Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Semin Nucl Med ; 42(3): 147-50, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22475423

RESUMEN

Although the term has been coined recently, the concepts underlying theranosis have been applied in patient care for more than one-half century. However, advanced technologies are used now. Theranosis describes processes used to tailor therapy for a patient. It is the use of diagnostic tests to identify those patients better-suited for a drug (or drugs) or to determine how well a drug is working. (131)I-iodide for imaging and for therapy of hyperthyroidism and thyroid cancer is an excellent example of personalized theranosis and has withstood challenge for more than 50 years. Radioimmunotherapy for non-Hodgkin lymphoma is a more recent example of theranosis. Either of 2 anti-CD20 monoclonal antibodies, one labeled with indium for imaging or (90)Y for radiotherapy or a second labeled with (131)I for both imaging and radiotherapy, is used for salvage and first-line therapy of multifocal non-Hodgkin lymphoma. The efficacy of these drugs is greater than that of alternative therapies. To mimic the molecular specificity and cell selectivity of a monoclonal antibody, smaller molecules that also bind to proteins upregulated by malignant cells can be used to transport cytotoxic agents to the malignant cells. Smaller carrier molecules like peptides, aptamers, affibodies, and selective, high-affinity ligands facilitate intensification of therapy because of their size. Personalized genomics, proteomics, and molecular imaging are among technologies currently used for theranosis. Molecular emission tomographic imaging with radiolabeled drugs has been used to examine the pharmacology of anticancer therapies and their effectiveness. Increased glycolysis, a molecular phenotype of many malignancies, can be imaged using (18)F-fluoro-2-deoxyglucose (FDG). Tomographic imaging using FDG allows stratification of patients into those responding and likely to respond to the therapy and those better treated in another manner. Prediction of therapeutic response avoids useless therapy so that FDG imaging is included in official response evaluation criteria. Although a fixed approach to therapy may be more practical, an individualized approach is more likely to ensure that each patient receives an effective drug and drug dose that has acceptable and definable tissue effects. Drugs that work in one individual may be ineffective or cause adverse events in others.


Asunto(s)
Linfoma no Hodgkin/diagnóstico , Linfoma no Hodgkin/terapia , Humanos , Radioisótopos de Yodo/uso terapéutico , Linfoma no Hodgkin/radioterapia , Medicina de Precisión
4.
Cancer Biother Radiopharm ; 23(4): 383-97, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18771343

RESUMEN

The cytocidal potency of a molecule can be augmented by conjugating a radionuclide for molecular targeted radionuclide therapy (MTRT) for cancer. Radioimmunotherapy (RIT) should be incorporated into the management of patients with B-cell non-Hodgkin's lymphoma (NHL) soon after the patients have proven incurable. Better drugs, strategies, and combinations with other drugs seem certain to make RIT integral to the management of patients with NHL and likely to lead to a cure of the currently incurable NHL. These improved drugs, strategies, and combinations thereof also offer opportunities for RIT to become part of the management of solid malignancies, including epithelial cancers. Smaller radionuclide carriers, such as those used for pretargeted strategies, provide dose intensification. The potential of pretargeted RIT to improve patient outcomes is striking.


Asunto(s)
Linfoma/radioterapia , Neoplasias/radioterapia , Radioterapia/métodos , Animales , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Relación Dosis-Respuesta en la Radiación , Trasplante de Células Madre Hematopoyéticas , Humanos , Linfoma/terapia , Linfoma no Hodgkin/radioterapia , Linfoma no Hodgkin/terapia , Neoplasias/terapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioinmunoterapia/efectos adversos , Radioinmunoterapia/métodos , Radioterapia/efectos adversos , Dosificación Radioterapéutica , Terapia Recuperativa/métodos
5.
Bioorg Med Chem Lett ; 18(17): 4802-5, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18701282

RESUMEN

Radioimmunotherapy (RIT) is a method for selectively delivering radionuclides to cancer cells while reducing the radiation dose to normal tissues. However, because of slow clearance of MAbs, normal tissues also received radiotoxicity. One of the promising strategies is linking on-demand cleavable (ODC) peptides between radiometal chelates and the tumor targeting agents. We have tested this proof-of-concept by using ODC peptides that are designed to be cleaved only by TNKase and are resistant to cleavage by enzymes present in the plasma and the tumor. TNKase-specific peptide linkers using l- and d-amino acids were screened by OBOC combinatorial peptide libraries. One of the best peptides was linked to radiometal chelate and ChL6-MAb to prepare radioimmunoconjugate (RIC). Optimization and characterization of the linker conjugation to MAb show (a) 1-2 peptides linked to each MAb; (b) immunoreactivity >80%; (c) specific activity of the RIC 0.7-1 microCi/microg; (d) RIC stable over 7 days in human plasma; and (e) radiometal-chelated ODC peptide cleaved from the RIC in plasma by TNKase at clinical dose levels of 10 microg/ml. The percent release of radiochelate from RIC was 50% at 24h and 85% over 7 2h in vitro. This novel ODC-linked RIC could be a potential molecule for RIT.


Asunto(s)
Inmunoconjugados/uso terapéutico , Péptidos/uso terapéutico , Radioinmunoterapia , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Humanos , Hidrólisis , Ratones , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/radioterapia , Péptidos/química , Péptidos/metabolismo , Radioinmunoterapia/métodos , Tenecteplasa , Activador de Tejido Plasminógeno/metabolismo , Células Tumorales Cultivadas
6.
Med Phys ; 35(7): 3062-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18697529

RESUMEN

Targeted radionuclide therapy (TRT) seeks molecular and functional targets within patient tumor sites. A number of agents have been constructed and labeled with beta, alpha, and Auger emitters. Radionuclide carriers spanning a broad range of sizes; e.g., antibodies, liposomes, and constructs such as nanoparticles have been used in these studies. Uptake, in percent-injected dose per gram of malignant tissue, is used to evaluate the specificity of the targeting vehicle. Lymphoma (B-cell) has been the primary clinical application. Extension to solid tumors will require raising the macroscopic absorbed dose by several-fold over values found in present technology. Methods that may effect such changes include multistep targeting, simultaneous chemotherapy, and external sequestration of the agent. Toxicity has primarily involved red marrow so that marrow replacement can also be used to enhance future TRT treatments. Correlation of toxicities and treatment efficiency has been limited by relatively poor absorbed dose estimates partly because of using standard (phantom) organ sizes. These associations will be improved in the future by obtaining patient-specific organ size and activity data with hybrid SPECT/CT and PET/CT scanners.


Asunto(s)
Neoplasias/terapia , Oncología por Radiación/métodos , Radioinmunoterapia/métodos , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Cámaras gamma , Humanos , Radioisótopos de Yodo/uso terapéutico , Oncología Médica/métodos , Neoplasias/diagnóstico por imagen , Fantasmas de Imagen , Tomografía de Emisión de Positrones/métodos , Radioisótopos/uso terapéutico , Dosificación Radioterapéutica , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada por Rayos X/métodos , Radioisótopos de Itrio/uso terapéutico
7.
Cancer Biother Radiopharm ; 23(2): 265-71, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18454696

RESUMEN

UNLABELLED: Nanoparticle thermotherapy (NPTT) uses monoclonal antibody-linked iron oxide magnetic nanoparticles (bioprobes) for the tumor-specific thermotherapy of cancer by hysteretic heating of the magnetic component of the probes through an externally applied alternating magnetic field (AMF). The present study investigated the effect of NPTT on a human prostate cancer cell line, DU145. The concept of total heat dose (THD) as a measure for NPTT was validated on a cellular level and THD was correlated to cell death in vitro. The study, furthermore, explored the potential enhancement of the NPTT effect through added external beam radiation therapy (EBRT), because both forms of treatment have a different, and potentially complementary, mechanism of causing cell death. METHODS: Using carbodiimide, (111)In-DOTA-ChL6 was conjugated to dextran iron oxide 20-nm particles with polyethylene glycol COOH groups on the surface and purified as (111)In-bioprobes. NPTT and EBRT were applied alone and combined to cells labeled with the bioprobes. Cell response was monitored by measuring lactate dehydrogenase (LDH), a product of cytolysis, in the medium. This distinct focus on the response to NPTT was possible, since we found in previous studies that the LDH assay was relatively insensitive to the response of cells (without bioprobes) to EBRT in the dose levels given here. RESULTS: NPTT showed a significantly increased cell death at a total calculated heat dose of 14.51 and 29.02 J/g cells (50% and 100% AMF duty, 350 Oe, 136 kHz, 12 cycles, 20 minutes total), compared with AMF exposure in the absence of bioprobes. Adding EBRT to NPTT did not increase cell death, as measured by LDH. However, EBRT given to cells labeled with bioprobes caused significant cell death at radiation doses of 10 Gy and higher. CONCLUSIONS: In human prostate cancer cell cultures, NPTT applied as a single modality caused cell death that correlated with THD estimation; complete cell death occurred at 14.51 J/g cells. Consequently, enhancement of the NPTT effect through the addition of EBRT could not be addressed. Interestingly, EBRT induced cell death on bioprobe-labeled cells at EBRT levels that did not show cell death in the absence of bioprobes; this phenomenon is worth investigating further.


Asunto(s)
Nanopartículas , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Humanos , Hipertermia Inducida , Lactato Deshidrogenasas/metabolismo , Masculino , Neoplasias de la Próstata/enzimología
8.
Cancer Biother Radiopharm ; 23(1): 82-91, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18298332

RESUMEN

BACKGROUND: Noninvasive, focused hyperthermia can be achieved by using an externally applied alternating magnetic field (AMF) if effective concentrations of nanoparticles can be delivered to the target cancer cells. Targeting agents, for example, monoclonal antibodies or peptides, linked to magnetic iron oxide nanoparticles (NP), represent a promising strategy to target cancer cells and hyperthermia. METHODS: We have developed a new radioconjugate NP ((111)In-DOTA-di-scFv-NP), using recombinantly generated antibody fragments, di-scFv-c, for the imaging and therapy of anti-MUC-1-expressing cancers, because aberrant MUC-1 is abundantly expressed on the majority of human epithelial cancers. Anti-MUC-1 di-scFv-c (50 kDa) were engineered, generated, and selected to link maleimide functionalized nanoparticles (NP-M). DOTA chelate was conjugated with di-scFv-c for radionuclide chelation to trace the radioimmunonanoparticles (RINPs) in vivo. RESULTS: Heat-inducing NP-M were prepared with maleimide density >15 per particle for site-specific thiolation. The specific activity of the RINP was 4-5 microCi (111)In/mg with >10 molecules of di-scFv per NP. We characterized the RINP by polyacrylamide gel electrophoresis, cellulose acetate electrophoresis, size-exclusion chromatography, and tumor-cell binding. RINP had a >90% di-scFv conjugated to NP and an immunoreactivity >80% relative to unmodified di-scFv-c on HBT 3477 and DU145 tumor cells. Pharmacokinetics and whole-body autoradiography studies demonstrated that a 5% injected dose was targeted in tumor after 24 hours. CONCLUSIONS: Further development of this new preparation of RINP may provide uniquely high tumor-targeting NP for AMF-driven tumor hyperthermia with less spleen and kidney accumulation.


Asunto(s)
Mucina-1/metabolismo , Nanopartículas/uso terapéutico , Neoplasias/diagnóstico por imagen , Neoplasias/radioterapia , Radioinmunoterapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Western Blotting , Células COS , Chlorocebus aethiops , Femenino , Compuestos Férricos/química , Compuestos Férricos/metabolismo , Calor , Humanos , Región Variable de Inmunoglobulina/inmunología , Radioisótopos de Indio/farmacocinética , Radioisótopos de Indio/uso terapéutico , Magnetismo , Maleimidas/química , Maleimidas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mucina-1/genética , Mucina-1/inmunología , Neoplasias/inmunología , Radiografía , Radioinmunodetección , Distribución Tisular , Células Tumorales Cultivadas/metabolismo
9.
Cancer Biother Radiopharm ; 23(6): 671-80, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20443694

RESUMEN

The promise of hyperthermia has yet to be realized, but the fundamental idea and the effects of heat on (cancer) cells are well known. Cell death from exposure to heat is a function of both the intensity of the heat and the length of the exposure. Cells die by necrosis and by apoptosis. Sublethal heat doses sensitize cancer cells to radiation and drugs. Because of advances in chemistry and physics, harnessing the power of heat to kill cancer cells seems achievable now! Using novel systems embodied in the combination of molecular-targeted nanoparticles and hysteretic heating of the nanoparticles with "focused" alternating magnetic frequencies (AMFs), heat delivery can be better controlled. Importantly, hyperthermia does not damage, and may actually enhance, the immune system. Trials in patients are needed to settle the clinical role of new thermal treatment.


Asunto(s)
Hipertermia Inducida , Neoplasias/terapia , Animales , Calor , Humanos
10.
Cancer Biother Radiopharm ; 23(6): 783-96, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20443696

RESUMEN

To mimic the molecular specificity and cell selectivity of monoclonal antibody (mAb) binding while decreasing size, nanomolecules (selective high-affinity ligands; SHALs), based on in silico modeling, have been created to bind to human leukocyte antigen-DR (HLA-DR10), a signaling receptor protein upregulated on the malignant B-lymphocytes of non-Hodgkin's lymphoma and chronic lymphocytic leukemia. SHALs were synthesized with a biotin or DOTA chelate (1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid), using a solid-phase lysine-polyethyleneglycol backbone to link sets of ligands shown previously to bind to HLA-DR10. Using cell-binding and death assays and confocal microscopy, SHAL uptake, residualization, and cytocidal activity were evaluated in HLA-DR10 expressing and nonexpressing live, human lymphoma cell lines. All of the SHALs tested were selective for, and accumulated in, expressing cells. Reflecting binding to HLA-DR10 inside the cells, SHALs having the Ct ligand (3-(2-([3-chloro-5-trifluoromethyl)-2-pyridinyl]oxy)-anilino)-3-oxopropanionic acid) residualized in expressing cells greater than 179 times more than accountable by cell-surface membrane HLA-DR10. Confocal microscopy confirmed the intracellular residualization of these SHALs. Importantly, SHALs with a Ct ligand had direct cytocidal activity, similar in potency to that of Lym-1 mAb and rituximab, selectively for HLA-DR10 expressing lymphoma cells and xenografts. The results show that SHALs containing the Ct ligand residualize intracellularly and have cytocidal effects mediated by HLA-DR10. These SHALs have extraordinary potential as novel molecules for the selective targeting of lymphoma and leukemia for molecular therapy and imaging. Further, these SHALs can be used to transport and residualize cytotoxic agents near critical sites inside these malignant cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Materiales Biomiméticos/farmacología , Antígenos HLA-DR/inmunología , Leucemia/terapia , Linfoma/terapia , Imitación Molecular/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino , Materiales Biomiméticos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Subtipos Serológicos HLA-DR , Humanos , Radioisótopos de Indio , Leucemia/inmunología , Linfoma/inmunología , Nanopartículas , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Radiofármacos , Rituximab , Espectrometría de Masa por Ionización de Electrospray , Distribución Tisular
12.
Int J Oncol ; 31(4): 729-40, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17786303

RESUMEN

Despite their large size, antibodies have proven to be suitable radioisotope carriers to deliver systemic radiotherapy, often molecular image-based, for lymphoma and leukemia. To mimic antibody (Ab) targeting behavior while decreasing size by 50-100x, a combination of computational and experimental methods were used to generate molecules that bind to unique sites within the HLA-DR epitopic region of Lym-1, an Ab shown effective in patients. Lym-1 Ab mimics (synthetic high afinity ligands; SHALs) were generated and studied in vitro, using live cell binding assays, and/or pharmacokinetic studies over 24 h in xenografted mice given 1 or 20 microg SHAL doses i.v. Multimilligram amounts of each of the dimeric (bis) SHALs were synthesized at high purity, and labeled with indium-111 at high specific activity and purity. These SHALs were selective for HLA-DR and HLA-DR expressing malignant cells and had functional affinities that ranged from 10(-9) M (nanomolar) to 10(-10) M. Blood clearances ranged from 3.6 to 9.5 h and body clearances ranged from 15.2 to 43.0 h for the 6 bis DOTA-SHALs studied in a mouse model for non-Hodgkin's lymphoma (NHL). While localization was shown in Raji NHL xenografts, biodistribution was influenced by 'sinks' for individual ligands of the SHALs. Highly pure, dimeric mimics for HLA-DR Ab were synthesized, biotinylated and radiolabeled, and showed selectivity in vitro. Pharmacokinetic behavior in mice was influenced by the ligands and by the linker length of the dimeric SHALs. Nanomolar or better functional affinity was observed when a suitably long linker was used to connect the two bidentate SHALs. The concept and methodology are of interest because applicable for targeting most proteins; the SHAL synthetic platform is highly efficient and adaptive.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos HLA-DR/inmunología , Linfoma no Hodgkin/diagnóstico por imagen , Imitación Molecular , Fragmentos de Péptidos/inmunología , Radiofármacos/farmacocinética , Animales , Anticuerpos Monoclonales de Origen Murino , Ensayo de Inmunoadsorción Enzimática , Femenino , Antígenos HLA-DR/metabolismo , Subtipos Serológicos HLA-DR , Humanos , Ligandos , Linfoma no Hodgkin/radioterapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/metabolismo , Radioinmunoterapia , Cintigrafía , Trasplante Heterólogo
13.
Cancer Biother Radiopharm ; 22(4): 521-30, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17803447

RESUMEN

Anti-CD20 monoclonal antibodies (mAbs), unlabeled rituximab (Rituxan, Biogen Idec Inc., Cambridge, MA; and Genentech Inc., South San Francisco, CA) or radiolabeled 90Y-ibritumomab (Zevalin, Biogen Idec Inc., Cambridge, MA) and 131I-tositumomab (Bexxar; Glaxo Smith Kline, Research Triangle Park, NC), have proven to be effective therapy for non-Hodgkin's lymphoma (NHL), but also induce immediate and persistent decreases in normal peripheral blood lymphocytes (PBLs). Lym-1, a mAb that selectively targets malignant lymphocytes, also has induced therapeutic responses and prolonged survival in patients with NHL when labeled with iodine-131 (131I). We have retrospectively examined its effect on PBLs in 41 NHL patients that had received 131I-Lym-1 therapy. Absolute lymphocyte counts (ALCs) were evaluated before and after the first and last 131I-Lym-1 infusion. Modest decreases in PBLs were observed in most of the patients. Using strict criteria to define recovery, time to recovery was determined for 19 patients, with the remainder censored because of insufficient follow-up (median follow up for censored patients: 22 days). Using Kaplan-Meier estimates, it would be predicted that 31% of patients would recover by 28 days and that median time to recovery would be 44 days after the last 131I-Lym-1 infusion. No predictors were found for time to recovery, considering such factors as the administered Lym-1 or 131I dose, spleen volume, or radiation doses to the body, marrow, or spleen. The data suggest that the effect of 131I-Lym-1 on ALC is the result of a nonspecific radiation effect, rather than a specific Lym-1 mAb effect. The shorter time required for ALC recovery after 131I-Lym-1 when compared to that reported for anti-CD20 mAbs, whether radiolabeled or otherwise, is probably related to differing mechanisms for lymphocytotoxicity and lesser Lym-1 antigenic density on normal B-lymphocytes.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Linfocitos/inmunología , Linfocitos/patología , Linfoma no Hodgkin/patología , Linfoma no Hodgkin/radioterapia , Adulto , Anciano , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales de Origen Murino , Anticuerpos Antineoplásicos/efectos adversos , Anticuerpos Antineoplásicos/inmunología , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Estimación de Kaplan-Meier , Linfoma no Hodgkin/inmunología , Masculino , Persona de Mediana Edad , Radioinmunoterapia
14.
Cancer Biother Radiopharm ; 22(4): 531-42, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17803448

RESUMEN

Selective high-affinity ligands (SHALs) were selected as substitutes for monoclonal antibodies (mAbs) to deliver radioisotopes to malignant tumors. Because a SHAL (5 KD) is considerably smaller in comparison to an antibody (150 KD), a significant therapeutic index (TI) enhancement for radioimmunotherapy (RIT) is anticipated. The antibody-antigen (Ab-Ag) model system chosen for the development of SHALs consists of Lym-1, a MAb with proven selectivity in non-Hodgkin's lymphoma (NHL) patients and its well-characterized Ag, the beta subunit of HLA DR10. Whereas Lym-1 is readily available, the subunit of HLA-DR10 is not. Native, heterodimeric (alpha and beta subunits) HLA-DR10 can be purified from Raji cells, which are known to overexpress this Ag. Inconsistent homogeneity between preparations of HLA-DR10 solubilized in the presence of detergents prompted us to express a recombinant form of the beta subunit of HLA-DR10 in Escherichia coli. Negligible production yields (

Asunto(s)
Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Linfoma/inmunología , Linfoma/metabolismo , Radioinmunoterapia/métodos , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino , Línea Celular Tumoral , Dicroismo Circular , Clonación Molecular , Epítopos/inmunología , Escherichia coli , Expresión Génica , Antígenos HLA-DR/química , Antígenos HLA-DR/inmunología , Subtipos Serológicos HLA-DR , Humanos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Sensibilidad y Especificidad , Homología Estructural de Proteína , Especificidad por Sustrato , Resonancia por Plasmón de Superficie
15.
Cancer Biother Radiopharm ; 22(4): 556-63, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17803451

RESUMEN

UNLABELLED: Protease degradable linkers have been proposed to improve the therapeutic index (TI) (i.e., tumor to normal tissue) of molecular targeted radioisotope therapy by reducing unbound radiotargeting agent in the blood and other normal tissues. If the radioisotope is detached from the circulating targeting agent once the radioisotope level in the tumors has been maximized, the success of this system depends on the ability to anticipate a preferred intervention time that will lead to significantly improved TIs. This paper presents a method to predict preferred intervention times and TIs by using pharmacokinetic tracer studies carried out without intervention. METHODS: Pharmacokinetic data for the blood and tumors from tracer doses of 111In-labeled chimeric and mouse monoclonal antibodies in patients and in mice were used as surrogates for corresponding 90Y radioimmunoconjugates. Data were fit with simple pharmacokinetic functions. A set of formulas was then developed to estimate the improvement in therapeutic index and the preferred intervention time, using simple modeling assumptions. RESULTS: A modeled introduction of enzymatic cleavable linkers resulted in an increase in the tumor-to-blood TI by a factor of 3.2-1.6 for the systems analyzed. As expected, the preferred intervention times varied depending on the pharmacokinetic data, but could be predicted based on a priori knowledge of the actual or anticipated pharmacokinetics in the absence of intervention. CONCLUSIONS: These results highlight the potential value of cleavable linkers in substantially increasing the TI, and provide an approach for estimating a preferred intervention time, using actual or predicted pharmacokinetic data obtained without intervention.


Asunto(s)
Vasos Sanguíneos/enzimología , Radioisótopos de Indio/metabolismo , Radioisótopos de Indio/farmacocinética , Péptidos/metabolismo , Radioisótopos de Itrio/metabolismo , Radioisótopos de Itrio/farmacocinética , Animales , Línea Celular Tumoral , Humanos , Radioisótopos de Indio/química , Radioisótopos de Indio/uso terapéutico , Ratones , Modelos Animales , Modelos Biológicos , Estudios Retrospectivos , Factores de Tiempo , Radioisótopos de Itrio/química , Radioisótopos de Itrio/uso terapéutico
16.
J Nucl Med ; 48(8): 1338-47, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17631545

RESUMEN

UNLABELLED: Despite their large size, antibodies (Abs) are suitable carriers to deliver systemic radiotherapy, often molecular image-based, for lymphoma and leukemia. Lym-1 Ab has proven to be an effective radioisotope carrier, even in small amounts, for targeting human leukocyte antigen DR (HLA-DR), a surface membrane protein overexpressed on B-cell lymphoma. Pairs of molecules (referred to as ligands), shown by computational and experimental methods to bind to each of 2 sites within the Lym-1 epitopic region, have been linked to generate small (<2 kDa) molecules (referred to as selective high-affinity ligands [SHALs]) to mimic the targeting properties of Lym-1 Ab. METHODS: A lysine-polyethylene glycol (PEG) backbone was used to synthetically link 2 of the following ligands: deoxycholate, 5-leuenkephalin, triiodothyronine, thyronine, dabsyl-L-valine, and N-benzoyl-L-arginyl-4-amino-benzoic acid to generate a series of 13 bidentate SHALs with a biotin or 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) chelate attached to the linker. These SHALs have been assessed for their selectivity in binding to HLA-DR10-expressing cells and for their pharmacokinetics and tissue biodistribution in mice. Biotinylated versions of these SHALs discriminated cell lines positive for HLA-DR10 expression with near-nanomolar affinity. The DOTA versions of 4 SHALs were labeled with (111)In for pharmacokinetic studies in mice with HLA-DR10-expressing malignant Raji xenografts. RESULTS: The bidentate, biotinylated, and DOTA-SHALs were synthesized in high-purity, multimilligram amounts. Mean radiochemical and product yields and purities were 90%, 75%, and 90% at mean specific activities of 3.9 MBq/microg (105 microCi/microg) for the (111)In-labeled SHALs. As expected, rapid blood clearance and tumor targeting were observed. The pharmacokinetics of the SHALs was influenced by the component ligands. Biliary clearance, kidney localization, and serum receptor binding contributed to less favorable tumor targeting. CONCLUSION: A series of SHALs was readily synthesized in multimilligram amounts and showed the expected selective binding in vitro. Better selection of the SHAL components should provide second-generation SHALs with improved properties to fulfill the substantial potential of these novel molecular carriers for targeting.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos HLA-DR/inmunología , Linfoma no Hodgkin/diagnóstico por imagen , Radiofármacos/farmacocinética , Animales , Anticuerpos Monoclonales de Origen Murino , Biotinilación , Femenino , Humanos , Ligandos , Linfoma no Hodgkin/radioterapia , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Tomografía de Emisión de Positrones , Radiofármacos/síntesis química , Trasplante Heterólogo
17.
Bioconjug Chem ; 18(3): 912-21, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17373772

RESUMEN

Selective high-affinity ligands (SHALs) were synthesized as molecular targeting agents for HLA-DR10, a cell surface receptor upregulated on malignant B-cell lymphocytes in non-Hodgkin's lymphoma and leukemia. SHALs are designed to mimic the affinity and selectivity of Lym-1, an antibody that binds to the beta-subunit of HLA-DR10. To bind selectively to HLA-DR10, SHALs were constructed to bind to two adjacent pockets on the surface of the beta-subunit of HLA-DR10 located within an epitope recognized by the Lym-1 antibody. A series of multivalent SHALs with molecular masses of 1500-3000 Da were synthesized using solid/polymer-supported synthesis on chlorotrityl chloride resin in 50-80% yield. To enable their use as radionuclide carriers in mouse studies, SHALs were conjugated to DOTA in a solution-phase reaction with 70-100% yield. 57Co/CoCl2 titrations revealed that 50-60% of the DOTA in the DOTA-conjugated SHALs was available for radiometal chelation. These DOTA-SHALs were labeled with 111In and used to carry out pharmacokinetic studies in mice. Radiolabeling reactions of DOTA-SHALs, with exactly one DOTA entity per targeting SHAL molecule, yielded products with greater than 90% radiochemical purity and specific activities ranging from 97 to 150 muCi/mug.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos HLA-DR/inmunología , Radioisótopos de Indio/química , Leucemia/inmunología , Linfoma no Hodgkin/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino , Linfocitos B/inmunología , Subtipos Serológicos HLA-DR , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Ligandos , Ratones
18.
J Nucl Med ; 48(3): 437-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17332622

RESUMEN

UNLABELLED: Antibody (mAb)-linked iron oxide nanoparticles (bioprobes) provide the opportunity to develop tumor specific thermal therapy (Rx) for metastatic cancer when inductively heated by an externally applied alternating magnetic field (AMF). To evaluate the potential of this Rx, in vivo tumor targeting, efficacy, and predictive radionuclide-based heat dosimetry were studied using (111)In-ChL6 bioprobes (ChL6 is chimeric L6) in a human breast cancer xenograft model. METHODS: Using carbodiimide, (111)In-DOTA-ChL6 (DOTA is dodecanetetraacetic acid) was conjugated to polyethylene glycol-iron oxide-impregnated dextran 20-nm particles and purified as (111)In-bioprobes. (111)In doses of 740-1,110 kBq (20-30 muCi) (2.2 mg of bioprobes) were injected intravenously into mice bearing HBT3477 human breast cancer xenografts. Pharmacokinetic (PK) data were obtained at 1, 2, 3, and 5 d. AMF was delivered 72 h after bioprobe injection at amplitudes of 1,410 (113 kA/m), 1,300 (104 kA/m), and 700 (56 kA/m) oersteds (Oe) at 30%, 60%, and 90% "on" time (duty), respectively, and at 1,050 Oe (84 kA/m) at 50% and 70% duty over the 20-min treatment. Treated and control mice were monitored for 90 d. Tumor total heat dose (THD) from activated tumor bioprobes was calculated for each Rx group using (111)In-bioprobe tumor concentration and premeasured particle heat response to AMF amplitudes. Tumor growth delay was analyzed by Wilcoxon rank sum comparison of time to double, triple, and quintuple tumor volume in each group, and all groups were compared with the controls. RESULTS: Mean tumor concentration of (111)In-bioprobes at 48 h was 14 +/- 2 percentage injected dose per gram; this concentration 24 h before AMF treatment was used to calculate THD. No particle-related toxicity was observed. Toxicity was observed at the highest AMF amplitude-duty combination of 1,300 Oe and 60% over 20 min; 6 of 10 mice died acutely. Tumor growth delay occurred in all of the other groups, correlated with heat dose and, except for the lowest heat dose group, was statistically significant when compared with the untreated group. Electron microscopy showed (111)In-bioprobes on tumor cells and cell death by necrosis at 24 and 48 h after AMF. CONCLUSION: mAb-guided bioprobes (iron oxide nanoparticles) effectively targeted human breast cancer xenografts in mice. THD, calculated using empirically observed (111)In-bioprobe tumor concentration and in vitro nanoparticle heat induction by AMF, correlated with tumor growth delay.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Calor/uso terapéutico , Radioisótopos de Indio/uso terapéutico , Magnetismo , Neoplasias Mamarias Experimentales/terapia , Nanopartículas , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias
19.
Chem Commun (Camb) ; (7): 695-7, 2007 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-17392953

RESUMEN

Heterofunctional azide and alkyne PEG-linkers have been synthesized and site specifically conjugated to scFv via a reactive thiol functionality; two scFv were coupled by copper catalyzed 1,3-dipolar cycloaddition to make divalent scFv (di-scFv) with an inter-scFv distance defined to provide divalent binding; antigen binding was maintained for the di-scFv construct and increased several times compared to that of the parent scFv; the cycloaddition reaction reported herein represents an important ligation strategy to covalently link macromolecular proteins and retain sensitive structural conformations.


Asunto(s)
Región Variable de Inmunoglobulina/química , Alquinos , Reacciones Antígeno-Anticuerpo , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Azidas , Línea Celular Tumoral , Reactivos de Enlaces Cruzados/química , Dimerización , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/química , Región Variable de Inmunoglobulina/uso terapéutico , Inmunohistoquímica , Masculino , Ingeniería de Proteínas
20.
Stat Med ; 26(5): 1114-35, 2007 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-16755532

RESUMEN

For molecularly targeted cancer therapy, potency of a novel monoclonal antibody is typically studied in vitro in order to observe directly the effect of the antibody on a malignant tumour cell population. Numbers of viable cells of the population are counted over time to evaluate how the antibody may change the population growth pattern. Usually, such count data is analysed by a log-linear model to estimate the average antibody effects, but it is based on a Poisson-like process and is not appropriate for modelling the growth pattern of the tumour cells. In this paper, we propose to analyse the count data from the point of view of a birth-and-death process, which is a more natural description of the biological process in these experiments. Assuming a simple birth-and-death process and a naïve regression model for the birth rate and death rate, we show that the log-linear relationship still holds for the average count and the covariates, although the linear predictor needs to satisfy a specific functional form. The estimation can be based on the quasi-likelihood method. The variance is not a simple function of the mean, but can be adequately approximated by a function of mean of either quadratic form or linear form, available in most standard statistical packages. We perform a simulation study to show that the proposed method provides consistent and robust estimations. The utility of the method is demonstrated by the analysis of a data set for experiment for anti-lymphoma monoclonal antibodies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Recuento de Células/estadística & datos numéricos , Neoplasias/tratamiento farmacológico , Muerte Celular , Modelos Lineales , Modelos Estadísticos , Neoplasias/mortalidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA