Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 325
Filtrar
1.
Life Sci ; 289: 120222, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34902436

RESUMO

In order to overcome limitations of conventional cancer therapy methods, immunotoxins with the capability of target-specific action have been designed and evaluated pre-clinically, and some of them are in clinical studies. Targeting cancer cells via antibodies specific for tumour-associated surface proteins is a new biomedical approach that could provide the selectivity that is lacking in conventional cancer therapy methods such as radiotherapy and chemotherapy. A successful example of an approved immunotoxin is represented by immunoRNases. ImmunoRNases are fusion proteins in which the toxin has been replaced by a ribonuclease. Conjugation of RNase molecule to monoclonal antibody or antibody fragment was shown to enhance specific cell-killing by several orders of magnitude, both in vitro and in animal models. There are several RNases obtained from different mammalian cells that are expected to be less immunogenic and systemically toxic. In fact, RNases are pro-toxins which become toxic only upon their internalization in target cells mediated by the antibody moiety. The structure and large size of the antibody molecules assembled with the immunoRNases have always been a challenge in the application of immunoRNases as an antitoxin. To overcome this obstacle, we have offered a new strategy for the application of immunoRNases as a promising approach for upgrading immunoRNAses with maximum affinity and high stability in the cell, which can ultimately act as an effective large-scale cancer treatment. In this review, we introduce the optimized antibody-like molecules with small size, approximately 10 kD, which are presumed to significantly enhance RNase activity and be a suitable agent with the potential for anti-cancer functionality. In addition, we also discuss new molecular entities such as monobody, anticalin, nonobody and affilin as refined versions in the development of immunoRNases. These small molecules express their functionality with the suitable small size as well as with low immunogenicity in the cell, as a part of immunoRNases.


Assuntos
Antineoplásicos Imunológicos , Antineoplásicos , Imunotoxinas , Neoplasias , Proteínas Recombinantes de Fusão , Ribonucleases , Animais , Antineoplásicos/imunologia , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Humanos , Imunotoxinas/genética , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Imunotoxinas/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Ribonucleases/genética , Ribonucleases/imunologia , Ribonucleases/farmacocinética , Ribonucleases/farmacologia
2.
Drug Metab Dispos ; 50(1): 8-16, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34649966

RESUMO

Intratumoral heterogeneity is a leading cause of treatment failure resulting in tumor recurrence. For the antibody-drug conjugate (ADC) ado-trastuzumab emtansine (T-DM1), two major types of resistance include changes in human epidermal growth factor receptor 2 (HER2) expression and reduced payload sensitivity, which is often exacerbated by heterogenous HER2 expression and ADC distribution during treatment. ADCs with bystander payloads, such as trastuzumab-monomethyl auristatin E (T-MMAE), can reach and kill adjacent cells with lower receptor expression that cannot be targeted directly with the ADC. Additionally, coadministration of T-DM1 with its unconjugated antibody, trastuzumab, can improve distribution and minimize heterogeneous delivery. However, the effectiveness of trastuzumab coadministration and ADC bystander killing in heterogenous tumors in reducing the selection of resistant cells is not well understood. Here, we use an agent-based model to predict outcomes with these different regimens. The simulations demonstrate that both T-DM1 and T-MMAE benefit from trastuzumab coadministration for tumors with high average receptor expression (up to 70% and 40% decrease in average tumor volume, respectively), with greater benefit for nonbystander payloads. However, the benefit decreases as receptor expression is reduced, reversing at low concentrations (up to 360% and 430% increase in average tumor volume for T-DM1 and T-MMAE, respectively) for this mechanism that impacts both ADC distribution and efficacy. For tumors with intrinsic payload resistance, coadministration uniformly exhibits better efficacy than ADC monotherapy (50%-70% and 19%-36% decrease in average tumor volume for T-DM1 and T-MMAE, respectively). Finally, we demonstrate that several regimens select for resistant cells at clinical tolerable doses, which highlights the need to pursue other mechanisms of action for durable treatment responses. SIGNIFICANCE STATEMENT: Experimental evidence demonstrates heterogeneity in the distribution of both the antibody-drug conjugate and the target receptor in the tumor microenvironment, which can promote the selection of resistant cells and lead to recurrence. This study quantifies the impact of increasing the antibody dose and utilizing bystander payloads in heterogeneous tumors. Alternative cell-killing mechanisms are needed to avoid enriching resistant cell populations.


Assuntos
Anticorpos Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Imunotoxinas/uso terapêutico , Receptor ErbB-2/genética , Ado-Trastuzumab Emtansina , Aminobenzoatos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados , Imunoterapia , Imunotoxinas/farmacocinética , Modelos Biológicos , Oligopeptídeos/uso terapêutico , Trastuzumab/uso terapêutico , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Front Immunol ; 11: 1261, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32695104

RESUMO

Immunotoxins are cytolytic fusion proteins developed for cancer therapy, composed of an antibody fragment that binds to a cancer cell and a protein toxin fragment that kills the cell. Pseudomonas exotoxin A (PE) is a potent toxin that is used for the killing moiety in many immunotoxins. Moxetumomab Pasudotox (Lumoxiti) contains an anti-CD22 Fv and a 38 kDa portion of PE. Lumoxiti was discovered in the Laboratory of Molecular Biology at the U.S. National Cancer Institute and co-developed with Medimmune/AstraZeneca to treat hairy cell leukemia. In 2018 Lumoxiti was approved by the US Food and Drug Administration for the treatment of drug-resistant Hairy Cell Leukemia. Due to the bacterial origin of the killing moiety, immunotoxins containing PE are highly immunogenic in patients with normal immune systems, but less immunogenic in patients with hematologic malignancies, whose immune systems are often compromised. LMB-100 is a de-immunized variant of the toxin with a humanized antibody that targets mesothelin and a PE toxin that was rationally designed for diminished reactivity with antibodies and B cell receptors. It is now being evaluated in clinical trials for the treatment of mesothelioma and pancreatic cancer and is showing somewhat diminished immunogenicity compared to its un modified parental counterpart. Here we review the immunogenicity of the original and de-immunized PE immunotoxins in mice and patients, the development of anti-drug antibodies (ADAs), their impact on drug availability and their effect on clinical efficacy. Efforts to mitigate the immunogenicity of immunotoxins and its impact on immunogenicity will be described including rational design to identify, remove, or suppress B cell or T cell epitopes, and combination of immunotoxins with immune modulating drugs.


Assuntos
Exotoxinas/imunologia , Imunotoxinas/imunologia , Pseudomonas/imunologia , ADP Ribose Transferases/imunologia , Animais , Formação de Anticorpos/imunologia , Toxinas Bacterianas/imunologia , Ensaios Clínicos como Assunto , Epitopos de Linfócito B/imunologia , Exotoxinas/química , Exotoxinas/farmacocinética , Humanos , Imunoensaio , Imunomodulação/efeitos dos fármacos , Imunotoxinas/química , Imunotoxinas/farmacocinética , Mesotelina , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/imunologia , Relação Estrutura-Atividade , Resultado do Tratamento , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
4.
Mol Cancer Ther ; 19(7): 1385-1394, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32398269

RESUMO

The EphA2 receptor is found at high levels in tumors and low levels in normal tissue and high EphA2 expression in biopsies is a predictor of poor outcome in patients. Drug discovery groups have therefore sought to develop EphA2-based therapies using small molecule, peptide, and nanoparticle-based approaches (1-3). However, until now only EphA2-targeting antibody-drug conjugates (ADC) have entered clinical development. For example, MEDI-547 is an EphA2-targeting ADC that displayed encouraging antitumor activity in preclinical models and progressed to phase I clinical testing in man. Here we describe the development of BT5528, a bicyclic peptide ("Bicycle") conjugated to the auristatin derivative maleimidocaproyl-monomethyl auristatin E to generate the Bicycle toxin conjugate BT5528. The report compares and contrasts the Pharmacokinetics (PK) characteristics of antibody and Bicycle-based targeting systems and discusses how the PK and payload characteristics of different delivery systems impact the efficacy-toxicity trade off which is key to the development of successful cancer therapies. We show that BT5528 gives rise to rapid update into tumors and fast renal elimination followed by persistent toxin levels in tumors without prolonged exposure of parent drug in the vasculature. This fast in, fast out kinetics gave rise to more favorable toxicology findings in rats and monkeys than were observed with MEDI-547 in preclinical and clinical studies.Graphical Abstract: http://mct.aacrjournals.org/content/molcanther/19/7/1385/F1.large.jpg.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Fibrossarcoma/tratamento farmacológico , Oligopeptídeos/química , Peptídeos Cíclicos/farmacologia , Receptor EphA2/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacocinética , Apoptose , Proliferação de Células , Feminino , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Humanos , Imunotoxinas/farmacocinética , Imunotoxinas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oligopeptídeos/administração & dosagem , Peptídeos Cíclicos/farmacocinética , Receptor EphA2/genética , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Proc Natl Acad Sci U S A ; 117(11): 6086-6091, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123080

RESUMO

Recombinant immunotoxins (RITs) are chimeric proteins composed of an Fv and a protein toxin being developed for cancer treatment. The Fv brings the toxin to the cancer cell, but most of the RITs do not reach the tumor and are removed by other organs. To identify cells responsible for RIT removal, and the pathway by which RITs reach these cells, we studied SS1P, a 63-kDa RIT that targets mesothelin-expressing tumors and has a short serum half-life. The major organs that remove RIT were identified by live mouse imaging of RIT labeled with FNIR-Z-759. Cells responsible for SS1P removal were identified by immunohistochemistry and intravital two-photon microscopy of kidneys of rats. The primary organ of SS1P removal is kidney followed by liver. In the kidney, SS1P passes through the glomerulus, is taken up by proximal tubular cells, and transferred to lysosomes. In the liver, macrophages are involved in removal. The short half-life of SS1P is due to its very rapid filtration by the kidney followed by degradation in proximal tubular cells of the kidney. In mice treated with SS1P, proximal tubular cells are damaged and albumin in the urine is increased. SS1P uptake by kidney is reduced by coadministration of l-lysine. Our data suggests that l-lysine administration to humans might prevent SS1P-mediated kidney damage, reduce albumin loss in urine, and alleviate capillary leak syndrome.


Assuntos
Albuminúria/patologia , Anticorpos Monoclonais/farmacocinética , Síndrome de Vazamento Capilar/patologia , Imunotoxinas/farmacocinética , Túbulos Renais Proximais/efeitos dos fármacos , Albuminúria/induzido quimicamente , Albuminúria/prevenção & controle , Albuminúria/urina , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Anticorpos Monoclonais/toxicidade , Síndrome de Vazamento Capilar/induzido quimicamente , Síndrome de Vazamento Capilar/prevenção & controle , Síndrome de Vazamento Capilar/urina , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes/química , Meia-Vida , Humanos , Imunotoxinas/administração & dosagem , Imunotoxinas/química , Imunotoxinas/toxicidade , Microscopia Intravital , Glomérulos Renais/metabolismo , Túbulos Renais Proximais/diagnóstico por imagem , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Lisina/administração & dosagem , Mesotelina , Camundongos , Microscopia de Fluorescência , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/toxicidade , Eliminação Renal/efeitos dos fármacos , Albumina Sérica/análise , Albumina Sérica/metabolismo , Coloração e Rotulagem
6.
Proc Natl Acad Sci U S A ; 115(15): E3501-E3508, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581296

RESUMO

Recombinant immunotoxins (RITs) are chimeric proteins consisting of a Fv that binds to a cancer cell and a portion of a protein toxin. One of these, Moxetumomab pasudotox, was shown to be effective in treating patients with some leukemias, where the cells are readily accessible to the RIT. However, their short half-life limits their efficacy in solid tumors, because penetration into the tumors is slow. Albumin and agents bound to albumin have a long half-life in the circulation. To increase the time tumor cells are exposed to RITs, we have produced and evaluated variants that contain either an albumin-binding domain (ABD) from Streptococcus or single-domain antibodies from Llama. We have inserted these ABDs into RITs targeting mesothelin, between the Fv and the furin cleavage site. We find that these proteins can be produced in large amounts, are very cytotoxic to mesothelin-expressing cancer cell lines, and have a high affinity for human or mouse serum albumin. In mice, the RIT containing an ABD from Streptococcus has a longer half-life and higher antitumor activity than the other two. Its half-life in the circulation of mice ranges from 113 to 194 min compared with 13 min for an RIT with no ABD. Cell uptake studies show the RIT enters the target cell bound to serum albumin. We conclude that RITs with improved half-lives and antitumor activity should be evaluated for the treatment of cancer in humans.


Assuntos
Imunotoxinas/farmacocinética , Animais , Toxinas Bacterianas/farmacocinética , Toxinas Bacterianas/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Modelos Animais de Doenças , Exotoxinas/farmacocinética , Exotoxinas/farmacologia , Feminino , Proteínas Ligadas por GPI/efeitos dos fármacos , Meia-Vida , Humanos , Imunotoxinas/imunologia , Leucemia/tratamento farmacológico , Mesotelina , Camundongos , Camundongos Nus , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Albumina Sérica/metabolismo , Albumina Sérica/uso terapêutico
7.
Blood ; 130(14): 1620-1627, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28983018

RESUMO

Novel therapies are needed to overcome chemotherapy resistance for children with relapsed/refractory acute lymphoblastic leukemia (ALL). Moxetumomab pasudotox is a recombinant anti-CD22 immunotoxin. A multicenter phase 1 study was conducted to determine the maximum-tolerated cumulative dose (MTCD) and evaluate safety, activity, pharmacokinetics, and immunogenicity of moxetumomab pasudotox in children, adolescents, and young adults with ALL (N = 55). Moxetumomab pasudotox was administered as a 30-minute IV infusion at doses of 5 to 50 µg/kg every other day for 6 (cohorts A and B) or 10 (cohort C) doses in 21-day cycles. Cohorts B and C received dexamethasone prophylaxis against capillary leak syndrome (CLS). The most common treatment-related adverse events were reversible weight gain, hepatic transaminase elevation, and hypoalbuminemia. Dose-limiting CLS occurred in 2 of 4 patients receiving 30 µg/kg of moxetumomab pasudotox every other day for 6 doses. Incorporation of dexamethasone prevented further dose-limiting CLS. Six of 14 patients receiving 50 µg/kg of moxetumomab pasudotox for 10 doses developed hemolytic uremic syndrome (HUS), thrombotic microangiopathy (TMA), or HUS-like events, exceeding the MTCD. Treatment expansion at 40 µg/kg for 10 doses (n = 11) exceeded the MTCD because of 2 HUS/TMA/HUS-like events. Dose level 6B (ie, 50 µg/kg × 6 doses) was the MTCD, selected as the recommended phase 2 dose. Among 47 evaluable patients, an objective response rate of 32% was observed, including 11 (23%) composite complete responses, 5 of which were minimal residual disease negative by flow cytometry. Moxetumomab pasudotox showed a manageable safety profile and evidence of activity in relapsed or refractory childhood ALL. This trial was registered at www.clinicaltrials.gov as #NCT00659425.


Assuntos
Toxinas Bacterianas/uso terapêutico , Exotoxinas/uso terapêutico , Imunotoxinas/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Adolescente , Adulto , Toxinas Bacterianas/efeitos adversos , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/farmacocinética , Síndrome de Vazamento Capilar/prevenção & controle , Criança , Pré-Escolar , Dexametasona/uso terapêutico , Exotoxinas/efeitos adversos , Exotoxinas/imunologia , Exotoxinas/farmacocinética , Feminino , Glucocorticoides/uso terapêutico , Síndrome Hemolítico-Urêmica/induzido quimicamente , Humanos , Hipoalbuminemia/induzido quimicamente , Imunotoxinas/efeitos adversos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Lactente , Masculino , Dose Máxima Tolerável , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Microangiopatias Trombóticas/induzido quimicamente , Aumento de Peso/efeitos dos fármacos , Adulto Jovem
8.
J Cancer Res Clin Oncol ; 143(10): 2025-2038, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28667390

RESUMO

PURPOSE: Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. METHODS: We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. RESULTS: Alexa Fluor® 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. CONCLUSIONS: Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.


Assuntos
ADP Ribose Transferases/administração & dosagem , Antígenos de Neoplasias/imunologia , Toxinas Bacterianas/administração & dosagem , Exotoxinas/administração & dosagem , Imunotoxinas/administração & dosagem , Proteínas de Neoplasias/imunologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Proteínas Recombinantes de Fusão/administração & dosagem , Fatores de Virulência/administração & dosagem , ADP Ribose Transferases/imunologia , Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Citometria de Fluxo , Proteínas Ligadas por GPI/imunologia , Células HEK293 , Humanos , Testes Imunológicos , Imunoterapia/métodos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Masculino , Terapia de Alvo Molecular , Neoplasias da Próstata/imunologia , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/imunologia , Fatores de Virulência/imunologia , Exotoxina A de Pseudomonas aeruginosa
9.
Cancer Biother Radiopharm ; 32(3): 83-89, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28380302

RESUMO

OBJECTIVES: Biodistribution of antibodies is vital to successful immunoscintigraphy/immunotherapy, and it is assumed to be similar to antigen distribution. We measured and compared the binding pattern of radiolabeled antibody to tissue antigen distribution in a nude mouse xenograft model of human melanoma. METHODS: We transplanted 107 FEM-XII human melanoma cells into the right flank of five nude mice. For the control, we transplanted 5 × 106 LS174T human colon cancer cells into the left flank. Two weeks later, 10 µCi of 131I-labeled melanoma-associated 96.5 monoclonal antibody (targeting p97 antigen) was intravenously injected. Three days later, we sacrificed the mice and evaluated 96.5 antibody binding and concentration in the tumors by ex vivo quantitative autoradiography (QAR). Two months later, we incubated adjacent tumor tissue slices in various concentrations of 125I-labeled 96.5 MoAb and evaluated the distribution/concentration of p97 antigen by in vitro QAR. RESULTS: p97 antigen distribution was homogeneous in the tumors (total antigen concentration [Bmax] = 17.36-38.36 pmol/g). In contrast, radiolabeled 96.5 antibody binding was heterogenous between location within the tumor (estimated bound antigen concentration = 0.7-6.6 pmol/g). No quantifiable parameters were found to be related with radiolabeled antibody binding and tumor antigen distribution. Antibody-bound tumor antigen to total antigen ratios ranged between 2% and 38%. CONCLUSIONS: Heterogeneous features of target antibody binding were observed in contrast to relatively homogenous feature of tumor antigen. We did not identify any correlations between p97 antigen distribution and 96.5 antibody binding in melanoma tissue. Radiolabeled 96.5 antibody binding patterns within melanoma cannot be predicted based on p97 antigen distribution in the tumor, which needs to be further studied with several other methods and more subjects in the future.


Assuntos
Anticorpos Monoclonais/imunologia , Imunotoxinas/farmacocinética , Antígenos Específicos de Melanoma/imunologia , Melanoma/imunologia , Animais , Reações Antígeno-Anticorpo , Humanos , Imunotoxinas/imunologia , Imunotoxinas/farmacologia , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/farmacocinética , Melanoma/radioterapia , Camundongos , Camundongos Nus , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Immunol Immunother ; 66(3): 367-378, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27933426

RESUMO

Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here, we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a tumor mouse model that has highly endogenous mTEM1 expression in the vasculature. Our data indicated that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.


Assuntos
Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Fragmentos de Imunoglobulinas/imunologia , Imunotoxinas/imunologia , Nanopartículas/administração & dosagem , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Sequência de Aminoácidos , Animais , Antígenos CD/biossíntese , Epitopos/imunologia , Humanos , Imunoterapia/métodos , Imunotoxinas/farmacocinética , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Nanopartículas/metabolismo , Proteínas de Neoplasias/biossíntese
11.
AAPS J ; 18(5): 1117-1130, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27287046

RESUMO

Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromolecular and small molecule properties. These issues range from systemic concerns, such as deconjugation of the small molecule drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Mathematical models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiologically based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quantitative in nature and cannot be handled qualitatively in isolation. For example, free antibody from deconjugation of the small molecule will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this mathematical model to analyze new experimental results on the distribution of the clinical antibody-drug conjugate Kadcyla in HER2-positive mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quantitative and mechanistic support to experimental studies trying to parse the impact of multiple mechanisms of action for these complex drugs.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Antineoplásicos/farmacocinética , Imunotoxinas/farmacocinética , Maitansina/análogos & derivados , Modelos Teóricos , Distribuição Tecidual/fisiologia , Trastuzumab/farmacocinética , Ado-Trastuzumab Emtansina , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Imunotoxinas/administração & dosagem , Maitansina/administração & dosagem , Maitansina/farmacocinética , Camundongos , Camundongos Nus , Distribuição Tecidual/efeitos dos fármacos , Trastuzumab/administração & dosagem , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Cancer Lett ; 372(2): 201-9, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-26806809

RESUMO

Triple-negative breast cancer (TNBC) is associated with poor prognosis and high prevalence among young premenopausal women. Unlike in other breast cancer subtypes, no targeted therapy is currently available. Overexpression of epithelial cell adhesion molecule (EpCAM) in 60% of TNBC tumors correlates with poorer prognosis and is associated with cancer stem cell phenotype. Thus, selective elimination of EpCAM(+) TNBC tumor cells is of clinical importance. Therefore, we constructed a fully human targeted cytolytic fusion protein, designated GbR201K-αEpCAM(scFv), in which an EpCAM-selective single-chain antibody fragment (scFv) is genetically fused to a granzyme B (Gb) mutant with reduced sensitivity to its natural inhibitor serpin B9. In vitro studies confirmed its specific binding, internalization and cytotoxicity toward a panel of EpCAM-expressing TNBC cells. Biodistribution kinetics and tumor-targeting efficacy using MDA-MB-468 cells in a human TNBC xenograft model in mice revealed selective accumulation of GbR201K-αEpCAM(scFv) in the tumors after i.v. injection. Moreover, treatment of tumor-bearing mice demonstrated a prominent inhibition of tumor growth of up to 50 % in this proof-of-concept study. Taken together, our results indicate that GbR201K-αEpCAM(scFv) is a promising novel targeted therapeutic for the treatment of TNBC.


Assuntos
Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular/antagonistas & inibidores , Granzimas/farmacologia , Imunoterapia/métodos , Imunotoxinas/farmacologia , Anticorpos de Cadeia Única/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Animais , Especificidade de Anticorpos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Molécula de Adesão da Célula Epitelial , Feminino , Granzimas/genética , Granzimas/farmacocinética , Células HEK293 , Humanos , Imunotoxinas/genética , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/farmacocinética , Distribuição Tecidual , Transfecção , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Cancer ; 138(4): 1013-23, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26334777

RESUMO

Fibroblast activation protein (FAP) is highly expressed in the tumor-associated fibroblasts (TAFs) of most human epithelial cancers. FAP plays a critical role in tumorigenesis and cancer progression, which makes it a promising target for novel anticancer therapy. However, mere abrogation of FAP enzymatic activity by small molecules is not very effective in inhibiting tumor growth. In this study, we have evaluated a novel immune-based approach to specifically deplete FAP-expressing TAFs in a mouse 4T1 metastatic breast cancer model. Depletion of FAP-positive stromal cells by FAP-targeting immunotoxin αFAP-PE38 altered levels of various growth factors, cytokines, chemokines and matrix metalloproteinases, decreased the recruitment of tumor-infiltrating immune cells in the tumor microenvironment and suppressed tumor growth. In addition, combined treatment with αFAP-PE38 and paclitaxel potently inhibited tumor growth in vivo. Our findings highlight the potential use of immunotoxin αFAP-PE38 to deplete FAP-expressing TAFs and thus provide a rationale for the use of this immunotoxin in cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Fibroblastos/metabolismo , Gelatinases/antagonistas & inibidores , Imunotoxinas/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Células 3T3 BALB , Modelos Animais de Doenças , Endopeptidases , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Imunotoxinas/farmacocinética , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Serina Endopeptidases
14.
Mol Cancer Ther ; 14(5): 1130-40, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25724665

RESUMO

Antibody-drug conjugates (ADC) are emerging as powerful cancer treatments that combine antibody-mediated tumor targeting with the potent cytotoxic activity of toxins. We recently reported the development of a novel ADC that delivers the cytotoxic payload monomethyl auristatin E (MMAE) to tumor cells expressing tissue factor (TF). By carefully selecting a TF-specific antibody that interferes with TF:FVIIa-dependent intracellular signaling, but not with the procoagulant activity of TF, an ADC was developed (TF-011-MMAE/HuMax-TF-ADC) that efficiently kills tumor cells, with an acceptable toxicology profile. To gain more insight in the efficacy of TF-directed ADC treatment, we compared the internalization characteristics and intracellular routing of TF with the EGFR and HER2. Both in absence and presence of antibody, TF demonstrated more efficient internalization, lysosomal targeting, and degradation than EGFR and HER2. By conjugating TF, EGFR, and HER2-specific antibodies with duostatin-3, a toxin that induces potent cytotoxicity upon antibody-mediated internalization but lacks the ability to induce bystander killing, we were able to compare cytotoxicity of ADCs with different tumor specificities. TF-ADC demonstrated effective killing against tumor cell lines with variable levels of target expression. In xenograft models, TF-ADC was relatively potent in reducing tumor growth compared with EGFR- and HER2-ADCs. We hypothesize that the constant turnover of TF on tumor cells makes this protein specifically suitable for an ADC approach.


Assuntos
Antineoplásicos/administração & dosagem , Receptores ErbB/metabolismo , Fator VIIa/metabolismo , Imunotoxinas/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Animais , Anticorpos , Antineoplásicos/farmacocinética , Apoptose , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Receptores ErbB/imunologia , Fator VIIa/imunologia , Humanos , Imunotoxinas/farmacocinética , Lisossomos/metabolismo , Camundongos , Neoplasias Experimentais/metabolismo , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Anticancer Res ; 34(7): 3263-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24982330

RESUMO

BACKGROUND: We herein report a comparison of the radiolabels 177Lu and 125I bound to the monoclonal antibody HH1 that targets the CD37 antigen expressed on non-Hodgkin B-cell lymphomas. MATERIALS AND METHODS: Mixtures of 177Lu and 125I-labeled HH1 antibody were co-injected into nude mice carrying Ramos xenografts and the biodistribution using the paired label format allowing tracer comparisons in each individual mouse. RESULTS: Products of the two radionuclides had very similar immunoractivity in vitro but showed different properties in vivo. Both products had relevant stability in blood and most normal tissues in nude mice carrying subcutaneous Ramos xenografts. However, both the tumor uptake and retention were significantly higher for 177Lu vs. 125I labeled HH1. The tumor to normal tissue ratios were several-fold improved for 177Lu compared to radioiodine labeled antibodies. CONCLUSION: The data presented herein support the evaluation of CD37 as a target for clinical 177Lu-based radioimmunotherapy against b-cell malignancies.


Assuntos
Imunotoxinas/farmacologia , Radioisótopos do Iodo/farmacologia , Lutécio/farmacologia , Linfoma de Células B/radioterapia , Radioisótopos/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Feminino , Compostos Heterocíclicos com 1 Anel/farmacocinética , Compostos Heterocíclicos com 1 Anel/farmacologia , Humanos , Imunotoxinas/farmacocinética , Radioisótopos do Iodo/farmacocinética , Lutécio/farmacocinética , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Radioimunoterapia/métodos , Radioisótopos/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Tetraspaninas/imunologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int J Cancer ; 135(6): 1497-508, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24523193

RESUMO

CMML (chronic myelomonocytic leukemia) belongs to the group of myeloid neoplasms known as myelodysplastic and myeloproliferative diseases. In some patients with a history of CMML, the disease transforms to acute myelomonocytic leukemia (AMML). There are no specific treatment options for patients suffering from CMML except for supportive care and DNA methyltransferase inhibitors in patients with advanced disease. New treatment strategies are urgently required, so we have investigated the use of immunotherapeutic directed cytolytic fusion proteins (CFPs), which are chimeric proteins comprising a selective domain and a toxic component (preferably of human origin to avoid immunogenicity). The human serine protease granzyme B is a prominent candidate for tumor immunotherapy because it is expressed in cytotoxic T lymphocytes and natural killer cells. Here, we report the use of CD64 as a novel target for specific CMML and AMML therapy, and correlate CD64 expression with typical surface markers representing these diseases. We demonstrate that CD64-specific human CFPs kill CMML and AMML cells ex vivo, and that the mutant granzyme B protein R201K is more cytotoxic than the wild-type enzyme in the presence of the granzyme B inhibitor PI9. Besides, the human CFP based on the granzyme B mutant was also able to kill AMML or CMML probes resistant to Pseudomonas exotoxin A.


Assuntos
Granzimas/administração & dosagem , Leucemia Mielomonocítica Aguda/tratamento farmacológico , Leucemia Mielomonocítica Crônica/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Estabilidade de Medicamentos , Feminino , Células HL-60 , Humanos , Imunotoxinas/administração & dosagem , Imunotoxinas/sangue , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Leucemia Mielomonocítica Aguda/imunologia , Leucemia Mielomonocítica Crônica/imunologia , Masculino , Pessoa de Meia-Idade , Receptores de IgG/biossíntese , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacocinética , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/farmacocinética
17.
Cancer Biother Radiopharm ; 29(2): 83-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24383763

RESUMO

Time-activity uptake curves [u(t) in % injected dose per gram of tissue] may be described by different--often complicated--functional forms. Because of the need to readily compare sequences of engineered radiopharmaceuticals, it is efficient to use mean residence time (MRT) as a one-parameter descriptor. In applying this computation to a sequence of five cognate anti-carcinoembryonic antigen (CEA) antibodies, it was found that the intact form had the longest MRT in the blood with the other four cognates having values less by approximately a factor of 10 or more. This difference probably follows from the lack of an intact Fc segment on the latter engineered molecules. MRT values for a sequence of six scFv-Fc engineered fragments demonstrated that the double mutant had the shortest blood residence time--30-fold less compared with the wild type. Whereas it is not possible to directly apply the MRT to nonbolus (tumor or organ) curves, a residence time (τ) may be assigned using the uptake function. Using τ, it was found that the intact (natural) form of the anti-CEA cognate set had the longest time at the tumor site in the human xenograft model in nude mice. The MRT and τ concept are proposed to also allow comparison of possible relative blood and tissue exposures, respectively, for cognate sets of unlabeled engineered antibodies used to treat malignancies although no data are yet available in the literature for this application.


Assuntos
Anticorpos Monoclonais/imunologia , Antígeno Carcinoembrionário/imunologia , Imunotoxinas/farmacologia , Imunotoxinas/farmacocinética , Modelos Biológicos , Compostos Radiofarmacêuticos/farmacologia , Compostos Radiofarmacêuticos/farmacocinética , Animais , Anticorpos Monoclonais/farmacocinética , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Radiother Oncol ; 110(2): 362-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24440046

RESUMO

PURPOSE: The effect of radioimmunotherapy (RIT) using the therapeutic radionuclide Y-90 bound to the anti-EGFR antibody cetuximab combined with external beam irradiation (EBRT) (EBRIT) on permanent local tumor control in vivo was examined. METHODS: Growth delay was evaluated in three human squamous cell carcinoma models after RIT with [(90)Y]Y-(CHX-A''-DTPA)4-cetuximab (Y-90-cetuximab). The EBRT dose required to cure 50% of the tumors (TCD50) for EBRT alone or EBRIT was evaluated in one RIT-responder (FaDu) and one RIT-non-responder (UT-SCC-5). EGFR expression and microenvironmental parameters were evaluated in untreated tumors, bioavailability was visualized by PET using ([(86)Y]Y-(CHX-A''-DTPA)4-cetuximab (Y-86-cetuximab) and biodistribution using Y-90-cetuximab. RESULTS: In UT-SCC-8 and FaDu but not in UT-SCC-5 radiolabeled cetuximab led to significant tumor growth delay. TCD50 after EBRT was significantly decreased by EGFR-targeted RIT in FaDu but not in UT-SCC-5. In contrast to EGFR expression, parameters of the tumor micromilieu and in particular the Y-90-cetuximab biodistribution or Y-86-cetuximab visualization in PET correlated with the responsiveness to RIT or EBRIT. CONCLUSION: EGFR-targeted EBRIT can improve permanent local tumor control compared to EBRT alone. PET imaging of bioavailability of labeled cetuximab appears to be a suitable predictor for response to EBRIT. This theragnostic approach should be further explored for clinical translation.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/radioterapia , Receptores ErbB/imunologia , Imunotoxinas/administração & dosagem , Radioisótopos de Ítrio/administração & dosagem , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacocinética , Carcinoma de Células Escamosas/imunologia , Linhagem Celular Tumoral , Cetuximab , Feminino , Humanos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Isotiocianatos/química , Masculino , Camundongos , Camundongos Nus , Ácido Pentético/análogos & derivados , Ácido Pentético/química , Radioimunoterapia , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/imunologia , Distribuição Aleatória , Distribuição Tecidual , Radioisótopos de Ítrio/química , Radioisótopos de Ítrio/farmacocinética
19.
Cancer Biother Radiopharm ; 29(2): 70-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24299067

RESUMO

Carcinoembryonic antigen (CEA) is an attractive target molecule of radioimmunotherapy (RIT). To enhance RIT's therapeutic efficacy, the fractionation of radiolabeled antibody doses is an attractive strategy. In this study, a fully human anti-CEA monoclonal antibody (mAb) C2-45 was selected by virtue of its lack of immunogenicity, and the effectiveness of fractionated RIT with yttrium-90 (9°Y)-labeled mAb C2-45 was evaluated. In LS180 tumor-bearing mice, indium-111 (¹¹¹In)-labeled mAb C2-45 showed high and persistent tumor accumulation. Therapeutic studies were performed with single doses of 9°Y-mAb C2-45 (100 or 200 µCi) or double doses of 100 µCi 9°Y-mAb C2-45 at different intervals (5, 10, and 15 days). All 9°Y-mAb C2-45-treated mice showed inhibition of tumor progression, while the time to tumor progression was much longer in both the 200-µCi-treated group and the double 100-µCi-treated group than in the single 100-µCi-treated group. The therapeutic effect of the double 100 µCi administration at days 0 and 15 lasted significantly longer than that in the other treatment groups. These findings indicate that 9°Y-mAb C2-45 may be a promising agent for the treatment of CEA-positive cancer and that the fractionation of 9°Y-labeled antibody doses could enhance the therapeutic effect if performed according to an appropriate protocol.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígeno Carcinoembrionário/imunologia , Neoplasias do Colo/radioterapia , Imunotoxinas/administração & dosagem , Radioimunoterapia/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Radioisótopos de Ítrio/administração & dosagem , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Humanos , Imunotoxinas/imunologia , Imunotoxinas/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Aleatória , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto , Radioisótopos de Ítrio/farmacocinética
20.
J Cancer Res Clin Oncol ; 140(1): 35-43, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24132461

RESUMO

PURPOSE: The extradomain B of fibronectin (ED-B) is a promising vascular target for selective pharmacodelivery in cancer patients. We analyzed a large series of prostatectomies from patients with prostate cancer, hyperplastic prostate disease, and normal prostates to study extent and tumor-selectivity of ED-B expression. METHODS: Using immunohistology, 68 adenocarcinomas of the prostate or prostate cancer-inflicted lymph nodes, 4 samples of benign prostatic hyperplasia, and 6 normal prostate glands were studied for ED-B expressing newly formed blood vessels. Further, we treated an advanced prostate cancer patient with the anti-ED-B antibody (131)I-L19SIP to study in vivo target accessibility. RESULTS: ED-B-positive blood vessels were found significantly more frequent in prostate cancers as compared with peritumoral prostate tissues or normal prostate glands, independent of tumor differentiation. The ED-B-positive blood vessels' density was 97 (±23), 65 (±9), and 59 (±9)/mm(2) in G3, G2, and G1 prostate cancers, respectively, and 7 (±5)/mm(2) in normal prostate glands. In high-grade (G3) prostate cancers, also the peritumoral tissue showed a higher density of ED-B vessels than normal prostate glands. Similar results were obtained when ED-B-positive vessel density was expressed as a fraction of CD34-positive vessel density. Finally, selective uptake of ED-B-binding (131)I-L19SIP to tumor lesions was found in an advanced prostate cancer patient by whole-body planar scintigraphy. CONCLUSIONS: ED-B-positive blood vessels were found to a large extent in prostate cancer tissues, but only rarely in normal prostates or benign prostatic hyperplasia. Whole-body planar scintigraphy in a prostate cancer patient confirmed selective uptake of (131)I-L19SIP in the prostate cancer tissues, qualifying ED-B as a promising target for selective pharmacodelivery of anticancer agents in prostate cancer.


Assuntos
Fibronectinas/biossíntese , Imunotoxinas/farmacocinética , Radioisótopos do Iodo/farmacocinética , Neoplasias da Próstata/metabolismo , Compostos Radiofarmacêuticos/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Anticorpos/imunologia , Vasos Sanguíneos/metabolismo , Epitopos/análise , Humanos , Imuno-Histoquímica , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Inclusão em Parafina , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Radioimunodetecção , Proteínas Recombinantes de Fusão/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA