Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 121(8): 1304-15, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23287859

RESUMEN

STAT3 operates in both cancer cells and tumor-associated immune cells to promote cancer progression. As a transcription factor, it is a highly desirable but difficult target for pharmacologic inhibition. We have recently shown that the TLR9 agonists CpG oligonucleotides can be used for targeted siRNA delivery to mouse immune cells. In the present study, we demonstrate that a similar strategy allows for targeted gene silencing in both normal and malignant human TLR9(+) hematopoietic cells in vivo. We have developed new human cell-specific CpG(A)-STAT3 siRNA conjugates capable of inducing TLR9-dependent gene silencing and activation of primary immune cells such as myeloid dendritic cells, plasmacytoid dendritic cells, and B cells in vitro. TLR9 is also expressed by several human hematologic malignancies, including B-cell lymphoma, multiple myeloma, and acute myeloid leukemia. We further demonstrate that oncogenic proteins such as STAT3 or BCL-X(L) are effectively knocked down by specific CpG(A)-siRNAs in TLR9(+) hematologic tumor cells in vivo. Targeting survival signaling using CpG(A)-siRNAs inhibits the growth of several xenotransplanted multiple myeloma and acute myeloid leukemia tumors. CpG(A)-STAT3 siRNA is immunostimulatory and nontoxic for normal human leukocytes in vitro. The results of the present study show the potential of using tumoricidal/immunostimulatory CpG-siRNA oligonucleotides as a novel 2-pronged therapeutic strategy for hematologic malignancies.


Asunto(s)
Terapia Genética/métodos , Neoplasias Hematológicas/terapia , Células Madre Hematopoyéticas/fisiología , ARN Interferente Pequeño/farmacocinética , Receptor Toll-Like 9/genética , Animales , Línea Celular Tumoral , Técnicas de Transferencia de Gen , Neoplasias Hematológicas/genética , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Linfoma/genética , Linfoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/terapia , Trasplante de Neoplasias , Oligodesoxirribonucleótidos/genética , ARN Interferente Pequeño/genética , Radioterapia , Factor de Transcripción STAT3/genética , Receptor Toll-Like 9/metabolismo , Trasplante Heterólogo
2.
Biol Blood Marrow Transplant ; 20(12): 2072-5, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25079874

RESUMEN

Transformation from indolent non-Hodgkin lymphoma (NHL) to diffuse large B cell lymphoma (DLBCL) has historically been associated with a poor prognosis. A small series of autologous stem cell transplantation (ASCT) studies using conventional conditioning regimens has demonstrated durable progression-free survival (PFS) rates ranging from 25% to 47%, but data in the rituximab era are lacking. Here we report the results of a multicenter retrospective trial evaluating ASCT in patients with transformed lymphoma using the Z-BEAM conditioning regimen, which combines yttrium-90-labeled ibritumomab tiuxetan (Zevalin) with high-dose BEAM (carmustine, etoposide, cytarabine, melphalan) chemotherapy. Sixty-three patients from 4 institutions were treated between 2003 and 2011. Histological confirmation of transformation was required and defined as a diagnosis of DLBCL in patients with either a prior history or concomitant diagnosis of low-grade B cell NHL. Median patient age at ASCT was 59.5 years, median number of prior regimens was 2, and all patients were exposed to rituximab. Disease status at ASCT was as follows: first complete remission (CR) (n = 30), first partial remission (n = 11), first relapse (n = 14), and at least second CR (n = 8). The median time from diagnosis of histological transformation to ASCT was 7.5 months (range, 2.8 to 116). Two-year nonrelapse mortality was 0%. Median follow-up for living patients was 28 months (range, 5 to 103). Two-year PFS was 68% (95% confidence interval, 58% to 75%), and overall survival was 90% (95% confidence interval, 80% to 95%). In conclusion, the Z-BEAM conditioning regimen for ASCT is well tolerated by patients with transformed lymphoma and demonstrates encouraging clinical outcomes.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Linfoma no Hodgkin/mortalidad , Linfoma no Hodgkin/terapia , Trasplante de Células Madre , Acondicionamiento Pretrasplante , Adulto , Anciano , Autoinjertos , Carmustina/administración & dosificación , Citarabina/administración & dosificación , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Podofilotoxina/administración & dosificación , Tasa de Supervivencia
3.
Blood ; 119(17): 3940-50, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22308288

RESUMEN

Cellular immune responses have the potential to elicit dramatic and sustained clinical remissions in lymphoma patients. Recent clinical trial data demonstrate that modification of T cells with chimeric antigen receptors (CARs) is a promising strategy. T cells containing CARs with costimulatory domains exhibit improved activity against tumors. We conducted a pilot clinical trial testing a "third-generation" CD20-specific CAR with CD28 and 4-1BB costimulatory domains in patients with relapsed indolent B-cell and mantle cell lymphomas. Four patients were enrolled, and 3 received T-cell infusions after cyclophosphamide lymphodepletion. Treatment was well tolerated, although one patient developed transient infusional symptoms. Two patients without evaluable disease remained progression-free for 12 and 24 months. The third patient had an objective partial remission and relapsed at 12 months after infusions. Modified T cells were detected by quantitative PCR at tumor sites and up to 1 year in peripheral blood, albeit at low levels. No evidence of host immune responses against infused cells was detected. In conclusion, adoptive immunotherapy with CD20-specific T cells was well tolerated and was associated with antitumor activity. We will pursue alternative gene transfer technologies and culture conditions in future studies to improve CAR expression and cell production efficiency.


Asunto(s)
Antígenos CD20/metabolismo , Antígenos CD28/genética , Inmunoterapia Adoptiva , Linfoma/terapia , Receptores de Antígenos/genética , Linfocitos T/trasplante , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Anciano , Anciano de 80 o más Años , Antígenos CD20/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD28/inmunología , Humanos , Linfoma/inmunología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Pronóstico , Receptores de Antígenos/inmunología , Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
4.
Biol Blood Marrow Transplant ; 18(3): 441-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21801706

RESUMEN

We conducted a matched-cohort analysis of autologous transplant conditioning regimens for diffuse large cell lymphoma in 92 patients treated with either radioimmunotherapy (RIT) or total body irradiation (TBI)-based conditioning regimens. The RIT regimen consisted of 0.4 mCi/kg of (90)Y-ibritumomab tiuxetan plus BEAM (BCNU, etoposide, cytarabine, melphalan). The TBI-based regimen combined fractionated TBI at 1200 cGy, with etoposide and cyclophosphamide. Five factors were matched between 46 patient pairs: age at transplant ±5 years, disease status at salvage, number of prior regimens, year of diagnosis ±5 years, and year of transplantation ±5 years. Patients in the TBI group had higher rates of cardiac toxicity and mucositis, whereas Z-BEAM patients had a higher incidence of pulmonary toxicity. Overall survival at 4 years was 81.0% for the Z-BEAM and 52.7% for the TBI group (P = .01). The 4-year cumulative incidence of relapse/progression was 40.4% and 42.1% for Z-BEAM and TBI, respectively (P = .63). Nonrelapse mortality was superior in the Z-BEAM group: 0% compared with 15.8% for TBI at 4 years (P < .01). Our data demonstrate that RIT-based conditioning had a similar relapse incidence to TBI, with lower toxicity, resulting in improved overall survival, particularly in patients with ≥2 prior regimens.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Linfoma de Células B Grandes Difuso/terapia , Radioinmunoterapia/métodos , Acondicionamiento Pretrasplante/métodos , Irradiación Corporal Total/métodos , Adulto , Anciano , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Estudios de Cohortes , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Linfoma de Células B Grandes Difuso/radioterapia , Linfoma de Células B Grandes Difuso/cirugía , Masculino , Persona de Mediana Edad , Radiofármacos/uso terapéutico , Rituximab , Terapia Recuperativa/métodos , Acondicionamiento Pretrasplante/efectos adversos , Trasplante Autólogo , Irradiación Corporal Total/efectos adversos , Adulto Joven
5.
J Immunol ; 184(8): 4284-94, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20220093

RESUMEN

We investigated relationships among chimeric TCR (cTCR) expression density, target Ag density, and cTCR triggering to predict lysis of target cells by cTCR(+) CD8(+) T human cells as a function of Ag density. Triggering of cTCR and canonical TCR by Ag could be quantified by the same mathematical equation, but cTCR represented a special case in which serial triggering was abrogated. The magnitude of target lysis could be predicted as a function of cTCR triggering, and the predicted minimum cTCR density required for maximal target lysis by CD20-specific cTCR was experimentally tested. cTCR density below approximately 20,000 cTCR/cell impaired target lysis, but increasing cTCR expression above this density did not improve target lysis or Ag sensitivity. cTCR downmodulation to densities below this critical minimum by interaction with Ag-expressing targets limited the sequential lysis of targets in a manner that could be predicted based on the number of cTCRs remaining. In contrast, acute inhibition of lysis of primary, intended targets (e.g., leukemic B cells) due to the presence of an excess of secondary targets (e.g., normal B cells) was dependent on the Ag density of the secondary target but occurred at Ag densities insufficient to promote significant cTCR downmodulation, suggesting a role for functional exhaustion rather than insufficient cTCR density. This suggests increasing cTCR density above a critical threshold may enhance sequential lysis of intended targets in isolation, but will not overcome the functional exhaustion of cTCR(+) T cells encountered in the presence of secondary targets with high Ag density.


Asunto(s)
Citotoxicidad Inmunológica/genética , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Modelos Inmunológicos , Proteínas Mutantes Quiméricas/antagonistas & inhibidores , Proteínas Mutantes Quiméricas/genética , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/genética , Antígenos CD20/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Línea Celular Transformada , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Relación Dosis-Respuesta Inmunológica , Humanos , Células Jurkat , Modelos Lineales , Proteínas Mutantes Quiméricas/biosíntesis , Valor Predictivo de las Pruebas , Receptores de Antígenos de Linfocitos T/biosíntesis , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
6.
Bioconjug Chem ; 22(4): 709-16, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21395337

RESUMEN

Optimal PET imaging of tumors with radiolabeled engineered antibodies requires, among other parameters, matching blood clearance and tumor uptake with the half-life of the engineered antibody. Although diabodies have favorable molecular sizes (50 kDa) for rapid blood clearance (t(1/2) = 30-60 min) and are bivalent, thereby increasing tumor uptake, they exhibit substantial kidney uptake as their major route of clearance, which is especially evident when they are labeled with the PET isotope (64)Cu (t(1/2) = 12 h). To overcome this drawback, diabodies may be conjugated to PEG, a modification that increases the apparent molecular size of the diabody and reduces kidney uptake without adversely affecting tumor uptake or the tumor to blood ratio. We show here that site-specific attachment of monodispersed PEGn of increasing molecular size (n = 12, 24, and 48) can uniformly increase the apparent molecular size of the PEG-diabody conjugate, decrease kidney uptake, and increase tumor uptake, the latter due to the increased residence time of the conjugate in the blood. Since the monodispersed PEGs were preconjugated to the chelator DOTA, the conjugates were able to bind radiometals such as (111)In and (64)Cu that can be used for SPECT and PET imaging, respectively. To allow conjugation of the DOTA-PEG to the diabody, the DOTA-PEG incorporated a terminal cysteine conjugated to a vinyl sulfone moiety. In order to control the conjugation chemistry, we have engineered a surface thiolated diabody that incorporates two cysteines per monomer (four per diabody). The thiolated diabody was expressed and purified from bacterial fermentation and only needs to be reduced prior to conjugation to the DOTA-PEGn-Cys-VS. This novel imaging agent (a diabody with DOTA-PEG48-Cys-VS attached to introduced thiols) gave up to 80%ID/g of tumor uptake with a tumor to blood ratio (T/B) of 8 at 24 h when radiolabeled with (111)In and 37.9% ID/g of tumor uptake (T/B = 8) at 44 h when radiolabeled with (64)Cu in PET imaging in an animal model. Tumor uptake was significantly improved from the 50% ID/g at 24 h observed with diabodies that were pegylated on surface lysine residues. Importantly, there was no loss of immunoreactivity of the site-specific Cys-conjugated diabody to its antigen (TAG-72) compared to the parent, unconjugated diabody. We propose that thiolated diabodies conjugated to DOTAylated monodisperse PEGs have the potential for superior SPECT and PET imaging in a clinical setting.


Asunto(s)
Compuestos Heterocíclicos con 1 Anillo , Riñón/metabolismo , Neoplasias/metabolismo , Polietilenglicoles , Tomografía de Emisión de Positrones , Radiofármacos , Compuestos de Sulfhidrilo/química , Animales , Sitios de Unión , Radioisótopos de Cobre/química , Radioisótopos de Cobre/farmacocinética , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Riñón/diagnóstico por imagen , Ratones , Ratones Desnudos , Estructura Molecular , Trasplante de Neoplasias , Neoplasias/diagnóstico por imagen , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Radiofármacos/química , Radiofármacos/farmacocinética , Distribución Tisular
7.
Blood ; 112(6): 2261-71, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18509084

RESUMEN

Adoptive immunotherapy with T cells expressing a tumor-specific chimeric T-cell receptor is a promising approach to cancer therapy that has not previously been explored for the treatment of lymphoma in human subjects. We report the results of a proof-of-concept clinical trial in which patients with relapsed or refractory indolent B-cell lymphoma or mantle cell lymphoma were treated with autologous T cells genetically modified by electroporation with a vector plasmid encoding a CD20-specific chimeric T-cell receptor and neomycin resistance gene. Transfected cells were immunophenotypically similar to CD8(+) effector cells and showed CD20-specific cytotoxicity in vitro. Seven patients received a total of 20 T-cell infusions, with minimal toxicities. Modified T cells persisted in vivo 1 to 3 weeks in the first 3 patients, who received T cells produced by limiting dilution methods, but persisted 5 to 9 weeks in the next 4 patients who received T cells produced in bulk cultures followed by 14 days of low-dose subcutaneous interleukin-2 (IL-2) injections. Of the 7 treated patients, 2 maintained a previous complete response, 1 achieved a partial response, and 4 had stable disease. These results show the safety, feasibility, and potential antitumor activity of adoptive T-cell therapy using this approach. This trial was registered at www.clinicaltrials.gov as #NCT00012207.


Asunto(s)
Antígenos CD20/genética , Inmunoterapia Adoptiva/métodos , Linfoma de Células del Manto/terapia , Linfoma no Hodgkin/terapia , Linfocitos T/trasplante , Adulto , Anciano , Femenino , Humanos , Transfusión de Linfocitos/métodos , Masculino , Persona de Mediana Edad , Inducción de Remisión , Terapia Recuperativa/métodos , Linfocitos T/metabolismo , Transfección , Trasplante Autólogo , Resultado del Tratamiento
8.
Clin Cancer Res ; 15(8): 2935-41, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351765

RESUMEN

PURPOSE: To determine the maximum tolerated dose of combined therapy using an yttrium-90-labeled anti-carcinoembryonic antigen (CEA) antibody with gemcitabine in patients with advanced CEA-producing solid tumors. EXPERIMENTAL DESIGN: The chimeric human/murine cT84.66 is an anti-CEA intact IgG1, with high affinity and specificity to CEA. This was given at a fixed yttrium-90-labeled dose of 16.6 mCi/m(2) to subjects who had and an elevated CEA in serum or in tumor by immunohistochemistry. Also required was a tumor that imaged with an (111)In-labeled cT84.66 antibody. Patients were treated with escalating doses of gemcitabine given i.v. over 30 minutes on day 1 and 3 after the infusion of the yttrium-90-labeled antibody. Patients were treated in cohorts of 3. The maximum tolerated dose was determined as the highest level at which no >1 of 6 patients experienced a dose limiting toxicity. RESULTS: A total of 36 patients were enrolled, and all but one had prior systemic therapy. The maximum tolerated dose of gemcitabine in this combination was 150 mg/m(2). Dose limiting toxicities at a gemcitabine dose of 165 mg/m(2) included a grade 3 rash and grade 4 neutropenia. One partial response was seen in a patient with colorectal cancer, and 4 patients had a >50% decrease in baseline CEA levels associated with stable disease. Human antichimeric antibody responses were the primary reason for stopping treatment in 12 patients. CONCLUSIONS: Feasibility of combining gemcitabine with an yttrium-90-labeled anti-CEA antibody is shown with preliminary evidence of clinical response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno Carcinoembrionario/inmunología , Desoxicitidina/análogos & derivados , Neoplasias Gastrointestinales/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Anciano , Anticuerpos Monoclonales/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica , Desoxicitidina/efectos adversos , Desoxicitidina/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Compuestos Organometálicos/efectos adversos , Resultado del Tratamiento , Gemcitabina
9.
J Nucl Med ; 50(9): 1500-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19690034

RESUMEN

UNLABELLED: The CD20 cell surface antigen is expressed at high levels by over 90% of B-cell non-Hodgkin lymphomas (NHL) and is the target of the anti-CD20 monoclonal antibody rituximab. To provide more sensitive, tumor-specific PET imaging of NHL, we sought to develop PET agents targeting CD20. METHODS: Two recombinant anti-CD20 rituximab fragments, a minibody (scFv-C(H)3 dimer; 80 kDa) and a modified scFv-Fc fragment (105 kDa), designed to clear rapidly, were generated. Both fragments were radiolabeled with (124)I, and the minibody was additionally labeled with (64)Cu (radiometal) after conjugation to 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA). The radioiodinated fragments and the radiometal-labeled minibody were evaluated in mice as small-animal PET imaging agents for the in vivo imaging of human CD20-expressing lymphomas. RESULTS: Rapid and specific localization to CD20-positive tumors was observed with the radioiodinated fragments. However, the tumor uptake levels and blood activities differed, resulting in different levels of contrast in the images. The better candidate was the minibody, with superior uptake (2-fold higher than that obtained with scFv-Fc) in CD20-positive tumors and low uptake in CD20-negative tumors. Ratios of CD20-positive tumors to CD20-negative tumors at 21 h were 7.0 +/- 3.1 (mean +/- SD) and 3.9 +/- 0.7 for the minibody and scFv-Fc, respectively. The ratio achieved with the (64)Cu-DOTA-minibody at 19 h was about 5-fold lower because of higher residual background activity in CD20-negative tumors. CONCLUSION: A radioiodinated minibody and a radioiodinated scFv-Fc fragment produced excellent, high-contrast images in vivo. These new immunoPET agents may prove useful for imaging CD20-positive lymphomas in preclinical models and in humans with NHL.


Asunto(s)
Anticuerpos Monoclonales , Linfoma de Células B/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/veterinaria , Radiofármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino , Modelos Animales de Enfermedad , Femenino , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Linfoma de Células B/inmunología , Ratones , Ratones Endogámicos C3H , Radiofármacos/inmunología , Reproducibilidad de los Resultados , Rituximab , Sensibilidad y Especificidad
10.
Clin Cancer Res ; 14(12): 3832-9, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559603

RESUMEN

PURPOSE: Clinical trials have commenced to evaluate the feasibility of targeting malignant gliomas with genetically engineered CTLs delivered directly to the tumor bed in the central nervous system. The objective of this study is to determine a suite of magnetic resonance imaging (MRI) measurements using an orthotopic xenograft murine model that can noninvasively monitor immunologically mediated tumor regression and reactive changes in the surrounding brain parenchyma. EXPERIMENTAL DESIGN: Our preclinical therapeutic platform is based on CTL genetic modification to express a membrane tethered interleukin-13 (IL-13) cytokine chimeric T-cell antigen receptor. This enables selective binding and signal transduction on encountering the glioma-restricted IL-13 alpha2 receptor (IL-13Ralpha2). We used MRI to visualize immune responses following adoptive transfer of IL-13Ralpha2-specific CD8+ CTL clones. RESULTS: Based on MRI measurements, several phases following IL-13Ralpha2-specific T-cell adoptive transfer could be distinguished, all of which correlated well with glioblastoma regression confirmed on histology. The first detectable changes, 24 hours post-treatment, were significantly increased T2 relaxation times and strongly enhanced signal on T1-weighted postcontrast images. In the next phase, the apparent diffusion coefficient was significantly increased at 2 and 3 days post-treatment. In the last phase, at day 3 after IL-13Ralpha2-specific T-cell injection, the volume of hyperintense signal on T1-weighted postcontrast image was significantly decreased, whereas apparent diffusion coefficient remained elevated. CONCLUSIONS: The present study indicates the feasibility of MRI to visualize different phases of immune response when IL-13Ralpha2-specific CTLs are administered directly to the glioma tumor bed. This will further the aim of better predicting clinical outcome following immunotherapy.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Citotoxicidad Inmunológica/genética , Glioblastoma/diagnóstico por imagen , Sistema Inmunológico/diagnóstico por imagen , Subunidad alfa2 del Receptor de Interleucina-13/genética , Linfocitos T Citotóxicos/inmunología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Diagnóstico por Imagen , Estudios de Factibilidad , Glioblastoma/genética , Glioblastoma/inmunología , Glioblastoma/terapia , Humanos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/fisiología , Inmunoterapia Adoptiva , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Radiografía , Linfocitos T Citotóxicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Res ; 67(2): 718-26, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17234783

RESUMEN

Antibody fragments with optimized pharmacokinetic profiles hold potential for detection and therapy of tumor malignancies. We studied the behavior of three anti-carcinoembryonic antigen (CEA) single-chain Fv-Fc (scFv-Fc) variants (I253A, H310A, and H310A/H435Q; Kabat numbering system) that exhibited differential serum persistence. Biodistribution studies done on CEA-positive tumor xenografted mice revealed that the 111In-labeled I253A fragment with the slowest clearance kinetics (T1/2beta, 27.7 h) achieved the highest tumor uptake (44.6% ID/g at 24 h), whereas the radiometal-labeled H310A/H435Q fragment with the most rapid elimination (T1/2beta, 7.05 h) reached a maximum of 28.0% ID/g at 12 h postinjection. The H310A protein was characterized by both intermediate serum half-life and tumor uptake. The 111In-based biodistribution studies showed that all three fragments were eliminated primarily through the liver, and hepatic radiometal activity correlated with the rate of fragment clearance. The 111In-labeled H310A/H435Q protein exhibited the highest liver uptake (23.5% ID/g at 24 h). Metabolism of the 125I-labeled scFv-Fc proteins resulted in low normal organ activity. Finally, the 125I/111In biodistribution data allowed for dose estimations, which suggest the 131I-labeled scFv-Fc H310A/H435Q as a promising candidate for radioimmunotherapy.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Inmunoconjugados/farmacocinética , Fragmentos de Inmunoglobulinas/metabolismo , Radioisótopos de Indio/farmacocinética , Radioisótopos de Yodo/farmacocinética , Radiofármacos/farmacocinética , Animales , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Femenino , Compuestos Heterocíclicos con 1 Anillo/química , Compuestos Heterocíclicos con 1 Anillo/farmacocinética , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Marcaje Isotópico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Distribución Tisular , Trasplante Heterólogo
12.
Cancer Res ; 67(6): 2872-80, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363611

RESUMEN

Currently, the lineage-specific cell-surface molecules CD19 and CD20 present on many B-cell malignancies are targets for both antibody- and cell-based therapies. Coupling these two treatment modalities is predicted to improve the antitumor effect, particularly for tumors resistant to single-agent biotherapies. This can be shown using an immunocytokine, composed of a CD20-specific monoclonal antibody fused to biologically active interleukin 2 (IL-2), combined with ex vivo expanded human umbilical cord blood-derived CD8(+) T cells, that have been genetically modified to be CD19 specific, for adoptive transfer after allogeneic hematopoietic stem-cell transplantation. We show that a benefit of targeted delivery of recombinant IL-2 by the immunocytokine to the CD19(+)CD20(+) tumor microenvironment is improved in vivo persistence of the CD19-specific T cells, and this results in an augmented cell-mediated antitumor effect. Phase I trials are under way using anti-CD20-IL-2 immunocytokine and CD19-specific T cells as monotherapies, and our results warrant clinical trials using combination of these two immunotherapies.


Asunto(s)
Antígenos CD19/inmunología , Inmunoconjugados/farmacología , Inmunoterapia Adoptiva/métodos , Interleucina-2/farmacología , Leucemia de Células B/terapia , Linfoma de Células B/terapia , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/inmunología , Interleucina-2/inmunología , Células K562 , Leucemia de Células B/inmunología , Linfoma de Células B/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID
13.
Bioconjug Chem ; 19(9): 1927-37, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18720981

RESUMEN

Increasing interest in the use of radiolabeled antibodies for cancer imaging and therapy drives the need for more efficient production of the antibody conjugates. Here, we illustrate a method for rapid and efficient production of radiolabeled antibody conjugates using vacuum diafiltration guided by mathematical modeling. We apply this technique to the production of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated antibodies at the milligram and gram production scale and achieve radiolabeling efficiencies >95% using In-111. Using vacuum diafiltration, antibody-chelate conjugation and purification can be accomplished within the same vessel, and the entire process can be completed in <24 h. Vacuum diafiltration also offers safer and gentler processing conditions by eliminating the need to keep the retentate vessel under positive pressure through applied gas pressure or shear-inducing restriction points in the retentate flow path. Experimental data and mathematical model calculations suggest there exists a weak binding affinity (approximately 10(4)M(-1)) between the charged chelate molecules (e.g., DOTA) and the antibodies that slows the removal of excess chelate during purification. By analyzing the radiolabeling efficiency as a function of the number of diavolumes, we demonstrate the importance of balancing the removal of free chelate with the introduction of metal contaminants from the diafiltration buffer and also illustrate how to optimize radiolabeling of antibody conjugates under a variety of operating conditions. This methodology is applicable to the production of antibody conjugates in general.


Asunto(s)
Anticuerpos Monoclonales , Filtración/métodos , Compuestos Heterocíclicos con 1 Anillo/química , Inmunoconjugados/química , Marcaje Isotópico/métodos , Radioinmunoterapia/métodos , Radiofármacos/química , Animales , Quelantes/química , Humanos , Modelos Teóricos , Vacio
14.
Nucl Med Biol ; 35(2): 151-8, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18312824

RESUMEN

Albumin fusion proteins have demonstrated the ability to prolong the in vivo half-life of small therapeutic proteins/peptides in the circulation and thereby potentially increase their therapeutic efficacy. To evaluate if this format can be employed for antibody-based imaging, an anticarcinoembryonic antigen (CEA) single-chain antibody(scFv)-albumin fusion protein was designed, expressed and radiolabeled for biodistribution and imaging studies in athymic mice bearing human colorectal carcinoma LS-174T xenografts. The [125 I]-T84.66 fusion protein demonstrated rapid tumor uptake of 12.3% injected dose per gram (ID/g) at 4 h that reached a plateau of 22.7% ID/g by 18 h. This was a dramatic increase in tumor uptake compared to 4.9% ID/g for the scFv alone. The radiometal [111 In]-labeled version resulted in higher tumor uptake, 37.2% ID/g at 18 h, which persisted at the tumor site with tumor: blood ratios reaching 18:1 and with normal tissues showing limited uptake. Based on these favorable imaging properties, a pilot [64 Cu]-positron emission tomography imaging study was performed with promising results. The anti-CEA T84.66 scFv-albumin fusion protein demonstrates highly specific tumor uptake that is comparable to cognate recombinant antibody fragments. The radiometal-labeled version, which shows lower normal tissue accumulation than these recombinant antibodies, provides a promising and novel platform for antibody-based imaging agents.


Asunto(s)
Albúminas/farmacocinética , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/diagnóstico por imagen , Radioinmunodetección/métodos , Proteínas Recombinantes de Fusión/farmacocinética , Albúminas/genética , Animales , Anticuerpos Antineoplásicos/genética , Antígeno Carcinoembrionario/genética , Neoplasias Colorrectales/inmunología , Femenino , Humanos , Inmunoconjugados/farmacocinética , Región Variable de Inmunoglobulina/genética , Radioisótopos de Indio/farmacocinética , Radioisótopos de Yodo/farmacocinética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Distribución Tisular/inmunología , Trasplante Heterólogo
15.
J Nucl Med ; 59(1): 38-43, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28637802

RESUMEN

The goal of this study was to characterize the relationship between tumor uptake of 64Cu-DOTA-trastuzumab as measured by PET/CT and standard, immunohistochemistry (IHC)-based, histopathologic classification of human epidermal growth factor receptor 2 (HER2) status in women with metastatic breast cancer (MBC). Methods: Women with biopsy-confirmed MBC and not given trastuzumab for 2 mo or more underwent complete staging, including 18F-FDG PET/CT. Patients were classified as HER2-positive (HER2+) or -negative (HER2-) based on fluorescence in situ hybridization (FISH)-supplemented immunohistochemistry of biopsied tumor tissue. Eighteen patients underwent 64Cu-DOTA-trastuzumab injection, preceded in 16 cases by trastuzumab infusion (45 mg). PET/CT was performed 21-25 (day 1) and 47-49 (day 2) h after 64Cu-DOTA-trastuzumab injection. Radiolabel uptake in prominent lesions was measured as SUVmax Average intrapatient SUVmax (pt) was compared between HER2+ and HER2- patients. Results: Eleven women were HER2+ (8 immunohistochemistry 3+; 3 immunohistochemistry 2+/FISH amplified), whereas 7 were HER2- (3 immunohistochemistry 2+/FISH nonamplified; 4 immunohistochemistry 1+). Median pt for day 1 and day 2 was 6.6 and 6.8 g/mL for HER 2+ and 3.7 and 4.3 g/mL for HER2- patients (P < 0.005 either day). The distributions of pt overlapped between the 2 groups, and interpatient variability was greater for HER2+ than HER2- disease (P < 0.005 and 0.001, respectively, on days 1 and 2). Conclusion: By 1 d after injection, uptake of 64Cu-DOTA-trastuzumab in MBC is strongly associated with patient HER2 status and is indicative of binding to HER2. The variability within and among HER2+ patients, as well as the overlap between the HER2+ and HER2- groups, suggests a role for 64Cu-DOTA-trastuzumab PET/CT in optimizing treatments that include trastuzumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Compuestos Organometálicos/metabolismo , Adulto , Anciano , Transporte Biológico , Neoplasias de la Mama/diagnóstico por imagen , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptor ErbB-2/metabolismo , Trastuzumab
16.
Hum Gene Ther ; 18(8): 712-25, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17685852

RESUMEN

We previously demonstrated the feasibility of generating therapeutic numbers of cytotoxic T lymphocyte (CTL) clones expressing a CD20-specific scFvFc:CD3zeta chimeric T cell receptor (cTCR), making them specifically cytotoxic for CD20+ B lymphoma cells. However, the process of generating and expanding he CTL clones was laborious, the CTL clones expressed the cTCR at low surface density, and they exhibited suboptimal proliferation and cytotoxicity. To improve the performance of the CTLs in vitro and in vivo, we engineered "second-generation'' plasmid constructs containing a translational enhancer (SP163) and CD28 and CD137 costimulatory domains in cis with the CD3zeta intracellular signaling domain of the cTCR gene. Furthermore, we verified the superiority of generating genetically modified polyclonal T cells expressing the second-generation cTCR rather than T cell clones. Our results demonstrate that SP163 enhances the surface expression of the cTCR; that the second-generation cTCR improves CTL activation, proliferation, and cytotoxicity; and that polyclonal T cells proliferate rapidly in vitro and mediate potent CD20-specific cytotoxicity. This study provides the preclinical basis for a clinical trial of adoptive T cell immunotherapy for patients with relapsed CD20+ mantle cell lymphoma and indolent lymphomas.


Asunto(s)
Antígenos CD28/genética , Inmunoterapia Adoptiva , Linfoma/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Antígenos CD28/metabolismo , Células Cultivadas , Elementos de Facilitación Genéticos , Humanos , Células Jurkat , Linfoma/inmunología , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/trasplante , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
17.
Cancer Res ; 65(2): 622-31, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15695407

RESUMEN

Antibody fragments are recognized as promising vehicles for delivery of imaging and therapeutic agents to tumor sites in vivo. The serum persistence of IgG1 and fragments with intact Fc region is controlled by the protective neonatal Fc receptor (FcRn) receptor. To modulate the half-life of engineered antibodies, we have mutated the Fc-FcRn binding site of chimeric anti-carcinoembryonic antigen (CEA) antibodies produced in a single-chain Fv-Fc format. The anti-CEA T84.66 single-chain Fv-Fc format wild-type and five mutants (I253A, H310A, H435Q, H435R, and H310A/H435Q, Kabat numbering system) expressed well in mammalian cell culture. After purification and characterization, effective in vitro antigen binding was shown by competition ELISA. Biodistribution studies in BALB/c mice using (125)I- and (131)I-labeled fragments revealed blood clearance rates from slowest to fastest as follows: wild-type > H435R > H435Q > I253A > H310A > H310A/H435Q. The terminal half-lives of the mutants ranged from 83.4 to 7.96 hours, whereas that of the wild-type was approximately 12 days. Additionally, (124)I-labeled wild-type, H435Q, I253A, H310A, and H310A/H435Q variants were evaluated in LS174T xenografted athymic mice by small animal positron emission tomography imaging, revealing localization to the CEA-positive xenografts. The slow clearing wild-type and H435Q constructs required longer to localize to the tumor and clear from the circulation. The I253A and H310A fragments showed intermediate behavior, whereas the H310A/H435Q variant quickly localized to the tumor site, rapidly cleared from the animal circulation and produced clear images. Thus, attenuating the Fc-FcRn interaction provides a way of controlling the antibody fragment serum half-life without compromising expression and tumor targeting.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Inmunoconjugados/farmacocinética , Fragmentos de Inmunoglobulinas/metabolismo , Radioisótopos de Yodo/farmacocinética , Radiofármacos/farmacocinética , Animales , Antígeno Carcinoembrionario/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Fragmentos de Inmunoglobulinas/biosíntesis , Fragmentos de Inmunoglobulinas/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mieloma Múltiple/metabolismo , Tomografía de Emisión de Positrones , Distribución Tisular , Trasplante Heterólogo
18.
Cancer Res ; 65(13): 5907-16, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15994969

RESUMEN

We have recently described the in vivo properties of an iodinated anti-p185HER2 engineered antibody fragment [minibody (scFv-C(H)3)2; 80 kDa], made from the internalizing 10H8 monoclonal antibody. Although the 10H8 minibody showed excellent binding to the target in vitro, only modest tumor uptake [5.6 +/- 1.7% injected dose per gram (ID/g) of tissue] was achieved in nude mice bearing MCF7/HER2 breast cancer tumors. Here, in an attempt to improve targeting, the 10H8 minibody was conjugated to 1,4,7,10-tetraazacyclododecane-N, N', N'', N'''-tetraacetic acid (DOTA), radiometal labeled, and evaluated in vivo. The tumor uptake of 111In-DOTA 10H8 minibody was 5.7 +/- 0.1% ID/g, similar to the radioiodinated 10H8 minibody. However, in addition to the expected liver clearance, the kidneys had unexpectedly high activity (34.0 +/- 4.0% ID/g). A minibody derived from a second anti-p185(HER2) antibody (trastuzumab; hu4D5v8) was also made. Tumor uptakes, evaluated by quantitative microPET using 64Cu-DOTA hu4D5v8 minibody, were 4.2 +/- 0.5% ID/g. Furthermore, in non-tumor-bearing mice, 111In-DOTA hu4D5v8 minibody exhibited similar elevated uptake in the kidneys (28.4 +/- 6.5% ID/g). Immunohistochemical staining of kidneys from non-tumor-bearing mice showed strong specific staining of the proximal tubules, and Western blot analysis of kidney lysate confirmed the presence of cross-reactive antigen. To further improve tumor uptake and normal tissue distribution, a larger hu4D5v8 fragment [(scFv-C(H)2-C(H)3)2; 105 kDa] was made, engineered to exhibit rapid clearance kinetics. This fragment, when evaluated by microPET, exhibited improved tumor targeting (12.2 +/- 2.4% ID/g) and reduced kidney uptake (13.1 +/- 1.5% ID/g). Thus, by manipulating the size and format of anti-p185(HER2) antibody fragments, the kidney activity was reduced and high or low expression of p185HER2 in xenografts could be distinguished by microPET imaging.


Asunto(s)
Inmunoconjugados/farmacocinética , Fragmentos de Inmunoglobulinas/metabolismo , Neoplasias/diagnóstico por imagen , Receptor ErbB-2/inmunología , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Antígeno Carcinoembrionario/inmunología , Radioisótopos de Cobre/farmacocinética , Femenino , Humanos , Inmunoconjugados/química , Fragmentos de Inmunoglobulinas/química , Radioisótopos de Indio/química , Radioisótopos de Indio/farmacocinética , Riñón/diagnóstico por imagen , Riñón/inmunología , Riñón/metabolismo , Ratones , Ratones Desnudos , Neoplasias/inmunología , Neoplasias/metabolismo , Tomografía de Emisión de Positrones , Radiofármacos/química , Radiofármacos/farmacocinética , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Distribución Tisular
19.
Cancer Biother Radiopharm ; 32(7): 258-265, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28910150

RESUMEN

OBJECTIVES: Report the feasibility, toxicities, and long-term results of a Phase I/II trial of 90Y-labeled anticarcinoembryonic antigen (anti-CEA) (cT84.66) radioimmunotherapy (RIT), gemcitabine, and hepatic arterial infusion (HAI) of fluorodeoxyuridine (FUdR) after maximal hepatic resection of metastatic colorectal cancer to the liver. METHODS: Patients with metastatic colorectal cancer to the liver postresection or ablation to minimum disease were eligible. Each cohort received HAI of FUdR for 14 days on a dose escalation schedule. The maximum HAI FUdR dose level planned was 0.2 mg/kg/day, which is the standard dose for HAI FUdR alone. On day 9, 90Y-cT84.66 anti-CEA at 16.6 mCi/m2 as an i.v. bolus infusion and on days 9-11 i.v. gemcitabine at 105 mg/m2 were given. Patients could receive up to three cycles every 6 weeks of protocol therapy. Four additional cycles of HAI FUdR were allowed after RIT. RESULTS: Sixteen patients were treated on this study. A maximum tolerated dose of 0.20 mg/kg/day of HAI FUdR combined with RIT at 16.6 mCi/m2 and gemcitabine at 105 mg/m2 was achieved with only 1 patient experiencing grade 3 reversible toxicity (mucositis). After surgery, 10 patients had no evidence of visible disease and remained without evidence of disease after completion of protocol therapy. The remaining 6 patients demonstrated radiological visible disease after surgery and after protocol therapy 2 patients had a CR, 1 patient had PR, 2 had stable disease, and 1 had progression. With a median follow-up of 41.8 months (18.7-114.6), median progression free survival was 9.6 months. Two patients demonstrated long-term disease control out to 45+ and 113+ months. CONCLUSION: This study demonstrates the safety, feasibility, and potential utility of HAI FUdR, RIT, and systemic gemcitabine. The trimodality approach does not have higher hematologic toxicities than seen in prior RIT-alone studies. Future efforts evaluating RIT in colorectal cancer should integrate RIT with systemic and regional therapies in the minimal tumor burden setting.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno Carcinoembrionario/uso terapéutico , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Adulto , Anciano , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Floxuridina/uso terapéutico , Humanos , Infusiones Intraarteriales/métodos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Radioinmunoterapia/métodos , Gemcitabina
20.
J Clin Oncol ; 23(21): 4669-78, 2005 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-16034043

RESUMEN

PURPOSE: Hodgkin's lymphoma (HL) has been demonstrated to be a good target for immunotherapy since lymphocyte activation markers such as CD30 are expressed in high numbers on the malignant cells. Thus, we developed a new radioimmunoconjugate consisting of the murine anti-CD30 monoclonal antibody (MAb) Ki-4 labeled with iodine-131 ((131)I). PATIENTS AND METHODS: The biodistribution of (131)I-Ki-4 was assessed via dosimetry after preinfusion of 5 mg native Ki-4 followed by 250 to 300 MBq (131)I-labeled Ki-4. Whole-body scintigraphy was performed 1 hour, 24 hours, 48 hours, 72 hours, and 6 days after the infusion. Dosimetry was calculated using the programs NucliDose ICON-IDL (version 5.0.2; Siemens, Erlanger, Germany) and MIRDOSE (version 3.1; Oak Ridge National Laboratories; Oak Ridge, TN). The therapeutic dose was given on day 8 after preinfusion of unlabeled Ki-4. RESULTS: We treated 22 patients with relapsed or refractory CD30-positive HL. Preinfusion of 5 mg native Ki-4 was sufficient to bind the soluble CD30. Imaging demonstrated localization of involved areas measuring 5 cm in diameter or more in four patients and 2.5 cm in one patient. Patients received total body doses of 0.035 Gy to 0.99 Gy. Acute toxicity was mild with grade 1 fatigue in 19 of 22 assessable patients. Seven patients experienced grade 4 degrees hematotoxicity 4 to 8 weeks after treatment. Response included one complete remission, five partial remissions, and three minor responses. CONCLUSION: Treatment with (131)I-Ki-4 is effective but can be associated with severe hematotoxicity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad de Hodgkin/terapia , Inmunoconjugados/uso terapéutico , Inmunoterapia/métodos , Antígeno Ki-1/inmunología , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/toxicidad , Humanos , Radioisótopos de Yodo , Ratones , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA