Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Anesth Analg ; 126(6): 1910-1913, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29570154

RESUMEN

The impact of volatile anesthetics on cancer progression has been observed for decades, but sex differences have not been described. Male and female immune systems vary considerably, and the immune system plays an important role in limiting cancer growth. Currently, mouse models describing the impact of volatile anesthetics on cancer growth are limited to same-sex models. In this brief report, we describe a sex-specific impact of isoflurane on melanoma growth observed in wild-type but not in immune-deficient mice. Future experimental designs related to anesthesia and cancer should evaluate the biological variable of sex in a systematic manner.


Asunto(s)
Anestésicos por Inhalación/efectos adversos , Inmunidad Celular/inmunología , Isoflurano/efectos adversos , Melanoma/inducido químicamente , Melanoma/inmunología , Caracteres Sexuales , Animales , Femenino , Inmunidad Celular/efectos de los fármacos , Masculino , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Blood ; 115(5): 985-94, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19965664

RESUMEN

Rituximab (RTX), a monoclonal antibody directed against the CD20 protein, is a drug commonly used in the treatment of B-cell-derived lymphoid neoplasias and of antibody-mediated autoimmune diseases. In addition to cell- and complement-mediated B-cell depletion, RTX is thought to inhibit B-cell survival and proliferation through negative regulation of canonical signaling pathways involving Akt, ERK, and mammalian target of rapamycin. However, surprisingly, although B-cell receptor (BCR) signaling has been considered critical for normal and more recently, for neoplastic B cells, the hypothesis that RTX could target BCR has never been investigated. Using follicular lymphoma cell lines as models, as well as normal B cells, we show here, for the first time, that pretreatment with RTX results in a time-dependent inhibition of the BCR-signaling cascade involving Lyn, Syk, PLC gamma 2, Akt, and ERK, and calcium mobilization. The inhibitory effect of RTX correlates with decrease of raft-associated cholesterol, complete inhibition of BCR relocalization into lipid raft microdomains, and down-regulation of BCR immunoglobulin expression. Thus, RTX-mediated alteration of BCR expression, dynamics, and signaling might contribute to the immunosuppressive activity of the drug.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Colesterol/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/efectos de los fármacos , Anticuerpos Monoclonales de Origen Murino , Western Blotting , Calcio/metabolismo , Línea Celular Tumoral , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Fosfolipasa C gamma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Rituximab , Quinasa Syk , Factores de Tiempo , Familia-src Quinasas/metabolismo
3.
Blood ; 113(20): 4875-84, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19278954

RESUMEN

In human blood, 1% to 5% of lymphocytes are gammadelta T cells; they mostly express the gammadelta T-cell receptor (TCR)Vgamma9, recognize nonpeptide phosphoantigens (PAgs) produced by microbes and tumor cells, and mediate different modes of lytic activities directed against tumor target cells. Antibody-dependent cell-mediated cytotoxicity (ADCC) mediated by cytolytic lymphoid cells is essential for the clinical activity of anticancer monoclonal antibodies (mAbs), but whether PAgs affect ADCC by gammadelta T cells is unknown. Here we report that, in association with the CD20(+)-specific mAb rituximab (RTX), the synthetic PAg bromohydrin pyrophosphate (BrHPP) increased TCRVgamma9(+) cell binding to CD20(+) lymphoma cells in vitro. This combination activated phospho-ZAP70 and phospho-ERK1/2 signaling in TCRVgamma9(+) cells and strongly enhanced their ADCC activity. We obtained similar results with BrHPP in the context of the mAbs alemtuzumab and trastuzumab. Furthermore, BrHPP enhanced RTX-mediated depletion of CD20(+) cells in vitro from peripheral blood mononuclear cells of healthy subjects and enhanced ADCC by gammadelta T cells from patients with chronic lymphocytic leukemia. In cynomolgus macaques, a regimen combining RTX, BrHPP, and IL2 activated TCRVgamma9(+) lymphocytes and enhanced B-cell depletion from blood and lymph nodes. Thus, the combination with BrHPP PAg is able to improve the efficacy of cancer immunotherapy by therapeutic mAbs.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Difosfatos/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Adulto , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales de Origen Murino , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Difosfatos/administración & dosificación , Difosfatos/inmunología , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Interleucina-2/administración & dosificación , Leucemia Linfocítica Crónica de Células B/sangre , Leucemia Linfocítica Crónica de Células B/inmunología , Macaca fascicularis , Masculino , Fosfatos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Rituximab , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
4.
Nat Commun ; 12(1): 565, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495474

RESUMEN

Accumulating evidence indicates that obesity with its associated metabolic dysregulation, including hyperinsulinemia and aberrant circadian rhythms, increases the risk for a variety of cancers including postmenopausal breast cancer. Caloric restriction can ameliorate the harmful metabolic effects of obesity and inhibit cancer progression but is difficult to implement and maintain outside of the clinic. In this study, we aim to test a time-restricted feeding (TRF) approach on mouse models of obesity-driven postmenopausal breast cancer. We show that TRF abrogates the obesity-enhanced mammary tumor growth in two orthotopic models in the absence of calorie restriction or weight loss. TRF also reduces breast cancer metastasis to the lung. Furthermore, TRF delays tumor initiation in a transgenic model of mammary tumorigenesis prior to the onset of obesity. Notably, TRF increases whole-body insulin sensitivity, reduces hyperinsulinemia, restores diurnal gene expression rhythms in the tumor, and attenuates tumor growth and insulin signaling. Importantly, inhibition of insulin secretion with diazoxide mimics TRF whereas artificial elevation of insulin through insulin pumps implantation reverses the effect of TRF, suggesting that TRF acts through modulating hyperinsulinemia. Our data suggest that TRF is likely to be effective in breast cancer prevention and therapy.


Asunto(s)
Neoplasias de la Mama/prevención & control , Modelos Animales de Enfermedad , Ayuno , Hiperinsulinismo/prevención & control , Obesidad/prevención & control , Posmenopausia/fisiología , Animales , Neoplasias de la Mama/sangre , Neoplasias de la Mama/fisiopatología , Restricción Calórica/métodos , Línea Celular Tumoral , Dieta Alta en Grasa , Femenino , Humanos , Hiperinsulinismo/sangre , Hiperinsulinismo/fisiopatología , Resistencia a la Insulina/fisiología , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Obesidad/fisiopatología , Ovariectomía , Posmenopausia/sangre
5.
Oncoimmunology ; 8(1): e1404212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30546937

RESUMEN

The cancer stem cell (CSC) paradigm posits that specific cells within a tumor, so-called CSC-like cells, have differing levels of tumorigenicity and chemoresistance. Original studies of CSCs identified them in human cancers and utilized mouse xenograft models to define the cancer initiating properties of these cells, thereby hampering the understanding of how immunity could affect CSCs. Indeed, few studies have characterized CSCs in the context of cancer immunoediting, and it is currently not clear how immunity could impact on the levels or stem-like behavior of CSCs. Using the well-studied 3'methylcholanthrene (MCA) model of primary sarcoma formation, we have defined a CSC-like population within MCA-induced sarcomas as expressing high levels of stem cell antigen-1 (Sca-1) and low levels of CD90. These Sca-1+CD90- CSC-like cells had higher tumor initiating ability, could spontaneously give rise to Sca-1-negative cells, and formed more sarcospheres than corresponding non-CSC-like cells. Moreover, when examining MCA-induced sarcomas that were in the equilibrium phase of cancer growth, higher levels of CSC-like cells were found compared to MCA-induced sarcomas in the escape phase of cancer progression. Notably, CSC-like cells also emerged during escape from anti-PD-1 or anti-CTLA4 therapy, thus suggesting that CSC-like cells could evade immune therapy. Finally, we demonstrate that paradoxically, interferon (IFN)-γ produced in vivo by immune cells could promote the emergence of CSC-like cells. Our findings define the existence of a Sca1+CD90- CSC-like population in the MCA-sarcoma model capable of differentiation, tumorsphere formation, and increased tumor initiation in vivo. These cells may also act as mediators of immune resistance during cancer immunoediting and immune therapy.

6.
Oncoimmunology ; 6(2): e1268310, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28344881

RESUMEN

Evidence of cancer immunosurveillance and immunoediting processes has been primarily demonstrated in mouse models of chemically induced oncogenesis. Although these models are very tractable, they are characterized by high mutational loads that represent a minority of human cancers. In this study, we sought to determine whether cancer immunosurveillance and immunoediting could be demonstrated in a more clinically relevant oncogene-induced model of carcinogenesis, the MMTV-PyMT (PyMT) mammary carcinoma model. This model system in the FVB/NJ strain background was previously used to demonstrate that adaptive immunity had no role in limiting primary cancer formation and in fact promoted metastasis, thus calling into question whether cancer immunosurveillance operated in preventing the development of breast cancer. Our current study in the C57BL/6 strain backgrounds provides a different conclusion, as we report here the existence of an adaptive immunosurveillance of PyMT mammary carcinomas using two independent models of immune deficiency. PyMT mice bred onto a Rag1-/- background or immune suppressed by chronic tacrolimus therapy both demonstrated accelerated development of mammary carcinomas. By generating a bank of cell lines from these animals, we further show that a subset of PyMT cell lines had delayed growth after transplantation into wild-type (WT) syngeneic, but not immune-deficient hosts. This reduced growth rate in immunocompetent animals was characterized by an increase in immune cell infiltration and tissue differentiation. Furthermore, loss of the immune cell infiltration that characterized immunoediting of slow growing cell lines, changed them into fast growing variants capable of progressing in the immunocompetent model. In conclusion, our study provides evidence that immunosurveillance and immunoediting of PyMT-derived cell lines modulate tumor progression in this oncogene-induced model of cancer.

7.
Cancer Res ; 77(8): 1997-2007, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381547

RESUMEN

In pancreatic ductal adenocarcinoma (PDAC), mutant KRAS stimulates the translation initiation factor eIF5A and upregulates the focal adhesion kinase PEAK1, which transmits integrin and growth factor signals mediated by the tumor microenvironment. Although eIF5A-PEAK1 signaling contributes to multiple aggressive cancer cell phenotypes, the downstream signaling processes that mediate these responses are uncharacterized. Through proteomics and informatic analyses of PEAK1-depleted PDAC cells, we defined protein translation, cytoskeleton organization, and cell-cycle regulatory pathways as major pathways controlled by PEAK1. Biochemical and functional studies revealed that the transcription factors YAP1 and TAZ are key targets of eIF5A-PEAK1 signaling. YAP1/TAZ coimmunoprecipitated with PEAK1. Interfering with eIF5A-PEAK1 signaling in PDAC cells inhibited YAP/TAZ protein expression, decreasing expression of stem cell-associated transcription factors (STF) including Oct4, Nanog, c-Myc, and TEAD, thereby decreasing three-dimensional (3D) tumor sphere growth. Conversely, amplified eIF5A-PEAK1 signaling increased YAP1/TAZ expression, increasing expression of STF and enhancing 3D tumor sphere growth. Informatic interrogation of mRNA sequence databases revealed upregulation of the eIF5A-PEAK1-YAP1-TEAD signaling module in PDAC patients. Taken together, our findings indicate that eIF5A-PEAK1-YAP signaling contributes to PDAC development by regulating an STF program associated with increased tumorigenicity. Cancer Res; 77(8); 1997-2007. ©2017 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Ciclo Celular/fisiología , Línea Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Iniciación de Péptidos/biosíntesis , Factores de Iniciación de Péptidos/genética , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Factor 5A Eucariótico de Iniciación de Traducción
8.
Cancer Immunol Res ; 4(9): 799-811, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27485135

RESUMEN

Despite clinical potential and recent advances, durable immunotherapeutic ablation of solid tumors is not routinely achieved. IL15 expands natural killer cell (NK), natural killer T cell (NKT) and CD8(+) T-cell numbers and engages the cytotoxic program, and thus is under evaluation for potentiation of cancer immunotherapy. We found that short-term therapy with IL15 bound to soluble IL15 receptor α-Fc (IL15cx; a form of IL15 with increased half-life and activity) was ineffective in the treatment of autochthonous PyMT murine mammary tumors, despite abundant CD8(+) T-cell infiltration. Probing of this poor responsiveness revealed that IL15cx only weakly activated intratumoral CD8(+) T cells, even though cells in the lung and spleen were activated and dramatically expanded. Tumor-infiltrating CD8(+) T cells exhibited cell-extrinsic and cell-intrinsic resistance to IL15. Our data showed that in the case of persistent viral or tumor antigen, single-agent systemic IL15cx treatment primarily expanded antigen-irrelevant or extratumoral CD8(+) T cells. We identified exhaustion, tissue-resident memory, and tumor-specific molecules expressed in tumor-infiltrating CD8(+) T cells, which may allow therapeutic targeting or programming of specific subsets to evade loss of function and cytokine resistance, and, in turn, increase the efficacy of IL2/15 adjuvant cytokine therapy. Cancer Immunol Res; 4(9); 799-811. ©2016 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Interleucina-15/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Citocinas/metabolismo , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Memoria Inmunológica , Interleucina-15/farmacología , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones , Ratones Transgénicos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Carga Tumoral
9.
Cell Rep ; 16(9): 2348-58, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27545889

RESUMEN

Cells undergoing xenobiotic or oxidative stress activate the transcription factor nuclear factor erythroid-derived 2-like 2 (Nrf2), which initiates an intrinsic "stress surveillance" pathway. We recently found that the cytokine IL-17D effects a form of extrinsic stress surveillance by inducing antitumor immunity, but how IL-17D is regulated remains unknown. Here, we show that Nrf2 induced IL-17D in cancer cell lines. Moreover, both Nrf2 and IL-17D were induced in primary tumors as well as during viral infection in vivo. Expression of IL-17D in tumors and virally infected cells is essential for optimal protection of the host as il17d(-/-) mice experienced a higher incidence of tumors and exacerbated viral infections compared to wild-type (WT) animals. Moreover, activating Nrf2 to induce IL-17D in established tumors led to natural killer cell-dependent tumor regression. These data demonstrate that Nrf2 can initiate both intrinsic and extrinsic stress surveillance pathways and highlight the use of Nrf2 agonists as immune therapies for cancer and infection.


Asunto(s)
Vigilancia Inmunológica , Interleucina-17/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Sarcoma/inmunología , Neoplasias de los Tejidos Blandos/inmunología , Animales , Carcinógenos , Línea Celular Tumoral , Chlorocebus aethiops , Regulación de la Expresión Génica , Humanos , Interleucina-17/genética , Metilcolantreno , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Muromegalovirus/crecimiento & desarrollo , Muromegalovirus/inmunología , Factor 2 Relacionado con NF-E2/genética , Sarcoma/inducido químicamente , Sarcoma/genética , Sarcoma/patología , Transducción de Señal , Neoplasias de los Tejidos Blandos/inducido químicamente , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/patología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Células Vero
10.
Oncoimmunology ; 3(12): e954853, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25964859

RESUMEN

Antitumor immunity suppresses tumorigenesis, but we do not understand how transformed cells initiate those immune responses that are essential for effective tumor immunosurveillance. Using the 3-MCA fibrosarcoma model, we identified IL-17D as a tumor-expressed cytokine that recruits natural killer cells, leading to the polarization of M1 macrophages and tumor rejection.

11.
Cell Rep ; 7(4): 989-98, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24794441

RESUMEN

The process of cancer immunoediting generates a repertoire of cancer cells that can persist in immune-competent hosts. In its most complex form, this process begins with the elimination of highly immunogenic unedited tumor cells followed by the escape of less immunogenic edited cells. Although edited tumors can release immunosuppressive factors, it is unknown whether unedited tumors produce cytokines that enhance antitumor function. Utilizing gene microarray analysis, we found the cytokine interleukin 17D (IL-17D) was highly expressed in certain unedited tumors but not in edited mouse tumor cell lines. Moreover, forced expression of IL-17D in edited tumor cells induced rejection by stimulating MCP-1 production from tumor endothelial cells, leading to the recruitment of natural killer (NK) cells. NK cells promoted M1 macrophage development and adaptive immune responses. IL-17D expression was also decreased in certain high-grade and metastatic human tumors, suggesting that it can be targeted for tumor immune therapy.


Asunto(s)
Interleucina-17/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/metabolismo , Humanos , Interleucina-17/biosíntesis , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Sarcoma/inmunología
12.
Cancer J ; 19(6): 483-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24270347

RESUMEN

Cancer immunosurveillance eradicates certain neoplasms, but the selective pressure exerted by this active surveillance leads to the emergence of immune evasive tumor clones in a process called cancer immunoediting. Natural killer (NK) cells are potent effectors of cancer immunoediting and can destroy tumors directly via exocytosis of cytotoxic granules or indirectly by producing interferon γ to activate M1 and TH1 immune responses. This review gathers current knowledge of NK immunosurveillance of primary tumors induced in mice and highlights the importance of NK immunosurveillance for human cancers. Evidence of NK immunoediting, as revealed by studies using NK-deficient models, demonstrates how exposure to NK cells engenders modification of cancer immunogenicity to permit survival and progression of the tumor clone in an immunocompetent environment.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Animales , Progresión de la Enfermedad , Humanos , Vigilancia Inmunológica/inmunología , Células Asesinas Naturales/patología , Ratones , Neoplasias/patología
13.
Leuk Res ; 37(4): 440-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23259986

RESUMEN

Rituximab (RTX, anti-CD20 antibody) combined with chemotherapy is currently standard treatment for chronic lymphocytic leukemia (CLL). Serum level of IL-8 is a prognostic factor for CLL that correlates with disease stage. We investigated whether endogenous IL-8 affects RTX or Obinutuzumab (GA101) B-leukemic depletion mediated by natural killers (NK). Using whole peripheral blood lymphocytes from untreated CLL patients, RTX, but most significantly GA101, were effective in B-cell depletion and NK activation. IL-8 inhibition completely inhibited B-cell depletion by RTX and reduced GA101-induced B-cell depletion. Altogether results underline IL-8 as an endogenous NK co-activator and confirm GA101 therapeutic potential for CLL treatment.


Asunto(s)
Antígenos CD20/inmunología , Linfocitos B/inmunología , Interleucina-8/fisiología , Células Asesinas Naturales/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Depleción Linfocítica , Receptores de IgG/agonistas , Anticuerpos Monoclonales/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos
14.
Cancers (Basel) ; 3(2): 1566-79, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24212774

RESUMEN

The concept of cancer stem cells has revolutionized our current vision of cancer development and was validated in solid tumors and cancers of the primitive hematopoietic compartment. Proof of the principle is still lacking, however, in malignancies of differentiated B-cells. We review here the current literature, which nevertheless suggests hierarchical organizations of the tumor clone for mostly incurable B-cell cancers such as multiple myeloma, lymphomas and B-chronic lymphocytic leukemia. We propose two models accounting for cancer stem cells in these contexts: a "top-to-bottom" clonal hierarchy from memory B-cells and a "bottom-to-top" model of clonal reprogramming. Selection pressure on the growing tumor can drive such reprogramming and increase its genetic diversity.

15.
Curr Drug Targets ; 11(7): 790-800, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20370648

RESUMEN

The development of therapeutic monoclonal antibodies (mAbs) has revolutionized the treatment of cancer along the last ten years. The best examples of their therapeutic efficacies have been obtained with rituximab for the treatment of CD20+ B-cell Non-Hodgkin Lymphoma (B-NHL), and several others antibodies with optimized bioactivities are now being developed for the treatment of various malignant hemopathies. We review here the main drugs developed in this field, and present some emerging concepts able to improve the bioactivities of the next generation of therapeutic mAbs.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hematológicas/tratamiento farmacológico , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Humanos , Modelos Biológicos , Modelos Moleculares , Proteínas Recombinantes de Fusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA