone marrow stromal cells (BMSC), BIRB 796 inhibited phosphorylation of p38 MAPK and secretion of interleukin-6 (IL-6) and vascular endothelial growth factor triggered by either tumour necrosis factor-alpha or tumour growth factor-beta1. BIRB 796 also inhibited IL-6 secretion induced in BMSCs by adherence to MM cells, thereby inhibiting tumour cell proliferation. These studies therefore suggest that BIRB 796 overcomes drug-resistance in the BM microenvironment, providing the framework for clinical trials of a p38 MAPK inhibitor, alone and in combination with bortezomib, Hsp90 inhibitor, or Dex, to improve patient outcome in MM.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Myélome multiple/traitement médicamenteux , Naphtalènes/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyrazoles/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Antinéoplasiques hormonaux/usage thérapeutique , Apoptose/effets des médicaments et des substances chimiques , Benzoquinones/usage thérapeutique , Acides boroniques/usage thérapeutique , Bortézomib , Adhérence cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Techniques de coculture , Tests de cytotoxicité immunologique , Dexaméthasone/usage thérapeutique , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Humains , Immunotransfert/méthodes , Interleukine-6/métabolisme , Protéines et peptides de signalisation intracellulaire , Lactames macrocycliques/usage thérapeutique , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Naphtalènes/pharmacologie , Phosphorylation , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Pyrazines/usage thérapeutique , Pyrazoles/pharmacologie , Cellules souches , Facteur de croissance transformant bêta/pharmacologie , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , p38 Mitogen-Activated Protein Kinases/métabolisme
18.
Rinsho Ketsueki ; 48(12): 1559-62, 2007 Dec.
Article de Japonais | MEDLINE | ID: mdl-18203517

RÉSUMÉ

Tumor lysis syndrome (TLS) is a severe complication of chemotherapy brought about by the rapid destruction of tumor cells. TLS is usually diagnosed by elevation of intracellular enzymes and no specific abnormality is found in complete blood counts. We present a 22-year-old woman with acute lymphoblastic leukemia (ALL) complicated with TLS, in whom elevation of leukocytes and platelet count was observed due to fragmented leukocytes. The day after initiating chemotherapy, a rapid increase in intracellular enzymes was found and a diagnosis of TLS was made. Her leukocyte and platelet counts increased from 8,400/ml to 42,600/ml. and from 43,000/ml to 231,000/ml, respectively. Many fragmented leukocytes were found in her peripheral blood picture. The automated hematology analyzer counted these fragments as leukocytes or platelets, with resulting pseudo-leukocytosis and pseudo-thrombocytosis. When evaluating laboratory data of TLS, it is necessary to focus on the peripheral blood picture to avoid misunderstanding the blood cell counts.


Sujet(s)
Hyperleucocytose/étiologie , Thrombocytose/étiologie , Syndrome de lyse tumorale/sang , Adulte , Femelle , Humains , Hyperleucocytose/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Thrombocytose/diagnostic
19.
Rinsho Ketsueki ; 47(7): 650-5, 2006 Jul.
Article de Japonais | MEDLINE | ID: mdl-16910576

RÉSUMÉ

We report a case of follicular lymphoma in which pulmonary cryptococcosis occurred with cladribine therapy. The case involved a 72-year-old man. He was diagnosed as having follicular lymphoma, grade 1, clinical stage IVA from a tongue tumor biopsy in January 2003. A total of 6 courses of R-CHOP therapy was performed, but no clear effect was found. A new cervical lesion appeared, so he was treated with a total of 2 courses of R-EPOCH therapy, and the effect was classed as stable disease. We started cladribine therapy (0.09 mg/kg, seven days of continuous infusion) from February 2004, and complete remission was achieved after 4 courses of cladribine therapy. In January 2005, an abnormal nodular shadow in the right S10 area was found on chest CT images which was diagnosed as pulmonary cryptococcosis by serum antigen and a trans-bronchial lung biopsy. We started fluconazole (200 mg a day, initially intravenous drip infusion, followed by oral intake), following which both the pulmonary shadow and serum antigen improved. Afterward, the fifth course of cladribine therapy and local radiation therapy were performed against a relapse of lymphoma, but cryptococcosis did not reappear. The prolonged bone marrow suppression after cladribine therapy was considered to be a severe adverse event. These findings suggest that it is very important to pay attention to any opportunistic infection such as pulmonary cryptococcosis.


Sujet(s)
Antinéoplasiques/effets indésirables , Chloro-2 désoxyadénosine/effets indésirables , Cryptococcose/étiologie , Mycoses pulmonaires/étiologie , Lymphome folliculaire/traitement médicamenteux , Infections opportunistes , Antinéoplasiques/pharmacologie , Moelle osseuse/effets des médicaments et des substances chimiques , Chloro-2 désoxyadénosine/pharmacologie , Cryptococcose/imagerie diagnostique , Humains , Mycoses pulmonaires/imagerie diagnostique , Mâle , Adulte d'âge moyen , Tomodensitométrie
20.
Clin Lymphoma ; 6(1): 31-6, 2005 Jun.
Article de Anglais | MEDLINE | ID: mdl-15989704

RÉSUMÉ

PURPOSE: To investigate the feasibility of high-dose chemotherapy (HDCT) followed by autologous stem cell transplantation (ASCT) for aggressive non-Hodgkin's lymphoma (NHL), we conducted HDCT with ASCT using a drug-only protocol without total body irradiation. Previously untreated and treated adult patients with aggressive lymphoma were enrolled onto this study. For the HDCT protocol, we developed the AECC regimen, a drug-only regimen consisting of etoposide, carboplatin, cyclophosphamide, and nimustine (ie, ACNU). Mobilized peripheral blood stem cells were used mainly as a source for ASCT and were used based on collection rates of CD34 cells. PATIENTS AND METHODS: Fifty-six patients were enrolled and assessed for this study. The median length of follow-up was 6.5 years, with a range of 0.2-12.5 years. Retrospective immunophenotypic examination indicated that the majority of the patients were diagnosed with B-cell lymphoma. RESULTS: Before HDCT, 37 patients still had disease (26 partial responses [PRs] and 11 cases of no response), and 19 patients exhibited a complete response (CR) before HDCT with ASCT. Among 56 patients, 37 (66%) exhibited a CR, including patients continuing their first CR and those experiencing a second or further CR, and 11 patients (19.6%) exhibited PR on HDCT with ASCT. Outcomes of patients without CR were significantly poorer than those of the patients with CR, and 7-year overall survival rates of patients with and without CR were 63% and 27.2%, respectively. No patients developed a second malignancy, including leukemia or myelodysplastic syndrome. CONCLUSIONS: High-dose chemotherapy followed by ASCT is one of the available consolidation therapies for aggressive NHL, and additional involved-field irradiation could play a role in the management of patients with NHL who do not exhibit a CR after treatment with HDCT containing a drug-only program.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Association thérapeutique , Lymphome malin non hodgkinien/thérapie , Transplantation de cellules souches , Adulte , Cyclophosphamide/usage thérapeutique , Survie sans rechute , Doxorubicine/usage thérapeutique , Femelle , Humains , Lymphome malin non hodgkinien/traitement médicamenteux , Lymphome malin non hodgkinien/mortalité , Lymphome malin non hodgkinien/anatomopathologie , Mâle , Adulte d'âge moyen , Prednisone/usage thérapeutique , Analyse de survie , Transplantation autologue , Vincristine/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE