Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cancer Res Commun ; 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38747612

RESUMO

Cancer-associated fibroblasts (CAFs) are a prominent cell type within the tumor microenvironment where they are known to promote cancer cell growth and survival, angiogenesis, drug resistance, and immunosuppression. The transmembrane prolyl protease Fibroblast Activation Protein (FAP) is expressed on the surface of highly pro-tumorigenic CAFs found in the stroma of nearly every cancer of epithelial origin. The widespread expression of FAP has made it an attractive therapeutic target based on the underlying hypothesis that eliminating pro-tumorigenic CAFs will disrupt the crosstalk between components of TME resulting in cancer cell death and immune infiltration. This hypothesis, however, has never been directly proven. To eliminate FAP-expressing CAFs, we developed an antibody-drug conjugate (ADC) using our anti-FAP antibody, huB12, coupled to a monomethyl auristatin E (huB12-MMAE) payload. After determining that huB12 was an effective targeting vector, we found that huB12-MMAE potently eliminated FAP-expressing cells as monocultures in vitro and significantly prolonged survival in vivo using a xenograft engineered to overexpress FAP. We investigated the effects of selectively eliminating CAFs using a layered, open microfluidic cell co-culture platform, known as the Stacks. Analysis of mRNA and protein expression found that treatment with huB12-MMAE resulted in the increased secretion of the pro-inflammatory cytokines IL6 and IL8 by CAFs and an associated increase in expression of pro-inflammatory genes in cancer cells. We also detected increased secretion of CSF1, a cytokine involved in myeloid recruitment and differentiation. Our findings suggest that the mechanism of FAP-targeted therapies are through effects on the immune microenvironment and anti-tumor immune response.

2.
Sci Adv ; 10(15): eadj1444, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38598637

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease resulting in irreversible scarring within the lungs. However, the lack of biomarkers that enable real-time assessment of disease activity remains a challenge in providing efficient clinical decision-making and optimal patient care in IPF. Fibronectin (FN) is highly expressed in fibroblastic foci of the IPF lung where active extracellular matrix (ECM) deposition occurs. Functional upstream domain (FUD) tightly binds the N-terminal 70-kilodalton domain of FN that is crucial for FN assembly. In this study, we first demonstrate the capacity of PEGylated FUD (PEG-FUD) to target FN deposition in human IPF tissue ex vivo. We subsequently radiolabeled PEG-FUD with 64Cu and monitored its spatiotemporal biodistribution via µPET/CT imaging in mice using the bleomycin-induced model of pulmonary injury and fibrosis. We demonstrated [64Cu]Cu-PEG-FUD uptake 3 and 11 days following bleomycin treatment, suggesting that radiolabeled PEG-FUD holds promise as an imaging probe in aiding the assessment of fibrotic lung disease activity.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Animais , Camundongos , Distribuição Tecidual , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/diagnóstico por imagem , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Peptídeos/metabolismo , Bleomicina
3.
Adv Funct Mater ; 33(33)2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37942189

RESUMO

The therapeutic efficacy of photodynamic therapy is limited by the ability of light to penetrate tissues. Due to this limitation, Cerenkov luminescence (CL) from radionuclides has recently been proposed as an alternative light source in a strategy referred to as Cerenkov radiation induced therapy (CRIT). Semiconducting polymer nanoparticles (SPNs) have ideal optical properties, such as large absorption cross-sections and broad absorbance, which can be utilized to harness the relatively weak CL produced by radionuclides. SPNs can be doped with photosensitizers and have nearly 100% energy transfer efficiency by multiple energy transfer mechanisms. Herein, we investigated an optimized photosensitizer doped SPN as a nanosystem to harness and amplify CL for cancer theranostics. We found that semiconducting polymers significantly amplified CL energy transfer efficiency. Bimodal PET and optical imaging studies showed high tumor uptake and retention of the optimized SPNs when administered intravenously or intratumorally. Lastly, we found that photosensitizer doped SPNs have excellent potential as a cancer theranostics nanosystem in an in vivo tumor therapy study. Our study shows that SPNs are ideally suited to harness and amplify CL for cancer theranostics, which may provide a significant advancement for CRIT that are unabated by tissue penetration limits.

4.
J Control Release ; 350: 284-297, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35995299

RESUMO

In breast cancer, the extracellular matrix (ECM) undergoes remodeling and changes the tumor microenvironment to support tumor progression and metastasis. Fibronectin (FN) assembly is an important step in the regulation of the tumor microenvironment since the FN matrix precedes the deposition of various other ECM proteins, controls immune cell infiltration, and serves as a reservoir for cytokines and growth factors. Therefore, FN is an attractive target for breast cancer therapy and imaging. Functional Upstream Domain (FUD) is a 6-kDa peptide targeting the N-terminal 70-kDa domain of FN, which is critical for fibrillogenesis. FUD has previously been shown to function as an anti-fibrotic peptide both in vitro and in vivo. In this work, we conjugated the FUD peptide with 20-kDa of PEG (PEG-FUD) and demonstrated its improved tumor exposure compared to non-PEGylated FUD in a murine breast cancer model via multiple imaging modalities. Importantly, PEG-FUD peptide retained a nanomolar binding affinity for FN and maintained in vitro plasma stability for up to 48 h. Cy5-labeled PEG-FUD bound to exogenous or endogenous FN assembled by fibroblasts. The in vivo fluorescence imaging with Cy5-labeled FUD and FUD conjugates demonstrated that PEGylation of the FUD peptide enhanced blood exposure after subcutaneous (SC) injection and significantly increased accumulation of FUD peptide in 4T1 mammary tumors. Intravital microscopy confirmed that Cy5-labeled PEG-FUD deposited mostly in the extravascular region of the tumor microenvironment after SC administration. Lastly, positron emission tomography/computed tomography imaging showed that 64Cu-labeled PEG-FUD preferentially accumulated in the 4T1 tumors with improved tumor uptake compared to 64Cu-labeled FUD (48 h: 1.35 ± 0.05 vs. 0.59 ± 0.03 %IA/g, P < 0.001) when injected intravenously (IV). The results indicate that PEG-FUD targets 4T1 breast cancer with enhanced tumor retention compared to non-PEGylated FUD, and biodistribution profiles of PEG-FUD after SC and IV injection may guide the optimization of PEG-FUD as a therapeutic and/or imaging agent for use in vivo.


Assuntos
Neoplasias da Mama , Fibronectinas , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Carbocianinas , Citocinas/metabolismo , Feminino , Fibronectinas/metabolismo , Humanos , Camundongos , Imagem Multimodal , Peptídeos/química , Distribuição Tecidual , Microambiente Tumoral
5.
Eur J Nucl Med Mol Imaging ; 49(11): 3705-3716, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35556159

RESUMO

PURPOSE: The lack of effective molecular biomarkers to monitor idiopathic pulmonary fibrosis (IPF) activity or treatment response remains an unmet clinical need. Herein, we determined the utility of fibroblast activation protein inhibitor for positron emission tomography (FAPI PET) imaging in a mouse model of pulmonary fibrosis. METHODS: Pulmonary fibrosis was induced by intratracheal administration of bleomycin (1 U/kg) while intratracheal saline was administered to control mice. Subgroups from each cohort (n = 3-5) underwent dynamic 1 h PET/CT after intravenously injecting FAPI-46 radiolabeled with gallium-68 ([68 Ga]Ga-FAPI-46) at 7 days and 14 days following disease induction. Animals were sacrificed following imaging for ex vivo gamma counting and histologic correlation. [68 Ga]Ga-FAPI-46 uptake was quantified and reported as percent injected activity per cc (%IA/cc) or percent injected activity (%IA). Lung CT density in Hounsfield units (HU) was also correlated with histologic examinations of lung fibrosis. RESULTS: CT only detected differences in the fibrotic response at 14 days post-bleomycin administration. [68 Ga]Ga-FAPI-46 lung uptake was significantly higher in the bleomycin group than in control subjects at 7 days and 14 days. Significantly (P = 0.0012) increased [68 Ga]Ga-FAPI-46 lung uptake in the bleomycin groups at 14 days (1.01 ± 0.12%IA/cc) vs. 7 days (0.33 ± 0.09%IA/cc) at 60 min post-injection of the tracer was observed. These findings were consistent with an increase in both fibrinogenesis and FAP expression as seen in histology. CONCLUSION: CT was unable to assess disease activity in a murine model of IPF. Conversely, FAPI PET detected both the presence and activity of lung fibrogenesis, making it a promising tool for assessing early disease activity and evaluating the efficacy of therapeutic interventions in lung fibrosis patients.


Assuntos
Fibrose Pulmonar Idiopática , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Animais , Bleomicina , Radioisótopos de Gálio , Humanos , Fibrose Pulmonar Idiopática/diagnóstico por imagem , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Tomografia por Emissão de Pósitrons , Quinolinas
6.
Adv Sci (Weinh) ; 8(10): 2001879, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34026426

RESUMO

Lymphoma is a heterogeneous disease with varying clinical manifestations and outcomes. Many subtypes of lymphoma, such as Burkitt's lymphoma and diffuse large B cell lymphoma, are highly aggressive with dismal prognosis even after conventional chemotherapy and radiotherapy. As such, exploring specific biomarkers for lymphoma is of high clinical significance. Herein, a potential marker, CD38, is investigated for differentiating lymphoma. A CD38-targeting monoclonal antibody (mAb, daratumumab) is then radiolabeled with Zr-89 and Lu-177 for theranostic applications. As the diagnostic component, the Zr-89-labeled mAb is highly specific in delineating CD38-positive lymphoma via positron emission tomography (PET) imaging, while the Lu-177-labeled mAb serves well as the therapeutic component to suppress tumor growth after a one-time administration. These results strongly suggest that CD38 is a lymphoma-specific marker and prove that 89Zr/177Lu-labeled daratumumab facilitates immunoPET imaging and radioimmunotherapy of lymphoma in preclinical models. Further clinical evaluation and translation of this CD38-targeted theranostics may be of significant help in lymphoma patient stratification and management.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Anticorpos Monoclonais/farmacologia , Lutécio/farmacocinética , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Glicoproteínas de Membrana/imunologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Medicina de Precisão/métodos , Radioisótopos/farmacocinética , Zircônio/farmacocinética , ADP-Ribosil Ciclase 1/metabolismo , Animais , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Humanos , Fatores Imunológicos/farmacocinética , Linfoma Difuso de Grandes Células B/imunologia , Linfoma Difuso de Grandes Células B/patologia , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos SCID , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/farmacologia , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nano Lett ; 21(11): 4692-4699, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34029471

RESUMO

Theranostic nanoparticles hold the potential to greatly improve cancer management by providing personalized medicine. Although many theranostic nanoconstructs have been successful in preclinical studies, clinical translation is still hampered by their limited targeting capability and lack of successful therapeutic efficacy. We report the use of novel ultrasmall porous silica nanoparticles (UPSN) with enhanced in vivo pharmacokinetics such as high target tissue accumulation (12% ID/g in the tumor) and evasion from the reticuloendothelial system (RES) organs. Herein, UPSN is conjugated with the isotopic pair 90/86Y, enabling both noninvasive imaging as well as internal radiotherapy. In vivo PET imaging demonstrates prolonged blood circulation and excellent tumor contrast with 86Y-DOTA-UPSN. Tumor-to-muscle and tumor-to-liver uptake values were significantly high (12.4 ± 1.7 and 1.5 ± 0.5, respectively), unprecedented for inorganic nanomaterials. 90Y-DOTA-UPSN significantly inhibits tumor growth and increases overall survival, indicating the promise of UPSN for future clinical translation as a cancer theranostic agent.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Porosidade , Medicina de Precisão , Dióxido de Silício
8.
J Nucl Med ; 62(3): 372-378, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32826320

RESUMO

CD20-overexpressed non-Hodgkin lymphoma typically indicates progressive malignancy. Obinutuzumab is a next-generation Food and Drug Administration-approved humanized monoclonal antibody that targets CD20. Previous studies with 89Zr-labeled obinutuzumab have successfully imaged CD20 in vivo. However, delayed tumor uptake and increased radioactive exposure caused by long blood circulation limit its clinical translation. This study aimed to develop 64Cu-labeled F(ab')2 fragments of obinutuzumab for imaging CD20 in lymphoma xenograft tumor models. Methods: F(ab')2 fragments were produced from obinutuzumab using an IgG-degrading enzyme of Streptococcus pyogenes (IdeS) enzyme and purified with protein A beads. Sodium dodecyl sulfate polyacrylamide gel electrophoresis and high-performance liquid chromatography were performed to evaluate the products and their stability. F(ab')2 products were conjugated with p-SCN-Bn-NOTA (NOTA) for 64Cu radiolabeling. Western blotting was performed to screen the CD20 expression levels of lymphoma cells. Enzyme-linked immunosorbent assay, flow cytometry, and confocal imaging were used to test the binding affinity in vitro. Serial PET imaging and biodistribution studies in subcutaneous lymphoma-bearing mice were performed using 64Cu-NOTA-F(ab')2-obinutuzumab or 64Cu-NOTA-F(ab')2-IgG. Results: F(ab')2-obinutuzumab and F(ab')2-IgG produced by the IdeS digestion system were confirmed with sodium dodecyl sulfate polyacrylamide gel electrophoresis and high-performance liquid chromatography. The radiochemical purity of 64Cu-labeled F(ab')2 fragments was no less than 98%, and the specific activity was 56.3 ± 7.9 MBq/mg (n = 6). Among the 5 lymphoma cell lines, Ramos showed the strongest expression of CD20, and CLL-155 showed the lowest, as confirmed by enzyme-linked immunosorbent assay, flow cytometry, and confocal imaging. PET imaging revealed rapid and sustained tumor uptake of 64Cu-NOTA-F(ab')2-obinutuzumab in Ramos tumor-bearing mice. The peak tumor uptake (9.08 ± 1.67 percentage injected dose per gram of tissue [%ID/g]) in the Ramos model was significantly higher than that in the CCL-155 model (2.78 ± 0.62 %ID/g) or the 64Cu-NOTA-F(ab')2-IgG control (1.93 ± 0.26 %ID/g, n = 4, P < 0.001). The tumor-to-blood and tumor-to-muscle ratios were 7.3 ± 1.6 and 21.9 ± 9.0, respectively, at 48 h after injection in the 64Cu-NOTA-F(ab')2-obinutuzumab group. Of the measured off-target organs, the kidneys showed the highest uptake. Ex vivo immunofluorescent staining verified the differential CD20 expression in the Ramos and CCL-155 tumor models. Conclusion: This study demonstrated that 64Cu-NOTA-F(ab')2-obinutuzumab had a rapid and sustained tumor uptake in CD20-positive lymphoma with high contrast, which could enable noninvasive evaluation of CD20 levels in the clinic.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Antígenos CD20/metabolismo , Radioisótopos de Cobre/química , Regulação Neoplásica da Expressão Gênica , Fragmentos Fab das Imunoglobulinas/química , Linfoma/metabolismo , Animais , Antígenos CD20/imunologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Humanos , Marcação por Isótopo , Linfoma/diagnóstico por imagem , Masculino , Camundongos
9.
Adv Healthc Mater ; 10(5): e2000690, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32691969

RESUMO

Advances in technology and nanomedicine have led to the development of nanoparticles that can be activated for multimodal imaging of cancer, where a stimulus induces a material modification that enhances image contrast. Multimodal imaging using nanomaterials with this capability can combine the advantages and overcome the limitations of any single imaging modality. When designed with stimuli-responsive abilities, the target-to-background ratio of multimodal imaging nanoprobes increases because specific stimuli in the tumor enhance the signal. Several aspects of the tumor microenvironment can be exploited as an internal stimulus response for multimodal imaging applications, such as the pH gradient, redox processes, overproduction of various enzymes, or combinations of these. In this review, design strategies are discussed and an overview of the recent developments of internally responsive multimodal nanomaterials is provided. Properly implementing this approach improves noninvasive cancer diagnosis and staging as well as provides a method to monitor drug delivery and treatment response.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , Imagem Multimodal , Nanomedicina , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral
10.
Acc Chem Res ; 53(9): 1869-1880, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32786331

RESUMO

Nanomedicine has benefited from recent advances in chemistry and biomedical engineering to produce nanoscale materials as theranostic agents. Well-designed nanomaterials may present optimal biological properties, influencing circulation, retention, and excretion for imaging and treatment of various diseases. As the understanding of nanomedicine pharmacokinetics expands continuously, efficient renal clearance of nanomedicines can significantly increase the signal-to-background ratio for precision diagnosis and lower potential toxicity for improved treatment. Studies on nanomaterial-kidney interactions have led to many novel findings on the underlying principles of nanomaterial renal clearance, targeting, and accumulation. In return, the optimized nanomedicines confer significant benefits to the detection and treatment of kidney dysfunction.In this Account, we present an overview of recent progress in the development of nanomaterials for kidney theranostics, aiming to speed up translation and expand possible applications. We start by introducing biological structures of the kidney and their influence on renal targeting, retention, and clearance. Several key factors regarding renal accumulation and excretion, including nanomaterial types, sizes, and shapes, surface charges, and chemical modifications, are identified and discussed. Next, we highlight our recent efforts investigating kidney-interacting nanomaterials and introduce representative nanomedicines for imaging and treatment of kidney diseases. Multiple renal-clearable and renal-accumulating nanomedicines were devised for kidney function imaging. By employing renal-clearable nanomedicines, including gold nanoparticles, porphyrin polymers, DNA frameworks, and polyoxometalate clusters, we were able to noninvasively evaluate split renal function in healthy and diseased mice. Further engineering of renal-accumulating nanosystems has shifted attention from renal diagnosis to precision kidney protection. Many biocompatible nanomedicines, such as DNA origami, selenium-doped carbon quantum dots, melanin nanoparticles, and black phosphorus have all played essential roles in diminishing excessive reactive oxygen species for kidney treatment and protection. Finally, we discuss the challenges and perspectives of nanomaterials for renal care, their future clinical translation, and how they may affect the current landscape of clinical practices. We believe that this Account updates our current understanding of nanomaterial-kidney interactions for further design and control of nanomedicines for specific kidney diagnosis and treatment. This timely Account will generate broad interest in integrating nanotechnology and nanomaterial-biological interaction for state-of-the-art theranostics of renal diseases.


Assuntos
Nefropatias/prevenção & controle , Nanomedicina , Nanoestruturas/química , Animais , Materiais Biocompatíveis/química , Meios de Contraste/química , DNA/química , Taxa de Filtração Glomerular , Glucocorticoides/uso terapêutico , Ouro/química , Rim/diagnóstico por imagem , Rim/fisiologia , Nefropatias/diagnóstico por imagem , Nefropatias/tratamento farmacológico , Imageamento por Ressonância Magnética , Nanopartículas Metálicas/química , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X
11.
Am J Transl Res ; 12(5): 1491-1514, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32509158

RESUMO

Cardiovascular diseases (CVDs) have been the leading cause of death in United States. While tremendous progress has been made for treating CVDs over the year, the high prevalence and substantial medical costs requires the necessity for novel methods for the early diagnosis and treatment monitoring of CVDs. Macrophages are a promising target due to its crucial role in the progress of CVDs (atherosclerosis, myocardial infarction and inflammatory cardiomyopathies). Positron emission tomography (PET) is a noninvasive imaging technique with high sensitivity and provides quantitive functional information of the macrophages in CVDs. Although 18F-FDG can be taken up by active macrophages, the PET imaging tracer is non-specific and susceptible to blood glucose levels. Thus, developing more specific PET tracers will help us understand the role of macrophages in CVDs. Moreover, macrophage-targeted PET imaging will further improve the diagnosis, treatment monitoring, and outcome prediction for patients with CVDs. In this review, we summarize various targets-based tracers for the PET imaging of macrophages in CVDs and highlight research gaps to advise future directions.

12.
Adv Sci (Weinh) ; 7(12): 2000420, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32596126

RESUMO

The manifestation of acute kidney injury (AKI) is associated with poor patient outcomes, with treatment options limited to hydration or renal replacement therapies. The onset of AKI is often associated with a surfeit of reactive oxygen species. Here, it is shown that selenium-doped carbon quantum dots (SeCQDs) have broad-spectrum antioxidant properties and prominent renal accumulation in both healthy and AKI mice. Due to these properties, SeCQDs treat or prevent two clinically relevant cases of AKI induced in murine models by either rhabdomyolysis or cisplatin using only 1 or 50 µg per mouse, respectively. The attenuation of AKI in both models is confirmed by blood serum measurements, kidney tissue staining, and relevant biomarkers. The therapeutic efficacy of SeCQDs exceeds amifostine, a drug approved by the Food and Drug Administration that also acts by scavenging free radicals. The findings indicate that SeCQDs show great potential as a treatment option for AKI and possibly other ROS-related diseases.

13.
Chem Rev ; 120(8): 3787-3851, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32202104

RESUMO

Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.


Assuntos
Técnicas Imunológicas/métodos , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Humanos , Neoplasias/imunologia , Compostos Radiofarmacêuticos/imunologia , Anticorpos de Domínio Único/imunologia
14.
Mol Pharm ; 17(5): 1697-1705, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32202792

RESUMO

Pancreatic cancer is highly aggressive, with a median survival time of less than 6 months and a 5-year overall survival rate of around 7%. The poor prognosis of PaCa is largely due to its advanced stage at diagnosis and the lack of efficient therapeutic options. Thus, the development of an efficient, multifunctional PaCa theranostic system is urgently needed. Overexpression of tissue factor (TF) has been associated with increased tumor growth, angiogenesis, and metastasis in many malignancies, including pancreatic cancer. Herein, we propose the use of a TF-targeted monoclonal antibody (ALT836) conjugated with the pair 86/90Y as a theranostic agent against pancreatic cancer. For methods, serial PET imaging with 86Y-DTPA-ALT836 was conducted to map the biodistribution the tracer in BXPC-3 tumor-bearing mice. 90Y-DTPA-ALT836 was employed as a therapeutic agent that also allowed tumor burden monitoring through Cherenkov luminescence imaging. The results were that the uptake of 86Y-DTPA-ALT836 in BXPC-3 xenograft tumors was high and increased over time up to 48 h postinjection (p.i.), corroborated through ex vivo biodistribution studies and further confirmed by Cherenkov luminescence Imaging. In therapeutic studies, 90Y-DTPA-ALT836 was found to slow tumor growth relative to the control groups and had significantly smaller (p < 0.05) tumor volumes 1 day p.i. Histological analysis of ex vivo tissues revealed significant damage to the treated tumors. The conclusion is that the use of the 86/90Y theranostic pair allows PET imaging with excellent tumor-to-background contrast and treatment of TF-expressing pancreatic tumors with promising therapeutic outcomes.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Tromboplastina/antagonistas & inibidores , Radioisótopos de Ítrio/farmacocinética , Animais , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Feminino , Camundongos , Neoplasias Pancreáticas/patologia , Tomografia por Emissão de Pósitrons , Distribuição Tecidual
16.
Am J Cancer Res ; 9(11): 2413-2427, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31815043

RESUMO

Clinical management of anaplastic thyroid cancer (ATC) is very challenging due to its dedifferentiation and aggressiveness. We aim to develop HER2-targeted multimodal imaging approaches and assess the diagnostic efficacies of these molecular imaging probes in preclinical ATC models. Flow cytometry was used to detect HER2 expression status in thyroid cancer cell lines. We then developed a HER2-specific immunoPET imaging probe 89Zr-Df-pertuzumab by radiolabeling a HER-2 specific monoclonal antibody (mAb) pertuzumab with 89Zr (t1/2=78.4 h) and a fluorescent imaging probe IRDye 800CW-pertuzumab. The diagnostic efficacies of the probes were assessed in subcutaneous and orthotopic ATC models, followed by ex vivo biodistribution profile and immunofluorescence staining studies. HER2 was highly expressed on the surface of all the four primary thyroid cancer cell lines examined, which included two ATC cell lines (i.e., 8505C and THJ-16T). PET imaging with 89Zr-Df-pertuzumab clearly visualized all the subcutaneous ATCs with a peak tumor uptake of 20.23±6.44 %ID/g (n=3), whereas the highest tumor uptake of the nonspecific probe 89Zr-Df-IgG in subcutaneous ATC models was 6.30±0.95 %ID/g (n=3). More importantly, 89Zr-Df-pertuzumab PET imaging strategy readily delineated all the orthotopic ATCs with a peak tumor uptake of 24.93±8.53 %ID/g (n=3). We also suggested that Cerenkov luminescence imaging (CLI) using 89Zr-Df-pertuzumab and fluorescence imaging using IRDye 800CW-pertuzumab are useful tools for image-guided removal of ATCs. We demonstrate that HER2 is a promising biomarker for ATC, and multimodal imaging using 89Zr-Df-pertuzumab and IRDye 800CW-pertuzumab is useful for identifying HER2-postive ATCs.

17.
Adv Mater ; 31(52): e1904894, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31709622

RESUMO

Cerenkov radiation (CR) from radionuclides can act as a built-in light source for cancer theranostics, opening a new horizon in biomedical applications. However, considerably low tumor-targeting efficiency of existing radionuclides and radionuclide-based nanomedicines limits the efficacy of CR-induced theranostics (CRIT). It remains a challenge to precisely and efficiently supply CR energy to the tumor site. Here, a "missile-detonation" strategy is reported, in which a high dose of p-SCN-Bn-deferoxamine-porphyrin-PEG nanocomplex (Df-PPN) is first adminstered as a CR energy receiver/missile to passively target to tumor, and then a low dose of the 89 Zr-labeled Df-PPN is administrated as a CR energy donor/detonator, which can be visualized and quantified by Cerenkov energy transfer imaging, positron-emission tomography, and fluorescence imaging. Based on homologous properties, the colocalization of Df-PPN and 89 Zr-Df-PPN in the tumor site is maximized and efficient CR energy transfer is enabled, which maximizes the tumor-targeted CRIT efficacy in an optimal spatiotemporal setting while also reducing adverse off-target effects from CRIT. This precise and efficient CRIT strategy causes significant tumor vascular damage and inhibited tumor growth.


Assuntos
Fármacos Fotossensibilizantes/química , Nanomedicina Teranóstica , Animais , Linhagem Celular Tumoral , Desferroxamina/química , Transferência de Energia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanoestruturas/química , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Estresse Oxidativo/efeitos dos fármacos , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Polietilenoglicóis/química , Porfirinas/química , Tomografia por Emissão de Pósitrons , Radioisótopos/química , Distribuição Tecidual , Transplante Heterólogo , Zircônio/química
18.
Adv Sci (Weinh) ; 6(22): 1901724, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31763153

RESUMO

Hydrogen sulfide (H2S) is of vital importance in several biological and physical processes. The significance of H2S-specific detection and monitoring is emphasized by its elevated levels in various diseases such as cancer. Nanotechnology enhances the performance of chemical sensing nanoprobes due to the enhanced efficiency and sensitivity. Recently, extensive research efforts have been dedicated to developing novel smart H2S-triggered/therapeutic system (SHTS) nanoplatforms for H2S-activated sensing, imaging, and therapy. Herein, the latest SHTS-based nanomaterials are summarized and discussed in detail. In addition, therapeutic strategies mediated by endogenous H2S as a trigger or exogenous H2S delivery are also included. A comprehensive understanding of the current status of SHTS-based strategies will greatly facilitate innovation in this field. Lastly, the challenges and key issues related to the design and development of SHTS-based nanomaterials (e.g., morphology, surface modification, therapeutic strategies, appropriate application, and selection of nanomaterials) are outlined.

19.
ACS Nano ; 13(11): 13382-13389, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31603304

RESUMO

Oxidative stress is one of the important mechanisms in cerebral ischemia/reperfusion (I/R) injury. Antioxidants with high brain accumulation are highly desired to help prevent cerebral I/R injury. Herein, intrathecal injection of polyoxometalate (POM) nanoclusters as nano-antioxidants with preferential brain uptake were applied for neuronal protection in cerebral I/R injury. Using powerful positron emission tomography imaging, the uptake of nano-antioxidants in the brain was non-invasively and real-timely monitored. Our results demonstrated that POM nanoclusters rapidly reached the ischemic penumbra after intrathecal injection and effectively scavenged reactive oxygen species (ROS) for inhibiting oxidative stress. The infarct size was reduced, and neurological function was restored in cerebral I/R injury rat models. As a proof-of-concept, the intrathecal injection of nano-antioxidants is an excellent therapeutic strategy to ameliorate cerebral I/R injury.


Assuntos
Antioxidantes/farmacologia , Isquemia Encefálica/tratamento farmacológico , Nanopartículas/química , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Compostos de Tungstênio/farmacologia , Animais , Antioxidantes/administração & dosagem , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Injeções Espinhais , Imageamento por Ressonância Magnética , Nanopartículas/administração & dosagem , Neurônios/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/diagnóstico por imagem , Traumatismo por Reperfusão/metabolismo , Compostos de Tungstênio/administração & dosagem
20.
Nano Lett ; 19(10): 7334-7341, 2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31518140

RESUMO

Effective therapy for protecting dying neurons against cerebral ischemia-reperfusion injury (IRI) represents a substantial challenge in the treatment of ischemic strokes. Oxidative stress coupled with excessive inflammation is the main culprit for brain IRI that results in neuronal damage and disability. Specifically, complement component 5a (C5a) exacerbates the vicious cycle between oxidative stress and inflammatory responses. Herein, we propose that a framework nucleic acid (FNA) conjugated with anti-C5a aptamers (aC5a) can selectively reduce C5a-mediated neurotoxicity and effectively alleviate oxidative stress in the brain. Intrathecal injection of the aC5a-conjugated FNA (aC5a-FNA) was applied for the treatment of rats with ischemic strokes. Positron emission tomography (PET) imaging was performed to investigate the accumulation of aC5a-FNA in the penumbra and its therapeutic efficacy. Results demonstrated that aC5a-FNA could rapidly penetrate different brain regions after brain IRI. Furthermore, aC5a-FNA effectively protected neurons from brain IRI, as verified by serum tests, tissue staining, biomarker detection, and functional assessment. The protective effect of aC5a-FNA against cerebral IRI in living animals may pave the way for the translation of FNA from bench to bedside and broaden the horizon of FNA in the field of biomedicine.


Assuntos
Aptâmeros de Nucleotídeos/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Complemento C5a/antagonistas & inibidores , Ácidos Nucleicos/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Aptâmeros de Nucleotídeos/administração & dosagem , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Complemento C5a/imunologia , Injeções Espinhais , Ácidos Nucleicos/administração & dosagem , Ratos Sprague-Dawley , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA