Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Blood Marrow Transplant ; 25(3): e76-e85, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30576834

RESUMEN

On August 30, 2017 the US Food and Drug Administration approved tisagenlecleucel (Kymriah; Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present, ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring, and disease assessments. To address these issues experts representing the American Society for Blood and Marrow Transplant, the European Society for Blood and Marrow Transplantation, the International Society of Cell and Gene Therapy, and the Foundation for the Accreditation of Cellular Therapy formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.


Asunto(s)
Testimonio de Experto , Inmunoterapia Adoptiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfocitos T/uso terapéutico , Antígenos CD19/inmunología , Niño , Vías Clínicas , Aprobación de Drogas , Humanos , Pautas de la Práctica en Medicina , Sociedades Médicas , Estados Unidos , Adulto Joven
2.
Biol Blood Marrow Transplant ; 20(3): 387-95, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24355521

RESUMEN

The number of survivors after allogeneic hematopoietic stem cell transplantation (HSCT) continues to increase, yet their survivorship experience has not been fully characterized. This study examines the health status and health-related quality of life (HRQL) of HSCT survivors. The aims of the study were to: (1) explore the baseline and change over time in these health outcomes, and (2) characterize subgroups experiencing adverse outcomes. In this longitudinal study, adults who survived >3 years from date of allogeneic HSCT completed a series of patient-reported outcome measures annually, including measures of health status, HRQL, and symptoms. Data were analyzed using hierarchical linear modeling. Subjects (N = 171) were on average 44 (±13.5) years of age and primarily male (62.6%); 40% were Hispanic. Mean scores for physical and mental health and HRQL were preserved relative to population norms. Hierarchical linear modeling revealed no significant change in the mean trajectories of these outcomes, although significant between-individual variability was observed. When controlling for demographic and clinical factors, physical symptom distress negatively affected all outcomes. The impact of symptom distress on physical health varied based on time since HSCT; impairment in physical health was greatest in survivors experiencing high symptom distress and who were within the first decade post transplantation. Extended treatment with systemic immunosuppressive therapy also predicted inferior physical health. These findings suggest that patient-centered outcomes are preserved relative to normative values and are generally stable after allogeneic HSCT, although survivors with persistent symptoms and those receiving systemic immunosuppression experience impairments in health status and HRQL.


Asunto(s)
Neoplasias Hematológicas/psicología , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Calidad de Vida/psicología , Estrés Psicológico/fisiopatología , Sobrevivientes/psicología , Adulto , Femenino , Estado de Salud , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Humanos , Inmunosupresores/efectos adversos , Estudios Longitudinales , Masculino , Salud Mental , Persona de Mediana Edad , Evaluación del Resultado de la Atención al Paciente , Trasplante Homólogo
3.
J Clin Pharmacol ; 61(2): 204-210, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32827162

RESUMEN

Eflapegrastim (Rolontis) is a long-acting granulocyte colony-stimulating factor (G-CSF) produced by conjugating a human G-CSF analogue and a human immunoglobulin G4 Fc fragment, linked via a polyethylene glycol linker. Weight-based doses of 45 to 270 µg/kg eflapegrastim (12.3-73.6 µg/kg as G-CSF) were evaluated in a phase 2 study in patients. Based on these results, a fixed dose of 13.2 mg eflapegrastim (3.6 mg G-CSF) was compared with pegfilgrastim (6 mg G-CSF) in 2 phase 3 studies and in a pharmacokinetic single-arm multicenter study. Absolute neutrophil count (ANC) data from these 3 studies were evaluated in patients with early-stage breast cancer who were treated with docetaxel and cyclophosphamide (n = 669). Serum concentrations of eflapegrastim were determined by enzyme-linked immunosorbent assay. Eflapegrastim systemic exposures were higher in cycle 1 than in cycle 3, likely attributable to the higher ANC in cycle 3, increasing neutrophil-mediated clearance. Eflapegrastim elicited a greater effect on ANC than pegfilgrastim in patients at ∼60% of the G-CSF dose. Body weight had no clinically significant effect on response, justifying administration of a fixed dose of eflapegrastim. The results from 2 phase 3 studies demonstrate that eflapegrastim at a fixed dose of 13.2 mg (3.6 mg G-CSF) administered once per chemotherapy cycle is effective in prophylactic treatment of chemotherapy-induced neutropenia.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Fármacos Hematológicos/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Neutropenia/inducido químicamente , Neutropenia/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Docetaxel/efectos adversos , Docetaxel/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Filgrastim/administración & dosificación , Semivida , Humanos , Tasa de Depuración Metabólica , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Polietilenglicoles/administración & dosificación
4.
Blood Rev ; 43: 100669, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32089398

RESUMEN

Acute and chronic graft-versus-host disease (GvHD) are the most important causes of treatment-related morbidity and mortality after allogeneic hematopoietic cell transplants for various diseases. Corticosteroids are an effective therapy in only about one-half of affected individuals and new therapy options are needed. We discuss novel strategies to treat GvHD using cellular-therapy including adoptive transfer of regulatory T-cells (Tregs), mesenchymal stromal cells (MSCs), cells derived from placental tissues, invariant natural killer T-cells (iNKTs), and myeloid-derived suppressor cells (MDSCs).These strategies may be more selective than drugs in modulating GvHD pathophysiology, and may be safer and more effective than conventional pharmacologic therapies. Additionally, these therapies have not been observed to substantially compromise the graft-versus-tumor effect associated with allotransplants. Many of these strategies are effective in animal models but substantial data in humans are lacking.


Asunto(s)
Enfermedad Injerto contra Huésped/terapia , Traslado Adoptivo/métodos , Animales , Enfermedad Injerto contra Huésped/fisiopatología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Supresoras de Origen Mieloide/trasplante , Células T Asesinas Naturales/trasplante , Linfocitos T Reguladores/trasplante
5.
Exp Hematol ; 92: 51-61, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33002567

RESUMEN

Eflapegrastim (Rolontis) is a long-acting granulocyte colony-stimulating factor (G-CSF) with an IgG4 Fc fragment and short polyethylene glycol linker. Current G-CSF products are administered 24 hours after chemotherapy. The present study compares the duration of neutropenia (DN) with eflapegrastim or pegfilgrastim at 0, 2, 5, or 24 hours post chemotherapy. Eflapegrastim was evaluated by G-CSF receptor binding and bone marrow cell proliferation assays in vitro. Eflapegrastim-Fc component binding to Fcγ receptors C1q and FcRn was assessed by enzyme-linked immunosorbent assay. Neutropenia was induced in rats via intraperitoneal cyclophosphamide or docetaxel/cyclophosphamide. Rats received chemotherapy followed by vehicle, pegfilgrastim, or eflapegrastim at 2, 5, or 24 hours. The difference in DN after treatment was assessed. In vitro binding to G-CSF receptor of both agents was similar. Binding to FcRn and no binding to Fcγ receptors or C1q were observed with eflapegrastim. Studies in chemotherapy-induced neutropenic rats revealed shorter DN with eflapegrastim versus pegfilgrastim. Increased levels of G-CSF in serum and marrow were observed in groups treated with eflapegrastim versus those treated with pegfilgrastim. Although eflapegrastim and pegfilgrastim have similar in vitro binding affinity, the Fc fragment in eflapegrastim increases the uptake into bone marrow, resulting in increased therapeutic potential for chemotherapy-induced neutropenia. Eflapegrastim's greater marrow resident time provided a pharmacodynamic advantage over pegfilgrastim, translating into shortened duration of neutropenia. Our findings support eflapegrastim same-day administration with chemotherapy, warranting further evaluation in patients undergoing myelosuppressive chemotherapy.


Asunto(s)
Filgrastim , Neutropenia/sangre , Neutropenia/tratamiento farmacológico , Polietilenglicoles , Animales , Ciclofosfamida/efectos adversos , Ciclofosfamida/farmacología , Docetaxel/efectos adversos , Docetaxel/farmacología , Filgrastim/farmacocinética , Filgrastim/farmacología , Antígenos de Histocompatibilidad Clase I/sangre , Humanos , Masculino , Ratones , Neutropenia/inducido químicamente , Neutropenia/patología , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Fc/sangre , Células U937
6.
Sci Transl Med ; 11(505)2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31413142

RESUMEN

Human interleukin-12 (hIL-12) is a cytokine with anticancer activity, but its systemic application is limited by toxic inflammatory responses. We assessed the safety and biological effects of an hIL-12 gene, transcriptionally regulated by an oral activator. A multicenter phase 1 dose-escalation trial (NCT02026271) treated 31 patients undergoing resection of recurrent high-grade glioma. Resection cavity walls were injected (day 0) with a fixed dose of the hIL-12 vector (Ad-RTS-hIL-12). The oral activator for hIL-12, veledimex (VDX), was administered preoperatively (assaying blood-brain barrier penetration) and postoperatively (measuring hIL-12 transcriptional regulation). Cohorts received 10 to 40 mg of VDX before and after Ad-RTS-hIL-12. Dose-related increases in VDX, IL-12, and interferon-γ (IFN-γ) were observed in peripheral blood, with about 40% VDX tumor penetration. Frequency and severity of adverse events, including cytokine release syndrome, correlated with VDX dose, reversing promptly upon discontinuation. VDX (20 mg) had superior drug compliance and 12.7 months median overall survival (mOS) at mean follow-up of 13.1 months. Concurrent corticosteroids negatively affected survival: In patients cumulatively receiving >20 mg versus ≤20 mg of dexamethasone (days 0 to 14), mOS was 6.4 and 16.7 months, respectively, in all patients and 6.4 and 17.8 months, respectively, in the 20-mg VDX cohort. Re-resection in five of five patients with suspected recurrence after Ad-RTS-hIL-12 revealed mostly pseudoprogression with increased tumor-infiltrating lymphocytes producing IFN-γ and programmed cell death protein 1 (PD-1). These inflammatory infiltrates support an immunological antitumor effect of hIL-12. This phase 1 trial showed acceptable tolerability of regulated hIL-12 with encouraging preliminary results.


Asunto(s)
Terapia Genética/métodos , Glioma/terapia , Interleucina-12/sangre , Corticoesteroides/farmacología , Adulto , Anciano , Dexametasona/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glioma/sangre , Glioma/tratamiento farmacológico , Glioma/mortalidad , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Interferón gamma/sangre , Interleucina-12/genética , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/terapia
7.
Bone Marrow Transplant ; 54(11): 1868-1880, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31092900

RESUMEN

On August 30, 2017, the U.S. Food and Drug Administration (US-FDA) approved tisagenlecleucel (KYMRIAH, Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor-T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring and disease assessments. To address these issues, experts representing the American Society for Blood and Marrow Transplant (ASBMT), the European Group for Blood and Marrow Transplantation (EBMT), the International Society of Cell and Gene Therapy (ISCT), and the Foundation for the Accreditation of Cellular Therapy (FACT), formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.

8.
J Nucl Med ; 49(6): 970-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18483087

RESUMEN

In animal models of cardiac disease and in human congestive heart failure, expression of angiotensin-converting enzyme (ACE) is upregulated in the failing heart and has been associated with disease progression leading to cardiac failure and fibrosis. To develop probes for imaging ACE expression, a series of di(2-pyridylmethyl)amine (D) chelates capable of binding M(CO)3+ (M = technetium, rhenium) was conjugated to lisinopril by acylation of the epsilon-amine of the lysine residue with a series of di(2-pyridylmethylamino)alkanoic acids where the distance of the chelator from the lisinopril core was investigated by varying the number of methylene spacer groups to produce di(2-pyridylmethyl)amine(Cx)lisinopril analogs: D(C4)lisinopril, D(C5)lisinopril, and D(C8)lisinopril. The inhibitory activity of each rhenium complex was evaluated in vitro against purified rabbit lung ACE and was shown to vary directly with the length of the methylene spacer: Re[D(C8)lisinopril], inhibitory concentration of 50% (IC50) = 3 nM; Re[D(C5)lisinopril], IC50 = 144 nM; and Re[D(C4)lisinopril], IC50 = 1,146 nM, as compared with lisinopril, IC50 = 4 nM. The in vivo specificity for ACE was determined by examining the biodistribution of the 99mTc-[D(C8)lisinopril] analog in rats with and without pretreatment with unlabeled lisinopril. Uptake in the lungs, a tissue that constitutively expresses ACE, was 15.2 percentage injected dose per gram at 10 min after injection and was dramatically reduced by pretreatment with lisinopril, supporting ACE-mediated binding in vivo. Planar anterior imaging analysis of 99mTc-[D(C8)lisinopril] corroborated these data. Thus, high-affinity 99mTc-labeled ACE inhibitor has been designed with potency similar to that of lisinopril and has been demonstrated to specifically localize to tissues that express ACE in vivo. This agent may be useful in monitoring ACE as a function of disease progression in relevant diseases such as heart failure.


Asunto(s)
Corazón/diagnóstico por imagen , Lisinopril/análogos & derivados , Miocardio/enzimología , Compuestos de Organotecnecio/farmacocinética , Peptidil-Dipeptidasa A/metabolismo , Animales , Perfilación de la Expresión Génica/métodos , Lisinopril/química , Lisinopril/farmacocinética , Tasa de Depuración Metabólica , Especificidad de Órganos , Compuestos de Organotecnecio/química , Cintigrafía , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Distribución Tisular
10.
Cancer Gene Ther ; 25(5-6): 106-116, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29755109

RESUMEN

The purpose of this study was to determine if localized delivery of IL-12 encoded by a replication-incompetent adenoviral vector engineered to express IL-12 via a RheoSwitch Therapeutic System® (RTS®) gene switch (Ad-RTS-IL-12) administered intratumorally which is inducibly controlled by the oral activator veledimex is an effective approach for glioma therapy. Mice bearing 5-10-day-old intracranial GL-261 gliomas were intratumorally administered Ad-RTS-mIL-12 in which IL-12 protein expression is tightly controlled by the activator ligand, veledimex. Local tumor viral vector levels concomitant with veledimex levels, IL-12-mRNA expression, local and systemic cytokine expression, tumor and systemic flow cytometry and overall survival were studied. Ad-RTS-mIL-12+veledimex elicited a dose-related increase in tumor IL-12 mRNA and IL-12 protein and discontinuation of veledimex resulted in a return to baseline levels. These changes correlated with local immune and antitumor responses. Veledimex crossed the blood-brain barrier in both orthotopic GL-261 mice and cynomolgus monkeys. We have demonstrated that this therapy induced localized controlled production of IL-12 which correlates with an increase in tumor-infiltrating lymphocytes (TILs) leading to the desired biologic response of tumor growth inhibition and regression. At day 85 (study termination), 65% of the animals that received veledimex at 10 or 30 mg/m2/day were alive and tumor free. In contrast, the median survival for the other groups were: vehicle 23 days, bevacizumab 20 days, temozolomide 33 days and anti-PD-1 37 days. These findings suggest that the controlled intratumoral production of IL-12 induces local immune cell infiltration and improved survival in glioma, thereby demonstrating that this novel regulated immunotherapeutic approach may be an effective form of therapy for glioma.


Asunto(s)
Neoplasias Encefálicas , Expresión Génica , Terapia Genética , Glioma , Interleucina-12/biosíntesis , Neoplasias Experimentales , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Glioma/genética , Glioma/metabolismo , Glioma/patología , Glioma/terapia , Interleucina-12/genética , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia
11.
Clin Pharmacol Drug Dev ; 6(3): 246-257, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27364771

RESUMEN

Major obstacles to developing effective immunotherapy are the ability of tumors to escape the immune system and the toxicity associated with systemic administration. To overcome these challenges, a gene delivery platform technology, RheoSwitch Therapeutic System (RTS), has been developed to enable the regulated expression of a target gene, Ad-RTS-IL-12, administered intratumorally, where IL-12 expression is controlled via the administration of an oral activator ligand, veledimex. Pharmacokinetics in healthy human subjects indicated that veledimex plasma exposure increased with increasing dose after single- and multiple-dose administration in Labrasol slurry and F-22 capsule formulations. No apparent formulation or sex-related difference in veledimex pharmacokinetics (PK) was observed. Minimal or no plasma accumulation of veledimex was observed after once-daily oral administration for 14 days. Veledimex steady state in plasma was reached after 5 daily doses. Food consumption prior to veledimex administration prolonged and enhanced absorption with no impact on the elimination rate and extent of metabolism of veledimex, resulting in significantly increased systemic exposure to veledimex and its 2 major circulating metabolites. Overall, veledimex was well tolerated and exhibited a PK profile supportive of once-daily dosing. For enhanced efficacy, veledimex should be taken under fed conditions to ensure optimal absorption and sufficient systemic exposure.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacocinética , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacocinética , Administración Oral , Adulto , Anciano , Área Bajo la Curva , Método Doble Ciego , Femenino , Semivida , Voluntarios Sanos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Biología Sintética , Adulto Joven
12.
Circulation ; 109(16): 2023-9, 2004 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15066940

RESUMEN

BACKGROUND: Plaque rupture with subsequent thrombosis is recognized as the underlying pathophysiology of most acute coronary syndromes and stroke. Thus, direct thrombus visualization may be beneficial for both diagnosis and guidance of therapy. We sought to test the feasibility of direct imaging of acute and subacute thrombosis using MRI together with a novel fibrin-binding gadolinium-labeled peptide, EP-1873, in an experimental animal model of plaque rupture and thrombosis. METHODS AND RESULTS: Fifteen male New Zealand White rabbits (weight, approximately 3.5 kg) were made atherosclerotic by feeding a high-cholesterol diet after endothelial aortic injury. Plaque rupture was then induced with the use of Russell's viper venom (RVV) and histamine. Subsequently, MRI of the subrenal aorta was performed before RVV, after RVV, and after EP-1873. Histology was performed on regions suggested by MRI to contain thrombus. Nine rabbits (60%) developed plaque rupture and thrombus, including 25 thrombi visually apparent on MRI as "hot spots" after injection of EP-1873. Histological correlation confirmed all 25 thrombi (100%), with no thrombi seen in the other regions of the aorta. In the remaining 6 rabbits (control) without plaque rupture, no thrombus was observed on the MR images or on histology. CONCLUSIONS: We demonstrate the feasibility of in vivo "molecular" MRI for the detection of acute and subacute thrombosis using a novel fibrin-binding MRI contrast agent in an animal model of atherosclerosis and acute/subacute thrombosis. Potential clinical applications include thrombus detection in acute coronary syndromes and stroke.


Asunto(s)
Medios de Contraste , Fibrina/metabolismo , Imagen por Resonancia Magnética/métodos , Péptidos , Trombosis/diagnóstico , Enfermedad Aguda , Animales , Arteriosclerosis/complicaciones , Arteriosclerosis/patología , Unión Competitiva , Gadolinio DTPA , Masculino , Péptidos/metabolismo , Conejos , Trombosis/etiología , Factores de Tiempo
13.
Clin Cancer Res ; 10(3): 1047-56, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14871984

RESUMEN

PURPOSE: To investigate potential immunotherapeutic strategies in B lymphocytic malignancies we looked for CTLs recognizing CD19 and CD20 epitopes. EXPERIMENTAL DESIGN: Three CD19 and CD20 peptides binding to HLA-A*0201 were identified and used to detect peptide specific CTLs by a quantitative real-time PCR to measure IFN-gamma mRNA expression in 23 healthy individuals and 28 patients (18 chronic lymphocytic leukemia (CLL), 7 follicular lymphoma, 2 acute lymphocytic leukemia, and 1 large cell lymphoma). Peptide-specific CTLs were expanded in culture with CD40-activated B cells to test lytic activity in three patients. RESULTS: In healthy individuals, CD8+ T-cell responses were detected in one to CD19(74-82), in three to CD20(127-135), and three to CD20(188-196). Seven of 27 patients (6 with CLL) had CD8+ T cells recognizing CD19(74-82). Seven patients responded to CD20(127-135) and three to CD20(188-196). All were CLL patients. CD19(74-82)-specific CTLs from three patients were expanded over 4 weeks. These cells were HLA-A*0201 specific and lytic for peptide-loaded antigen-presenting cells but not to malignant or unpulsed B cells. CONCLUSIONS: CTLs that recognize CD19 and CD20 epitopes exist in healthy individuals and may be increased in CLL patients. They are of low avidity and require high doses of peptide for activation. Strategies to increase T-cell avidity would be necessary for T-cell immunotherapeutic approaches using the peptides studied.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfoma de Células B/metabolismo , Péptidos/química , Linfocitos T Citotóxicos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos/química , Antígenos CD19/biosíntesis , Antígenos CD19/metabolismo , Antígenos CD20/biosíntesis , Antígenos CD20/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Linfocitos B/inmunología , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , ADN Complementario/metabolismo , Femenino , Citometría de Flujo , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Interferón gamma/metabolismo , Linfoma de Células B/inmunología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Unión Proteica , ARN/química , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Exp Hematol ; 30(8): 943-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12160846

RESUMEN

OBJECTIVE: Recent studies suggest that primitive bone marrow-derived cells contribute to regeneration of many tissues, including muscle, endothelium, myocardium, neural tissues, liver, and skin. Conversely, primitive cells resident in muscle and other tissues have been reported to reconstitute hematopoiesis. We investigated the contribution of cells with a primitive hematopoietic phenotype to human epidermal skin formation in recipients of allogeneic mobilized peripheral blood hematopoietic stem cell (HSC) transplantation. PATIENTS AND METHODS: Our study population included female patients who had received granulocyte colony-stimulating factor mobilized peripheral blood HSC transplants from male donors for a variety of benign and malignant hematologic disorders at least 6 months before study entry, with a history of skin graft-vs-host disease. Epidermal skin cells (keratinocytes) obtained from punch biopsies of the skin were cultured under conditions specific for growth and expansion of homogenous populations of keratinocytes from keratinocyte stem cells. After multiple passages, DNA was extracted from cultured cells and evaluated by two different polymerase chain reaction (PCR) method for detection of Y chromosome specific sequences. RESULTS: Neither sensitive PCR-based technique revealed the presence of male donor-derived keratinocyte stem cells in keratinocytes cultured from skin biopsies of female allogeneic transplantation recipients. CONCLUSIONS: We could not confirm the contribution of donor mobilized peripheral blood hematopoietic stem cells to keratinocyte stem cell populations after HSC transplantation. These results cannot explain the presence of donor-derived cells with keratinocyte phenotypic markers in tissue sections of HSC transplant recipients.


Asunto(s)
Células Epidérmicas , Supervivencia de Injerto , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Queratinocitos/citología , Células Madre/citología , Trasplante Homólogo , Adulto , Amelogenina , Biopsia , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteínas del Esmalte Dental/genética , Femenino , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Hibridación Fluorescente in Situ , Antígenos Comunes de Leucocito/análisis , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Especificidad de Órganos , Donantes de Tejidos , Quimera por Trasplante , Cromosoma X/genética , Cromosoma Y/genética
15.
Inorg Chem ; 38(6): 1326-1335, 1999 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11670921

RESUMEN

Ternary ligand technetium complexes of a hydrazinonicotinamide-conjugated cyclic peptide (HYNICtide: cyclo(D-Val-NMeArg-Gly-Asp-Mamb(5-(6-(6-hydrazinonicotinamido)hexanamide)))) and 2-hydrazinopyridine (HYPY) were prepared and characterized by various spectroscopic methods. The HPLC concordance experiments for (99m)Tc and (99)Tc analogues show clearly that the same complexes are prepared on the no-carrier-added ((99m)Tc) and the carrier-added ((99)Tc) levels. Using a chirality experiment, it was demonstrated that the presence of two radiometric peaks in the HPLC chromatograms of RP444, RP445, and RP446 is due to the resolution of diastereomers, which result from the presence of chiral cyclic peptide and the formation of two enantiomers of the technetium chelate. In a ligand challenge experiment, we found that the high solution stability of these ternary ligand [(99m)Tc]HYNICtide complexes is due to their kinetic inertness. The 1:1:1:1 composition for Tc:HYNICtide:L:tricine (L = TPPTS, TPPDS, and TPPMS) in these ternary ligand [(99)Tc]HYNICtide complexes is confirmed by (1)H NMR and FAB mass spectral data and is completely consistent with that determined on the tracer ((99m)Tc) level. In addition, the IC(50) values of RP444, RP445, and RP446 and the two isomeric forms of RP444 were determined using a platelet IIb/IIIa binding assay. Both isomeric forms of RP444 were found to have the same binding affinity (IC(50) = 13 +/- 2 nM). Complexes [(99)Tc(HYPY)(PPh(3))(2)Cl(2)] and [(99)Tc(HYPY)(PPh(3))(tricine)] were isolated from the reaction of HYPY with [n-Bu(4)N][TcOCl(4)(-)] in the presence of excess tricine and triphenylphosphine. [(99)Tc(HYPY)(PPh(3))(tricine)] serves as a model for ternary ligand [(99m)Tc]HYNICtide complexes. Both complexes have been characterized by HPLC, spectroscopic (IR, NMR, and FAB-MS) methods, and elemental analysis. The HPLC concordance for complexes [(99m)Tc(HYPY)(PPh(3))(tricine)] and [(99)Tc(HYPY)(PPh(3))(tricine)] shows that the two complexes are identical. The NMR ((1)H and (13)C) data suggests that the complex [(99)Tc(HYPY)(PPh(3))(tricine)] have an octahedral coordination geometry with a monodentate diazenido HYPY, a tetradentate tricine, and a monodentate triphenylphosphine coligand.

16.
Clin Nucl Med ; 28(6): 480-4, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12911097

RESUMEN

PURPOSE: Infective endocarditis (IE) is characterized by aggregation of activated platelets, fibrin, and bacteria. DMP444, a high-affinity glycoprotein IIb/IIIa receptor antagonist, binds to the fibrinogen-binding domain of activated platelets, depicting a key feature of IE. Tc-99m DMP444 scintigraphy was studied in a group of patients with possible IE. METHODS: Tc-99m DMP444 (600 MBq; 16 mCi) planar and SPECT images of the heart were recorded in patients with possible IE for as long as 6 hours after injection. Results were compared to echocardiography and the Duke classification. RESULTS: Sixteen patients (age range, 37 to 78 years) participated. DMP444 imaging was positive on SPECT in five patients, and all had definite endocarditis (affecting both prosthetic and native valves). Eleven patients were DMP444 negative, seven with no proof of IE. The remaining four patients were classified as having IE, but three had been receiving adequate intravenous antibiotic regimens for > or = 2 weeks at the time of scintigraphy and one had Q-fever endocarditis. CONCLUSIONS: DMP444 SPECT allows in vivo visualization of IE if it is performed within 1 to 2 weeks after the start of antibiotic treatment. Given the high affinity of DMP444 for activated platelets, the results indicate the involvement of activated platelets in early IE.


Asunto(s)
Endocarditis Bacteriana/diagnóstico por imagen , Oligopéptidos , Compuestos de Organotecnecio , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Tomografía Computarizada de Emisión de Fotón Único , Adulto , Anciano , Ecocardiografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Radiofármacos
17.
J Nucl Med ; 54(3): 380-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23303962

RESUMEN

UNLABELLED: This phase 1 study was performed to determine the pharmacokinetics and ability to visualize prostate cancer in bone, soft-tissue, and the prostate gland using (123)I-MIP-1072 and (123)I-MIP-1095, novel radiolabeled small molecules targeting prostate-specific membrane antigen. METHODS: Seven patients with a documented history of prostate cancer by histopathology or radiologic evidence of metastatic disease were intravenously administered 370 MBq (10 mCi) of (123)I-MIP-1072 and (123)I-MIP-1095 2 wk apart in a crossover trial design. (123)I-MIP-1072 was also studied in 6 healthy volunteers. Whole-body planar and SPECT/CT imaging was performed and pharmacokinetics studied over 2-3 d. Target-to-background ratios were calculated. Absorbed radiation doses were estimated using OLINDA/EXM. RESULTS: (123)I-MIP-1072 and (123)I-MIP-1095 visualized lesions in soft tissue, bone, and the prostate gland within 0.5-1 h after injection, with retention beyond 48 h. Target-to-background ratios from planar images averaged 2:1 at 1 h, 3:1 at 4-24 h, and greater than 10:1 at 4 and 24 h for SPECT/CT. Both agents cleared the blood in a biphasic manner; clearance of (123)I-MIP-1072 was approximately 5 times faster. (123)I-MIP-1072 was excreted in the urine, with 54% and 74% present by 24 and 72 h, respectively. In contrast, only 7% and 20% of (123)I-MIP-1095 had been renally excreted by 24 and 72 h, respectively. Estimated absorbed radiation doses were 0.054 versus 0.110 mGy/MBq for the kidneys and 0.024 versus 0.058 mGy/MBq for the liver, for (123)I-MIP-1072 and (123)I-MIP-1095, respectively. CONCLUSION: (123)I-MIP-1072 and (123)I-MIP-1095 detect lesions in soft tissue, bone, and the prostate gland at as early as 1-4 h. These novel radiolabeled small molecules have excellent pharmacokinetic and pharmacodynamic profiles and warrant further development as diagnostic and potentially when labeled with (131)I therapeutic radiopharmaceuticals.


Asunto(s)
Glutamatos , Neoplasias de la Próstata/diagnóstico por imagen , Urea/análogos & derivados , Anciano , Anciano de 80 o más Años , Estudios Cruzados , Glutamatos/farmacocinética , Humanos , Radioisótopos de Yodo/farmacocinética , Masculino , Persona de Mediana Edad , Imagen Multimodal , Tomografía de Emisión de Positrones , Dosis de Radiación , Radiofármacos/farmacocinética , Distribución Tisular , Tomografía Computarizada por Rayos X , Urea/farmacocinética
18.
Cancer Biother Radiopharm ; 25(3): 299-308, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20578835

RESUMEN

Metaiodobenzylguanidine (MIBG) is an enzymatically stable synthetic analog of norepinephrine that when radiolabled with diagnostic ((123)I) or therapeutic ((131)I) isotopes has been shown to concentrate highly in sympathetically innervated tissues such as the heart and neuroendocrine tumors that possesses high levels of norepinephrine transporter (NET). As the transport of MIBG by NET is a saturable event, the specific activity of the preparation may have dramatic effects on both the efficacy and safety of the radiodiagnostic/radiotherapeutic. Using a solid labeling approach (Ultratrace), noncarrier-added radiolabeled MIBG can be efficiently produced. In this study, specific activities of >1200 mCi/micromol for (123)I and >1600 mCi/micromol for (131)I have been achieved. A series of studies were performed to assess the impact of cold carrier MIBG on the tissue distribution of (123/131)I-MIBG in the conscious rat and on cardiovascular parameters in the conscious instrumented dog. The present series of studies demonstrated that the carrier-free Ultratrace MIBG radiolabeled with either (123)I or (131)I exhibited similar tissue distribution to the carrier-added radiolabeled MIBG in all nontarget tissues. In tissues that express NETs, the higher the specific activity of the preparation the greater will be the radiopharmaceutical uptake. This was reflected by greater efficacy in the mouse neuroblastoma SK-N-BE(2c) xenograft model and less appreciable cardiovascular side-effects in dogs when the high-specific-activity radiopharmaceutical was used. The increased uptake and retention of Ultratrace (123/131)I-MIBG may translate into a superior diagnostic and therapeutic potential. Lastly, care must be taken when administering therapeutic doses of the current carrier-added (131)I-MIBG because of its potential to cause adverse cardiovascular side-effects, nausea, and vomiting.


Asunto(s)
3-Yodobencilguanidina/análogos & derivados , 3-Yodobencilguanidina/farmacocinética , 3-Yodobencilguanidina/uso terapéutico , Radioisótopos de Yodo/química , Radiofármacos/farmacocinética , Radiofármacos/uso terapéutico , 3-Yodobencilguanidina/química , 3-Yodobencilguanidina/farmacología , Estructuras Animales/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Médula Ósea/metabolismo , Bradicardia/inducido químicamente , Perros , Electrocardiografía/efectos de los fármacos , Femenino , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Radioisótopos de Yodo/uso terapéutico , Marcaje Isotópico/métodos , Masculino , Ratones , Ratones Desnudos , Miocardio/metabolismo , Neuroblastoma/patología , Neuroblastoma/radioterapia , Radiofármacos/síntesis química , Radiofármacos/farmacología , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 70(10): 4045-53, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20442292

RESUMEN

Radiolabeled benzamides are attractive candidates for targeted radiotherapy of metastatic melanoma as they bind melanin and exhibit high tumor uptake and retention. One such benzamide, N-(2-diethylamino-ethyl)-4-(4-fluoro-benzamido)-5-iodo-2-methoxy-benzamide (MIP-1145), was evaluated for its ability to distinguish melanin-expressing from amelanotic human melanoma cells, and to specifically localize to melanin-containing tumor xenografts. The binding of [(131)I]MIP-1145 to melanoma cells in vitro was melanin dependent, increased over time, and insensitive to mild acid treatment, indicating that it was retained within cells. Cold carrier MIP-1145 did not reduce the binding, consistent with the high capacity of melanin binding of benzamides. In human melanoma xenografts, [(131)I]MIP-1145 exhibited diffuse tissue distribution and washout from all tissues except melanin-expressing tumors. Tumor uptake of 8.82% injected dose per gram (ID/g) was seen at 4 hours postinjection and remained at 5.91% ID/g at 24 hours, with tumor/blood ratios of 25.2 and 197, respectively. Single photon emission computed tomography imaging was consistent with tissue distribution results. The administration of [(131)I]MIP-1145 at 25 MBq or 2.5 GBq/m(2) in single or multiple doses significantly reduced SK-MEL-3 tumor growth, with multiple doses resulting in tumor regression and a durable response for over 125 days. To estimate human dosimetry, gamma camera imaging and pharmacokinetic analysis was performed in cynomolgus monkeys. The melanin-specific binding of [(131)I]MIP-1145 combined with prolonged tumor retention, the ability to significantly inhibit tumor growth, and acceptable projected human dosimetry suggest that it may be effective as a radiotherapeutic pharmaceutical for treating patients with metastatic malignant melanoma.


Asunto(s)
Benzamidas/uso terapéutico , Radioisótopos de Yodo/uso terapéutico , Melaninas/metabolismo , Melanoma Experimental/radioterapia , Radiofármacos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Macaca fascicularis , Masculino , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Dosificación Radioterapéutica , Tasa de Supervivencia , Tomografía Computarizada de Emisión de Fotón Único
20.
Bioanalysis ; 1(2): 321-56, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-21083171

RESUMEN

The era of 'modern medicine' has changed its name to 'molecular medicine', and reflects a new age based on personalized medicine utilizing molecular biomarkers in the diagnosis, staging and monitoring of therapy. Alzheimer's disease has a classical biomarker determined at autopsy with the histologic staining of amyloid accumulation in the brain. Today we can diagnose Alzheimer's disease using the same classical pathologic biomarker, but now using a noninvasive imaging probe to image the amyloid deposition in a patient and potentially provide treatment strategies and measure their effectiveness. Molecular medicine is the exploitation of biomarkers to detect disease before overt expression of pathology. Physicians can now find, fight and follow disease using imaging, and the need for other disease biomarkers is in high demand. This review will discuss the innovative physical and molecular biomarker probes now being developed for imaging systems and we will introduce the concepts needed for validation and regulatory acceptance of surrogate biomarkers in the detection and treatment of disease.


Asunto(s)
Biomarcadores/análisis , Diagnóstico por Imagen , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Animales , Biomarcadores/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Farmacocinética , Conejos , Cintigrafía , Ratas , Investigación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA