Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (204)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38407494

ABSTRACT

The glomeruli are fundamental units in the kidney; hence, studying the glomeruli is pivotal for understanding renal function and pathology. Biological imaging provides intuitive information; thus, it is of great significance to label and observe the glomeruli. However, the glomeruli observation methods currently in use require complicated operations, and the results may lose label details or three-dimensional (3D) information. The clear, unobstructed brain imaging cocktails and computational analysis (CUBIC) tissue clearing technology has been widely used in renal research, allowing for more accurate detection and deeper detection depth. We found that mouse glomeruli can be rapidly and effectively labeled by tail vein injection of medium molecular weight FITC-Dextran followed by the CUBIC clearing method. The cleared mouse kidney could be scanned by a light-sheet microscope (or a confocal microscope when sliced) to obtain three-dimensional image stacks of all the glomeruli in the entire kidney. Processed with appropriate software, the glomeruli signals could be easily digitized and further analyzed to measure the number, volume, and frequency of the glomeruli.


Subject(s)
Kidney Glomerulus , Kidney , Animals , Mice , Kidney Glomerulus/diagnostic imaging , Brain/diagnostic imaging , Molecular Weight , Software
2.
Eur J Nucl Med Mol Imaging ; 51(6): 1530-1543, 2024 May.
Article in English | MEDLINE | ID: mdl-38189910

ABSTRACT

PURPOSE: Noninvasive quantifying activated hepatic stellate cells (aHSCs) by molecular imaging is helpful for assessing disease progression and therapeutic responses of liver fibrosis. Our purpose is to develop platelet-derived growth factor receptor ß (PDGFRß)-targeted radioactive tracer for assessing liver fibrosis by positron emission tomography (PET) imaging of aHSCs. METHODS: Comparative transcriptomics, immunofluorescence staining and flow cytometry were used to evaluate PDGFRß as biomarker for human aHSCs and determine the correlation of PDGFRß with the severity of liver fibrosis. The high affinity affibody for PDGFRß (ZPDGFRß) was labeled with gallium-68 (68Ga) for PET imaging of mice with carbon tetrachloride (CCl4)-induced liver fibrosis. Binding of the [68Ga]Ga-labeled ZPDGFRß ([68Ga]Ga-DOTA-ZPDGFRß) for aHSCs in human liver tissues was measured by autoradiography. RESULTS: PDGFRß overexpressed in aHSCs was highly correlated with the severity of liver fibrosis in patients and CCl4-treated mice. The 68Ga-labeled ZPDGFRß affibody ([68Ga]Ga-DOTA-ZPDGFRß) showed PDGFRß-dependent binding to aHSCs. According to the PET imaging, hepatic uptake of [68Ga]Ga-DOTA-ZPDGFRß increased with the accumulation of aHSCs and collagens in the fibrotic livers of mice. In contrast, hepatic uptake of [68Ga]Ga-DOTA-ZPDGFRß decreased with spontaneous recovery or treatment of liver fibrosis, indicating that the progression and therapeutic responses of liver fibrosis in mice could be visualized by PDGFRß-targeted PET imaging. [68Ga]Ga-DOTA-ZPDGFRß also bound human aHSCs and visualized fibrosis in patient-derived liver tissues. CONCLUSIONS: PDGFRß is a reliable biomarker for both human and mouse aHSCs. PDGFRß-targeted PET imaging could be used for noninvasive monitoring of liver fibrosis in mice and has great potential for clinical translation.


Subject(s)
Gallium Radioisotopes , Liver Cirrhosis , Positron-Emission Tomography , Receptor, Platelet-Derived Growth Factor beta , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/metabolism , Animals , Positron-Emission Tomography/methods , Humans , Receptor, Platelet-Derived Growth Factor beta/metabolism , Mice , Male , Hepatic Stellate Cells/metabolism , Heterocyclic Compounds, 1-Ring/chemistry
3.
Eur J Nucl Med Mol Imaging ; 50(10): 2952-2961, 2023 08.
Article in English | MEDLINE | ID: mdl-37256321

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is a highly vascularized solid carcinoma and tumor vessel-targeted molecular imaging might be effective for early diagnosis of HCC. Herein, we developed a novel trimeric affibody (ZTRI) with highly specific binding to the platelet-derived growth factor receptor beta (PDGFRß). The aim of this study is to evaluate the feasibility of 68Ga-radiolabeled ZTRI ([68Ga]Ga-DOTA-ZTRI) as PET tracer for diagnosis of HCC. METHODS: The bioinformatics analysis of clinical database and immunoblotting of clinical specimens were performed to validate the potential of PDGFRß as HCC biomarker. The trimeric affibody ZTRI was conjugated with DOTA-NHS-ester and radiolabeled with 68Ga to produce [68Ga]Ga-DOTA-ZTRI conjugate. Immunoreactivity and specific uptake of [68Ga]Ga-DOTA-ZTRI were assessed by dose-dependent cell binding, autoradiography, and biodistribution analysis. [68Ga]Ga-DOTA-ZTRI PET/CT scanning of diethylnitrosamine (DEN)-induced primary HCC rats and a rare case of idiopathical HCC rhesus monkey was performed to evaluate the imaging capability and radiation dosimetry of [68Ga]Ga-DOTA-ZTRI in vivo. RESULTS: Excessive PDGFRß was validated as a representative biomarker of HCC neovascularization. The radiolabeling of [68Ga]Ga-DOTA-ZTRI was achieved at more than 95% radiochemical yield. In vitro assays showed specific uptake of [68Ga]Ga-DOTA-ZTRI in HCC tumor vessels by autoradiography. Animal PET/CT imaging with [68Ga]Ga-DOTA-ZTRI successfully visualized the tumor lesions in primary HCC rats and rhesus monkey, and indicated radiation absorbed dose of 2.03E-02 mSv/MBq for each scanning. CONCLUSIONS: Our results demonstrated that [68Ga]Ga-DOTA-ZTRI conjugate could be applied as a promising PET tracer for early diagnosis of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Rats , Animals , Positron Emission Tomography Computed Tomography/methods , Carcinoma, Hepatocellular/diagnostic imaging , Gallium Radioisotopes/chemistry , Tissue Distribution , Macaca mulatta , Cell Line, Tumor , Liver Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Biomarkers
4.
Biomaterials ; 295: 121994, 2023 04.
Article in English | MEDLINE | ID: mdl-36775789

ABSTRACT

Prompting higher-order death receptor (DR) clustering by increasing the valency of DR agonist is efficient to induce apoptosis of tumor cells. As an attractive DR agonist with superior biosafety, the trimeric tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exerts limited antitumor effect in patients, which is predominantly attributed to its low DR clustering ability and short serum half-life. Previous antibody scaffolds-based engineering strategies to increase the valency and/or prolong the serum half-life of TRAIL improve apoptosis induction, however, often produce large proteins with poor tumor penetration. Covalent protein ligation mediated by small molecular superglues such as SpyTag/SpyCatcher might be a novel strategy to assemble higher-order TRAIL variants. Upon fusion to TRAIL promotor, SpyTag/SpyCatcher molecular superglue preferentially ligated two trimeric TRAIL to produce a hexameric TRAIL variant, HexaTR, exhibiting a significantly increased apoptosis induction. In addition, an albumin-binding HexaTR, ABD-HexaTR, with a prolonged serum half-life by binding to endogenous albumin was also produced using the same strategy. Compared to the trimeric TRAIL, the hexameric HexaTR and ABD-HexaTR showed 20-50 times greater in vivo antitumor effect, resulting in eradication of several types of large (150-300 mm3) tumor xenografts. Combination with bortezomib carried by liposome further improved the antitumor effects of the hexavalent HexaTR and ABD-HexaTR in refractory cancer. Our results indicate that the superglue-mediated higher-order assembly is promising to improve the DR clustering and proapoptotic signaling of TRAIL, showing great advantages in constructing the next generation of DR agonists for cancer therapy.


Subject(s)
Apoptosis , TNF-Related Apoptosis-Inducing Ligand , Humans , Cell Line, Tumor , Ligands , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Albumins/pharmacology
5.
Drug Deliv ; 29(1): 1698-1711, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35635308

ABSTRACT

Multidrug resistance (MDR), which is common in colorectal cancer (CRC), induces high mortality in patients. Due to its robust and selective apoptosis induction in some CRC cells with MDR, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is attractive as a novel tool for CRC therapy. However, TRAIL is limited by its poor tumor-homing ability and inefficient apoptosis induction in CRC cells expressing low levels of death receptor (DR). Here, the tumor-homing RGR peptide (CRGRRST) was fused to TRAIL to produce RGR-TRAIL. Compared with TRAIL, RGR-TRAIL showed greater cell binding and cytotoxicity in CRC cells. In addition, RGR-TRAIL exerted significantly enhanced tumor uptake and growth suppression in mice bearing CRC tumor xenografts. Notably, RGR-TRAIL eradicated all tumor xenografts of DR-overexpressing COLO205 cells. However, TRAIL only showed mild tumor growth suppression under the same conditions, indicating that RGR fusion significantly increased the antitumor effect of TRAIL in DR-overexpressing CRC cells by improving tumor homing. Nevertheless, RGR fusion did not significantly enhance the antitumor effect of TRAIL in HT29 cells expressing low levels of DR. We found that DR expression in HT29 cells was enhanced by epidermal growth factor receptor (EGFR)-targeted photodynamic therapy (PDT). Moreover, both the in vitro and in vivo antitumor effects of RGR-TRAIL were significantly improved by combination with PDT. HT29 tumor xenografts (∼20%) were even eradicated by combination therapy. These results indicate that it is valuable to further evaluate the combination therapy of RGR-TRAIL and tumor-targeted PDT for clinical therapy of CRC with MDR.


Subject(s)
Colorectal Neoplasms , Photochemotherapy , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Heterografts , Humans , Mice , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand/pharmacology
6.
J Control Release ; 340: 243-258, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34752799

ABSTRACT

Immunotherapies based on immune checkpoint-blocking antibodies have been considered the most attractive cancer treatments in recent years. However, the systemic administration of immune checkpoint-blocking antibodies is limited by low response rates and high risk of inducing immune-related adverse events (irAEs), which might be overcome by the tumor-targeted delivery of these antibodies. To achieve tumor-targeted delivery, immune checkpoint-blocking antibodies are usually modified with tumor-homing ligands through difficult genetic fusion or chemical conjugation. As most immune checkpoint-blocking antibodies are immunoglobin G (IgG) antibodies, we hypothesize that these IgG antibodies might be noncovalently modified with a tumor-homing ligand fused to an IgG-binding domain (IgBD). To test this hypothesis, the tumor-homing ZPDGFRß affibody, which targets platelet-derived growth factor receptor ß (PDGFRß), was fused to the Fab-selective IgBD in a trimeric format. After mixing ZPDGFRß fused to the IgBD with immune checkpoint-blocking IgG against programmed death-ligand 1 (αPD-L1), a novel homogenous complex was formed, indicating that αPD-L1 had been successfully modified with ZPDGFRß fused to the IgBD. ZPDGFRß-modified αPD-L1 bound to both PDGFRß and PD-L1, thus leading to greater tumor uptake and antitumor effects in mice bearing PDGFRß+PD-L1+ tumor grafts. In addition, due to the broad spectrum of IgBD for IgG, immune checkpoint-blocking IgG antibodies against cytotoxic T-lymphocyte-associated protein 4 (αCTLA-4) and signal regulatory protein alpha (αSIRPα) were also modified with ZPDGFRß fused to the IgBD. These results demonstrated that a tumor-homing ligand fused to the IgBD might be developed as a versatile platform for the modification of immune checkpoint-blocking IgG antibodies to achieve tumor-targeted delivery.


Subject(s)
Drug Delivery Systems , Immune Checkpoint Inhibitors , Immunoglobulin G/immunology , Neoplasms , Animals , Immunotherapy , Mice , Neoplasms/drug therapy , Receptor, Platelet-Derived Growth Factor beta
7.
Theranostics ; 11(9): 4281-4297, 2021.
Article in English | MEDLINE | ID: mdl-33754061

ABSTRACT

Chemotherapeutic multidrug resistance (MDR) is the major hindrance for clinical therapy of colorectal cancer (CRC). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) with selective cytotoxicity might overcome MDR of CRC cells. Unfortunately, cross-resistance to TRAIL has been detected in many CRC cells, suggesting the need to combine TRAIL with sensitizers to combat refractory CRC. Our purpose is to explore the potential of combination therapy of TRAIL and tumor-cell targeted photodynamic therapy (PDT) in combating CRC with both chemotherapeutic MDR and TRAIL resistance. Methods: Tumor cell-targeted PDT was performed using a Ze-IR700 photosensitizer with high affinity for epidermal growth factor receptor (EGFR). The impact of PDT on the gene expression of CRC cells was revealed by RNA sequencing. The synergistic antitumor effect of long-acting TRAIL and PDT was evaluated in mice bearing tumor grafts of CRC cells with both chemotherapeutic MDR and TRAIL resistance. Results: Chemotherapeutic MDR and TRAIL resistance are common in CRC cells. Pretreatment of CRC cells with tumor cell-targeted PDT significantly (10-60 times) increased the sensitivity of these CRC cells to TRAIL by upregulating death receptors. Combination therapy, but not monotherapy, of long-acting TRAIL and PDT greatly induced apoptosis of CRC cells, thus efficiently eradicated large (~150 mm3) CRC tumor xenografts in mice. Conclusions: Tumor cell-targeted PDT extensively sensitizes CRC cells to TRAIL. Combination therapy of long-acting TRAIL and PDT is promising to combat CRC with both chemotherapeutic MDR and TRAIL resistance, which might be developed as a novel strategy for precision therapy of refractory CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Female , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Photochemotherapy/methods , Photosensitizing Agents/pharmacology
8.
Appl Microbiol Biotechnol ; 105(4): 1447-1460, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33528691

ABSTRACT

Due to their potent immune stimulation, tumor necrosis factor alpha (TNFα) variants with tumor-homing activity are attractive as novel antitumor drugs. The promising antitumor effect of NGR-TNFα in clinical trials triggered extensive interest in developing novel tumor-homing TNFα variants in recent years. Owing to its promising antitumor effect, NGR-TNFα is usually used as a control for newly developed tumor-homing TNFα variants. In our previous works, we produced a pericyte-targeting Z-TNFα at high levels using the Escherichia coli (E. coli) M15-pQE30 system. To further compare Z-TNFα and NGR-TNFα, we attempted to express NGR-TNFα using the same system. Surprisingly, native NGR-TNFα was expressed at a low (~ 0.2 mg/L) level in E. coli M15 containing the pQE30 plasmid. However, a single nucleotide mutation of C to G, resulting in a substitution of leucine (L) with valine (V) at the start of TNFα, increased the expression of NGR-TNFα by ~ 100 times through improving transcription. In addition, the amino acid substitution showed a little impact on the receptor binding, in vitro cytotoxicity, and in vivo antitumor effect of NGR-TNFα. As fusing NGR to the N-terminus of TNFα with a valine substitution did not reduce the protein yield, the TNFα gene with a C > G mutation might be used to prepare novel tumor-homing TNFα when the native TNFα-based variant is expressed at an extremely low level in E. coli. Notably, in addition to the mutated valine, the impact of N-terminal additional amino acids provided by pQE30 vector on the function of TNFα variant must be carefully evaluated. KEY POINTS : • A single nucleotide mutation increased the expression of NGR-TNFα by two orders. • Nucleotide mutation-induced amino acid substitution did not reduce NGR-TNFα activity.


Subject(s)
Escherichia coli , Tumor Necrosis Factor-alpha , Cell Line, Tumor , Escherichia coli/genetics , Galanin/analogs & derivatives , Mutation , Nucleotides , Oligopeptides/genetics , Substance P/analogs & derivatives , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
9.
Biomacromolecules ; 21(10): 4017-4029, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32804484

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive antitumor drug candidate for precision cancer therapy due to its superior selective cytotoxicity in a variety of tumor cells. However, the clinical application of TRAIL in cancer therapy has been limited by its poor tumor-homing capacities and short half-life. Herein, we designed a tridomain TRAIL variant, Z-ABD-TRAIL, by sequentially fusing the platelet-derived growth factor receptor beta (PDGFRß)-specific affibody ZPDGFRß and an albumin-binding domain (ABD) to the N-terminus of TRAIL. The fusion protein Z-ABD-TRAIL was produced as a soluble protein with high yield in Escherichia coli (E. coli). The ZPDGFRß domain provided Z-ABD-TRAIL with PDGFRß-binding properties and thus promoted its tumor homing via the engagement of PDGFRß-expressing pericytes on tumor microvessels. ABD-mediated binding of Z-ABD-TRAIL to albumin in the blood endowed TRAIL with long-lasting (>72 h for Z-ABD-TRAIL vs <0.5 h for TRAIL) abilities to kill tumor cells. Although the in vitro cytotoxicity of Z-ABD-TRAIL in tumor cells was similar to that of the parent TRAIL, the in vivo tumor uptake, apoptosis-inducing ability, and antitumor effect of Z-ABD-TRAIL were much greater than those of TRAIL, indicating that ZPDGFRß-mediated tumor homing and ABD-introduced albumin binding significantly improved the pharmacodynamics of TRAIL. In addition, repeated injection of high-dose Z-ABD-TRAIL showed no obvious acute toxicity in mice. These results demonstrate that the newly designed tridomain Z-ABD-TRAIL is a promising agent for precision cancer therapy.


Subject(s)
Escherichia coli , TNF-Related Apoptosis-Inducing Ligand , Albumins , Animals , Apoptosis , Cell Line, Tumor , Delayed-Action Preparations , Mice , TNF-Related Apoptosis-Inducing Ligand/genetics
10.
Mol Pharm ; 17(7): 2508-2517, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32396000

ABSTRACT

Antibody-based near-infrared photoimmunotherapy (NIR-PIT) is an attractive strategy for cancer treatment. Tumor cells can be selectively and efficiently killed by the targeted delivery of an antibody-photoabsorber complex followed by exposure to NIR light. Glycoprotein A33 antigen (GPA33) is highly expressed in most human colorectal cancers (CRCs) and is an ideal diagnostic and therapeutic target. We previously produced a single-chain fragment of a variable antibody against GPA33 (A33scFv antibody). Here, we investigate the efficacy of NIR-PIT by combining A33scFv with the NIR photoabsorber IR700 (A33scFv-IR700). In vitro, recombinant A33scFv displayed specific binding and delivery of an NIR dye to GPA33-positive tumor cells. Furthermore, A33scFv-IR700-mediated NIR-PIT was successful in rapidly and specifically killing GPA33-positive colorectal tumor cells. NIR-PIT treatment induced the release of lactate dehydrogenase from tumor cells, followed by cell necrosis, rather than apoptosis, through the promotion of reactive oxygen species accumulation in tumor cells. In mice bearing LS174T tumor grafts, A33scFv selectively accumulated in GPA33-positive tumors. Following only a single injection of the conjugate and subsequent illumination, A33scFv-IR700-mediated NIR-PIT induced a significant increase in therapeutic response in LS174T-tumor mice compared with that in the non-NIR-PIT groups (p < 0.001). Because the GPA33 antigen is specifically expressed in CRC tumors, A33scFv-IR700 might be a promising antibody fragment-photoabsorber conjugate for NIR-PIT of CRC.


Subject(s)
Cell Death/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Immunoconjugates/therapeutic use , Immunotherapy/methods , Membrane Glycoproteins/immunology , Phototherapy/methods , Single-Chain Antibodies/immunology , Animals , Cell Death/immunology , Colorectal Neoplasms/immunology , HT29 Cells , Humans , L-Lactate Dehydrogenase/metabolism , Mass Spectrometry , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Necrosis/metabolism , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Single-Chain Antibodies/radiation effects , Single-Chain Antibodies/toxicity , Xenograft Model Antitumor Assays
11.
Apoptosis ; 25(1-2): 105-119, 2020 02.
Article in English | MEDLINE | ID: mdl-31894448

ABSTRACT

Liver fibrosis usually progresses to liver cirrhosis and even hepatocellular carcinoma. Since activated hepatic stellate cells (aHSCs) are responsible for liver fibrosis, reducing the quantity of aHSCs was considered the essential strategy for clinical antihepatofibrotic therapy. Due to the overexpression of TRAIL receptor 2 (DR5) in aHSCs, human TNF-related apoptosis-inducing ligand (hTRAIL) that could induce aHSCs apoptosis might be feasible for antihepatofibrotic therapy. However, the in vivo aHSCs-apoptosis-induction of hTRAIL is limited by its poor cell-targeting and a short half-life. In this study, we found that platelet-derived growth factor receptor ß (PDGFRß) was co-expressed with DR5 in aHSCs. And the ZPDGFRß affibody with high affinity for PDGFRß could bind aHSCs and, thus, accumulate in the fibrotic liver. ZPDGFRß was fused to hTRAIL to produce the fusion protein Z-hTRAIL. Compared to hTRAIL, Z-hTRAIL showed greater in vitro cell binding and apoptosis-induction in aHSCs. In addition, Z-hTRAIL induced apoptosis of aHSCs but spared other normal liver cells. In vivo, Z-hTRAIL accumulated preferentially in fibrotic livers and exerted greater effects than hTRAIL in inducing aHSCs apoptosis and reducing extracellular matrix (ECM) deposition. These results demonstrated that the antihepatofibrotic effect of hTRAIL was improved by PDGFRß-targeted delivery. To enhance its pharmacokinetics, Z-hTRAIL was modified with 10 kDa polyethylene glycol (PEG), which significantly (30-40 times) prolonged its half-life. The PEGylated long-acting Z-hTRAIL was more potent than the native Z-hTRAIL in regressing liver fibrosis. These results suggest that the aHSC-targeting and long-acting Z-hTRAIL might serve as a novel tool for antihepatofibrotic therapy.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Cell Line, Tumor , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Hepatic Stellate Cells/cytology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/physiopathology , Mice , Mice, Inbred C57BL , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics
12.
Int J Nanomedicine ; 14: 8543-8560, 2019.
Article in English | MEDLINE | ID: mdl-31802868

ABSTRACT

BACKGROUND: Nanoparticles exhibit great promise for improving the solubility and tissue-specific distribution of chemotherapeutic agents; however, the passive and highly variable enhanced permeability and retention (EPR) effects observed in tumors frequently leads to insufficient delivery of nanodrugs into tumors. The tumor-penetrating peptide iRGD can actively enhance tumor-selective delivery of nanoparticles into tumors by binding to integrin and interacting with tissue-penetrating receptor neuropilin-1. MATERIALS AND METHODS: To improve colorectal cancer treatment, in this study, we prepared a paclitaxel (PTX)-loaded PLGA nanoparticle (PLGA-PTX) and evaluated its tumor-targeting and antitumor activity by co-administration with iRGD. RESULTS: Compared to free PTX, encapsulated PTX retained preferential cytotoxicity toward various colorectal cancer cells while effectively sparing healthy cells. PLGA-PTX treatment resulted in cell cycle arrest at the G2/M phase and apoptosis, leading to inhibition of cancer cell migration and invasion. PLGA-PTX combined with iRGD displayed little enhancement of cytotoxicity in vitro. Despite this, iRGD receptors integrin and neuropilin-1 were found to be primarily overexpressed on abundant tumor vessels in mice bearing colorectal tumors. Consequently, co-administration of nanoparticles with iRGD promoted the selective delivery of nanoparticles into tumor tissues in vivo. Additionally, the combined regimen enhanced the antitumor effects compared to those of each individual reagent. CONCLUSION: Our findings suggest that PLGA nanoparticles combined with the iRGD peptide provide a promising drug delivery strategy for facilitating active drug accumulation into tumors, given that iRGD receptors are overexpressed on tumor vessels. This co-administration system lacking covalent conjugation provides a more convenient means to combine various therapeutic agents with iRGD to achieve personalized nanotherapy.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Delivery Systems , Nanoparticles/chemistry , Oligopeptides/administration & dosage , Paclitaxel/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/administration & dosage , Neoplasm Invasiveness , Oligopeptides/chemistry , Paclitaxel/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tissue Distribution
13.
Mol Pharm ; 16(5): 1950-1957, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30986347

ABSTRACT

Platelet-derived growth factor receptor ß (PDGFRß) is overexpressed in a variety of malignant cancers, plays a critical role in tumor angiogenesis, and has been proven as a valuable target for cancer treatment. In this pilot study, a dimeric affibody molecule, ZPDGFRß, was prepared and radiolabeled with positron emission radionuclide zirconium-89 for PET imaging of colorectal tumors by targeting PDGFRß expression in vivo. The PDGFRß-binding capability of dimeric affibody was evaluated by flow cytometry, immunofluorescent staining, and whole-body optical imaging. Then, ZPDGFRß was conjugated with DFO-Bn-NCS and radiolabeled with 89Zr. Targeted binding capability of 89Zr-DFO-ZPDGFRß to PDGFRß expressing cells was investigated by cellular assay in vitro and microPET/CT imaging in vivo. Dimeric ZPDGFRß affibody had specifically higher binding capability with PDGFRß expressing pericytes rather than LS-174T cancer cells, and well colocalized with tumor neovasculature by flow cytometry and immunofluorescent assay. ZPDGFRß was successfully labeled with 89Zr by DFO chelating with yield of 94.1 ± 3.53%. 89Zr-DFO-ZPDGFRß indicated preserved specific binding ability with PDGFRß expressing cells and effective inhibiting capability to PDGF-ß ligands ( P < 0.05) in vitro. Biodistribution indicated that tumor uptake of 89Zr-DFO-ZPDGFRß reached the peak of 6.93 ± 0.64%ID/g, and the tumor-to-blood ratio was 5.5 ± 0.6 at 2 h post-injection. LS-174T xenografts were clearly visualized by microPET/CT imaging through 1 to 4 h post-injection of 89Zr-DFO-ZPDGFRß affibody conjugate. In conclusion, the 89Zr-DFO-ZPDGFRß conjugate demonstrated specific and high binding ability with colorectal tumor, which indicated its use as a potential radiopharmaceutical for diagnostic imaging of tumor associate vasculatures with PET/CT.


Subject(s)
Antibodies, Monoclonal/chemistry , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Positron Emission Tomography Computed Tomography/methods , Radioisotopes/chemistry , Receptor, Platelet-Derived Growth Factor beta/metabolism , Xenograft Model Antitumor Assays/methods , Zirconium/chemistry , Animals , Antibodies, Monoclonal/metabolism , BALB 3T3 Cells , Cell Line, Tumor , Deferoxamine/analogs & derivatives , Deferoxamine/chemistry , Humans , Mice , Mice, Nude , Pericytes/metabolism , Pilot Projects , Radioisotopes/metabolism , Receptor, Platelet-Derived Growth Factor beta/immunology , Tissue Distribution , Zirconium/metabolism
14.
J Control Release ; 302: 63-78, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30930215

ABSTRACT

The delivery of anticancer drugs is hampered by tumor vessels with abnormal structure and function, which requires that vessel normalization be mediated by pharmaceutics. The current strategies for vessel normalization focus on direct modulation of endothelial cells (ECs), which frequently affect vessels in normal tissues. Modulating EC-supporting cells, such as pericytes (PCs), is a new direction. Here, we produced a fusion protein, Z-TNFα, by fusing the platelet-derived growth factor receptor ß (PDGFRß)- antagonistic affibody ZPDGFRß to tumor necrosis factor α (TNFα). Owing to the affinity of fused ZPDGFRß for PDGFRß, Z-TNFα binds PDGFRß+ PCs but not PDGFRß- ECs. Low-dose (1 µg/mouse) Z-TNFα treatment remodeled the tumor vessels, thus reducing vessel permeability and increasing vessel perfusion. As a result, the Z-TNFα treatment improved the delivery of doxorubicin (DOX) and enhanced its antitumor effect, indicating that Z-TNFα induced normalization of tumor vessels. Mechanically, the tumor vessel normalization mediated by Z-TNFα might be attributed to the reduction of vascular endothelial growth factor (VEGF) secretion by PCs and the elevated expression of intercellular cell adhesion molecule-1 (ICAM-1) in PCs, which might suppress the proliferation and migration of ECs and simultaneously trigger interaction between perivascular macrophages and PCs. These results demonstrated that tumor-associated PCs could be considered novel target cells for vessel normalization, and Z-TNFα might be developed as a potential tool for antitumor combination therapy.


Subject(s)
Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Pericytes/chemistry , Receptor, Platelet-Derived Growth Factor beta/chemistry , Recombinant Fusion Proteins/chemistry , Tumor Necrosis Factor-alpha/chemistry , Animals , Antineoplastic Agents/pharmacology , Biological Transport , Cell Line, Tumor , Combinatorial Chemistry Techniques/methods , Doxorubicin/pharmacology , Drug Liberation , Endothelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred ICR , Neoplasms, Experimental , Protein Kinase Inhibitors/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A
15.
Theranostics ; 8(9): 2459-2476, 2018.
Article in English | MEDLINE | ID: mdl-29721092

ABSTRACT

The inefficiency of recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based clinical regimens has been dominantly attributed to the short half-life of TRAIL. Affinity-controlled release using endogenous long-acting proteins, such as IgG and albumin, as carriers is extremely attractive for improving the pharmacokinetics of TRAIL. Up to now, it is unclear whether IgG-binding is efficient for affinity-controlled release of TRAIL. Methods: An IgG-binding affibody, IgBD, was genetically fused to the N-terminus of TRAIL to produce IgBD-TRAIL.The IgG-binding ability, cytotoxicity, serum half-life, and in vivo antitumor effect of IgBD-TRAIL were compared with that of TRAIL. In addition, an albumin-binding affibody, ABD, was fused to TRAIL to produce ABD-TRAIL. The cytototoxicity, serum half-life, and antitumor effect of IgBD-TRAIL and ABD-TRAIL were compared. Results: IgBD fusion endowed TRAIL with high affinity (nM) for IgG without interference with its cytotoxicity. The serum half-life of IgBD-TRAIL is 50-60 times longer than that of TRAIL and the tumor uptake of IgBD-TRAIL at 8-24 h post-injection was 4-7-fold that of TRAIL. In vivo antitumor effect of IgBD-TRAIL was at least 10 times greater than that of TRAIL. Owing to the high affinity (nM) for albumin, the serum half-life of ABD-TRAIL was 80-90 times greater than that of TRAIL. However, after binding to albumin, the cytotoxicity of ABD-TRAIL was reduced more than 10 times. In contrast, binding to IgG had little impact on the cytotoxicity of IgBD-TRAIL. Consequently, intravenously injected IgBD-TRAIL showed antitumor effects superior to those of ABD-TRAIL. Conclusions: Endogenous long-acting proteins, particularly IgG-based affinity-controlled release, prolonged the serum half-life as well as significantly enhanced the antitumor effect of TRAIL. IgBD-mediated endogenous IgG binding might be a novel approach for the affinity-controlled release of other protein drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Delayed-Action Preparations/pharmacology , Immunoglobulin G/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , Cell Line , Cell Line, Tumor , HCT116 Cells , Half-Life , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Recombinant Fusion Proteins/pharmacology , Serum Albumin/pharmacology
16.
Drug Deliv ; 24(1): 1818-1830, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29182023

ABSTRACT

Vascular-targeted photodynamic therapy (PDT) is an important strategy for cancer therapy. Conventional vascular-targeted PDT has been achieved by passive photosensitizer (PS) delivery, which involves a high risk of adverse effects. Active PS delivery is urgently required for vascular-targeted PDT. Although endothelial cells and pericytes are major cellular components of tumor blood vessels, little attention has been paid to pericyte-targeted PDT for cancer therapy. PDGFRß is abundantly expressed in the pericytes of various tumors. In this experiment, a dimeric ZPDGFRß affibody with a 0.9 nM affinity for PDGFRß was produced. The ZPDGFRß affibody showed PDGFRß-dependent pericyte binding. Intravenously injected ZPDGFRß affibody was predominantly distributed on pericytes and thus accumulated in LS174T tumor grafts. The conjugate of the ZPDGFRß affibody and IR700 dye, i.e. ZIR700, bound to PDGFRß+ pericytes but not to PDGFRß- LS174T tumor cells. Accordingly, ZIR700-mediated PDT in vitro induced the death of pericytes but not of LS174T tumor cells. In mice bearing LS174T tumor grafts, ZIR700-mediated PDT damaged tumor blood vessels, thus inducing tumor destruction by intensifying tissue hypoxia. The average mass of tumor grafts administered with ZIR700-mediated PDT was approximately 20-30% of that of the control, indicating that pericyte-targeted PDT is efficient for cancer therapy. In addition, ZIR700-mediated PDT increased the tumor uptake of TNF-related apoptosis-inducing ligand (TRAIL) injected post-illumination. Consequently, combination therapy of ZIR700-mediated PDT and TRAIL showed greater tumor suppression than ZIR700-mediated PDT- or TRAIL-based monotherapy. These results demonstrated that active vascular-targeted PDT could be achieved by using ZPDGFRß affibody-directed delivery of PS.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Colorectal Neoplasms/drug therapy , Photosensitizing Agents/administration & dosage , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Humans , Light , Mice , Mice, Inbred BALB C , Mice, Nude , Pericytes/drug effects , Photochemotherapy/methods , TNF-Related Apoptosis-Inducing Ligand/administration & dosage
17.
Theranostics ; 7(8): 2261-2276, 2017.
Article in English | MEDLINE | ID: mdl-28740549

ABSTRACT

Human tumor necrosis factor-related apoptosis-inducing ligand (hTRAIL) has exhibited superior in vitro cytotoxicity in a variety of tumor cells. However, hTRAIL showed a disappointing anticancer effect in clinical trials, although hTRAIL-based regimens were well tolerated. One important reason might be that hTRAIL was largely trapped by its decoy receptors, which are ubiquitously expressed on normal cells. Tumor-targeted delivery might improve the tumor uptake and thus enhance the antitumor effect of hTRAIL. Platelet-derived growth factor receptor ß (PDGFRß)-expressing pericytes are enriched in tumor tissues derived both from patients with colon cancer and from mice bearing colorectal tumor xenografts. A ZPDGFRß affibody showed high affinity (nM) for PDGFRß and was predominantly distributed on tumor-associated PDGFRß-positive pericytes. Co-administration with the ZPDGFRß affibody did not significantly enhance the antitumor effect of hTRAIL in mice bearing tumor xenografts. Fusion to the ZPDGFRß affibody endows hTRAIL with PDGFRß-binding ability but does not interfere with its death receptor binding and activation. The fused ZPDGFRß affibody mediated PDGFRß-dependent binding of hTRAIL to pericytes. In addition, hTRAIL bound on pericytes could kill tumor cells through juxtatropic activity or exhibit cytotoxicity in tumor cells after being released from pericytes. Intravenously injected hTRAIL fused to ZPDGFRß affibody initially accumulated on tumor-associated pericytes and then diffused to the tumor parenchyma over time. Fusion to the ZPDGFRß affibody increased the tumor uptake of hTRAIL, thus enhancing the antitumor effect of hTRAIL in mice bearing tumor xenografts. These results demonstrate that pericyte-targeted delivery mediated by a ZPDGFRß affibody is an alternative strategy for tumor-targeted delivery of anticancer agents.


Subject(s)
Autoantibodies/metabolism , Biological Products/pharmacology , Biological Products/pharmacokinetics , Colorectal Neoplasms/therapy , Pericytes/drug effects , Receptor, Platelet-Derived Growth Factor beta/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Administration, Intravenous , Animals , Autoantibodies/genetics , Biological Products/administration & dosage , Cells, Cultured , Disease Models, Animal , Heterografts , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Pericytes/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Treatment Outcome
18.
Mol Pharm ; 14(2): 502-512, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28029256

ABSTRACT

The poor in vivo potencies of most therapeutic proteins might be attributed to their short serum half-lives. PEGylation is a well-established method and has been clinically proven to improve pharmacokinetics. mmTRAIL exhibited supercytotoxicity in a variety of tumor cells, but its serum half-life was less than 10 min in mice. Here, mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were produced by N-terminus-specific PEGylation of mmTRAIL with 5, 10, or 20 kDa mPEG, respectively. The particle sizes of mmTRAIL-5K, mmTRAIL-10K, and mmTRAIL-20K were 9.09 ± 2.76, 12.62 ± 4.05, and 15.68 ± 4.95 nm, which were higher than the threshold (∼7 nm) of renal clearance. Accordingly, mmTRAIL-5K exhibited a serum half-life of 30 min only 3 times longer than that of mmTRAIL. However, both mmTRAIL-10K and mmTRAIL-20K exhibited similar serum half-lives ranging from 350 to 400 min, indicating that PEGylation with 10 or 20 kDa mPEG significantly improved the pharmacokinetics of mmTRAIL. However, death receptor binding of mmTRAIL-20K was reduced 5- to 8-fold, resulting in a 3-fold reduction of cytotoxicity. Additionally, repeated administration of mmTRAIL-20K elicited both mPEG-specific IgG and IgM antibody responses in rats. In contrast, the receptor binding and cytotoxicity of mmTRAIL-10K were similar to those of mmTRAIL. Repeated administration of mmTRAIL-10K did not obviously stimulate mPEG-specific antibody responses in rats and rhesus monkeys. Of the three PEGylated mmTRAIL analogues, mmTRAIL-10K exerted the greatest tumor suppression in mice bearing human tumor xenografts. These results demonstrated that conjugation of mmTRAIL to 10 kDa mPEG was better than that to 5 or 20 kDa mPEG for enhancing antitumor effects.


Subject(s)
Antibodies/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Polyethylene Glycols/chemistry , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/blood , Cell Line, Tumor , Half-Life , Humans , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Male , Mice , Mice, Inbred BALB C , Protein Binding/physiology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...