Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Acta Haematol ; 147(1): 8-21, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37717577

RESUMEN

BACKGROUND: B-cell receptor (BCR) signaling is crucial for normal B-cell development and adaptive immunity. In chronic lymphocytic leukemia (CLL), the malignant B cells display many features of normal mature B lymphocytes, including the expression of functional B-cell receptors (BCRs). Cross talk between CLL cells and the microenvironment in secondary lymphatic organs results in BCR signaling and BCR-driven proliferation of the CLL cells. This critical pathomechanism can be targeted by blocking BCR-related kinases (BTK, PI3K, spleen tyrosine kinase) using small-molecule inhibitors. Among these targets, Bruton tyrosine kinase (BTK) inhibitors have the highest therapeutic efficacy; they effectively block leukemia cell proliferation and generally induce durable remissions in CLL patients, even in patients with high-risk disease. By disrupting tissue homing receptor (i.e., chemokine receptor and adhesion molecule) signaling, these kinase inhibitors also mobilize CLL cells from the lymphatic tissues into the peripheral blood (PB), causing a transient redistribution lymphocytosis, thereby depriving CLL cells from nurturing factors within the tissue niches. SUMMARY: The clinical success of the BTK inhibitors in CLL underscores the central importance of the BCR in CLL pathogenesis. Here, we review CLL pathogenesis with a focus on the role of the BCR and other microenvironment cues. KEY MESSAGES: (i) CLL cells rely on signals from their microenvironment for proliferation and survival. (ii) These signals are mediated by the BCR as well as chemokine and integrin receptors and their respective ligands. (iii) Targeting the CLL/microenvironment interaction with small-molecule inhibitors provides a highly effective treatment strategy, even in high-risk patients.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Transducción de Señal , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/uso terapéutico , Proliferación Celular , Biología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Microambiente Tumoral
2.
Blood ; 129(9): 1155-1165, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28031181

RESUMEN

Targeting B-cell receptor (BCR) signaling is a successful therapeutic strategy in mature B-cell malignancies. Precursor BCR (pre-BCR) signaling, which is critical during normal B lymphopoiesis, also plays an important role in pre-BCR+ B cell acute lymphoblastic leukemia (B-ALL). Here, we investigated the activity and mechanism of action of the BTK inhibitor ibrutinib in preclinical models of B-ALL. Pre-BCR+ ALL cells were exquisitely sensitive to ibrutinib at therapeutically relevant drug concentrations. In pre-BCR+ ALL, ibrutinib thwarted autonomous and induced pre-BCR signaling, resulting in deactivation of PI3K/Akt signaling. Ibrutinib modulated the expression of pre-BCR regulators (PTPN6, CD22, CD72, and PKCß) and substantially reduced BCL6 levels. Ibrutinib inhibited ALL cell migration toward CXCL12 and beneath marrow stromal cells and reduced CD44 expression. CRISPR-Cas9 gene editing revealed that both BTK and B lymphocyte kinase (BLK) are relevant targets of ibrutinib in pre-BCR+ ALL. Consequently, in mouse xenograft models of pre-BCR+ ALL, ibrutinib treatment significantly prolonged survival. Combination treatment of ibrutinib with dexamethasone or vincristine demonstrated synergistic activity against pre-BCR+ ALL. These data corroborate ibrutinib as a promising targeted agent for pre-BCR+ ALL and highlight the importance of ibrutinib effects on alternative kinase targets.


Asunto(s)
Antineoplásicos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Piperidinas , Leucemia-Linfoma Linfoblástico de Células Precursoras B/enzimología , Proteínas Tirosina Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Br J Haematol ; 174(3): 425-36, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27071778

RESUMEN

B cell acute lymphoblastic leukaemia (B-ALL) cells express high levels of CXCR4 chemokine receptors for homing and retention within the marrow microenvironment. Bone marrow stromal cells (BMSC) secrete CXCL12, the ligand for CXCR4, and protect B-ALL cells from cytotoxic drugs. Therefore, the therapeutic use of CXCR4 antagonists has been proposed to disrupt cross talk between B-ALL cells and the protective stroma. Because CXCR4 antagonists can have activating agonistic function, we compared the genetic and pharmacological deletion of CXCR4 in B-ALL cells, using CRISPR-Cas9 gene editing and CXCR4 antagonists that are in clinical use (plerixafor, BKT140). Both genetic and pharmacological CXCR4 inhibition significantly reduced B-ALL cell migration to CXCL12 gradients and beneath BMSC, and restored drug sensitivity to dexamethasone, vincristine and cyclophosphamide. NOD/SCID/IL-2rγnull mice injected with CXCR4 gene-deleted B-ALL cells had significant delay in disease progression and superior survival when compared to control mice injected with CXCR4 wild-type B-ALL cells. These findings indicate that anti-leukaemia activity of CXCR4 antagonists is primarily due to CXCR4 inhibition, rather than agonistic activity, and corroborate that CXCR4 is an important target to overcome stroma-mediated drug resistance in B-ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores CXCR4/antagonistas & inhibidores , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Edición Génica , Humanos , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Ratones , Ratones Endogámicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptor Cross-Talk/efectos de los fármacos , Receptores CXCR4/genética , Células del Estroma/metabolismo , Células Tumorales Cultivadas
4.
Clin Adv Hematol Oncol ; 14(1): 55-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27057669

RESUMEN

B-cell receptor (BCR) signaling has emerged as a key pathway for the expansion of neoplastic B-cell clones in several B-cell malignancies. The mechanisms that activate BCR signaling differ substantially among subtypes of B-cell lymphoma and leukemia. These include BCR stimulation by foreign or self-antigens, or the acquisition of mutations in components of the BCR pathway that result in autonomous or enhanced antigen-induced BCR signaling. Targeting BCR signaling with selective inhibitors of the BCR-associated kinases Bruton's tyrosine kinase, spleen tyrosine kinase, and phosphoinositide 3-kinase delta induces high response rates in patients with chronic lymphocytic leukemia, mantle cell lymphoma, Waldenström macroglobulinemia, and diffuse large B-cell lymphoma of the activated B-cell-like subtype and is currently transforming the therapeutic landscape in these diseases. Here we review the mechanisms of BCR activation that govern growth and survival of malignant B cells. We also summarize recent clinical trials of BCR inhibitors, with a focus on the most clinically advanced agents.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/metabolismo , Linfoma de Células B/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Linfoma de Células B/química , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de Antígenos de Linfocitos B/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal/efectos de los fármacos
5.
Clin Cancer Res ; 23(9): 2313-2324, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27697994

RESUMEN

Purpose: B-cell receptor (BCR)-associated kinase inhibitors, such as ibrutinib, have revolutionized the treatment of chronic lymphocytic leukemia (CLL). However, these agents are not curative, and resistance is already emerging in a proportion of patients. IL4, expressed in CLL lymph nodes, can augment BCR signaling and reduce the effectiveness of BCR kinase inhibitors. Therefore, simultaneous targeting of the IL4- and BCR signaling pathways by cerdulatinib, a novel dual Syk/JAK inhibitor currently in clinical trials (NCT01994382), may improve treatment responses in patients.Experimental Design: PBMCs from patients with CLL were treated in vitro with cerdulatinib alone or in combination with venetoclax. Cell death, chemokine, and cell signaling assay were performed and analyzed by flow cytometry, immunoblotting, q-PCR, and ELISA as indicated.Results: At concentrations achievable in patients, cerdulatinib inhibited BCR- and IL4-induced downstream signaling in CLL cells using multiple readouts and prevented anti-IgM- and nurse-like cell (NLC)-mediated CCL3/CCL4 production. Cerdulatinib induced apoptosis of CLL cells, in a time- and concentration-dependent manner, and particularly in IGHV-unmutated samples with greater BCR signaling capacity and response to IL4, or samples expressing higher levels of sIgM, CD49d+, or ZAP70+ Cerdulatinib overcame anti-IgM, IL4/CD40L, or NLC-mediated protection by preventing upregulation of MCL-1 and BCL-XL; however, BCL-2 expression was unaffected. Furthermore, in samples treated with IL4/CD40L, cerdulatinib synergized with venetoclax in vitro to induce greater apoptosis than either drug alone.Conclusions: Cerdulatinib is a promising therapeutic for the treatment of CLL either alone or in combination with venetoclax, with the potential to target critical survival pathways in this currently incurable disease. Clin Cancer Res; 23(9); 2313-24. ©2016 AACR.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Pirimidinas/administración & dosificación , Receptores de Antígenos de Linfocitos B/efectos de los fármacos , Sulfonas/administración & dosificación , Adenina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de las Cinasas Janus/administración & dosificación , Quinasas Janus/antagonistas & inhibidores , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteínas de Neoplasias/genética , Piperidinas , Proteínas Proto-Oncogénicas c-bcr/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcr/genética , Pirazoles/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Quinasa Syk/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos
6.
J Invest Dermatol ; 131(2): 495-503, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21048785

RESUMEN

The phosphatidyl inositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway has been shown to be involved in the development of melanoma. PI-103 is a kinase inhibitor blocking PI3K class IA and mTOR complex 1 and 2. Here, we studied the effect of targeting the PI3K/mTORC1/mTORC2 pathway by PI-103 and rapamycin in melanoma cells and in a melanoma mouse model. Dual targeting of PI3K and mTOR by PI-103 induced apoptosis and cell-cycle arrest, and inhibited viability of melanoma cells in vitro. Combined treatment with PI-103 and the prototypic mTORC1 inhibitor rapamycin led to the synergistic suppression of AKT and ribosomal S6 protein phosphorylation and to the induction of apoptosis. In vivo, PI-103 and rapamycin displayed only modest single-agent activity, but the combination significantly reduced the tumor growth compared with both single agents. These data show that blocking the PI3K/mTORC1/mTORC2 pathway using the combination of two distinct small-molecule inhibitors ("vertical inhibition") leads to superior efficacy against malignant melanoma in vitro and in vivo.


Asunto(s)
Melanoma/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas/antagonistas & inhibidores , Transducción de Señal/fisiología , Neoplasias Cutáneas/patología , Transactivadores/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Furanos/farmacología , Humanos , Técnicas In Vitro , Diana Mecanicista del Complejo 1 de la Rapamicina , Melanoma/metabolismo , Melanoma/fisiopatología , Ratones , Ratones Desnudos , Complejos Multiproteicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/fisiopatología , Serina-Treonina Quinasas TOR , Transactivadores/metabolismo , Factores de Transcripción , Trasplante Heterólogo
7.
Cancer Lett ; 296(2): 249-56, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20471160

RESUMEN

VEGF receptor blockage has been reported to increase serum VEGF. We hypothesized that mTOR inhibition by everolimus counteracts VEGF induction by sunitinib resulting in an improved anti-tumor activity of sunitinib. In vitro, sunitinib in combination with everolimus did not outperform the respective monotherapies. In vivo, monotherapies reduced tumor growth by 60%, whereas the combination of sunitinib and everolimus led to an almost complete tumor growth inhibition. This superior anti-tumor activity coincided with attenuation of VEGF peaks. In conclusion mTOR inhibition by everolimus counteracts VEGF induction by sunitinib and results in significant reduction of tumor burden and long-lasting tumor growth control.


Asunto(s)
Antineoplásicos/farmacología , Inmunosupresores/uso terapéutico , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Pirroles/farmacología , Sirolimus/análogos & derivados , Neoplasias Gástricas/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Everolimus , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Sirolimus/uso terapéutico , Neoplasias Gástricas/patología , Sunitinib , Serina-Treonina Quinasas TOR , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
8.
Cancer Biol Ther ; 9(11): 919-27, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20404549

RESUMEN

The vascular endothelial growth factor (VEGF) is a central mediator of tumor-induced angiogenesis. Everolimus, a mammalian target of rapamycin (mTOR) inhibitor, decreases VEGF-secretion of cancer cells. Vatalanib is a selective inhibitor of VEGF receptors 1-3. In the present study it was hypothesized that dual inhibition of VEGF signaling by inhibition of VEGF production and VEGF receptor signaling leads to synergistic anti-tumor effects. In vitro, effects of vatalanib and everolimus on cell proliferation, cell cycle, apoptosis and signal transduction were examined in three gastric cancer cell lines. Effects on angiogenesis were assessed using tube formation assays of cultured human umbilical vein endothelial cells (HUVECs). In vivo, the antitumor effect of compounds was studied using a gastric cancer xenograft nude mouse model. VEGF of murine origin (mVEGF) and human cancer cell-derived VEGF (hVEGF) were studied separately by specific ELISAs. Tumor vascularization and proliferation were quantified by immunohistochemistry. In vitro, everolimus but not vatalanib decreased gastric cancer proliferation without inducing apoptosis. Vatalanib abolished endothelial cell tube formation, whereas inhibition of tube formation by everolimus was incomplete. In vivo, the combination of vatalanib with everolimus was superior to single agent treatments and reduced tumor size by about 50% relative to everolimus monotherapy (p < 0.005). Pharmacodynamic analysis of VEGF plasma level showed a decrease of hVEGF by everolimus and indicated a trend towards lower mVEGF level only in the combination group. In line, there was a tendency for lower vascular density and proliferation for combination treatment. We conclude that in a preclinical model of gastric cancer the antitumor activity of vatalanib can be augmented by everolimus.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ftalazinas/farmacología , Piridinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Ensayo de Inmunoadsorción Enzimática , Everolimus , Humanos , Inmunosupresores/farmacología , Ratones , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/prevención & control , Ftalazinas/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Sirolimus/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA