Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
J Nanobiotechnology ; 20(1): 64, 2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35109860

RESUMO

BACKGROUND: While immune checkpoint inhibitors such as anti-PD-L1 antibodies have revolutionized cancer treatment, only subgroups of patients show durable responses. Insight in the relation between clinical response, PD-L1 expression and intratumoral localization of PD-L1 therapeutics could improve patient stratification. Therefore, we present the modular synthesis of multimodal antibody-based imaging tools for multiscale imaging of PD-L1 to study intratumoral distribution of PD-L1 therapeutics. RESULTS: To introduce imaging modalities, a peptide containing a near-infrared dye (sulfo-Cy5), a chelator (DTPA), an azide, and a sortase-recognition motif was synthesized. This peptide and a non-fluorescent intermediate were used for site-specific functionalization of c-terminally sortaggable mouse IgG1 (mIgG1) and Fab anti-PD-L1. To increase the half-life of the Fab fragment, a 20 kDa PEG chain was attached via strain-promoted azide-alkyne cycloaddition (SPAAC). Biodistribution and imaging studies were performed with 111In-labeled constructs in 4T1 tumor-bearing mice. Comparing our site-specific antibody-conjugates with randomly conjugated antibodies, we found that antibody clone, isotype and method of DTPA conjugation did not change tumor uptake. Furthermore, addition of sulfo-Cy5 did not affect the biodistribution. PEGylated Fab fragment displayed a significantly longer half-life compared to unPEGylated Fab and demonstrated the highest overall tumor uptake of all constructs. PD-L1 in tumors was clearly visualized by SPECT/CT, as well as whole body fluorescence imaging. Immunohistochemistry staining of tumor sections demonstrated that PD-L1 co-localized with the fluorescent and autoradiographic signal. Intratumoral localization of the imaging agent could be determined with cellular resolution using fluorescent microscopy. CONCLUSIONS: A set of molecularly defined multimodal antibody-based PD-L1 imaging agents were synthesized and validated for multiscale monitoring of PD-L1 expression and localization. Our modular approach for site-specific functionalization could easily be adapted to other targets.


Assuntos
Imunoconjugados , Neoplasias , Animais , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Humanos , Imunoconjugados/metabolismo , Imuno-Histoquímica , Camundongos , Neoplasias/diagnóstico por imagem , Distribuição Tecidual
3.
J Nucl Med ; 61(2): 270-275, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31519800

RESUMO

Arteriovenous malformations (AVMs) have an inherent capacity to form new blood vessels, resulting in excessive lesion growth, and this process is further triggered by the release of angiogenic factors. 68Ga-labeled arginine-glycine-aspartate tripeptide sequence (RGD) PET/CT imaging may provide insight into the angiogenic status and treatment response of AVMs. This clinical feasibility study was performed to demonstrate that 68Ga-RGD PET/CT imaging can be used to quantitatively assess angiogenesis in peripheral AVMs. Methods: Ten patients with a peripheral AVM (mean age, 40 y; 4 men and 6 women) and scheduled for endovascular embolization treatment were prospectively included. All patients underwent 68Ga-RGD PET/CT imaging 60 min after injection (mean dose, 207 ± 5 MBq). Uptake in the AVM, blood pool, and muscle was quantified as SUVmax and SUVpeak, and a descriptive analysis of the PET/CT images was performed. Furthermore, immunohistochemical analysis was performed on surgical biopsy sections of peripheral AVMs to investigate the expression pattern of integrin αvß3Results:68Ga-RGD PET/CT imaging showed enhanced uptake in all AVM lesions (mean SUVmax, 3.0 ± 1.1; mean SUVpeak, 2.2 ± 0.9). Lesion-to-blood and lesion-to-muscle ratios were 3.5 ± 2.2 and 4.6 ± 2.8, respectively. Uptake in blood and muscle was significantly higher in AVMs than in background tissue (P = 0.0006 and P = 0.0014, respectively). Initial observations included uptake in multifocal AVM lesions and enhanced uptake in intraosseous components in those AVM cases affecting bone integrity. Immunohistochemical analysis revealed cytoplasmatic and membranous integrin αvß3 expression in the endothelial cells of AVMs. Conclusion: This feasibility study showed increased uptake in AVMs with angiogenic activity, compared with surrounding tissue without angiogenic activity, suggesting that 68Ga-RGD PET/CT imaging can be used as a tool to quantitatively determine angiogenesis in AVMs. Further studies will be conducted to explore the potential of 68Ga-RGD PET/CT imaging for guiding current treatment decisions and for assessing response to antiangiogenic treatment.


Assuntos
Malformações Arteriovenosas/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem , Oligopeptídeos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Adulto , Malformações Arteriovenosas/complicações , Malformações Arteriovenosas/metabolismo , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/complicações , Oligopeptídeos/metabolismo , Estudos Prospectivos , Transporte Proteico , Adulto Jovem
4.
Cancer Immunol Res ; 7(1): 150-161, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30459153

RESUMO

Antibodies that block the interaction between programmed death ligand 1 (PD-L1) and PD-1 have shown impressive responses in subgroups of patients with cancer. PD-L1 expression in tumors seems to be a prerequisite for treatment response. However, PD-L1 is heterogeneously expressed within tumor lesions and may change upon disease progression and treatment. Imaging of PD-L1 could aid in patient selection. Previously, we showed the feasibility to image PD-L1+ tumors in immunodeficient mice. However, PD-L1 is also expressed on immune cell subsets. Therefore, the aim of this study was to assess the potential of PD-L1 micro single-photon emission tomography/computed tomography (microSPECT/CT) using radiolabeled PD-L1 antibodies to (i) measure PD-L1 expression in two immunocompetent tumor models (syngeneic mice and humanized mice harboring PD-L1 expressing immune cells) and (ii) monitor therapy-induced changes in tumor PD-L1 expression. We showed that radiolabeled PD-L1 antibodies accumulated preferentially in PD-L1+ tumors, despite considerable uptake in certain normal lymphoid tissues (spleen and lymph nodes) and nonlymphoid tissues (duodenum and brown fat). PD-L1 microSPECT/CT imaging could also distinguish between high and low PD-L1-expressing tumors. The presence of PD-L1+ immune cells did not compromise tumor uptake of the human PD-L1 antibodies in humanized mice, and we demonstrated that radiotherapy-induced upregulation of PD-L1 expression in murine tumors could be monitored with microSPECT/CT imaging. Together, these data demonstrate that PD-L1 microSPECT/CT is a sensitive technique to detect variations in tumor PD-L1 expression, and in the future, this technique may enable patient selection for PD-1/PD-L1-targeted therapy.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Animais , Anticorpos Monoclonais/farmacocinética , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Radioisótopos de Índio , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único
5.
Cancer Biother Radiopharm ; 32(2): 67-73, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28301262

RESUMO

BACKGROUND: Up to now, prostate-specific membrane antigen (PSMA)-targeted radionuclide therapy mainly focused on ß-emitting radionuclides. Herein, two new 213Bi-labeled agents for PSMA-targeted α therapy of prostate cancer (PCa) are reported. METHODS: The biodistribution of 213Bi-labeled small-molecule inhibitor PSMA I&T and nanobody JVZ-008 was evaluated in mice bearing PSMA-positive LNCaP xenografts. DNA damage response was followed using LNCaP cells and LNCaP xenografts. RESULTS: In vitro, 213Bi-PSMA I&T and 213Bi-JVZ-008 therapy of LNCaP cells led to increased number of DNA double-strand breaks (DSBs), detected as 53BP1 and γH2AX nuclear foci. In vivo, tumor uptake of 213Bi-PSMA I&T and 213Bi-JVZ-008 was 5.75% ± 2.70%ID/g (injected dose per gram) and 2.68% ± 0.56%ID/g, respectively, with similar tumor-to-kidney ratios. Furthermore, both agents induced in vivo DSBs in the tumors, which were detected between 1 hour and 24 hours after injection. 213Bi-PSMA I&T induced significantly more DSBs than 213Bi-JVZ-008 (p < 0.01). CONCLUSIONS: 213Bi-PSMA I&T and 213Bi-JVZ-008 showed efficient and rapid tumor targeting and produced DSBs in PSMA-expressing LNCaP cells and xenografts. These promising results require further evaluation of 213Bi-labeled agents with regard to their therapeutic efficacy and toxicity for PCa therapy.


Assuntos
Bismuto/química , Quebras de DNA de Cadeia Dupla , Neoplasias da Próstata/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/química , Radioisótopos/química , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Nanopartículas/química , Transplante de Neoplasias , Neoplasias da Próstata/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Nucl Med ; 58(6): 926-933, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28232604

RESUMO

Pretargeted radioimmunotherapy (PRIT) with the ß-emitting radionuclide 177Lu is an attractive approach to treat carcinoembryonic antigen (CEA)-expressing tumors. The therapeutic efficacy of PRIT might be improved using α-emitting radionuclides such as 213Bi. Herein, we report and compare the tumor-targeting properties and therapeutic efficacy of 213Bi and 177Lu for PRIT of CEA-expressing xenografts, using the bispecific monoclonal antibody TF2 (anti-CEA × anti-histamine-succinyl-glycine [HSG]) and the di-HSG-DOTA peptide IMP288. Methods: The in vitro binding characteristics of 213Bi-IMP288 were compared with those of 177Lu-IMP288. Tumor targeting of 213Bi-IMP288 and 177Lu-IMP288 was studied in mice bearing subcutaneous LS174T tumors that were pretargeted with TF2. Finally, the effect of 213Bi-IMP288 (6, 12, or 17 MBq) and 177Lu-IMP288 (60 MBq) on tumor growth and survival was assessed. Toxicity was determined by monitoring body weight, analyzing blood samples for hematologic and renal toxicity (hemoglobin, leukocytes, platelets, creatinine), and immunohistochemical analysis of the kidneys. Results: The in vitro binding characteristics of 213Bi-IMP288 (dissociation constant, 0.45 ± 0.20 nM) to TF2-pretargeted LS174T cells were similar to those of 177Lu-IMP288 (dissociation constant, 0.53 ± 0.12 nM). In vivo accumulation of 213Bi-IMP288 in LS174T tumors was observed as early as 15 min after injection (9.2 ± 2.0 percentage injected dose [%ID]/g). 213Bi-IMP288 cleared rapidly from the circulation; at 30 min after injection, the blood levels were 0.44 ± 0.28 %ID/g. Uptake in normal tissues was low, except for the kidneys, where uptake was 1.8 ± 1.1 %ID/g at 30 min after injection. The biodistribution of 213Bi-IMP288 was comparable to that of 177Lu-IMP288. Mice treated with a single dose of 213Bi-IMP288 or 177Lu-IMP288 showed significant inhibition of tumor growth. Median survival for the groups treated with phosphate-buffered saline, 6 MBq 213Bi-IMP288, 12 MBq 213Bi-IMP288, and 60 MBq 177Lu-IMP288 was 22, 31, 45, and 42 d, respectively. Mice receiving 17 MBq 213Bi-IMP288 showed significant weight loss, resulting in a median survival of only 24 d. No changes in hemoglobin, platelets, or leukocytes were observed in the treatment groups. However, immunohistochemical analysis of the kidneys of mice treated with 17 or 12 MBq 213Bi-IMP288 showed signs of tubular damage, indicating nephrotoxicity. Conclusion: To our knowledge, this study shows for the first time that PRIT with TF2 and 213Bi-IMP288 is feasible and at least as effective as 177Lu-IMP288. However, at higher doses, kidney toxicity was observed. Future studies are warranted to determine the optimal dosing schedule to improve therapeutic efficacy while reducing renal toxicity.


Assuntos
Antígeno Carcinoembrionário/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/radioterapia , Terapia de Alvo Molecular/métodos , Radioimunoterapia/métodos , Radioisótopos/uso terapêutico , Partículas alfa/uso terapêutico , Animais , Partículas beta/uso terapêutico , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Resultado do Tratamento
7.
Mol Imaging Biol ; 19(4): 540-549, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27798786

RESUMO

PURPOSE: The aim of the study was to investigate the potential of diffusion-weighted magnetic resonance imaging (DW-MRI) and 3'-dexoy-3'-[18F]fluorothymidine ([18F]FLT) positron emission tomography (PET) as early biomarkers of treatment response of 5-fluorouracil (5-FU) in a syngeneic rat model of colorectal cancer liver metastases. PROCEDURES: Wag/Rij rats with intrahepatic syngeneic CC531 tumors were treated with 5-FU (15, 30, or 60 mg/kg in weekly intervals). Before treatment and at days 1, 3, 7, and 14 after treatment rats underwent DW-MRI and [18F]FLT PET. Tumors were analyzed immunohistochemically for Ki67, TK1, and ENT1 expression. RESULTS: 5-FU inhibited the growth of CC531 tumors in a dose-dependent manner. Immunohistochemical analysis did not show significant changes in Ki67, TK1, and ENT1 expression. However, [18F]FLT SUVmean and SUVmax were significantly increased at days 4 and 7 after treatment with 5-FU (60 mg/kg) and returned to baseline at day 14 (SUVmax at days -1, 4, 7, and 14 was 1.1 ± 0.1, 2.3 ± 0.5, 2.3 ± 0.6, and 1.5 ± 0.4, respectively). No changes in [18F]FLT uptake were observed in the nontreated animals. Furthermore, the apparent diffusion coefficient (ADCmean) did not change in 5-FU-treated rats compared to untreated rats. CONCLUSION: This study suggests that 5-FU treatment induces a flare in [18F]FLT uptake of responsive CC531 tumors in the liver, while the ADCmean did not change significantly. Future studies in larger groups are warranted to further investigate whether [18F]FLT PET can discriminate between disease progression and treatment response.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Didesoxinucleosídeos/uso terapêutico , Imagem de Difusão por Ressonância Magnética , Fluoruracila/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Tomografia por Emissão de Pósitrons , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Didesoxinucleosídeos/farmacologia , Modelos Animais de Doenças , Imuno-Histoquímica , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Ratos , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Resultado do Tratamento
8.
Theranostics ; 6(6): 849-61, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27162555

RESUMO

Prostate-specific membrane antigen (PSMA) is a well-established target for nuclear imaging and therapy of prostate cancer (PCa). Radiolabeled small-molecule PSMA inhibitors are excellent candidates for PCa theranostics-they rapidly and efficiently localize in tumor lesions. However, high tracer uptake in kidneys and salivary glands are major concerns for therapeutic applications. Here, we present the preclinical application of PSMA I&T, a DOTAGA-chelated urea-based PSMA inhibitor, for SPECT/CT imaging and radionuclide therapy of PCa. (111)In-PSMA I&T showed dose-dependent uptake in PSMA-expressing tumors, kidneys, spleen, adrenals, lungs and salivary glands. Coadministration of 2-(phosphonomethyl)pentane-1,5-dioic acid (2-PMPA) efficiently reduced PSMA-mediated renal uptake of (111)In-PSMA I&T, with the highest tumor/kidney radioactivity ratios being obtained using a dose of 50 nmol 2-PMPA. SPECT/CT clearly visualized subcutaneous tumors and sub-millimeter intraperitoneal metastases; however, high renal and spleen uptake in control mice (no 2-PMPA) interfered with visualization of metastases in the vicinity of those organs. Coadministration of 2-PMPA increased the tumor-to-kidney absorbed dose ratio during (177)Lu-PSMA I&T radionuclide therapy. Hence, at equivalent absorbed dose to the tumor (36 Gy), coinjection of 2-PMPA decreased absorbed dose to the kidneys from 30 Gy to 12 Gy. Mice injected with (177)Lu-PSMA I&T only, showed signs of nephrotoxicity at 3 months after therapy, whereas mice injected with (177)Lu-PSMA I&T + 2-PMPA did not. These data indicate that PSMA I&T is a promising theranostic tool for PCa. PSMA-specific uptake in kidneys can be successfully tackled using blocking agents such as 2-PMPA.


Assuntos
Glutamato Carboxipeptidase II/antagonistas & inibidores , Medicina de Precisão/métodos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/terapia , Radioterapia/métodos , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único/métodos , Nanomedicina Teranóstica/métodos , Animais , Antígenos de Superfície , Modelos Animais de Doenças , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Masculino , Camundongos , Radioisótopos/administração & dosagem , Radioisótopos/efeitos adversos , Radioterapia/efeitos adversos
9.
Cancer Res ; 75(14): 2928-36, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25977331

RESUMO

Antibodies that block the interaction between programmed death ligand 1 (PD-L1) and PD-1 have shown impressive antitumor activity. Patients with tumors expressing PD-L1 are most likely to respond to this treatment. The aim of our study was to develop a noninvasive imaging technique to determine tumor PD-L1 expression in vivo. This could allow selection of patients that are most likely to benefit from anti-PD-1/PD-L1 treatment and to monitor PD-L1 expression during therapy. The monoclonal antibody PD-L1.3.1 was radiolabeled with Indium-111 ((111)In) and characterized using PD-L1-expressing MDA-MB-231 cells. Subsequently, the optimal antibody dose and time point for imaging was determined in mice with MDA-MB-231 xenografts. Finally, SPECT/CT imaging was performed in xenograft models with different PD-L1 expression levels and tumor sections were analyzed for PD-L1 expression using IHC. The optimal antibody dose of (111)In-PD-L1.3.1 (Kd = 1 nmol/L) for SPECT/CT imaging was ≤1 µg. Highest tumor-to-normal tissue contrast was obtained at days 3 and 7 after injection. (111)In-PD-L1.3.1 SPECT/CT showed efficient accumulation in high PD-L1-expressing tumors (MDA-MB-231 and SK-Br-3), whereas no specific uptake was observed in tumors with low or no detectable levels of PD-L1 (SUM149, BT474, and MCF-7). SPECT/CT and autoradiography showed a very heterogeneous distribution of (111)In-PD-L1.3.1 within the tumor. In conclusion, this is the first study showing the feasibility of noninvasive in vivo imaging of PD-L1 expression in tumors. (111)In-PD-L1.3.1 showed efficient and specific uptake in PD-L1 expressing xenografts. This technique may enable patient selection for PD-1 and PD-L1-targeted therapy.


Assuntos
Anticorpos Monoclonais , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Neoplasias da Mama/diagnóstico por imagem , Radioisótopos de Índio , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Xenoenxertos , Humanos , Radioisótopos de Índio/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ligação Proteica , Distribuição Tecidual , Células Tumorais Cultivadas
10.
PLoS One ; 10(2): e0117745, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25680198

RESUMO

INTRODUCTION: The insulin-like growth factor 1 receptor (IGF-1R) may be involved in the development of resistance against conventional cancer treatment. The aim of this study was to assess whether IGF-1R expression of breast tumors changes during neoadjuvant therapy and to study whether these changes were associated with survival. METHODS: Paraffin embedded tumor tissue was collected from pretreatment biopsies and surgical resections of 62 breast cancer patients who were treated with neoadjuvant chemotherapy or endocrine therapy. IGF-1R expression was determined immunohistochemically and compared before and after treatment. RESULTS: High membranous IGF-1R expression at diagnosis correlated significantly with ER positivity, low tumor stage (stage I/II) and longer overall survival (p < 0.05). After neoadjuvant treatment, membranous IGF-1R expression remained the same in 41 (65%) tumors, was upregulated in 11 (18%) tumors and downregulated in 11 (18%) tumors. Changes in membranous IGF-1R expression were associated with overall survival (log-rank test: p = 0.013, multivariate cox-regression: p = 0.086). Mean overall survival time for upregulation, no change, and downregulation in IGF-1R expression was 3.0 ± 0.5 years, 7.3 ± 1.0 years and 15.0 ± 1.8 years, respectively. Changes in other parameters were not significantly associated with survival. CONCLUSION: Neoadjuvant therapy can induce changes in IGF-1R expression. Upregulation of IGF-1R expression after neoadjuvant treatment is a poor prognostic factor in breast cancer patients, providing a rationale for incorporating anti-IGF-1R drugs in the management of these patients.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Receptor IGF Tipo 1/metabolismo , Idoso , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Feminino , Seguimentos , Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias , Prognóstico , Receptor IGF Tipo 1/genética , Regulação para Cima
11.
Mol Pharm ; 11(11): 4249-57, 2014 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-25294389

RESUMO

Bevacizumab and cetuximab are approved for the treatment of cancer. However, in advanced colorectal cancer, addition of cetuximab to chemotherapy with bevacizumab did not improve survival. The reason for the lack of activity remains unclear. The aim of this study was to determine the effect of cetuximab on VEGF expression and targeting of bevacizumab to the tumor. Mice with subcutaneous SUM149 or WiDr xenografts were treated with cetuximab, bevacizumab, or a combination of the two. Before the start of cetuximab treatment and after 7 and 21 days of treatment, the uptake of radiolabeled bevacizumab in the tumor was measured by immunoSPECT/CT. Tumor growth of SUM149 xenografts was significantly inhibited by cetuximab, bevacizumab, or their combination, whereas growth of WiDr xenografts was not affected. Cetuximab caused a significant reduction of bevacizumab uptake in SUM149 xenografts, whereas tumor-to-blood ratios in mice with WiDr xenografts did not change. Biodistribution studies with an irrelevant antibody in the SUM149 model also showed significantly reduced tumor-to-blood ratios. Cetuximab treatment did not decrease VEGF expression. Without decreasing VEGF levels, cetuximab reduces tumor targeting of bevacizumab. This could, at least partly, explain why the combination of bevacizumab and cetuximab does not result in improved therapeutic efficacy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Receptores ErbB/metabolismo , Terapia de Alvo Molecular , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Cetuximab , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Multimodal , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Imaging Biol ; 16(4): 529-37, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24532107

RESUMO

PURPOSE: The aim was to assess changes in insulin-like growth factor 1 receptor (IGF-1R) expression with immunoSPECT/CT and to study the dynamics of IGF-1R expression of human breast tumors during endocrine treatment. PROCEDURES: Mice with MCF-7 xenografts were treated with estradiol or tamoxifen, and IGF-1R expression was measured by immunohistochemistry and immunoSPECT/CT using (111)In-R1507 (anti-IGF-1R antibody). Moreover, IGF-1R expression was analyzed immunohistochemically on 22 human breast tumors, treated preoperatively with endocrine therapy. RESULTS: Estradiol resulted in an increased expression of IGF-1R, as measured by immunohistochemistry and immunoSPECT/CT. In contrast, tamoxifen resulted in a downregulation of IGF-1R, whereas this could not be measured with immunoSPECT/CT. A downregulation was also detectable in 9 out of 22 (41 %) human breast tumors after endocrine therapy. CONCLUSIONS: Anti-estrogen treatment can cause a reduction in membranous IGF-1R expression. Based on these results, a combination of anti-IGF-1R antibodies with anti-estrogen therapy might not be a rational treatment strategy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Estradiol/uso terapêutico , Receptor IGF Tipo 1/metabolismo , Tamoxifeno/uso terapêutico , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/patologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Estradiol/farmacologia , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Receptor IGF Tipo 1/farmacologia , Tamoxifeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Eur J Cancer ; 49(13): 2851-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23664098

RESUMO

PURPOSE: To investigate whether F(ab')2-fragments of the monoclonal Insulin-like Growth Factor-1 Receptor (IGF-1R) antibody R1507 (F(ab')2-R1507) can successfully target IGF-1R in Ewing sarcomas (ES). MATERIALS AND METHODS: BALB/c nude mice were subcutaneously implanted with IGF-1R-expressing human ES xenografts (EW-5 and EW-8) which previously showed heterogeneous or no uptake of indium-111-labelled R1507 IgG ((111)In-R1507), respectively. Mice were injected with (111)In-F(ab')2-R1507 or (111)In-R1507 as a reference. Biodistribution and immuno-SPECT/computed tomography (CT) imaging studies were carried out 2, 4, 8 and 24 h post-injection (p.i.) for (111)In-F(ab')2-R1507 and 24 h p.i. for (111)In-R1507. RESULTS: Biodistribution studies showed specific accumulation of (111)In-F(ab')2-R1507 in EW-5 xenografts from t=2 h p.i. onwards (3.6 ± 0.2%ID/g at t = 24 h p.i.) and (111)In-F(ab')2-R1507 immuno-SPECT showed almost homogeneous intratumoural distribution at t=24h p.i. Tumour-to-blood ratios of (111)In-F(ab')2-R1507 were significantly higher than those of (111)In-R1507 at t=24 h p.i. (2.4 ± 0.4 versus 0.5 ± 0.1, respectively; p<0.05). More importantly, (111)In-F(ab')2-R1507 also specifically accumulated in EW-8 tumours (3.7 ± 0.7%ID/g at t = 24 h p.i). In both EW-5 and EW-8 tumours, there was a good spatial correlation between IGF-1R expression and (111)In-F(ab')2-R1507 tumour distribution. CONCLUSION: (111)In-F(ab')2-R1507 fragments can successfully target IGF-1R in ES models and have superior tumour penetrating and IGF-1R-targeting properties as compared to (111)In-R1507. This suggests that anti-IGF-1R therapies in ES and other tumours may be improved by using smaller therapeutic compounds, although further in vivo studies addressing this topic are warranted.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Fragmentos Fab das Imunoglobulinas/farmacologia , Receptor IGF Tipo 1/imunologia , Sarcoma de Ewing/tratamento farmacológico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Fragmentos Fab das Imunoglobulinas/metabolismo , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sarcoma de Ewing/diagnóstico por imagem , Sarcoma de Ewing/imunologia , Sarcoma de Ewing/metabolismo , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Cancer ; 133(2): 307-14, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23335047

RESUMO

Bevacizumab (antivascular endothelial growth factor [anti-VEGF]) and cetuximab (antiepidermal growth factor receptor [anti-EGFR]) are approved antibodies for treatment of cancer. However, in advanced colorectal cancer, the combination fails to improve survival. As the reason for the lack of activity is unknown, our study aims to determine the effect of bevacizumab on targeting of anti-EGFR and insulin-like growth factor 1 receptor (IGF-1R) antibodies in tumors with single-photon emission computed tomography (SPECT)/CT imaging. Mice with subcutaneous EGFR and IGF-1R-expressing SUM149 xenografts received a single dose of bevacizumab (10 mg/kg) or saline. After 4 days, mice were injected with radiolabeled cetuximab or R1507, an anti-IGF-1R antibody. A control group received a radiolabeled irrelevant IgG (hLL2). Three days later, SPECT/CT images were acquired and mice were dissected to determine the concentration of antibodies in the tissues. Tumors were analyzed immunohistochemically to determine vascular density (CD34), VEGF, EGFR and IGF-1R expression. SPECT/CT imaging revealed that bevacizumab treatment significantly reduced tumor targeting of radiolabeled cetuximab by 40% from 33.1 ± 1.1 %ID/g to 19.8 ± 5.7 %ID/g (p = 0.009) for untreated and bevacizumab-treated tumors, respectively. A similar effect was found for (111) In-R1507: tumor targeting of R1507 decreased by 35%. No significant differences in tumor uptake were observed in mice that received an irrelevant IgG. Uptake in normal organs was not altered by bevacizumab. Immunohistochemical analysis showed that vascular density decreased with 43%, whereas EGFR and IGF-1R expression was unaltered. In conclusion, bevacizumab treatment significantly reduces tumor targeting of anti-EGFR and anti-IGF-1R antibodies. This emphasizes the importance of timing and sequencing of bevacizumab in combination with other antibodies.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Receptores ErbB/imunologia , Terapia de Alvo Molecular , Receptor IGF Tipo 1/imunologia , Animais , Antígenos CD34/metabolismo , Bevacizumab , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cetuximab , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Imagem Multimodal , Transplante de Neoplasias , Tomografia por Emissão de Pósitrons , Receptor IGF Tipo 1/antagonistas & inibidores , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Tomografia Computadorizada por Raios X , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
15.
Mol Pharm ; 9(8): 2314-21, 2012 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-22747077

RESUMO

The insulin-like growth factor 1 receptor (IGF-1R) is a potential new target for the treatment of breast cancer. Patients with breast cancer lesions that express IGF-1R may benefit from treatment with anti-IGF-1R antibodies. IGF-1R expression can be visualized using radiolabeled R1507, a monoclonal antibody directed against IGF-1R. However, antibodies clear slowly from the circulation, resulting in low tumor-to-background ratios early after injection. Therefore, we aimed to accelerate targeting of IGF-1R using radiolabeled R1507 F(ab')2 and Fab fragments. In vitro, immunoreactivity, binding affinity and internalization of R1507 IgG, F(ab')2 and Fab were determined using the triple negative IGF-1R-expressing breast cancer cell line SUM149. In vivo, pharmacokinetics of (111)In-labeled R1507 IgG, F(ab')2 and Fab were studied in mice bearing subcutaneous SUM149 xenografts. SPECT/CT images were acquired and the biodistribution was measured ex vivo. The in vitro binding characteristics of radiolabeled R1507 IgG and F(ab')2 were comparable, whereas the affinity of Fab fragments was significantly lower (Kd: 0.6 nM, 0.7 nM and 3.0 nM for R1507 IgG, F(ab')2 and Fab, respectively). Biodistribution studies showed that the maximum tumor uptake of (111)In-R1507 IgG, F(ab')2 and Fab was 31.8% ID/g (72 h p.i.), 10.0% ID/g (6 h p.i.), and 1.8% ID/g (1 h p.i.), respectively. However, maximal tumor-to-blood ratios for F(ab')2 (24 h p.i.: 7.5) were more than twice as high as those obtained with R1507 (72 h p.i.: 2.8) and Fab (6 h p.i.: 2.8). Injection of an excess of unlabeled R1507 significantly reduced tumor uptake, suggesting that the uptake of R1507 IgG and F(ab')2 was specific for IGF-1R, while the major fraction of the tumor uptake of Fab was nonspecific. IGF-1R-expressing xenografts were visualized with (111)In-F(ab')2 SPECT/CT as early as 6 h p.i., while with R1507 IgG, the tumor could be visualized after 24 h. No specific targeting was observed with (111)In-Fab. (111)In-F(ab')2 fragments showed improved targeting of IGF-1R expressing tumors. Tumor-to-blood ratios were twice as high as those obtained with (111)In-R1507, and adequate tumor targeting on SPECT/CT images was observed as early as 6 h p.i. For individualization and optimization of IGF-1R targeted therapy, (111)In-F(ab')2 may be the tracer of choice.


Assuntos
Anticorpos Monoclonais/química , Fragmentos Fab das Imunoglobulinas/química , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/diagnóstico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus
16.
J Control Release ; 164(3): 283-90, 2012 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-22800584

RESUMO

Clinical application of calcium phosphate cement (CPC; with incorporated polymeric porogens) in an injectable form implicates that loading methods for growth factors are limited. In view of this, the current study evaluated the in vitro and in vivo release kinetics of bone morphogenetic protein-2 (BMP-2) loaded on poly(d,l-lactic-co-glycolic acid) (PLGA) microparticles (CPC/PLGA), BMP-2 incorporation into the liquid phase of CPC (CPC/liquid), and BMP-2 absorbed to the surface of preset, porous CPC (CPC/surface) as a control via an in vitro release experiment and in vivo using microSPECT imaging with (125)I-labeled BMP-2. In addition, the osteoinductive capacity of scaffolds generated via the different BMP-2 loading methods was assessed in a subcutaneous rat model. Additional controls consisted of porous CPC scaffolds (CPC/porous) and CPC/PLGA (CPC/control) without BMP-2 loading. The results revealed that it is feasible to load BMP-2 into CPC via adsorption to PLGA-microparticles or the liquid phase of CPC, which resulted in a similar release profile over the course of 28 days, despite distinct protein distribution patterns. Compared to CPC-scaffolds with surface-loaded BMP-2, these loading methods showed a similar release profile, except for a significantly decreased burst release. As such, the observed osteoinductive capacity for only CPC-scaffolds with surface-loaded BMP-2 is likely to be related to this difference in burst release. It remains unclear to what extent the differential BMP-2 loading methods for injectable CPC can affect the biological response in a bone environment.


Assuntos
Cimentos Ósseos/química , Proteína Morfogenética Óssea 2/administração & dosagem , Fosfatos de Cálcio/química , Ácido Láctico/química , Ácido Poliglicólico/química , Animais , Proteína Morfogenética Óssea 2/química , Composição de Medicamentos , Implantes de Medicamento , Injeções Subcutâneas , Masculino , Microscopia Eletrônica de Varredura , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Porosidade , Estrutura Secundária de Proteína , Ratos , Ratos Wistar , Solubilidade , Propriedades de Superfície , Tomografia Computadorizada de Emissão de Fóton Único
17.
Clin Cancer Res ; 17(24): 7693-703, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22038993

RESUMO

PURPOSE: To investigate whether indium-111-labeled R1507 ((111)In-R1507) immuno-SPECT (single-photon emission computed tomography), a novel noninvasive, in vivo screening method to visualize membranous insulin-like growth factor 1 receptor (IGF-1R) expression and accessibility, can be used to predict IGF-1R treatment (R1507) response in bone sarcomas. EXPERIMENTAL DESIGN: BALB/c nude mice were subcutaneously implanted with IGF-1R-expressing human bone sarcoma xenografts (OS-1, EW-5, and EW-8) which showed high, modest, or no response, respectively, to R1507, a monoclonal antibody targeting the extracellular domain of IGF-1R. An IGF-1R-negative tumor (OS-33), unresponsive to IGF-1R inhibitors, was examined as well. Mice were injected with (111)In-R1507. Biodistribution and immuno-SPECT/computed tomography imaging studies were carried out 1, 3, and 7 days p.i. in mice with OS-1 and EW-5 xenografts and 3 days p.i. in mice with EW-8 and OS-33 xenografts. RESULTS: Biodistribution studies showed specific accumulation of (111)In-R1507 in OS-1 and EW-5 xenografts (27.5 ± 6.5%ID/g and 14.0 ± 2.8%ID/g, 3 days p.i., respectively). Most importantly, (111)In-R1507 uptake in IGF-1R positive, but unresponsive, EW-8 xenografts (6.5 ± 1.5%ID/g, 3 days p.i.) was similar to that of the IGF-1R-negative OS-33 tumor (5.5 ± 0.6%ID/g, 3 days p.i.). Uptake in normal tissues was low and nonspecific. Corresponding immuno-SPECT images clearly discriminated between high, modest, and nonresponding tumors by showing a homogeneous (OS-1), heterogeneous (EW-5), or nonspecific (EW-8 and OS-33) tumor uptake of (111)In-R1507. CONCLUSIONS: (111)In-R1507 immuno-SPECT is an excellent method to visualize membranous IGF-1R expression and target accessibility in vivo in human bone sarcoma xenografts and may serve as an independent marker to predict IGF-1R therapy (R1507) response in bone sarcoma patients.


Assuntos
Neoplasias Ósseas/diagnóstico por imagem , Receptor IGF Tipo 1/metabolismo , Sarcoma/diagnóstico por imagem , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Radioisótopos de Índio , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos , Prognóstico , Receptor IGF Tipo 1/antagonistas & inibidores , Reprodutibilidade dos Testes , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Int J Cancer ; 129(4): 870-8, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20957635

RESUMO

Noninvasive imaging of the epidermal growth factor receptor (EGFR) in head-and-neck squamous cell carcinoma could be of value to select patients for EGFR-targeted therapy. We assessed dose optimization of (111) Indium-DTPA-cetuximab ((111) In-cetuximab) for EGFR imaging in a head-and-neck squamous cell carcinoma xenograft model. (111) In-cetuximab slowly internalized into FaDu cells in vitro, amounting to 1.0 × 10(4) molecules cetuximab per cell after 24 hr (15.8% of added activity). In nude mice with subcutaneous FaDu xenograft tumors, a protein dose escalation study with (111) In-cetuximab showed highest specific accumulation in tumors at protein doses between 1 and 30 µg per mouse (mean tumor uptake 33.1 ± 3.1%ID/g, 3 days postinjection (p.i.)). The biodistribution of (111) In-cetuximab and (125) I-cetuximab was determined at 1, 3 and 7 days p.i. at optimal protein dose. Tumor uptake was favorable for (111) In-cetuximab compared to (125) I-cetuximab. With pixel-by-pixel analysis, good correlations were found between intratumoral distribution of (111) In-cetuximab as determined by autoradiography and EGFR expression in the same tumor sections as determined immunohistochemically (mean r = 0.74 ± 0.14; all correlations p < 0.0001). Micro Single Photon Emission Computed Tomography (MicroSPECT) scans clearly visualized FaDu tumors from 1 day p.i. onward and tumor-to-background contrast increased until 7 days p.i. (tumor-to-liver ratios 0.58 ± 0.24, 3.42 ± 0.66, 8.99 ± 4.66 and 16.33 ± 11.56, at day 0, day 1, day 3 and day 7 p.i., respectively). Our study suggests that, at optimal cetuximab imaging dose, (111) In-cetuximab can be used for visualization of EGFR expression in head-and-neck squamous cell carcinoma using SPECT.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Carcinoma de Células Escamosas/diagnóstico por imagem , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/diagnóstico por imagem , Radioisótopos de Índio , Ácido Pentético , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacocinética , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Cetuximab , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Imunofluorescência , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Técnicas Imunoenzimáticas , Radioisótopos de Índio/farmacocinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ácido Pentético/farmacocinética , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Nucl Med ; 51(10): 1565-72, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20847162

RESUMO

UNLABELLED: The insulinlike growth factor 1 receptor (IGF-1R) is a new target for the treatment of breast cancer. Patients with breast cancer lesions that express IGF-1R may benefit from treatment with anti-IGF-1R antibodies. Therefore, the aim of the present study was to develop a noninvasive, in vivo imaging method, using radiolabeled antibodies, to visualize IGF-1R expression. METHODS: R1507 is a monoclonal antibody directed against the IGF-1R. In vitro, the affinity and internalization kinetics of (111)In-R1507 were determined using the IGF-1R-expressing triple-negative breast cancer cell line SUM149. In vivo, the pharmacodynamics of (111)In-R1507 and (125)I-R1507 were determined in mice with subcutaneous SUM149 tumors. (111)In-R1507 SPECT and (89)Zr-R1507 PET images of mice with subcutaneous SUM149 tumors were acquired at 1, 3, and 7 d after injection. RESULTS: (111)In-R1507 (concentration required to inhibit binding by 50%, 0.1 nM) was slowly internalized by SUM149 cells. (111)In-R1507 specifically and efficiently accumulated in the SUM149 xenografts: the tumor uptake was 20 percentage injected dose per gram (%ID/g), 33 %ID/g, and 31 %ID/g at 1, 3, and 7 d after injection, respectively. (125)I-R1507 accumulated in the tumor less efficiently. Small-animal SPECT and small-animal PET of mice clearly visualized the subcutaneous SUM149 xenograft, with increasing contrast at later time points. CONCLUSION: (111)In-R1507 and (89)Zr-R1507 are new tracers to noninvasively determine IGF-1R expression in vivo in breast cancer xenografts using SPECT and PET. In the future, these techniques may enable patient selection for IGF-1R-targeted therapy.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/imunologia , Perfilação da Expressão Gênica/métodos , Radioimunodetecção/métodos , Receptor IGF Tipo 1/imunologia , Animais , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Feminino , Humanos , Marcação por Isótopo , Camundongos , Compostos Radiofarmacêuticos/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA