Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Adv Mater ; 35(10): e2209603, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36524741

ABSTRACT

Glutathione (GSH)-activatable probes hold great promise for in vivo cancer imaging, but are restricted by their dependence on non-selective intracellular GSH enrichment and uncontrollable background noise. Here, a holographically activatable nanoprobe caging manganese tetraoxide is shown for tumor-selective contrast enhancement in magnetic resonance imaging (MRI) through cooperative GSH/albumin-mediated cascade signal amplification in tumors and rapid elimination in normal tissues. Once targeting tumors, the endocytosed nanoprobe effectively senses the lysosomal microenvironment to undergo instantaneous decomposition into Mn2+ with threshold GSH concentration of ≈ 0.12 mm for brightening MRI signals, thus achieving high contrast tumor imaging and flexible monitoring of GSH-relevant cisplatin resistance during chemotherapy. Upon efficient up-regulation of extracellular GSH in tumor via exogenous injection, the relaxivity-silent interstitial nanoprobe remarkably evolves into Mn2+ that are further captured/retained and re-activated into ultrahigh-relaxivity-capable complex by stromal albumin in the tumor, and simultaneously allows the renal clearance of off-targeted nanoprobe in the form of Mn2+ via lymphatic vessels for suppressing background noise to distinguish tiny liver metastasis. These findings demonstrate the concept of holographic tumor activation via both tumor GSH/albumin-mediated cascade signal amplification and simultaneous background suppression for precise tumor malignancy detection, surveillance, and surgical guidance.


Subject(s)
Albumins , Glutathione , Magnetic Resonance Imaging , Metal Nanoparticles , Molecular Probes , Neoplasms , Glutathione/administration & dosage , Glutathione/pharmacokinetics , Glutathione/pharmacology , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Molecular Probes/pharmacology , Albumins/administration & dosage , Albumins/pharmacokinetics , Albumins/pharmacology , Magnetic Resonance Imaging/methods , Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Image Enhancement/methods , Holography/methods , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , Metal Nanoparticles/administration & dosage , Transferrin/administration & dosage , Transferrin/pharmacokinetics , Transferrin/pharmacology , Tissue Distribution , A549 Cells , Humans , Animals , Mice , Mice, Inbred BALB C , Mice, Nude , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Cisplatin/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology
2.
Molecules ; 26(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34500775

ABSTRACT

Thioredoxin-interacting protein (TXNIP) is involved in multiple disease-associated functions related to oxidative stress, especially by inhibiting the anti-oxidant- and thiol-reducing activity of thioredoxin (TXN). Shiga-Y5 (SY5), a fluorine-19 magnetic resonance probe for detecting amyloid-ß deposition in the brain, previously showed therapeutic effects in a mouse model of Alzheimer's disease; however, the mechanism of action of SY5 remains unclear. SY5 passes the blood-brain barrier and then undergoes hydrolysis to produce a derivative, Shiga-Y6 (SY6), which is a TXNIP-negative regulator. Therefore, this study investigates the therapeutic role of SY5 as the prodrug of SY6 in the thioredoxin system in the brain of a mouse model of Alzheimer's disease. The intraperitoneal injection of SY5 significantly inhibited TXNIP mRNA (p = 0.0072) and protein expression (p = 0.0143) induced in the brain of APP/PS1 mice. In contrast, the levels of TXN mRNA (p = 0.0285) and protein (p = 0.0039) in the brain of APP/PS1 mice were increased after the injection of SY5. The ratio of TXN to TXNIP, which was decreased (p = 0.0131) in the brain of APP/PS1 mice, was significantly increased (p = 0.0072) after the injection of SY5. These results suggest that SY5 acts as a prodrug of SY6 in targeting the thioredoxin system and could be a potential therapeutic compound in oxidative stress-related diseases in the brain.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Carrier Proteins/antagonists & inhibitors , Curcumin/pharmacology , Disease Models, Animal , Molecular Probes/pharmacology , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Curcumin/administration & dosage , Curcumin/analogs & derivatives , Fluorine , Injections, Intraperitoneal , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Molecular Structure
3.
Med Sci Monit ; 27: e930257, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34375323

ABSTRACT

BACKGROUND Albumin level does not precisely reflect nutritional status. We aimed to investigate the impact of a nutrition intervention on hemodialysis patients by use of fluorescence-based plasma albumin (FPA) detection. MATERIAL AND METHODS Eighty patients underwent maintenance hemodialysis for more than half a year and had a mean albumin <3.5 g/dL for over 3 months. The subjects were randomly divided into either a Control Group (CG) or an Intervention Group (IG). The IG received nutritional supplementation, and the CG group received routine nutritional support for 12 months. FPA and plasma albumin (PA) concentrations were measured. The fluorescence probe 1,3-Dichloro-7-hydroxy-9,9-dimethyl-2(9H)-acridone methyl biphenyl benzoate was used in FPA detection. Quality of life was estimated using WHOQOL-BREF (Quality of Life Scale developed through the World Health Organization), the 36-Item Short-Form Survey (SF-36), and the 6-minute walking test (6MWT). RESULTS After a 6-month and a 12-month intervention, PA and FPA concentrations increased, and the increase in FPA concentration was higher than that of PA in the IG group (P<0.05). Comparatively, the parameters of quality of life and 6MWT were improved in the IG group (P<0.05) but there were only minor changes in the CG group (P>0.05). There is an obvious association between the changes in FPA concentration and the parameters of quality of life and 6MWT but not PA. CONCLUSIONS Use of the fluorescence probe improves the detection sensitivity of plasma albumin and provides a potential method to assess clinical outcomes in hemodialysis patients.


Subject(s)
Kidney Failure, Chronic/therapy , Malnutrition/diagnosis , Nursing Care/methods , Nutritional Support/methods , Serum Albumin, Human/analysis , Adult , Female , Fluorescence , Humans , Kidney Failure, Chronic/complications , Male , Malnutrition/blood , Malnutrition/etiology , Malnutrition/therapy , Middle Aged , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Quality of Life , Renal Dialysis/adverse effects , Serum Albumin, Human/chemistry , Treatment Outcome , Walk Test
4.
Mol Pharm ; 18(5): 2082-2090, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33797932

ABSTRACT

The glypican-3 (GPC3) receptor is a membrane protein that is highly expressed in tumor tissues but rarely expressed in the normal liver and can be used as a target for early diagnosis of hepatocellular carcinoma (HCC). Herein, we developed a GPC3-targeted 99mTc-labeled probe for SPECT imaging in HCC. 99mTc-HPG was rapidly radiosynthesized within 20 min with an excellent radiochemical purity (>98%), possessing good stability. Results from in vitro cell binding assays indicated that the binding specificity of 99mTc-HPG to GPC3-positive HepG2 cells was acceptable. For SPECT/CT imaging, the HepG2 tumors were clearly visualized with the highest tumor/muscle ratio (11.55 ± 0.54) at 1 h post-injection, and the tumor uptake of 99mTc-HPG reduced from 2.99 ± 0.15 to 1.17 ± 0.09% ID/g in the blocking study. Convenient preparation, excellent GPC3 specificity in HCC, rapid clearance from normal organs, and good biosafety profiles of 99mTc-HPG warrant further investigations for clinical translation.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Glypicans/metabolism , Liver Neoplasms/diagnosis , Radiopharmaceuticals/administration & dosage , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Carcinoma, Hepatocellular/pathology , Female , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Mice , Molecular Imaging/methods , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Organotechnetium Compounds/administration & dosage , Organotechnetium Compounds/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Technetium , Tissue Distribution , Xenograft Model Antitumor Assays
5.
Nat Commun ; 12(1): 2385, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888701

ABSTRACT

Nanoparticle internalisation is crucial for the precise delivery of drug/genes to its intracellular targets. Conventional quantification strategies can provide the overall profiling of nanoparticle biodistribution, but fail to unambiguously differentiate the intracellularly bioavailable particles from those in tumour intravascular and extracellular microenvironment. Herein, we develop a binary ratiometric nanoreporter (BiRN) that can specifically convert subtle pH variations involved in the endocytic events into digitised signal output, enabling the accurately quantifying of cellular internalisation without introducing extracellular contributions. Using BiRN technology, we find only 10.7-28.2% of accumulated nanoparticles are internalised into intracellular compartments with high heterogeneity within and between different tumour types. We demonstrate the therapeutic responses of nanomedicines are successfully predicted based on intracellular nanoparticle exposure rather than the overall accumulation in tumour mass. This nonlinear optical nanotechnology offers a valuable imaging tool to evaluate the tumour targeting of new nanomedicines and stratify patients for personalised cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/analysis , Molecular Imaging/methods , Nanoparticles/analysis , Neoplasms/drug therapy , Animals , Cell Line, Tumor/transplantation , Disease Models, Animal , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , Fluorescent Dyes/chemistry , Humans , Intravital Microscopy , Mice , Molecular Probes/administration & dosage , Molecular Probes/analysis , Molecular Probes/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods , Paclitaxel/administration & dosage , Patient Selection , Reproducibility of Results , Theranostic Nanomedicine/methods , Tissue Distribution , Tumor Microenvironment/drug effects
6.
Future Oncol ; 17(6): 711-722, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33136464

ABSTRACT

In recent years, cancer immunotherapy has evolved as an exciting novel strategy for researchers and clinicians worldwide. Immunotherapeutic agents such as immune checkpoint blockers have changed the standard-of-care treatment provided for many tumors. Unfortunately, only a small proportion of patients respond effectively to these checkpoint inhibitors. Moreover, the immunosuppressive pathways for cancer are probably too complicated to achieve optimal outcome with immune checkpoint inhibitors alone. Combining current therapeutic options and immunotherapy-based approaches is being explored as an effective strategy to treat cancer. The use of nanotechnology-based platforms for delivery of immunotherapeutic agents or combination therapy could offer a major advantage over conventional anticancer treatment options. This review highlights the potential role of different nanotechnology-based strategies in improving the efficacy of immune checkpoint blockade therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems/methods , Immune Checkpoint Inhibitors/administration & dosage , Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Liposomes , Molecular Imaging/methods , Molecular Probes/administration & dosage , Nanoparticles/administration & dosage , Neoplasms/diagnostic imaging , Neoplasms/immunology , Theranostic Nanomedicine/trends , Treatment Outcome
7.
Sci Rep ; 10(1): 20777, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247180

ABSTRACT

Radionuclide molecular imaging of cancer-specific targets is a promising method to identify patients for targeted antibody therapy. Radiolabeled full-length antibodies however suffer from slow clearance, resulting in high background radiation. To overcome this problem, a pretargeting system based on complementary peptide nucleic acid (PNA) probes has been investigated. The pretargeting relies on sequential injections of primary, PNA-tagged antibody and secondary, radiolabeled PNA probe, which are separated in time, to allow for clearance of non-bound primary agent. We now suggest to include a clearing agent (CA), designed for removal of primary tumor-targeting agent from the blood. The CA is based on the antibody cetuximab, which was conjugated to PNA and lactosaminated by reductive amination to improve hepatic clearance. The CA was evaluated in combination with PNA-labelled trastuzumab, T-ZHP1, for radionuclide HER2 pretargeting. Biodistribution studies in normal mice demonstrated that the CA cleared ca. 7 times more rapidly from blood than unmodified cetuximab. Injection of the CA 6 h post injection of the radiolabeled primary agent [131I]I-T-ZHP1 gave a moderate reduction of the radioactivity concentration in the blood after 1 h from 8.5 ± 1.8 to 6.0 ± 0.4%ID/g. These proof-of-principle results could guide future development of a more efficient CA.


Subject(s)
Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/chemistry , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/chemistry , Radioimmunotherapy/methods , Animals , Antibodies, Neoplasm/metabolism , Cell Line, Tumor , Cetuximab/administration & dosage , Cetuximab/blood , Cetuximab/chemistry , Female , Humans , Immunoconjugates/pharmacokinetics , Mice , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Molecular Probes/pharmacokinetics , Molecular Targeted Therapy/methods , Peptide Nucleic Acids/pharmacokinetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Tissue Distribution , Trastuzumab/administration & dosage , Trastuzumab/blood , Trastuzumab/chemistry
8.
Pharm Res ; 37(12): 237, 2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33151373

ABSTRACT

PURPOSE: Hyaluronan (HA) based biomaterials are widely used as tissue scaffolds, drug formulations, as well as targeting ligands and imaging probes for diagnosis and drug delivery. However, because of the presence of abundant endogenous HA presented in various tissues in vivo, the pharmacokinetic behavior and biodistribution patterns of exogenously administered HAs have not been well characterized. METHODS: The HA backbone was modified with Diethylenetriamine (DTPA) to enable the chelation of gadolinium (Gd) and aluminum (Al) ions. Series of PET and MR imaging were taken after the injection of HA-DTPA-Gd and HA-DTPA-Al18F while using18F-FDG and Magnevist(DTPA-Gd) as controls. The Tomographic images were analyzed and quantified to reveal the distribution and locations of HA in tumor-bearing mice. RESULTS: The labeled HAs had good stability in plasma. They retained binding affinity towards CD44s on tumor cell surface. The injected HAs distributed widely in various organs, but were found to be cleared quickly except inside tumor tissues where the signals were higher and persisted longer. CONCLUSION: Medical imaging tools, including MR and PET, can be highly valuable for examining biomaterial distribution non-invasively. The HA tumor accumulation properties may be explored for the development of active targeting drug carriers and molecular probes.


Subject(s)
Biocompatible Materials/pharmacokinetics , Drug Carriers/pharmacokinetics , Hyaluronic Acid/pharmacokinetics , Molecular Probes/pharmacokinetics , A549 Cells , Animals , Biocompatible Materials/administration & dosage , Contrast Media , Drug Carriers/administration & dosage , Humans , Hyaluronic Acid/administration & dosage , Magnetic Resonance Imaging , Male , Mice , Molecular Probes/administration & dosage , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/pathology , Positron-Emission Tomography , Tissue Distribution , Xenograft Model Antitumor Assays
9.
Discov Med ; 29(158): 169-180, 2020.
Article in English | MEDLINE | ID: mdl-33007192

ABSTRACT

Aptamers are short single-stranded oligonucleotides that have attracted considerable attention due to their favorable biological characteristics. Aptamers can specifically target and bind to proteins or tumor cells, achieving tumor diagnosis and therapy in vitro and in vivo. Following an introduction of methodologies of producing aptamers and the recent advances of aptamers being applied to clinical samples or xenograft tumors, tumor diagnosis using aptamers will be reviewed, including fluorescence imaging, radionuclide-based imaging, MRI, histochemical imaging, and multimodality imaging. Preclinical applications in tumor therapy in vivo will also be discussed, covering different kinds of treatment mechanisms, including aptamer therapeutics, chemotherapy, gene therapy, immunotherapy, and combination therapy. Safety and efficacy of tumor-targeting therapeutics via aptamers, as well as the current challenges and future perspectives about aptamers' clinical applications, will be summarized.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , Drug Carriers/chemical synthesis , Molecular Probes/administration & dosage , Neoplasms/therapy , Animals , Antineoplastic Agents/chemical synthesis , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/pharmacology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Combined Modality Therapy/methods , Combined Modality Therapy/trends , Disease Models, Animal , Drug Screening Assays, Antitumor , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Immunotherapy/methods , Immunotherapy/trends , Molecular Imaging/methods , Molecular Imaging/trends , Molecular Probes/chemical synthesis , Molecular Probes/pharmacology , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/pathology , SELEX Aptamer Technique/trends
10.
Theranostics ; 10(25): 11607-11621, 2020.
Article in English | MEDLINE | ID: mdl-33052236

ABSTRACT

The hypoxic microenvironment in solid tumors severely limits the efficacy of photodynamic therapy (PDT). Therefore, the development of nanocarriers co-loaded with photosensitizers and oxygen, together with imaging guidance ability, is of great significance in cancer therapy. However, previously reported synthetic methods for these multi-functional probes are complicated, and the raw materials used are toxic. Methods: Herein, the human endogenous protein, hemoglobin (Hb), was used for the simultaneous biomimetic synthesis of Gd-based nanostructures and co-loading of Chlorine e6 (Ce6) and oxygen for alleviating the hypoxic environment of tumors and accomplishing magnetic resonance imaging (MRI)-guided enhanced PDT. The Gd@HbCe6-PEG nanoprobes were synthesized via a green and protein biomimetic approach. The physicochemical properties, including relaxivity, oxygen-carrying/release capability, and PDT efficacy of Gd@HbCe6-PEG, were measured in vitro and in vivo on tumor-bearing mice after intravenous injection. Morphologic and functional MRI were carried out to evaluate the efficacy of PDT. Results: The results demonstrated the successful synthesis of compact Gd@HbCe6-PEG nanostructures with desired multi-functionalities. Following treatment with the nanoparticles, the embedded MR moiety was effective in lighting tumor lesions and guiding therapy. The oxygen-carrying capability of Hb after biomimetic synthesis was confirmed by spectroscopic analysis and oxygen detector in vitro. Further, tumor oxygenation for alleviating tumor hypoxia in vivo after intravenous injection of Gd@HbCe6-PEG was verified by photoacoustic imaging and immunofluorescence staining. The potent treatment efficacy of PDT on early-stage was observed by the morphologic and functional MR imaging. Importantly, rapid renal clearance of the particles was observed after treatment. Conclusion: In this study, by using a human endogenous protein, we demonstrated the biomimetic synthesis of multi-functional nanoprobes for simultaneous tumor oxygenation and imaging-guided enhanced PDT. The therapeutic efficacy could be quantitatively confirmed at 6 h post PDT with diffusion-weighted imaging (DWI).


Subject(s)
Antineoplastic Agents/administration & dosage , Metal Nanoparticles/administration & dosage , Neoplasms/drug therapy , Photochemotherapy/methods , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemical synthesis , Biomimetic Materials/pharmacokinetics , Cell Line, Tumor/transplantation , Chlorophyllides , Diffusion Magnetic Resonance Imaging , Disease Models, Animal , Female , Gadolinium/administration & dosage , Gadolinium/chemistry , Green Chemistry Technology , Hemoglobins/administration & dosage , Hemoglobins/chemistry , Humans , Injections, Intravenous , Metal Nanoparticles/chemistry , Mice , Molecular Probes/administration & dosage , Molecular Probes/chemical synthesis , Molecular Probes/pharmacokinetics , Neoplasms/diagnostic imaging , Oxygen/administration & dosage , Oxygen/chemistry , Photoacoustic Techniques , Porphyrins/administration & dosage , Porphyrins/chemistry , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects
11.
Mol Pharm ; 17(12): 4589-4602, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33108189

ABSTRACT

Prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer cells and therefore is an attractive target for prostate cancer diagnosis and radionuclide therapy. Recently, published results from clinical studies using a new PSMA-targeting PET imaging agent, [68Ga]Ga-PSMA-093 ([68Ga]Ga-HBED-CC-O-carboxymethyl-Tyr-CO-NH-Glu), support the development of this agent for the diagnosis of prostate cancer. In this study, the HBED-CC chelating group in PSMA-093 was replaced by stereoselective (R)- or (S)-DOTAGA. This chelating group serves not only for chelating 68Ga but is also amendable for complexing other radioactive metals for radionuclide therapy. The corresponding optically pure (R)- and (S)-[68Ga/177Lu]-DOTAGA derivatives, (R)-[68Ga/177Lu]-13 and (S)-[68Ga/177Lu]-13, were successfully prepared. Comparison of radiolabeling, binding affinity, cell uptake, and biodistribution between the two isomers was performed. Radiolabeling of (R)-[177Lu]Lu-13 and (S)-[177Lu]Lu-13 at 50 °C suggested that rates of complex formation were time-dependent and the formation of (S)-[177Lu]Lu-13 was distinctly faster. The rates of complex formation for the corresponding 68Ga agents were comparable between structural isomers. The natGa and natLu equivalents showed high binding PSMA affinity (IC50 = 24-111 nM), comparable to that of the parent agent, [natGa]Ga-PSMA-093 (IC50 = 34.0 nM). Results of cell uptake and biodistribution studies in PSMA-expressing PC3-PIP tumor-bearing mice appeared to show no difference between the labeled (R)- and (S)-isomers. This is the first time that a pair of [68Ga/177Lu]-(R)- and (S)-DOTAGA isomers of PSMA agents were evaluated. Results of biological studies between the isomers showed no noticeable difference; however, the distinctions on the rate of Lu complex formation should be considered in the development of new 177Lu-DOTAGA-based radionuclide therapy agents in the future.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Molecular Probes/pharmacokinetics , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Animals , Cell Line, Tumor , Chelating Agents/administration & dosage , Chelating Agents/chemical synthesis , Chelating Agents/pharmacokinetics , Gallium Radioisotopes , Humans , Inhibitory Concentration 50 , Lutetium , Male , Mice , Molecular Imaging/methods , Molecular Probes/administration & dosage , Molecular Probes/chemical synthesis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Radioisotopes , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemical synthesis , Stereoisomerism , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 26(18): 5007-5018, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32611647

ABSTRACT

PURPOSE: To evaluate the prognostic value of posttreatment fibrosis in human PDAC patients, and to compare a type I collagen targeted MRI probe, CM-101, to the standard contrast agent, Gd-DOTA, for their abilities to identify FOLFIRINOX-induced fibrosis in a murine model of PDAC. EXPERIMENTAL DESIGN: Ninety-three chemoradiation-treated human PDAC samples were stained for fibrosis and outcomes evaluated. For imaging, C57BL/6 and FVB mice were orthotopically implanted with PDAC cells and FOLFIRINOX was administered. Mice were imaged with Gd-DOTA and CM-101. RESULTS: In humans, post-chemoradiation PDAC tumor fibrosis was associated with longer overall survival (OS) and disease-free survival (DFS) on multivariable analysis (OS P = 0.028, DFS P = 0.047). CPA increased the prognostic accuracy of a multivariable logistic regression model comprised of previously established PDAC risk factors [AUC CPA (-) = 0.76, AUC CPA (+) = 0.82]. In multiple murine orthotopic PDAC models, FOLFIRINOX therapy reduced tumor weight (P < 0.05) and increased tumor fibrosis by collagen staining (P < 0.05). CM-101 MR signal was significantly increased in fibrotic tumor regions. CM-101 signal retention was also increased in the more fibrotic FOLFIRINOX-treated tumors compared with untreated controls (P = 0.027), consistent with selective probe binding to collagen. No treatment-related differences were observed with Gd-DOTA imaging. CONCLUSIONS: In humans, post-chemoradiation tumor fibrosis is associated with OS and DFS. In mice, our MR findings indicate that translation of collagen molecular MRI with CM-101 to humans might provide a novel imaging technique to monitor fibrotic response to therapy to assist with prognostication and disease management.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Chemoradiotherapy, Adjuvant , Neoadjuvant Therapy/methods , Neoplasm Recurrence, Local/epidemiology , Pancreas/pathology , Pancreatic Neoplasms/therapy , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Collagen/analysis , Collagen/metabolism , Disease-Free Survival , Female , Fibrosis , Fluorouracil/administration & dosage , Follow-Up Studies , Heterocyclic Compounds/administration & dosage , Humans , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Magnetic Resonance Imaging/methods , Male , Mice , Middle Aged , Molecular Imaging/methods , Molecular Probes/administration & dosage , Neoplasm Recurrence, Local/prevention & control , Organometallic Compounds/administration & dosage , Oxaliplatin/administration & dosage , Pancreas/diagnostic imaging , Pancreas/surgery , Pancreatectomy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Analysis , Xenograft Model Antitumor Assays
13.
Clin Cancer Res ; 26(17): 4581-4589, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32499234

ABSTRACT

PURPOSE: Targeted therapies for cancer have accelerated the need for functional imaging strategies that inform therapeutic efficacy. This study assesses the potential of functional genetic screening to integrate therapeutic target identification with imaging probe selection through a proof-of-principle characterization of a therapy-probe pair using dynamic nuclear polarization (DNP)-enhanced magnetic resonance spectroscopic imaging (MRSI). EXPERIMENTAL DESIGN: CRISPR-negative selection screens from a public dataset were used to identify the relative dependence of 625 cancer cell lines on 18,333 genes. Follow-up screening was performed in hepatocellular carcinoma with a focused CRISPR library targeting imaging-related genes. Hyperpolarized [1-13C]-pyruvate was injected before and after lactate dehydrogenase inhibitor (LDHi) administration in male Wistar rats with autochthonous hepatocellular carcinoma. MRSI evaluated intratumoral pyruvate metabolism, while T2-weighted segmentations quantified tumor growth. RESULTS: Genetic screening data identified differential metabolic vulnerabilities in 17 unique cancer types that could be imaged with existing probes. Among these, hepatocellular carcinoma required lactate dehydrogenase (LDH) for growth more than the 29 other cancer types in this database. LDH inhibition led to a decrease in lactate generation (P < 0.001) and precipitated dose-dependent growth inhibition (P < 0.01 overall, P < 0.05 for dose dependence). Intratumoral alanine production after inhibition predicted the degree of growth reduction (P < 0.001). CONCLUSIONS: These findings demonstrate that DNP-MRSI of LDH activity using hyperpolarized [1-13C]-pyruvate is a theranostic strategy for hepatocellular carcinoma, enabling quantification of intratumoral LDHi pharmacodynamics and therapeutic efficacy prediction. This work lays the foundation for a novel theranostic platform wherein functional genetic screening informs imaging probe selection to quantify therapeutic efficacy on a cancer-by-cancer basis.


Subject(s)
Carbon-13 Magnetic Resonance Spectroscopy/methods , Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms, Experimental/diagnosis , Liver Neoplasms/diagnosis , Molecular Imaging/methods , Animals , CRISPR-Cas Systems/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Datasets as Topic , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Early Detection of Cancer/methods , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Liver/diagnostic imaging , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Molecular Probes/administration & dosage , Molecular Probes/pharmacokinetics , Precision Medicine/methods , Proof of Concept Study , Pyruvic Acid/metabolism , Rats
14.
Trends Cancer ; 6(8): 645-659, 2020 08.
Article in English | MEDLINE | ID: mdl-32448754

ABSTRACT

Tumor metastases, that is, the development of secondary tumors in organs distant from the primary tumor, and their treatment remain a serious problem in cancer therapy. The unique challenges for tracking and treating tumor metastases lie in the small size, high heterogeneity, and wide dispersion to distant organs of metastases. Recently, nanomedicines, with the capacity to precisely deliver therapeutic agents to both primary and secondary tumors, have demonstrated many potential benefits for metastatic cancer theranostics. Given the remarkable progression in emerging nanotherapeutics for antimetastatic treatment, it is timely to summarize the latest advances in this field. This review highlights the rationale, advantages, and challenges for integrating biomedical nanotechnology with cancer biology to develop antimetastatic nanotherapeutics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Nanoparticles/administration & dosage , Neoplasm Metastasis/drug therapy , Theranostic Nanomedicine/methods , Tumor Microenvironment/drug effects , Humans , Molecular Imaging/methods , Molecular Probes/administration & dosage , Neoplasm Metastasis/diagnosis , Neoplastic Cells, Circulating/drug effects
15.
Sci Rep ; 10(1): 6244, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32277103

ABSTRACT

Glutathione (GSH) is often upregulated in cancer, where it serves to mitigate oxidative stress. γ-glutamyl-transferase (GGT) is a key enzyme in GSH homeostasis, and compared to normal brain its expression is elevated in tumors, including in primary glioblastoma. GGT is therefore an attractive imaging target for detection of glioblastoma. The goal of our study was to assess the value of hyperpolarized (HP) γ-glutamyl-[1-13C]glycine for non-invasive imaging of glioblastoma. Nude rats bearing orthotopic U87 glioblastoma and healthy controls were investigated. Imaging was performed by injecting HP γ-glutamyl-[1-13C]glycine and acquiring dynamic 13C data on a preclinical 3T MR scanner. The signal-to-noise (SNR) ratios of γ-glutamyl-[1-13C]glycine and its product [1-13C]glycine were evaluated. Comparison of control and tumor-bearing rats showed no difference in γ-glutamyl-[1-13C]glycine SNR, pointing to similar delivery to tumor and normal brain. In contrast, [1-13C]glycine SNR was significantly higher in tumor-bearing rats compared to controls, and in tumor regions compared to normal-appearing brain. Importantly, higher [1-13C]glycine was associated with higher GGT expression and higher GSH levels in tumor tissue compared to normal brain. Collectively, this study demonstrates, to our knowledge for the first time, the feasibility of using HP γ-glutamyl-[1-13C]glycine to monitor GGT expression in the brain and thus to detect glioblastoma.


Subject(s)
Brain/diagnostic imaging , Glioblastoma/diagnosis , Magnetic Resonance Imaging/methods , Molecular Imaging/methods , gamma-Glutamyltransferase/metabolism , Animals , Brain/pathology , Carbon Isotopes/administration & dosage , Carbon Isotopes/chemistry , Cell Line, Tumor , Dipeptides/administration & dosage , Dipeptides/chemistry , Feasibility Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Male , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Rats , Up-Regulation , Xenograft Model Antitumor Assays
16.
Nat Commun ; 11(1): 1250, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144257

ABSTRACT

Currently, there are no non-invasive tools to accurately diagnose wound and surgical site infections before they become systemic or cause significant anatomical damage. Fluorescence and photoacoustic imaging are cost-effective imaging modalities that can be used to noninvasively diagnose bacterial infections when paired with a molecularly targeted infection imaging agent. Here, we develop a fluorescent derivative of maltotriose (Cy7-1-maltotriose), which is shown to be taken up in a variety of gram-positive and gram-negative bacterial strains in vitro. In vivo fluorescence and photoacoustic imaging studies highlight the ability of this probe to detect infection, assess infection burden, and visualize the effectiveness of antibiotic treatment in E. coli-induced myositis and a clinically relevant S. aureus wound infection murine model. In addition, we show that maltotriose is an ideal scaffold for infection imaging agents encompassing better pharmacokinetic properties and in vivo stability than other maltodextrins (e.g. maltohexose).


Subject(s)
Fluorescent Dyes/administration & dosage , Molecular Imaging/methods , Myositis/diagnostic imaging , Surgical Wound Infection/diagnostic imaging , Trisaccharides/administration & dosage , Animals , Carbocyanines/administration & dosage , Carbocyanines/chemistry , Disease Models, Animal , Drug Stability , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Injections, Intravenous , Luminescent Measurements/methods , Mice , Microscopy, Fluorescence/methods , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Molecular Probes/metabolism , Myositis/microbiology , Photoacoustic Techniques/methods , Rats , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Surgical Wound Infection/microbiology , Trisaccharides/chemistry , Trisaccharides/metabolism
17.
Chem Commun (Camb) ; 56(29): 4106-4109, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32163087

ABSTRACT

Herein we report a pair of redox-responsive manganese complexes Mn(iii)/(ii)-N,N'-bis(2-hydroxy-4-trifluoromethylbenzyl)ethylenediamine-N,N'-diacetate (HTFBED, L1), which are water soluble and biologically interconvertible, as reversible redox-responsive probes in 1H/19F MRI for detecting and imaging biological redox species, offering a means to access valuable redox information associated with various diseases.


Subject(s)
Coordination Complexes , Edetic Acid , Manganese , Molecular Probes , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Edetic Acid/administration & dosage , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Hep G2 Cells , Humans , Magnetic Resonance Imaging , Manganese/administration & dosage , Manganese/chemistry , Molecular Probes/administration & dosage , Molecular Probes/chemistry , Oxidation-Reduction , Pyocyanine/pharmacology
18.
Chem Commun (Camb) ; 56(28): 3999-4002, 2020 Apr 11.
Article in English | MEDLINE | ID: mdl-32154536

ABSTRACT

Novel red Zn(ii) complex-based fluorescent probes featuring cryptolepine-curcumin derivatives, namely, [Zn(BQ)Cl2] (BQ-Zn) and [Zn(BQ)(Cur)]Cl (BQCur-Zn), were developed for the simple and fluorescent label-free detection of apoptosis, an important biological process. The probes could synergistically promote mitochondrion-mediated apoptosis and enhance tumor therapeutic effects in vitro and vivo.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Curcumin/administration & dosage , Fluorescent Dyes/administration & dosage , Indole Alkaloids/administration & dosage , Molecular Probes/administration & dosage , Quinolines/administration & dosage , Zinc/administration & dosage , Animals , Cell Line, Tumor , Humans , Mice , Neoplasms/drug therapy , Photochemotherapy
19.
J Pharm Pharm Sci ; 22(1): 567-575, 2019.
Article in English | MEDLINE | ID: mdl-31804919

ABSTRACT

PURPOSE: Probe substrates are used routinely to assess transporter function in vitro. Administration of multiple probe substrates together as a "cocktail" in sandwich-cultured human hepatocytes (SCHH) could increase the throughput of transporter function assessment in a physiologically-relevant in vitro system. This study was designed to compare transporter function between cocktail and single agent administration in SCHH. METHODS: Rosuvastatin, digoxin, and metformin were selected as probe substrates of hepatic transporters OATP1B1, OATP1B3, BCRP, P-gp, and OCT1. Total accumulation (Cells+Bile) and biliary excretion index (BEI) values derived from administration of the cocktail were compared to values obtained after administration of single agents in the absence and presence of a model inhibitor, erythromycin estolate. RESULTS: For rosuvastatin and metformin accumulation, the ratio of means [90% confidence interval (CI)] for cocktail to single agent administration was 100% [94%, 106%] and 90% [82%, 99%], respectively. Therefore, the cocktail and single-agent mode of administration were deemed equivalent per standard equivalence criterion of 80-120% for rosuvastatin and metformin accumulation, but not for digoxin accumulation (77% [62%, 92%]). The ratio of means [90% CI] for rosuvastatin BEI values between the two administration modes (105% [97%, 114%]) also was deemed equivalent. The ratio for digoxin BEI values between the two administration modes was 99% [78%, 120%]. In the presence of erythromycin estolate, the two administration modes were deemed equivalent for evaluation of rosuvastatin, digoxin, and metformin accumulation; the ratio of means [90% CI] was 104% [94%, 115%], 94% [82%, 105%], and 100% [88%, 111%], respectively. However, rosuvastatin and digoxin BEI values were low and quite variable in the presence of the inhibitor, so the BEI results were inconclusive. CONCLUSIONS: These data suggest that rosuvastatin and metformin can be administered as a cocktail to evaluate the function of OATP1B1, OATP1B3, BCRP, and OCT1 in SCHH, and that digoxin may not be an ideal component of such a cocktail.


Subject(s)
Cell Culture Techniques , Hepatocytes/metabolism , Membrane Transport Proteins/metabolism , Molecular Probes/chemistry , Biological Transport , Cells, Cultured , Digoxin/administration & dosage , Digoxin/chemistry , Digoxin/metabolism , Erythromycin Estolate/administration & dosage , Erythromycin Estolate/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Metformin/administration & dosage , Metformin/chemistry , Metformin/metabolism , Molecular Probes/administration & dosage , Molecular Probes/metabolism , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/metabolism
20.
Theranostics ; 9(23): 6962-6975, 2019.
Article in English | MEDLINE | ID: mdl-31660080

ABSTRACT

Purpose: Positron emission tomography (PET) imaging of apoptosis is very important for early evaluation of tumor therapeutic efficacy. A stimuli-responsive probe based on the peptide sequence Asp-Glu-Val-Asp (DEVD), [18F]DEVD-Cys(StBu)-PPG(CBT)-AmBF3 ([18F]1), for PET imaging of tumor apoptosis was designed and prepared. This study aimed to develop a novel smart probe using a convenient radiosynthesis method and to fully examine the sensitivity and specificity of the probe response to the tumor treatment. Methods: The radiolabelling precursor DEVD-Cys(StBu)-PPG(CBT)-AmBF3 (1) was synthesized through multistep reactions. The reduction together with caspase-controlled macrocyclization and self-assembly of 1 was characterized and validated in vitro. After [18F]fluorination in the buffer (pH= 2.5), the radiolabelling yield (RLY), radiochemical purity (RCP) and stability of the probe [18F]1 in PBS and mouse serum were investigated by radio-HPLC. The sensitivity and specificity of [18F]1 for detecting the drug-induced apoptosis was fully evaluated in vitro and in vivo. The effect of cold precursor 1 on the cell uptake and tumor imaging of [18F]1 was also assessed. The level of activated caspase-3 in Hela cells and tumors with or without apoptosis induction was analyzed and compared by western blotting and histological staining. Results: The whole radiosynthesis process of [18F]1 was around 25 min with RLY of 50%, RCP of over 99% and specific activity of 1.45 ± 0.4 Ci/µmol. The probe was very stable in both PBS and mouse serum within 4 h. It can be activated by caspase-3 and then undergo an intermolecular cyclization to form nanosized particles. The retained [18F]1 in DOX-treated HeLa cells was 2.2 folds of that in untreated cells. Within 1 h microPET imaging of the untreated Hela-bearing mice, the injection of [18F]1 resulted in the increase of the uptake ratio of tumor to muscle (T/M) only from 1.74 to 2.18, while in the DOX-treated Hela-bearing mice T/M increased from 1.88 to 10.52 and the co-injection of [18F]1 and 1 even led to the increase of T/M from 3.08 to 14.81. Conclusions: A caspase-responsive smart PET probe [18F]1 was designed and prepared in a kit-like manner. Co-injection of [18F]1 and 1 generated remarkably enhanced tumor uptake and signal-to-noise ratio in the tumor-bearing mice with drug-induced apoptosis, which correlated well with the expression level of activated caspase-3. This early readout of treatment response ensured that the probe [18F]1 could serve as a promising PET imaging probe for timely and noninvasive evaluation of tumor therapy.


Subject(s)
Apoptosis , Caspase 3/metabolism , Fluorine Radioisotopes/chemistry , Molecular Probes/chemistry , Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Doxorubicin/administration & dosage , Fluorine Radioisotopes/administration & dosage , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Molecular Probes/administration & dosage , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/physiopathology , Positron-Emission Tomography/instrumentation , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...