Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Leukemia ; 23(12): 2222-32, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19741729

RESUMEN

Although outcomes for patients with multiple myeloma (MM) have improved over the past decade, the disease remains incurable and even patients who respond well to induction therapy ultimately relapse and require additional treatment. Conventional chemotherapy and high-dose therapy with stem cell transplantation (SCT) have historically been utilized in the management of relapsed MM, but in recent years the immunomodulatory drugs (IMiDs) thalidomide and lenalidomide, as well as the proteasome inhibitor bortezomib, have assumed a primary role in this setting. This review focuses on the role of thalidomide, lenalidomide and bortezomib in relapsed and refractory MM, with additional discussion dedicated to emerging drugs in relapsed MM that may prove beneficial to patients with this disease.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Terapia Recuperativa/métodos , Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Bortezomib , Humanos , Inmunosupresores/uso terapéutico , Lenalidomida , Pirazinas/uso terapéutico , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Resultado del Tratamiento
3.
Bone Marrow Transplant ; 35(1): 77-83, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15502851

RESUMEN

The diagnosis and treatment of cancer is often associated with high levels of psychosocial distress, yet exploration of these issues is rarely included in routine oncologic care. We conducted a pilot study to evaluate the feasibility of screening for psychosocial distress after autologous and allogeneic stem cell transplantation. A total of 80 adults were enrolled in Boston, MA, USA. Subjects completed self-administered assessments prior to hospital admission, at their first clinic visit after hospital discharge, and at 100 days post transplant. Assessments included validated instruments assessing psychosocial distress and quality of life (QOL). Elevated levels of anxiety and/or depression were detected in 55% of those providing pre-transplant assessments and were associated with compromised QOL. Post transplant screening was successfully performed in 69% of subjects and identified that 44% had symptoms of depression, anxiety or post traumatic stress disorder. Pre-transplant distress was associated with detection of distress after transplantation (81 vs 13%, P< 0.0001). In summary, we detected high levels of distress in transplant patients using self-administered tools. Pre-transplant distress appears to be highly predictive of distress post transplant and is a feasible marker to target screening and intervention programs.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Estrés Psicológico/diagnóstico , Adaptación Psicológica , Adulto , Ansiedad , Depresión , Emociones , Femenino , Humanos , Acontecimientos que Cambian la Vida , Masculino , Tamizaje Masivo , Persona de Mediana Edad , Proyectos Piloto , Calidad de Vida , Rol del Enfermo , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/psicología , Trastornos por Estrés Postraumático , Encuestas y Cuestionarios , Factores de Tiempo
4.
Leukemia ; 15(12): 1950-61, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11753617

RESUMEN

Increased angiogenesis has recently been recognized in active multiple myeloma (MM). Since vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are two key mediators of angiogenesis, we characterized the production of VEGF, b-FGF and interleukin-6 (IL-6) (a MM growth and survival factor) in MM cell lines and Epstein-Barr virus (EBV) transformed B cell lines from MM patients, patient MM cells, as well as bone marrow stromal cells (BMSCs) from normal healthy donors and MM patients. We detected secretion of VEGF, but no bFGF and IL-6, in MM cell lines (MM.1S, RPMI 8226 and U266); EBV transformed B cell lines from MM patients (IM-9, HS-Sultan and ARH77); MM cell lines resistant to doxorubicin (RPMI-DOX40), mitoxantrone (RPMI-MR20), melphalan (RPMI-LR5) and dexamethasone (MM.1R); and patient MM cells (MM1 and MM2). BMSCs from MM patients and normal donors secreted VEGF, b-FGF and IL-6. Importantly, when MM cells were adhered to BMSCs, there was a significant increase in VEGF (1.5- to 3.1-fold) and IL-6 (1.9- to 56-fold) secretion. In contrast, the bFGF decreased in co-cultures of BMSCs and MM cells. Paraformaldehyde fixation of BMSCs or MM cells prior to adhesion revealed that VEGF was produced both from BMSCs and MM cells, though it may come primarily from BMSCs in some cultures. IL-6 was produced exclusively in BMSCs, rather than MM cells. Moreover, when MM cells were placed in Transwell insert chambers to allow their juxtaposition to BMSCs without cell to cell contact, induction of VEGF and IL-6 secretion persisted, suggesting the importance of humoral factors. Addition of exogenous IL-6 (10 ng/ml) increased VEGF secretion by BMSCs. Conversely, VEGF (100 ng/ml) significantly increased IL-6 secretion by BMSCs. Moreover, anti-human VEGF (1 microg/ml) and anti-human IL-6 (10 microg/ml) neutralizing antibodies reduced IL-6 and VEGF secretion, respectively, in cultures of BMSCs alone and co-cultures of BMSCs and MM cells. Finally, thalidomide (100 microM) and its immunomodulatory analog IMiD1-CC4047 (1 microM) decreased the upregulation of IL-6 and VEGF secretion in cultures of BMSCs, MM cells and co-cultures of BMSCs with MM cells. These data demonstrate the importance of stromal-MM cell interactions in regulating VEGF and IL-6 secretion, and suggest additional mechanisms whereby thalidomide and IMiD1-CC4047 act against MM cells in the BM millieu.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Mieloma Múltiple/patología , Células del Estroma/citología , Inhibidores de la Angiogénesis/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular , Comunicación Celular/fisiología , Técnicas de Cocultivo , Interacciones Farmacológicas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/fisiopatología , Células del Estroma/metabolismo , Células del Estroma/fisiología , Talidomida/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
Semin Oncol ; 28(6): 607-12, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11740818

RESUMEN

Novel therapies in multiple myeloma (MM) target not only the tumor cell but also the bone marrow (BM) microenvironment. Thalidomide (Thal), as well as derivative immunomodulatory drugs (IMiDs), directly induce apoptosis or G1 growth arrest in MM cell lines and patient's MM cells which are resistant to melphalan (Mel), doxorubicin (Dox), and dexamethasone (Dex). Although Thal and IMiDs do not alter adhesion of MM cells to bone marrow stromal cells (BMSCs), they inhibit the upregulation of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) secretion triggered by the binding of MM cells to BMSCs. Proteasome inhibitors represent another potential anticancer therapy targeting the MM cell and the BM microenvironment. The proteasome inhibitor PS-341 directly inhibits proliferation and induces apoptosis in both human MM cell lines and freshly isolated patient's MM cells which are resistant to Mel, Dox, and Dex. PS-341 inhibits p44/42 mitogen-activated protein kinase (MAPK) growth signaling triggered by IL-6 and induces apoptosis, despite induction of p21 and p27, in p53 wild-type and p53 mutant MM cells. PS-341 adds to the anti-MM activity of dexamethasone and overcomes IL-6-mediated protection against dexamethasone-induced apoptosis. PS-341 blocks the paracrine growth of human MM cells by decreasing their adherence to BMSCs and related NF-kappaB-dependent induction of IL-6 secretion in BMSCs. Moreover, proliferation and MAPK growth signaling of those residual adherent MM cells is also inhibited. Tumor necrosis factor-alpha (TNF-alpha), which is produced by some MM cells, induces only low-level MM proliferation and MAPK activation in MM cells, but markedly upregulates IL-6 secretion from BMSCs and upregulates expression of adhesion molecules (VLA-4 and LFA-1) on MM cells and their receptors (VCAM-1 and ICAM-1) on BMSCs, with resultant increased binding of MM cells to BMSCs. Inhibition of TNF-alpha-induced NF-kappaB activation with PS-341 inhibits both the upregulation of these molecules on MM cells and BMSCs and the resultant increased adhesion. Therefore, inhibiting TNF-alpha and its sequelae may be useful treatment strategies in MM. Our data show that VEGF causes proliferation and enhances migration of MM as well as plasma cell leukemia (PCL) cells. VEGF induced twofold activation of cell migration in MM cells and more than 100-fold activation of cell migration in PCL cells, suggesting an important role of VEGF in the progression of MM to PCL. These data indicate that VEGF plays a pivotal role not only in neoangiogenesis in MM BM but also in proliferation and migration of tumor cells.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Médula Ósea/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Talidomida/uso terapéutico , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Adyuvantes Inmunológicos/uso terapéutico , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Médula Ósea/irrigación sanguínea , Ciclo Celular/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Cisteína Endopeptidasas , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Humanos , Linfocinas/antagonistas & inhibidores , Complejos Multienzimáticos/antagonistas & inhibidores , Mieloma Múltiple/patología , Neovascularización Patológica , Inhibidores de Fosfodiesterasa/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Complejo de la Endopetidasa Proteasomal , Talidomida/análogos & derivados , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
6.
Blood ; 98(1): 210-6, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11418482

RESUMEN

The antiangiogenic activity of thalidomide (Thal), coupled with an increase in bone marrow angiogenesis in multiple myeloma (MM), provided the rationale for the use of Thal in MM. Previously, the direct anti-MM activity of Thal and its analogues (immunomodulatory drugs, IMiDs) on MM cells was demonstrated, suggesting multiple mechanisms of action. In this study, the potential immunomodulatory effects of Thal/IMiDs in MM were examined. It was demonstrated that Thal/IMiDs do not induce T-cell proliferation alone but act as costimulators to trigger proliferation of anti-CD3-stimulated T cells from patients with MM, accompanied by an increase in interferon-gamma and IL-2 secretion. However, an increase in autologous T-cell killing of patient MM cells could not be demonstrated. A role for natural killer (NK)- and LAK-cell-mediated killing is suggested because IL-2-primed peripheral blood mononuclear cells (PBMCs) treated with Thal/IMiDs demonstrated significantly increased lysis of MM cell lines. Cold target inhibition assays suggested NK- rather than LAK-cell-mediated killing. Furthermore, this killing was not major histocompatibility complex-class restricted, and the depletion of CD56(+) cells blocked the drug-induced MM cell lysis. It was significant that increased killing of patient MM cells by autologous PBMCs treated with Thal/IMiDs was also observed. Although the in vivo relevance of NK-cell-mediated MM cell killing is unknown, phenotypic analysis performed in MM patients receiving Thal therapy demonstrated an increase in CD3(-)CD56(+) cells in patients responding to therapy. Thus in vitro and in vivo data support the hypothesis that Thal may mediate its anti-MM effect, at least in part, by modulating NK cell number and function.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Células Asesinas Naturales/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Talidomida/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Estudios de Casos y Controles , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Recuento de Linfocitos , Mieloma Múltiple/sangre , Mieloma Múltiple/inmunología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Talidomida/administración & dosificación , Células Tumorales Cultivadas/efectos de los fármacos
7.
J Clin Oncol ; 19(4): 1152-9, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11181681

RESUMEN

PURPOSE: The role of donor marrow T-cell depletion (TCD) in preventing graft-versus-host disease (GVHD) after transplantation of unrelated allogeneic marrow remains undefined. Because different TCD methodologies differ in the degree and specificity with which T cells are removed, it is likely that transplant outcomes would depend on which technique is used. Herein, we report results in the first 48 recipients of unrelated marrow using CD6+ TCD as the sole form of GVHD prophylaxis. PATIENTS AND METHODS: Median age of patients was 46 years (20 to 58 years). Donors were matched at A/B HLA loci. Ablation consisted of cyclophosphamide and fractionated total-body irradiation (TBI; 14 Gy). To facilitate engraftment, patients also received 7.5 Gy (22 patients) [corrected] or 4.5 Gy (26 patients) [corrected] of total lymphoid irradiation (TLI) before admission. No additional immune suppressive prophylaxis was administered. Granulocyte colony-stimulating factor was administered daily from day +1 to engraftment. RESULTS: All 48 patients demonstrated neutrophil engraftment. An absolute neutrophil count of 500 x 10(6)/L was achieved at a median of 12 days (range, 9 to 23 days). There were no cases of late graft failure. The number of CD34+ cells infused/kg was associated with speed of platelet and neutrophil recovery. The dose of TLI did not influence engraftment. Grades 2-4 acute GVHD occurred in 42% of patients (95% confidence interval [CI], 0.28 to 0.57). Mortality at day 100 was 19%. There have been only five relapses. Estimated 2-year survival was 44% (95% CI, 0.28 to 0.59) for the entire group, 58% for patients less than 50 years of age. In multivariable analysis, age less than 50 years (P =.002), cytomegalovirus seronegative status (P =.04), and early disease status at bone marrow transplant (P =.05) were associated with superior survival. CONCLUSION: CD6+ TCD does not impede engraftment of unrelated bone marrow after low-dose TLI, cyclophosphamide, and TBI. CD6+ TCD as the sole form of GVHD prophylaxis results in an incidence of GVHD that compares favorably with many adult studies of unrelated transplantation using unmanipulated marrow and immune-suppressive medications, especially in light of the median age of our patients (46 years). Although event-free survival in patients less than 50 years of age is very encouraging, older patients experience frequent transplantation-related complications despite TCD.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Trasplante de Médula Ósea/efectos adversos , Enfermedad Injerto contra Huésped/prevención & control , Depleción Linfocítica/métodos , Linfocitos T/inmunología , Adulto , Terapia Combinada , Femenino , Humanos , Leucemia/terapia , Subgrupos Linfocitarios , Masculino , Persona de Mediana Edad
8.
J Clin Oncol ; 19(1): 242-52, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11134219

RESUMEN

PURPOSE: Although the number of autologous and allogeneic stem-cell transplantations (SCT) is increasing, relatively little information about recovery after transplantation is available. Quantitative information appropriate for patient counseling is difficult to discern from the literature. We sought to suggest reasonable expectations for recovery and symptoms after SCT for hematologic malignancies and other disorders using the following measures: (1) objective measures of health status, such as frequency of clinic visits, need for rehospitalization, medication usage, work status, and overall and event-free survival; (2) qualitative assessment of quality of life, such as returning to a normal life, resumption of normal activities, satisfaction with appearance, and whether recovery has occurred; and (3) quantification of specific bothersome symptoms. PATIENTS AND METHODS: Autologous and allogeneic SCT recipients at a tertiary-care transplant center participated in the prospective, longitudinal questionnaire study. RESULTS: Three hundred twenty patients were studied. Questionnaire response rates at 6, 12, and 24 months range from 85% to 88% among survivors. Although autologous patients had better event-free and overall survival, fewer symptoms, and more complete recovery at 6 months, these advantages had largely equalized by 12 months. Specific bothersome symptoms were reported by less than 24% of patients after transplantation, except for fatigue and financial and sexual difficulties, which were more prevalent. CONCLUSION: These findings may help counsel patients considering transplantation and educate them about reasonable expectations for recovery. Overall, the low level of bothersome symptoms and continued recovery through the first year after transplantation are encouraging.


Asunto(s)
Enfermedades Hematológicas/rehabilitación , Enfermedades Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Recuperación de la Función , Actividades Cotidianas , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Estado de Salud , Enfermedades Hematológicas/mortalidad , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Calidad de Vida , Estadísticas no Paramétricas , Tasa de Supervivencia , Trasplante Autólogo , Trasplante Homólogo , Resultado del Tratamiento , Estados Unidos/epidemiología
9.
Blood ; 96(9): 2943-50, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11049970

RESUMEN

Although thalidomide (Thal) was initially used to treat multiple myeloma (MM) because of its known antiangiogenic effects, the mechanism of its anti-MM activity is unclear. These studies demonstrate clinical activity of Thal against MM that is refractory to conventional therapy and delineate mechanisms of anti-tumor activity of Thal and its potent analogs (immunomodulatory drugs [IMiDs]). Importantly, these agents act directly, by inducing apoptosis or G1 growth arrest, in MM cell lines and in patient MM cells that are resistant to melphalan, doxorubicin, and dexamethasone (Dex). Moreover, Thal and the IMiDs enhance the anti-MM activity of Dex and, conversely, are inhibited by interleukin 6. As for Dex, apoptotic signaling triggered by Thal and the IMiDs is associated with activation of related adhesion focal tyrosine kinase. These studies establish the framework for the development and testing of Thal and the IMiDs in a new treatment paradigm to target both the tumor cell and the microenvironment, overcome classical drug resistance, and achieve improved outcome in this presently incurable disease.


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Inmunosupresores/toxicidad , Mieloma Múltiple/patología , Talidomida/toxicidad , Talidomida/uso terapéutico , Ciclo Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Dexametasona/toxicidad , Doxorrubicina/toxicidad , Femenino , Fase G1/efectos de los fármacos , Humanos , Masculino , Melfalán/toxicidad , Talidomida/análogos & derivados , Células Tumorales Cultivadas
10.
Semin Oncol ; 26(5 Suppl 14): 97-106, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10561024

RESUMEN

The use of serotherapy to treat patients with plasma cell dyscrasias (PCDs) has been sought by us and others. Candidate antigens that have been targeted or proposed for targeting in PCDs include the immunoglobulin idiotype, CD19, CD38, CD54, CD126, HM1.24, and Muc-1 core protein. Unfortunately, many of these antigens are not ideal for use in serotherapy since they are not selectively expressed, are either shed or secreted, or have not been fully characterized. Serotherapy with an anti-CD19 monoclonal antibody (B4) conjugated to a blocked ricin toxin had no significant activity in patients with multiple myeloma (MM). Circulating CD20+ clonotypic B cells have been detected in the circulation of most MM and Waldenstrom's macroglobulinemia (WM) patients. Plasma cells from most WM patients express CD20, but most MM patient plasma cells either lack CD20 or express it weakly. In view of recent successes with anti-CD20-directed serotherapy in other B-cell malignancies, we initiated a phase II trial to study the anti-CD20 monoclonal antibody rituximab (Rituxan; IDEC Pharmaceuticals, San Diego, CA, and Genentech, Inc, San Francisco, CA) in patients with MM. We describe two PCD patients (one with WM and one with MM) who responded to therapy. By flow cytometric analysis, CD20+ plasma cells and B cells present in the bone marrow and peripheral blood of a patient with MM disappeared with response to rituximab therapy. However, residual CD20- tumor cells remained in the bone marrow following rituximab therapy, and after 6 months this patient progressed with CD20- myeloma cells. As a potential strategy to overcome this limitation, we demonstrated that interferon-gamma at pharmacologically achievable levels induced CD20 expression on these CD20- plasma cells, consistent with our recent findings that interferon-gamma is a potent inducer of CD20 expression on MM patient plasma cells and B cells. We also characterize a response to rituximab with a decrease in paraprotein and resolution of anemia in a patient with WM whose response to rituximab is ongoing after 19+ months. This preliminary experience supports the potential use of serotherapy targeting CD20 in PCDs. Our studies further suggest that interferon-gamma may enhance CD20 expression on MM plasma cells, thereby increasing their susceptibility to anti-CD20 monoclonal antibody therapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Macroglobulinemia de Waldenström/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales de Origen Murino , Antígenos CD20/biosíntesis , Linfocitos B/inmunología , Ensayos Clínicos Fase II como Asunto , Citometría de Flujo , Humanos , Inmunización Pasiva , Interferón gamma/farmacología , Masculino , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Paraproteinemias/tratamiento farmacológico , Paraproteinemias/inmunología , Fenotipo , Rituximab , Macroglobulinemia de Waldenström/inmunología , Macroglobulinemia de Waldenström/patología
11.
Curr Opin Oncol ; 11(2): 102-8, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10188074

RESUMEN

Experience from around the world now suggests that high response rates can be achieved in patients with multiple myeloma treated with high-dose therapy followed by hematopoietic stem cell transplantation. However, patients are destined to relapse and few, if any, are cured. Major obstacles to cure are the excessive toxicity noted after allografting in multiple myeloma, contaminating tumor cells in multiple myeloma autografts, and most importantly, the persistence of minimal residual disease after high-dose therapy and either allogeneic or autologous stem cell transplantation. In this context, strategies are being developed to decrease the toxicity of allografting and to enhance allogeneic anti-multiple myeloma immunity after transplantation to improve outcome. To improve autografting, better ablative regimens, more efficacious purging of tumor cells from autografts, and enhancement of autologous anti-multiple myeloma immunity post-transplantation are modalities being tested to enhance overall and event-free survival.


Asunto(s)
Trasplante de Médula Ósea , Mieloma Múltiple/terapia , Humanos , Trasplante Autólogo/fisiología , Trasplante Homólogo/fisiología
12.
Bone Marrow Transplant ; 21(12): 1177-81, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9674848

RESUMEN

For patients with non-Hodgkin's lymphoma (NHL) undergoing blood or bone marrow transplantation (BMT), the use of autologous grafts has often been preferred to that of allogeneic stem cells because of a significantly lower incidence of non-relapse mortality. If complications associated with allo-BMT could be minimized without compromising efficacy, then it might become a preferred strategy for certain subsets of patients. In this report, we describe the toxicity and long-term efficacy of T cell-depleted allogeneic BMT using anti-CD6 monoclonal antibody and complement alone to reduce the risk of GVHD and its sequelae. Twenty-two patients, aged 18-60 years, with high (n = 10), intermediate (n = 9), or low (n = 3) grade NHL underwent HLA-identical allogeneic BMT from siblings. Patients had either relapsed after at least one remission or never achieved a full remission with chemotherapy. Twenty patients had a history of marrow involvement. Bone marrow was depleted of CD6+ T cells with T12 monoclonal antibody and complement as the sole form of GVHD prophylaxis. Stable hematopoietic engraftment occurred in all 22 patients. Four patients developed grade 2 and 1 patient grade 3 GVHD (23% grades 2-4 GVHD). Chronic GVHD has occurred in three patients. Treatment-related mortality was very low. Only one patient died while in remission. Thirteen patients are alive and free of disease with a median follow-up of 30 months. Estimated event-free and overall survivals are 54 and 59%, respectively. CD6 allogeneic marrow transplantation is associated with a low risk of transplant-related complications and may offer advantages for certain patients with recurrent NHL felt to be at high risk for relapse after autologous transplantation.


Asunto(s)
Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Trasplante de Médula Ósea , Depleción Linfocítica , Linfoma no Hodgkin/terapia , Adolescente , Adulto , Trasplante de Médula Ósea/efectos adversos , Femenino , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Linfoma no Hodgkin/mortalidad , Masculino , Persona de Mediana Edad , Recurrencia , Trasplante Homólogo
13.
Blood ; 90(5): 1982-92, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9292533

RESUMEN

The murine double minute 2 (MDM2) protein facilitates G1 to S phase transition by activation of E2F-1 and can enhance cell survival by suppressing wild-type p53 (wtp53) function. In this study, we examined MDM2 expression and function in multiple myeloma (MM) cells. MDM2 is strongly and constitutively expressed in MM cell lines (ARH-77, RPMI 8226, and OCI-My5) and in the cells of plasma cell leukemia (PCL) patients, but is not expressed in normal bone marrow mononuclear cells (BM MNCs). Treatment of MM cells with MDM2 antisense, but not sense, nonsense, or scrambled, oligodeoxyribonucleotides (ODNs) decreased DNA synthesis and cell viability; it also induced G1 growth arrest, as evidenced by propidium iodide (PI) staining and induction of retinoblastoma protein (pRB) to E2F-1 binding. Moreover, inhibition of MDM2 using antisense ODNs also triggered MM cell apoptosis as evidenced by acridine orange-ethidium bromide staining. We next studied the association of MDM2 with wtp53 and/or mutant p53 (mtp53), E2F-1, CDK4, and p21. MDM2 constitutively binds to E2F-1 in all MM cells, to both wtp53 and mtp53, and to p21 in tumor cells lacking p53. These data suggest that MDM2 may enhance cell-cycle progression in MM cells both by activating E2F-1 and by downregulating cell-cycle inhibitory proteins (wtp53 and p21). Overexpression of MDM2 may therefore contribute to both growth and survival of MM cells, suggesting the potential utility of treatment strategies targeting MDM2 in MM.


Asunto(s)
Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Proteínas Proto-Oncogénicas/metabolismo , Apoptosis , División Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Neoplasias/genética , Oligonucleótidos Antisentido , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Células Tumorales Cultivadas
14.
J Immunol ; 159(5): 2212-21, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9278309

RESUMEN

IL-6 mediates growth of some human multiple myeloma (MM) cells and IL-6-dependent cell lines. Although three IL-6 signaling pathways (STAT1, STAT3, and Ras-dependent MAPK cascade) have been reported, cascades mediating IL-6-triggered growth of MM cells and cell lines are not defined. In this study, we therefore characterized IL-6 signaling cascades in MM cell lines, MM patient cells, and IL-6-dependent B9 cells to determine which pathway mediates IL-6-dependent growth. IL-6 induced phosphorylation of JAK kinases and gp130, regardless of the proliferative response of MM cells to this growth factor. Accordingly, we next examined downstream IL-6 signaling via the STAT3, STAT1, and Ras-dependent mitogen-activated protein kinase (MAPK) cascades. IL-6 triggered phosphorylation of STAT1 and/or STAT3 in MM cells independent of their proliferative response to IL-6. In contrast, IL-6 induced phosphorylation of Shc and its association with Sos1, as well as phosphorylation of MAPK, only in MM cells and B9 cells that proliferated in response to IL-6. Moreover, MAPK antisense, but not sense, oligonucleotide inhibited IL-6-induced proliferation of these cells. These data suggest that STAT1 and/or STAT3 activation may occur independently of the proliferative response to IL-6, and that activation of the MAPK cascade is an important distal pathway for IL-6-mediated growth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Interleucina-6/farmacología , Proteínas Quinasas Activadas por Mitógenos , Mieloma Múltiple/patología , Proteínas de Neoplasias/fisiología , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas , Transducción de Señal/efectos de los fármacos , Proteínas ras/fisiología , Antígenos CD/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , División Celular/efectos de los fármacos , Receptor gp130 de Citocinas , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Janus Quinasa 1 , Janus Quinasa 2 , Glicoproteínas de Membrana/metabolismo , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Oligonucleótidos Antisentido/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Represoras/metabolismo , Proteína SOS1 , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal/fisiología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , TYK2 Quinasa , Transactivadores/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos
15.
Blood ; 90(1): 279-89, 1997 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-9207463

RESUMEN

Interleukin-6 (IL-6) is a growth factor for multiple myeloma (MM) cells and can inhibit MM cell apoptosis. Our recent studies show that IL-6 facilitates MM cell growth via phosphorylation of retinoblastoma protein (pRB); however, the effects of IL-6 on those cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors (CDIs) that are known to regulate phosphorylation of pRB have not been defined in MM cells. In the present report, we cultured MM cell lines and patient cells with IL-6 and/or dexamethasone (Dex) and characterized changes in cell cycle; expression and association of cyclins, CDKs, and CDIs; and phosphorylation of pRB. Dex induced G1 growth arrest in MM cells, whereas IL-6 facilitated G1 to S phase transition; moreover, the effect of Dex was blocked by IL-6. p21WAF1 (p21) protein was constitutively expressed in the majority of MM cells independent of the status of p53. Its expression was upregulated by Dex and downregulated by IL-6; again, IL-6 inhibited the increase in p21 triggered by Dex. These alterations in p21 expression in MM cells were associated with changes in p21 binding to CDK2, CDK4, and CDK6; CDK2, CDK4, and CDK6 kinase activities; and phosphorylation of pRB. In contrast, expression of G1 cell cycle regulatory proteins, including p27KIP1, cyclin D2, and cyclin E, was not altered in MM cells cultured with Dex and/or IL-6. Finally, interferon-gamma (IFN-gamma) also induced G1 growth arrest and upregulated p21 protein expression; as with Dex, affects of IFN-gamma were inhibited by IL-6. Our results therefore show that changes in cell cycle distribution in MM cells triggered by Dex, IL-6, and IFN-gamma correlate with changes in p21 protein expression and implicate p21 in the coupling of Dex-, IL-6-, and IFN-gamma-related signals to G1 cell cycle regulation in MM cells.


Asunto(s)
Antineoplásicos/farmacología , Ciclinas/biosíntesis , Dexametasona/farmacología , Fase G1/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-6/farmacología , Mieloma Múltiple , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Interacciones Farmacológicas , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Tumorales Cultivadas , Regulación hacia Arriba
16.
Blood ; 89(8): 3039-47, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9108425

RESUMEN

The appropriate timing of bone marrow transplantation (BMT) for adults with acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) is controversial. Although allogeneic transplantation results in a lower risk of disease recurrence than intensive chemotherapy alone, overall outcome following BMT may not be improved due to the higher incidence of therapy-related fatal complications, frequently as a result of the development of graft-versus-host disease (GVHD). Selective T-cell depletion of donor marrow can reduce the incidence of GVHD and thereby limit transplant-related toxicity. Herein we report the risk of GVHD, incidence of transplant related mortality (TRM), likelihood of disease relapse, and overall survival in adult patients undergoing BMT with CD6 depleted allogeneic marrow for acute leukemia in first remission. Forty-one consecutive allogeneic transplants were performed on patients with acute leukemia and high-risk features (28 AML, 13 ALL) using T12 monoclonal antibody and complement to remove CD6+ T cells from donor marrow. No pre- or posttransplant immune suppressive medications for GVHD prophylaxis were administered. The actuarial estimated risk of grade 2 to 4 acute GVHD was 15% in patients receiving HLA identical grafts. Chronic GVHD developed in five patients. The estimated risk of TRM for patients in first complete remission was 5% at Day +100 and 16% at 2 years. Fatalities attributable to infection with cytomegalovirus or Epstein-Barr virus occurred in only three patients. Estimated probabilities of relapse, overall survival, and event-free survival at 4 years were 25%, 71%, and 63%, respectively. No significant differences in GVHD, TRM, relapse rate, or survival was observed for patients with AML compared with those with ALL. Allogeneic transplantation with CD6 depleted bone marrow is effective in consolidating remissions of high-risk patients with acute leukemia in first remission without excessive toxicity.


Asunto(s)
Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Trasplante de Médula Ósea , Enfermedad Injerto contra Huésped/prevención & control , Leucemia/terapia , Depleción Linfocítica , Subgrupos de Linfocitos T , Enfermedad Aguda , Adulto , Antibióticos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Purgación de la Médula Ósea , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/mortalidad , Terapia Combinada , Citarabina/administración & dosificación , Supervivencia sin Enfermedad , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/epidemiología , Humanos , Infecciones/etiología , Infecciones/mortalidad , Leucemia/tratamiento farmacológico , Leucemia/inmunología , Leucemia/mortalidad , Tablas de Vida , Masculino , Persona de Mediana Edad , Neoplasias Primarias Secundarias/terapia , Inducción de Remisión , Riesgo , Análisis de Supervivencia , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/inmunología , Trasplante Homólogo/mortalidad , Resultado del Tratamiento
18.
Blood ; 88(6): 2219-27, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8822942

RESUMEN

Interleukin-6 (IL-6) mediates autocrine and paracrine growth of multiple myeloma (MM) cells and inhibits tumor cell apoptosis. Abnormalities of retinoblastoma protein (pRB) and mutations of RB gene have been reported in up to 70% of MM patients and 80% of MM-derived cell lines. Because dephosphorylated (activated) pRB blocks transition from G1 to S phase of the cell cycle whereas phosphorylated (inactivated) pRB releases this growth arrest, we characterized the role of pRB in IL-6-mediated MM cell growth. Both phosphorylated and dephosphorylated pRB were expressed in all serum-starved MM patient cells and MM-derived cell lines, but pRB was predominantly in its phosphorylated form. In MM cells that proliferated in response to IL-6, exogenous IL-6 downregulated dephosphorylated pRB and decreased dephosphorylated pRB-E2F complexes. Importantly, culture of MM cells with RB antisense, but not RB sense, oligonucleotide (ODN) triggered IL-6 secretion and proliferation in MM cells; however, proliferation was only partially inhibited by neutralizing anti-IL-6 monoclonal antibody (MoAb). In contrast to MM cells, normal splenic B cells express dephosphorylated pRB. Although CD40 ligand (CD40L) triggers a shift from dephosphorylated to phosphorylated pRB and proliferation of B cells, the addition of exogenous IL-6 to CD40L-treated B cells does not alter either pRB or proliferation, as observed in MM cells. These results suggest that phosphorylated pRB is constitutively expressed in MM cells and that IL-6 further shifts pRB from its dephosphorylated to its phosphorylated form, thereby promoting MM cell growth via two mechanisms; by decreasing the amount of E2F bound by dephosphorylated pRB due to reduced dephosphorylated pRB, thereby releasing growth arrest; and by upregulating IL-6 secretion by MM cells and related IL-6-mediated autocrine tumor cell growth.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Interleucina-6/farmacología , Mieloma Múltiple/metabolismo , Proteína de Retinoblastoma/metabolismo , Linfocitos B/metabolismo , División Celular/efectos de los fármacos , ADN/biosíntesis , Factores de Transcripción E2F , Sustancias de Crecimiento/farmacología , Humanos , Mieloma Múltiple/patología , Oligodesoxirribonucleótidos/metabolismo , Oligonucleótidos Antisentido/metabolismo , Fosforilación , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas
19.
Blood ; 87(5): 1928-38, 1996 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8634441

RESUMEN

Interleukin-6 (IL-6), a product of bone marrow stromal cells (BMSCs), is a growth factor for multiple myeloma (MM) cells. Transforming growth factor-beta1 (TGF-beta1) is also produced by BMSCs and can regulate IL-6 secretion by several tissues, including BMSCs. The present study was designed to characterize in vitro tumor growth regulation by TGF-beta1 in MM. Sorted CD38+CD45RA- MM cells secreted significantly more TGF-beta1 (8.2 +/- 2.0 ng/mL) than peripheral blood mononuclear cells (P < .001), splenic B cells (P < .001), and CD40 ligand (CD40L) pretreated B cells (P < .05). TGF-beta1 secretion by MM-BMMCs (3.8 +/- 0.9 ng/mL) was significantly greater than by N-BMMCs (1.2 +/- 0.1 ng/mL, P < .001). MM-BMSCs also secreted significantly more TGF-beta1 (6.6 +/- 2.5 ng/mL, n = 11) than N-BMSCs (4.4 +/- 0.6 ng/mL, P < .02, n = 10) and N-BMSC lines (3.9 +/- 0.2 ng/mL, P < .02, n = 6). TGF-beta1 secretion was correlated with IL-6 secretion in MM-BMSCs. Anti-TGF-beta1 monoclonal antibody both blocked IL-6 secretion by BMSCs and inhibited the increments in IL-6 secretion by BMSCs induced by MM cell adhesion. Moreover, exogenous TGF-beta1 upregulated IL-6 secretion by MM-BMSCs, normal BMSCs, and CD38+ CD45RA- MM cells, as well as tumor cell proliferation. This is in contrast to the inhibitory effect of TGF-beta1 on proliferation and Ig secretion of normal splenic B cells. Finally, retinoblastoma proteins (pRB) are constitutively phosphorylated in MM cells; TGF-beta1 either did not alter or increased pRB phosphorylation. pRB are dephosphorylated in splenic B cells and phosphorylated in CD40L triggered B cells in contrast to its effects on MM cells, TGF-beta1 decreased phosphorylation of pRB in CD40L treated B cells. These results suggest that TGF-beta1 is produced in MM by both tumor cells and BMSCs, with related tumore cell growth. Moreover, MM cell growth may be enhanced by resistance of tumor cells to the inhibitory effects of TGF-beta1 on normal B-cell proliferation and Ig secretion.


Asunto(s)
Linfocitos B/efectos de los fármacos , Interleucina-6/metabolismo , Mieloma Múltiple/patología , Células Madre Neoplásicas/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Linfocitos B/metabolismo , Médula Ósea/patología , Ligando de CD40 , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Humanos , Interleucina-10/farmacología , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/farmacología , Células Madre Neoplásicas/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
20.
Stem Cells ; 13(6): 588-96, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8590860

RESUMEN

Platelet transfusions have long had an important role in the treatment of patients with thrombocytopenia due to disease or myelotoxic treatment or in patients with reduced platelet function. However, platelet transfusions are associated with numerous risks, both immunologic (e.g., transfusion reactions, alloimmunization, immunosuppression) and infectious (e.g., viral, bacterial). In addition, several laboratory and clinical factors can influence post-transfusion platelet recovery. Recent technological advances have introduced the potential for using alternatives to platelet transfusions, such as cytokines or platelet substitutes, which may avoid the risks of transfusion. Platelet development from megakaryocytes is a process that is highly regulated by cytokines and animal research suggests that selected cytokines involved in this process may be useful in the treatment of thrombocytopenia. Newer developments, including the utilization of recombinant cytokines with relatively selective stimulation of platelet production (e.g., interleukin 6 [IL-6]) and the recent discovery of a megakaryocyte colony stimulating factor (thrombopoietin), represent major therapeutic opportunities in the treatment of thrombocytopenia. Platelet substitutes, e.g., thromboerythrocytes, also show promise in the management of platelet deficiencies.


Asunto(s)
Transfusión Sanguínea , Neoplasias/terapia , Transfusión de Plaquetas , Trombocitopenia/terapia , Humanos , Neoplasias/complicaciones , Recuento de Plaquetas , Factores de Riesgo , Trombocitopenia/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...