Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Pharmaceutics ; 13(12)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34959463

ABSTRACT

Despite the recent successes in siRNA therapeutics, targeted delivery beyond the liver remains the major hurdle for the widespread application of siRNA in vivo. Current cationic liposome or polymer-based delivery agents are restricted to the liver and suffer from off-target effects, poor clearance, low serum stability, and high toxicity. In this study, we genetically engineered a non-cationic non-viral tumor-targeted universal siRNA nanocarrier (MW 26 KDa). This protein nanocarrier consists of three function domains: a dsRNA binding domain (dsRBD) (from human protein kinase R) for any siRNA binding, 18-histidine for endosome escape, and two RGD peptides at the N- and C-termini for targeting tumor and tumor neovasculature. We showed that cloned dual-RGD-dsRBD-18his (dual-RGD) protein protects siRNA against RNases, induces effective siRNA endosomal escape, specifically targets integrin αvß3 expressing cells in vitro, and homes siRNA to tumors in vivo. The delivered siRNA leads to target gene knockdown in the cell lines and tumor xenografts with low toxicity. This multifunctional and biomimetic siRNA carrier is biodegradable, has low toxicity, is suitable for mass production by fermentation, and is serum stable, holding great potential to provide a widely applicable siRNA carrier for tumor-targeted siRNA delivery.

2.
Med Oncol ; 37(11): 98, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33034761

ABSTRACT

Prostate-specific membrane antigen (PSMA; also termed glutamate carboxypeptidase II (GCP II)) is abundantly expressed in prostate cancer. It has been shown recently that PSMA is expressed in neovasculature of differentiated thyroid cancer. In this study, we show that 18F-DCFPyl might detect neovasculature in advanced, metastatic differentiated thyroid cancer (DTC). We first stained the preserved lymph node samples of three patients with DTC who had undergone total thyroidectomy and neck dissection for cervical lymph node metastatic disease to identify PSMA expression, with the PSMA antibody (DAKO Monoclonal). Then, we performed 18F-DCFPyl imaging in two other advanced DTC patients with elevated serum thyroglobulin (Tg), indicative of residual disease. We compared the findings with contemporaneous FDG PET/CT scan, conventional Imaging (CT,MRI) and whole-body scan performed with I123/I131. All the three lymph node samples stained positive for PSMA expression in the neovasculature. In the first imaged patient, 18F-DCFPyl detected activity within the retropharyngeal CT contrast-enhancing lymph node. Compared to FDG PET/CT, the 18F-DCFPyl scan showed a greater SUV (3.1 vs 1.8). In the second imaged patient, 18F-DCFPyl showed intense uptake in the L3 vertebra (not seen on the post treatment 131I scan or the 18F-FDG PET/CT). MRI of the lumbar spine confirmed the presence of sclerotic-lytic lesion at the location, consistent with metastatic disease. Our exploratory study is proof of principle, that the prostate cancer imaging agent 18F-DCFPyl may prove useful for the localization of metastases, in patients with metastatic RAI-refractory DTC by detecting neoangiogenesis within the tumor.


Subject(s)
Antigens, Surface/metabolism , Fluorine Radioisotopes/therapeutic use , Glutamate Carboxypeptidase II/metabolism , Neovascularization, Pathologic/diagnostic imaging , Radiopharmaceuticals/therapeutic use , Thyroid Neoplasms/diagnostic imaging , Adult , Drug Resistance, Neoplasm , Fluorine Radioisotopes/metabolism , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/therapeutic use , Humans , Iodine Radioisotopes/metabolism , Iodine Radioisotopes/therapeutic use , Male , Middle Aged , Neoplasm, Residual , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Whole Body Imaging
3.
Annu Rev Med ; 70: 461-477, 2019 01 27.
Article in English | MEDLINE | ID: mdl-30691373

ABSTRACT

In recent years, small-molecule inhibitors of prostate-specific membrane antigen (PSMA) labeled with radionuclides that allow for positron emission tomography (PET) imaging have been extensively studied in many clinical contexts in men with prostate cancer (PCa). The high sensitivity and specificity of these agents for identifying sites of PCa has quickly led to their widespread adoption as a de facto clinical standard of care throughout much of the world. PSMA-targeted PET radiotracers have been particularly well-studied in preoperatively staging men with high-risk PCa, evaluating biochemical recurrence following definitive therapy, and guiding metastasis-directed therapy in patients suspected of having oligorecurrent/oligometastatic disease. Furthermore, the expression of PSMA on the tumor neovasculature of many nonprostate malignancies has enabled a burgeoning subfield concentrated on delineating the potential utility of PSMA-targeted PET agents for imaging other cancers. In this review, we highlight the preclinical development of key small molecules that are now being clinically utilized for PCa imaging, discuss the roles of PSMA-targeted agents in guiding patient management, and consider the role these compounds may play in imaging nonprostate cancers.


Subject(s)
Antigens, Surface/analysis , Glutamate Carboxypeptidase II/analysis , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Radioisotopes/pharmacology , Aged , Animals , Antigens, Surface/radiation effects , Cohort Studies , Glutamate Carboxypeptidase II/radiation effects , Humans , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Preoperative Care/methods , Prostatectomy/methods , Prostatic Neoplasms/surgery , Retrospective Studies , Sensitivity and Specificity
4.
J Nucl Med ; 59(7): 1007-1013, 2018 07.
Article in English | MEDLINE | ID: mdl-29674422

ABSTRACT

In this review, we cover the evolution of knowledge on the biology of prostate-specific membrane antigen (PSMA) and its translation to therapy. The usual key to discovery is a realistic model for experimentation and for testing a hypothesis. A realistic model is especially needed in the case of the human prostate, which differs significantly from the prostate of species often used as research models. We will emphasize the genetic characterization of PSMA, the nature of the PSMA protein, and its role as a carboxypeptidase, with differing important substrates and products in different tissues. We give special prominence to the importance of PSMA as a target for imaging and therapy in prostate cancer and its underdeveloped role for imaging and targeting the neovasculature of tumors other than prostate cancer. Lastly, we bring attention to its importance in other nonprostatic tissues.


Subject(s)
Diagnostic Imaging/methods , Glutamate Carboxypeptidase II/metabolism , Radiotherapy/methods , Folic Acid/metabolism , Humans , Male , Molecular Targeted Therapy , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism
5.
J Neurooncol ; 138(3): 581-589, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29524126

ABSTRACT

Tumor angiogenesis has attracted increasing attention because of its potential as a valuable marker in the differential diagnosis of brain tumors as well as a novel therapeutic target. Prostate-specific membrane antigen (PSMA) is expressed by the neovasculature endothelium of some tumors, with little to no expression by the tumor cells or normal vasculature endothelium. The aim of this study was to investigate the potential of PSMA for the evaluation of the tumor neovasculature of various brain tumors and the possibility of detecting PSMA expression in brain tumors using PET imaging with 89Zr-Df-IAB2M (anti-PSMA minibody). Eighty-three tissue specimens including gliomas, metastatic brain tumors, primary central nervous system lymphomas (PCNSL), or radiation necroses were analyzed by immunohistochemical staining with PSMA antibody. 89Zr-Df-IAB2M PET scans were performed in three patients with recurrent high-grade gliomas or metastatic brain tumor. PSMA was highly expressed in the vascular endothelium of high-grade glioma and metastatic brain tumor, whereas PSMA was poorly expressed in the vascular endothelium of PCNSL and radiation necrosis. PSMA expression in high-grade gliomas and a metastatic brain tumor was clearly visualized by PET imaging with 89Zr-Df-IAB2M. Furthermore, a trend toward a positive correlation between the degree of 89Zr-Df-IAB2M uptake and PSMA expression levels in tumor specimens was observed. PET imaging of PSMA using 89Zr-Df-IAB2M may have potential value in the differential diagnosis of high-grade glioma from PCNSL or radiation necrosis as well as in the prediction of treatment efficacy and assessment of treatment response to bevacizumab therapy for high-grade glioma.


Subject(s)
Antigens, Surface/metabolism , Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Endothelium, Vascular/metabolism , Glutamate Carboxypeptidase II/metabolism , Neovascularization, Pathologic/diagnostic imaging , Positron-Emission Tomography , Adult , Aged , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Endothelium, Vascular/pathology , Female , Glioma/blood supply , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/pathology , Humans , Immunoglobulins/administration & dosage , Immunoglobulins/analysis , Lung Neoplasms/blood supply , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphoma/diagnostic imaging , Lymphoma/metabolism , Lymphoma/pathology , Male , Middle Aged , Necrosis/diagnostic imaging , Necrosis/etiology , Necrosis/metabolism , Necrosis/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Radiation Injuries/diagnostic imaging , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radioisotopes/administration & dosage , Radioisotopes/analysis , Radiopharmaceuticals , Zirconium/administration & dosage , Zirconium/analysis
6.
ACS Biomater Sci Eng ; 4(6): 1986-1999, 2018 Jun 11.
Article in English | MEDLINE | ID: mdl-33445269

ABSTRACT

Breast cancer has been the first killer among women. In this study, combretastatin A-4 (CA-4) loaded 5-amino acid peptide Ala-Pro-Arg-Pro-Gly (APRPG) modified PEG-PDLLA mixed micelles was developed to target tumor neovasculature for breast cancer therapy. CA-4 is an effective vascular disrupting agent. The APRPG-modified PEG-PDLLA polymer was successfully synthesized and thin-film hydration method was used to prepare APRPG-PEG-PDLLA/MPEG-PDLLA mixed micelles. Drug loading capacity (DL), encapsulation efficiency (EE), and the optimized ratio of APRPG-PEG-PDLLA: MPEG-PDLLA for efficient drug loading was investigated. The particle size, zeta potential, morphology, and the crystallographic study were carried out to characterize the micelles. In vitro release study revealed a sustained release of CA-4 from the mixed micelles while compared to free CA-4. Moreover, the cytotoxicity data of blank and drug loaded mixed micelles suggested that the APRPG-PEG-PDLLA/MPEG-PDLLA mixed micelles were safe drug carriers and the encapsulated CA-4 remained potent antitumor effect. The cellular uptake study and the in vivo imaging and biodistribution study demonstrated that the APRPG peptide modified mixed micelles has the higher cellular uptake efficiency and could significantly facilitate the accumulation at tumor site. Furthermore, the micelles were slowly extravasated from blood vessels and inhibited embryonic angiogenesis in the transgenic zebrafish model. Consequently, the CA-4 loaded APRPG-PEG-PDLLA/MPEG-PDLLA mixed micelles group demonstrated a significant inhibition of tumor growth in 4T1 breast cancer model. In short, the CA-4 loaded APRPG-PEG-PDLLA/MPEG-PDLLA mixed micelles might have great potential for breast cancer therapy.

7.
Theranostics ; 7(19): 4862-4876, 2017.
Article in English | MEDLINE | ID: mdl-29187909

ABSTRACT

Background: Tumor vessels can potentially serve as diagnostic, prognostic and therapeutic targets for solid tumors. Fluorescent dyes are commonly used as biological indicators, while photobleaching seriously hinders their application. In this study, we aim to generate a fluorescent silica nanoparticles (FSiNPs) theranostic system marked by the mouse endgolin (mEND) aptamer, YQ26. Methods: A highly specific YQ26 was selected by using gene-modified cell line-based SELEX technique. FSiNPs were prepared via the reverse microemulsion method. The YQ26-FSiNPs theranostic system was developed by combining YQ26 with the FSiNPs for in vivo tumor imaging, treatment and monitoring. Results: Both in vitro experiments (i.e. cellular and tumor tissue targeting assays) and in vivo animal studies (i.e. in vivo imaging and antitumor efficacy of YQ26-FSiNPs) clearly demonstrated that YQ26-FSiNPs could achieve prominently high targeting efficiency and therapeutic effects via aptamer YQ26-mediated binding to endoglin (END) molecule. Conclusion: This simple, sensitive, and specific YQ26-FSiNPs theranostic system has a great potential for clinical tumor targeting imaging and treatment.


Subject(s)
Aptamers, Nucleotide/pharmacokinetics , Endoglin/metabolism , Nanoparticles/metabolism , Neovascularization, Pathologic/therapy , Theranostic Nanomedicine/methods , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/therapeutic use , Cell Line, Tumor , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neovascularization, Pathologic/diagnostic imaging , Protein Binding , SELEX Aptamer Technique , Silicon Dioxide/chemistry , Tissue Distribution
8.
Biomaterials ; 139: 1-11, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28578297

ABSTRACT

Attacking the supportive vasculature network of a tumor offers an important new avenue for cancer therapy. Herein, a near-infrared (NIR) laser-activated "nanobomb" was developed as a noninvasive and targeted physical therapeutic strategy to effectively disrupt tumor neovasculature in an accurate and expeditious manner. This "nanobomb" was rationally fabricated via the encapsulation of vinyl azide (VA) into c(RGDfE) peptide-functionalized, hollow copper sulfide (HCuS) nanoparticles. The resulting RGD@HCuS(VA) was selectively internalized into integrin αvß3-expressing tumor vasculature endothelial cells and dramatically increased the photoacoustic signals from the tumor neovasculature, achieving a maximum signal-to-noise ratio at 4 h post-injection. Upon NIR irradiation, the local temperature increase triggered VA to release N2 bubbles rapidly. Subsequently, these N2 bubbles could instantly explode to destroy the neovasculature and further induce necrosis of the surrounding tumor cells. A single-dose injection of RGD@HCuS(VA) led to complete tumor regression after laser irradiation, with no tumor regrowth for 30 days. More importantly, high-resolution photoacoustic angiography, combined with excellent biodegradability, facilitated the precise destruction of tumor neovasculature by RGD@HCuS(VA) without damaging normal tissues. These results demonstrate the great potential of this "nanobomb" for clinical translation to treat cancer patients with NIR laser-accessible orthotopic tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Laser Therapy/methods , Nanocapsules/therapeutic use , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Animals , Antineoplastic Agents/chemical synthesis , Azides/chemistry , Cell Line, Tumor , Copper/chemistry , Female , Hot Temperature , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/chemistry , Mice , NIH 3T3 Cells , Nanocapsules/chemistry , Nitrogen/chemistry , Peptides, Cyclic/chemistry , Photoacoustic Techniques , Signal-To-Noise Ratio , Sulfides/chemistry , Xenograft Model Antitumor Assays
9.
Biomaterials ; 137: 23-36, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28528300

ABSTRACT

Metastasis of cancer makes up the vast majority of cancer-related deaths, and it usually initiates from tumor cells invasiveness and develops through tumor neovasculature. In this work, we have fabricated a CD44/neuropilin dual receptor-targeting nanoparticulate system (tLyP-1-HT NPs) with endogenous or FDA approved components for treating metastatic triple negative breast cancer (TNBC). The enhanced specific targeting of tLyP-1-HT NPs to both metastatic tumor cells and metastasis-supporting tumor neovasculature was contributed by means of CD44/neuropilin dual receptor-mediated interaction. The NPs not only effectively suppress the invasive capability of tumor cells themselves, but also significantly restrain the metastasis incidence via extravasation as well as the eventual colonization in lungs. In all the three types of TNBC-bearing mice models, orthotopic, post-metastasis and metastasis prevention models, the docetaxel-loaded tLyP-1-HT NPs exhibited markedly enhanced anti-tumor and anti-metastasis efficacy. The inhibitory rates of tLyP-1-HT NPs against orthotopic tumor growth and lung metastasis achieved 79.6% and 100%, respectively. The metastasis inhibition rate and life extension rate of the tLyP-1-HT NPs against post-pulmonary metastasis mice reached 85.1% and up to 62.5%, respectively. All the results demonstrated the designed dual receptor-targeting multifunctional NPs hold great potential in treating metastatic TNBC and lung metastasis.


Subject(s)
Hyaluronan Receptors/metabolism , Nanoparticles/chemistry , Neuropilins/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Docetaxel , Drug Carriers , Drug Liberation , Female , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Particle Size , Polymers/chemistry , Surface Properties , Taxoids/therapeutic use , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
10.
Methods Mol Biol ; 1430: 375-86, 2016.
Article in English | MEDLINE | ID: mdl-27172968

ABSTRACT

In vivo preclinical assays are required to screen potential agents that target the tumor vasculature. Here a hollow fiber-based assay for the quantification of neovasculature in the presence or absence of an agent that potentially targets tumor neovasculature is described. The neovasculature is developed as a consequence of the presence of tumor cells encapsulated in hollow fibers, which are transplanted sub-cutaneously in the dorsal flanks of mice.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Humans , MCF-7 Cells , Mice , Neoplasm Transplantation , Neoplasms/blood supply , Xenograft Model Antitumor Assays
11.
Int J Cancer ; 137(10): 2478-91, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-25951495

ABSTRACT

Low-intensity ultrasound-microbubble (LIUS-MB) treatment is a promising antivascular therapy for tumors. We sought to determine whether LIUS-MB treatment with an appropriate ultrasound pressure could achieve substantial and persistent cessation of tumor perfusion without having significant effects on normal tissue. Further, we investigated the mechanisms underlying this treatment. Murine S-180 sarcomas, thigh muscles, and skin tissue from 60 tumor-bearing mice were subjected to sham therapy, an ultrasound application combined with microbubbles in four different ultrasound pressures (0.5, 1.5, 3.0, 5.0 MPa), or ultrasound at 5.0 MPa alone. Subsequently, contrast-enhanced ultrasonic imaging and histological studies were performed. Tumor microvessels, tumor cell necrosis, apoptosis, tumor growth, and survival were evaluated in 85 mice after treatment with the selected ultrasound pressure. We found that twenty-four hours after LIUS-MB treatment at 3.0 MPa, blood perfusion and microvessel density of the tumor had substantially decreased by 84 ± 8% and 84%, respectively (p < 0.01). Similar reductions were not observed in the muscle or skin. Additionally, an extreme reduction in the number of immature vessels was observed in the tumor (reduced by 90%, p < 0.01), while the decrease in mature vessels was not significant. Further, LIUS-MB treatment at 3.0 MPa promoted tumor cell necrosis and apoptosis, delayed tumor growth, and increased the survival rate of tumor-bearing mice (p < 0.01). These findings indicate that LIUS-MB treatment with an appropriate ultrasound pressure could selectively and persistently reduce tumor perfusion by depleting the neovasculature. Therefore, LIUS-MB treatment offers great promise for clinical applications in antivascular therapy for solid tumors.


Subject(s)
Microbubbles/therapeutic use , Neovascularization, Pathologic/therapy , Sarcoma 180/therapy , Skin/pathology , Thigh/pathology , Ultrasonic Therapy/methods , Animals , Cell Line, Tumor , Male , Mice , Neovascularization, Pathologic/pathology , Sarcoma 180/blood supply , Sarcoma 180/pathology , Treatment Outcome
12.
Int J Pharm ; 493(1-2): 380-9, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26004003

ABSTRACT

EphA2 is a transmembrane receptor tyrosine kinase that is highly expressed on both tumor neovasculature and some kinds of tumor cells. Here, a homing peptide with a sequence of YSAYPDSVPMMSK (YSA) that binds specifically with EphA2 was utilized to modify the stealth liposomes (YSA-LP). With a particle size of about 85 nm, this functionalized nanocarrier was loaded with fluorescent probe or doxorubicin (DOX) and investigated in vitro and in vivo. In the cellular endocytosis studies in vitro, coumarin-6 loaded YSA-LP exhibited significant specificity to both EphA2-overexpressing tumor cells (MDA-MB-231) and human umbilical vein endothelial cells (HUVEC) via a YSA mediated interaction. In a MDA-MB-231 xenograft tumor mouse model, DiR-loaded YSA-LP showed more lasting accumulation in tumor tissue by small animal imaging compared to unmodified liposomes (LP). Further, YSA-LP greatly facilitated the efficacy of DOX loaded against both tumor cells and tumor angiogenesis in the same mouse model, evidenced by inhibiting tumor growth, metastasis and CD31 expression as well as inducing cancer cell apoptosis. Additionally, YSA-LP (DOX) showed relatively low systemic and cardiac toxicity compared with control groups. In conclusion, YSA might be a promising targeting motif for EphA2-overexpressing tumor cells and tumor neovasculature, which could be used to mediate drug delivery for chemotherapy agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Peptides/administration & dosage , Receptor, EphA2/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Female , Human Umbilical Vein Endothelial Cells , Humans , Liposomes , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Peptides/chemistry , Peptides/therapeutic use , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Tumor Burden/drug effects
13.
Biomaterials ; 42: 161-71, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25542804

ABSTRACT

Tumor angiogenesis is a multistep process involved with multiple molecular events in cancer microenvironment. Several molecular-targeted agents aiming to suppress tumor angiogenesis have been successfully translated into cancer clinic. However, new strategies are still urgently desired to be excavated to overcome the poor response and resistance in some antiangiogenic therapies. Recently, Delta-like ligand 4 (Dll4) is identified to be specifically over-expressed on tumor vascular endothelial cells (EC), and the Dll4-Notch pathway serves as a critical regulator in the development and maintenance of tumor angiogenesis. The intensively up-regulated phenotype of Dll4 on the membrane of tumor vascular EC implies that Dll4 may act as a targetable address for drug delivery system (DDS) to achieve targeted antiangiogenic cancer therapy. Here, a nano-DDS, GD16 peptide (H2N-GRCTNFHNFIYICFPD-CONH2, containing a disulfide bond between Cys3 and Cys13) conjugated nanoparticles loading paclitaxel (GD16-PTX-NP), which can specifically target the angiogenic marker Dll4, was fabricated for the investigation of antiangiogenic therapeutic efficacy in human head and neck cancer FaDu (Dll4-negative) xenograft in nude mice. The results demonstrate that GD16-PTX-NP achieved controlled drug release and exhibited favorable in vivo long-circulating feature. GD16-PTX-NP exerted enhanced antiangiogenic activity in the inhibition of human umbilical vein endothelial cell (HUVEC) viability, motility, migration, and tube formation, and in the Matrigel plug model as well, which can be definitely ascribed to the active internalization mediated by the interaction of GD16 and the over-expressed Dll4 on EC. GD16-PTX-NP showed accurate in vivo tumor neovasculature targeting property in FaDu tumor, where the paclitaxel was specifically delivered into the tumor vascular EC, leading to significant apoptosis of tumor vascular EC and necrosis of tumor tissues. The antiangiogenic activity of GD16-PTX-NP significantly contributed to its in vivo anticancer efficacy in Fadu tumor; moreover, no overt toxicity to the mice was observed. Our research firstly presents the potency and significance of a Dll4-targeted nanomedicine in antiangiogenic cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Drug Delivery Systems , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nanomedicine/methods , Neovascularization, Pathologic/drug therapy , Amino Acid Sequence , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Survival/drug effects , Collagen , Coumarins/metabolism , Drug Combinations , Endocytosis/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Laminin , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Peptides/chemistry , Proteoglycans , Rats, Sprague-Dawley , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/pathology , Thiazoles/metabolism , Xenograft Model Antitumor Assays
14.
J Mol Biol ; 426(21): 3606-18, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25073100

ABSTRACT

Fibrin formation from fibrinogen is a rare process in the healthy organism but is a pathological feature of thrombotic events, cancer and a wide range of inflammatory conditions. We have designed and constructed an antibody phage display library (containing 13 billion clones) for the selective recognition of the N-terminal peptide of fibrin alpha chain. The key structural feature for selective fibrin binding was a K94E mutation in the VH domain. From this library, an antibody was isolated (termed AP2), which recognizes the five N-terminal amino acids of fibrin with high affinity (Kd=44nM), but does not bind to fibrinogen. The AP2 antibody could be expressed in various formats (scFv, small immune protein and IgG) and inhibited fibrin clot formation in a concentration-dependent manner. Moreover, the AP2 antibody stained the fibrin-rich provisional stroma in solid tumors but did not exhibit any detectable staining toward normal tissues. Using a radioiodinated antibody preparation and quantitative biodistribution studies in tumor-bearing mice, AP2 was shown to selectively localize to fibrin-rich F9 murine teratocarcinomas, but not to SKRC-52 human kidney cancer xenografts. Collectively, the experiments indicate that the AP2 antibody recognizes fibrin in vitro and in vivo. The antibody may facilitate the development of fibrin-specific therapeutic agents.


Subject(s)
Antibodies, Monoclonal/chemistry , Blood Coagulation , Fibrin/chemistry , Neoplasms/immunology , Peptide Library , Amino Acid Sequence , Animals , Cell Line, Tumor , Fibrinogen/chemistry , Humans , Immunoglobulin G/chemistry , Mice , Molecular Sequence Data , Mutation , Neoplasm Transplantation , Neoplasms/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Thrombosis
15.
Biomaterials ; 35(4): 1215-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24231414

ABSTRACT

Antiangiogenic therapy is a validated approach for colorectal cancer (CRC) treatment. However, diverse adverse effects inevitably appear due to the off-target effect of the approved antiangiogenic inhibitors on the physiological functions and homeostasis. This study was to investigate a new tumor vessel targeting nanoparticulate drug delivery system, F56 peptide conjugated nanoparticles loading vincristine (F56-VCR-NP), for the effective treatment of CRC subcutaneous xenograft and experimental lung metastasis model. The controlled release behavior and in vivo pharmacokinetic profile of F56-VCR-NP were characterized. The tumor vessel targeting and antiangiogenic activity of F56-VCR-NP was evaluated in human umbilical vein endothelial cells (HUVEC, a classical cell model mimicking tumor vascular EC), subcutaneous human HCT-15 xenograft in immunodeficient nude mice, and experimental CT-26 lung metastasis model in immunocompetent mice. The therapeutic efficacy (animal survival and toxicity) was further investigated in the model of CT-26 lung metastasis in mice. F56-VCR-NP could achieve 30-day controlled drug release in PBS (pH 7.4) and exhibited favorable long-circulating feature in vivo. F56-VCR-NP could accurately target the CRC neovasculature and elicit nanoparticle internalization in the tumor vascular EC, where the antiangiogenic VCR-induced dramatic EC apoptosis and necrosis of CRC tissue. F56-VCR-NP significantly prolonged the mouse survival with no obvious toxicity (weight loss and anepithymia) in the CT-26 lung metastasis mice model, and this pronounced antitumor effect was closely related with the decreased microvessel density in the metastases. The present nanoparticle-based targeted antiangiogenic therapy may provide a new promising approach for the therapy of CRC and lung metastasis, which deserves further translational research.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Vincristine/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Movement/drug effects , Colorectal Neoplasms/pathology , Drug Delivery Systems , Human Umbilical Vein Endothelial Cells , Humans , Lung/blood supply , Lung/drug effects , Lung/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Oligopeptides/chemistry , Rats, Sprague-Dawley , Vincristine/administration & dosage , Vincristine/pharmacokinetics
16.
Int J Cancer ; 133(10): 2483-92, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23661285

ABSTRACT

Converting T cells into tumor cell killers by grafting them with a chimeric antigen receptor (CAR) has shown promise as a cancer immunotherapeutic. However, the inability of these cells to actively migrate and extravasate into tumor parenchyma has limited their effectiveness in vivo. Here we report the construction of a CAR containing an echistatin as its targeting moiety (eCAR). As echistatin has high binding affinity to αvß3 integrin that is highly expressed on the surface of endothelial cells of tumor neovasculature, T cells engrafted with eCAR (T-eCAR) can efficiently lyse human umbilical vein endothelial cells and tumor cells that express αvß3 integrin when tested in vitro. Systemic administration of T-eCAR led to extensive bleeding in tumor tissues with no evidence of damage to blood vessels in normal tissues. Destruction of tumor blood vessels by T-eCAR significantly inhibited the growth of established bulky tumors. Moreover, when T-eCAR was codelivered with nanoparticles in a strategically designed temporal order, it dramatically increased nanoparticle deposition in tumor tissues, pointing to the possibility that it may be used together with nanocarriers to increase their capability to selectively deliver antineoplastic drugs to tumor tissues.


Subject(s)
Melanoma, Experimental/blood supply , Melanoma, Experimental/therapy , Nanoparticles/administration & dosage , T-Lymphocytes/physiology , Amino Acid Sequence , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Blood Vessels/pathology , Cell Line , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/pathology , Humans , Integrin alphaVbeta3/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutant Chimeric Proteins/biosynthesis , Mutant Chimeric Proteins/genetics , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Antigen/metabolism , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL